National Technology Initiative Project Support Fund
Yekaterinburg, Russian Federation
Ural State Forestry Engineering University
Ekaterinburg, Ekaterinburg, Russian Federation
Peptides offer a promising analogue to synthetic drugs in treating hyperuricemia. This article introduces reliable amino acid patterns that cause the inhibitory xanthine oxidase (CSR) activity of peptides. The research objective was to propose new antihyperuricemic peptides and prove their effectiveness by predictive analytics in silico. The study featured peptides with inhibitory xanthine oxidase activity. The authors developed a protocol for searching, identifying, and quantifying patterns of amino acid residues in target peptide sequences. The identified peptides were tested for physicochemical properties, pharmacokinetic profile, inhibitory xanthine oxidase activity, general and target biological activity, and toxicity. The research revealed amino acid patterns responsible for inhibiting the xanthine oxidase enzyme, as well as generated new peptide sequences. Forty-nine non-toxic peptides with different lengths of amino acid sequences demonstrated high antimicrobial and inhibitory potential against the targeted drugs used to treat hyperuricemia and type 2 diabetes mellitus. The peptides were low-molecular compounds of predominantly hydrophilic and hydrophobic nature, 4-7 amino acids long. They contained negatively charged amino acid residues of proline, tryptophan, and phenylalanine with an average molecular weight of 723 Da. The study offers an important insight into the molecular mechanisms of xanthine oxidase inhibition and opens up new prospects for developing novel antihyperuricemic peptide drugs.
Peptides, hyperuricemia, xanthine oxidase inhibitors, amino acid patterns, IC50
1. Shalnova SA, Deev AD, Artamonova GV, Duplyakov DV, Efanov AYu, Zhernakova YuV, et al. Hyperuricemia and its correlates in the Russian population: ESSE-RF epidemiological study. Rational Pharmacotherapy In Cardiology. 2014;10 (2):153–159. (In Russ.).
2. Zhernakova YuV. Hyperuricemia as risk factor for cardiovascular disease – what’s new? Medical Alphabet. 2020;(13): 5–11. (In Russ.). https://doi.org/10.33667/2078-5631-2020-13-5-11; https://elibrary.ru/MUQHSO
3. Yeliseyev MS, Yeliseyeva MYe. Modern aspects of pathogenesis and correction of hyperuricemia and associated conditions. Effective Pharmacotherapy. 2019;15(8):32–40. (In Russ). https://doi.org/10.33978/2307-3586-2019-15-8-32-40; https://elibrary.ru/SNGWTM
4. Li Q, Kang X, Shi C, Li Y, Majumder K, Ninga Z. Moderation of hyperuricemia in rats via consuming walnut protein hydrolysate diet and identification of new antihyperuricemic peptides. Food and Function. 2018;9(1):107–116. https://doi.org/https://doi.org/10.1039/c7fo01174a
5. Lai S-W, Hwang B-F, Kuo Y-H, Liu C-S, Liao K-F. Allopurinol use and the risk of dementia a meta-analysis of case-control studies. Medicine.2022;101(26):e29827. http://doi.org/10.1097/md.0000000000029827
6. van der Pol KH, Koenderink J, van den Heuvel JJMW, van den Broek P, Peters J, van Bunningen IDW, et al. Effects of allopurinol and febuxostat on uric acid transport and transporter expression in human umbilical vein endothelial cells. PLoS ONE. 2024;19(6):e0305906. https://doi.org/10.1371/journal.pone.0305906
7. Matsumoto K, Okamoto K, Ashizawa N, Nishino T. FYX-051: A novel and potent hybrid-type inhibitor of xanthine oxidoreductase. Journal of Pharmacology and Experimental Therapeutics. 2011;336(1):95–103. https://doi.org/10.1124/ jpet.110.174540
