DOI QR코드

DOI QR Code

Ethanol Extract of Ganoderma lucidum Augments Cellular Anti-oxidant Defense through Activation of Nrf2/HO-1

  • Lee, Yoo-hwan (Departments of Acupuncture and Moxibustion, Dong-Eui University College of Korean Medicine) ;
  • Kim, Jung-hee (Departments of Acupuncture and Moxibustion, Dong-Eui University College of Korean Medicine) ;
  • Song, Choon-ho (Departments of Acupuncture and Moxibustion, Dong-Eui University College of Korean Medicine) ;
  • Jang, Kyung-jeon (Departments of Acupuncture and Moxibustion, Dong-Eui University College of Korean Medicine) ;
  • kim, Cheol-hong (Departments of Acupuncture and Moxibustion, Dong-Eui University College of Korean Medicine) ;
  • Kang, Ji-Sook (Anti-Aging Research Center, Dong-Eui University) ;
  • Choi, Yung-hyun (Anti-Aging Research Center, Dong-Eui University) ;
  • Yoon, Hyun-Min (Departments of Acupuncture and Moxibustion, Dong-Eui University College of Korean Medicine)
  • Received : 2016.01.24
  • Accepted : 2016.02.17
  • Published : 2016.03.31

Abstract

Objectives: The mushroom Ganoderma lucidum has been widely used as a traditional herbal medicine for many years. Although several studies have focused on the anti-oxidative activity of this mushroom, the molecular mechanisms underlying its activity have not yet been clearly established. The present study investigated the cytoprotective effect of ethanol extract of Ganoderma lucidum (EGL) against oxidative stress (hydrogen peroxide, $H_2O_2$) and elucidated the underlying mechanisms in a C2C12 myoblast cell line. Methods: Oxidative stress markers were determined by using the comet assay to measure reactive oxygen species (ROS) generation and deoxyribonucleic acid (DNA) damage. Cell viability and Western blotting analyses were employed to evaluate the cellular response to EGL and $H_2O_2$ in C2C12 cells. Transfection with nuclear factor erythroid 2-related factor 2 (Nrf2)-specific small interfering ribonucleic acid (siRNA) was conducted to understand the relationship between Nrf2 expression and $H_2O_2$-induced growth inhibition. Results: The results showed that EGL effectively inhibited $H_2O_2$-induced growth and the generation of ROS. EGL markedly suppressed $H_2O_2$-induced comet-like DNA formation and phosphorylation of histone H2AX at serine 139 ($p-{\gamma}H2AX$), a widely used marker of DNA damage, suggesting that EGL prevented $H_2O_2$-induced DNA damage. Furthermore, the EGL treatment effectively induced the expression of Nrf2, as well as heme oxygenase-1 (HO-1), with parallel phosphorylation and nuclear translocation of Nrf2 in the C2C12 myoblasts. However, zinc protoporphyrin IX, a HO-1 inhibitor, significantly abolished the protective effects of EGL against $H_2O_2$-induced accumulation of ROS and reduced cell growth. Notably, transient transfection with Nrf2-specific siRNA attenuated the cytoprotective effects and HO-1 induction by EGL, indicating that EGL induced the expression of HO-1 in an Nrf2-dependent manner. Conclusion: Collectively, these results demonstrate that EGL augments the cellular anti-oxidant defense capacity through activation of Nrf2/HO-1, thereby protecting C2C12 myoblasts from $H_2O_2$-induced oxidative cytotoxicity.

