DOI QR코드

DOI QR Code

Administration of red ginseng ameliorates memory decline in aged mice

  • Lee, Yeonju (Department of Molecular Medicine and TIDRC, School of Medicine, Ewha Womans University) ;
  • Oh, Seikwan (Department of Molecular Medicine and TIDRC, School of Medicine, Ewha Womans University)
  • Received : 2014.08.06
  • Accepted : 2015.01.05
  • Published : 2015.07.15

Abstract

Background: It has been known that ginseng can be applied as a potential nutraceutical for memory impairment; however, experiments with animals of old age are few. Methods: To determine the memory enhancing effect of red ginseng, C57BL/6 mice (21 mo old) were given experimental diet pellets containing 0.12% red ginseng extract (approximately 200 mg/kg/d) for 3 mo. Young and old mice (4 mo and 21 mo old, respectively) were used as the control group. The effect of red ginseng, which ameliorated memory impairment in aged mice, was quantified using Y-maze test, novel objective test, and Morris water maze. Red ginseng ameliorated age-related declines in learning and memory in older mice. In addition, red ginseng's effect on the induction of inducible nitric oxide synthase and proinflammatory cytokines was investigated in the hippocampus of aged mice. Results: Red ginseng treatment suppressed the production of age-processed inducible nitric oxide synthase, cyclooxygenase-2, tumor necrosis factor-${\alpha}$, and interleukin-$1{\beta}$ expressions. Moreover, it was observed that red ginseng had an antioxidative effect on aged mice. The suppressed glutathione level in aged mice was restored with red ginseng treatment. The antioxidative-related enzymes Nrf2 and HO-1 were increased with red ginseng treatment. Conclusion: The results revealed that when red ginseng is administered over long periods, age-related decline of learning and memory is ameliorated through anti-inflammatory activity.

