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

Abstract

Bovine whey proteins are highly valued dairy ingredients. This is primarily due to their amino acid content, digestibility, bioactivities and their processing characteristics. One of the reported bioactivities of whey proteins is antioxidant activity. Numerous dietary intervention trials with humans and animals indicate that consumption of whey products can modulate redox biomarkers to reduce oxidative stress. This bioactivity has in part been assigned to whey peptides using a range of biochemical or cellular assays in vitro. Superimposing whey peptide sequences from gastrointestinal samples, with whey peptides proven to be antioxidant in vitro, allows us to propose peptides from whey likely to exhibit antioxidant activity in the diet. However, whey proteins themselves are targets of oxidation during processing particularly when exposed to high thermal loads and/or extensive processing (e.g. infant formula manufacture). Oxidative damage of whey proteins can be selective with regard to the residues that are modified and are associated with the degree of protein unfolding, with α-Lactalbumin more susceptible than β-Lactoglobulin. Such oxidative damage may have adverse effects on human health. This review summarises how whey proteins can modulate cellular redox pathways and conversely how whey proteins can be oxidised during processing. Given the extensive processing steps that whey proteins are often subjected to, we conclude that oxidation during processing is likely to compromise the positive health attributes associated with whey proteins.

Introduction

Milk proteins (whey proteins (∼20%) and caseins (80%)) are high-quality sources of amino acids (AA) in the human diet. Bovine milk proteins and peptides play important roles in human health not just in terms of nutrition but also in terms of their notable bioactivities. The major bovine whey proteins are α-Lactalbumin (α-Lac) and β-Lactoglobulin (β-Lg) with immunoglobulins, bovine serum albumin (BSA) and lactoferrin as minor proteins (). Whey proteins provide a complete protein source and are rich in both sulphur-containing and branched-chain AAs. Whey proteins do not coagulate under the acidic conditions present in the stomach, and are considered to be “fast proteins” since they reach the jejunum shortly after entering the gastrointestinal tract and have a digestible indispensable AA score of 1.09 [Citation1]. Whey proteins exhibit a wide range of bioactivities including antioxidant, antibacterial, antifungal, antiviral, antihypertensive, antithrombotic, opioid and immunomodulatory properties [Citation2]. Consequently, whey products are recognised as value-added ingredients and are commonly used in the sports nutrition market, nutritional beverages for the elderly and infant formula (IF) [Citation3]. These products are also prized in food formulation as they improve product quality by water-binding, stabilising aerated food products and acting as emulsifying agents [Citation4]. Whey products can be supplied as ingredients for food formulations as whey protein concentrate (WPC), whey protein isolate (WPI) or as whey hydrolysates (WH) all of which differ in the degree of processing and protein content (). However, numerous processing steps from milk to whey powder to food formulation inherently expose whey proteins themselves to redox modifications. This review summarises how whey proteins can modulate cellular redox pathways and conversely how whey proteins can be oxidised during processing.

Table 1. Bovine whey proteins.

Table 2. Composition of different bovine whey products.

Whey proteins and their ability to modify redox pathways – studies in animal models and humans

The antioxidant and detoxifying bioactivities of whey proteins are most likely linked to their contribution to glutathione (GSH) synthesis, recently reviewed in Corrochano et al. [Citation5]. Whey proteins are rich in Cys with β-Lg containing 5 Cys residues, α-Lac has 8 Cys, BSA has 35 Cys and lactoferrin contains 34 Cys, although in each case the majority of Cys is present as disulphide bonds. The thiol (R-SH) group of Cys reacts rapidly with many oxidants. This AA when present with Gly and Gln in the tripeptide, GSH, is an important cofactor and antioxidant in mammalian cells and tissues. Reduced GSH, is readily oxidised to the disulphide species, oxidised glutathione (GSSG), with the latter then readily recycled by the enzyme GSH reductase, at the expense of nicotinamide adenine dinucleotide phosphate (NADPH), back to its reduced form. Other oxidised species can however also be formed from GSH including GSH sulphonamide and oxy acids, with the latter species being irreversible products. GSH detoxifies a number of endogenous and exogenous toxins including toxic metals, petroleum distillates, lipid peroxides, quinones, bilirubin and prostaglandins through direct conjugation. Cell lines (e.g. C2C12, MRC-5, PC12, Caco2, HUVEC, 9HTEo, HepG2, and REPE-1) exposed to various whey products (WPC and WPI) have documented increases in GSH levels with some exceptions [Citation5]. Whey products have also been reported to increase activities of the protective enzymes superoxide dismutase (which removes superoxide radicals) and catalase (which reduces H2O2 to water) and decrease levels of reactive oxygen species, lipid peroxidation and DNA damage in cellular assays [Citation5]. However using cell lines with whole proteins has its limitations, not least of which is the altered redox homeostasis of immortalised cell lines [Citation6] and the nonphysiological exposure of cells to intact dietary proteins.

