DOI QR코드

DOI QR Code

Ginsenoside Rb2 suppresses the glutamate-mediated oxidative stress and neuronal cell death in HT22 cells

  • Kim, Dong Hoi (Convergence Research Center for Dementia, KIST) ;
  • Kim, Dae Won (Department of Biochemistry, College of Dentistry, Gangneung Wonju National University) ;
  • Jung, Bo Hyun (Department of Oral Anatomy, College of Dentistry, Gangneung Wonju National University) ;
  • Lee, Jong Hun (Department of Oral Anatomy, College of Dentistry, Gangneung Wonju National University) ;
  • Lee, Heesu (Department of Oral Anatomy, College of Dentistry, Gangneung Wonju National University) ;
  • Hwang, Gwi Seo (College of Korean Medicine, Gacheun University) ;
  • Kang, Ki Sung (College of Korean Medicine, Gacheun University) ;
  • Lee, Jae Wook (Convergence Research Center for Dementia, KIST)
  • Received : 2018.07.09
  • Accepted : 2018.12.07
  • Published : 2019.04.15

Abstract

Background: The objective of our study was to analyze the neuroprotective effects of ginsenoside derivatives Rb1, Rb2, Rc, Rd, Rg1, and Rg3 against glutamate-mediated neurotoxicity in HT22 hippocampal mouse neuron cells. Methods: The neuroprotective effect of ginsenosides were evaluated by measuring cell viability. Protein expressions of mitogen-activated protein kinase (MAPK), Bcl2, Bax, and apoptosis-inducing factor (AIF) were determined by Western blot analysis. The occurrence of apoptotic and death cells was determined by flow cytometry. Cellular level of $Ca^{2+}$ and reactive oxygen species (ROS) levels were evaluated by image analysis using the fluorescent probes Fluor-3 and 2',7'-dichlorodihydrofluorescein diacetate, respectively. In vivo efficacy of neuroprotection was evaluated using the Mongolian gerbil of ischemic brain injury model. Result: Reduction of cell viability by glutamate (5 mM) was significantly suppressed by treatment with ginsenoside Rb2. Phosphorylation of MAPKs, Bax, and nuclear AIF was gradually increased by treatment with 5 mM of glutamate and decreased by co-treatment with Rb2. The occurrence of apoptotic cells was decreased by treatment with Rb2 ($25.7{\mu}M$). Cellular $Ca^{2+}$ and ROS levels were decreased in the presence of Rb2, and in vivo data indicated that Rb2 treatment (10 mg/kg) significantly diminished the number of degenerated neurons. Conclusion: Our results suggest that Rb2 possesses neuroprotective properties that suppress glutamate-induced neurotoxicity. The molecular mechanism of Rb2 is by suppressing the MAPKs activity and AIF translocation.

