Korean J Physiol Pharmacol.  2020 Sep;24(5):373-384. 10.4196/kjpp.2020.24.5.373.

Four active monomers from Moutan Cortex exert inhibitoryeffects against oxidative stress by activating Nrf2/Keap1signaling pathway

Affiliations
  • 1College of Pharmaceutical Sciences, Southwest University, Beibei, Chongqing 400716, P. R. China

Abstract

Paeonol, quercetin, -sitosterol, and gallic acid extracted from MoutanCortex had been reported to possess anti-oxidative, anti-inflammatory, and antitumoractivities. This work aimed to illustrate the potential anti-oxidative mechanismof monomers in human liver hepatocellular carcinoma (HepG2) cells-induced byhydrogen peroxide (H2O2) and to evaluate whether the hepatoprotective effect ofmonomers was independence or synergy in mice stimulated by carbon tetrachloride(CCl4). Monomers protected against oxidative stress in HepG2 cells in a doseresponsemanner by inhibiting the generation of reactive oxygen species, increasingtotal antioxidant capacity, catalase and superoxide dismutase (SOD) activities, andactivating the antioxidative pathway of nuclear factor E2-related factor 2/KelchlikeECH-associated protein 1 (Nrf2/Keap1) signaling pathway. We found that thein vitro antioxidant capacities of paeonol and quercetin were better than those of-sitosterol and gallic acid. Furthermore, paeonol apparently diminished the levelsof alanine transaminase and aspartate aminotransferase, augmented the contentsof glutathione and SOD, promoted the expressions of Nrf2 and heme oxygenase-1proteins in mice stimulated by CCl4. In HepG2 cells, paeonol, quercetin, -sitosterol,and gallic acid play a defensive role against H2O2-induced oxidative stress throughactivating Nrf2/Keap1 pathway, indicating that these monomers have anti-oxidativeproperties. Totally, paeonol and quercetin exerted anti-oxidative and hepatoprotectiveeffects, which is independent rather than synergy.

Keyword

Kelch like ECH-associated protein 1; Liver failure; Nuclear factor E2-related factor 2; Oxidative stress; Signal transduction

Figure

  • Fig. 1 Effects of four monomers and hydrogen peroxide (H2O2) on cell viability of human liver hepatocellular carcinoma (HepG2) cells. (A) The effects of monomers (paeonol, quercetin, β-sitosterol, gallic acid, and vitamin E) on HepG2 cell viability. (B) The effect of H2O2 on HepG2 cell viability at different concentration of H2O2. (C) The effects of monomers on HepG2 cell viabilities at the concentration of 7 mM H2O2. The data from three independent experiments are presented as the mean ± standard deviation. *p < 0.05, **p < 0.01, and ***p < 0.001, the H2O2 vs. control group; ##p < 0.01, and ###p < 0.001, the monomers vs. H2O2 group.

  • Fig. 2 Effects of four monomers on reactive oxygen species (ROS) generation of human liver hepatocellular carcinoma (HepG2) cells induced by hydrogen peroxide (H2O2). (A, C) ROS generation of HepG2 cells was measured at the different incubated time by fluorescence microscope assay. (B, D) ROS generation of HepG2 cells was inhibited by four monomers at 15 min incubated time through fluorescence microscope assay. The data from three independent experiments are presented as the mean ± standard deviation. **p < 0.01, and ***p < 0.001, the H2O2 vs. control group; ###p < 0.001, the monomers vs. H2O2 group.

  • Fig. 3 Activation of four monomers on the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor E2-related factor 2 (Nrf2) signaling pathway in human liver hepatocellular carcinoma (HepG2) cells induced by hydrogen peroxide (H2O2). (A) The levels of Nrf2, Keap1, NQO1, and HO-1 expressions were measured by western blot assay. (B) Translocation of Nrf2 protein from the cytosol to the nucleus was also evaluated by western blot assay. (C) Effects of four monomers on the binding capacities of Nrf2-ARE complexes in HepG2 cells after H2O2 treatment. The data from three independent experiments are presented as the mean ± standard deviation. NQO1, quinone oxidoreductase 1; HO-1, heme oxygenase-1; ARE, antioxidant-responsive element. *p < 0.05, **p < 0.01, and ***p < 0.001, the H2O2 vs. control group; #p < 0.05, ##p < 0.01, and ###p < 0.001, the monomers vs. H2O2 group.