Human or animal intervention trials with diets that include whey products are the best assessment of impact on cellular redox pathways although reliable and consistent data with well-defined biomarkers is rather limited. In a significant number of cases generic and non-specific assays of antioxidant activity have been used, which is not ideal. lists rat and mouse studies with a focus on redox parameters, where animals consumed different whey products (WPC and WPI) at a dosage of 0.02–1 g/kg body mass over a period of 7–84 d. Redox biomarkers were measured in liver, brain, erythrocyte, muscle, serum, kidney, colon, salivary gland or parathyroid gland in the presence or absence of various stressors (exercise, diabetes, heat, brain injury, hepatotoxicity, dyslipidaemia, schizophrenia or aflatoxin). Although conflicting data have been reported [Citation7], the majority of trials have reported increases in GSH and other antioxidant markers. However exactly which species, and what concentrations, are responsible for these changes is unclear in most cases. Human dietary whey intervention trials with a focus on redox readouts have been reviewed previously by Corrochano et al. [Citation5], with additional trials detailed in . These intervention trials generally recruit participants for, on average, a 3-week study with or without an exercise routine, measuring plasma GSH as an indicator of whole body redox status. Many studies report increases in plasma GSH from time zero with whey intervention but others observe no effect on plasma GSH levels () [Citation8,Citation9]. Whether or not a single redox analyte detected at very low levels in the plasma is a good biomarker of redox state at a global or local tissue level is debatable [Citation10,Citation11].

Table 3. Animal studies with different whey products and the antioxidant responses reported.

Table 4. Human intervention trials with whey products and physiological response.

There is also some evidence from intervention trials that whey proteins may decrease plasma GSH levels, cause heart damage and liver injury [Citation12,Citation13]. Oral gavage of adult male Sprague Dawley rats (n = 6) with the environmental pollutant acrolein (0.005 g/kg body weight/d) for 30 d resulted in a significant decrease in GSH levels (8.38 ± 1.17 nmol/mg protein) in red blood cells compared to the control group (11.31 ± 1.63 nmol/mg protein) (p < .05) [Citation12]. Addition of whey protein at a dosage of 0.2 g/kg body weight/d did not halt the loss of GSH. In addition, this co-treatment with whey protein exacerbated an observed increase in plasma homocysteine levels and creatine kinase levels induced by acrolein, such that the levels of these markers in the co-treatment group were significantly higher than in the controls. This led the authors to hypothesise that intake of acrolein together with whey proteins may cause heart damage in rats [Citation12]. Liver may also be adversely affected by whey treatment. Gürgen et al. [Citation13] investigated liver health in Wistar albino male rats (n = 10) after a whey protein diet for 5 d (short-term) or 4 weeks (long-term). Hepatic injury was observed by abnormal hepatocyte histology and significantly increased levels of serum aspartate amino transferase and hepatic interluekin-1β in rats that consumed whey compared to the control group, with the markers of liver injury worsening with increasing time of exposure to whey protein diet (p < .05).

