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 (3 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Open Access

Exploration of cyclooxygenase-2 inhibitory peptides from walnut dreg proteins based on in silico and in vitro analysis

Zishan Honga,b,c,1,Jing Xiea,b,c,1Liang Taoa,b,c,1Jing-Jing Daib,d,eTingting Lia,b,eLi Zhanga,c,eYuying Baia,b,eXia Hua,c,eJinlian Chena,b,eJun Shenga,f( )Yang Tiana,b,c,d,e( )
College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
Yunnan Provincial Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
School of Tea and Coffee, Pu’er University, Pu’er 665000, China
Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming 650201, China
Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China

1 These authors contributed equally to this study.

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Highlights

• Peptides in walnut dreg proteins are efficiently released by alcalase.

• Alcalase hydrolysate (Al-WPH) inhibits cyclooxygenase-2 (COX-2) activity.

• Four novel peptides (AGFP, FPGA, LFPD, and VGFP) were identified in Al-WPH.

• AGFP, FPGA, LFPD, and VGFP showed potential COX-2 inhibitory activity.

• Walnut dreg protein is an excellent low-cost source of anti-inflammatory peptides.

Graphical Abstract

Abstract

Walnut dreg protein hydrolysates (WDPHs) exhibit a variety of biological activities, however, the cyclooxygenase-2 (COX-2) inhibitory peptide of WDPHs remain unclear. The aim of this study was to rapidly screen for such peptides in WDPHs through a combination of in silico and in vitro analysis. In total, 1262 peptide sequences were observed by nano liquid chromatography/tandem mass spectrometry (nano LC-MS/MS) and 4 novel COX-2 inhibitory peptides (AGFP, FPGA, LFPD, and VGFP) were identif ied. Enzyme kinetic data indicated that AGFP, FPGA, and LFPD displayed mixed-type COX-2 inhibition, whereas VGFP was a non-competitive inhibitor. This is mainly because the peptides form hydrogen bonds and hydrophobic interactions with residues in the COX-2 active site. These results demonstrate that computer analysis combined with in vitro evaluation allows for rapid screening of COX-2 inhibitory peptides in walnut protein dregs.

Electronic Supplementary Material

Download File(s)
fshw-13-3-1636_ESM1.docx (613.4 KB)

References

[1]

K. Uchida, HNE as an inducer of COX-2, Free Radic. Biol. Med. 111 (2017) 169-172. https://doi.org/10.1016/j.freeradbiomed.2017.02.004.

[2]

M.A.H. Khan, S.H. Hwang, S.D. Barnett, et al., Multitarget molecule, PTUPB, to treat diabetic nephropathy in rats, Br. J. Pharmacol. 178 (2021) 4468-4484. https://doi.org/10.1111/bph.15623.

[3]

C. Cozowicz, J. Poeran, N. Zubizarreta, et al., Non-opioid analgesic modes of pain management are associated with reduced postoperative complications and resource utilisation: a retrospective study of obstructive sleep apnoea patients undergoing elective joint arthroplasty, Br. J. Anaesth. 122 (2019) 131-140. https://doi.org/10.1016/j.bja.2018.08.027.

[4]

Y.H. Luan, D. Wang, Q. Yu, et al., Action of β-endorphin and nonsteroidal anti-inflammatory drugs, and the possible effects of nonsteroidal antiinflammatory drugs on β-endorphin, J. Clin. Anesth. 37 (2017) 123-128. https://doi.org/10.1016/j.jclinane.2016.12.016.

[5]

A. Karateev, A. Tsurgan, N. Gontarenko, AB0987 Recurrence of Nsaid -induced ulcers: Has the situation changed in recent years? Ann. Rheum. Dis. 75 (2016) 1238. https://doi.org/10.1136/annrheumdis-2016-eular.4708.

[6]

K.B. Sondergaard, G. Gislason, NSAIDs and cardiac arrest: non-steroidal anti-inflammatory drug use is associated with increased risk of out-ofhospital cardiac arrest: a nationwide case–time–control study, Eur. Heart J. 38 (2017) 1788-1789. https://doi.org/10.1093/ehjcvp/pvw041.

[7]

D. Liu, Y. Guo, H. Ma, Production, bioactivities and bioavailability of bioactive peptides derived from walnut origin by-products: a review, Crit. Rev. Food Sci. Nutr. (2022) 1-16. https://doi.org/10.1080/10408398.2022.2054933.

