AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.7 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Identification of egg protein-derived peptides as xanthine oxidase inhibitors: virtual hydrolysis, molecular docking, and in vitro activity evaluation

Zhipeng YuaYaxin CaobRuotong KanbHuizhuo JibWenzhu Zhaoa( )Sijia WucJingbo LiucDavid Shiuand
School of Food Science and Engineering, Hainan University, Haikou 570228, China
College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
Lab of Nutrition and Functional Food, Jilin University, Changchun 130062, China
Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Show Author Information

Abstract

The purpose of this study was to screen the xanthine oxidase (XO) inhibitory peptides from egg white proteins through virtual hydrolysis, in vitro activity validation, and molecular docking. The results demonstrated that tripeptide EEK from ovalbumin exhibited potent XO inhibitory activity with an IC50 value of 141 µmol/L. The molecular docking results showed that tripeptide EEK bound with the active center of XO via 3 carbon hydrogen bond interactions, 2 salt bridges, 5 conventional hydrogen bond interactions, and 4 attractive charge interactions. The residues Glu802, Phe1009, and Arg880 may play key roles in the XO catalytic reaction. Especially, the key intermolecular forces of inhibiting XO activity may be special type of hydrogen bonds including carbon hydrogen bond interactions and attraction charge interactions. The novel tripeptide EEK is potential candidates for controlling hyperuricemia.

References

[1]

R.J. Johnson, S. Titte, J.R. Cade, et al., Uric acid, evolution and primitive cultures, Semin. Nephrol. 25(1) (2005) 3-8. https://doi.org/10.1016/j.semnephrol.2004.09.002.

[2]

T.R. Merriman, N. Dalbeth, The genetic basis of hyperuricaemia and gout, Joint Bone Spine 78(1) (2011) 35-40. https://doi.org/10.1016/j.jbspin.2010.02.027.

[3]

E. Suresh, Diagnosis and management of gout: a rational approach, Postgrad. Med. J. 81(959) (2005) 572-579. https://doi.org/10.1136/pgmj.2004.030692.

[4]

D. Conen, V. Wietlisbach, P. Bovet, et al., Prevalence of hyperuricemia and relation of serum uric acid with cardiovascular risk factors in a developing country, BMC Public Health 4(1) (2004) 1-9. https://doi.org/10.1186/1471-2458-4-9.

[5]

D. Khanna, J.D. Fitzgerald, P.P. Khanna, et al., 2012 American College of Rheumatology Guidelines for management of gout. Part 1: systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricemia, Arthritis Care Res. 64(10) (2012) 1431-1446. https://doi.org/10.1002/acr.21772.

[6]

R.P. Obermayr, C. Temml, G. Gutjahr, et al., Elevated uric acid increases the risk for kidney disease, J. Am. Soc. Nephrol. 19(12) (2008) 2407-2413. https://doi.org/10.1681/ASN.2008010080.

[7]

Y. Li, X. Kang, Q. Li, et al, Anti-hyperuricemic peptides derived from bonito hydrolysates based on in vivo hyperuricemic model and in vitro xanthine oxidase inhibitory activity, Peptides 107 (2018) 45-53. https://doi.org/10.1016/j.peptides.2018.08.001.

[8]

V. Lavelli, C. Peri, A. Rizzolo, Antioxidant activity of tomato products as studied by model reactions using xanthine oxidase, myeloperoxidase, and copper-induced lipid peroxidation, J. Agr. Food Chem. 48(5) (2000) 1442-1448. https://doi.org/10.1021/jf990782j.

[9]

J.K. Lin, P.C. Chen, C.T. Ho, et al, Inhibition of xanthine oxidase and suppression of intracellular reactive oxygen species in HL-60 cells by theaflavin-3, 3'-digallate,(−)-epigallocatechin-3-gallate, and propyl gallate, J. Agr. Food Chem. 48(7) (2000) 2736-2743. https://doi.org/10.1021/jf000066d.

[10]

C.M. Burns, R.L. Wortmann, Gout therapeutics: new drugs for an old disease, The Lancet 377(9760) (2011) 165-177. https://doi.org/10.1016/S0140-6736(10)60665-4.

[11]

Q. Li, X. Kang, C. Shi, et al., Moderation of hyperuricemia in rats via consuming walnut protein hydrolysate diet and identification of new antihyperuricemic peptides, Food Funct. 9(1) (2018) 107-116. https://doi.org/10.1039/c7fo01174a.

[12]

N. Dalbeth, T. Bardin, M. Doherty, et al., Discordant American College of Physicians and international rheumatology guidelines for gout management: consensus statement of the Gout, Hyperuricemia and Crystal-Associated Disease Network (G-CAN), Nat. Rev. Rheumatol. 13(9) (2017) 561-568. https://doi.org/10.1038/nrrheum.2017.126.

