DOI QR코드

DOI QR Code

Multitarget effects of Korean Red Ginseng in animal model of Parkinson's disease: antiapoptosis, antioxidant, antiinflammation, and maintenance of blood-brain barrier integrity

  • Choi, Jong Hee (Department of Science in Korean Medicine, Graduate School, Kyung Hee University) ;
  • Jang, Minhee (Brain Korea 21 Plus Program and Institute of Korean Medicine, Graduate School, Kyung Hee University) ;
  • Nah, Seung-Yeol (Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University) ;
  • Oh, Seikwan (Department of Neuroscience, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University) ;
  • Cho, Ik-Hyun (Department of Science in Korean Medicine, Graduate School, Kyung Hee University)
  • Received : 2017.09.01
  • Accepted : 2018.01.17
  • Published : 2018.07.15

Abstract

Background: Ginsenosides are the main ingredients of Korean Red Ginseng. They have extensively been studied for their beneficial value in neurodegenerative diseases such as Parkinson's disease (PD). However, the multitarget effects of Korean Red Ginseng extract (KRGE) with various components are unclear. Methods: We investigated the multitarget activities of KRGE on neurological dysfunction and neurotoxicity in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. KRGE (37.5 mg/ kg/day, 75 mg/kg/day, or 150 mg/kg/day, per os (p.o.)) was given daily before or after MPTP intoxication. Results: Pretreatment with 150 mg/kg/day KRGE produced the greatest positive effect on motor dysfunction as assessed using rotarod, pole, and nesting tests, and on the survival rate. KRGE displayed a wide therapeutic time window. These effects were related to reductions in the loss of tyrosine hydroxylase-immunoreactive dopaminergic neurons, apoptosis, microglial activation, and activation of inflammatory factors in the substantia nigra pars compacta and/or striatum after MPTP intoxication. In addition, pretreatment with KRGE activated the nuclear factor erythroid 2-related factor 2 pathways and inhibited phosphorylation of the mitogen-activated protein kinases and nuclear factor-kappa B signaling pathways, as well as blocked the alteration of blood-brain barrier integrity. Conclusion: These results suggest that KRGE may effectively reduce MPTP-induced neurotoxicity with a wide therapeutic time window through multitarget effects including antiapoptosis, antiinflammation, antioxidant, and maintenance of blood-brain barrier integrity. KRGE has potential as a multitarget drug or functional food for safe preventive and therapeutic strategies for PD.

