Journal Home > Volume 12 , Issue 1

Traditional Chinese preserved egg products have exhibited some anti-inflammatory effects, but their mechanisms of action remain unknown. This study aimed to investigate the anti-inflammatory effects of preserved egg white (PEW) treatment on dextran sulfate sodium (DSS)-induced colitis in mice and the underlying mechanisms. The results showed that treatment with PEW in mice with DSS-induced colitis for 14 days effectively improved the clinical signs, inhibited the secretion and gene expression of pro-inflammatory cytokines, and reduced myeloperoxidase (MPO) activity and oxidative stress levels. In addition, western blotting results showed that PEW significantly suppressed DSS-induced phosphorylation levels of nuclear factor-kappa B (NF-кB) p65 and p38 mitogen-activated protein kinase (MAPK) in colon tissues of mice with colitis. PEW also enhanced the production of short-chain fatty acids (SCFAs) and modulated gut microbiota composition in mice with DSS-induced colitis, including increasing the relative abundance of beneficial bacteria Lachnospiraceae, Ruminococcaceae and Muribaculaceae, and reducing the relative abundance of harmful bacteria Proteobacteria. Taken together, our study demonstrated that preserved egg white could alleviate DSS-induced colitis in mice through the reduction of oxidative stress, modulation of inflammatory cytokines, NF-κB, MAPK and gut microbiota composition.


menu
Abstract
Full text
Outline
About this article

Preserved egg white alleviates DSS-induced colitis in mice through the reduction of oxidative stress, modulation of infl ammatory cytokines, NF-κB, MAPK and gut microbiota composition

Show Author's information Lingyu LiaNing Qiua( )Yaqi MengaChenyan WangaYoshinori MinebRussell KeastcVincent Guyonnetd
Key Laboratory of Environment Correlative Dietology, Ministry of Education, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
Department of Food Science, University of Guelph, Guelph, Ontario N1G 2W1, Canada
CASS Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria 3125, Australia
FFI Consulting Ltd, 2488 Lyn Road, Brockville ON K6V 5T3, Canada

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

Abstract

Traditional Chinese preserved egg products have exhibited some anti-inflammatory effects, but their mechanisms of action remain unknown. This study aimed to investigate the anti-inflammatory effects of preserved egg white (PEW) treatment on dextran sulfate sodium (DSS)-induced colitis in mice and the underlying mechanisms. The results showed that treatment with PEW in mice with DSS-induced colitis for 14 days effectively improved the clinical signs, inhibited the secretion and gene expression of pro-inflammatory cytokines, and reduced myeloperoxidase (MPO) activity and oxidative stress levels. In addition, western blotting results showed that PEW significantly suppressed DSS-induced phosphorylation levels of nuclear factor-kappa B (NF-кB) p65 and p38 mitogen-activated protein kinase (MAPK) in colon tissues of mice with colitis. PEW also enhanced the production of short-chain fatty acids (SCFAs) and modulated gut microbiota composition in mice with DSS-induced colitis, including increasing the relative abundance of beneficial bacteria Lachnospiraceae, Ruminococcaceae and Muribaculaceae, and reducing the relative abundance of harmful bacteria Proteobacteria. Taken together, our study demonstrated that preserved egg white could alleviate DSS-induced colitis in mice through the reduction of oxidative stress, modulation of inflammatory cytokines, NF-κB, MAPK and gut microbiota composition.

Keywords: Gut microbiota, Oxidative stress, MAPK, NF-κB, Colitis, Preserved egg white

References(65)

[1]

M.F. Neurath, Cytokines in inflammatory bowel disease, Nat Rev Immunol. 14(5) (2014) 329-342. https://doi.org/10.1038/nri3661.

[2]

P. Hindryckx, G. Novak, N. Vande Casteele, et al., Review article: dose optimisation of infliximab for acute severe ulcerative colitis, Aliment Pharmacol Ther. 45(5) (2017) 617-630. https://doi.org/10.1111/apt.13913.

