Browse > Article
http://dx.doi.org/10.4196/kjpp.2021.25.4.321

Protective effects of lutein against vancomycin-induced acute renal injury in mice via upregulation of peroxisome proliferator-activated receptor gamma/nuclear factor erythroid 2-related factor 2 and inhibition nuclear factor-kappaB/caspase 3  

Emeka, Promise M. (Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University)
Rasool, Sahibzada T. (Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University)
Morsy, Mohamed A. (Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University)
Islam, Mohamed I. Hairul (Department of Biological Sciences, College of Science, King Faisal University)
Chohan, Muhammad S. (Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University)
Publication Information
The Korean Journal of Physiology and Pharmacology / v.25, no.4, 2021 , pp. 321-331 More about this Journal
Abstract
Vancomycin, an antibiotic used occasionally as a last line of treatment for methicillin-resistant Staphylococcus aureus, is reportedly associated with nephrotoxicity. This study aimed at evaluating the protective effects of lutein against vancomycin-induced acute renal injury. Peroxisome proliferator-activated receptor gamma (PPARγ) and its associated role in renoprotection by lutein was also examined. Male BALB/c mice were divided into six treatment groups: control with normal saline, lutein (200 mg/kg), vancomycin (250 mg/kg), vancomycin (500 mg/kg), vancomycin (250 mg/kg) with lutein, and vancomycin (500 mg/kg) with lutein groups; they were euthanized after 7 days of treatment. Thereafter, samples of blood, urine, and kidney tissue of the mice were analyzed, followed by the determination of levels of N-acetyl-β-D-glucosaminidase (NAG) in the urine, renal creatine kinase; protein carbonyl, malondialdehyde, and caspase-3 in the kidney; and the expression of PPARγ, nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor-kappaB (NF-κB) in renal tissue. Results showed that the levels of protein carbonyl and malondialdehyde, and the activity of NAG, creatine kinase and caspase-3, were significantly increased in the vancomycin-treatment groups. Moreover, the levels of Nrf2 significantly decreased, while NF-κB expression increased. Lutein ameliorated these effects, and significantly increased PPARγ expression. Furthermore, it attenuated vancomycin-induced histological alterations such as, tissue necrosis and hypertrophy. Therefore, we conclude that lutein protects against vancomycin-induced renal injury by potentially upregulating PPARγ/Nrf2 expression in the renal tissues, and consequently downregulating the pathways: inflammation by NF-κB and apoptosis by caspase-3.
Keywords
Acute renal injury; Caspase-3; Lutein; PPAR gamma; Vancomycin;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Hughes BP. A method for the estimation of serum creatine kinase and its use in comparing creatine kinase and aldolase activity in normal and pathological sera. Clin Chim Acta. 1962;7:597-603.   DOI
2 Velmurugan G, Venkatesh Babu DD, Ramasamy S. Prolonged monocrotophos intake induces cardiac oxidative stress and myocardial damage in rats. Toxicology. 2013;307:103-108.   DOI
3 Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem. 1979;95:351-358.   DOI
4 Rojas-Franco P, Franco-Colin M, Camargo MEM, Carmona MME, Ortiz-Butron M del R E, Blas-Valdivia V, Cano-Europa E. Phycobiliproteins and phycocyanin of Arthrospira maxima (Spirulina) reduce apoptosis promoters and glomerular dysfunction in mercury-related acute kidney injury. Toxicol Res Appl. 2018;2:1-10.
5 Ahn SY, Cho CH, Park KG, Lee HJ, Lee S, Park SK, Lee IK, Koh GY. Tumor necrosis factor-alpha induces fractalkine expression preferentially in arterial endothelial cells and mithramycin A suppresses TNF-alpha-induced fractalkine expression. Am J Pathol. 2004;164:1663-1672.   DOI
6 Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods. 2001;25:402-408.   DOI
7 Ouyang B, Li Z, Ji X, Huang J, Zhang H, Jiang C. The protective role of lutein on isoproterenol-induced cardiac failure rat model through improving cardiac morphology, antioxidant status via positively regulating Nrf2/HO-1 signalling pathway. Pharm Biol. 2019;57:529-535.   DOI
8 Cheng F, Zhang Q, Yan FF, Wan JF, Lin CS. Lutein protects against ischemia/reperfusion injury in rat skeletal muscle by modulating oxidative stress and inflammation. Immunopharmacol Immunotoxicol. 2015;37:329-334.   DOI
9 Buonocore D, Rucci S, Vandoni M, Negro M, Marzatico F. Oxidative system in aged skeletal muscle. Muscles Ligaments Tendons J. 2012;1:85-90.
