4,485
Views
24
CrossRef citations to date
0
Altmetric
Review Articles

Whey proteins: targets of oxidation, or mediators of redox protection

ORCID Icon, ORCID Icon, ORCID Icon, , , ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 1136-1152 | Received 04 Mar 2019, Accepted 06 Jun 2019, Published online: 12 Sep 2019

References

  • Rutherfurd SM, Fanning AC, Miller BJ, et al. Protein digestibility-corrected amino acid scores and digestible indispensable amino acid scores differentially describe protein quality in growing male rats. J Nutr. 2015;145:372–379.
  • Pereira PC. Milk nutritional composition and its role in human health. Nutrition. 2014;30:619–627.
  • Solak BB, Akin N. Health benefits of whey protein: a review. J Food Sci Eng. 2012;2:129–137.
  • De Wit JN. Marschall Rhône-Poulenc award lecture. Nutritional and functional characteristics of whey proteins in food products. J Dairy Sci. 1998;81:597–608.
  • Corrochano AR, Buckin V, Kelly PM, et al. Invited review: whey proteins as antioxidants and promoters of cellular antioxidant pathways. J Dairy Sci. 2018;101:4747–4761.
  • Michels AJ, Frei B. Myths, artifacts, and fatal flaws: identifying limitations and opportunities in vitamin C research. Nutrients. 2013;5:5161–5192.
  • Kim J, Paik HD, Yoon YC, et al. Whey protein inhibits iron overload-induced oxidative stress in rats. J Nutr Sci Vitaminol. 2013;59:198–205.
  • Lands LC, Iskandar M, Beaudoin N, et al. Dietary supplementation with pressurized whey in patients with cystic fibrosis. J Med Food. 2010;13:77–82.
  • Middleton N, Jelen P, Bell G. Whole blood and mononuclear cell glutathione response to dietary whey protein supplementation in sedentary and trained male human subjects. Int J Food Sci Nutr. 2004;55:131–141.
  • Giustarini D, Colombo G, Garavaglia ML, et al. Assessment of glutathione/glutathione disulphide ratio and S-glutathionylated proteins in human blood, solid tissues, and cultured cells. Free Radic Biol Med. 2017;112:360–375.
  • Oliveira PVS, Laurindo F. Implications of plasma thiol redox in disease. Clin Sci. 2018;132:1257–1280.
  • Aydın B, Atlı Şekeroğlu Z, Şekeroğlu V. Effects of whey protein and conjugated linoleic acid on acrolein-induced cardiac oxidative stress, mitochondrial dysfunction and dyslipidemia in rats. Biomed Pharmacother. 2018;107:901–907.
  • Gürgen SG, Yücel AT, Karakuş AÇ, et al. Usage of whey protein may cause liver damage via inflammatory and apoptotic responses. Hum Exp Toxicol. 2015;34:769–779.
  • Corrochano AR, Arranz E, De Noni I, et al. Intestinal health benefits of bovine whey proteins after simulated gastrointestinal digestion. J Funct Foods. 2018;49:526–535.
  • Corrochano AR, Ferraretto A, Arranz E, et al. Bovine whey peptides transit the intestinal barrier to reduce oxidative stress in muscle cells. Food Chem. 2019;288:306–314.
  • Giromini C, Cheli F, Rebucci R, et al. Invited review: dairy proteins and bioactive peptides: modeling digestion and the intestinal barrier. J Dairy Sci. 2019;102:929–942.
  • Bourlieu C, Ménard O, De La Chevasnerie A, et al. The structure of infant formulas impacts their lipolysis, proteolysis and disintegration during in vitro gastric digestion. Food Chem. 2015;182:224–235.
  • Egger L, Ménard O, Baumann C, et al. Digestion of milk proteins: comparing static and dynamic in vitro digestion systems with in vivo data. Food Res Int. 2019;118:32–39.
  • Furlund CB, Ulleberg EK, Devold TG, et al. Identification of lactoferrin peptides generated by digestion with human gastrointestinal enzymes. J Dairy Sci. 2013;96:75–88.
  • Tagliazucchi D, Helal A, Verzelloni E, et al. Composition and properties of peptides that survive standardised in vitro gastro-pancreatic digestion of bovine milk. Int Dairy J. 2016;61:196–204.
  • Tari NR, Fan MZ, Archbold T, et al. Effect of milk protein composition of a model infant formula on the physicochemical properties of in vivo gastric digestates. J Dairy Sci. 2018;101:2851–2861.
