684
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Ameliorative impacts of Rhodiola rosea against hepatic toxicity induced by monosodium glutamate: role of inflammation-, oxidative-stress-, and apoptosis-associated markers

ORCID Icon, &
Article: 2244749 | Received 27 Mar 2023, Accepted 01 Aug 2023, Published online: 01 Nov 2023

References

  • Risi R, Postic C, Watanabe M. Editorial: The liver as an endocrine organ: hepatokines and ketone bodies, novel hormones to be acknowledged. Front Endocrinol (Lausanne). 2023;13:1117773. doi:10.3389/fendo.2022.1117773
  • Halliwell B. Free radicals, antioxidants, and human disease: curiosity, cause, or consequence? Lancet (British Edition). 1994;344(8924):721–724. doi:10.1016/s0140-6736(94)92211-x
  • Samarghandian S, et al. Crocus sativus L. (saffron) stigma aqueous extract induces apoptosis in alveolar human lung cancer cells through caspase-dependent pathways activation. BioMed Res Int. 2013. doi:10.1155/2020/3516010
  • Chakraborty SP. Patho-physiological and toxicological aspects of monosodium glutamate. Toxicol Mech Methods. 2019;29(6):389–396. doi:10.1080/15376516.2018.1528649
  • Garattini S. Glutamic acid, twenty years later. J Nutr. 2000;130(4S Suppl):901s–909s. doi:10.1093/jn/130.4.901S
  • Zhou Y, Danbolt NC. Glutamate as a neurotransmitter in the healthy brain. J Neural Transm. 2014;121(8):799–817. doi:10.1007/s00702-014-1180-8
  • Sharma A. Monosodium glutamate-induced oxidative kidney damage and possible mechanisms: a mini-review. J Biomed Sci. 2015;22:93. doi:10.1186/s12929-015-0192-5
  • Pu W-L, Zhang M-Y, Bai R-Y, et al. Anti-inflammatory effects of Rhodiola rosea L.: a review. Biomed Pharmacother. 2020;121:109552. doi:10.1016/j.biopha.2019.109552
  • Marchev AS, Dinkova-Kostova AT, György Z, et al. Rhodiola rosea L.: from golden root to green cell factories. Phytochem Rev. 2016;15(4):515–536. doi:10.1007/s11101-016-9453-5
  • Ganzera M, Yayla Y, Khan IA. Analysis of the marker compounds of Rhodiola rosea L. (golden root) by reversed phase high performance liquid chromatography. Chem Pharm Bull. 2001;49(4):465–467. doi:10.1248/cpb.49.465
  • Yoshikawa M, Shimada H, Shimoda H, et al. Bioactive constituents of Chinese natural medicines. II. Rhodiolae Radix. (1). Chemical structures and antiallergic activity of Rhodiocyanosides A and B from the underground part of Rhodiola quadrifida (PALL.) FISCH. et MEY. (Crassulaceae). Chem Pharm Bull. 1996;44(11):2086–2091. doi:10.1248/cpb.44.2086
  • Zhu L, Wei T, Gao J, et al. The cardioprotective effect of salidroside against myocardial ischemia reperfusion injury in rats by inhibiting apoptosis and inflammation. Apoptosis. 2015;20(11):1433–1443. doi:10.1007/s10495-015-1174-5
  • Nabavi SF, Braidy N, Orhan IE, et al. Rhodiola rosea L. and Alzheimer's disease: from farm to pharmacy. Phytother Res. 2016;30(4):532–539. doi:10.1002/ptr.5569
  • Senthilkumar R, Chandran R, Parimelazhagan T. Hepatoprotective effect of Rhodiola imbricata rhizome against paracetamol-induced liver toxicity in rats. Saudi J Biol Sci. 2014;21(5):409–416. doi:10.1016/j.sjbs.2014.04.001
  • Farombi EO, Onyema OO. Monosodium glutamate-induced oxidative damage and genotoxicity in the rat: modulatory role of vitamin C, vitamin E and quercetin. Hum Exp Toxicol. 2006;25(5):251–259. doi:10.1191/0960327106ht621oa
  • Bhatia A, Saikia PP, Dkhar B, et al. Anesthesia protocol for ear surgery in Wistar rats (animal research). Anim Models Exp Med. 2022;5(2):183–188. doi:10.1002/ame2.12198
  • Lowry OH, Rosebrough NJ, Farr AL, et al. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193(1):265–275. doi:10.1016/S0021-9258(19)52451-6
  • Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem. 1979;95(2):351–358. doi:10.1016/0003-2697(79)90738-3
  • Nishikimi M, Appaji N, Yagi K. The occurrence of superoxide anion in the reaction of reduced phenazine methosulfate and molecular oxygen. Biochem Biophys Res Commun. 1972;46(2):849–854. doi:10.1016/S0006-291X(72)80218-3
  • Beutler E, Duron O, Kelly BM. Improved method for the determination of blood glutathione. J Lab Clin Med. 1963;61:882–888. https://pubmed.ncbi.nlm.nih.gov/13967893/
  • Hadwan MH, Ali SK. New spectrophotometric assay for assessments of catalase activity in biological samples. Anal Biochem. 2018;542:29–33. doi:10.1016/j.ab.2017.11.013
  • Soliman MM, Attia HF, El-Ella GA. Genetic and histopathological alterations induced by cypermethrin in rat kidney and liver: protection by sesame oil. Int J Immunopathol Pharmacol. 2015;28(4):508–520. doi:10.1177/0394632015575950
  • Slaoui M, Fiette L. Methods in molecular biology. Methods Mol Biol. 2011;691:69–82. doi:10.1007/978-1-60761-849-2_4
  • Songserm T, Engel B, van Roozelaar DJ, et al. Cellular immune response in the small intestine of two broiler chicken lines orally inoculated with malabsorption syndrome homogenates. Vet Immunol Immunopathol. 2002;85(1-2):51–62. doi:10.1016/S0165-2427(01)00411-1
  • Shi Z, Taylor AW, Yuan B, et al. Monosodium glutamate intake is inversely related to the risk of hyperglycemia. Clin Nutr. 2014;33(5):823–828. doi:10.1016/j.clnu.2013.10.018
  • Inetianbor J, Yakubu JM, Stephen E. Effects of food additives and preservatives on man-a review. 2015;6:18. https://www.researchgate.net/publication/277714512
  • DelRaso N, et al. Cadmium uptake kinetics in rat hepatocytes: correction for albumin binding. 2003;72(1):19–30. doi:10.1093/toxsci/kfg009
  • Koyu A, et al. Evaluation of the effects of cadmium on rat liver. 2006;284:81–85. doi:10.1007/s11010-005-9017-2
  • Noor KK, Ijaz MU, Ehsan N, et al. Hepatoprotective role of vitexin against cadmium-induced liver damage in male rats: a biochemical, inflammatory, apoptotic and histopathological investigation. Biomed Pharmacother. 2022;150:112934. doi:10.1016/j.biopha.2022.112934
  • Jomova K, Valko MJT. Advances in metal-induced oxidative stress and human disease. 2011;283(2-3):65–87. doi:10.1016/j.tox.2011.03.001
  • Begic A, et al. Disulfiram moderately restores impaired hepatic redox status of rats subchronically exposed to cadmium. 2017;32(1):478–489. doi:10.1080/14756366.2016.1261132
  • Ortiz G, Bitzer-Quintero OK, Zárate CB, et al. Monosodium glutamate-induced damage in liver and kidney: a morphological and biochemical approach. Biomed Pharmacother. 2006;60(2):86–91. doi:10.1016/j.biopha.2005.07.012
  • Shukry M, El-Shehawi AM, El-Kholy WM, et al. Ameliorative effect of graviola (Annona muricata) on mono sodium glutamate-induced hepatic injury in rats: antioxidant, apoptotic, anti-inflammatory, lipogenesis markers, and histopathological studies. Animals (Basel). 2020;10(11):1996. doi:10.3390/ani10111996
  • Tawfik MS, Al-Badr N. Adverse effects of monosodium glutamate on liver and kidney functions in adult rats and potential protective effect of vitamins C and E. Food Nutr Sci. 2012;03(05):651. doi:10.4236/fns.2012.35089
  •  Ijaz MU, Anwar H, Iqbal S, et al. Protective effect of myricetin on nonylphenol-induced testicular toxicity: biochemical, steroidogenic, hormonal, spermatogenic, and histological-based evidences. Environ Sci Pollut Res. 2021;28(18):22742–22757. doi:10.1007/s11356-020-12296-5.
