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 (892.1 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Differentially expressed whey proteins of donkey and bovine colostrum revealed with a label-free proteomics approach

Mohan Lia,1Qilong Lib,1Haikun YuaXiumin ZhangcDehao LidWanying SongaYan Zhenga( )Xiqing Yuea( )
College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
Beijing Academy of Food Sciences, Beijing 100068, China
The Fourth Affiliated Hospital of China Medical University, Shenyang 110866, China

1 These authors contributed equally to this work and should be regarded as co-first author.Peer review under responsibility of KeAi Communications Co., Ltd.]]>

Show Author Information

Abstract

This study aimed to analyze and compare the differentially expressed whey proteins (DEWPs) of donkey and bovine colostrum using high-performance liquid chromatography with tandem mass spectrometry-based proteomics. A total of 620 and 696 whey proteins were characterized in the donkey and bovine colostrum, respectively, including 383 common whey proteins. Among these common proteins, 80 were identified as DEWPs, including 21 upregulated and 59 downregulated DEWPs in donkey colostrum compared to bovine colostrum. Gene Ontology analysis revealed that these DEWPs were mainly related to cellular components, such as extracellular exosome, plasma membrane, and mitochondrion; biological processes, such as oxidation-reduction process, cell-cell adhesion, and small guanosine triphosphate (GTP) ase-mediated signal transduction; and molecular functions, such as GTP binding, GTPase activity, and soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor activity. Metabolic pathway analysis suggested that the majority of the DEWPs were associated with soluble NSF factor attachment protein receptor interactions in vesicular transport, fatty acid biosynthesis, and estrogen signaling pathways. Our results provide a vital insight into the differences between donkey and bovine colostrum, along with important information on the significant components as nutritional and functional factors to be included in infant formula based on multiple milk sources.

References

[1]

O. Ballard, A.L. Morrow, Human milk composition, Pediatr. Clin. N. Am. 60 (2013) 49-74. https://doi.org/10.1016/j.pcl.2012.10.002.

[2]

M. Li, Q. Li, Y. Zheng, et al., New insights into the alterations of full spectrum amino acids in human colostrum and mature milk between different domains based on metabolomics, Eur. Food Res. Technol. 246 (2020) 1119-1128. https://doi.org/10.1007/s00217-020-03470-7.

[3]

K. Stelwagen, E. Carpenter, B. Haigh, et al., Immune components of bovine colostrum and milk, J. Anim. Sci. 87 (2009) 3-9. https://doi.org/10.2527/jas.2008-1377.

[4]

M. Rathe, K. Müller, P.T. Sangild, et al., Clinical applications of bovine colostrum therapy: a systematic review, Nutr. Rev. 72 (2014) 237-254. https://doi.org/10.1111/nure.12089.

[5]

B.A. McGrath, P.F. Fox, P.L.H. McSweeney, et al., Composition and properties of bovine colostrum: a review, Dairy Sci. Technol. 96 (2015) 133-158. https://doi.org/10.1007/s13594-015-0258-x.

[6]

M. Li, W. Li, F. Kong, et al., Metabolomics methods to analyze full spectrum of amino acids in different domains of bovine colostrum and mature milk, Eur. Food Res. Technol. 246 (2019) 213-224. https://doi.org/10.1007/s00217-019-03385-y.

[7]

L.H. Ulfman, J.H.W. Leusen, H.F.J. Savelkoul, et al., Effects of bovine immunoglobulins on immune function, allergy, and infection, Front. Nutr. 5(2018) 52. https://doi.org/10.3389/fnut.2018.00052.

[8]

I.E. Filipescu, L. Leonardi, L. Menchetti, et al., Preventive effects of bovine colostrum supplementation in TNBS-induced colitis in mice, PLoS One 13(2018) e0202929. https://doi.org/10.1371/journal.pone.0202929.

[9]

S.H. Barakat, M.A. Meheissen, O.M. Omar, et al., Bovine colostrum in the treatment of acute diarrhea in children: a double-blinded randomized controlled trial, J. Trop. Pediatrics 66 (2020) 46-55. https://doi.org/10.1093/tropej/fmz029.

[10]

W.R.D. Duff, P.D. Chilibeck, J.J. Rooke, et al., The effect of bovine colostrum supplementation in older adults during resistance training, Int. J. Sport Nutr. Exe. 24 (2014) 276-285. https://doi.org/10.1123/ijsnem.2013-0182.

[11]

M.C. Vieira, M.B. Morais, J.V. Spolidoro, et al., A survey on clinical presentation and nutritional status of infants with suspected cow' milk allergy, BMC Pediatr. 10 (2010) 25. https://doi.org/10.1186/1471-2431-10-25.

[12]

Y. Ma, L. Zhang, Y. Wu, et al., Changes in milk fat globule membrane proteome after pasteurization in human, bovine and caprine species, Food Chem. 279 (2018) 209-215. https://doi.org/10.1016/j.foodchem.2018.12.015.

