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 (5.9 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

Donkey whey proteins ameliorate dextran sulfate sodium-induced ulcerative colitis in mice by downregulating the S100A8-TRAF6-NF-κB axis-mediated inflammatory response

Mohan Lia,1Qilong Lib,1Rayhnigul AbdllaaJiali ChenaXiqing Yuea( )Siew Young Quekc,d( )
College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
Food Science, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
Riddet Institute, Centre for Research Excellence in Food Research, Palmerston North 4474, New Zealand

1 These authors contributed equally to this work.

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

Show Author Information

Abstract

Donkey milk has a variety of physiological functions, including antibacterial and anti-inflammatory. Donkey whey proteins (DWPs), as the main functional component in donkey milk, its inhibitory effect on colitis is still unclear. In this study, the inhibitory effect and potential mechanism of DWPs on dextran sulfate sodium (DSS)-induced colitis were investigated. Firstly, the DWPs and bovine milk whey proteins (BWPs) were characterized using proteomics. Then, we administered DWPs and BWPs to mice with colitis via oral gavage. The results of immunohistochemistry and flow cytometry indicated that DWPs increased T regulatory cell accumulation and increased the abundance of the cluster of differentiation 205+ (CD205+) macrophages compared to those with BWPs and in model groups. In addition, DWPs exhibited a more remarkable ability to inhibit pro-inflammatory proteins (S100A8, TRAF6, and NF-κB) expression and inflammatory secretion than BWPs. In addition, DWPs significantly decreased NF-κB and CD86 levels more than BWPs or the negative control in both LPS-stimulated human peripheral blood mononuclear cells or cell lines. These findings indicate that DWPs comprise a promising anti-colitis functional food, and this work has established a foundation for future research on these compounds.

References

[1]

A.C. Ford, P. Moayyedi, S.B. Hanauer, Ulcerative colitis, BMJ 346 (2013) f432. https://doi.org/10.1136/bmj.f432.

[2]

M.H. Wanderås, B.A. Moum, M.L. Høivik, et al., Predictive factors for a severe clinical course in ulcerative colitis: results from population-based studies, World J. Gastrointest. Pharmacol. Ther. 7 (2016) 235-241. https://doi.org/10.4292/wjgpt.v7.i2.235.

[3]

A. Lavelle, H. Sokol, Gut microbiota-derived metabolites as key actors in inflammatory bowel disease, Nat. Rev. Gastroenterol. Hepatol. 17 (2020) 223–237.

[4]

X. Chang, S.L. Wang, S.B. Zhao, et al., Extracellular vesicles with possible roles in gut intestinal tract homeostasis and IBD, Mediators Inflamm. 2020 (2020).

[5]

J. Meier, A. Sturm, Current treatment of ulcerative colitis, World J. Gastroenterol. 17 (2011) 3204.

[6]

S. Singh, M. Fumery, W. Sandborn, et al., Systematic review with network meta-analysis: first-and second-line pharmacotherapy for moderate-severe ulcerative colitis, Aliment. Pharmacol. Ther. 47 (2018) 162-175.

[7]

Q. Chen, H. Wang, C. Zhu, et al., Anti-apoptotic effects of milk-derived casein glycomacropeptide on mice with ulcerative colitis, Food Agric. Immunol. 25 (2014) 453-466.

[8]

K. Matsuoka, Y. Uemura, T. Kanai, et al., Efficacy of Bifidobacterium breve fermented milk in maintaining remission of ulcerative colitis, Dig. Dis. Sci. 63 (2018) 1910-1919.

[9]

R. Pastuszka, J. Barłowska, Z. Litwińczuk, Allergenicity of milk of different animal species in relation to human milk, Postepy. Hig. Med. Dosw. 70 (2016) 1451-1459. https://doi.org/10.5604/17322693.1227842.

[10]

M. Li, W. Li, J. Wu, et al., Quantitative lipidomics reveals alterations in donkey milk lipids according to lactation, Food Chem. 310 (2020) 125866. https://doi.org/10.1016/j.foodchem.2019.125866.

[11]

M. Li, Y. Liu, Q. Li, et al., Comparative exploration of free fatty acids in donkey colostrum and mature milk based on a metabolomics approach, J. Dairy Sci. 103 (2020) 6022-6031. https://doi.org/10.3168/jds.2019-17720.

[12]

S. Vincenzetti, P. Polidori, P. Mariani, et al., Donkey's milk protein fractions characterization, Food Chem. 106 (2008) 640-649. https://doi.org/10.1016/j.foodchem.2007.06.026.

[13]

F. Tidona, A. Criscione, T.G. Devold, et al., Protein composition and micelle size of donkey milk with different protein patterns: effects on digestibility, Int. Dairy J. 35 (2014) 57-62. https://doi.org/10.1016/j.idairyj.2013.10.018.