[13]

S. Jarjour, M. Barrette, V. Normand, et al., Genetic markers associated with cutaneous adverse drug reactions to allopurinol: a systematic review, Pharmacogenomics 16(7) (2015) 755-767. https://doi.org/10.2217/pgs.15.21.

[14]

Z.J. Li, C.H. Cho, Development of peptides as potential drugs for cancer therapy, Curr. Pharm. Design 16(10) (2010) 1180-1189. https://doi.org/10.2174/138161210790945913.

[15]

I. Murota, S. Taguchi, N. Sato, et al., Identification of antihyperuricemic peptides in the proteolytic digest of shark cartilage water extract using in vivo activity-guided fractionation, J. Agric. Food Chem. 62(11) (2014) 2392-2397. https://doi.org/10.1021/jf405504u.

[16]

Z. Yu, R. Kan, S. Wu, et al., Xanthine oxidase inhibitory peptides derived from tuna protein: virtual screening, inhibitory activity, and molecular mechanisms, J. Sci. Food Agric. 101(4) (2021) 1349-1354. https://doi.org/10.1002/jsfa.10745.

[17]

H. Ji, W. Zhao, Z. Yu, et al., Mechanism of interactions between egg protein-derived tri-peptides and cellular membrane by molecular dynamic simulation and isothermal titration calorimetry, Int. J. Food. Sci. Tech. (2022). https://doi.org/10.1111/ijfs.15736.

[18]

K.S. Bhullar, J. Wu, Dietary peptides in aging: evidence and prospects, Food Sci. Hum. Well. 9(1) (2020) 1-7. https://doi.org/10.1016/j.fshw.2020.01.001.

[19]

A. Dávalos, M. Miguel, B. Bartolome, et al., Antioxidant activity of peptides derived from egg white proteins by enzymatic hydrolysis, J. Food Protect. 67(9) (2004) 1939-1944. https://doi.org/10.4315/0362-028x-67.9.1939.

[20]

Y. Mine, F. Ma, S. Lauriau, Antimicrobial peptides released by enzymatic hydrolysis of hen egg white lysozyme, J. Agr. Food Chem. 52(5) (2004) 1088-1094. https://doi.org/10.1021/jf0345752.

[21]

Z. Yu, S. Wu, W. Zhao, et al., Biological evaluation and interaction mechanism of beta-site APP cleaving enzyme 1 inhibitory pentapeptide from egg albumin, Food Sci. Hum. Well. 9(2) (2020) 162-167. https://doi.org/10.1016/j.fshw.2020.01.004.

[22]

W. Zhao, D. Zhang, Z. Yu, et al., Novel membrane peptidase inhibitory peptides with activity against angiotensin converting enzyme and dipeptidyl peptidase IV identified from hen eggs, J. Funct. Foods 64 (2020). https://doi.org/10.1016/j.jff.2019.103649.

[23]

E. Holen, B. Bolann, S. Elsayed, Novel B and T cell epitopes of chicken ovomucoid (Gal d 1) induce T cell secretion of IL-6, IL-13, and IFN-γ, Clin. Exp. Allergy 31(6) (2001) 952-964. https://doi.org/10.1046/j.1365-2222.2001.01102.x.

[24]

Y. Fan, Z. Yu, W. Zhao, et al., Identification and molecular mechanism of angiotensin-converting enzyme inhibitory peptides from Larimichthys crocea titin, Food Sci. Hum. Well. 9(3) (2020) 257-263. https://doi.org/10.1016/j.fshw.2020.04.001.

[25]

Z. Yu, L. Kang, W. Zhao, et al., Identification of novel umami peptides from myosin via homology modeling and molecular docking, Food Chem. 344 (2021) 128728. https://doi.org/10.1016/j.foodchem.2020.128728.

[26]

Z. Yu, H. Ji, J. Shen, et al., Identification and molecular docking study of fish roe-derived peptides as potent BACE 1, AChE, and BChE inhibitors, Food Funct. 11(7) (2020) 6643-6651. https://doi.org/10.1039/d0fo00971g.

[27]

M. Barati, F. Javanmardi, S.M.H.Mousavi Jazayeri, et al., Techniques, perspectives, and challenges of bioactive peptide generation: a comprehensive systematic review, Compr. Rev. Food Sci. Food Saf. 19(4) (2020) 1488-1520. https://doi.org/10.1111/1541-4337.12578.

[28]

E. Gasteiger, C. Hoogland, A. Gattiker, et al., Protein identification and analysis tools on the ExPASy server, The Proteomics Protocols Handbook (2005) 571-607. https://doi.org/10.1385/1-59259-584-7:531.

[29]

Z. Yu, R. Kan, H. Ji, et al., Identification of tuna protein-derived peptides as potent SARS-CoV-2 inhibitors via molecular docking and molecular dynamic simulation, Food Chem. 342 (2021) 128366. https://doi.org/10.1016/j.foodchem.2020.128366.