[3]

J.Z. Liu, S.V. Sommeren, H. Huang, et al., Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations, Nat Genet. 47(9) (2015) 979-986. https://doi.org/10.1038/ng.3359.

[4]

S.C. Ng, W. Tang, R.W. Leong, et al., Environmental risk factors in inflammatory bowel disease: a population-based case-control study in Asia-Pacific, Gut. 64(7) (2015) 1063-1071. http://dx.doi.org/10.1136/gutjnl-2014-307410.

[5]

L. Guidi, D. Pugliese, A. Armuzzi, Update on the management of inflammatory bowel disease: specific role of adalimumab, Clin Exp Gastroenterol. 4 (2011) 163-172. http://dx.doi.org/10.2147/CEG.S14558.

[6]

L.S. Celiberto, F.A. Graef, G.R. Healey, et al., Inflammatory bowel disease and immunonutrition: novel therapeutic approaches through modulation of diet and the gut microbiome, Immunology 155 (2018) 36-52. https://doi.org/10.1111/imm.12939.

[7]

J.H. Shin, Y.K. Lee, W.J. Shon, et al., Gut microorganisms and their metabolites modulate the severity of acute colitis in a tryptophan metabolism-dependent manner, Eur J Nutr. 59(8) (2020) 3591-3601. https://doi.org/10.1007/s00394-020-02194-4.

[8]

T. Requena, M.C. Martinez-Cuesta, C. Pelaez, Diet and microbiota linked in health and disease, Food Funct. 9(2) (2018) 688-704. https://doi.org/10.1039/C7FO01820G.

[9]

D. Gevers, S. Kugathasan, L.A. Denson, et al., The treatment-naive microbiome in new-onset Crohn's disease, Cell Host Microbe. 15(3) (2014) 382-392. http://dx.doi.org/10.1016/j.chom.2014.02.005.

[10]

J. Tan, C. McKenzie, M. Potamitis, et al., The role of short-chain fatty acids in health and disease, Adv Immunol. 121 (2014) 91-119. https://doi.org/10.1016/B978-0-12-800100-4.00003-9.

[11]

J. Wang, G. Zhu, C. Sun, et al., TAK-242 ameliorates DSS-induced colitis by regulating the gut microbiota and the JAK2/STAT3 signaling pathway, Microb Cell Fact. 19(158) (2020) 1-17. https://doi.org/10.1186/s12934-020-01417-x.

[12]

S. Kanwal, T.P. Joseph, S. Aliya, et al., Attenuation of DSS induced colitis by dictyophora indusiata polysaccharide (DIP) via modulation of gut microbiota and inflammatory related signaling pathways, J Funct Foods. 64 (2020) 103641. https://doi.org/10.1016/j.jff.2019.103641.

[13]

H. Yu, N. Qiu, Y. Meng, et al., A comparative study of the modulation of the gut microbiota in rats by dietary intervention with different sources of egg-white proteins, J Sci Food Agric. 100(9) (2020) 3622-3629. https://doi.org/10.1002/jsfa.10387.

[14]

Y. Wang, Q. Xie, Y. Zhang, et al., Combination of probiotics with different functions alleviate DSS-induced colitis by regulating intestinal microbiota, IL-10, and barrier function, Appl Microbiol Biotechnol. 104(1) (2020) 335-349. https://doi.org/10.1007/s00253-019-10259-6.

[15]

C. Wang, S. Li, K. Hong, et al., The roles of different Bacteroides fragilis strains in protecting against DSS-induced ulcerative colitis and related functional genes, Food Funct. 12(18) (2021) 8300-8313. https://doi.org/10.1039/D1FO00875G.

[16]

Y. Kobayashi, P. Rupa, J. Kovacs-Nolan, et al., Oral administration of hen egg white ovotransferrin attenuates the development of colitis induced by dextran sodium sulfate in mice, J Agric Food Chem. 63(5) (2015) 1532-1539. https://doi.org/10.1021/jf505248n.