Bioavailable antioxidant whey peptides

In addition to providing essential AAs and reduced thiols (Cys residues), whey proteins also contribute peptides with potential antioxidant activity [Citation5,Citation14,Citation15]. However to be bioactive beyond the gut, whey peptides must survive gut transit and be bioavailable to their target [Citation16]. The harsh conditions of the upper gastrointestinal tract function to hydrolyse proteins into individual AAs for transport across the intestinal barrier. Several recent studies have tracked the fate of whey proteins during upper gastrointestinal digestion using in vitro digestion models [Citation14,Citation17–20] or gastric [Citation18,Citation21] and jejunal effluents [Citation22,Citation23] from pigs [Citation18,Citation21,Citation24] and humans [Citation19,Citation22,Citation23] post consumption of various dairy foods (IF, WPI, skim milk powder, unpasteurised milk, whey powder, and lactoferrin). Based on these studies, lists locations within individual whey proteins from which peptides have been identified in the intestinal phase. β-Lg has four gut-resistant “hotspots;” consisting of fragments (f) with the following amino acid residues from the primary sequence f(41–58), f(92–100), f(126–138) and f(149–154). α-Lac also has 4 hotspots; f(17–27), f(63–68), f(80–90) and f(97–102). BSA has 5; f(11–18), f(107–114), f(219–224), f(489–495) and f(514–518). Lactoferrin has 7; f(67–77), f(140–145), f(216–228), f(289–295), f(309–318), f(332–337) and f(592–594). This suggests that these regions of the primary sequence are somewhat resistant to gastrointestinal digestion and peptides from these hotspots may survive the gut long enough to be transported across the intestinal barrier. Certainly, the presence of proline and/or aspartic acid, or glutamic acid residues within a peptide appears to confer a resistance to gastrointestinal digestion [Citation23]. It should also be noted that peptide profiles differ according to degree of processing [Citation24]. also details whey peptides that have been reported to have potential redox activity encrypted within these hotspots [Citation5,Citation25–35]. These whey peptides have been reported to show redox activity in the ferric-reducing antioxidant power assay (FRAP), 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) assay, oxygen radical absorbance capacity assay (ORAC) or the 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical assay. It should however be noted that all of these assays are generic in vitro assays conducted in the absence of alternative targets, and hence the data cannot be readily translated to more complex systems. In some cases, however, they have also been shown to boost cellular antioxidant status, albeit in vitro [Citation5,Citation15]. Cross matching gastrointestinal resistant hotspots to antioxidant bioactivity allows us to propose a list of potential antioxidant whey peptides likely to arrive in the bloodstream, post whey consumption. Little is known about peptides derived from whey circulating in the bloodstream. Jakobsson et al. [Citation36] quantified α-Lac (140–250 µg/L serum/L human milk/kg body weight) by radioimmunoassay in blood plasma from 1 month old breastfed infants (n = 3), 30–60 min after feeding. Kuitunen et al. [Citation37] also reported that plasma from 20 full term infants was positive for α-Lac and β-Lg, levels of which declined over time (almost 60% reduction by 8 months of age) as the infant gut barrier matured. No information on the sequences of the bioavailable peptides was provided. However, we have recently identified a number of antioxidant whey peptides post in vitro gastrointestinal digestion capable of traversing the tight junctions of Caco2-HT29 monolayers (a widely accepted model for the intestinal barrier) and arriving in the basolateral compartment [Citation15].

Table 5. Whey peptide sequences resistant to upper gastrointestinal digestion and antioxidant peptides encrypted within these sequences.

Redox modification of whey during processing

As whey products typically undergo several processing steps from milk to final food matrix, there is substantial evidence that processing can induce or exacerbate redox reactions that modify whey proteins. At the outset, milk produced for consumer consumption is heat-treated to kill bacteria and increase shelf life. Multiple methods are used including low (e.g. 15 s at 74 °C), high (e.g. 15 s at 90 °C), and ultrahigh (e.g. 145 °C for a few seconds) temperature treatments. Most commercial milk is also homogenised by high pressure treatment to reduce fat globule size. Bovine liquid whey is then produced by either the enzymatic treatment of milk (sweet whey) or by the addition of acids or minerals (acid whey) both of which result in the precipitation, and therefore removal, of caseins. Significant modifications on whey proteins occur with processing, due to heat treatment (pasteurisation and spray drying), exposures to high pressures (e.g. during homogenisation), light exposure, the use of sterilisation/disinfection agents such as H2O2 (permitted at concentrations ≤ 16 mM in the USA [Citation38]), high pH values (employed to give hydrolysed protein samples e.g. for IF) and long-term exposure to reducing sugars (e.g. lactose, glucose, galactose. and products from these).