[30]

W. Shen, T. Matsui, Current knowledge of intestinal absorption of bioactive peptides, Food Funct. 8(12) (2017) 4306-4314. https://doi.org/10.1039/c7fo01185g.

[31]

S. Gupta, P. Kapoor, K. Chaudhary, et al., In silico approach for predicting toxicity of peptides and proteins, PLoS ONE 8(9) (2013) e73957. https://doi.org/10.1371/journal.pone.0073957.

[32]

J. He, N.L. Zhu, J. Kong, et al., A newly discovered phenylethanoid glycoside from stevia rebaudiana bertoni affects insulin secretion in rat INS-1 islet beta cells, Molecules 24(22) (2019) 4178. https://doi.org/10.3390/molecules24224178.

[33]

H. Cao, J.M. Pauff, R. Hille, X-ray crystal structure of a xanthine oxidase complex with the flavonoid inhibitor quercetin, J. Nat. Prod. 77(7) (2014) 1693-1699. https://doi.org/10.1021/np500320g.

[34]

W. He, G. Su, D. Sun-Waterhouse, et al., In vivo anti-hyperuricemic and xanthine oxidase inhibitory properties of tuna protein hydrolysates and its isolated fractions, Food Chem. 272 (2019) 453-461. https://doi.org/10.1016/j.foodchem.2018.08.057.

[35]

R. Ou, L. Lin, M. Zhao, et al, Action mechanisms and interaction of two key xanthine oxidase inhibitors in galangal: combination of in vitro and in silico molecular docking studies, Int. J. Biol. Macromol. 162 (2020) 1526-1535. https://doi.org/10.1016/j.ijbiomac.2020.07.297.

[36]

M.D. Santi, M.P. Zunini, B. Vera, et al., Xanthine oxidase inhibitory activity of natural and hemisynthetic flavonoids from Gardenia oudiepe (Rubiaceae) in vitro and molecular docking studies, Eur. J. Med. Chem. 143 (2018) 577-582. https://doi.org/10.1016/j.ejmech.2017.11.071.

[37]

A.B. Nongonierma, C. Mooney, D.C. Shields, et al., Inhibition of dipeptidyl peptidase Ⅳ and xanthine oxidase by amino acids and dipeptides, Food Chem. 141(1) (2013) 644-653. https://doi.org/10.1016/j.foodchem.2013.02.115.

[38]

K. Okamoto, K. Matsumoto, R. Hille, et al., The crystal structure of xanthine oxidoreductase during catalysis: implications for reaction mechanism and enzyme inhibition, Proc. Natl. Acad. Sci. USA 101(21) (2004) 7931-7936. https://doi.org/10.1073/pnas.0400973101.

[39]

Y. Wang, G. Zhang, J. Pan, et al., Novel insights into the inhibitory mechanism of kaempferol on xanthine oxidase, J. Agric. Food Chem. 63(2) (2015) 526-534. https://doi.org/10.1021/jf505584m.

[40]

Y. Yamaguchi, T. Matsumura, K. Ichida, et al., Human xanthine oxidase changes its substrate specificity to aldehyde oxidase type upon mutation of amino acid residues in the active site: roles of active site residues in binding and activation of purine substrate, J. Biochem. 141(4) (2007) 513-524. https://doi.org/10.1093/jb/mvm053.

[41]

J. Yan, G. Zhang, Y. Hu, et al., Effect of luteolin on xanthine oxidase: inhibition kinetics and interaction mechanism merging with docking simulation, Food Chem. 141(4) (2013) 3766-3773. https://doi.org/10.1016/j.foodchem.2013.06.092.

[42]

S. Lin, G. Zhang, Y. Liao, et al., Dietary flavonoids as xanthine oxidase inhibitors: structure-affinity and structure-activity relationships, J. Agric. Food Chem. 63(35) (2015) 7784-7794. https://doi.org/10.1021/acs.jafc.5b03386.

[43]

A.C. Pierce, K.L. Sandretto, G.W. Bemis, Kinase inhibitors and the case for CH.O hydrogen bonds in protein-ligand binding, Proteins 49(4) (2002) 567-576. https://doi.org/10.1002/prot.10259.

Food Science and Human Wellness
Pages 1591-1597
Cite this article:
Yu Z, Cao Y, Kan R, et al. Identification of egg protein-derived peptides as xanthine oxidase inhibitors: virtual hydrolysis, molecular docking, and in vitro activity evaluation. Food Science and Human Wellness, 2022, 11(6): 1591-1597. https://doi.org/10.1016/j.fshw.2022.06.017

490

Views

43

Downloads

24

Crossref

25

Web of Science

27

Scopus

1

CSCD

Altmetrics

Received: 02 April 2021
Revised: 31 May 2021
Accepted: 05 September 2021
Published: 18 July 2022
© 2022 Beijing Academy of Food Sciences.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return