[17]

M. Lee, J. Kovacs-Nolan, C. Yang, et al., Hen egg lysozyme attenuates inflammation and modulates local gene expression in a porcine model of dextran sodium sulfate (DSS)-induced colitis, J Agric Food Chem. 57(6) (2009) 2233-2240. https://doi.org/10.1021/jf803133b.

[18]

Y. Chen, B. Yang, R.P. Ross, et al., Orally administered CLA ameliorates DSS-induced colitis in mice via intestinal barrier improvement, oxidative stress reduction, and inflammatory cytokine and gut microbiota modulation, J Agric Food Chem. 67(48) (2019) 13282-13298. https://doi.org/10.1021/acs.jafc.9b05744.

[19]

Y. Han, M. Song, M. Gu, et al., Dietary intake of whole strawberry inhibited colonic inflammation in dextran-sulfate-sodium-treated mice via restoring immune homeostasis and alleviating gut microbiota dysbiosis, J Agric Food Chem. 67(33) (2019) 9168-9177. https://doi.org/10.1021/acs.jafc.8b05581.

[20]

H. Ge, Z. Cai, J. Chai, et al., Egg white peptides ameliorate dextran sulfate sodium-induced acute colitis symptoms by inhibiting the production of pro-inflammatory cytokines and modulation of gut microbiota composition, Food Chem. 360 (2021) 129981. https://doi.org/10.1016/j.foodchem.2021.129981.

[21]

H. Zhang, C.A. Hu, J. Kovacs-Nolan, et al., Bioactive dietary peptides and amino acids in inflammatory bowel disease, Amino Acids. 47(10) (2015) 2127-2141. https://doi.org/10.1007/s00726-014-1886-9.

[22]

K. Majumder, S. Chakrabarti, S.T. Davidge, et al., Structure and activity study of egg protein ovotransferrin derived peptides (IRW and IQW) on endothelial inflammatory response and oxidative stress, J Agric Food Chem. 61(9) (2013) 2120-2129. https://doi.org/10.1021/jf3046076.

[23]

J. Kovacs-Nolan, M. Phillips, Y. Mine, Advances in the value of eggs and egg components for human health, J Agric Food Chem. 53(22) (2005) 8421-8431. https://doi.org/10.1021/jf050964f.

[24]

J.H. Lee, S.H. Moon, H.S. Kim, et al., Antioxidant and anticancer effects of functional peptides from ovotransferrin hydrolysates, J Sci Food Agric. 97(14) (2017) 4857-4864. https://doi.org/10.1002/jsfa.8356.

[25]

H.R. Ibrahim, M.I. Hoq, T. Aoki, Ovotransferrin possesses SOD-like superoxide anion scavenging activity that is promoted by copper and manganese binding, Int J Biol Macromol. 41(5) (2007) 631-640. https://doi.org/10.1016/j.ijbiomac.2007.08.005.

[26]

Y. Zhao, Y. Yao, M. Xu, et al., Simulated gastrointestinal digest from preserved egg white exerts anti-inflammatory effects on Caco-2 cells and a mouse model of DSS-induced colitis, J Funct Foods. 35 (2017) 655-665. https://doi.org/10.1016/j.jff.2017.06.028.

[27]

M. Zhang, Y. Zhao, Y. Yao, et al., Isolation and identification of peptides from simulated gastrointestinal digestion of preserved egg white and their anti-inflammatory activity in TNF-alpha-induced Caco-2 cells, J Nutr Biochem. 63 (2019) 44-53. https://doi.org/10.1016/j.jnutbio.2018.09.019.

[28]

M. Zhang, Y. Zhao, N. Wu, et al., The anti-inflammatory activity of peptides from simulated gastrointestinal digestion of preserved egg white in DSS-induced mouse colitis, Food Funct. 9(12) (2018) 6444-6454. https://doi.org/10.1039/C8FO01939H.