Oxidation, glycation, and racemisation of whey proteins

Thermal treatment of milk and milk products can result in a significant increase in the level of oxidative modification and formation of protein carbonyls (with the sites of some of these characterised [Citation39]) and crosslinks. The levels of these materials have been proposed as a marker of milk powder quality [Citation40]. The highest levels of protein oxidation products have been reported to be present in powdered IF [Citation39,Citation41–44]. α-Lac has been reported to show enhanced oxidation compared to β-Lg [Citation45]. A recent study has reported that thermal treatment can induce reducible (disulphide) crosslinks in isolated β-Lg (100 µM), but not isolated α-Lac (150 µM) in both the absence and presence of H2O2 (500 µM), which appears to be associated with the protein unfolding and the accessibility of the free Cys-121 residue on β-Lg [Citation46,Citation47]. Blocking this thiol prevented cross-link formation, underlining the importance of this residue. Mixed cross-links between β-Lg and α-Lac were detected when both proteins were co-treated [Citation47]. Disulphide cross-links can also be generated via thiol-disulphide exchange (i.e. nonoxidative) reactions, with these occurring via attack of a free thiol (and usually the more reactive anion form) on a disulphide bond, with this resulting in an exchange of partners, and hence cross-link formation [Citation48,Citation49]. This process occurs more rapidly at higher temperatures and on protein unfolding, as this increases the accessibility of reaction sites [Citation48]. These reactions can be enhanced by the addition of free thiols [Citation50] and higher Ca2+ concentrations [Citation51].

Exposure to both heat and H2O2 appears to have a greater effect than either agent alone, with this ascribed to the occurrence of a mechanism involving the formation of a sulfenic acid (RSOH species) at Cys121 in β-Lg mediated by H2O2, once this residue is made accessible by thermal unfolding [Citation46,Citation47]. Limited loss of Met and Trp (at very high H2O2 concentration) has also been detected with both β-Lg and α-Lac on treatment with heat and H2O2 [Citation46,Citation47]. Increased exposure to heat (longer times and higher temperatures) and higher concentrations of H2O2 have been shown to enhance the extent of modification, as do combinations of these two factors [Citation46,Citation47]. No protection against damage was detected when radical trapping agents were included, indicating that these are molecular (two electron) and not radical (one electron) reactions [Citation46]. These data indicate that oxidative damage can be selective with regard to the residues that are modified, and that unfolding of the proteins is a critical factor with regard to the extent of damage. Decreasing heat loads and oxidant exposure would therefore be expected to minimise whey protein modification.

Photo-oxidation arising from light exposure of milk preparations containing riboflavin (vitamin B2, an endogenous component of milk) can induce changes in milk protein structure (e.g. polymerisation, secondary and tertiary structure of specific proteins), as well as inducing the formation of protein carbonyls, the Tyr oxidation products, di-tyrosine (di-Tyr) and 3,4-dihydroxyphenylalanine (DOPA), and the Trp-derived species, N-formylkynurenine (NFK) and kynurenine (Kyn) [Citation52,Citation53]. Different oxidant systems can therefore generate different patterns and extents of damage, and involve alternative mechanisms, though the overall effects (e.g. aggregation) may be similar. Light exposure has been used as a nonthermal technology to control pathogens and extend product shelf-life but this may result in increased formation of carbonyls and hence protein damage [Citation54].