8. Li Q, Kang X, Shi C, Li Y, Majumder K, Ninga Z. Moderation of hyperuricemia in rats via consuming walnut protein hydrolysate diet and identification of new antihyperuricemic peptides. Food and Function. 2018;9(1):107–116. https:// doi.org/10.1039/c7fo01174a
9. Mehmood A, Zhao L, Wang C, Nadeem M, Raza A, Ali N, et al. Management of hyperuricemia through dietary polyphenols as a natural medicament. Critical Reviews in Food Science and Nutrition. 2019;59(9):1433–1455. https://doi.org/https://doi.org/10.1080/10408398.2017.1412939
10. Gao Y-F, Liu M-Q, Li Z-H, Zhang H-L, Hao J-Q, Liu B-H, Li X-Y, et al. Purification and identification of xanthine oxidase inhibitory peptides from enzymatic hydrolysate of α-lactalbumin and bovine colostrum casein. Food Research International. 2023;169:112882. https://doi.org/10.1016/j.foodres.2023.112882
11. Nongonierma AB, Fitzgerald RG. Tryptophan-containing milk protein-derived dipeptides inhibit xanthine oxidase. Peptides. 2012;37(2):263–272. https://doi.org/10.1016/j.peptides.2012.07.030
12. Qi X, Chen H, Guan K, Sun Y, Wang R, Li Q, et al. Novel xanthine oxidase inhibitory peptides derived from whey protein: identification, in vitro inhibition mechanism and in vivo activity validation. Bioorganic Chemistry. 2022;128:106097. https://doi.org/10.1016/j.bioorg.2022.106097
13. Xu Y, Gong H, Zou Y, Mao X. Antihyperuricemic activity and inhibition mechanism of xanthine oxidase inhibitory peptides derived from whey protein by virtual screening. Journal Dairy Science.2024;107(4):1877–1886. https://doi.org/10.3168/ jds.2023-24028
14. Ahmed AS, El-Bassiony T, Elmalt LM, Ibrahim HR. Identification of potent antioxidant bioactive peptides from goat milk proteins. Food Research International. 2015;74:80–88. https://doi.org/10.1016/j.foodres.2015.04.032
15. Yu Zh, Cao Y, Kan R, Ji H, Zhao W, Wu S, 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
16. Thaha A, Wang B-S, Chang Y-W, Hsia S-M, Huang T-C, Shiau C-Y, et al. Food-derived bioactive peptides with antioxidative capacity, xanthine oxidase and tyrosinase inhibitory activity. Processes. 2021;9(5):747. https://doi.org/10.3390/ pr9050747
17. Bu Y, Wang F, Zhu W, Li X. Combining bioinformatic prediction and assay experiment to identify novel xanthine oxidase inhibitory peptides from Pacific bluefin tuna (Thunnus Orientalis). E3S Web of Conferences. 2020;185:04062. https:// doi.org/10.1051/e3sconf/202018504062
18. Zhang P, Jiang Z, Lei J, Yan Q, Chang C. Novel hemoglobin-derived xanthine oxidase inhibitory peptides: Enzymatic preparation and inhibition mechanisms. Journal of Functional Foods. 2023;102:105459. https://doi.org/10.1016/j.jff.2023.105459
19. Murota I, Taguchi S, Sato N, Park EY, Nakamura Y, Sato K. Identification of antihyperuricemic peptides in the proteolytic digest of shark cartilage water extract using in vivo activity-guided fractionation. Journal of Agricultural and Food Chemistry. 2014;62(11):2392–2397. https://doi.org/10.1021/jf405504u
20. Li Y, Kang X, Li Q, Shi C, Lian Y, Yuan E, et al. Anti-hyperuricemic peptides derived from bonito hydrolysates based on in vivo hyperuricemic model and in vitro xanthine oxidase inhibitory activity. Peptides. 2018;107:45–53. https:// doi.org/10.1016/j.peptides.2018.08.001