[14]

R. Derdak, S. Sakoui, O.L. Pop, et al., Insights on health and food applications of Equus asinus (donkey) milk bioactive proteins and peptides— an overview, Foods 9 (2020) 1302. https://doi.org/10.3390/foods9091302.

[15]

D. Brumini, A. Criscione, S. Bordonaro, et al., Whey proteins and their antimicrobial properties in donkey milk: a brief review, Dairy Sci. Technol. 96 (2016) 1-14. https://doi.org/10.1007/s13594-015-0246-1.

[16]

D. Brumini, C.B. Furlund, I. Comi, et al., Antiviral activity of donkey milk protein fractions on echovirus type 5, Int. Dairy J. 28 (2013) 109-111. https://doi.org/10.1016/j.idairyj.2012.08.010.

[17]

X. Mao, J. Gu, Y. Sun, et al., Anti-proliferative and anti-tumour effect of active components in donkey milk on A549 human lung cancer cells, Int. Dairy J. 19 (2009) 703-708. https://doi.org/10.1016/j.idairyj.2009.05.007.

[18]

S. Yvon, M. Olier, M. Leveque, et al., Donkey milk consumption exerts antiinflammatory properties by normalizing antimicrobial peptides levels in Paneth's cells in a model of ileitis in mice, Eur. J. Nutr. 57 (2018) 155-166.

[19]

A. Tafaro, T. Magrone, F. Jirillo, et al., Immunological properties of donkeys milk: its potential use in the prevention of atherosclerosis, Curr. Pharm. Design 13 (2007) 3711-3717. https://doi.org/10.2174/138161207783018590.

[20]

F. Lamine, J. Fioramonti, L. Bueno, et al., Nitric oxide released by lactobacillus farciminis improves TNBS-induced colitis in rats, Scand. J. Gastroenterol. 39 (2004) 37-45.

[21]

M. Li, H. Yu, J. Chen, et al., Novel insights into whey protein differences between donkey and bovine milk, Food Chem. 365 (2021) 130397. https://doi.org/10.1016/j.foodchem.2021.130397.

[22]

M.L. Molina, J. Guerrero, J.A. Cidlowski, et al., LPS regulates the expression of glucocorticoid receptor α and β isoforms and induces a selective glucocorticoid resistance in vitro, J. Inflamm. Lond Engl. 14 (2017) 22. https://doi.org/10.1186/s12950-017-0169-0.

[23]

J. Cox, M. Mann, MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification, Nat. Biotechnol. 26 (2008) 1367-1372. https://doi.org/10.1038/nbt.1511.

[24]

G.O. Consortium, The Gene Ontology (GO) database and informatics resource, Nucleic Acids Res. 32 (2004) D258-D261. https://doi.org/10.1093/nar/gkh036.

[25]

M. Kanehisa, S. Goto, KEGG: Kyoto encyclopedia of genes and genomes, Nucleic Acids Res. 28 (2000) 27-30. https://doi.org/10.1093/nar/28.1.27.

[26]

D.W. Huang, B.T. Sherman, R.A. Lempicki, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists, Nucleic Acids Res. 37 (2009) 1-13. https://doi.org/10.1093/nar/gkn923.

[27]

D.F.S. Araújo, G.C.B. Guerra, M.M.E. Pintado, et al., Intestinal antiinflammatory effects of goat whey on DNBS-induced colitis in mice, PLoS One 12 (2017) e0185382. https://doi.org/10.1371/journal.pone.0185382.

[28]

M.K. Jeengar, D. Thummuri, M. Magnusson, et al., Uridine ameliorates dextran sulfate sodium (DSS)-induced colitis in mice, Sci Rep. Uk. 7 (2017) 3924. https://doi.org/10.1038/s41598-017-04041-9.

[29]

B.T. Kurien, R.H. Scofield, Western blotting, Methods 38 (2006) 283-293. https://doi.org/10.1016/j.ymeth.2005.11.007.

[30]

J. Wang, B. Lei, J. Yan, et al., Donkey milk oligosaccharides influence the growth-related characteristics of intestinal cells and induce G2/M growth arrest via the p38 pathway in HT-29 cells, Food Funct. 10 (2019) 4823-4833. https://doi.org/10.1039/c8fo02584c.

[31]

S.H. Lee, J.E. Kwon, M.L. Cho, Immunological pathogenesis of inflammatory bowel disease, Intestinal. Res. 16 (2018) 26-42. https://doi.org/10.5217/ir.2018.16.1.26.

[32]

H. Abo, K.L. Flannigan, D. Geem, et al., Combined IL-2 Immunocomplex and Anti-IL-5 mAb treatment expands Foxp3+ Treg cells in the absence of Eosinophilia and Ameliorates experimental colitis, Front. Immunol. 10 (2019) 459. https://doi.org/10.3389/fimmu.2019.00459.