[29]

R. Gao, Y. Shen, W. Shu, et al., Sturgeon hydrolysates alleviate DSS-induced colon colitis in mice by modulating NF-κB, MAPK, and microbiota composition, Food Funct. 11(8) (2020) 6987-6999. https://doi.org/10.1039/C9FO02772F.

[30]

H. Cui, Y. Cai, L. Wang, et al., Berberine regulates Treg/Th17 balance to treat ulcerative colitis through modulating the gut microbiota in the colon, Front Pharmacol. 9 (2018) 571. https://doi.org/10.3389/fphar.2018.00571.

[31]

C. Quast, E. Pruesse, P. Yilmaz, et al., The SILVA ribosomal RNA gene database project: improved data processing and web-based tools, Nucleic Acids Res. 41(D1) (2013) D590-D596. https://doi.org/10.1093/nar/gks1219.

[32]

Z. Xu, W. Chen, Q. Deng, et al., Flaxseed oligosaccharides alleviate DSS-induced colitis through modulation of gut microbiota and repair of the intestinal barrier in mice, Food Funct. 11(9) (2020) 8077-8088. https://doi.org/10.1039/D0FO01105C.

[33]

A.K. Pandurangan, S. Ismail, Z. Saadatdoust, et al., Allicin alleviates dextran sodium sulfate- (DSS-) induced ulcerative colitis in BALB/c mice, Oxid Med Cell Longev. 2015 (2015) 1-13. https://doi.org/10.1155/2015/605208.

[34]

D. Muthas, A. Reznichenko, C.A. Balendran, et al., Neutrophils in ulcerative colitis: a review of selected biomarkers and their potential therapeutic implications, Scand J Gastroenterol. 52(2) (2017) 125-135. https://doi.org/10.1080/00365521.2016.1235224.

[35]

M. Wu, P. Li, Y. An, et al., Phloretin ameliorates dextran sulfate sodium-induced ulcerative colitis in mice by regulating the gut microbiota, Pharmacol Res. 150 (2019) 104489. https://doi.org/10.1016/j.phrs.2019.104489.

[36]

M. Guo, Z. Li, Polysaccharides isolated from Nostoc commune Vaucher inhibit colitis-associated colon tumorigenesis in mice and modulate gut microbiota, Food Funct. 10 (2019) 6873-6881. https://doi.org/10.1039/C9FO00296K.

[37]

Z. Gu, Y. Zhu, S. Jiang, et al., Tilapia head glycolipids reduce inflammation by regulating the gut microbiota in dextran sulphate sodium-induced colitis mice, Food Funct. 11(4) (2020) 3245-3255. https://doi.org/10.1039/D0FO00116C.

[38]

X. Shao, C. Sun, X. Tang, et al., Anti-inflammatory and intestinal microbiota modulation properties of Jinxiang Garlic (Allium sativum L.) polysaccharides toward dextran sodium sulfate-induced colitis, J Agric Food Chem. 68(44) (2020) 12295-12309. https://doi.org/10.1021/acs.jafc.0c04773.

[39]

Y. Shi, P. Rupa, B. Jiang, et al., Hydrolysate from eggshell membrane ameliorates intestinal inflammation in mice, Int J Mol Sci. 15(12) (2014) 22728-22742. https://doi.org/10.3390/ijms151222728.

[40]

F. Li, Y. Han, X. Cai, et al., Dietary resveratrol attenuated colitis and modulated gut microbiota in dextran sulfate sodium-treated mice, Food Funct. 11(1) (2020) 1063-1073. https://doi.org/10.1039/C9FO01519A.

[41]

Y. Peng, Y. Shi, H. Zhang, et al., Anti-inflammatory and anti-oxidative activities of daidzein and its sulfonic acid ester derivatives, J Funct Foods. 35 (2017) 635-640. https://doi.org/10.1016/j.jff.2017.06.027.