  • Fig. 4 The expressions of Nrf2, NQO1, and HO-1 proteins were determined in the presence or absence of paeonol, quercetin and ML385 in HepG2 cells induced by H2O2. The data from three independent experiments are presented as the mean ± standard deviation. Nrf2, nuclear factor E2-related factor 2; NQO1, quinone oxidoreductase 1; HO-1, heme oxygenase-1; HepG2, human liver hepatocellular carcinoma; H2O2, hydrogen peroxide. **p < 0.01, and ***p < 0.001, the H2O2 vs. control group; ###p < 0.001, the monomers vs. H2O2 group.

  • Fig. 5 Effects of separate or combined administration of paeonol and quercetin on liver tissues and serum markers of mice after carbon tetrachloride (CCl4) intraperitoneal injection. (A) Representative histopathological changes in mice livers were observed with H&E staining. Magnification 200× (1, central venous hyperemia; 2, inflammatory infiltration; 3, hepatocyte necrosis). (B–F) Effects of active monomers on the levels of AST, ALT, AST/ALT ratio, GSH, and SOD in mice serums induced by CCl4. Paeonol and quercetin groups were pretreated with 100 mg/kg paeonol and 100 mg/kg quercetin, respectively. L-(paeonol + quercetin) group was pretreated with 50 mg/kg paeonol and 50 mg/kg quercetin. H-(paeonol + quercetin) group was pretreated with 100 mg/kg paeonol and 100 mg/kg quercetin. The data from three independent experiments are presented as the mean ± standard deviation. AST, aspartate aminotransferase; ALT, alanine transaminase; GSH, glutathione; SOD, superoxide dismutase. *p < 0.05, ***p < 0.001, the CCl4 vs. control group; #p < 0.05, ##p < 0.01, and ###p < 0.001, the monomers vs. CCl4 group.

  • Fig. 6 Effects of separate or combined administration of paeonol and quercetin on the expressions of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins in mice livers induced by carbon tetrachloride (CCl4). (A) Representative changes of Nrf2 in mice livers were obtained by immunohistochemical staining assay (magnification 200×). (B) The expressions of Nrf2 and HO-1 proteins were detected by western blot. The data from three independent experiments are presented as the mean ± standard deviation. *p < 0.05, the CCl4 vs. control group; #p < 0.05, ##p < 0.01, and ###p < 0.001, the monomers vs. CCl4 group.