[33]

S. Wang, R. Song, Z. Wang, et al., S100A8/A9 in Inflammation, Front. Immunol. 9 (2018) 1298. https://doi.org/10.3389/fimmu.2018.01298.

[34]

D.X. Hou, T. Yanagita, T. Uto, et al., Anthocyanidins inhibit cyclooxygenase-2 expression in LPS-evoked macrophages: structure–activity relationship and molecular mechanisms involved, Biochem. Pharmacol. 70 (2005) 417-425. https://doi.org/10.1016/j.bcp.2005.05.003.

[35]

B. Chassaing, J.D. Aitken, M. Malleshappa, et al., Dextran sulfate sodium (DSS)-induced colitis in mice, Curr. Protoc. Immunol. 104 (2014) 15.25.1-15.25.14. https://doi.org/10.1002/0471142735.im1525s104.

[36]

S. Wang, P. Xia, Y. Chen, et al., Regulatory innate lymphoid cells control innate intestinal inflammation, Cell 171 (2017) 201-216.e18. https://doi.org/10.1016/j.cell.2017.07.027.

[37]

K.K. Sanchez, G.Y. Chen, A.M.P. Schieber, et al., Cooperative metabolic adaptations in the host can favor asymptomatic infection and select for attenuated virulence in an enteric pathogen, Cell 175 (2018) 146-158.e15. https://doi.org/10.1016/j.cell.2018.07.016.

[38]

X. Song, X. Sun, S.F. Oh, et al., Microbial bile acid metabolites modulate gut RORγ+ regulatory T cell homeostasis, Nature 577 (2020) 410-415. https://doi.org/10.1038/s41586-019-1865-0.

[39]

A. Bordoni, F. Danesi, D. Dardevet, et al, Dairy products and inflammation: a review of the clinical evidence, Crit. Rev. Food Sci. 57 (2015) 2497-2525. https://doi.org/10.1080/10408398.2014.967385.

[40]

J.R. Kanwar, R.K. Kanwar, S. Stathopoulos, et al., Comparative activities of milk components in reversing chronic colitis, J. Dairy Sci. 99 (2016) 2488-2501. https://doi.org/10.3168/jds.2015-10122.

[41]

E. Janssen, R.S. Geha, Primary immunodeficiencies caused by mutations in actin regulatory proteins, Immunol. Rev. 287 (2019) 121-134. https://doi.org/10.1111/imr.12716.

[42]

F. Liu, A.D. Smith, G. Solano-Aguilar, et al., Mechanistic insights into the attenuation of intestinal inflammation and modulation of the gut microbiome by krill oil using in vitro and in vivo models, Microbiome 8 (2020) 83. https://doi.org/10.1186/s40168-020-00843-8.

[43]

Y. Shimomura, E. Mizoguchi, K. Sugimoto, et al., Regulatory role of B-1 B cells in chronic colitis, Int. Immunol. 20 (2008) 729-737. https://doi.org/10.1093/intimm/dxn031.

[44]

D. Bauché, B. Joyce-Shaikh, R. Jain, et al., LAG3+ regulatory T cells restrain interleukin-23-producing CX3CR1+ gut-resident macrophages during group 3 innate lymphoid cell-driven colitis, Immunity 49 (2018) 342-352.e5. https://doi.org/10.1016/j.immuni.2018.07.007.

[45]

M.D. Giovangiulio, A. Rizzo, E. Franzè, et al., Tbet expression in regulatory T cells is required to initiate Th1-mediated colitis, Front. Immunol. 10 (2019) 2158. https://doi.org/10.3389/fimmu.2019.02158.

[46]

Y.L. Chen, Y.L. Zhang, Y.C. Dai, et al., Systems pharmacology approach reveals the antiinflammatory effects of Ampelopsis grossedentata on dextran sodium sulfate-induced colitis, World J. Gastroentero. 24 (2018) 1398-1409. https://doi.org/10.3748/wjg.v24.i13.1398.

[47]

S.L. Doyle, L.A.J. O'Neill, Toll-like rfeceptors: from the discovery of NFκB to new insights into transcriptional regulations in innate immunity, Biochem. Pharmacol. 72 (2006) 1102-1113. https://doi.org/10.1016/j.bcp.2006.07.010.

Food Science and Human Wellness
Pages 1809-1819
Cite this article:
Li M, Li Q, Abdlla R, et al. Donkey whey proteins ameliorate dextran sulfate sodium-induced ulcerative colitis in mice by downregulating the S100A8-TRAF6-NF-κB axis-mediated inflammatory response. Food Science and Human Wellness, 2023, 12(5): 1809-1819. https://doi.org/10.1016/j.fshw.2023.02.045

481

Views

63

Downloads

10

Crossref

10

Web of Science

10

Scopus

0

CSCD

Altmetrics

Received: 21 December 2021
Revised: 03 February 2022
Accepted: 03 April 2021
Published: 21 March 2023
© 2023 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