Keywords

References

  1. Szczesny B, Olah G, Walker DK, Volpi E, Rasmussen BB, Szabo C, et al. Deficiency in repair of the mitochondrial genome sensitizes proliferating myoblasts to oxidative damage. PLoS One. 2013;8(9):e75201. https://doi.org/10.1371/journal.pone.0075201
  2. Miller CJ, Gounder SS, Kannan S, Goutam K, Muthusamy VR, Firpo MA, et al. Disruption of Nrf2/ARE signaling impairs antioxidant mechanisms and promotes cell degradation pathways in aged skeletal muscle. Biochim Biophys Acta. 2012;1822(6):1038-50. https://doi.org/10.1016/j.bbadis.2012.02.007
  3. Ji LL, Gomez-Cabrera MC, Vina J. Exercise and hormesis: activation of cellular antioxidant signaling pathway. Ann N Y Acad Sci. 2006;1067:425-35. https://doi.org/10.1196/annals.1354.061
  4. Forman HJ. Use and abuse of exogenous $H_2O_2$ in studies of signal transduction. Free Radic Biol Med. 2007;42(7):926-32. https://doi.org/10.1016/j.freeradbiomed.2007.01.011
  5. Halliwell B. Oxidative stress and neurodegeneration: where are we now?. J Neurochem. 2006;97(6):1634-58. https://doi.org/10.1111/j.1471-4159.2006.03907.x
  6. Song W, Kwak HB, Lawler JM. Exercise training attenuates age-induced changes in apoptotic signaling in rat skeletal muscle. Antioxid Redox Signal. 2006;8(3-4):517-28. https://doi.org/10.1089/ars.2006.8.517
  7. Powers SK, Jackson MJ. Exercise-induced oxidative stress: cellular mechanisms and impact on muscle force production. Physiol Rev. 2008;88(4):1243-76. https://doi.org/10.1152/physrev.00031.2007
  8. Sanodiya BS, Thakur GS, Baghel RK, Prasad GB, Bisen PS. Ganoderma lucidum: a potent pharmacological macrofungus. Curr Pharm Biotechnol. 2009;10(8):717-42. https://doi.org/10.2174/138920109789978757
  9. Wachtel-Galor S, Tomlinson B, Benzie IF. Ganoderma lucidum ("Lingzhi"), a Chinese medicinal mushroom: biomarker responses in a controlled human supplementation study. Br J Nutr. 2004;91(2):263-9. https://doi.org/10.1079/BJN20041039
  10. Cheng S, Sliva D. Ganoderma lucidum for cancer treatment: we are close but still not there. Integr Cancer Ther. 2015;14(3):249-57. https://doi.org/10.1177/1534735414568721
  11. Yuen JW, Gohel MD, Ng CF. The differential immunological activities of Ganoderma lucidum on human pre-cancerous uroepithelial cells. J Ethnopharmacol. 2011;135(3):711-8. https://doi.org/10.1016/j.jep.2011.04.005
  12. Bhardwaj N, Katyal P, Sharma AK. Suppression of inflammatory and allergic responses by pharmacologically potent fungus Ganoderma lucidum. Recent Pat Inflamm Allergy Drug Discov. 2014;8(2):104-17. https://doi.org/10.2174/1872213X08666140619110657
  13. Ma HT, Hsieh JF, Chen ST. Anti-diabetic effects of Ganoderma lucidum. Phytochemistry. 2015;114:109-13. https://doi.org/10.1016/j.phytochem.2015.02.017
  14. Ha do T, Oh J, Khoi NM, Dao TT, Dung le V, Do TN, et al. In vitro and in vivo hepatoprotective effect of ganodermanontriol against t-BHP-induced oxidative stress. J Ethnopharmacol. 2013;150(3):875-85. https://doi.org/10.1016/j.jep.2013.09.039
  15. Lin ZB, Zhang HN. Anti-tumor and immunoregulatory activities of Ganoderma lucidum and its possible mechanisms. Acta Pharmacol Sin. 2004;25(11):1387-95.