Reference

1. Yu BP. 1994; Cellular defenses against damage from reactive oxygen species. Physiol Rev. 74:139–162. DOI: 10.1152/physrev.1994.74.1.139. PMID: 8295932.
Article
2. Berndt C, Lillig CH, Flohé L. 2014; Redox regulation by glutathione needs enzymes. Front Pharmacol. 5:168. DOI: 10.3389/fphar.2014.00168. PMID: 25100998. PMCID: PMC4101335.
Article
3. Wang P, Gao YM, Sun X, Guo N, Li J, Wang W, Yao LP, Fu YJ. 2017; Hepatoprotective effect of 2'-O-galloylhyperin against oxidative stress-induced liver damage through induction of Nrf2/ARE-mediated antioxidant pathway. Food Chem Toxicol. 102:129–142. DOI: 10.1016/j.fct.2017.02.016. PMID: 28213291.
4. Zitka O, Skalickova S, Gumulec J, Masarik M, Adam V, Hubalek J, Trnkova L, Kruseova J, Eckschlager T, Kizek R. 2012; Redox status expressed as GSH:GSSG ratio as a marker for oxidative stress in paediatric tumour patients. Oncol Lett. 4:1247–1253. DOI: 10.3892/ol.2012.931. PMID: 23205122. PMCID: PMC3506742.
Article
5. Lin TA, Ke BJ, Cheng CS, Wang JJ, Wei BL, Lee CL. 2019; Red quinoa bran extracts protects against carbon tetrachloride-induced liver injury and fibrosis in mice via activation of antioxidative enzyme systems and blocking TGF-β1 pathway. Nutrients. 11:395. DOI: 10.3390/nu11020395. PMID: 30781895. PMCID: PMC6412755.
Article
6. Taguchi K, Motohashi H, Yamamoto M. 2011; Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution. Genes Cells. 16:123–140. DOI: 10.1111/j.1365-2443.2010.01473.x. PMID: 21251164.
Article
7. Su C, Xia X, Shi Q, Song X, Fu J, Xiao C, Chen H, Lu B, Sun Z, Wu S, Yang S, Li X, Ye X, Song E, Song Y. 2015; Neohesperidin dihydrochalcone versus CCl4-induced hepatic injury through different mechanisms: the implication of free radical scavenging and Nrf2 activation. J Agric Food Chem. 63:5468–5475. DOI: 10.1021/acs.jafc.5b01750. PMID: 25978654.
8. Liu J, Sun H, Zhang A, Yan G, Han Y, Xue C, Zhou X, Shi H, Wang X. 2014; Serum pharmacochemistry combined with multiple data processing approach to screen the bioactive components and their metabolites in Mutan Cortex by ultra‐performance liquid chromatography tandem mass spectrometry. Biomed Chromatogr. 28:500–510. DOI: 10.1002/bmc.3060.
Article
9. Lin MY, Lee YR, Chiang SY, Li YZ, Chen YS, Hsu CD, Liu YW. 2013; Cortex Moutan induces bladder cancer cell death via apoptosis and retards tumor growth in mouse bladders. Evid Based Complement Alternat Med. 2013:207279. DOI: 10.1155/2013/207279. PMID: 24282433. PMCID: PMC3824643.
Article
10. Oh GS, Pae HO, Oh H, Hong SG, Kim IK, Chai KY, Yun YG, Kwon TO, Chung HT. 2001; In vitro anti-proliferative effect of 1,2,3,4,6-penta-O-galloyl-beta-D-glucose on human hepatocellular carcinoma cell line, SK-HEP-1 cells. Cancer Lett. 174:17–24. DOI: 10.1016/S0304-3835(01)00680-2. PMID: 11675148.
Article
11. Chen G, Zhang L, Zhu Y. 2006; Determination of glycosides and sugars in Moutan Cortex by capillary electrophoresis with electrochemical detection. J Pharm Biomed Anal. 41:129–134. DOI: 10.1016/j.jpba.2005.11.001. PMID: 16343840.
Article
12. Li J, Li Y, Pan S, Zhang L, He L, Niu Y. 2019; Paeonol attenuates ligation-induced periodontitis in rats by inhibiting osteoclastogenesis via regulating Nrf2/NF-κB/NFATc1 signaling pathway. Biochimie. 156:129–137. DOI: 10.1016/j.biochi.2018.09.004. PMID: 30213522.
Article
13. Li XY, Xu JD, Zhou SS, Kong M, Xu YY, Zou YT, Tang Y, Zhou L, Xu MZ, Xu J, Li SL. 2018; Time segment scanning-based quasi-multiple reaction monitoring mode by ultra-performance liquid chromatography coupled with quadrupole/time-of-flight mass spectrometry for quantitative determination of herbal medicines: Moutan Cortex, a case study. J Chromatogr A. 1581(1582):33–42. DOI: 10.1016/j.chroma.2018.10.047. PMID: 30389211.
Article