Keywords

References

  1. Barnes CA, McNaughton BL. An age comparison of the rates of acquisition and forgetting of spatial information in relation to long-term enhancement of hippocampal synapses. Behav Neurosci 1985;99:1040-8. https://doi.org/10.1037/0735-7044.99.6.1040
  2. Gallagher M, Pelleymounter MA. An age-related spatial learning deficit: choline uptake distinguishes "impaired" and "unimpaired" rats. Neurobiol Aging 1988;9:363-9. https://doi.org/10.1016/S0197-4580(88)80082-4
  3. Rapp PR, Gallagher M. Preserved neuron number in the hippocampus of aged rats with spatial learning deficits. Proc Natl Acad Sci U S A 1996;93:9926-30. https://doi.org/10.1073/pnas.93.18.9926
  4. Zhong YM, Nishijo H, Uwano T, Tamura R, Kawanishi K, Ono T. Red ginseng ameliorated place navigation deficits in young rats with hippocampal lesions and aged rats. Physiol Behav 2000;69:511-25. https://doi.org/10.1016/S0031-9384(00)00206-7
  5. Ott BR, Saver JL. Unilateral amnesic stroke. Six new cases and a review of the literature. Stroke 1993;24:1033-42. https://doi.org/10.1161/01.STR.24.7.1033
  6. Heinonen O, Soininen H, Sorvari H, Kosunen O, Paljarvi L, Koivisto E, Riekkinen Sr PJ. Loss of synaptophysin-like immunoreactivity in the hippocampal formation is an early phenomenon in Alzheimer's disease. Neurosci 1995;64: 375-84. https://doi.org/10.1016/0306-4522(94)00422-2
  7. Mizutani T, Kashara M. Degeneration of the intrahippocampal routes of the perforant and alvear pathways in senile dementia of Alzheimer type. Neurosci Lett 1995;184:141-4. https://doi.org/10.1016/0304-3940(94)11190-T
  8. Carr DB, Goate A, Phil D, Morris JC. Current concepts in the pathogenesis of Alzheimer's disease. Am J Med 1997;103:3S-10S.
  9. Corey-Bloom J, Sabbagh MN, Bondi MW, Hansen L, Alford MF, Masliah E, Thal LJ. Hippocampal sclerosis contributes to dementia in the elderly. Neurology 1997;48:154-60. https://doi.org/10.1212/WNL.48.1.154
  10. Ames BN, Shigenaga MK, Hagen TM. Oxidants, antioxidants, and the degenerative diseases of aging. Proc Natl Acad Sci U S A 1993;90:7915-22. https://doi.org/10.1073/pnas.90.17.7915
  11. Goto S, Takahashi R, Kumiyama AA, Radak Z, Hayashi T, Takenouchi M, Abe R. Implications of protein degradation in aging. Ann N Y Acad Sci 2001;928:54-64.
  12. Johnson FB, Sinclair DA, Guarente L. Molecular biology of aging. Cell 1999;96: 291-302. https://doi.org/10.1016/S0092-8674(00)80567-X
  13. Chung HY, Kim HJ, Kim KW, Choi JS, Yu BP. Molecular inflammation hypothesis of aging based on the anti-aging mechanism of calorie restriction. Microsc Res Tech 2002;59:264-72. https://doi.org/10.1002/jemt.10203
  14. Terasawa K, Shimada Y, Kita T, Yamamoto T, Tosa H, Tanaka N, Saito Y, Kanaki E, Goto S, Mizushima N, et al. Choto-san in the treatment of vascular dementia: a double-blind, placebo-controlled study. Phytomedicine 1997;4: 15-22. https://doi.org/10.1016/S0944-7113(97)80022-0
  15. Tang W, Eisenbrand G. Chinese drugs of plant origin: chemistry, pharmacology, and use in raditional and modern medicine. 1st ed. Berlin: Springer- Verlag; 1992. p. 711.
  16. Oliynyk S, Oh S. Actoprotective effect of ginseng: improving mental and physical performance. J Ginseng Res 2013;37:144-66. https://doi.org/10.5142/jgr.2013.37.144
  17. Kang KS, Kim HY, Pyo JS, Yokozawa T. Increase in the free radical scavenging activity of ginseng by heat-processing. Biol Pharm Bull 2006;29:750-4. https://doi.org/10.1248/bpb.29.750
  18. Konoshima T, Takasaki M, Tokuda H, Nishino H, Duc NM, Kasai R, Yamasaki K. Anti-tumor-promoting activity of majonoside-R2 from Vietnamese ginseng, Panax vietnamensis Ha et Grushv. (I). Biol Pharm Bull 1998;21:834-8. https://doi.org/10.1248/bpb.21.834
  19. Park JD. Recent studies on the chemical constituents of Korean ginseng (Panax ginseng C. A. Meyer). Korean J Ginseng Sci 1996;20:389-415.