Presence of oxidation, glycation, and racemised whey proteins in IF

Whey protein modification is a potential health risk for infants fed with IF, and there is convincing data that indicates that breastmilk has considerable health benefits (reviewed [Citation55]). summarises the large number of different types and levels of protein modifications detected in IF samples. At a macromolecular level, these include both reducible (presumed to be disulphide-linked species, due to their loss on diothiothreitol treatment) and (multiple types of) nonreducible protein aggregates [Citation56]. Evidence has been presented for the cross-linked species di-Tyr (a species arising from radical reactions), as well as lysinoalanine (LAL), lanthionine (LAN), and the precursor species for the latter two products, dehydroalanine (DHA) () [Citation56,Citation57]. Whether the DHA arises from base-catalysed or radical-mediated reactions are unknown. Significant levels of protein carbonyls are also present in the samples, as detected by assaying total carbonyl content and also by using immunoblotting on proteins separated by SDS-PAGE, with the latter experiments indicating that a significant quantity of the carbonyls is present on high-molecular-mass aggregates [Citation56]. The exact nature of the proteins involved and the sites within these proteins remain to be determined, though it is clear that there are significant structural changes to the IF proteins induced by processing [Citation58].

Table 6. Selected values of concentrations (nmol/mg protein except where otherwise indicated) of protein oxidation products, advanced glycation endproducts (AGEs) and racemised AAs present in powdered infant formula samples.

Modifications have also been detected on IF proteins at the AA level, with evidence reported for the formation of Trp oxidation products (N-formylkynurenine, NFK; kynurenine, Kyn; and 3-hydroxykynurenine, 3OHKyn), di-Tyr, Phe-derived materials (3-hydroxyPhe, m-Tyr) and the Met oxidation product, methionine sulfoxide [Citation56,Citation59]. Some of these species are consistent with the species detected on isolated whey proteins exposed to specific oxidant systems (e.g. heat/H2O2 and also light in the presence of riboflavin) [Citation46,Citation47,Citation52,Citation60,Citation61]. A number of the species detected are consistent with the occurrence of radical reactions (e.g. di-Tyr, which appears to be only formed by radical reactions), but others (e.g. methionine sulfoxide) can be formed by both radical and molecular (two electron) processes (e.g. direct oxidation by H2O2) [Citation62]. A number of these materials may undergo additional reactions (e.g. redox cycling) that may exacerbate damage [Citation63]. Furthermore, some of these products (e.g. those from Trp) may have multiple biological activities and contribute to disease, if taken up [Citation64].

IF also contain significant levels of advanced glycation endproducts (AGE) materials including the early stage product furosine, and well-characterised AGEs including Nɛ-(carboxymethyl)lysine (CML), Nɛ-(carboxyethyl)lysine (CEL), pyrraline, and protein crosslinks, such as pentosidine (from Lys and Arg residue linked by a pentose), glyoxal lysine dimer (GOLD), methylglyoxal lysine dimer (MOULD), and Arg-derived products such as argpyrimidine and hydroimidazolone isomers [Citation56,Citation65–72]. The levels of these materials have been examined using a wide variety of both direct and indirect methodologies [Citation66,Citation73,Citation74]. AGE is a chemically heterogeneous group of compounds. AGE formation in dairy products involve Maillard reactions, but the reaction conditions differ greatly depending on thermal load during processing [Citation75,Citation76]. Multiple AGEs have been characterised in foods (for a review of experimental methods, see Aalaei et al. [Citation77]) and in human tissues, possibly as a result of dietary exposure, including the Lys-derived products: Nɛ-(carboxymethyl)lysine (CML), Nɛ-(carboxyethyl)lysine (CEL), pyrraline, and protein crosslinks such as pentosidine (from Lys and Arg residue linked by a pentose), GOLD, MOULD and Arg-derived products including argpyrimidine and hydroimidazolone isomers. Changes in AGE levels, most commonly measured by mass spectrometry [Citation65], have been used to assess glycation/glycooxidation status in foods [Citation78,Citation79].

Analyses for the presence of D-AAs showed that significant levels of these un-natural AA isomers are also present, though whether these arise via oxidation or high temperature/high pH reactions is unclear [Citation56]. In some cases the levels of these species are very high, but it is impossible to eliminate the possibility that some of these arise during processing for analysis, rather than these being present endogenously. Further work needs to be carried out to clarify this point.