  • Boutrou R, Gaudichon C, Dupont D, et al. Sequential release of milk protein-derived bioactive peptides in the jejunum in healthy humans. Am J Clin Nutr. 2013;97:1314–1323.
  • Sanchón J, Fernández-Tomé S, Miralles B, et al. Protein degradation and peptide release from milk proteins in human jejunum. Comparison with in vitro gastrointestinal simulation. Food Chem. 2018;239:486–494.
  • Barbé F, Le Feunteun S, Rémond D, et al. Tracking the in vivo release of bioactive peptides in the gut during digestion: mass spectrometry peptidomic characterization of effluents collected in the gut of dairy matrix fed mini-pigs. Food Res Int. 2014;63:147–156.
  • Athira S, Mann B, Saini P, et al. Production and characterisation of whey protein hydrolysate having antioxidant activity from cheese whey. J Sci Food Agric. 2015;95:2908–2915.
  • Bamdad F, Bark S, Kwon CH, et al. Anti-inflammatory and antioxidant properties of peptides released from beta-Lactoglobulin by high hydrostatic pressure-assisted enzymatic hydrolysis. Molecules. 2017;22:pii: E949.
  • Bertucci JI, Liggieri CS, Colombo ML, et al. Application of peptidases from Maclura pomifera fruit for the production of active biopeptides from whey protein. LWT Food Sci Technol. 2015;64:157–163.
  • Contreras MDM, Hernández-Ledesma B, Amigo L, et al. Production of antioxidant hydrolyzates from a whey protein concentrate with thermolysin: optimization by response surface methodology. LWT Food Sci Technol. 2011;44:9–15.
  • Conway V, Gauthier SF, Pouliot Y. Antioxidant activities of buttermilk proteins, whey proteins, and their enzymatic hydrolysates. J Agric Food Chem. 2013;61:364–372.
  • Cruz-Huerta E, Maqueda DM, de la Hoz L, et al. Short communication: identification of iron-binding peptides from whey protein hydrolysates using iron (III) Immobilized metal ion affinity chromatography and reversed phase-HPLC-tandem mass spectrometry. J Dairy Sci. 2016;99:77–82.
  • Hernandez-Ledesma B, Davalos A, Bartolome B, et al. Preparation of antioxidant enzymatic hydrolysates from (alpha-lactalbumin and beta-lactoglobulin. Identification of active peptides by HPLC-MS/MS. J Agr Food Chem. 2005;53: 588–593.
  • Mann B, Kumari A, Kumar R, et al. Antioxidant activity of whey protein hydrolysates in milk beverage system. J Food Sci Technol. 2015;52:3235–3241.
  • Nongonierma AB, FitzGerald RJ. Inhibition of dipeptidyl peptidase IV (DPP-IV) by tryptophan containing dipeptides. Food Funct. 2013;4:1843–1849.
  • Sadat L, Cakir-Kiefer C, N’Negue MA, et al. Isolation and identification of antioxidative peptides from bovine alpha-lactalbumin. Int Dairy J. 2011;21:214–221.
  • Suetsuna K, Ukeda H, Ochi H. Isolation and characterization of free radical scavenging activities peptides derived from casein. J Nutr Biochem. 2000;11:128–131.
  • Jakobsson I, Axelsson I, Juvonen P, et al. Human alpha-lactalbumin as a marker of macromolecular absorption in early infancy. Acta Paediatr Scand. 1989;351:42–47.
  • Kuitunen M, Savilahti E, Sarnesto A. Human alpha-lactalbumin and bovine beta-lactoglobulin absorption in infants. Allergy. 1994;49:354–360.
  • USA Food and Drug Administration. Direct food substances affirmed as generally recognized as safe. The Code of Federal Regulations of the USA; 2018. 21CFR184.13669(c).
  • Milkovska-Stamenova S, Mnatsakanyan R, Hoffmann R. Protein carbonylation sites in bovine raw milk and processed milk products. Food Chem. 2017;229:417–424.
  • Scheidegger D, Radici PM, Vergara-Roig VA, et al. Evaluation of milk powder quality by protein oxidative modifications. J Dairy Sci. 2013;96:3414–3423.
  • Fenaille F, Parisod V, Tabet JC, et al. Carbonylation of milk powder proteins as a consequence of processing conditions. Proteomics. 2005;5:3097–3104.
  • Fenaille F, Parisod V, Visani P, et al. Modifications of milk constituents during processing: a preliminary benchmarking study. Int Dairy J. 2006;16:728–739.