  • Hassan ZA, et al. The effects of monosodium glutamate on thymic and splenic immune functions and role of recovery (biochemical and histological study). J Cytol Histol. 2014;5(6):1. doi:10.4172/2157-7099.1000283
  • Ibrahim MA, et al. Amelioration of monosodium glutamate-induced hepatotoxicity by vitamin C. Eur J Sci Res. 2011;60(1):159–165. https://www.researchgate.net/publication/289794360. http://www.eurojournals.com/ejsr.htm
  • Sharma A, Wongkham C, Prasongwattana V, et al. Proteomic analysis of kidney in rats chronically exposed to monosodium glutamate. PloS one. 2014;9(12):e116233. doi:10.1371/journal.pone.0116233
  • Calis IU, et al. The effects of monosodium glutamate and tannic acid on adult rats. Iran Red Crescent Med J. 2016;18(10). doi:10.5812/ircmj.37912
  • Singh K, Ahluwalia P. Effect of monosodium glutamate on lipid peroxidation and certain antioxidant enzymes in cardiac tissue of alcoholic adult male mice. J Cardiovasc Dis Res. 2012;3(1):12–18. doi:10.4103/0975-3583.91595
  • Ahmadiasl N, et al. The anti-inflammatory effect of erythropoietin and melatonin on renal ischemia reperfusion injury in male rats. Adv Pharm Bull. 2014;4(1):49. doi:10.5681/apb.2014.008
  • Weinstein DM, Mihm MJ, Bauer JA. Cardiac peroxynitrite formation and left ventricular dysfunction following doxorubicin treatment in mice. J Pharmacol Exp Ther. 2000;294(1):396–401. https://pubmed.ncbi.nlm.nih.gov/10871338/
  • Yilmaz HR, Uz E, Yucel N, et al. Protective effect of caffeic acid phenethyl ester (CAPE) on lipid peroxidation and antioxidant enzymes in diabetic rat liver. J Biochem Mol Toxicol. 2004;18(4):234–238. doi:10.1002/jbt.20028
  • Kim SH, Hyun SH, Choung SY. Antioxidative effects of Cinnamomi cassiae and Rhodiola rosea extracts in liver of diabetic mice. Biofactors. 2006;26(3):209–219. doi:10.1002/biof.5520260306
  • Manonmani G, Bhavapriya V, Kalpana S, et al. Antioxidant activity of Cassia fistula (Linn.) flowers in alloxan induced diabetic rats. J Ethnopharmacol. 2005;97(1):39–42. doi:10.1016/j.jep.2004.09.051
  • Drayton DL, Liao S, Mounzer RH, et al. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol. 2006;7(4):344–353. doi:10.1038/ni1330
  • Wang Q, Kuang H, Su Y, et al. Naturally derived anti-inflammatory compounds from Chinese medicinal plants. J Ethnopharmacol. 2013;146(1):9–39. doi:10.1016/j.jep.2012.12.013
  • Jing W, Jin R-G, Xiao L, et al. Anti-asthma effects of synthetic salidroside through regulation of Th1/Th2 balance. Chin J Nat Med. 2014;12(7):500–504. doi:10.1016/S1875-5364(14)60078-9
  • Furuya DT, Poletto AC, Favaro RR, et al. Anti-inflammatory effect of atorvastatin ameliorates insulin resistance in monosodium glutamate–treated obese mice. Metabolism. 2010;59(3):395–399. doi:10.1016/j.metabol.2009.08.011
  • Eid RA, Al-Shraim M, Zaki MS, et al. Vitamin E protects against monosodium glutamate-induced acute liver injury and hepatocyte ultrastructural alterations in rats. Ultrastruct Pathol. 2019;43(4-5):199–208. doi:10.1080/01913123.2019.1673860
  • Falalyeyeva T, et al. Probiotic strains of lactobacilli and bifidobacteria alter pro-and anti-inflammatory cytokines production in rats with monosodium glutamate-induced obesity. Receptor. 2017;4:2. doi:10.15407/fz63.01.017
  • Guan S, Song B, Song Y, et al. Protective effects of salidroside from Rhodiola rosea on LPS-induced acute lung injury in mice. Immunopharmacol Immunotoxicol. 2012;34(4):667–672. doi:10.3109/08923973.2011.650175
  • Schriner SE, Avanesian A, Liu Y, et al. Protection of human cultured cells against oxidative stress by Rhodiola rosea without activation of antioxidant defenses. Free Radical Biol Med. 2009;47(5):577–584. doi:10.1016/j.freeradbiomed.2009.05.025
  • Falalyeyeva TM, Leschenko IV, Beregova TV, et al. Neuroprotective effects of Rhodiola sacra on transient global cerebral ischemia through activating AMPK/Nrf2 pathway in rats. Antioxid Redox Signaling. 2022;36(7-9):567–591. doi:10.1089/ars.2020.8224
  • Yan Z-Q, Chen J, Xing G-X, et al. Salidroside prevents cognitive impairment induced by chronic cerebral hypoperfusion in rats. J Int Med Res. 2015;43(3):402–411. doi:10.1177/0300060514566648
  • Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler, Thromb, Vasc Biol. 2011;31(5):986–1000. doi:10.1161/ATVBAHA.110.207449
  • Jiang J, Dai J, Cui H. Vitexin reverses the autophagy dysfunction to attenuate MCAO-induced cerebral ischemic stroke via mTOR/Ulk1 pathway. Biomed Pharmacother. 2018;99:583–590. doi:10.1016/j.biopha.2018.01.067
  • Dooley S, ten Dijke P. TGF-β in progression of liver disease. Cell Tissue Res. 2012;347(1):245–256. doi:10.1007/s00441-011-1246-y
  • Yi J-H, Park S, Kapadia R, et al. Role of transcription factors in mediating post-ischemic cerebral inflammation and brain damage. Neurochem Int. 2007;50(7-8):1014–1027. doi:10.1016/j.neuint.2007.04.019