Comparison of IF samples containing native bovine proteins with those that contain hydrolysed proteins (hypoallergenic brands, which contain predominantly small peptides and free AAs), shows that most markers of modification are consistently present at higher levels in IF with hydrolysed proteins [Citation56]. This has been ascribed to the additional processing required to generate these samples (i.e. longer and more complex production times) as well as the decreased steric and electronic hindrance for reactions at free AAs/small peptides when compared to intact proteins. Thus many modification reactions occur more rapidly with small peptides/free AAs, than with intact proteins, though this is not always the case. For glycation reactions, a further factor that may enhance the extent of modification in the hydrolysed protein brands, is the elevated level of N-terminal amines generated on proteolysis of the original protein peptide bonds [Citation80]. This may be a significant contributor to the higher levels of AGEs present in these samples. However, the levels of side-chain derived species, such as furosine and CML (formed from reaction at the ε-amine group of Lys side-chains) have also been reported to be approximately double those present in the intact protein brands [Citation56].

Biological effects of oxidation, glycation, and racemised AAs

The impact of modified proteins on human health has been investigated to only a limited extent [Citation81], but there is increasing interest in the effects of oxidised proteins on human health [Citation82]. Consumption of oxidised food components can increase the level of oxidative stress in living tissues and this appears to contribute to the development of some diseases [Citation83–85]. This area has been recently reviewed by Delgado-Andrade and Fogliano [Citation86]. This is considered to be associated with the exposure of the gastrointestinal tract and internal organs to potentially cytotoxic and mutagenic materials [Citation87–89]. Oxidised AAs/peptides may impair homeostasis and cell toxicity via multiple different mechanisms. For example, L-Phe can be oxidised by HO. to give m-Tyr, which is cytotoxic by a pathway that appears to involve incorporation of the oxidised AA into proteins [Citation90]. Dietary intake of oxidised Tyr (e.g. di-Tyr) in rats has been reported to induce oxidative damage and hepatic fibrosis via MAPK/TGF-β1 pathway [Citation91]. Exposure to various Kyn species (Trp oxidation products) appears to be injurious as these have been associated with both neurotoxicity and pathogenesis of intestinal diseases [Citation44,Citation92–94]. The high levels of methionine sulfoxide (up to 74% of parent Met) in some milk products appears to be responsible for inducing changes in redox homeostasis, thus showing a toxic potential [Citation59,Citation95]. This may arise from the reduction of the methionine sulfoxide by the family of methionine sulfoxide reductase enzymes present in most organisms (including humans). These enzymes require reducing equivalents from the thioredoxin/thioredoxin reductase/NADPH system, with the consumption of NADPH resulting in a depletion of reducing equivalents, an oxidative stress, and a change in the redox homeostasis of the cell. Oral intake of some oxidised AAs has been shown to induce hepatic and renal fibrosis in mice, possibly via impairment of antioxidant defence systems and modification of the Nrf2-ARE gene pathway [Citation96]. In addition, increased levels of protein carbonylation, di-Tyr and advanced protein oxidation products have been found in the liver, kidney and blood of mice, in response to the intake of modified materials, consistent with these causing oxidative injury in vivo [Citation96].

The safety of Maillard reaction products (MRPs) is a concern, as some of these (e.g. acrylamide) are potential, or known, mutagens or carcinogens [Citation97,Citation98]. This area has been recently reviewed [Citation99,Citation100]. However, some MRPs have also been proposed to exert antioxidative, antibiotic, and antiallergic activity [Citation101,Citation102]. Thus reductones have been shown to possess strong antioxidative capacity through electron transfer, hydrogen atom donation and metal ion chelation (e.g. of Cu and Fe ions which might otherwise act as pro-oxidants) [Citation101–104]. Food-derived AGEs have, however, also been reported to promote oxidative stress and inflammation, and contribute to chronic disease [Citation100,Citation105]. A recent study has demonstrated that oral intake of CML promotes its accumulation in the gastrointestinal tract of rats, stimulates inflammatory responses by downregulating enzymatic antioxidative pathways and increases the levels of inflammatory cytokines [Citation106]. Long-term exposure of rats to dietary AGEs has been reported to increase colon permeability, modulate gut microbial ecology in a detrimental manner and induce host metabolic disturbances which can adversely impact host health [Citation107,Citation108]. In contrast, other studies have reported an absence of deleterious effects [Citation109], and even beneficial effects of dietary AGEs [Citation99], with a significant increase in the total antioxidant capacity of plasma from rats observed after oral administration of a diet supplemented with pronyllysine (a Lys product) [Citation110]. Whether dietary AGEs have a detrimental or beneficial impact on human health is controversial, due to difficulty in carrying out controlled trials [Citation109,Citation111].