[8]

T. Zhi, D. Hong, Z. Zhang, et al., Anti-inflammatory and gut microbiota regulatory effects of walnut protein derived peptide LPF in vivo, Food Res. Int. 152 (2022) 110875. https://doi.org/10.1016/j.foodres.2021.110875.

[9]

X. Kong, L. Zhang, W. Song, et al., Separation, identification and molecular binding mechanism of dipeptidyl peptidase IV inhibitory peptides derived from walnut (Juglans regia L.) protein, Food Chem. 347 (2021) 129062. https://doi.org/10.1016/j.foodchem.2021.129062.

[10]

D. Liu, Y. Guo, P. Wu, et al., The necessity of walnut proteolysis based on evaluation after in vitro simulated digestion: ACE inhibition and DPPH radical-scavenging activities, Food Chem. 311 (2020) 125960. https://doi.org/10.1016/j.foodchem.2019.125960.

[11]

S. Sun, X. Xu, X. Sun, et al., Preparation and identification of ACE inhibitory peptides from the marine macroalga Ulva intestinalis, Mar. Drugs 17 (2019) 179. https://doi.org/10.3390/md17030179.

[12]

H. Sun, Q. Chang, L. Liu, et al., High-throughput and rapid screening of novel ACE inhibitory peptides from sericin source and inhibition mechanism by using in silico and in vitro prescriptions, J. Agric. Food Chem. 65 (2017)10020-10028. https://doi.org/10.1021/acs.jafc.7b04043.

[13]

C. Yan, Z. Zhou, Walnut pellicle phenolics greatly influence the extraction and structural properties of walnut protein isolates, Food Res. Int. 141 (2021) 110163. https://doi.org/10.1016/j.foodres.2021.110163.

[14]

S. Wang, G. Su, J. Fan, et al., Arginine-containing peptides derived from walnut protein against cognitive and memory impairment in scopolamineinduced zebrafish: design, release, and neuroprotection, J. Agric. Food Chem. 70 (2022) 11579-11590. https://doi.org/10.1021/acs.jafc.2c05104.

[15]

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

[16]

Q. Li, C. Zhang, H. Chen, et al., BioPepDB: an integrated data platform for food-derived bioactive peptides, Int. J. Food Sci. Nutr. 69 (2018) 963-968. https://doi.org/10.1080/09637486.2018.1446916.

[17]

A.A. Zamyatnin, A.S. Borchikov, M.G. Vladimirov, et al., The EROPMoscow oligopeptide database, Nucleic Acids Res. 34 (2006) D261-D266. https://doi.org/10.1093/nar/gkj008.

[18]

H. Zhang, G. Chen, Y. Zhang, et al., Potential hypoglycemic, hypolipidemic, and anti-inflammatory bioactive components in Nelumbo nucifera leaves explored by bioaffinity ultrafiltration with multiple targets, Food Chem. 375 (2022) 131856. https://doi.org/10.1016/j.foodchem.2021.131856.

[19]

M. Yan, H. Tao, S. Qin, Effect of enzyme type on the antioxidant activities and functional properties of enzymatic hydrolysates from sea cucumber (Cucumaria frondosa) viscera, J. Aquat. Food Prod. Technol. 25 (2016) 940-952. https://doi.org/10.1080/10498850.2014.994083.

[20]

C. Yu, L. Zheng, Y. Cai, et al., Desirable characteristics of casein peptides with simultaneously enhanced emulsion forming ability and antioxidative capacity in O/W emulsion, Food Hydrocoll. 131 (2022) 107812. https://doi.org/10.1016/j.foodhyd.2022.107812.

[21]

N. Lapsongphon, J. Yongsawatdigul, Production and purification of antioxidant peptides from a mungbean meal hydrolysate by Virgibacillus sp.SK37 proteinase, Food Chem. 141 (2013) 992-999. https://doi.org/10.1016/j.foodchem.2013.04.054.

[22]

M. Merz, T. Eisele, P. Berends, et al., Flavourzyme, an enzyme preparation with industrial relevance: automated nine-step purification and partial characterization of eight enzymes, J. Agric. Food Chem. 63 (2015) 5682-5693. https://doi.org/10.1021/acs.jafc.5b01665.

[23]

R. Jin, X. Teng, J. Shang, et al., Identification of novel DPP-IV inhibitory peptides from Atlantic salmon (Salmo salar) skin, Food Res. Int. 133 (2020) 109161. https://doi.org/10.1016/j.foodres.2020.109161.