14. Feng RB, Wang Y, He C, Yang Y, Wan JB. 2018; Gallic acid, a natural polyphenol, protects against tert-butyl hydroperoxide- induced hepatotoxicity by activating ERK-Nrf2-Keap1-mediated antioxidative response. Food Chem Toxicol. 119:479–488. DOI: 10.1016/j.fct.2017.10.033. PMID: 29066411.
Article
15. Peng Z, Gong X, Yang Y, Huang L, Zhang Q, Zhang P, Wan R, Zhang B. 2017; Hepatoprotective effect of quercetin against LPS/d-GalN induced acute liver injury in mice by inhibiting the IKK/NF-κB and MAPK signal pathways. Int Immunopharmacol. 52:281–289. DOI: 10.1016/j.intimp.2017.09.022. PMID: 28963941.
16. Chen F, Mo K, Zhang Q, Fei S, Zu Y, Yang L. 2017; A novel approach for distillation of paeonol and simultaneous extraction of paeoniflorin by microwave irradiation using an ionic liquid solution as the reaction medium. Sep Purif Technol. 183:73–82. DOI: 10.1016/j.seppur.2017.03.069.
Article
17. Li YL, Li J, Wang NL, Yao XS. 2008; Flavonoids and a new polyacetylene from Bidens parviflora Willd. Molecules. 13:1931–1941. DOI: 10.3390/molecules13081931. PMID: 18794794. PMCID: PMC6244959.
Article
18. Li WH, Chang ST, Chang SC, Chang HT. 2008; Isolation of antibacterial diterpenoids from Cryptomeria japonica bark. Nat Prod Res. 22:1085–1093. DOI: 10.1080/14786410802267510. PMID: 18780250.
19. He L, She Z. 2017; Molecular structure identification and properties of gallic acid from galla chinensis. Chem Fiber Text Techol. 46:5–9.
20. Sun X, Wang P, Yao LP, Wang W, Gao YM, Zhang J, Fu YJ. 2018; Paeonol alleviated acute alcohol-induced liver injury via SIRT1/Nrf2/NF-κB signaling pathway. Environ Toxicol Pharmacol. 60:110–117. DOI: 10.1016/j.etap.2018.04.016. PMID: 29704732.
Article
21. Surh YJ. 2003; Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer. 3:768–780. DOI: 10.1038/nrc1189. PMID: 14570043.
Article
22. Pinheiro-Sant'ana HM, Guinazi M, Oliveira Dda S, Della Lucia CM, Reis Bde L, Brandão SC. 2011; Method for simultaneous analysis of eight vitamin E isomers in various foods by high performance liquid chromatography and fluorescence detection. J Chromatogr A. 1218:8496–8502. DOI: 10.1016/j.chroma.2011.09.067. PMID: 22014383.
23. Yen GC, Duh PD, Tsai HL. 2002; Antioxidant and pro-oxidant properties of ascorbic acid and gallic acid. Food Chem. 79:307–313. DOI: 10.1016/S0308-8146(02)00145-0.
Article
24. Ho HH, Chang CS, Ho WC, Liao SY, Wu CH, Wang CJ. 2010; Anti-metastasis effects of gallic acid on gastric cancer cells involves inhibition of NF-kappaB activity and downregulation of PI3K/AKT/small GTPase signals. Food Chem Toxicol. 48:2508–2516. DOI: 10.1016/j.fct.2010.06.024. PMID: 20600540.
25. Hsiang CY, Hseu YC, Chang YC, Kumar KJ, Ho TY, Yang HL. 2013; Toona sinensis and its major bioactive compound gallic acid inhibit LPS-induced inflammation in nuclear factor-κB transgenic mice as evaluated by in vivo bioluminescence imaging. Food Chem. 136:426–434. DOI: 10.1016/j.foodchem.2012.08.009. PMID: 23122080.
Article
26. Sies H, Berndt C, Jones DP. 2017; Oxidative stress. Annu Rev Biochem. 86:715–748. DOI: 10.1146/annurev-biochem-061516-045037. PMID: 28441057.
Article
27. Wei S, Chi J, Zhou M, Li R, Li Y, Luo J, Kong L. 2019; Anti-inflammatory lindenane sesquiterpeniods and dimers from Sarcandra glabra and its upregulating AKT/Nrf2/HO-1 signaling mechanism. Ind Crops Prod. 137:367–376. DOI: 10.1016/j.indcrop.2019.05.041.
Article
28. Chen B, Lu Y, Chen Y, Cheng J. 2015; The role of Nrf2 in oxidative stress-induced endothelial injuries. J Endocrinol. 225:R83–R99. DOI: 10.1530/JOE-14-0662. PMID: 25918130.
Article
29. Zhuang Y, Ma Q, Guo Y, Sun L. 2017; Protective effects of rambutan (Nephelium lappaceum) peel phenolics on H2O2-induced oxidative damages in HepG2 cells and D-galactose-induced aging mice. Food Chem Toxicol. 108(Pt B):554–562. DOI: 10.1016/j.fct.2017.01.022. PMID: 28137606.
