DOI QR코드

DOI QR Code

Protective effect of Cordyceps militaris against hydrogen peroxide-induced oxidative stress in vitro

  • He, Mei Tong (Department of Food Science and Nutrition, Pusan National University) ;
  • Lee, Ah Young (Department of Food Science, Gyeongnam National University of Science and Technology) ;
  • Park, Chan Hum (Department of Medicinal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration) ;
  • Cho, Eun Ju (Department of Food Science and Nutrition, Pusan National University)
  • Received : 2018.09.05
  • Accepted : 2019.05.14
  • Published : 2019.08.01

Abstract

BACKGROUND/OBJECTIVES: Excessive production of reactive oxygen species (ROS) such as hydroxyl (${\cdot}OH$), nitric oxide (NO), and hydrogen peroxide ($H_2O_2$) is reported to induce oxidative stress. ROS generated by oxidative stress can potentially damage glial cells in the nervous system. Cordyceps militaris (CM), a kind of natural herb widely found in East Asia. In this study, we investigated the free radical scavenging activity of the CM extract and its neuroprotective effects in $H_2O_2$-induced C6 glial cells. MATERIALS/METHODS: The ethanol extract of CM ($100-1,000{\mu}g/mL$) was used to measure DPPH, ${\cdot}OH$, and NO radical scavenging activities. In addition, hydrogen peroxide ($H_2O_2$)-induced C6 glial cells were treated with CM at $0.5-2.5{\mu}g/mL$ for measurement of cell viability, ROS production, and protein expression resulting from oxidative stress. RESULTS: The CM extract showed high scavenging activities against DPPH, ${\cdot}OH$, and NO radicals at concentration of $1,000{\mu}g/mL$. Treatment of CM with $H_2O_2$-induced oxidative stress in C6 glial cells significantly increased cell viability, and decreased ROS production. Cyclooxygenase-2 and inducible nitric oxide synthase protein expression was down-regulated in CM-treated groups. In addition, the protein expression level of phospho-p38 mitogen-activated protein kinase (p-p38 MAPK), phospho-c-Jun N-terminal kinase (p-JNK), and phospho-extracellular regulated protein kinases (p-ERK) in $H_2O_2$-induced C6 glial cells was down-regulated upon CM administration. CONCLUSION: CM exhibited radical scavenging activity and protective effect against $H_2O_2$ as indicated by the increased cell viability, decreased ROS production, down-regulation of inflammation-related proteins as well as p-p38, p-JNK, and p-ERK protein levels. Therefore, we suggest that CM could play the protective role from oxidative stress in glial cells.