[42]

B. Li, R. Alli, P. Vogel, et al., IL-10 modulates DSS-induced colitis through a macrophage-ROS-NO axis, Mucosal Immunol. 7(4) (2014) 869-878. https://doi.org/10.1038/mi.2013.103.

[43]

X. Wang, N. Yu, Z. Wang, et al., Akebia trifoliata pericarp extract ameliorates inflammation through NF-kappaB/MAPK signaling pathways and modifies gut microbiota, Food Funct. 11(5) (2020) 4682-4696. https://doi.org/10.1039/C9FO02917F.

[44]

H. Zhang, C.N. Xu, Y. Mine, Synthetic phosphoserine dimer attenuates lipopolysaccharide‐induced inflammatory response in human intestinal epithelial cells via activation of NF‐κB and MAPKs cell signalling pathways, INT J FOOD SCI TECH. 55(1) (2019) 82-91. https://doi.org/10.1111/ijfs.14246.

[45]

A. Sharma, N.V. Tirpude, M. Kumari, et al., Rutin prevents inflammation-associated colon damage via inhibiting the p38/MAPKAPK2 and PI3K/Akt/GSK3beta/NF-kappaB signalling axes and enhancing splenic Tregs in DSS-induced murine chronic colitis, Food Funct. 12(18) (2021) 8492-8506. https://doi.org/10.1039/D1FO01557E.

[46]

C.W. Espelin, A. Goldsipe, P.K. Sorger, et al., Elevated GM-CSF and IL-1beta levels compromise the ability of p38 MAPK inhibitors to modulate TNF-alpha levels in the human monocytic/macrophage U937 cell line, Mol Biosyst. 6(10) (2010) 1956-1972. https://doi.org/10.1039/C002848G.

[47]

L. Lian, S. Zhang, Z. Yu, et al., The dietary freeze-dried fruit powder of actinidia arguta ameliorates dextran sulphate sodium-induced ulcerative colitis in mice by inhibiting the activation of MAPKs, Food Funct. 10(9) (2019) 5768-5778. https://doi.org/10.1039/C9FO00664H.

[48]

Y. Ren, Y. Geng, Y. Du, et al., Polysaccharide of hericium erinaceus attenuates colitis in C57BL/6 mice via regulation of oxidative stress, inflammation-related signaling pathways and modulating the composition of the gut microbiota, J Nutr Biochem. 57 (2018) 67-76. https://doi.org/10.1016/j.jnutbio.2018.03.005.

[49]

G. Wang, Y. Liu, Z. Lu, et al., The ameliorative effect of a Lactobacillus strain with good adhesion ability against dextran sulfate sodium-induced murine colitis, Food Funct. 10(1) (2019) 397-409. https://doi.org/10.1039/C8FO01453A.

[50]

C. Hernandez-Chirlaque, C.J. Aranda, B. Ocon, et al., Germ-free and antibiotic-treated mice are highly susceptible to epithelial injury in DSS colitis, J Crohns Colitis. 10(11) (2016) 1324-1335. https://doi.org/10.1093/ecco-jcc/jjw096.

[51]

L. Zhu, L.Z. Xu, S. Zhao, et al., Protective effect of baicalin on the regulation of Treg/Th17 balance, gut microbiota and short-chain fatty acids in rats with ulcerative colitis, Appl Microbiol Biotechnol. 104(12) (2020) 5449-5460. https://doi.org/10.1007/s00253-020-10527-w.

[52]

X. Zhu, S. Xiang, X. Feng, et al., Impact of cyanocobalamin and methylcobalamin on inflammatory bowel disease and the intestinal microbiota composition, J Agric Food Chem. 67(3) (2019) 916-926. https://pubs.acs.org/doi/10.1021/acs.jafc.8b05730.

[53]

J. Zhang, Z. Guo, Z. Xue, et al., A phylo-functional core of gut microbiota in healthy young Chinese cohorts across lifestyles, geography and ethnicities, ISME J. 9(9) (2015) 1979-1990. https://doi.org/10.1038/ismej.2015.11.