Keywords

References

  1. Riedel G, Platt P, Micheau J. Glutamate receptor function in learning and memory. Behav Brain Res 2003;140:1-47. https://doi.org/10.1016/S0166-4328(02)00272-3
  2. Reynolds IJ, Hastings TG. Glutamate induces the production of reactive oxygen species in cultured forebrain neurons following NMDA receptor activation. J Neurosci 1995;15:3318-27. https://doi.org/10.1523/JNEUROSCI.15-05-03318.1995
  3. Choi DW. Glutamate neurotoxicity and diseases of the nervous system. Neuron 1988;1:623-34. https://doi.org/10.1016/0896-6273(88)90162-6
  4. Dong XX, Wang Y, Qin ZH. Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol Sin 2009;30:379-87. https://doi.org/10.1038/aps.2009.24
  5. Coyle TJ, Puttfarcken P. Oxidative stress, glutamate, and neurodegerative disorders. Science 1993;262:689-95. https://doi.org/10.1126/science.7901908
  6. Niikura T, Tajima H, Kita Y. Curr Neuropharmacol 2006;4:139-47. https://doi.org/10.2174/157015906776359577
  7. Cookson MR. Mol Neurodegener 2009;4:9. https://doi.org/10.1186/1750-1326-4-9
  8. Cowan CM, Raymond LA. Curr Top Dev Biol 2006;75:25-71. https://doi.org/10.1016/S0070-2153(06)75002-5
  9. Murphy TH, Miyamoto M, Sastre A, Schnaar RL, Coyle JT. Glutamate toxicity in a neuronal cell line involves inhibition of cysteine transport leading to oxidative stress. Neuron 1989;2:1547-58. https://doi.org/10.1016/0896-6273(89)90043-3
  10. Choi DW. Glutamate neurotoxicity and diseases of the nervous system. Neuron 1988;1:623-34. https://doi.org/10.1016/0896-6273(88)90162-6
  11. Tan S, Schubert D, Maher P. Oxytosis: a novel form of programmed cell death. Curr Top Med Chem 2001;1:497-506. https://doi.org/10.2174/1568026013394741
  12. Kang Y, Tiziani S, Park G, Kaul M, Paternostro G. Cellular protection using Flt3 and $PI3K{\alpha}$ inhibitors demonstrates multiple mechanisms of oxidative glutamate toxicity. Nat Comm 2014;5:1-12.
  13. Yang E-J, Song K-S. Polyozellin, a key constituent of the edible mushroom Polyozellus multiplex, attenuates glutamate-induced mouse hippocampal neuronal HT22 cell death. Food Funct 2015;6:3678-86. https://doi.org/10.1039/C5FO00636H
  14. Tobaben S, Grohm J, Seiler A, Conrad M, Plesnila N, Culmsee C. Bid-mediated mitochondrial damage is a key mechanism in glutamate-induced oxidative stress and AIF-dependent cell death in immortalized HT-22 hippocampal neurons. Cell Death Differ 2011;18:282-92. https://doi.org/10.1038/cdd.2010.92
  15. Culmsee C, Plesnila N. Targeting Bid to prevent programmed cell death in neurons. Biochem Soc Trans 2006;34:1334-40. https://doi.org/10.1042/BST0341334
  16. Xu X, Chua CC, Kong J, Kostrzewa RM, Kumaraguru U, Hamdy RC, Chua BH. Necrostatin-1 protects against glutamate-induced glutathione depletion and caspase-independent cell death in HT-22 cells. J Neurochem 2007;103:2004-14. https://doi.org/10.1111/j.1471-4159.2007.04884.x
  17. Landshamer S, Hoehn M, Barth N, Duvezin-Caubet S, Schwake G, Tobaben S, Kazhdan I, Becattini B, Zahler S, Vollmar A, et al. Bid-induced release of AIF from mitochondria causes immediate neuronal cell death. Cell Death Differ 2008;15:1553-63. https://doi.org/10.1038/cdd.2008.78
  18. Susin SA1, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999;397:441-6. https://doi.org/10.1038/17135
  19. Daugas E1, Susin SA, Zamzami N, Ferri KF, Irinopoulou T, Larochette N, Prevost MC, Leber B, Andrews D, Penninger J, et al. Mitochondrio-nuclear translocation of AIF in apoptosis and necrosis. FASEB J 2000;14:729-39. https://doi.org/10.1096/fasebj.14.5.729