  16. Yoon HM, Jang KJ, Han MS, Jeong JW, Kim GY, Lee JH, et al. Ganoderma lucidum ethanol extract inhibits the inflammatory response by suppressing the NF-${\kappa}B$ and toll-like receptor pathways in lipopolysaccharide-stimulated BV2 microglial cells. Exp Ther Med. 2013;5(3):957-63. https://doi.org/10.3892/etm.2013.895
  17. Jang KJ, Son IS, Shin DY, Yoon HM, Choi YH. Anti-invasive activity of ethanol extracts of Ganoderma lucidum through tightening of tight junctions and inhibition of matrix metalloproteinase activities in human gastric carcinoma cells. J Acupunct Meridian Stud. 2011;4(4):225-35. https://doi.org/10.1016/j.jams.2011.09.013
  18. Lai CS, Yu MS, Yuen WH, So KF, Zee SY, Chang RC. Antagonizing beta-amyloid peptide neurotoxicity of the anti-aging fungus Ganoderma lucidum. Brain Res. 2008;1190:215-24. https://doi.org/10.1016/j.brainres.2007.10.103
  19. Chen LW, Wang YQ, Wei LC, Shi M, Chan YS. Chinese herbs and herbal extracts for neuroprotection of dopaminergic neurons and potential therapeutic treatment of Parkinson's disease. CNS Neurol Disord Drug Targets. 2007;6(4):273-81. https://doi.org/10.2174/187152707781387288
  20. Li B, Lee DS, Kang Y, Yao NQ, An RB, Kim YC. Protective effect of ganodermanondiol isolated from the Lingzhi mushroom against tert-butyl hydroperoxide-induced hepatotoxicity through Nrf2-mediated antioxidant enzymes. Food Chem Toxicol. 2013;53:317-24. https://doi.org/10.1016/j.fct.2012.12.016
  21. Ha TB, Gerhauser C, Zhang WD, Ho-Chong-Line N, Fouraste I. New lanostanoids from Ganoderma lucidum that induce NAD(P)H:quinone oxidoreductase in cultured hepalcic7 murine hepatoma cells. Planta Med. 2000;66(7):681-4. https://doi.org/10.1055/s-2000-8647
  22. Choi S, Nguyen VT, Tae N, Lee S, Ryoo S, Min BS, et al. Anti-inflammatory and heme oxygenase-1 inducing activities of lanostane triterpenes isolated from mushroom Ganoderma lucidum in RAW264.7 cells. Toxicol Appl Pharmacol. 2014;280(3):434-42. https://doi.org/10.1016/j.taap.2014.09.007
  23. Hsieh TC, Wu JM. Suppression of proliferation and oxidative stress by extracts of Ganoderma lucidum in the ovarian cancer cell line OVCAR-3. Int J Mol Med. 2011;28(6):1065-9. https://doi.org/10.3892/ijmm.2011.788
  24. Oluba OM, Adebisi KE, Eidangbe GO, Odutuga AA, Onyeneke EC. Modulatory effect of crude aqueous extract of Lingzhi or Reishi medicinal mushroom, Ganoderma lucidum (higher Basidiomycetes), on hematological and antioxidant indices in plasmodium berghei-infected mice. Int J Med Mushrooms. 2014;16(5):499-506. https://doi.org/10.1615/IntJMedMushrooms.v16.i5.90
  25. Li C, Shi L, Chen D, Ren A, Gao T, Zhao M. Functional analysis of the role of glutathione peroxidase (GPx) in the ROS signaling pathway, hyphal branching and the regulation of ganoderic acid biosynthesis in Ganoderma lucidum. Fungal Genet Biol. 2015;82:168-80. https://doi.org/10.1016/j.fgb.2015.07.008
  26. Sakata Y, Zhuang H, Kwansa H, Koehler RC, Dore S. Resveratrol protects against experimental stroke: putative neuroprotective role of heme oxygenase 1. Exp Neurol. 2010;224(1):325-9. https://doi.org/10.1016/j.expneurol.2010.03.032
  27. Kobayashi M. Yamamoto M. Molecular mechanisms activating the Nrf2-Keap1 pathway of antioxidant gene regulation. Antioxid Redox Signal. 2005;7(3-4):385-94. https://doi.org/10.1089/ars.2005.7.385
