DOI QR코드

DOI QR Code

The preventive effect of Mori Ramulus on oxidative stress-induced cellular damage in skeletal L6 myoblasts through Nrf2-mediated activation of HO-1

  • Cheol, Park (Division of Basic Sciences, College of Liberal Studies, Dong-eui University) ;
  • Hyesook, Lee (Anti-Aging Research Center, Dong-eui University) ;
  • Sung Ok, Kim (Department of Food and Nutrition, College of Life and Health, Kyungsung University) ;
  • Eun‑Woo, Lee (Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-eui University) ;
  • Hyun‑Tai, Lee (Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-eui University) ;
  • Hyun Ju, Kwon (Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-eui University) ;
  • Byung Woo, Kim (Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-eui University) ;
  • Gi‑Young, Kim (Department of Marine Life Science, Jeju National University) ;
  • Mi Ryeo, Kim (Department of Pharmacology, College of Korean Medicine, Daegu Haany University) ;
  • Yung Hyun, Choi (Anti-Aging Research Center, Dong-eui University)
  • 투고 : 2022.03.17
  • 심사 : 2022.06.20
  • 발행 : 2023.01.15

초록

The aim of the present study is to investigate the preventive effect of water extract of Mori Ramulus (MRWE) on oxidative stress-mediated cellular damages in rat skeletal L6 myoblasts. Our results demonstrated that MRWE pretreatment markedly improved cell survival and suppressed cell cycle arrest at the G2/M phase and apoptosis in hydrogen peroxide (H2O2)-treated L6 cells. H2O2-triggered DNA damage was also notably reduced by MRWE, which since it was correlated with protection of reactive oxygen species (ROS) production. Additionally, H2O2 stimulated cytosolic release of cytochrome c and up-regulation of Bax/Bcl-2 ratio, whereas MRWE suppressed these changes following by H2O2. Moreover, MRWE inhibited the cleavage of poly(ADP-ribose) polymerase as well as the activity of caspase-3 by H2O2. Furthermore, MRWE enhanced H2O2-mediated expression of nuclear factor erythroid 2-associated factor 2 (Nrf2) and its representative downstream enzyme, heme oxygenase-1 (HO-1). However, the protective effects of MRWE on H2O2-induced ROS production, cell cycle arrest and apoptosis were significantly attenuated by HO-1 inhibitor. In conclusion, our present results suggests that MRWE could protect L6 myoblasts from H2O2-induced cellular injury by inhibiting ROS generation along with Nrf2-mediated activation of HO-1, indicating this finding may expand the scope of application of Mori Ramulus in medicine.

키워드

과제정보

This research was funded by the National Research Foundation of Korea Grant (NRF-2021R1A2C201471711 and 2021R1A2C200954911) and Korea Basic Science Institute Grant funded (2020R1A6C101A201)