Keywords

References

  1. Kalia LV, Lang AE. Parkinson's disease. Lancet 2015;386:896-912. https://doi.org/10.1016/S0140-6736(14)61393-3
  2. Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE, Lang AE. Parkinson disease. Nat Rev Dis Primers 2017;3:17013. https://doi.org/10.1038/nrdp.2017.13
  3. Shulman JM, De Jager PL, Feany MB. Parkinson's disease: genetics and pathogenesis. Annu Rev Pathol 2011;6:193-222. https://doi.org/10.1146/annurev-pathol-011110-130242
  4. Kalinderi K, Bostantjopoulou S, Fidani L. The genetic background of Parkinson's disease: current progress and future prospects. Acta Neurol Scand 2016;134:314-26.
  5. Gubellini P, Kachidian P. Animal models of Parkinson's disease: an updated overview. Rev Neurol (Paris) 2015;171:750-61. https://doi.org/10.1016/j.neurol.2015.07.011
  6. Marsden CD. Problems with long-term levodopa therapy for Parkinson's disease. Clin Neuropharmacol 1994;17(Suppl. 2):S32-44. https://doi.org/10.1097/00002826-199402000-00003
  7. Calabresi P, Di Filippo M. Multitarget disease-modifying therapy in Parkinson's disease? Lancet Neurol 2015;14:975-6. https://doi.org/10.1016/S1474-4422(15)00227-6
  8. Dias KS, Viegas Jr C. Multi-target directed drugs: a modern approach for design of new drugs for the treatment of Alzheimer's disease. Curr Neuropharmacol 2014;12:239-55. https://doi.org/10.2174/1570159X1203140511153200
  9. Bajda M, Guzior N, Ignasik M, Malawska B. Multi-target-directed ligands in Alzheimer's disease treatment. Curr Med Chem 2011;18:4949-75. https://doi.org/10.2174/092986711797535245
  10. Zheng H, Fridkin M, Youdim M. From single target to multitarget/network therapeutics in Alzheimer's therapy. Pharmaceuticals (Basel) 2014;7:113-35. https://doi.org/10.3390/ph7020113
  11. Li XZ, Zhang SN, Liu SM, Lu F. Recent advances in herbal medicines treating Parkinson's disease. Fitoterapia 2013;84:273-85. https://doi.org/10.1016/j.fitote.2012.12.009
  12. Athauda D, Foltynie T. The ongoing pursuit of neuroprotective therapies in Parkinson disease. Nat Rev Neurol 2015;11:25-40. https://doi.org/10.1038/nrneurol.2014.226
  13. Newman DJ, Cragg GM. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J Nat Prod 2012;75:311-35. https://doi.org/10.1021/np200906s
  14. Cho IH. Effects of Panax ginseng in neurodegenerative diseases. J Ginseng Res 2012;36:342-53. https://doi.org/10.5142/jgr.2012.36.4.342
  15. Lee YM, Yoon H, Park HM, Song BC, Yeum KJ. Implications of red Panax ginseng in oxidative stress associated chronic diseases. J Ginseng Res 2017;41:113-9. https://doi.org/10.1016/j.jgr.2016.03.003
  16. Radad K, Gille G, Moldzio R, Saito H, Rausch WD. Ginsenosides Rb1 and Rg1 effects on mesencephalic dopaminergic cells stressed with glutamate. Brain Res 2004;1021:41-53. https://doi.org/10.1016/j.brainres.2004.06.030
  17. Lin WM, Zhang YM, Moldzio R, Rausch WD. Ginsenoside Rd attenuates neuroinflammation of dopaminergic cells in culture. J Neural Transm Suppl 2007:105-12.
  18. 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
  19. Chen XC, Chen Y, Zhu YG, Fang F, Chen LM. Protective effect of ginsenoside Rg1 against MPTP-induced apoptosis in mouse substantia nigra neurons. Acta Pharmacol Sin 2002;23:829-34.
  20. Hu JF, Song XY, Chu SF, Chen J, Ji HJ, Chen XY, Yuan YH, Han N, Zhang JT, Chen NH. Inhibitory effect of ginsenoside Rg1 on lipopolysaccharide-induced microglial activation in mice. Brain Res 2011;1374:8-14. https://doi.org/10.1016/j.brainres.2010.11.069
  21. Radad K, Moldzio R, Rausch WD. Ginsenosides and their CNS targets. CNS Neurosci Ther 2011;17:761-8. https://doi.org/10.1111/j.1755-5949.2010.00208.x
  22. 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.
  23. Jun YL, Bae CH, Kim D, Koo S, Kim S. Korean red ginseng protects dopaminergic neurons by suppressing the cleavage of p35 to p25 in a Parkinson's disease mouse model. J Ginseng Res 2015;39:148-54. https://doi.org/10.1016/j.jgr.2014.10.003
  24. Kim D, Jeon H, Ryu S, Koo S, Ha KT, Kim S. Proteomic analysis of the effect of Korean red ginseng in the striatum of a Parkinson's disease mouse model. PLoS One 2016;11:e0164906. https://doi.org/10.1371/journal.pone.0164906
  25. Landis SC, Amara SG, Asadullah K, Austin CP, Blumenstein R, Bradley EW, Crystal RG, Darnell RB, Ferrante RJ, Fillit H, et al. A call for transparent reporting to optimize the predictive value of preclinical research. Nature 2012;490:187-91. https://doi.org/10.1038/nature11556
  26. Jang M, Lee MJ, Kim CS, Cho IH. Korean red ginseng extract attenuates 3-nitropropionic acid-induced Huntington's-like symptoms. Evid Based Complement Alternat Med 2013;2013:237207.
  27. Lee MJ, Jang M, Choi J, Chang BS, Kim DY, Kim SH, Kwak YS, Oh S, Lee JH, Chang BJ, et al. Korean red ginseng and ginsenoside-Rb1/-Rg1 alleviate experimental autoimmune encephalomyelitis by suppressing Th1 and Th17 cells and upregulating regulatory T cells. Mol Neurobiol 2016;53:1977-2002. https://doi.org/10.1007/s12035-015-9131-4
  28. Jackson-Lewis V, Przedborski S. Protocol for the MPTP mouse model of Parkinson's disease. Nat Protoc 2007;2:141-51. https://doi.org/10.1038/nprot.2006.342
  29. Deacon RM. Assessing nest building in mice. Nat Protoc 2006;1:1117-9. https://doi.org/10.1038/nprot.2006.170