30. Lyu Z, Ji X, Chen G, An B. 2019; Atractylodin ameliorates lipopolysaccharide and D-galactosamine-induced acute liver failure via the suppression of inflammation and oxidative stress. Int Immunopharmacol. 72:348–357. DOI: 10.1016/j.intimp.2019.04.005. PMID: 31030090.
Article
31. Auten RL, Davis JM. 2009; Oxygen toxicity and reactive oxygen species: the devil is in the details. Pediatr Res. 66:121–127. DOI: 10.1203/PDR.0b013e3181a9eafb. PMID: 19390491.
Article
32. Weber LW, Boll M, Stampfl A. 2003; Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol. 33:105–136. DOI: 10.1080/713611034. PMID: 12708612.
Article
33. Pham-Huy LA, He H, Pham-Huy C. 2008; Free radicals, antioxidants in disease and health. Int J Biomed Sci. 4:89–96. PMID: 23675073. PMCID: PMC3614697.
34. Johnston DE, Kroening C. 1998; Mechanism of early carbon tetrachloride toxicity in cultured rat hepatocytes. Pharmacol Toxicol. 83:231–239. DOI: 10.1111/j.1600-0773.1998.tb01475.x. PMID: 9868740.
Article
35. Vuda M, D'Souza R, Upadhya S, Kumar V, Rao N, Kumar V, Boillat C, Mungli P. 2012; Hepatoprotective and antioxidant activity of aqueous extract of Hybanthus enneaspermus against CCl4-induced liver injury in rats. Exp Toxicol Pathol. 64:855–859. DOI: 10.1016/j.etp.2011.03.006. PMID: 21478003.
36. Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, Yamamoto M, Nabeshima Y. 1997; An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 236:313–322. DOI: 10.1006/bbrc.1997.6943. PMID: 9240432.
Article
37. Chun KS, Kundu J, Kundu JK, Surh YJ. 2014; Targeting Nrf2-Keap1 signaling for chemoprevention of skin carcinogenesis with bioactive phytochemicals. Toxicol Lett. 229:73–84. DOI: 10.1016/j.toxlet.2014.05.018. PMID: 24875534.
Article
38. Keum YS. 2011; Regulation of the Keap1/Nrf2 system by chemopreventive sulforaphane: implications of posttranslational modifications. Ann N Y Acad Sci. 1229:184–189. DOI: 10.1111/j.1749-6632.2011.06092.x. PMID: 21793854.
Article
39. Lou Y, Guo Z, Zhu Y, Kong M, Zhang R, Lu L, Wu F, Liu Z, Wu J. 2019; Houttuynia cordata Thunb. and its bioactive compound 2-undecanone significantly suppress benzo(a)pyrene-induced lung tumorigenesis by activating the Nrf2-HO-1/NQO-1 signaling pathway. J Exp Clin Cancer Res. 38:242. DOI: 10.1186/s13046-019-1255-3. PMID: 31174565. PMCID: PMC6556055.
Article
40. Nguyen T, Nioi P, Pickett CB. 2009; The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem. 284:13291–13295. DOI: 10.1074/jbc.R900010200. PMID: 19182219. PMCID: PMC2679427.
Article
41. Calabrese V, Cornelius C, Dinkova-Kostova AT, Calabrese EJ, Mattson MP. 2010; Cellular stress responses, the hormesis paradigm, and vitagenes: novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid Redox Signal. 13:1763–1811. DOI: 10.1089/ars.2009.3074. PMID: 20446769. PMCID: PMC2966482.
Article
42. Bucolo C, Drago F, Maisto R, Romano GL, D'Agata V, Maugeri G, Giunta S. 2019; Curcumin prevents high glucose damage in retinal pigment epithelial cells through ERK1/2-mediated activation of the Nrf2/HO-1 pathway. J Cell Physiol. 234:17295–17304. DOI: 10.1002/jcp.28347. PMID: 30770549.
Article
43. Sharath Babu GR, Anand T, Ilaiyaraja N, Khanum F, Gopalan N. 2017; Pelargonidin modulates Keap1/Nrf2 pathway gene expression and ameliorates citrinin-induced oxidative stress in HepG2 cells. Front Pharmacol. 8:868. DOI: 10.3389/fphar.2017.00868. PMID: 29230174. PMCID: PMC5711834.
Article
44. Dinkova-Kostova AT, Talalay P. 2010; NAD(P)H:quinone acceptor oxidoreductase 1 (NQO1), a multifunctional antioxidant enzyme and exceptionally versatile cytoprotector. Arch Biochem Biophys. 501:116–123. DOI: 10.1016/j.abb.2010.03.019. PMID: 20361926. PMCID: PMC2930038.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr