Journal Home > Volume 1 , Issue 2

Whey protein, an essential component of milk, possesses a rich repertoire of bioactive proteins with unique nutritional and functional properties. Employing liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS), we conducted a comparative analysis of whey proteins derived from enzyme curd Esprion300HS (E300HS) and acid curd Textrion PROGEL 800 (TP800) methods. Our investigation identified 216 proteins, of which 32 exhibited differential expression between E300HS and TP800. Notably, E300HS displayed significantly higher levels of calcium-binding protein, lactoperoxidase, and lactadherin compared to TP800. Peptide profiling revealed 162 peptides, with 12 upregulated and 14 downregulated in E300HS relative to TP800. Furthermore, E300HS exhibited abundant lactoferrin-derived peptides, while TP800 displayed a higher prevalence of casein-like peptides. These proteins and peptides encompass a broad range of biological functions, including anticancer, antimicrobial, and bone remodeling activities. Our findings underscore the vast potential of whey protein in food industry applications and its significance in cancer research, bone development, and age-related degenerative diseases.


menu
Abstract
Full text
Outline
About this article

Differences in proteomic and peptide profiles of whey protein by acid curd and enzyme curd process from bovine milk

Show Author's information He Li1,2Zheng Li2Ying Ma1( )
School of Medicine and Health, Harbin Institute of Technology, Harbin 150090, China
Jiangsu Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, China

Abstract

Whey protein, an essential component of milk, possesses a rich repertoire of bioactive proteins with unique nutritional and functional properties. Employing liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS), we conducted a comparative analysis of whey proteins derived from enzyme curd Esprion300HS (E300HS) and acid curd Textrion PROGEL 800 (TP800) methods. Our investigation identified 216 proteins, of which 32 exhibited differential expression between E300HS and TP800. Notably, E300HS displayed significantly higher levels of calcium-binding protein, lactoperoxidase, and lactadherin compared to TP800. Peptide profiling revealed 162 peptides, with 12 upregulated and 14 downregulated in E300HS relative to TP800. Furthermore, E300HS exhibited abundant lactoferrin-derived peptides, while TP800 displayed a higher prevalence of casein-like peptides. These proteins and peptides encompass a broad range of biological functions, including anticancer, antimicrobial, and bone remodeling activities. Our findings underscore the vast potential of whey protein in food industry applications and its significance in cancer research, bone development, and age-related degenerative diseases.

Keywords: peptide, whey protein, proteomic, active ingredient

References(41)

[1]

M. D. Pendon, V. Jose, Jr. Romanin, et al., A biorefinery concept for the production of fuel ethanol, probiotic yeast, and whey protein from a by-product of the cheese industry, Appl. Microbiol. Biotechnol. 105 (2021) 3859–3871. https://doi.org/10.1007/s00253-021-11278-y.

[2]

J. Giraldo, C. Igualada, R. Cabizza, et al., Transfer of antibiotics from goat’s milk to rennet curd and whey fractions during cheese-making, Food Chem. 392 (2022) 133218. https://doi.org/10.1016/j.foodchem.2022.133218.

[3]

A. Vasiliauskaite, J. Mileriene, E. Songisepp, et al., Application of edible coating based on liquid acid whey protein concentrate with indigenous Lactobacillus helveticus for acid-curd cheese quality improvement, Foods 21 (2022) 3353. https://doi.org/10.3390/foods11213353.

[4]

S. S. Li, L. Ling, Q. K. Zeng, et al., Quantitative differences in whey proteins among Murrah, Nili-Ravi and Mediterranean buffaloes using a TMT proteomic approach, Food Chem. 269 (2018) 228–235. https://doi.org/10.1016/j.foodchem.2018.06.122.

[5]

R. J. S. D. Castro, A. Ohara, P. K. Okuro, et al., Whey protein as a key component in food systems: physicochemical properties, production technologies and applications, Food Struct. 14 (2017) 17–29. https://doi.org/10.1016/j.foostr.2017.05.004.

[6]

M. Yang, X. Cao, R. Wu, et al., Comparative proteomic exploration of whey proteins in human and bovine colostrum and mature milk using iTRAQ-coupled LC-MS/MS, Int. J. Food Sci. Nutr. 68 (2017) 671–681. https://doi.org/10.1080/09637486.2017.1279129.

[7]

W. Zhong, J. Li, J. Dai, et al., Digestibility of polymerized whey protein using in vitro digestion model and antioxidative property of its hydrolysate, Food Biosci. 42 (2021) 101109. https://doi.org/10.1016/j.fbio.2021.101109.

[8]

S. Gilmartin, N. O’Brien, L. Giblin, et al., Whey for sarcopenia; can whey peptides, hydrolysates or proteins play a beneficial role?, Foods 9 (2020) 750. https://doi.org/10.3390/foods9060750.

[9]
H. R. Chen, X. F. Qi, K. F. Guan, et al., Peptides released from bovine α-lactalbumin by simulated digestion alleviated free fatty acids-induced lipid accumulation in HepG2 cells, J. Funct. Foods 85 (2021) 104618. https://doi.org/10.1016/j.jff.2021.104618.
DOI
[10]

X. F. Qi, H. R. Chen, K. F. Guan, et al., Identification, inhibitory mechanism and transepithelial transport of xanthine oxidase inhibitory peptides from whey protein after simulated gastrointestinal digestion and intestinal absorption, Food Res. Int. 162 (2022) 111959. https://doi.org/10.1016/j.foodres.2022.111959.

[11]

F. Arrutia, R. Rubio, F. A. Riera, Production and membrane fractionation of bioactive peptides from a whey protein concentrate, J. Food Eng. 184 (2016) 1–9. https://doi.org/10.1016/j.jfoodeng.2016.03.010.

[12]

H. Li, K. F. Guan, M. Liu, et al., Novel antioxidant peptides from MFGM protein hydrolysates: separation, identification and effects on dexamethasone-induced mitochondrial dysfunction, Food Chem. 403 (2023) 134473. https://doi.org/10.1016/j.foodchem.2022.134473.

[13]
J. Théolier, R. Hammami, P. Labelle, et al., Isolation and identification of antimicrobial peptides derived by peptic cleavage of whey protein isolate, J. Funct. Foods 5 (2013) 706–714. https://doi.org/10.1016/j.jff.2013.01.014.
DOI
[14]

Y. Yang, D. Bu, X. Zhao, et al., Proteomic analysis of cow, yak, buffalo, goat and camel milk whey proteins: quantitative differential expression patterns, J. Proteome Res. 12 (2013) 1660. https://doi.org/10.1021/pr301001m.

[15]
K. F. Guan, H. Li, H. R. Chen, et al., TMT-based quantitative proteomics analysis reveals the effect of bovine derived MFG-E8 against oxidative stress on rat L6 cells, Food Funct. 12 (2021) 7310–7320. https://doi.org/10.1039/d1fo01135a.
DOI
[16]
H. Li, K. F. Guan, X. Li, et al., MFG-E8 induced differences in proteomic profiles in mouse C2C12 cells and its effect on PI3K/Akt and ERK signal pathways, Int. J. Biol. Macromol. 124 (2019) 681–688. https://doi.org/10.1016/j.ijbiomac.2018.11.265.
DOI
[17]
A. R. Corrochano, E. Arranz, E. D. Noni, et al., Intestinal health benefits of bovine whey proteins after simulated gastrointestinal digestion, J. Funct. Foods 49 (2018) 526–535. https://doi.org/10.1016/j.jff.2018.08.043.
DOI
[18]

S. Pisanu, D. Pagnozzi, M. Pes, et al., Differences in the peptide profile of raw and pasteurised ovine milk cheese and implications for its bioactive potential, Int. Dairy J. 42 (2015) 26–33. https://doi.org/10.1016/j.idairyj.2014.10.007.

[19]
N. Urtasun, M. F. Baieli, D. B. Hirsch, et al., Lactoperoxidase purification from whey by using dye affinity chromatography, Food Bioprod. Process 103 (2017) 58–65. https://doi.org/10.1016/j.fbp.2017.02.011.
DOI
[20]

J. Carlsson, Catalatic activity of lactoperoxidase in the presence of SCN, Biochem. Biophys. Res. Commun. 116 (1983) 568–573. https://doi.org/10.1016/0006-291x(83)90561-2.

[21]

I. Pascal, G. Juliane, K. Katharina, et al., Class switch toward noninflammatory, spike-specific IgG4 antibodies after repeated SARS-CoV-2 mRNA vaccination, Sci. Immunol. 79 (2023) 2798. https://doi.org/10.1126/sciimmunol.ade2798.

[22]

H. Ohnuki, A. Mizutani, H. Otani, et al., Oral ingestion of cow’s milk immunoglobulin G stimulates some cellular immune systems and suppresses humoral immune responses in mouse, Int. Immunopharmacol. 6 (2006) 1315–1322. https://doi.org/10.1016/j.intimp.2006.04.006.

[23]

B. H. Sliker, B. T. Goetz, H. L. Peters, et al., Beta 2-microglobulin regulates amyloid precursor-like protein 2 expression and the migration of pancreatic cancer cells, Cancer Biol. Ther. 20 (2019) 931–940. https://doi.org/10.1080/15384047.2019.1580414.

[24]

P. H. Anborgh, J. C. Mutrie, A. B. Tuck, et al., Pre- and post-translational regulation of osteopontin in cancer, J. Cell Commun. Signal. 5 (2011) 111–122. https://doi.org/10.1007/s12079-011-0130-6.

[25]

L. Liu, Q. Luo, J. Sun, et al., Decreased nuclear stiffness via FAK-ERK1/2 signaling is necessary for osteopontin-promoted migration of bone marrow-derived mesenchymal stem cells, Exp. Cell Res. 355 (2017) 172–181. https://doi.org/10.1016/j.yexcr.2017.04.004.