10 Havasi A, Borkan SC. Apoptosis and acute kidney injury. Kidney Int. 2011;80:29-40.   DOI
11 Yang B, El Nahas AM, Thomas GL, Haylor JL, Watson PF, Wagner B, Johnson TS. Caspase-3 and apoptosis in experimental chronic renal scarring. Kidney Int. 2001;60:1765-1776.   DOI
12 Nataraj J, Manivasagam T, Thenmozhi AJ, Essa MM. Lutein protects dopaminergic neurons against MPTP-induced apoptotic death and motor dysfunction by ameliorating mitochondrial disruption and oxidative stress. Nutr Neurosci. 2016;19:237-246.   DOI
13 Fung FK, Law BY, Lo AC. Lutein attenuates both apoptosis and autophagy upon cobalt (II) chloride-induced hypoxia in rat Muller cells. PLoS One. 2016;11:e0167828.   DOI
14 Yamashita M, Yoshida T, Suzuki S, Homma K, Hayashi M. Podocyte-specific NF-κB inhibition ameliorates proteinuria in adriamycin-induced nephropathy in mice. Clin Exp Nephrol. 2017;21:16-26.   DOI
15 Xiao Z, Shan J, Li C, Luo L, Lu J, Li S, Long D, Li Y. Mechanisms of cyclosporine-induced renal cell apoptosis: a systematic review. Am J Nephrol. 2013;37:30-40.   DOI
16 Jung ME, Gatch MB, Simpkins JW. Estrogen neuroprotection against the neurotoxic effects of ethanol withdrawal: potential mechanisms. Exp Biol Med (Maywood). 2005;230:8-22.   DOI
17 Buhrmann C, Mobasheri A, Busch F, Aldinger C, Stahlmann R, Montaseri A, Shakibaei M. Curcumin modulates nuclear factor kappaB (NF-kappaB)-mediated inflammation in human tenocytes in vitro: role of the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem. 2011;286:28556-28566.   DOI
18 Kvandova M, Barancik M, Balis P, Puzserova A, Majzunova M, Dovinova I. The peroxisome proliferator-activated receptor gamma agonist pioglitazone improves nitric oxide availability, renin-angiotensin system and aberrant redox regulation in the kidney of prehypertensive rats. J Physiol Pharmacol. 2018;69:231-243.
19 Corrales P, Izquierdo-Lahuerta A, Medina-Gomez G. Maintenance of kidney metabolic homeostasis by PPAR gamma. Int J Mol Sci. 2018;19:2063.   DOI
20 Zhang YF, Wang Q, Su YY, Wang JL, Hua BJ, Yang S, Feng JX, Li HY. PPAR-γ agonist rosiglitazone protects rat peritoneal mesothelial cells against peritoneal dialysis solution-induced damage. Mol Med Rep. 2017;15:1786-1792.   DOI
21 Cho HY, Gladwell W, Wang X, Chorley B, Bell D, Reddy SP, Kleeberger SR. Nrf2-regulated PPARγ expression is critical to protection against acute lung injury in mice. Am J Respir Crit Care Med. 2010;182:170-182.   DOI
22 Lee C. Collaborative power of Nrf2 and PPARγ activators against metabolic and drug-induced oxidative injury. Oxid Med Cell Longev. 2017;2017:1378175.