Keywords

References

  1. Liochev SI. Reactive oxygen species and the free radical theory of aging. Free Radic Biol Med 2013;60:1-4. https://doi.org/10.1016/j.freeradbiomed.2013.02.011
  2. Juranek I, Bezek S. Controversy of free radical hypothesis: reactive oxygen species--cause or consequence of tissue injury? Gen Physiol Biophys 2005;24:263-78.
  3. Bayir H. Reactive oxygen species. Crit Care Med 2005;33:S498-501. https://doi.org/10.1097/01.CCM.0000186787.64500.12
  4. 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
  5. Ott M, Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria, oxidative stress and cell death. Apoptosis 2007;12:913-22. https://doi.org/10.1007/s10495-007-0756-2
  6. Siddique YH, Beg T, Afzal M. Protective effect of ascorbic acid against oxidative damage induced by hydrogen peroxide in cultured human peripheral blood lymphocytes. Indian J Clin Biochem 2009;24:294-300. https://doi.org/10.1007/s12291-009-0055-5
  7. Chandra K, Salman AS, Mohd A, Sweety R, Ali KN. Protection against FCA induced oxidative stress induced DNA damage as a model of arthritis and in vitro anti-arthritic potential of Costus speciosus rhizome extract. Inter J Pharm Phytochem Res 2015;7:383-9.
  8. Gille JJ, Joenje H. Cell culture models for oxidative stress: superoxide and hydrogen peroxide versus normobaric hyperoxia. Mutat Res 1992;275:405-14. https://doi.org/10.1016/0921-8734(92)90043-O
  9. Spitz DR, Li GC, McCormick ML, Sun Y, Oberley LW. Stable H2O2-resistant variants of Chinese hamster fibroblasts demonstrate increases in catalase activity. Radiat Res 1988;114:114-24. https://doi.org/10.2307/3577149
  10. Rojkind M, Dominguez-Rosales JA, Nieto N, Greenwel P. Role of hydrogen peroxide and oxidative stress in healing responses. Cell Mol Life Sci 2002;59:1872-91. https://doi.org/10.1007/PL00012511
  11. Minghetti L, Polazzi E, Nicolini A, Levi G. Opposite regulation of prostaglandin E2 synthesis by transforming growth $factor-{\beta}1$ and interleukin 10 in activated microglial cultures. J Neuroimmunol 1998;82:31-9. https://doi.org/10.1016/S0165-5728(97)00185-9
  12. Prajeeth CK, Huehn J, Stangel M. Regulation of neuroinflammatory properties of glial cells by T cell effector molecules. Neural Regen Res 2018;13:234-6. https://doi.org/10.4103/1673-5374.226385
  13. Barnham KJ, Masters CL, Bush AI. Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov 2004;3:205-14. https://doi.org/10.1038/nrd1330
  14. Beal MF. Mitochondria take center stage in aging and neurodegeneration. Ann Neurol 2005;58:495-505. https://doi.org/10.1002/ana.20624
  15. Loh KP, Huang SH, De Silva R, Tan BK, Zhu YZ. Oxidative stress: apoptosis in neuronal injury. Curr Alzheimer Res 2006;3:327-37. https://doi.org/10.2174/156720506778249515
  16. Li YZ, Ren S, Yan XT, Li HP, Li W, Zheng B, Wang Z, Liu YY. Improvement of Cisplatin-induced renal dysfunction by Schisandra chinensis stems via anti-inflammation and anti-apoptosis effects. J Ethnopharmacol 2018;217:228-37. https://doi.org/10.1016/j.jep.2018.01.033
  17. Choi YJ, Kim HS, Lee J, Chung J, Lee JS, Choi JS, Yoon TR, Kim HK, Chung HY. Down-regulation of oxidative stress and COX-2 and iNOS expressions by dimethyl lithospermate in aged rat kidney. Arch Pharm Res 2014;37:1032-8. https://doi.org/10.1007/s12272-014-0332-6
  18. Kumamoto H, Ooya K. Immunohistochemical detection of phosphorylated JNK, p38 MAPK, and ERK5 in ameloblastic tumors. J Oral Pathol Med 2007;36:543-9. https://doi.org/10.1111/j.1600-0714.2007.00555.x
  19. Wu XF, Zhang M, Bhandari B, Li Z. Effects of microwave-assisted pulse-spouted bed freeze-drying (MPSFD) on volatile compounds and structural aspects of Cordyceps militaris. J Sci Food Agric 2018;98:4634-43. https://doi.org/10.1002/jsfa.8993