  30. Franklin KBJ, Paxinos G. The mouse brain in stereotaxic coordinates. San Iego: Elsevier Academic Press; 2008.
  31. Choi JH, Jang M, Kim EJ, Kim H, Ye SK, Cho IH. Oriental medicine Woohwangchungsimwon attenuates Kainic acid-induced seizures and neuronal cell death in the Hippocampus. Rejuvenation Res 2016.
  32. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using realtime quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 2001;25:402-8. https://doi.org/10.1006/meth.2001.1262
  33. Hirsch EC, Hunot S, Faucheux B, Agid Y, Mizuno Y, Mochizuki H, Tatton WG, Tatton N, Olanow WC, et al. Dopaminergic neurons degenerate by apoptosis in Parkinson's disease. Mov Disord 1999;14:383-5. https://doi.org/10.1002/1531-8257(199903)14:2<383::AID-MDS1037>3.0.CO;2-F
  34. Lobsiger CS, Cleveland DW. Glial cells as intrinsic components of non-cellautonomous neurodegenerative disease. Nat Neurosci 2007;10:1355-60. https://doi.org/10.1038/nn1988
  35. Du L, Zhang Y, Chen Y, Zhu J, Yang Y, Zhang HL. Role of microglia in neurological disorders and their potentials as a therapeutic target. Mol Neurobiol 2016.
  36. Jang M, Cho IH. Sulforaphane ameliorates 3-nitropropionic acid-induced striatal toxicity by activating the Keap1-nrf2-are pathway and inhibiting the MAPKs and NF-kappaB pathways. Mol Neurobiol 2016;53:2619-35. https://doi.org/10.1007/s12035-015-9230-2
  37. Willis CL. Glia-induced reversible disruption of blood-brain barrier integrity and neuropathological response of the neurovascular unit. Toxicol Pathol 2010;39:172-85.
  38. 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(2):106-11. https://doi.org/10.1016/j.neulet.2012.08.022
  39. Liu Q, Kou JP, Yu BY. Ginsenoside Rg1 protects against hydrogen peroxideinduced cell death in PC12 cells via inhibiting NF-kappaB activation. Neurochem Int 2011;58:119-25. https://doi.org/10.1016/j.neuint.2010.11.004
  40. Blesa J, Trigo-Damas I, Quiroga-Varela A, Jackson-Lewis VR. Oxidative stress and Parkinson's disease. Front Neuroanat 2015;9:91.
  41. Guneysel O, Onultan O, Onur O. Parkinson's disease and the frequent reasons for emergency admission. Neuropsychiatr Dis Treat 2008;4:711-4.
  42. Cohen GM. Caspases: the executioners of apoptosis. Biochem J 1997;326(Pt 1):1-16. https://doi.org/10.1042/bj3260001
  43. Li XY, Liang J, Tang YB, Zhou JG, Guan YY. Ginsenoside Rd prevents glutamateinduced apoptosis in rat cortical neurons. Clin Exp Pharmacol Physiol 2010;37:199-204. https://doi.org/10.1111/j.1440-1681.2009.05286.x
  44. Hu G, Wu Z, Yang F, Zhao H, Liu X, Deng Y, Shi M, Zhao G. Ginsenoside Rd blocks AIF mitochondrio-nuclear translocation and NF-kappaB nuclear accumulation by inhibiting poly(ADP-ribose) polymerase-1 after focal cerebral ischemia in rats. Neurol Sci 2013;34:2101-6. https://doi.org/10.1007/s10072-013-1344-6
  45. Ye R, Zhang X, Kong X, Han J, Yang Q, Zhang Y, Chen Y, Li P, Liu J, Shi M, et al. Ginsenoside Rd attenuates mitochondrial dysfunction and sequential apoptosis after transient focal ischemia. Neuroscience 2011;178:169-80. https://doi.org/10.1016/j.neuroscience.2011.01.007
  46. Nabavi SF, Sureda A, Habtemariam S, Nabavi SM. Ginsenoside Rd and ischemic stroke; a short review of literatures. J Ginseng Res 2015;39:299-303. https://doi.org/10.1016/j.jgr.2015.02.002
  47. Im GJ, Chang JW, Choi J, Chae SW, Ko EJ, Jung HH. Protective effect of Korean red ginseng extract on cisplatin ototoxicity in HEI-OC1 auditory cells. Phytother Res 2010;24:614-21.
  48. Kim EH, Jang MH, Shin MC, Shin MS, Kim CJ. Protective effect of aqueous extract of Ginseng radix against 1-methyl-4-phenylpyridinium-induced apoptosis in PC12 cells. Biol Pharm Bull 2003;26:1668-73. https://doi.org/10.1248/bpb.26.1668
  49. Du X, Xu H, Jiang H, Xie J. Akt/Nrf2 activated upregulation of heme oxygenase-1 involves in the role of Rg1 against ferrous iron-induced neurotoxicity in SKN-SH cells. Neurotox Res 2013;24:71-9. https://doi.org/10.1007/s12640-012-9362-3
  50. Lee YY, Park JS, Lee EJ, Lee SY, Kim DH, Kang JL, Kim HS. Anti-inflammatory mechanism of ginseng saponin metabolite Rh3 in lipopolysaccharide-stimulated microglia: critical role of 5'-adenosine monophosphate-activated protein kinase signaling pathway. J Agric Food Chem 2015;63:3472-80. https://doi.org/10.1021/jf506110y
  51. Kortekaas R, Leenders KL, van Oostrom JC, Vaalburg W, Bart J, Willemsen AT, Hendrikse NH. Blood-brain barrier dysfunction in Parkinsonian midbrain in vivo. Ann Neurol 2005;57(2):176-9. https://doi.org/10.1002/ana.20369
  52. Guo Q, Li P, Wang Z, Cheng Y, Wu H, Yang B, Du S, Lu Y. Brain distribution pharmacokinetics and integrated pharmacokinetics of Panax notoginsenoside R1, ginsenosides Rg1, Rb1, Re and Rd in rats after intranasal administration of Panax notoginseng saponins assessed by UPLC/MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2014;969:264-71. https://doi.org/10.1016/j.jchromb.2014.08.034
  53. Li M, Guan YM, Liu N, Shao C, Liu ZB, Chen JB, Wang Q, Pan X, Sun H, Zhang Y. Brain concentration of ginsenosides and pharmacokinetics after oral administrationof mountain-cultivated ginseng. J Chin Chem Soc 2017;64(4):395-403. https://doi.org/10.1002/jccs.201600783