  20. Gong X, Sucher NJ. Stroke therapy in traditional Chinese medicine (TCM): prospects for drug discovery and development. Trends Pharmacol Sci 1999;20:191-6. https://doi.org/10.1016/S0165-6147(98)01276-0
  21. He B, Chen P, Yang J, Yun Y, Zhang X, Yang R, Shen Z. Neuroprotective effect of 20(R)-ginsenoside Rg(3) against transient focal cerebral ischemia in rats. Neurosci Lett 2012;526:106-11. https://doi.org/10.1016/j.neulet.2012.08.022
  22. Cheon SY, Cho KJ, Lee JE, Kim HW, Lee SK, Kim HJ, Kim GW. Cerebroprotective effects of red ginseng extract pretreatment against ischemia-induced oxidative stress and apoptosis. Int J Neurosci 2013;123:269-77. https://doi.org/10.3109/00207454.2012.758120
  23. Tachikawa E, Kudo K, Kashimoto T, Takahashi E. Effects of ginseng saponins on receptor stimulation-responses. Nihon Yakurigaku Zasshi 1997;110:126-31 [In Japanese].
  24. Kim HS, Hwang SL, Nah SY, Oh S. Changes of [$^{3}H$]MK-801, [3H]muscimol and [$^{3}H$]flunitrazepam binding in rat brain by the prolonged ventricular infusion of ginsenoside Rc and Rg1. Pharmacol Res 2001;43:473-9. https://doi.org/10.1006/phrs.2001.0809
  25. Park HJ, Rhee MH, Park KM, Nam KY, Park KH. Effect of non-saponin fraction from Panax ginseng on cGMP and thromboxane A2 in human platelet aggregation. J Ethnopharmacol 1995;49:157-62. https://doi.org/10.1016/0378-8741(95)01317-2
  26. Nitta H, Matsumoto K, Shimizu M, Ni XH, Watanabe H. Panax ginseng extract improves the scopolamine-induced disruption of 8-arm radial maze performance in rats. Biol Pharm Bull 1995;18:1439-42. https://doi.org/10.1248/bpb.18.1439
  27. Nitta H, Matsumoto K, Shimizu M, Ni XH, Watanabe H. Panax ginseng extract improves the performance of aged Fischer 344 rats in radial maze task but not in operant brightness discrimination task. Biol Pharm Bull 1995;18:1286-8. https://doi.org/10.1248/bpb.18.1286
  28. Zhao R, McDaniel WF. Ginseng improves strategic learning by normal and brain-damaged rats. Neuroreport 1998;9:1619-24. https://doi.org/10.1097/00001756-199805110-00066
  29. Yamaguchi Y, Haruta K, Kobayashi H. Effects of ginsenosides on impaired performance induced in the rat by scopolamine in a radial-arm maze. Psychoneuroendocrinology 1995;20:645-53. https://doi.org/10.1016/0306-4530(95)00008-C
  30. Mook-Jung I, Hong HS, Boo JH, Lee KH, Yun SH, Cheong MY, Joo I, Huh K, Jung MW. Ginsenoside Rb1 and Rg1 improve spatial learning and increase hippocampal synaptophysin level in mice. J Neurosci Res 2001;63:509-15. https://doi.org/10.1002/jnr.1045
  31. Kim DH, Jung JS, Suh HW, Huh SO, Min SK, Son BK, Park JH, Kim ND, Kim YH, Song DK. Inhibition of stress-induced plasma corticosterone levels by ginsenosides in mice: involvement of nitric oxide. Neuroreport 1998;9:2261-4. https://doi.org/10.1097/00001756-199807130-00021
  32. Dou DQ, Zhang YW, Zhang L, Chen YJ, Yao XS. The inhibitory effects of ginsenosides on protein tyrosine kinase activated by hypoxia/reoxygenation in cultured human umbilical vein endothelial cells. Planta Med 2001;67:19-23. https://doi.org/10.1055/s-2001-10629
  33. Lee ST, Chu K, Sim JY, Heo JH, Kim M. Panax ginseng enhances cognitive performance in Alzheimer disease. Alzheimer Dis Assoc Disord 2008;22:222-6. https://doi.org/10.1097/WAD.0b013e31816c92e6
  34. 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;31:207-23. https://doi.org/10.3233/JAD-2012-120439
  35. Wu CF, Bi XL, Yang JY, Zhan JY, Dong YX, Wang JH, Wang JM, Zhang R, Li X. Differential effects of ginsenosides on NO and TNF-alpha production by LPSactivated N9 microglia. Int Immunopharmacol 2007;7:313-20. https://doi.org/10.1016/j.intimp.2006.04.021
  36. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci 2000;908:244-54.