21. Hu X, Zhou Y, Zhou S, Chen S, Wu Y, Li L, et al. Purification and identification of novel xanthine oxidase inhibitory peptides derived from round scad (Decapterus Maruadsi) protein hydrolysates. Marine Drugs. 2021;19(10):538. https:// doi.org/10.3390/md19100538
22. Song M. Screening of xanthine oxidase inhibiting peptides in meat Protein of bluespot mackerel based on ligand fishing. BoHai University. 2020. [Internet]. [cited 2023 Dec 18]. Available from: https://cdmd.cnki.com.cn/Article/CDMD- 10167-1020750025.htm
23. Cui F, Xi L, Zhao G, Wang D, Tan X, Li J, et al. Screening of xanthine oxidase inhibitory peptides by ligand fishing and molecular docking technology. Food Bioscience. 2022;50:102152. https://doi.org/10.1016/j.fbio.2022.102152
24. Zhong H, Abdullah, Zhang Y, Deng L, Zhao M, Tang J, et al. Exploring the potential of novel xanthine oxidase inhibitory peptide (ACECD) derived from Skipjack tuna hydrolysates using affinity-ultrafiltration coupled with HPLC-MALDI- TOF/TOF-MS. Food Chemistry. 2021;347:129068. https://doi.org/10.1016/j.foodchem.2021.129068
25. Zhang H, Saravanan KM, Zhang JZH, Wu X. Deep-learning based bioactive peptides generation and screening against Xanthine oxidase. bioRxiv. 2023;11:523536. https://doi.org/10.1101/2023.01.11.523536
26. Hou M, Xiang H, Hu X, Chen Sh, Wu Y, Xu J, et al. Novel potential XOD inhibitory peptides derived from Trachi- notus ovatus: isolation, identification and structure-function analysis. Food Bioscience. 2022;47:101639. https://doi.org/10.1016/ j.fbio.2022.101639
27. Wei L, Hongwu JI, Song W, Peng S, Zhan S, Qu Y, et al. Identification and molecular docking of two novel peptides with xanthine oxidase inhibitory activity from Auxis thazard. Food Science and Technology. 2022;42:e106921. https:// doi.org/10.1590/fst.106921
28. Chen X, Guan W, Li Y, Zhang J, Cai L. Xanthine oxidase inhibitory peptides from Larimichthys polyactis: characterization and in vitro/in silico evidence. Foods. 2023;2:982. https://doi.org/10.3390/foods12050982
29. Zhao Q, Meng Y, Liu J, Hu Z, Du Y, Sun J, et al. Separation, identification and docking analysis of xanthine oxidase inhibitory peptides from pacific cod bone-flesh mixture. LWT. 2022;167:113862. https://doi.org/10.1016/j.lwt. 2022.113862
30. Yu Z, Kan R, Wu S, Guo H, Zhao W, Ding L, et al. Xanthine oxidase inhibitory peptides derived from tuna protein: virtual screening, inhibitory activity, and molecular mechanisms. Journal of the Science of Food and Agriculture. 2021; 101(4):1349–1354. https://doi.org/10.1002/jsfa.10745
31. Zhao Q, Jiang X, Mao Zh, Zhang J, Sun J, Mao X. Exploration, sequence optimization and mechanism analysis of novel xanthine oxidase inhibitory peptide from Ostrea rivularis Gould. Food Chemistry. 2023;404:134537. https://doi.org/https://doi.org/10.1016/j.foodchem.2022.134537
32. Mao Zh, Jiang H, Sun J, Mao X. Virtual screening and structure optimization of xanthine oxidase inhibitory peptides from whole protein sequences of Pacific white shrimp via molecular docking. Food Chemistry. 2023;429:136837. https:// doi.org/10.1016/j.foodchem.2023.136837
33. Liu N, Wang Y, Yang M, Bian W, Zeng L, Yin S, et al. A new rice-derived short peptide potently alleviated hyperuricemia induced by potassium oxonate in rats. Journal of Agricultural and Food Chemistry.2019;67(1):220–228. https:// doi.org/10.1021/acs.jafc.8b05879
34. Li QY. Study on the structure-activity mechanism of targeting inhibition of xanthine oxidase by uric acid-lowering peptides derived from walnut. [dissertation]. China: South China University of Technology, 2018.