  20. Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal 2012;24:981-90. https://doi.org/10.1016/j.cellsig.2012.01.008
  21. Choi JH, Choi AY, Yoon H, Choe W, Yoon K-S, Ha J, Yeo E-J, Kang I. Baicalein protects HT22 murine hippocampal neuronal cells against endoplasmic reticulum stress-induced apoptosis through inhibition of reactive oxygen species production and CHOP induction. Exp Mol Med 2010;42:811-22. https://doi.org/10.3858/emm.2010.42.12.084
  22. Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K, Cobb MH. Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev 2001;22:153-83. https://doi.org/10.1210/er.22.2.153
  23. Tibbles LA, Woodgett JR. The stress-activated protein kinase pathways. Cell Mol Lif Sci 1999;55:1230-54. https://doi.org/10.1007/s000180050369
  24. Park EH, Kim YJ, Yamabe N, Park SH, Kim HK, Jang HJ, Kim JH, Cheon GJ, Ham J, Kang KS. Stereospecific anticancer effects of ginsenoside Rg3 epimers isolated from heat-processed American ginseng on human gastric cancer cell. J Ginseng Res 2014;38:22-7. https://doi.org/10.1016/j.jgr.2013.11.007
  25. Park JY, Choi P, Kim HK, Kang KS, Ham J. Increase in apoptotic effect of Panax ginseng by microwave processing in human prostate cancer cells: in vitro and in vivo studies. J Ginseng Res 2016;40:62-7. https://doi.org/10.1016/j.jgr.2015.04.007
  26. Jang HJ, Han IH, Kim YJ, Yamabe N, Lee D, Hwang GS, Oh M, Choi KC, Kim SN, Ham J, et al. Anticarcinogenic effects of products of heat-processed ginsenoside Re, a major constituent of ginseng berry, on human gastric cancer cells. J Agric Food Chem 2014;62:2830-6. https://doi.org/10.1021/jf5000776
  27. Dong X, Zheng L, Lu S, Yang Y. Neuroprotective effects of pretreatment of ginsenoside Rb1 on severe cerebral ischemia-induced injuries in aged mice: involvement of anti-oxidant signaling. Geriatr Gerontol Int 2017;17:338-45. https://doi.org/10.1111/ggi.12699
  28. Gao XQ, Yang CX, Chen GJ, Wang GY, Chen B, Tan SK, Liu J, Yuan QL. Ginsenoside Rb1 regulates the expressions of brain-derived neurotrophic factor and caspase-3 and induces neurogenesis in rats with experimental cerebral ischemia. J Ethnopharmacol 2010;132:393-9. https://doi.org/10.1016/j.jep.2010.07.033
  29. Wang L, Zhao H, Zhai ZZ, Qu LX. Protective effect and mechanism of ginsenoside Rg1 in cerebral ischaemia-reperfusion injury in mice. Biomed Pharmacother 2018;99:876-82. https://doi.org/10.1016/j.biopha.2018.01.136
  30. Zhang G, Xia F, Zhang Y, Zhang X, Cao Y, Wang L, Liu X, Zhao G, Shi M. Ginsenoside Rd is efficacious against acute ischemic stroke by suppressing microglial proteasome-mediated inflammation. Mol Neurobiol 2016;53: 2529-40. https://doi.org/10.1007/s12035-015-9261-8
  31. Kim JH, Cho SY, Lee JH, Jeong SM, Yoon IS, Lee BH, Lee JH, Pyo MK, Lee SM, Chung JM, et al. Neuroprotective effects of ginsenoside Rg3 against homocysteine-induced excitotoxicity in rat hippocampus. Brain Res 2007;1136:190-9. https://doi.org/10.1016/j.brainres.2006.12.047
  32. Tian J, Fu F, Geng M, Jiang Y, Yang J, Jiang W, Wang C, Liu K. Neuroprotective effect of 20(S)-ginsenoside Rg3 on cerebral ischemia in rats. Neurosci Lett 2005;374:92-7. https://doi.org/10.1016/j.neulet.2004.10.030
  33. Tan S, Wood M, Maher P. Oxidative stress induces a form of programmed cell death with characteristics of both apoptosis and necrosis in neuronal cells. J Neurochem 1998;71:95-105. https://doi.org/10.1046/j.1471-4159.1998.71010095.x