  20. Huang SJ, Huang FK, Li YS, Tsai SY. The quality improvement of solid-state fermentation with Cordyceps militaris by UVB irradiation. Food Technol Biotechnol 2017;55:445-53.
  21. Das SK, Masuda M, Sakurai A, Sakakibara M. Medicinal uses of the mushroom Cordyceps militaris: current state and prospects. Fitoterapia 2010;81:961-8. https://doi.org/10.1016/j.fitote.2010.07.010
  22. Chu HL, Chien JC, Duh PD. Protective effect of Cordyceps militaris against high glucose-induced oxidative stress in human umbilical vein endothelial cells. Food Chem 2011;129:871-6. https://doi.org/10.1016/j.foodchem.2011.05.037
  23. Ng TB, Wang HX. Pharmacological actions of Cordyceps, a prized folk medicine. J Pharm Pharmacol 2005;57:1509-19. https://doi.org/10.1211/jpp.57.12.0001
  24. Ramesh T, Yoo SK, Kim SW, Hwang SY, Sohn SH, Kim IW, Kim SK. Cordycepin (3'-deoxyadenosine) attenuates age-related oxidative stress and ameliorates antioxidant capacity in rats. Exp Gerontol 2012;47:979-87. https://doi.org/10.1016/j.exger.2012.09.003
  25. Bawadekji A, Al Ali K, Al Ali M. A review of the bioactive compound and medicinal value of Cordyceps militaris. J North Basic Appl Sci 2016;347:1-3.
  26. Li XT, Li HC, Li CB, Dou DQ, Gao MB. Protective effects on mitochondria and anti-aging activity of polysaccharides from cultivated fruiting bodies of Cordyceps militaris. Am J Chin Med 2010;38:1093-106. https://doi.org/10.1142/S0192415X10008494
  27. Lin R, Liu H, Wu S, Pang L, Jia M, Fan K, Jia S, Jia L. Production and in vitro antioxidant activity of exopolysaccharide by a mutant, Cordyceps militaris SU5-08. Int J Biol Macromol 2012;51:153-7. https://doi.org/10.1016/j.ijbiomac.2012.04.011
  28. Park JM, Lee JS, Lee KR, Ha SJ, Hong EK. Cordyceps militaris extract protects human dermal fibroblasts against oxidative stress-induced apoptosis and premature senescence. Nutrients 2014;6:3711-26. https://doi.org/10.3390/nu6093711
  29. Hatano T, Edamatsu R, Hiramatsu M, Mori A, Fujita Y, Yasuhara T, Yoshida T, Okuda T. Effects of the interaction of tannins with co-existing substances. VI.: effects of tannins and related polyphenols on superoxide anion radical, and on 1, 1-Diphenyl-2-picrylhydrazyl radical. Chem Pharm Bull (Tokyo) 1989;37:2016-21. https://doi.org/10.1248/cpb.37.2016
  30. Chung SK, Osawa T, Kawakishi S. Hydroxyl radical-scavenging effects of spices and scavengers from brown mustard (Brassica nigra). Biosci Biotechnol Biochem 1997;61:118-23. https://doi.org/10.1271/bbb.61.118
  31. Marcocci L, Maguire JJ, Droy-Lefaix MT, Packer L. The nitric oxide-scavenging properties of Ginkgo biloba extract EGb 761. Biochem Biophys Res Commun 1994;201:748-55. https://doi.org/10.1006/bbrc.1994.1764
  32. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983;65:55-63. https://doi.org/10.1016/0022-1759(83)90303-4
  33. Ma WW, Hou CC, Zhou X, Yu HL, Xi YD, Ding J, Zhao X, Xiao R. Genistein alleviates the mitochondria-targeted DNA damage induced by ${\beta}$-amyloid peptides 25-35 in C6 glioma cells. Neurochem Res 2013;38:1315-23. https://doi.org/10.1007/s11064-013-1019-y
  34. Blois MS. Antioxidant determinations by the use of a stable free radical. Nature 1958;181:1199-200. https://doi.org/10.1038/1811199a0
  35. Hayyan M, Hashim MA, AlNashef IM. Superoxide ion: generation and chemical implications. Chem Rev 2016;116:3029-85. https://doi.org/10.1021/acs.chemrev.5b00407
  36. Patel Rajesh M, Patel Natvar J. In vitro antioxidant activity of coumarin compounds by DPPH, Superoxide and nitric oxide free radical scavenging methods. J Adv Pharm Technol Res 2011;1:52-68.
  37. Aruoma OI. Free radicals, oxidative stress, and antioxidants in human health and disease. J Am Oil Chem Soc 1998;75:199-212. https://doi.org/10.1007/s11746-998-0032-9
  38. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 2007;39:44-84. https://doi.org/10.1016/j.biocel.2006.07.001