35. Wu Y, He H, Hou T. Purification, identification, and computational analysis of xanthine oxidase inhibitory peptides from kidney bean. Journal of Food Science. 2021;86(3):1081–1088. https://doi.org/10.1111/1750-3841.15603 15603
36. Dong Y, Sun N, Ge Q, Lv R, Lin S. Antioxidant soy peptide can inhibit xanthine oxidase activity and improve LO2 cell damage. Food Bioscience. 2023;52:102455. https://doi.org/10.1016/j.fbio.2023.102455
37. Jang I-T, Hyun S-H, Shin J-W, Lee Y-H, Ji J-H, Lee J-S. Characterization of an antigout xanthine oxidase inhibitor from pleurotusostreatus. Mycobiology.2014;42(3):296–300. https://doi.org/10.5941/myco.2014.42.3.296
38. Serba EM, Yuraskina TV, Rimareva LV, Tadzibova PYu, Sokolova EN, Volkova GS. Microbial Biomass as a Bioresource of Functional Food Ingredients: A Review. Food Processing: Techniques and Technology. 2023;53(3):426–444. (In Russ.). https://doi.org/10.21603/2074-9414-2023-3-2446; https://www.elibrary.ru/OYPVMI
39. Halavach TM, Kurchenko VP, Tarun EI, Romanovich RV, Mushkevich NV, Kazimirov AD, et al. Chitosan complexes with amino acids and whey peptides: Sensory and antioxidant properties. Foods and Raw Materials. 2024;12(1):13–21. https:// doi.org/10.21603/2308-4057-2024-1-584; https://elibrary.ru/XMDORK
40. Classical and molecular biology [Internet]. [cited 2023 Dec 21]. Available from: https://molbiol.ru/?&langid=en
41. Kyte J, Doolittle RF. A simple method for displaying the hydropathic character of a protein. Journal of Molecular Biology. 1982;157(1):105–132. https://doi.org/10.1016/0022-2836(82)90515-0
42. Kumar N, Kaur K, Bedi PMS. Hybridization of molecular docking studies with machine learning based QSAR model for prediction of xanthine oxidase activity. Computational and Theoretical Chemistry. 2023;1227:114262. https://doi.org/https://doi.org/10.1016/J.COMPTC.2023.114262
43. Thakur A, Kumar A, Sharma V, Mehta V. PIC50: An open source tool for interconversion of PIC50 values and IC50 for efficient data representation and analysis. https://doi.org/10.13140/RG.2.2.18440.70408
44. Du Z, Ding X, Xu Y, Li Y. UniDL4BioPep: a universal deep learning architecture for binary classification in peptide bioactivity. Briefings in Bioinformatics. 2023;24(3):bbad135. https://doi.org/10.1093/bib/bbad135
45. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports. 2017;7:42717. https://doi.org/10.1038/srep42717
46. Manzoor M, Singh J, Gani A. Exploration of bioactive peptides from various origin as promising nutraceutical treasures: in vitro, in silico and in vivo studies. Food Chemistry. 2021;373:131395. https://doi.org/10.1016/j.foodchem.2021.131395
47. Nongonierma AB, Mooney C, Shields DC, FitzGerald RJ. Inhibition of dipeptidyl peptidase IV and xanthine oxidase by amino acids and dipeptides. Food Chemistry. 2013;141(1):644–653. https://doi.org/10.1016/j.foodchem.2013.02.115
48. Bellaver EH, Kempka AP. Potential of milk-derived bioactive peptides as antidiabetic, antihypertensive, and xanthine oxidase inhibitors: a comprehensive bibliometric analysis and updated review. Amino Acids. 2023;55:1829–1855. https:// doi.org/10.1007/s00726-023-03351-9