  34. Wen T-C, Yoshimura H, Matsuda S, Lim J-H, Sakanaka M. Ginseng root prevents learning disability and neuronal loss in gerbils with 5-minute forebrain ischemia. Acta Neuropathol 1996;91:15-22. https://doi.org/10.1007/s004010050387
  35. Lee JC, Park JH, Ahn JH, Kim IH, Cho JH, Choi JH, Yoo KY, Lee CH, Hwang IK, Cho JH, et al. New GABAergic neurogenesis in the hippocampal CA1 region of a gerbil mdel of long-term survival after transient cerebral ischemic injury. Brain Pathol 2016;26:581-92. https://doi.org/10.1111/bpa.12334
  36. Schmued LC, Hopkins KJ, Fluoro-Jade B. A high affinity fluorescent marker for the localization of neuronal degeneration. Brain Res 2000;874:123-30. https://doi.org/10.1016/S0006-8993(00)02513-0
  37. Park JH, Joo HS, Yoo KY, Shin BN, Kim IH, Lee CH, Choi JH, Byun K, Lee B, Lim SS, et al. Extract from Terminalia chebula seeds protect against experimental ischemic neuronal damage via maintaining SODs and BDNF levels. Neurochem Res 2011;36:2043-50. https://doi.org/10.1007/s11064-011-0528-9
  38. Park JH, Shin BN, Chen BH, Kim IH, Ahn JH, Cho JH, Tae HJ, Lee JC, Lee CH, Kim YM, et al. Neuroprotection and reduced gliosis by atomoxetine pretreatment in a gerbil model of transient cerebral ischemia. J Neurol Sci 2015;359:373-80. https://doi.org/10.1016/j.jns.2015.11.028
  39. Kim YC, Kim SR, Markelonis GJ, Oh TH. Ginsenosides Rb1 and Rg3 protect cultured rat cortical cells from glutamate-induced neurodegeneration. J Neurosci Res 1998;53:426-32. https://doi.org/10.1002/(SICI)1097-4547(19980815)53:4<426::AID-JNR4>3.0.CO;2-8
  40. Fukui M, Song JM, Choi JY, Choi HJ, Zhu BT. Mechanism of glutamate-induced neurotoxicity inTH22mouse hippocampal cells. Euro J of Pharm 2009;617:1-11. https://doi.org/10.1016/j.ejphar.2009.06.059
  41. Son Y, Cheong YK, Kim NH, Chung HT, Kang DG, Pae HO. Mitogen-activated protein kinases and reactive oxygen species; how can ROS activate MAPK pathways? J Signal Transduct 2011;2011:792639.
  42. Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signaling 2012;24:981-90. https://doi.org/10.1016/j.cellsig.2012.01.008
  43. Pettmann B1, Henderson CE. Neuronal cell death. Neuron 1998;20:633-47. https://doi.org/10.1016/S0896-6273(00)81004-1
  44. Bredesen DE. Neural apoptosis. Ann Neurol 1995;38:839-51. https://doi.org/10.1002/ana.410380604
  45. Zhang Y, Lu X, Bhavnani BR. Equine estrogens differentially inhibit DNA fragmentation induced by glutamate in neuronal cells by modulation of regulatory proteins involved in programmed cell death. BMC Neurosci 2003;4:32. https://doi.org/10.1186/1471-2202-4-32
  46. Yang E-J, Min JS, Ku H-Y, Choi H-S, Park M, Kim MK, Song K-S, Lee D-S. Isoliquirituugenin isolated from Glycyrrhiza uralensis protects neuronal cells against glutamate-induced mitochondrial dysfunction. Biochem Biophy Res Comm 2012;421:658-64. https://doi.org/10.1016/j.bbrc.2012.04.053
  47. Landshamer S, Hoehn M, Barth N, Duvezin-Caubet S, Schwake G, Tobaben S, Kazhdan I, Becattini B, Zahler S, Vollmar A, et al. Bid-induced release of AIF from mitochondria causes immediate neuronal cell death. Cell Death Diff 2008;15:1553-63. https://doi.org/10.1038/cdd.2008.78
  48. Tobaben S, Grohm J, Seiler A, Conrad M, Plesnila N, Culmsee C. Bid-mediated mitochondrial damage is a key mechanism in glutamate-induced oxidative stress and AIF-dependent cell death in immortalized HT-22 hippocampal neurons. Cell Death Diff 2011;18:282-92. https://doi.org/10.1038/cdd.2010.92
  49. Ong W-Y, Farooqui T, Koh H-L, Farooqui AA, Ling E-A. Protective effects of ginseng on neurological disorders. Front Aging Neurosci 2015;7:129.