참고문헌

  1. Chen C, Mohamad Razali UH, Saikim FH, Mahyudin A, Mohd Noor NQI (2021) Morus alba L. plant: Bioactive compounds and potential as a functional food ingredient. Foods 10:689. https://doi.org/10.3390/foods10030689 
  2. Chan EW, Lye PY, Wong SK (2016) Phytochemistry, pharmacology, and clinical trials of Morus alba. Chin J Nat Med 14:17-30. https://doi.org/10.3724/SP.J.1009.2016.00017 
  3. He X, Fang J, Ruan Y, Wang X, Sun Y, Wu N, Zhao Z, Chang Y, Ning N, Guo H, Huang L (2018) Structures, bioactivities and future prospective of polysaccharides from Morus alba (white mulberry): a review. Food Chem 245:899-910. https://doi.org/10.1016/j.foodchem.2017.11.084 
  4. Han T, Ko E, Kim M, Choi M, Lee C, Kim IH, Shin S, Um MY (2021) Mori Ramulus inhibits pancreatic beta-cell apoptosis and prevents insulin resistance by restoring hepatic mitochondrial function. Antioxidants (Basel) 10:901. https://doi.org/10.3390/antiox10060901 
  5. Park SY, Jin B, Shin JH, Adisakwattana S, Kwon O (2017) Standardized Mori ramulus extract improves insulin secretion and insulin sensitivity in C57BLKS/J db/db mice and INS-1 cells. Biomed Pharmacother 92:308-315. https://doi.org/10.1016/j.biopha.2017.05.080 
  6. Xu L, Yang F, Wang J, Huang H, Huang Y (2015) Anti-diabetic effect mediated by Ramulus mori polysaccharides. Carbohydr Polym 117:63-69. https://doi.org/10.1016/j.carbpol.2014.09.052 
  7. Kim TI, Kwon EB, Oh YC, Go Y, Choi JG (2021) Mori ramulus and its major component morusin inhibit herpes simplex virus type 1 replication and the virus-induced reactive oxygen species. Am J Chin Med 49:163-179. https://doi.org/10.1142/S0192415X21500099 
  8. Wan LZ, Ma B, Zhang YQ (2014) Preparation of morusin from Ramulus mori and its effects on mice with transplanted H22 hepatocarcinoma. BioFactors 40:636-645. https://doi.org/10.1002/biof.1191 
  9. Jiang Q, Li X, Tian Y, Lin Q, Xie H, Lu W, Chi Y, Chen D (2017) Lyophilized aqueous extracts of Mori Fructus and Mori Ramulus protect mesenchymal stem cells from ·OH-treated damage: bioassay and antioxidant mechanism. BMC Complement Altern Med 17:242. https://doi.org/10.1186/s12906-017-1730-3 
  10. Chen Z, Du X, Yang Y, Cui X, Zhang Z, Li Y (2018) Comparative study of chemical composition and active components against α-glucosidase of various medicinal parts of Morus alba L. Biomed Chromatogr 32:e4328. https://doi.org/10.1002/bmc.4328 
  11. Kwon DH, Jeong JW, Choi EO, Lee HW, Lee KW, Kim KY, Kim SG, Hong SH, Kim GY, Park C, Hwang HJ, Son CG, Choi YH (2017) Inhibitory effects on the production of inflammatory mediators and reactive oxygen species by Mori folium in lipopolysaccharide-stimulated macrophages and zebrafish. An Acad Bras Cienc 89:661-674. https://doi.org/10.1590/0001-3765201720160836 
  12. Park HM, Hong JH (2014) Effect of extraction methods on antioxidant activities of Mori Ramulus. J Korean Soc Food Sci Nutr 43:1709-1715. https://doi.org/10.3746/jkfn.2014.43.11.1709 
  13. Li X, Wang L, Gao X, Li G, Cao H, Song D, Cai S, Liang T, Zhang B, Du G (2015) Mechanisms of protective effect of Ramulus Mori polysaccharides on renal injury in high-fat diet/streptozotocin-induced diabetic rats. Cell Physiol Biochem 37:2125-2134. https://doi.org/10.1159/000438570 
  14. Guo C, Li R, Zheng N, Xu L, Liang T, He Q (2013) Anti-diabetic effect of ramulus mori polysaccharides, isolated from Morus alba L., on STZ-diabetic mice through blocking inflammatory response and attenuating oxidative stress. Int Immunopharmacol 16:93-99. https://doi.org/10.1016/j.intimp.2013.03.029 
  15. Xia P, Gao X, Duan L, Zhang W, Sun YF (2018) Mulberrin (Mul) reduces spinal cord injury (SCI)-induced apoptosis, inflammation and oxidative stress in rats via miroRNA-337 by targeting Nrf-2. Biomed Pharmacother 107:1480-1487. https://doi.org/10.1016/j.biopha.2018.07.082 