  37. Godbout JP, Chen J, Abraham J, Richwine AF, Berg BM, Kelley KW, Johnson RW. Exaggerated neuroinflammation and sickness behavior in aged mice following activation of the peripheral innate immune system. FASEB J 2005;19:1329-31. https://doi.org/10.1096/fj.05-3776fje
  38. Maher FO, Nolan Y, Lynch MA. Downregulation of IL-4-induced signalling in hippocampus contributes to deficits in LTP in the aged rat. Neurobiol Aging 2005;26:717-28. https://doi.org/10.1016/j.neurobiolaging.2004.07.002
  39. Chen J, Buchanan JB, Sparkman NL, Godbout JP, Freund GG, Johnson RW. Neuroinflammation and disruption in working memory in aged mice after acute stimulation of the peripheral innate immune system. Brain Behav Immun 2008;22:301-11. https://doi.org/10.1016/j.bbi.2007.08.014
  40. Mrak RE, Griffin WS. Glia and their cytokines in progression of neurodegeneration. Neurobiol Aging 2005;26:349-54. https://doi.org/10.1016/j.neurobiolaging.2004.05.010
  41. Hirsch EC, Hunot S. Neuroinflammation in Parkinson's disease: a target for neuroprotection? Lancet Neurol 2009;8:382-97. https://doi.org/10.1016/S1474-4422(09)70062-6
  42. Rikans LE, Hornbrook KR. Lipid peroxidation, antioxidant protection and aging. Biochim Biophys Acta 1997;1362:116-27. https://doi.org/10.1016/S0925-4439(97)00067-7
  43. Lai LS, Chou ST, Chao WW. Studies on the antioxidative activities of Hsian-tsao (Mesona procumbens Hemsl) leaf gum. J Agric Food Chem 2001;49:963-8. https://doi.org/10.1021/jf001146k
  44. Liu R, Choi J. Age-associated decline in gamma-glutamylcysteine synthetase gene expression in rats. Free Radic Biol Med 2000;28:566-74. https://doi.org/10.1016/S0891-5849(99)00269-5
  45. Wu JL, Wu QP, Yang XF, Wei MK, Zhang JM, Huang Q, Zhou XY. l-Malate reverses oxidative stress and antioxidative defenses in liver and heart of aged rats. Physiol Res 2008;57:261-8.
  46. Aydin AF, Kucukgergin C, Ozdemirler-Erata G, Kocak-Toker N, Uysal M. The effect of carnosine treatment on prooxidant-antioxidant balance in liver, heart and brain tissues of male aged rats. Biogerontology 2010;11:103-9. https://doi.org/10.1007/s10522-009-9232-4
  47. Zhu D, Wu L, Li CR, Wang XW, Ma YJ, Zhong ZY, Zhao HB, Cui J, Xun SF, Huang XL, et al. Ginsenoside Rg1 protects rat cardiomyocyte from hypoxia/ reoxygenation oxidative injury via antioxidant and intracellular calcium homeostasis. J Cell Biochem 2009;108:117-24. https://doi.org/10.1002/jcb.22233
  48. Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun 1997;236:313-22. https://doi.org/10.1006/bbrc.1997.6943
  49. Kwak MK, Itoh K, Yamamoto M, Sutter TR, Kensler TW. Role of transcription factor Nrf2 in the induction of hepatic phase 2 and antioxidative enzymes in vivo by the cancer chemoprotective agent, 3H-1, 2-dimethiole-3-thione. Mol Med 2001;7:135-45.
  50. Lee JM, Calkins MJ, Chan K, Kan YW, Johnson JA. Identification of the NF-E2- related factor-2-dependent genes conferring protection against oxidative stress in primary cortical astrocytes using oligonucleotide microarray analysis. J Biol Chem 2003;278:12029-38. https://doi.org/10.1074/jbc.M211558200
  51. Shih AY, Johnson DA, Wong G, Kraft AD, Jiang L, Erb H, Johnson JA, Murphy TH. Coordinate regulation of glutathione biosynthesis and release by Nrf2- expressing glia potently protects neurons from oxidative stress. J Neurosci 2003;23:3394-406. https://doi.org/10.1523/JNEUROSCI.23-08-03394.2003
  52. Lee JM, Johnson JA. An important role of Nrf2-ARE pathway in the cellular defense mechanism. J Biochem Mol Biol 2004;37:139-43. https://doi.org/10.5483/BMBRep.2004.37.2.139
  53. Vargas MR, Johnson DA, Sirkis DW, Messing A, Johnson JA. Nrf2 activation in astrocytes protects against neurodegeneration in mouse models of familial amyotrophic lateral sclerosis. J Neurosci 2008;28:13574-81. https://doi.org/10.1523/JNEUROSCI.4099-08.2008