[24]

S. Rawdkuen, N. Rodzi, S. Pinijsuwan, Characterization of sacha inchi protein hydrolysates produced by crude papain and Calotropis proteases, LWT-Food Sci. Technol. 98 (2018) 18-24. https://doi.org/10.1016/j.lwt.2018.08.008.

[25]

Z.Y. Li, W. Youravong, A.H. Kittikun, Protein hydrolysis by protease isolated from tuna spleen by membrane filtration: a comparative study with commercial proteases, LWT-Food Sci. Technol. 43 (2010) 166-172. https://doi.org/10.1016/j.lwt.2009.07.002.

[26]

S. Wang, L. Zheng, T. Zhao, et al., Inhibitory effects of walnut (Juglans regia) peptides on neuroinflammation and oxidative stress in lipopolysaccharide-induced cognitive impairment mice, J. Agric. Food Chem. 68 (2020) 2381-2392. https://doi.org/10.1021/acs.jafc.9b07670.

[27]

S.K. Sharma, B.J. Al-Hourani, M. Wuest, et al., Synthesis and evaluation of fluorobenzoylated di- and tripeptides as inhibitors of cyclooxygenase-2 (COX-2), Bioorg. Med. Chem. 20 (2012) 2221-2226. https://doi.org/10.1016/j.bmc.2012.02.021.

[28]

S.A. Nankar, A.H. Pande, Properties of apolipoprotein E derived peptide modulate their lipid-binding capacity and influence their anti-inflammatory function, Biochim. Biophys. Acta 1841 (2014) 620-629. https://doi.org/10.1016/j.bbalip.2014.01.006.

[29]

R. Suttisuwan, S. Phunpruch, T. Saisavoey, et al., Isolation and characterization of anti-inflammatory peptides derived from trypsin hydrolysis of microalgae protein (Synechococcus sp. VDW), Food Biotechnol. 33 (2019) 303-324. https://doi.org/10.1080/08905436.2019.1673171.

[30]

V.G. Tacias-Pascacio, R. Morellon-Sterling, E.H. Siar, et al., Use of alcalase in the production of bioactive peptides: a review, Int. J. Biol. Macromol. 165 (2020) 2143-2196. https://doi.org/10.1016/j.ijbiomac.2020.10.060.

[31]

J. Fernández-Lucas, D. Castañeda, D. Hormigo, New trends for a classical enzyme: papain, a biotechnological success story in the food industry, Trends Food Sci. Technol. 68 (2017) 91-101. https://doi.org/10.1016/j.tifs.2017.08.017.

[32]

G. Rajasekaran, R. Kamalakannan, S.Y. Shin, Enhancement of the antiinflammatory activity of temporin-1Tl-derived antimicrobial peptides by tryptophan, arginine and lysine substitutions, J. Pept. Sci. 21 (2015) 779-785. https://doi.org/10.1002/psc.2807.

[33]

S.J. Walmsley, P.A. Rudnick, Y. Liang, et al., Comprehensive analysis of protein digestion using six trypsins reveals the origin of trypsin as a significant source of variability in proteomics, J. Proteome Res. 12 (2013) 5666-5680. https://doi.org/10.1021/pr400611h.

[34]
L. Mora, M.C. Aristoy, F. Toldrá, Bioactive peptides in foods, in: B. Caballero, P.M. Finglas and F. Toldrá (Eds.), Encyclopedia of Food and Health, Academic Press, Oxford, 2016, pp. 395-400, https://doi.org/10.1016/B978-0-12-384947-2.00067-2.
[35]

C. Millan-Linares Mdel, M. Yust Mdel, J.M. Alcaide-Hidalgo, et al., Lupine protein hydrolysates inhibit enzymes involved in the inflammatory pathway, Food Chem. 151 (2014) 141-147. https://doi.org/10.1016/j.foodchem.2013.11.053.

[36]

J.L. Zhang, F.F. Zhou, Y.Z. Li, et al., New sesquiterpenoids with COX-2 inhibitory activity from the medical plant Physalis alkekengi L. var. franchetii, Fitoterapia 141 (2020) 104470. https://doi.org/10.1016/j.fitote.2020.104470.

[37]

W. Liu, H. Li, Y. Wen, et al., Molecular mechanism for the α-glucosidase inhibitory effect of wheat germ peptides, J. Agric. Food Chem. 69 (2021) 15231-15239. https://doi.org/10.1021/acs.jafc.1c06098.