  28. Zhang Y, Gordon GB. A strategy for cancer prevention: stimulation of the Nrf2-ARE signaling pathway. Mol Cancer Ther. 2004;3(7):885-93.
  29. Niture SK, Khatri R, Jaiswal AK. Regulation of Nrf2-an update. Free Radic Biol Med. 2014;66:36-44. https://doi.org/10.1016/j.freeradbiomed.2013.02.008
  30. Ishii T, Itoh K, Takahashi S, Sato H, Yanagawa T, Katoh Y, et al. Transcription factor Nrf2 coordinately regulates a group of oxidative stress-inducible genes in macrophages. J Biol Chem. 2000;275(21):16023-9. https://doi.org/10.1074/jbc.275.21.16023
  31. Scapagnini G, Butterfield DA, Colombrita C, Sultana R, Pascale A, Calabrese V. Ethyl ferulate, a lipophilic polyphenol, induces HO-1 and protects rat neurons against oxidative stress. Antioxid Redox Signal. 2004;6(5):811-8. https://doi.org/10.1089/ars.2004.6.811
  32. Katori M, Anselmo DM, Busuttil RW, Kupiec-Weglinski JW. A novel strategy against ischemia and reperfusion injury: cytoprotection with heme oxygenase system. Transpl Immunol. 2002;9(2-4):227-33. https://doi.org/10.1016/S0966-3274(02)00043-6
  33. Motterlini R, Foresti R. Heme oxygenase-1 as a target for drug discovery. Antioxid Redox Signal. 2014;20(11):1810-26. https://doi.org/10.1089/ars.2013.5658
  34. Chapple SJ, Siow RC, Mann GE. Crosstalk between Nrf2 and the proteasome: therapeutic potential of Nrf2 inducers in vascular disease and aging. Int J Biochem Cell Biol. 2012;44(8):1315-20. https://doi.org/10.1016/j.biocel.2012.04.021
  35. Jeong WS, Jun M, Kong AN. Nrf2: A potential molecular target for cancer chemoprevention by natural compounds. Antioxid Redox Signal. 2006;8(1-2):99-106. https://doi.org/10.1089/ars.2006.8.99
  36. Pi J, Bai Y, Reece JM, Williams J, Liu D, Freeman ML, et al. Molecular mechanism of human Nrf2 activation and degradation: role of sequential phosphorylation by protein kinase CK2. Free Radic Biol Med. 2007;42(12):1797-806. https://doi.org/10.1016/j.freeradbiomed.2007.03.001
  37. Apopa PL, He X, Ma Q. Phosphorylation of Nrf2 in the transcription activation domain by casein kinase 2 (CK2) is critical for the nuclear translocation and transcription activation function of Nrf2 in IMR-32 neuroblastoma cells. J Biochem Mol Toxicol. 2008;22(1):63-76. https://doi.org/10.1002/jbt.20212
  38. Jang KJ, Han MH, Lee BH, Kim BW, Kim CH, Yoon HM, et al. Induction of apoptosis by ethanol extracts of Ganoderma lucidum in human gastric carcinoma cells. J Acupunct Meridian Stud. 2010;3(1):24-31. https://doi.org/10.1016/S2005-2901(10)60004-0
  39. Osman AG, Mekkawy IA, Verreth J, Wuertz S, Kloas W, Kirschbaum F. Monitoring of DNA breakage in embryonic stages of the African catfish Clarias gariepinus (Burchell, 1822) after exposure to lead nitrate using alkaline comet assay. Environ Toxicol. 2008;23(6):679-87. https://doi.org/10.1002/tox.20373
  40. Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 1998;273(10):5858-68. https://doi.org/10.1074/jbc.273.10.5858

Cited by

  1. Overexpression of HO-1 assisted PM2.5-induced apoptosis failure and autophagy-related cell necrosis vol.145, 2017, https://doi.org/10.1016/j.ecoenv.2017.07.047
  2. Comparison of Antioxidative Effects of Insect Tea and Its Raw Tea (Kuding Tea) Polyphenols in Kunming Mice vol.23, pp.1, 2018, https://doi.org/10.3390/molecules23010204