[54]

X. Liu, B. Mao, J. Gu, et al., Blautia-a new functional genus with potential probiotic properties?, Gut Microbes 13(1) (2021) 1-21. https://doi.org/10.1080/19490976.2021.1875796.

[55]

A.E. Reeves, M.J. Koenigsknecht, I.L. Bergin, et al., Suppression of clostridium difficile in the gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family Lachnospiraceae, Infection and Immunity 80(11) (2012) 3786-3794. https://doi.org/10.1128/IAI.00647-12.

[56]

I. Lagkouvardos, T.R. Lesker, T.C.A. Hitch, et al., Sequence and cultivation study of Muribaculaceae reveals novel species, host preference, and functional potential of this yet undescribed family, Microbiome 7(28) (2019) 1-15. https://doi.org/10.1186/s40168-019-0637-2.

[57]

M.A. Vinolo, H.G. Rodrigues, R.T. Nachbar, et al., Regulation of inflammation by short chain fatty acids, Nutrients 3(10) (2011) 858-876. https://doi.org/10.3390/nu3100858.

[58]

K.E. Bach Knudsen, H.N. Laerke, M.S. Hedemann, et al., Impact of diet-modulated butyrate production on intestinal barrier function and inflammation, Nutrients 10(10) (2018) 1499. https://doi.org/10.3390/nu10101499.

[59]

D. Lee, F. Bamdad, K. Khey, et al., Antioxidant and anti-inflammatory properties of chicken egg vitelline membrane hydrolysates, Poult Sci. 96(9) (2017) 3510-3516. https://doi.org/10.3382/ps/pex125.

[60]

C. Xu, C. Yang, Y. Yin, et al., Phosphopeptides (PPPs) from hen egg yolk phosvitin exert anti-inflammatory activity via modulation of cytokine expression, J Funct Foods. 4(4) (2012) 718-726. https://doi.org/10.1016/j.jff.2012.04.011.

[61]

D. Young, F. Nau, M. Pasco, et al., Identification of hen egg yolk-derived phosvitin phosphopeptides and their effects on gene expression profiling against oxidative stress-induced Caco-2 cells, J Agric Food Chem. 59(17) (2011) 9207-9218. https://doi.org/10.1021/jf202092d.

[62]

N. Ding, C. Mao, Z. Cai, et al., Anti-inflammatory effect of preserved egg with simulated gastrointestinal digestion on LPS-stimulated RAW264.7 cells, Poult Sci. 98(10) (2019) 4401-4407. https://doi.org/10.3382/ps/pez243.

[63]

B. Hernández-Ledesma, A. Dávalos, B. Bartolomé, et al., Preparation of antioxidant enzymatic hydrolysates from α-lactalbumin and β-lactoglobulin. Identification of active peptides by HPLC-MS/MS, J Agric Food Chem. 53(3) (2005) 588-593. https://doi.org/10.1021/jf048626m.

[64]

Y. Meng, C. Chen, N. Qiu, et al., Modulation of gut microbiota in rats fed whole egg diets by processing duck egg to preserved egg, J Biosci Bioeng. 130(1) (2020) 54-62. https://doi.org/10.1016/j.jbiosc.2020.02.015.

[65]

A.M. Davila, F. Blachier, M. Gotteland, et al., Re-print of “Intestinal luminal nitrogen metabolism: role of the gut microbiota and consequences for the host”, Pharmacol Res. 69(1) (2013) 114-126. https://doi.org/10.1016/j.phrs.2012.11.005.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 06 October 2021
Revised: 15 October 2021
Accepted: 06 November 2021
Published: 09 August 2022
Issue date: January 2023

Copyright

© 2023 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

Acknowledgements

This work was financially supported by the Chinese National Natural Science Funds (31772043), and the Fundamental Research Funds for the Central Universities (2662018JC021).

Rights and permissions

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

Return