Cited by

  1. Reduction of Mitophagy-Related Oxidative Stress and Preservation of Mitochondria Function Using Melatonin Therapy in an HT22 Hippocampal Neuronal Cell Model of Glutamate-Induced Excitotoxicity vol.10, 2019, https://doi.org/10.3389/fendo.2019.00550
  2. Partial-root Harvest of American Ginseng (Panax quinquefolius L.): A Non-Destructive Method for Harvesting Root Tissue for Ginsenoside Analysis vol.84, pp.2, 2019, https://doi.org/10.2179/0008-7475.84.2.310
  3. Neuroprotective Effects of Tetrahydrocurcumin against Glutamate-Induced Oxidative Stress in Hippocampal HT22 Cells vol.25, pp.1, 2020, https://doi.org/10.3390/molecules25010144
  4. Effects of Ginseng on Neurological Disorders vol.14, 2019, https://doi.org/10.3389/fncel.2020.00055
  5. Analysis and Identification of Active Compounds from Salviae miltiorrhizae Radix Toxic to HCT-116 Human Colon Cancer Cells vol.10, pp.4, 2019, https://doi.org/10.3390/app10041304
  6. Hormesis and Ginseng: Ginseng Mixtures and Individual Constituents Commonly Display Hormesis Dose Responses, Especially for Neuroprotective Effects vol.25, pp.11, 2020, https://doi.org/10.3390/molecules25112719
  7. Protective Effects of Active Compounds from Salviae miltiorrhizae Radix against Glutamate-Induced HT-22 Hippocampal Neuronal Cell Death vol.8, pp.8, 2019, https://doi.org/10.3390/pr8080914
  8. Mitochondrial connection to ginsenosides vol.43, pp.10, 2019, https://doi.org/10.1007/s12272-020-01279-2
  9. Antihypoxic Action of Panax Japonicus, Tribulus Terrestris and Dioscorea Deltoidea Cell Cultures: In Silico and Animal Studies vol.39, pp.11, 2019, https://doi.org/10.1002/minf.202000093
  10. 1-Methoxylespeflorin G11 Protects HT22 Cells from Glutamate-Induced Cell Death through Inhibition of ROS Production and Apoptosis vol.31, pp.2, 2019, https://doi.org/10.4014/jmb.2011.11032
  11. Neuroprotective Effect of Gallocatechin Gallate on Glutamate-Induced Oxidative Stress in Hippocampal HT22 Cells vol.26, pp.5, 2019, https://doi.org/10.3390/molecules26051387
  12. Ginsenoside from ginseng: a promising treatment for inflammatory bowel disease vol.73, pp.3, 2019, https://doi.org/10.1007/s43440-020-00213-z
  13. Inhibition of Alveolar Bone Destruction by Red Ginseng Extract in an Experimental Animal Periodontitis Model vol.50, pp.7, 2021, https://doi.org/10.3746/jkfn.2021.50.7.672
  14. Azaphilones from an Endophytic Penicillium sp. Prevent Neuronal Cell Death via Inhibition of MAPKs and Reduction of Bax/Bcl-2 Ratio vol.84, pp.8, 2021, https://doi.org/10.1021/acs.jnatprod.1c00298
  15. Metabolomic analysis of untargeted bovine uterine secretions in dairy cows with endometritis using ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry vol.139, 2019, https://doi.org/10.1016/j.rvsc.2021.07.006
  16. Tandem Mass Spectrometry for the Analysis of Ginsenosides in a Phytoadaptogene Composition with Antitumor Properties vol.55, pp.6, 2019, https://doi.org/10.1134/s0040579521050225
  17. The Neuroprotective Effect of Increased PINK1 Expression Following Glutamate Excitotoxicity in Neuronal Cells vol.480, 2019, https://doi.org/10.1016/j.neuroscience.2021.11.020