  • Meltretter J, Becker CM, Pischetsrieder M. Identification and site-specific relative quantification of beta-lactoglobulin modifications in heated milk and dairy products. J Agric Food Chem. 2008;56:5165–5171.
  • Meltretter J, Wüst J, Pischetsrieder M. Modified peptides as indicators for thermal and nonthermal reactions in processed milk. J Agric Food Chem. 2014;62:10903–10915.
  • Meyer B, Baum F, Vollmer G, et al. Distribution of protein oxidation products in the proteome of thermally processed milk. J Agric Food Chem. 2012;60:7306–7311.
  • Krämer AC, Thulstrup PW, Lund MN, et al. Key role of cysteine residues and sulfenic acids in thermal- and H2O2-mediated modification of β-lactoglobulin. Free Radic Biol Med. 2016;97:544–555.
  • Krämer AC, Torreggiani A, Davies MJ. Effect of oxidation and protein unfolding on cross linking of β-lactoglobulin and α-lactalbumin. J Agric Food Chem. 2017;65:10258–10269.
  • Creamer LK, Bienvenue A, Nilsson H, et al. Heat-induced redistribution of disulfide bonds in milk proteins. 1. Bovine beta-lactoglobulin. J Agric Food Chem. 2004;52:7660–7668.
  • Nagy P. Kinetics and mechanisms of thiol-disulfide exchange covering direct substitution and thiol oxidation-mediated pathways. Antioxid Redox Signal. 2013;18:1623–1641.
  • Nielsen LR, Lund MN, Davies MJ, et al. Effect of free Cys on the denaturation and aggregation of holo α-lactalbumin. Int Dairy J. 2018;79:52–61.
  • Nielsen LR, Nielsen SB, Zhao Z, et al. Control of α-lactalbumin aggregation by modulation of temperature and concentration of calcium and cysteine. J Agric Food Chem. 2018;66:7110–7120.
  • Dalsgaard TK, Nielsen JH, Brown BE, et al. Dityrosine, 3,4-dihydroxyphenylalanine (DOPA), and radical formation from tyrosine residues on milk proteins with globular and flexible structures as a result of riboflavin-mediated photo-oxidation. J Agric Food Chem. 2011;59:7939–7947.
  • Dalsgaard TK, Otzen D, Nielsen JH, et al. Changes in structures of milk proteins upon photo-oxidation. J Agric Food Chem. 2007;55:10968–10976.
  • Fernández M, Ganan M, Guerra C, et al. Protein oxidation in processed cheese slices treated with pulsed light technology. Food Chem. 2014;159:388–390.
  • Boué G, Cummins E, Guillou S, et al. Public health risks and benefits associated with breast milk and infant formula consumption. Crit Rev Food Sci Nutr. 2018;58:126–145.
  • Chen Z, Leinisch F, Greco I, et al. Characterization and quantification of protein oxidative modifications and amino acid racemization in powdered infant milk formula. Free Radic Biol Med. 2018;120:S47.
  • D’Agostina A, Boschin G, Rinaldi A, et al. Updating on the lysinoalanine content of commercial infant formulae and beicost products. Food Chem. 2003;80:483–488.
  • Sun X, Wang C, Wang H, Guo M. Effects of processing on structure and thermal properties of powdered preterm infant formula. J Food Sci. 2018;83:1685–1694.
  • Baxter JH, Lai CS, Phillips RR, et al. Direct determination of methionine sulfoxide in milk proteins by enzyme hydrolysis/high-performance liquid chromatography. J Chromatogr A. 2007;1157:10–16.
  • Fuentes-Lemus E, Silva E, Barrias P, et al. Aggregation of α-and β-caseins induced by peroxyl radicals involves secondary reactions of carbonyl compounds as well as di-tyrosine and di-tryptophan formation. Free Radic Biol Med. 2018;124:176–188.
  • Fuentes-Lemus E, Silva E, Leinisch F, et al. Alpha- and beta-casein aggregation induced by riboflavin-sensitized photo-oxidation occurs via di-tyrosine cross-links and is oxygen concentration dependent. Food Chem. 2018;256:119–128.
  • Schöneich C. Methionine oxidation by reactive oxygen species: reaction mechanisms and relevance to Alzheimer’s disease. Biochim Biophys Acta. 2005;1703:111–119.
  • Boatright WL, Crum AD. Redox cycling and generation of reactive oxygen species in commercial infant formulas. Food Chem. 2016;196:189–195.
  • Cervenka I, Agudelo LZ, Ruas JL. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science. 2017;357:eaaf9794.
  • Ahmed N, Mirshekar-Syahkal B, Kennish L, et al. Assay of advanced glycation endproducts in selected beverages and food by liquid chromatography with tandem mass spectrometric detection. Mol Nutr Food Res. 2005;49:691–699.
  • Birlouez-Aragon I, Pischetsrieder M, Leclere J. Assessment of protein glycation markers in infant formulas. Food Chem. 2004;87:253–259.
  • Cardoso HB, Wierenga PA, Gruppen H, et al. Maillard induced aggregation of individual milk proteins and interactions involved. Food Chem. 2019;276:652–661.
  • Ciesarová Z, Kukurová K, Bednáriková A, et al. Effect of heat treatment and dough formulation on the formation of maillard reaction products in fine bakery products-benefits and weak points. J Food Nutr Res. 2009;48:20–30.
  • Guerra-Hernandez E, Corzo N, Garcia-Villanova B. Maillard reaction evaluation by furosine determination during infant cereal processing. J Cereal Sci. 1999;29:171–176.
  • Prosser CG, Carpenter EA, Hodgkinson AJ. Nε-carboxymethyllysine in nutritional milk formulas for infants. Food Chem. 2019;274:886–890. [doi:]
  • Rufián-Henares JA, García-Villanova B, Guerra-Hernández E. Generation of furosine and color in infant/enteral formula-resembling systems. J Agric Food Chem. 2004;52:5354–5358.
  • Silván JM, van de Lagemaat J, Olano A, et al. Analysis and biological properties of amino acid derivates formed by Maillard reaction in foods. J Pharm Biomed Anal. 2006;41:1543–1551.
  • Birlouez-Aragon I, Locquet N, de St Louvent E, et al. Evaluation of the maillard reaction in infant formulas by means of front-face fluorescence. Ann N Y Acad Sci. 2005;1043:308–318.
  • Contreras-Calderón J, Guerra-Hernández E, García-Villanova B. Utility of some indicators related to the Maillard browning reaction during processing of infant formulas. Food Chem. 2009;114:1265–1270.
  • Richardson P, Thermal technologies in food processing. Milton Park, Abingdon: Taylor & Francis; 2001.
  • Sund-Levander M, Forsberg C, Wahren LK. Normal oral, rectal, tympanic and axillary body temperature in adult men and women: a systematic literature review. Scand J Caring Sci. 2002;16:122–128.
  • Aalaei K, Rayner M, Sjöholm I. Chemical methods and techniques to monitor early Maillard reaction in milk products; A review. Crit Rev Food Sci Nutr. 2018;59:1829–1839.
  • Brownlee, MDM. Advanced protein glycosylation in diabetes and aging. Annu Rev Med. 1995;46:223–234.
  • Wells-Knecht KJ, Brinkmann E, Wells-Knecht MC, et al. New biomarkers of Maillard reaction damage to proteins. Nephrol Dial Transplant. 1996;11:41–47.
  • Penndorf I, Biedermann D, Maurer SV, Henle T. Studies on N-terminal glycation of peptides in hypoallergenic infant formulas: quantification of alpha-N-(2-furoylmethyl) amino acids. J Agric Food Chem. 2007;55:723–727.
  • Estévez M, Luna C. Dietary protein oxidation: a silent threat to human health? Crit Rev Food Sci Nutr. 2017;57:3781–3793.
  • Committee ES. Guidance on human health risk-benefit assessment of foods. EFSA J. 2010;8:1673.
  • Kanner J. Dietary advanced lipid oxidation endproducts are risk factors to human health. Mol Nutr Food Res. 2007;51:1094–1101.
  • Turek JJ, Watkins BA, Schoenlein IA, et al. Oxidized lipid depresses canine growth, immune function, and bone formation. J Nutr Biochem. 2003;14:24–31.
  • Zhang W, Xiao S, Lee EJ, et al. Effects of dietary oxidation on the quality of broiler breast meat. Anim Ind Rep. 2011;657:48.
  • Delgado-Andrade C, Fogliano V. Dietary advanced glycosylation end-products (dAGEs) and melanoidins formed through the Maillard reaction: physiological consequences of their intake. Annu Rev Food Sci Technol. 2018;9:271–291.
  • Biswas MS, Mano J. Reactive carbonyl species activate caspase-3-like protease to initiate programmed cell death in plants. Plant Cell Physiol. 2016;57:1432–1442.