  16. Tran PL, Tran PT, Tran HNK, Lee S, Kim O, Min BS, Lee JH (2018) A prenylated flavonoid, 10-oxomornigrol F, exhibits anti-inflammatory effects by activating the Nrf2/heme oxygenase-1 pathway in macrophage cells. Int Immunopharmacol 55:165-173. https://doi.org/10.1016/j.intimp.2017.12.015 
  17. Gao XH, Zhang SD, Wang LT, Yu L, Zhao XL, Ni HY, Wang YQ, Wang JD, Shan CH, Fu YJ (2020) Anti-inflammatory effects of neochlorogenic acid extract from mulberry leaf (Morus alba L.) against LPS-stimulated inflammatory response through mediating the AMPK/Nrf2 signaling pathway in A549 cells. Molecules 25:1385. https://doi.org/10.3390/molecules25061385 
  18. Hu D, Bao T, Lu Y, Su H, Ke H, Chen W (2020) Polysaccharide from mulberry fruit (Morus alba L.) protects against palmitic-acid-induced hepatocyte lipotoxicity by activating the Nrf2/ARE signaling pathway. J Agric Food Chem 68:13016-13024. https://doi.org/10.1021/acs.jafc.9b03335 
  19. Wen L, Shi D, Zhou T, Tu J, He M, Jiang Y, Yang B (2020) Identification of two novel prenylated flavonoids in mulberry leaf and their bioactivities. Food Chem 315:126236. https://doi.org/10.1016/j.foodchem.2020.126236 
  20. He L, He T, Farrar S, Ji L, Liu T, Ma X (2017) Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell Physiol Biochem 44:532-553. https://doi.org/10.1159/000485089 
  21. Benhar M (2020) Oxidants, antioxidants and thiol redox switches in the control of regulated cell death pathways. Antioxidants (Basel) 9:309. https://doi.org/10.3390/antiox9040309 
  22. Bouviere J, Fortunato RS, Dupuy C, Werneck-de-Castro JP, Carvalho DP, Louzada RA (2021) Exercise-stimulated ROS sensitive signaling pathways in skeletal muscle. Antioxidants (Basel) 10:537. https://doi.org/10.3390/antiox10040537 
  23. Mosca N, Petrillo S, Bortolani S, Monforte M, Ricci E, Piemonte F, Tasca G (2021) Redox homeostasis in muscular dystrophies. Cells 10:1364. https://doi.org/10.3390/cells10061364 
  24. Qaisar R, Bhaskaran S, Premkumar P, Ranjit R, Natarajan KS, Ahn B, Riddle K, Claflin DR, Richardson A, Brooks SV, Van Remmen H (2018) Oxidative stress-induced dysregulation of excitation-contraction coupling contributes to muscle weakness. J Cachexia Sarcopenia Muscle 9:1003-1017. https://doi.org/10.1002/jcsm.12339 
  25. Majumder A, Singh M, George AK, Tyagi SC (2019) Restoration of skeletal muscle homeostasis by hydrogen sulfide during hyperhomocysteinemia-mediated oxidative/ER stress condition (1). Can J Physiol Pharmacol 97:441-456. https://doi.org/10.1139/cjpp-2018-0501 
  26. Lian D, Chen MM, Wu H, Deng S, Hu X (2022) The role of oxidative stress in skeletal muscle myogenesis and muscle disease. Antioxidants (Basel) 11:755. https://doi.org/10.3390/antiox11040755 
  27. Brillo V, Chieregato L, Leanza L, Muccioli S, Costa R (2021) Mitochondrial dynamics, ROS, and cell signaling: a blended overview. Life (Basel) 11:332. https://doi.org/10.3390/life11040332 
  28. Park C, Ji SY, Lee H, Choi SH, Kwon CY, Kim SY, Lee ET, Choo ST, Kim GY, Choi YH, Kim MR (2021) Mori Ramulus suppresses hydrogen peroxide-induced oxidative damage in murine myoblast C2C12 cells through activation of AMPK. Int J Mol Sci 22:11729. https://doi.org/10.3390/ijms222111729 
  29. Sim KH, Shu MS, Kim S, Kim JY, Choi BH, Lee YJ (2021) Cilostazol induces apoptosis and inhibits proliferation of hepatocellular carcinoma cells by activating AMPK. Biotechnol Bioprocess Eng 26:776-785. https://doi.org/10.1007/s12257-021-0002-8 
  30. Park C, Lee H, Hong SH, Kim JH, Park SK, Jeong JW, Kim GY, Hyun JW, Yun SJ, Kim BW, Kim WJ, Choi YH (2019) Protective effect of diphlorethohydroxycarmalol against oxidative stress-induced DNA damage and apoptosis in retinal pigment epithelial cells. Cutan Ocul Toxicol 38:298-308. https://doi.org/10.1080/15569527.2019.1613425 
  31. Choi MJ, Mukherjee S, Yun JW (2021) Loss of ADAMTS15 promotes browning in 3T3-L1 white adipocytes via activation of β3-adrenergic receptor. Biotechnol Bioprocess Eng 26:188-200. https://doi.org/10.1007/s12257-021-0036-y 