23 Mahmoud AM, Germoush MO, Alotaibi MF, Hussein OE. Possible involvement of Nrf2 and PPARγ up-regulation in the protective effect of umbelliferone against cyclophosphamide-induced hepatotoxicity. Biomed Pharmacother. 2017;86:297-306.   DOI
24 Jorgensen SCJ, Murray KP, Lagnf AM, Melvin S, Bhatia S, Shamim MD, Smith JR, Brade KD, Simon SP, Nagel J, Williams KS, Ortwine JK, Veve MP, Truong J, Huang DB, Davis SL, Rybak MJ. A multicenter evaluation of vancomycin-associated acute kidney injury in hospitalized patients with acute bacterial skin and skin structure infections. Infect Dis Ther. 2020;9:89-106.   DOI
25 Garcia-Rojas P, Antaramian A, Gonzalez-Davalos L, Villarroya F, Shimada A, Varela-Echavarria A, Mora O. Induction of peroxisomal proliferator-activated receptor gamma and peroxisomal proliferator-activated receptor gamma coactivator 1 by unsaturated fatty acids, retinoic acid, and carotenoids in preadipocytes obtained from bovine white adipose tissue1,2. J Anim Sci. 2010;88:1801-1808.   DOI
26 Dieterich C, Puey A, Lin S, Swezey R, Furimsky A, Fairchild D, Mirsalis JC, Ng HH. Gene expression analysis reveals new possible mechanisms of vancomycin-induced nephrotoxicity and identifies gene markers candidates. Toxicol Sci. 2009;107:258-269.   DOI
27 Lodise TP, Lomaestro B, Graves J, Drusano GL. Larger vancomycin doses (at least four grams per day) are associated with an increased incidence of nephrotoxicity. Antimicrob Agents Chemother. 2008;52:1330-1336.   DOI
28 Makarov SS. NF-kappaB as a therapeutic target in chronic inflammation: recent advances. Mol Med Today. 2000;6:441-448.   DOI
29 Selvaraj RK, Shanmugasundaram R, Klasing KC. Effects of dietary lutein and PUFA on PPAR and RXR isomer expression in chickens during an inflammatory response. Comp Biochem Physiol A Mol Integr Physiol. 2010;157:198-203.   DOI
30 Jin XH, Ohgami K, Shiratori K, Suzuki Y, Hirano T, Koyama Y, Yoshida K, Ilieva I, Iseki K, Ohno S. Inhibitory effects of lutein on endotoxin-induced uveitis in Lewis rats. Invest Ophthalmol Vis Sci. 2006;47:2562-2568.   DOI
31 Jesse CR, Bortolatto CF, Wilhelm EA, Roman SS, Prigol M, Nogueira CW. The peroxisome proliferator-activated receptor-γ agonist pioglitazone protects against cisplatin-induced renal damage in mice. J Appl Toxicol. 2014;34:25-32.   DOI
32 Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1:a001651.   DOI
33 Qin X, Tsoi MF, Zhao X, Zhang L, Qi Z, Cheung BMY. Vancomycin-associated acute kidney injury in Hong Kong in 2012-2016. BMC Nephrol. 2020;21:41.   DOI
34 Rybak MJ, Le J, Lodise TP, Levine DP, Bradley JS, Liu C, Mueller BA, Pai MP, Wong-Beringer A, Rotschafer JC, Rodvold KA, Maples HD, Lomaestro BM. Therapeutic monitoring of vancomycin for serious methicillin-resistant Staphylococcus aureus infections: a revised consensus guideline and review of the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists. Pharmacotherapy. 2020;40:363-367.   DOI
35 Yoshiyama Y, Yazaki T, Wong PC, Beauchamp D, Kanke M. The effect of fosfomycin on glycopeptide antibiotic-induced nephrotoxicity in rats. J Infect Chemother. 2001;7:243-246.   DOI
36 Diep QN, Schiffrin EL. Increased expression of peroxisome proliferator-activated receptor-alpha and -gamma in blood vessels of spontaneously hypertensive rats. Hypertension. 2001;38:249-254.   DOI
37 Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci. 2014;39:199-218.   DOI
38 Ibrahim MA, El-Sheikh AA, Khalaf HM, Abdelrahman AM. Protective effect of peroxisome proliferator activator receptor (PPAR)-α and -γ ligands against methotrexate-induced nephrotoxicity. Immunopharmacol Immunotoxicol. 2014;36:130-137.   DOI
39 Rafi MM, Kanakasabai S, Gokarn SV, Krueger EG, Bright JJ. Dietary lutein modulates growth and survival genes in prostate cancer cells. J Med Food. 2015;18:173-181.   DOI
40 Rybak M, Lomaestro B, Rotschafer JC, Moellering R Jr, Craig W, Billeter M, Dalovisio JR, Levine DP. Therapeutic monitoring of vancomycin in adult patients: a consensus review of the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists. Am J Health Syst Pharm. 2009;66:82-98.   DOI
41 Nishino Y, Takemura S, Minamiyama Y, Hirohashi K, Ogino T, Inoue M, Okada S, Kinoshita H. Targeting superoxide dismutase to renal proximal tubule cells attenuates vancomycin-induced nephrotoxicity in rats. Free Radic Res. 2003;37:373-379.   DOI
42 Suntres ZE. Liposomal antioxidants for protection against oxidant-induced damage. J Toxicol. 2011;2011:152474.   DOI
43 Stanton RC. Oxidative stress and diabetic kidney disease. Curr Diab Rep. 2011;11:330-336.   DOI
44 Ratliff BB, Abdulmahdi W, Pawar R, Wolin MS. Oxidant mechanisms in renal injury and disease. Antioxid Redox Signal. 2016;25:119-146.   DOI
45 Liu ZG, Qi ZC, Liu WL, Wang WZ. Lutein protects against ischemia/reperfusion injury in rat kidneys. Mol Med Rep. 2015;11:2179-2184.   DOI
46 Kim DH, Kwack SJ, Yoon KS, Choi JS, Lee BM. 4-hydroxynonenal: a superior oxidative biomarker compared to malondialdehyde and carbonyl content induced by carbon tetrachloride in rats. J Toxicol Environ Health A. 2015;78:1051-1062.   DOI
47 Korbecki J, Bobinski R, Dutka M. Self-regulation of the inflammatory response by peroxisome proliferator-activated receptors. Inflamm Res. 2019;68:443-458.   DOI
48 Nair SV, Ghanam K, Deshpande J, Juturu V. Lutein and zeaxanthin isomers induces mitochondrial biogenesis and improves endurance capacity in muscle cells. EC Ophthalmol. 2018;9:658-668.
49 Kim JE, Clark RM, Park Y, Lee J, Fernandez ML. Lutein decreases oxidative stress and inflammation in liver and eyes of guinea pigs fed a hypercholesterolemic diet. Nutr Res Pract. 2012;6:113-119.   DOI
50 Ma Y, Shi M, Wang Y, Liu J. PPARγ and its agonists in chronic kidney disease. Int J Nephrol. 2020;2020:2917474.
51 Selvaraj RK, Klasing KC. Lutein and eicosapentaenoic acid interact to modify iNOS mRNA levels through the PPARgamma/RXR pathway in chickens and HD11 cell lines. J Nutr. 2006;136:1610-1616.   DOI
52 Vasudeva V, Tenkanidiyoor YS, Peter AJ, Shetty J, Lakshman SP, Fernandes R, Patali KA. Radioprotective efficacy of lutein in ameliorating electron beam radiation-induced oxidative injury in Swiss albino mice. Iran J Med Sci. 2018;43:41-51.
53 Zhang B, Lo C, Shen L, Sood R, Jones C, Cusmano-Ozog K, ParkSnyder S, Wong W, Jeng M, Cowan T, Engleman EG, Zehnder JL. The role of vanin-1 and oxidative stress-related pathways in distinguishing acute and chronic pediatric ITP. Blood. 2011;117:4569-4579.   DOI
54 Qu S, Dai C, Lang F, Hu L, Tang Q, Wang H, Zhang Y, Hao Z. Rutin attenuates vancomycin-induced nephrotoxicity by ameliorating oxidative stress, apoptosis, and inflammation in rats. Antimicrob Agents Chemother. 2018;63:e01545-e01518.
55 Nakamura T, Hashimoto Y, Kokuryo T, Inui KI. Effects of fosfomycin and imipenem/cilastatin on nephrotoxicity and renal excretion of vancomycin in rats. Pharm Res. 1998;15:734-738.   DOI
56 Horak E, Hopfer SM, Sunderman FW Jr. Spectrophotometric assay for urinary N-acetyl-beta-D-glucosaminidase activity. Clin Chem. 1981;27:1180-1185.   DOI