Cited by

  1. Adeno-Associated Viral Vector Serotype DJ-Mediated Overexpression of N171-82Q-Mutant Huntingtin in the Striatum of Juvenile Mice Is a New Model for Huntington’s Disease vol.12, pp.None, 2018, https://doi.org/10.3389/fncel.2018.00157
  2. 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.None, 2018, https://doi.org/10.3389/fphar.2018.00515
  3. 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, 2018, https://doi.org/10.3389/fphar.2018.01444
  4. Antiepileptic and anti-neuroinflammatory effects of red ginseng in an intrahippocampal kainic acid model of temporal lobe epilepsy demonstrated by electroencephalography vol.35, pp.2, 2018, https://doi.org/10.12701/yujm.2018.35.2.192
  5. Red Ginseng Reduces Inflammatory Response via Suppression MAPK/P38 Signaling and p65 Nuclear Proteins Translocation in Rats and Raw 264.7 Macrophage vol.47, pp.7, 2018, https://doi.org/10.1142/s0192415x19500812
  6. Red Ginseng Inhibits Tau Aggregation and Promotes Tau Dissociation In Vitro vol.2020, pp.None, 2020, https://doi.org/10.1155/2020/7829842
  7. Natural Products and Their Bioactive Compounds: Neuroprotective Potentials against Neurodegenerative Diseases vol.2020, pp.None, 2020, https://doi.org/10.1155/2020/6565396
  8. Analysis and Identification of Active Compounds from Salviae miltiorrhizae Radix Toxic to HCT-116 Human Colon Cancer Cells vol.10, pp.4, 2018, https://doi.org/10.3390/app10041304
  9. Ferulic Acid Ameliorates MPP+/MPTP-Induced Oxidative Stress via ERK1/2-Dependent Nrf2 Activation: Translational Implications for Parkinson Disease Treatment vol.57, pp.7, 2018, https://doi.org/10.1007/s12035-020-01934-1
  10. Protective Effects of Active Compounds from Salviae miltiorrhizae Radix against Glutamate-Induced HT-22 Hippocampal Neuronal Cell Death vol.8, pp.8, 2018, https://doi.org/10.3390/pr8080914
  11. Korean Red Ginseng Regulates Intestinal Tight Junction and Inflammation in the Colon of a Parkinson's Disease Mouse Model vol.23, pp.12, 2018, https://doi.org/10.1089/jmf.2019.4640
  12. 파킨슨병 in vivo 모델에서 한약재 및 기능성 식품의 항산화 효과에 대한 고찰 vol.41, pp.6, 2018, https://doi.org/10.22246/jikm.2020.41.6.993
  13. Korean red ginseng suppresses 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced inflammation in the substantia nigra and colon vol.94, pp.None, 2018, https://doi.org/10.1016/j.bbi.2021.02.028
  14. Ovarian cysts disappear after 14-day oral regimen of Korean red ginseng extract in letrozole-induced polycystic ovarian syndrome vol.64, pp.3, 2018, https://doi.org/10.5468/ogs.20094