  32. Liang Y, Kong D, Zhang Y, Li S, Li Y, Ramamoorthy S, Ma J (2020) Fisetin inhibits cell proliferation and induces apoptosis via JAK/STAT3 signaling pathways in human thyroid TPC 1 cancer cells. Biotechnol Bioprocess Eng 25:197-205. https://doi.org/10.1007/s12257-019-0326-9 
  33. Shi T, van Soest DMK, Polderman PE, Burgering BMT, Dansen TB (2021) DNA damage and oxidant stress activate p53 through difierential upstream signaling pathways. Free Radic Biol Med 172:298-311. https://doi.org/10.1016/j.freeradbiomed.2021.06.013 
  34. Beyfuss K, Hood DA (2018) A systematic review of p53 regulation of oxidative stress in skeletal muscle. Redox Rep 23:100-117. https://doi.org/10.1080/13510002.2017.1416773 
  35. Panieri E, Gogvadze V, Norberg E, Venkatesh R, Orrenius S, Zhivotovsky B (2013) Reactive oxygen species generated in different compartments induce cell death, survival, or senescence. Free Radic Biol Med 57:176-187. https://doi.org/10.1016/j.freeradbiomed.2012.12.024 
  36. Santa-Gonzalez GA, Gomez-Molina A, Arcos-Burgos M, Meyer JN, Camargo M (2016) Distinctive adaptive response to repeated exposure to hydrogen peroxide associated with upregulation of DNA repair genes and cell cycle arrest. Redox Biol 9:124-133. https://doi.org/10.1016/j.redox.2016.07.004 
  37. Oyama K, Takahashi K, Sakurai K (2011) Hydrogen peroxide induces cell cycle arrest in cardiomyoblast H9c2 cells, which is related to hypertrophy. Biol Pharm Bull 34:501-506. https://doi.org/10.1248/bpb.34.501 
  38. Jiang Q, Yin J, Chen J, Ma X, Wu M, Liu G, Yao K, Tan B, Yin Y (2020) Mitochondria-targeted antioxidants: A step towards disease treatment. Oxid Med Cell Longev 2020:8837893. https://doi.org/10.1155/2020/8837893 
  39. Cordelli E, Bignami M, Pacchierotti F (2021) Comet assay: a versatile but complex tool in genotoxicity testing. Toxicol Res (Camb) 10:68-78. https://doi.org/10.1093/toxres/tfaa093 
  40. Raavi V, Perumal V, Paul SFD (2021) Potential application of γ-H2AX as a biodosimetry tool for radiation triage. Mutat Res Rev Mutat Res 787:108350. https://doi.org/10.1016/j.mrrev.2020.108350 
  41. Joo MS, Kim WD, Lee KY, Kim JH, Koo JH, Kim SG (2016) AMPK facilitates nuclear accumulation of Nrf2 by phosphorylating at serine 550. Mol Cell Biol 36:1931-1942. https://doi.org/10.1128/MCB.00118-16 
  42. Chen W, Zhang YN, Jia QQ, Ji A, Shao SX, Zhang L, Gong M, Yin Q, Huang XL (2020) MicroRNA-214 protects L6 skeletal myoblasts against hydrogen peroxide-induced apoptosis. Free Radic Res 54:162-172. https://doi.org/10.1080/10715762.2020.1730828 
  43. Dam AD, Mitchell AS, Quadrilatero J (2013) Induction of mitochondrial biogenesis protects against caspase-dependent and caspase-independent apoptosis in L6 myoblasts. Biochim Biophys Acta 1833:3426-3435. https://doi.org/10.1016/j.bbamcr.2013.04.014 
  44. Dadsena S, King LE, Garcia-Saez AJ (2021) Apoptosis regulation at the mitochondria membrane level. Biochim Biophys Acta Biomembr 1863:183716. https://doi.org/10.1016/j.bbamem.2021.183716 
  45. Xiong S, Mu T, Wang G, Jiang X (2014) Mitochondria-mediated apoptosis in mammals. Protein Cell 5:737-749. https://doi.org/10.1007/s13238-014-0089-1 
  46. Liu T, Lv YF, Zhao JL, You QD, Jiang ZY (2021) Regulation of Nrf2 by phosphorylation: Consequences for biological function and therapeutic implications. Free Radic Biol Med 168:129-141. https://doi.org/10.1016/j.freeradbiomed.2021.03.034 
  47. Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J (2016) Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cell Mol Life Sci 73:3221-3247. https://doi.org/10.1007/s00018-016-2223-0 
  48. Yu C, Xiao JH (2021) The Keap1-Nrf2 system: a mediator between oxidative stress and aging. Oxid Med Cell Longev 2021:6635460. https://doi.org/10.1155/2021/6635460