  • Esterbauer H. Cytotoxicity and genotoxicity of lipid-oxidation products. Am J Clin Nutr. 1993;57:779S–785S.
  • Xie F, Sun S, Xu A, et al. Advanced oxidation protein products induce intestine epithelial cell death through a redox-dependent, c-jun N-terminal kinase and poly (ADP-ribose) polymerase-1-mediated pathway. Cell Death Dis. 2014;5:e1006.
  • Gurer-Orhan H, Ercal N, Mare S, et al. Misincorporation of free m-tyrosine into cellular proteins: a potential cytotoxic mechanism for oxidized amino acids. Biochem J. 2006;395:277–284.
  • Li ZL, Shi Y, Le G, et al. 24-week exposure to oxidized tyrosine induces hepatic fibrosis involving activation of the mapk/tgf-β1 signaling pathway in Sprague-Dawley rats model. Oxid Med Cell Longev. 2016;2016:3123294.
  • Chen Y, Guillemin GJ. Kynurenine pathway metabolites in humans: disease and healthy states. Int J Tryptophan Res. 2009;2:1–19.
  • Keszthelyi D, Troost FJ, Masclee AA. Understanding the role of tryptophan and serotonin metabolism in gastrointestinal function. Neurogastroenterol Motil. 2009;21:1239–1249.
  • Turski MP, Turska M, Zgrajka W, et al. Presence of kynurenic acid in food and honeybee products. Amino Acids. 2009;36:75–80.
  • Dever JT, Elfarra AA. L-methionine-dl-sulfoxide metabolism and toxicity in freshly isolated mouse hepatocytes: gender differences and inhibition with aminooxyacetic acid. Drug Metab Dispos. 2008;36:2252–2260.
  • Li ZL, Mo L, Le G, et al. Oxidized casein impairs antioxidant defense system and induces hepatic and renal injury in mice. Food Chem Toxicol. 2014;64:86–93.
  • Mottram DS, Wedzicha BL, Dodson AT. Acrylamide is formed in the Maillard reaction. Nature. 2002;419:448–449.
  • Van Boekel MA. Formation of flavour compounds in the Maillard reaction. Biotechnol Adv. 2006;24:230–233.
  • Tamanna N, Mahmood N. Food processing and maillard reaction products: effect on human health and nutrition. Int J Food Sci. 2015;2015:1.
  • Uribarri J, del Castillo MD, de la Maza MP, et al. Dietary advanced glycation end products and their role in health and disease. Adv Nutr. 2015;6:461–473.
  • Friedman M. Food browning and its prevention: an overview. J Agric Food Chem. 1996;44:631–653.
  • Vhangani LN, Van Wyk J. Antioxidant activity of Maillard reaction products (MRPs) derived from fructose–lysine and ribose–lysine model systems. Food Chem. 2013;137:92–98.
  • Damodaran S, Parkin KL, Fennema’s food chemistry. Boca Raton (FL): CRC Press; 2017.
  • Yilmaz Y, Toledo R. Antioxidant activity of water-soluble Maillard reaction products. Food Chem. 2005;93:273–278.
  • Palimeri S, Palioura E, Diamanti-Kandarakis E. Current perspectives on the health risks associated with the consumption of advanced glycation end products: recommendations for dietary management. Diabetes Metab Syndr Obes Targets Ther. 2015;8:415–426.
  • Yuan X, Zhao J, Qu W, et al. Accumulation and effects of dietary advanced glycation end products on the gastrointestinal tract in rats. Int J Food Sci Technol. 2018;53;2273–2281.
  • Badr G, Sayed LH, Omar HEDM, et al. Camel whey protein protects B and T cells from apoptosis by suppressing activating transcription factor-3 (ATF-3)-mediated oxidative stress and enhancing phosphorylation of AKT and IκB-α in type I diabetic mice. Cell Physiol Biochem. 2017;41:41–54.
  • Qu W, Nie C, Zhao J, et al. Microbiome–metabolomics analysis of the impacts of long-term dietary advanced-glycation-end-product consumption on C57BL/6 mouse fecal microbiota and metabolites. J Agric Food Chem. 2018;66:8864–8875.
  • Ames JM. Evidence against dietary advanced glycation endproducts being a risk to human health. Mol Nutr Food Res. 2007;51:1085–1090.
  • Somoza V, Wenzel E, Lindenmeier M, et al. Influence of feeding malt, bread crust, and a pronylated protein on the activity of chemopreventive enzymes and antioxidative defense parameters in vivo. J Agric Food Chem. 2005;53:8176–8182.