[13]

A. Høst, Frequency of cow's milk allergy in childhood, Ann. Allergy Asthma. Immunol. 89 (2002) 33-37. https://doi.org/10.1016/s1081-1206(10)62120-5.

[14]

A. Murgia, P. Scano, M. Contu, et al., Characterization of donkey milk and metabolite profile comparison with human milk and formula milk, LWT-Food Sci. Technol. 74 (2016) 427-433. https://doi.org/10.1016/j.lwt.2016.07.070.

[15]

M. Li, S. Kang, Y. Zheng, et al., Comparative metabolomics analysis of donkey colostrum and mature milk using ultra-high-performance liquid tandem chromatography quadrupole time-of-flight mass spectrometry, J. Dairy Sci. 103 (2020) 992-1001. https://doi.org/10.3168/jds.2019-17448.

[16]

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

[17]

H.Y. Guo, K. Pang, X.Y. Zhang, et al., Composition, physiochemical properties, nitrogen fraction distribution, and amino acid profile of donkey milk, J. Dairy Sci. 90 (2007) 1635-1643. https://doi.org/10.3168/jds.2006-600.

[18]

M.O. Swar, Donkey milk-based formula: a substitute for patients with cow's milk protein allergy, Sudan J. Paediatr. 11 (2011) 21-24.

[19]

W.L. Hurley, P.K. Theil, Perspectives on immunoglobulins in colostrum and milk, Nutrients 3 (2011) 442-474. https://doi.org/10.3390/nu3040442.

[20]

P.C. Pereira, Milk nutritional composition and its role in human health, Nutrition 30 (2014) 619-627. https://doi.org/10.1016/j.nut.2013.10.011.

[21]

A.R. Madureira, C.I. Pereira, A.M.P. Gomes, et al., Bovine whey proteins -overview on their main biological properties, Food Res. Int. 40 (2007) 1197-1211. https://doi.org/10.1016/j.foodres.2007.07.005.

[22]

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.

[23]

X.Y. Zhang, L. Zhao, L. Jiang, et al., The antimicrobial activity of donkey milk and its microflora changes during storage, Food Control 19 (2008)1191-1195. https://doi.org/10.1016/j.foodcont.2008.01.005.

[24]

Q. Li, M. Li, J. Zhang, et al., Donkey milk inhibits triple-negative breast tumor progression and is associated with increased cleaved-caspase-3 expression, Food Funct. 11 (2020) 3053-3065. https://doi.org/10.1039/c9fo02934f.

[25]

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.

[26]

M. Shariatikia, M. Behbahani, H. Mohabatkar, Anticancer activity of cow, sheep, goat, mare, donkey and camel milks and their caseins and whey proteins and in silico comparison of the caseins, Mol. Biology Res. Commun. 6 (2017) 57-64.

[27]

L. Chianese, M.G. Calabrese, P. Ferranti, et al., Proteomic characterization of donkey milk "caseome, " J. Chromatogr. A 1217 (2010) 4834-4840.https://doi.org/10.1016/j.chroma.2010.05.017.

[28]

X. Zhang, H. Li, J. Yu, et al., Label-free based comparative proteomic analysis of whey proteins between different milk yields of Dezhou donkey, Biochem. Bioph. Res. Commun. 508 (2018) 237-242.https://doi.org/10.1016/j.bbrc.2018.11.130.

[29]

W. Li, M. Li, X. Cao, et al., Comparative analysis of whey proteins in donkey colostrum and mature milk using quantitative proteomics, Food Res.Int. 127 (2019) 108741. https://doi.org/10.1016/j.foodres.2019.108741.

[30]

W. Li, M. Li, X. Cao, et al., Quantitative proteomic analysis of milk fat globule membrane (MFGM) proteins from donkey colostrum and mature milk, Food Funct. 10 (2019) 4256-4268. https://doi.org/10.1039/c9fo00386j.

[31]

T.G.O. Consortium, The Gene Ontology project in 2008, Nucleic. Acids Res.36 (2008) D440-D444. https://doi.org/10.1093/nar/gkm883.

[32]

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.

[33]

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.

[34]

Y.J. Wang, J.X. Xiao, S. Li, et al., Protein metabolism and signal pathway regulation in rumen and mammary gland, Curr. Protein Pept. Sci. 18 (2017)636-651. https://doi.org/10.2174/1389203717666160627075021.

[35]

C.K. Reynolds, N.B. Kristensen, Nitrogen recycling through the gut and the nitrogen economy of ruminants: an asynchronous symbiosis1, J. Anim. Sci.86 (2008) E293-E305. https://doi.org/10.2527/jas.2007-0475.

[36]

M. Li, Q. Li, S. Kang, et al., Characterization and comparison of lipids in bovine colostrum and mature milk based on UHPLC-QTOF-MS lipidomics, Food Res Int. 136 (2020) 109490. https://doi.org/10.1016/j.foodres.2020.109490.