49. Li Y, Kang X, Li Q, Shi C, Lian Y,Yuan E, et al. Anti-hyperuricemic peptides derived from bonito hydrolysates based on in vivo hyperuricemic model and in vitro xanthine oxidase inhibitory activity. Peptides. 2018;107:45–53. https:// doi.org/10.1016/j.peptides.2018.08.001
50. He W, Su G, Sun-Waterhouse D, Waterhouse GIN, Zhao M, Liu Y. In vivo anti-hyperuricemic and xanthine oxidase inhibitory properties of tuna protein hydrolysates and its isolated fractions. Food Chemistry. 2019;272:453–461. https://doi. org/10.1016/j.foodchem.2018.08.057
51. Li Q, Shi C, Wang M, Zhou M, Liang M, Zhang T, et al. Tryptophan residue enhances in vitro walnut protein- derived peptides exerting xanthine oxidase inhibition and antioxidant activities. Journal of Functional Foods. 2019;53:276–285. https://doi.org/10.1016/j.jff.2018.11.024
52. Zhao L, Ai X, Pan F, Zhou N, Zhao L, Cai S, et al. Novel peptides with xanthine oxidase inhibitory activity identified from macadamia nuts: integrated in silico and in vitro analysis. European Food Research and Technology. 2022;248:2031–2042. https://doi.org/10.1007/s00217-022-04028-5
53. Xu Y, Gong H, Zou Y, Mao X. Antihyperuricemic activity and inhibition mechanism of xanthine oxidase inhibitory peptides derived from whey protein by virtual screening. Journal Dairy Science. 2023;107(4):1877–1886. https://doi.org/10.3168/ jds.2023-24028
54. Huang X-N, Zhang Y-M, Wen Y, Jiang Y, Wang C-H. Protease-catalyzed rational synthesis of uric acid-lowering peptides in non-aqueous medium. International Journal of Peptide Research and Therapeutics. 2022;28:61. https://doi.org/10.1007/ s10989-022-10367-4
55. Allahyari M, Samadi-Noshahr Z, Hosseinian S, Salmani H, Noras M, Khajavi-Rad A. Camel milk and allopurinol attenuated adenine-induced acute renal failure in rats Iranian Journal of Science. 2021;45:1539–1548. https://doi.org/10.1007/ s40995-021-01155-8
56. Li Q, Li X, Wang J, Liu H, Kwong JS-W, Chen H, et al. Diagnosis and treatment for hyperuricemia and gout: a systematic review of clinical practice guidelines and consensus statements. BMJ Open. 2019;9:e026677. http://doi.org/10.1136/ bmjopen-2018-026677
57. Huang Y, Fan S, Lu G, Sun N, Wang R, Lu C, et al. Systematic investigation of the amino acid profiles that are correlated with xanthine oxidase inhibitory activity: Effects, mechanism and applications in protein source screening. Free Radical Biology and Medicine. 2021;177:326–336. https://doi.org/10.1016/j.freeradbiomed.2021.11.004
58. Miller MB, Bassler BL. Quorum sensing in bacteria. Annual Review of Microbiology. 2001;55:165–199. https:// doi.org/10.1146/annurev.micro.55.1.165
59. Shirvani-Rad S, Khatibzade-Nasari N, Ejtahed H-S, Larijani B. Exploring the role of gut microbiota dysbiosis in gout pathogenesis: a systematic review. Frontiers in Medicine. 2023;10:1163778. https://doi.org/10.3389/fmed.2023.1163778
60. Zheliabina OV, Eliseev MS, Glukhova SI, Nasonov EL. Contributing Factors of Diabetes Mellitus among Patients wth Gout (Results of the Long-Term Prospective Study). Doklady Biochemistry and Biophysics. 2023;511(1):195–202. https:// doi.org/10.1134/S1607672923700321; https://elibrary.ru/TWNBMB