  • Klenovics KS, Boor P, Somoza V, et al. Advanced glycation end products in infant formulas do not contribute to insulin resistance associated with their consumption. PLoS One. 2013;8:e53056.
  • Dworschák E. Nonenzyme browning and its effect on protein nutrition. Crit Rev Food Sci Nutr. 1980;13:1–40.
  • Chen Z, Kondrashina A, Greco I, et al. Effects of protein-derived amino acid modification products present in infant formula on metabolic function, oxidative stress, and intestinal permeability in cell models. J Agric Food Chem. 2019;67:5634–5646.
  • Dunlop RA, Dean RT, Rodgers KJ. The impact of specific oxidized amino acids on protein turnover in J774 cells. Biochem J. 2008;410:131–140.
  • Rodgers KJ, Wang H, Fu S, et al. Biosynthetic incorporation of oxidized amino acids into proteins and their cellular proteolysis. Free Radic Biol Med. 2002;32:766–775.
  • Li ZL, Shi Y, Ding Y, et al. Dietary oxidized tyrosine (O-Tyr) stimulates TGF-β1-induced extracellular matrix production via the JNK/p38 signaling pathway in rat kidneys. Amino Acids. 2017;49:241–260.
  • Hubatsch I, Ragnarsson EG, Artursson P. Determination of drug permeability and prediction of drug absorption in Caco-2 monolayers. Nat Protoc. 2007;2:2111–2119.
  • Hellwig M, Geissler S, Peto A, et al. Transport of free and peptide-bound pyrraline at intestinal and renal epithelial cells. J Agric Food Chem. 2009;57:6474–6480.
  • Devés R, Boyd CA. Transporters for cationic amino acids in animal cells: discovery, structure, and function. Physiol Rev. 1998;78:487–545.
  • Geissler S, Hellwig M, Zwarg M, et al. Transport of the advanced glycation end products alanylpyrraline and pyrralylalanine by the human proton-coupled peptide transporter hPEPT1. J Agric Food Chem. 2010;58:2543–2547.
  • Grunwald S, Krause R, Bruch M, et al. Transepithelial flux of early and advanced glycation compounds across Caco-2 cell monolayers and their interaction with intestinal amino acid and peptide transport systems. Br J Nutr. 2006;95:1221–1228.
  • Hellwig M, Geissler S, Matthes R, et al. Transport of free and peptide-bound glycated amino acids: synthesis, transepithelial flux at Caco-2 cell monolayers, and interaction with apical membrane transport proteins. ChemBioChem. 2011;12:1270–1279.
  • Šebeková K, Saavedra G, Zumpe C, et al. Plasma concentration and urinary excretion of N ɛ -(carboxymethyl)lysine in breast milk- and formula-fed infants. Ann NY Acad Sci. 2008;1126:177–180.
  • Steinerová A, Racek J, Rajdl D, et al. Significant increase in antibodies against oxidized LDL particles (IgoxLDL) in three-month old infants who received milk formula. Atherosclerosis. 2004;173:147–148.
  • Elmhiri G, Mahmood DFD, Niquet-Leridon C, et al. Formula-derived advanced glycation end products are involved in the development of long-term inflammation and oxidative stress in kidney of IUGR piglets. Mol Nutr Food Res. 2015;59:939–947.
  • Cai W, He JC, Zhu L, et al. High levels of dietary advanced glycation end products transform low-density lipoprotein into a potent redox-sensitive mitogen-activated protein kinase stimulant in diabetic patients. Circulation. 2004;110:285–291.
  • Hummel S, Beyerlein A, Tamura R, et al. First infant formula type and risk of islet autoimmunity in the environmental determinants of diabetes in the young (TEDDY) study. Dia Care. 2017;40:398–404.
  • Writing Group for the TRIGR Study Group Knip M, Åkerblom HK, et al. Effect of hydrolyzed infant formula vs. conventional formula on risk of type 1 diabetes: the TRIGR randomized clinical trial. JAMA. 2018;319:38–48.
  • Saukkonen T, Virtanen SM, Karppinen M, et al. Significance of cow’s milk protein antibodies as risk factor for childhood IDDM: interactions with dietary cow’s milk intake and HLA-DQB1 genotype. Childhood diabetes in Finland study group. Diabetologia. 1998;41:72–78.