  39. Apel K, Hirt H. Reactive oxygen species: metabolism, oxidative stress, and signal transduction. Annu Rev Plant Biol 2004;55:373-99. https://doi.org/10.1146/annurev.arplant.55.031903.141701
  40. Dong CH, Yang T, Lian T. A comparative study of the antimicrobial, antioxidant, and cytotoxic activities of methanol extracts from fruit bodies and fermented mycelia of caterpillar medicinal mushroom Cordyceps militaris (Ascomycetes). Int J Med Mushrooms 2014;16:485-95. https://doi.org/10.1615/IntJMedMushrooms.v16.i5.70
  41. Jessen KR, Mirsky R. Glial cells in the enteric nervous system contain glial fibrillary acidic protein. Nature 1980;286:736-7. https://doi.org/10.1038/286736a0
  42. Sadigh-Eteghad S, Majdi A, Mahmoudi J, Golzari SE, Talebi M. Astrocytic and microglial nicotinic acetylcholine receptors: an overlooked issue in Alzheimer's disease. J Neural Transm (Vienna) 2016;123:1359-67. https://doi.org/10.1007/s00702-016-1580-z
  43. Choi EO, Jeong JW, Park C, Hong SH, Kim GY, Hwang HJ, Cho EJ, Choi YH. Baicalein protects C6 glial cells against hydrogen peroxide-induced oxidative stress and apoptosis through regulation of the Nrf2 signaling pathway. Int J Mol Med 2016;37:798-806. https://doi.org/10.3892/ijmm.2016.2460
  44. Valori CF, Brambilla L, Martorana F, Rossi D. The multifaceted role of glial cells in amyotrophic lateral sclerosis. Cell Mol Life Sci 2014;71:287-97. https://doi.org/10.1007/s00018-013-1429-7
  45. Puves D, Augustine GJ, Fitzpatrick D, Hall WC, LaMantia AS, White LE. Neuroscience. 5th ed. Sunderland (MA): Sinauer Associates, Inc.;2012. p.560-80.
  46. McGeer PL, McGeer EG. Glial cell reactions in neurodegenerative diseases: pathophysiology and therapeutic interventions. Alzheimer Dis Assoc Disord 1998;12 Suppl 2:S1-6. https://doi.org/10.1097/00002093-199803000-00001
  47. Yu H, Liu Z, Zhou H, Dai W, Chen S, Shu Y, Feng J. JAK-STAT pathway modulates the roles of iNOS and COX-2 in the cytoprotection of early phase of hydrogen peroxide preconditioning against apoptosis induced by oxidative stress. Neurosci Lett 2012;529:166-71. https://doi.org/10.1016/j.neulet.2012.09.013
  48. Minghetti L. Cyclooxygenase-2 (COX-2) in inflammatory and degenerative brain diseases. J Neuropathol Exp Neurol 2004;63:901-10. https://doi.org/10.1093/jnen/63.9.901
  49. Landino LM, Crews BC, Timmons MD, Morrow JD, Marnett LJ. Peroxynitrite, the coupling product of nitric oxide and superoxide, activates prostaglandin biosynthesis. Proc Natl Acad Sci U S A 1996;93:15069-74. https://doi.org/10.1073/pnas.93.26.15069
  50. Xie Z, Smith CJ, Van Eldik LJ. Activated glia induce neuron death via MAP kinase signaling pathways involving JNK and p38. Glia 2004;45:170-9. https://doi.org/10.1002/glia.10314
  51. Kwon SH, Kim JA, Hong SI, Jung YH, Kim HC, Lee SY, Jang CG. Loganin protects against hydrogen peroxide-induced apoptosis by inhibiting phosphorylation of JNK, p38, and ERK 1/2 MAPKs in SH-SY5Y cells. Neurochem Int 2011;58:533-41. https://doi.org/10.1016/j.neuint.2011.01.012
  52. Choi YH, Kim GY, Lee HH. Anti-inflammatory effects of cordycepin in lipopolysaccharide-stimulated RAW 264.7 macrophages through Toll-like receptor 4-mediated suppression of mitogen-activated protein kinases and $NF-{\kappa}B$ signaling pathways. Drug Des Devel Ther 2014;8:1941-53. https://doi.org/10.2147/DDDT.S71957
  53. Shin JW, Seol IC, Son CG. Interpretation of animal dose and human equivalent dose for drug development. J Korean Orient Med 2010;31:1-7.

Cited by

  1. Sweet Cherry Byproducts Processed by Green Extraction Techniques as a Source of Bioactive Compounds with Antiaging Properties vol.9, pp.5, 2019, https://doi.org/10.3390/antiox9050418
  2. Hexarelin Modulation of MAPK and PI3K/Akt Pathways in Neuro-2A Cells Inhibits Hydrogen Peroxide-Induced Apoptotic Toxicity vol.14, pp.5, 2019, https://doi.org/10.3390/ph14050444