Cited by

  1. Role of the Red Ginseng in Defense against the Environmental Heat Stress in Sprague Dawley Rats vol.20, pp.11, 2015, https://doi.org/10.3390/molecules201119692
  2. Effects of Grape Skin Extract on Age-Related Mitochondrial Dysfunction, Memory and Life Span in C57BL/6J Mice vol.18, pp.3, 2016, https://doi.org/10.1007/s12017-016-8428-4
  3. Bioconversion, health benefits, and application of ginseng and red ginseng in dairy products vol.26, pp.5, 2015, https://doi.org/10.1007/s10068-017-0159-2
  4. Ginseng: An Nonnegligible Natural Remedy for Healthy Aging vol.8, pp.6, 2017, https://doi.org/10.14336/ad.2017.0707
  5. The immune-enhancing activity of Cervus nippon mantchuricus extract (NGE) in RAW264.7 macrophage cells and immunosuppressed mice vol.99, pp.1, 2015, https://doi.org/10.1016/j.foodres.2017.06.053
  6. Korean Red Ginseng Enhances Neurogenesis in the Subventricular Zone of 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Treated Mice vol.10, pp.None, 2015, https://doi.org/10.3389/fnagi.2018.00355
  7. Protective effects of cultured and fermented ginseng extracts against scopolamine-induced memory loss in a mouse model vol.34, pp.1, 2015, https://doi.org/10.5625/lar.2018.34.1.37
  8. Anti‐ageing effect of red ginseng revealed by urinary metabonomics using RRLC‐Q‐TOF‐MS vol.29, pp.4, 2018, https://doi.org/10.1002/pca.2758
  9. Brain Ageing, Cognition and Diet: A Review of the Emerging Roles of Food-Based Nootropics in Mitigating Age-Related Memory Decline vol.12, pp.1, 2015, https://doi.org/10.2174/1874609812666190311160754
  10. Screening and determination for potential acetylcholinesterase inhibitory constituents from ginseng stem–leaf saponins using ultrafiltration (UF)‐LC‐ESI‐MS2 vol.30, pp.1, 2015, https://doi.org/10.1002/pca.2787
  11. Red Ginseng Attenuates Aβ-Induced Mitochondrial Dysfunction and Aβ-mediated Pathology in an Animal Model of Alzheimer’s Disease vol.20, pp.12, 2019, https://doi.org/10.3390/ijms20123030
  12. Korean red ginseng promotes hippocampal neurogenesis in mice vol.15, pp.5, 2015, https://doi.org/10.4103/1673-5374.268905
  13. Red Ginseng Inhibits Tau Aggregation and Promotes Tau Dissociation In Vitro vol.2020, pp.None, 2020, https://doi.org/10.1155/2020/7829842
  14. Kamikihito Enhances Cognitive Functions and Reward-Related Behaviors of Aged C57BL/6J Mice in an Automated Behavioral Assay System vol.11, pp.None, 2015, https://doi.org/10.3389/fphar.2020.01037
  15. Anti‐ageing effects of red ginseng on female Drosophila melanogaster vol.24, pp.6, 2015, https://doi.org/10.1111/jcmm.15029
  16. Protective effect of Polygonatum sibiricum Polysaccharide on D-galactose-induced aging rats model vol.10, pp.None, 2015, https://doi.org/10.1038/s41598-020-59055-7
  17. 국내 한의학 학술지에 발표된 항염증 한약재 및 한약처방 연구동향 - 2015년 이후 발표된 실험논문을 중심으로 - vol.36, pp.1, 2015, https://doi.org/10.6116/kjh.2021.36.1.19.
  18. Panax Ginseng alleviates thioacetamide-induced liver injury in ovariectomized rats: Crosstalk between inflammation and oxidative stress vol.16, pp.11, 2015, https://doi.org/10.1371/journal.pone.0260507
  19. The effects of agomelatine on endoplasmic reticulum stress related to mitochondrial dysfunction in hippocampus of aging rat model vol.351, pp.None, 2015, https://doi.org/10.1016/j.cbi.2021.109703