  • Vaarala O, Ilonen J, Ruohtula T, et al. Removal of bovine insulin from cow’s milk formula and early initiation of beta-cell autoimmunity in the FINDIA pilot study. Arch Pediatr Adolesc Med. 2012;166:608–614.
  • Falkowski M, Maciejczyk M, Koprowicz T, et al. Whey protein concentrate WPC-80 improves antioxidant defense systems in the salivary glands of 14-month Wistar rats. Nutrients. 2018;10: pii: 782.
  • Żebrowska-Gamdzyk M, Maciejczyk M, Zalewska A, et al. Whey protein concentrate WPC-80 intensifies glycoconjugate catabolism and induces oxidative stress in the liver of rats. Nutrients. 2018;10:pii: E1178.
  • Gad AS, Khadrawy YA, El-Nekeety AA, et al. Antioxidant activity and hepatoprotective effects of whey protein and Spirulina in rats. Nutrition. 2011;27:582–589.
  • Balbis E, Patriarca S, Furfaro AL, et al. Whey proteins influence hepatic glutathione after CCl4 intoxication. Toxicol Ind Health. 2009;25:325–328.
  • Ashoush IS, El-Batawy OI, El-Shourbagy GA. Antioxidant activity and hepatoprotective effect of pomegranate peel and whey powders in rats. Ann Agric Sci. 2013;58:27–32.
  • Abdel-Wahhab KG, Mannaa FA, Abdel-Wahhab MA. The protective effects of whey protein and Spirulina Against CCl4-induced erythrocyte damage in rats. J Appl Sci Res. 2013;9:2063–2071.
  • Mansour DF, Nada SA, El-Denshary ES, et al. Milk whey proteins modulate endotoxemia-induced hepatotoxicity in rats. Int J Pharm Pharm Sci. 2015;7:65–71.
  • Abdel-Aziem SH, Hassan AM, Abdel-Wahhab MA. Dietary supplementation with whey protein and ginseng extract counteracts oxidative stress and DNA damage in rats fed an aflatoxin-contaminated diet. Mutat Res. 2011;723:65–71.
  • Hassan AM, Abdel-Aziem SH, Abdel-Wahhab MA. Modulation of DNA damage and alteration of gene expression during aflatoxicosis via dietary supplementation of Spirulina (Arthrospira) and Whey protein concentrate. Ecotoxicol Environ Saf. 2012;79:294–300.
  • Haraguchi FK, Silva ME, Neves LX, et al. Whey protein precludes lipid and protein oxidation and improves body weight gain in resistance-exercised rats. Eur J Nutr. 2011;50:331–339.
  • Elia D, Stadler K, Horváth V, et al. Effect of soy- and whey protein-isolate supplemented diet on the redox parameters of trained mice. Eur J Nutr. 2006;45:259–266.
  • Garg G, Singh S, Singh AK, et al. Whey protein concentrate supplementation protects rat brain against aging-induced oxidative stress and neurodegeneration. Appl Physiol Nutr Metab. 2018;43:437–444.
  • Hamad EM, Taha SH, Abou Dawood AGI, et al. Protective effect of whey proteins against nonalcoholic fatty liver in rats. Lipids Health Dis. 2011;10:57.
  • Ignowski E, Winter AN, Duval N, et al. The cysteine-rich whey protein supplement, Immunocal®, preserves brain glutathione and improves cognitive, motor, and histopathological indices of traumatic brain injury in a mouse model of controlled cortical impact. Free Radic Biol Med. 2018;124:328–341.
  • Shertzer HG, Krishan M, Genter MB. Dietary whey protein stimulates mitochondrial activity and decreases oxidative stress in mouse female brain. Neurosci Lett. 2013;548:159–164.
  • Ross E, Winter A, Wilkins H, et al. A cystine-rich whey supplement (Immunocal(®)) delays disease onset and prevents spinal cord glutathione depletion in the hSOD1(G93A) Mouse model of amyotrophic lateral sclerosis. Antioxidants (Basel). 2014;3:843–865.
  • Song W, Tavitian A, Cressatti M, et al. Cysteine-rich whey protein isolate (Immunocal®) ameliorates deficits in the GFAP.HMOX1 mouse model of schizophrenia. Free Radic Biol Med. 2017;110:162–175.
  • Oner OZ, Oğünç AV, Cingi A, et al. Whey feeding suppresses the measurement of oxidative stress in experimental burn injury. Surg Today. 2006;36:376–381.