[26]

S. J. Lee, S. E. Baek, M. A. Jang, et al., Osteopontin plays a key role in vascular smooth muscle cell proliferation via EGFR-mediated activation of AP-1 and C/EBPβ pathways, Pharmacol. Res. 108 (2016) 1–8. https://doi.org/10.1016/j.phrs.2016.03.041.

[27]
Y. C. Fong, S. C. Liu, C. Y. Huang, et al., Osteopontin increases lung cancer cells migration via activation of the αvβ3 integrin/FAK/Akt and NF-κB-dependent pathway, Lung Cancer 64 (2009) 263–270. https://doi.org/10.1016/j.lungcan.2008.09.003.
DOI
[28]
S. Li, J. Sheng, J. K. Hu, et al., Ribonuclease 4 protects neuron degeneration by promoting angiogenesis, neurogenesis, and neuronal survival under stress, Angiogenesis 16 (2013) 387–404. https://doi.org/10.1007/s10456-012-9322-9.
DOI
[29]

X. Gao, Z. Xu, Mechanisms of action of angiogenin, Acta. Biochim. Biophys. Sin. 40 (2008) 619–624. https://doi.org/10.1111/j.1745-7270.2008.00442.x.

[30]
L. Zhou, W. Ma, F. Zhang, et al., VEGF165 and angiopoietin-1 decreased myocardium infarct size through phosphatidylinositol-3 kinase and Bcl-2 pathways, Gene Ther. 12 (2005) 196–202. https://doi.org/10.1038/sj.gt.3302416.
DOI
[31]

W. Xiao, S. Qi, H. Wei, Carvedilol protects against the H2O2-induced cell damages in rat myoblasts by regulating the Circ_NFIX/miR-125b-5p/TLR4 signal axis, J. Cardiovasc. Pharmacol. 78 (2021) 604–614. https://doi.org/10.1097/FJC.0000000000001095.

[32]
E. S. Ji, S. J. Park, S. I. Ahn, et al., Soluble whey protein hydrolysate ameliorates muscle atrophy induced by immobilization via regulating the PI3K/Akt pathway in C57BL/6 mice, Nutrients 12 (2020) 3362. https://doi.org/10.3390/nu12113362.
DOI
[33]

D. Ripolles, S. Harouna, J. A. Parrón, et al., Antibacterial activity of bovine milk lactoferrin and its hydrolysates prepared with pepsin, chymosin and microbial rennet against foodborne pathogen Listeria monocytogenes, Int. Dairy J. 45 (2015) 15–22. https://doi.org/10.1016/j.idairyj.2015.01.007.

[34]

X. Wang, F. Liu, Q. An, et al., Lactoferrin deficiency impairs proliferation of satellite cells via downregulating the ERK1/2 signaling pathway, Int. J. Mol. Sci. 23 (2022) 7478. https://doi.org/10.3390/ijms23137478.

[35]

D. Ripollés, J. A. Parrón, J. Fraguas, et al., Determination of lactadherin concentration in dairy by-products by ELISA: effect of heat treatment and hydrolysis, J. Dairy Sci. 101 (2018) 912–923. https://doi.org/10.3168/jds.2017-13608.

[36]

H. Li, W. L. Xu, Y. Ma, et al., Milk fat globule membrane protein promotes C2C12 cell proliferation through the PI3K/Akt signaling pathway, Int. J. Biol. Macromol. 114 (2018) 1305–1314. https://doi.org/10.1016/j.ijbiomac.2018.04.026.

[37]

A. Mishra, S. Liu, J. Promes, et al., Perilipin 2 downregulation in β cells impairs insulin secretion under nutritional stress and damages mitochondria, JCI Insight 6 (2021) 144341. https://doi.org/10.1172/jci.insight.144341.

[38]
B. Lucia, A. Giovanni, S. Andrea, et al., On the interaction of the highly charged peptides casocidins with biomimetic membranes, Bioelectrochemistry 123 (2018) 1–8. https://doi.org/10.1016/j.bioelechem.2018.04.012.
DOI
[39]

H. Chiba, F. Tani, M. Yoshikawa, et al., Opioid antagonist peptides derived from kappa-casein, J. Dairy Res. 56 (1989) 363–366. https://doi.org/10.1017/s0022029900028818.

[40]

E. Lahov, W. Regelson, Antibacterial and immunostimulating casein-derived substances from milk: casecidin, isracidin peptides, Food Chem. Toxicol. 34 (1996) 131–145. https://doi.org/10.1016/0278-6915(95)00097-6.

[41]

B. M. Donida, E. Mrak, C. Gravaghi, et al., Casein phosphopeptides promote calcium uptake and modulate the differentiation pathway in human primary osteoblast-like cells, Peptides 30 (2009) 2233–2241. https://doi.org/10.1016/j.peptides.2009.08.003.

Publication history
Copyright
Rights and permissions

Publication history

Received: 15 March 2023
Revised: 18 May 2023
Accepted: 25 May 2023
Published: 03 July 2023
Issue date: June 2023

Copyright

© Beijing Academy of Food Sciences 2023.

Rights and permissions

Food Science of Animal Products published 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