[38]

G. Fricker, C. Bruns, J. Munzer, et al., Intestinal absorption of the octapeptide SMS 201–995 visualized by fluorescence derivatization, Gastroenterology 100 (1991) 1544-1552. https://doi.org/10.1016/0016-5085(91)90651-z.

[39]

A.M. Ghribi, A. Sila, R. Przybylski, et al., Purification and identification of novel antioxidant peptides from enzymatic hydrolysate of chickpea (Cicer arietinum L.) protein concentrate, J. Funct. Foods 12 (2015) 516-525. https://doi.org/10.1016/j.jff.2014.12.011.

[40]

H. You, T. Wu, W. Wang, et al., Preparation and identification of dipeptidyl peptidase IV inhibitory peptides from quinoa protein, Food Res. Int. 156 (2022) 111176. https://doi.org/10.1016/j.foodres.2022.111176.

[41]

J. Feng, Y.L. Ma, P. Sun, et al., Purification and characterisation of α-glucosidase inhibitory peptides from defatted camellia seed cake, Int. J. Food Sci. Technol. 56 (2020) 138-147. https://doi.org/10.1111/ijfs.14613.

[42]

R. Jarapula, K. Gangarapu, S. Manda, et al., Synthesis, in vivo antiinflammatory activity, and molecular docking studies of new isatin derivatives, Int. J. Med. Chem. 2016 (2016) 2181027. https://doi.org/10.1155/2016/2181027.

[43]

F.A. Ragab, H.I. Heiba, M.G. El-Gazzar, et al., Anti-inflammatory, analgesic and COX-2 inhibitory activity of novel thiadiazoles in irradiated rats, J. Photochem. Photobiol. B: Biol. 166 (2016) 285-300. https://doi.org/10.1016/j.jphotobiol.2016.12.007.

[44]

D.K. Yadav, Saloni, P. Sharma, et al., Studies of the benzopyran class of selective COX-2 inhibitors using 3D-QSAR and molecular docking, Arch. Pharm. Res. 41 (2018) 1178-1189. https://doi.org/10.1007/s12272-017-0945-7.

[45]

T. Sumaryada, A. Astarina, L. Ambarsari, Molecular docking and physicochemical analysis of the active compounds of soursop (Annona muricata Linn) for an anti-breast cancer agent, Biointerface Res. Appl. Chem. 11 (2021) 11380-11389. https://doi.org/10.33263/BRIAC114.1138011389.

[46]

M.A. Ahmaditaba, S. Shahosseini, B. Daraei, et al., Design, synthesis, and biological evaluation of new peptide analogues as selective COX-2 inhibitors, Arch. Pharm. 350 (2017) 1700158. https://doi.org/10.1002/ardp.201700158.

[47]

S. Gupta, A.K. Sharma, V. Shastri, et al., Prediction of anti-inflammatory proteins/peptides: an insilico approach, J. Transl. Med. 15 (2017) 7. https://doi.org/10.1186/s12967-016-1103-6.

[48]

S.R.C.K. Rajendran, B. Mason, C.C. Udenigwe, Peptidomics of peptic digest of selected potato tuber proteins: Post-translational modifications and limited cleavage specificity, J. Agric. Food Chem. 64 (2016) 2432-2437. https://doi.org/10.1021/acs.jafc.6b00418.

[49]

A.B. Nongonierma, S. Paolella, P. Mudgil, et al., Identification of novel dipeptidyl peptidase IV (DPP-IV) inhibitory peptides in camel milk protein hydrolysates, Food Chem. 244 (2017) 340-348. https://doi.org/10.1016/j.foodchem.2017.10.033.

[50]

M.M. Wang, Y.N. Li, W.K. Ming, et al., Bioassay-guided isolation of human carboxylesterase 2 inhibitory and antioxidant constituents from Laportea bulbifera: inhibition interactions and molecular mechanism, Arab. J. Chem. 15 (2022) 103723. https://doi.org/10.1016/j.arabjc.2022.103723.

Food Science and Human Wellness
Pages 1636-1644
Cite this article:
Hong Z, Xie J, Tao L, et al. Exploration of cyclooxygenase-2 inhibitory peptides from walnut dreg proteins based on in silico and in vitro analysis. Food Science and Human Wellness, 2024, 13(3): 1636-1644. https://doi.org/10.26599/FSHW.2022.9250143

596

Views

146

Downloads

1

Crossref

0

Web of Science

0

Scopus

0

CSCD

Altmetrics

Received: 25 December 2022
Revised: 12 January 2023
Accepted: 05 February 2023
Published: 08 February 2024
© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

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

Return