  • Velioglu Ogünç A, Manukyan M, Cingi A, et al. Dietary whey supplementation in experimental models of wound healing. Int J Vitam Nutr Res. 2008;78:70–73.
  • Toden S, Bird AR, Topping DL, et al. Differential effects of dietary whey, casein and soya on colonic DNA damage and large bowel SCFA in rats fed diets low and high in resistant starch. Br J Nutr. 2007;97:535–543.
  • Bartfay WJ, Davis MT, Medves JM, et al. Milk whey protein decreases oxygen free radical production in a murine model of chronic iron-overload cardiomyopathy. Can J Cardiol. 2003;19:1163–1168.
  • Chitapanarux T, Tienboon P, Pojchamarnwiputh S, et al. Open-labeled pilot study of cysteine-rich whey protein isolate supplementation for nonalcoholic steatohepatitis patients. J Gastroenterol Hepatol. 2009;24:1045–1050.
  • Bumrungpert A, Pavadhgul P, Nunthanawanich P, et al. Whey protein supplementation improves nutritional status, glutathione levels, and immune function in cancer patients: a randomized, double-blind controlled trial. J Med Food. 2018;21:612–616.
  • Grey V, Mohammed SR, Smountas AA, et al. Improved glutathione status in young adult patients with cystic fibrosis supplemented with whey protein. J Cyst Fibros. 2003;2:195–198.
  • Smiles WJ, Areta JL, Coffey VG, et al. Modulation of autophagy signaling with resistance exercise and protein ingestion following short-term energy deficit. Am J Physiol Regul Integr Comp Physiol. 2015;309:R603–R612.
  • Flaim C, Kob M, Di Pierro AM, et al. Effects of a whey protein supplementation on oxidative stress, body composition and glucose metabolism among overweight people affected by diabetes mellitus or impaired fasting glucose: a pilot study. J Nutr Biochem. 2017;50:95–102.
  • Sheikholeslami Vatani D, Ahmadi Kani Golzar F. Changes in antioxidant status and cardiovascular risk factors of overweight young men after six weeks supplementation of whey protein isolate and resistance training. Appetite. 2012;59:673–678.
  • Tosukhowong P, Boonla C, Dissayabutra T, et al. Biochemical and clinical effects of Whey protein supplementation in Parkinson’s disease: a pilot study. J Neurol Sci. 2016;367:162–170.
  • Micke P, Beeh KM, Schlaak JF, et al. Oral supplementation with whey proteins increases plasma glutathione levels of HIV-infected patients. Eur J Clin Invest. 2001;31:171–178.
  • Moreno YF, Sgarbieri VC, da Silva MN, et al. Features of whey protein concentrate supplementation in children with rapidly progressive HIV infection. J Trop Pediatr. 2006;52:34–38.
  • Zavorsky GS, Kubow S, Grey V, et al. An open-label dose-response study of lymphocyte glutathione levels in healthy men and women receiving pressurized whey protein isolate supplements. Int J Food Sci Nutr. 2007;58:429–436.
  • Egger L, Schlegel P, Baumann C, et al. Mass spectrometry data of in vitro and in vivo pig digestion of skim milk powder. Data Brief. 2018;21:911–917.
  • Bellomonte G, Costantini A, Giammarioli S. Comparison of modified automatic Dumas method and the traditional Kjeldahl method for nitrogen determination in infant food. J Assoc off Anal Chem. 1987;70:227–229.
  • Friedman M. Chemistry, biochemistry, nutrition, and microbiology of lysinoalanine, lanthionine, and histidinoalanine in food and other proteins. J Agric Food Chem. 1999;47:1295–1319.
  • Pompei C, Rossi M, Marè F. Protein quality in commercial milk-based infant formulas. J Food Quality. 1988;10:375–391.
  • Martysiak-Zurowska D, Stolyhwo A. Content of furosine in infant formulae and follow-on formulae. Pol J Food Nutr Sci. 2007;57:185–190.
  • Baptista JAB, Carvalho R. Indirect determination of Amadori compounds in milk-based products by HPLC/ELSD/UV as an index of protein deterioration. Food Res Int. 2004;37:739–747.
  • Morales V, Olano A, Corzo N. Ratio of maltose to maltulose and furosine as quality parameters for infant formula. J Agric Food Chem. 2004; 52:6732–6736.
  • Guerra-Hernández E, Ramirez-Jiménez A, García-Villanova B. Glucosylisomaltol, a new indicator of browning reaction in baby cereals and bread. J Agric Food Chem. 2002;50:7282–7287.