[37]

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.

[38]

M. Li, W. Li, J. Wu, et al., Quantitative lipidomics reveals alterations in donkey milk lipids according to lactation, Food Chem. 310 (2019) 125866.https://doi.org/10.1016/j.foodchem.2019.125866.

[39]

X. Zou, J. Huang, Q. Jin, et al., Lipid composition analysis of milk fats from different mammalian species: potential for use as human milk fat substitutes, J. Agr. Food Chem. 61 (2013) 7070-7080. https://doi.org/10.1021/jf401452y.

[40]

P.F. Fox, T. Uniacke-Lowe, P.L.H. McSweeney, et al., Enzymology of Milk and Milk Products, Dairy Chemistry and Biochemistry, 2015, pp. 377-414.https://doi.org/10.1007/978-3-319-14892-2.

[41]

L.W. Xiang, L.D. Melton, I.K.H. Leung, Reference module in food science, Mycotoxins (2018) 560-565. https://doi.org/10.1016/b978-0-08-100596-5.21488-1.

[42]

G. Kontopidis, C. Holt, L. Sawyer, Invited review: β-lactoglobulin: binding properties, structure, and function, J. Dairy Sci. 87 (2004) 785-796.https://doi.org/10.3168/jds.s0022-0302(04)73222-1.

[43]

L. Sawyer, G. Kontopidis, The core lipocalin, bovine β-lactoglobulin, Biochimica. Et. Biophysica. Acta. Bba. - Protein Struct. Mol. Enzym. 1482(2000) 136-148. https://doi.org/10.1016/s0167-4838(00)00160-6.

[44]

F. Roth-Walter, L.F. Pacios, C. Gomez-Casado, et al., The major cow milk allergen Bos d 5 manipulates T-helper cells depending on its load with siderophore-bound iron, PLoS One 9 (2014) e104803.https://doi.org/10.1371/journal.pone.0104803.

[45]

G.A. Perkins, B. Wagner, The development of equine immunity: current knowledge on immunology in the young horse, Equine. Vet. J. 47 (2015)267-274. https://doi.org/10.1111/evj.12387.

[46]

M. Yáñez-Mó, P.R.M. Siljander, Z. Andreu, et al., Biological properties of extracellular vesicles and their physiological functions, J. Extracell Vesicles.4 (2015) 27066. https://doi.org/10.3402/jev.v4.27066.

[47]

G. Raposo, W. Stoorvogel, Extracellular vesicles: exosomes, microvesicles, and friends, J. Cell Biology. 200 (2013) 373-383. https://doi.org/10.1083/jcb.201211138.

[48]

M. Witkowska-Zimny, E. Kaminska-El-Hassan, Cells of human breast milk, Cell Mol. Biol. Lett. 22 (2017) 11. https://doi.org/10.1186/s11658-017-0042-4.

[49]

E.T. Kavalali, SNARE interactions in membrane trafficking: a perspective from mammalian central synapses, Bioessays 24 (2002) 926-936.https://doi.org/10.1002/bies.10165.

[50]

R.J. Kusuma, S. Manca, T. Friemel, et al., Human vascular endothelial cells transport foreign exosomes from cow's milk by endocytosis, Am. J. Physiol-Cell Ph. 310 (2016) C800-C807. https://doi.org/10.1152/ajpcell.00169.2015.

[51]

T. Wolf, S.R. Baier, J. Zempleni, The intestinal transport of bovine milk exosomes is mediated by endocytosis in human colon carcinoma Caco-2 cells and rat small intestinal IEC-6 Cells, J. Nutr. 145 (2015) 2201-2206.https://doi.org/10.3945/jn.115.218586.

[52]

P.R. Pryor, B.M. Mullock, N.A. Bright, et al., Combinatorial SNARE complexes with VAMP7 or VAMP8 define different late endocytic fusion events, Embo. Rep. 5 (2004) 590-595. https://doi.org/10.1038/sj.embor.7400150.

[53]

C.C. Wang, H. Shi, K. Guo, et al., VAMP8/Endobrevin as a general vesicular SNARE for regulated exocytosis of the exocrine system, Mol. Biol.Cell. 18 (2007) 1056-1063. https://doi.org/10.1091/mbc.e06-10-0974.

Food Science and Human Wellness
Pages 1224-1231
Cite this article:
Li M, Li Q, Yu H, et al. Differentially expressed whey proteins of donkey and bovine colostrum revealed with a label-free proteomics approach. Food Science and Human Wellness, 2023, 12(4): 1224-1231. https://doi.org/10.1016/j.fshw.2022.10.004

702

Views

52

Downloads

15

Crossref

14

Web of Science

14

Scopus

0

CSCD

Altmetrics

Received: 03 January 2021
Revised: 09 February 2021
Accepted: 07 March 2021
Published: 18 November 2022
© 2023 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

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

Return