As non-native AAs may be absorbed across the gastrointestinal tract, the presence of modified whey protein species in foods or IF may pose a health risk to infants, and consequently a number of studies have examined both the uptake of these modified AAs and peptides across cell monolayers, and the effects of these materials on cells in culture and animal growth [Citation112].

Exposure (for 24–48 h) of nondifferentiated Caco2 cells (a human intestinal epithelial cell line) to a range of modified AAs species (oxidised, glycated, D-isomers; either singly or as mixtures), at concentrations similar to those reported to be present in IFs (e.g. 20 µM), does not appear to have deleterious effects on cell growth [Citation113]. In some cases, increased metabolic activity (as measured by MTT assay) was detected compared to control cells [Citation113]. However, exposure to combined oxidation products, AGEs or D-AAs for 2 h at concentrations of 20 or 200 µM, increased protein carbonyl levels in the cells in a dose-dependent manner, consistent with the induction of oxidative stress [Citation113]. Studies with individual protein modification products suggest that m-Tyr may be a driver of this change, possibly via mis-incorporation (in place of Phe or Tyr) into new proteins. Such incorporation has been shown in previous studies to result in the formation and/or accumulation of truncated and dysfunctional proteins [Citation90,Citation114,Citation115]. Other studies have reported increased extracellular matrix production induced by oxidised Tyr species in rat kidneys [Citation116].

Long term (e.g. 21 d) incubation of Caco2 cells on filter inserts in transwells results in the formation of intact monolayers and expression of transport proteins and brush border hydrolases [Citation117]. At the equivalent concentrations to those detected in IF, and a 2-h exposure time (to mimic gut transit times [Citation117]) the modified AAs did not induce changes in the transepithelial electrical resistance (TEER), or permeability as assessed using lucifer yellow [Citation113]. These data indicate that these concentrations of modified materials do not induce gross changes to the monolayers, or cell toxicity, under the conditions employed. Quantification of modified AAs added to the apical side of the transwells, at 20 or 200 µM, showed that there was no significant decrease in the concentration of these species over the 2-h period, indicating that these species do not rapidly equilibrate across the monolayer, and that these materials are stable over this incubation period [Citation113]. However, some material was detected (by LC-MS) in the basolateral compartment, consistent with limited monolayer penetration. The permeability was between 0.003 and 0.095% for the 20 µM concentration group, with 3OHKyn being the least abundant, and m-Tyr the highest. This high permeability of m-Tyr is consistent with the induction of protein carbonyls by this compound (see above). The permeability increased as the apical concentration was increased, with di-Tyr giving the highest basolateral concentrations [Citation113]. The levels of the modified AAs present in lysates from the Caco2 cells, after 2-h exposure, were too low to be detected [Citation113]. AGEs appear to be only slowly transported across Caco2 cell monolayers [Citation113,Citation118]. Whether transepithelial transport occurs via diffusion, AA transporters (eg B°, +, b°, +, and y+ [Citation119]), endocytosis, or other mechanisms remains to be established. Previous studies have shown that AGEs can penetrate across cell monolayers [Citation118,Citation120–122], but this appears to occur, in at least some cases, by uptake of di-peptides rather than free AAs. Thus, evidence has been presented for uptake of di-peptides containing CML, CEL, ArgP, and pyrraline [Citation118,Citation122], via specific peptide transporters (e.g. the proton-coupled peptide transporter 1, PEPT1). These di-peptides are then subject to intracellular hydrolysis to the free modified AAs, which subsequently arrive in the basolateral compartment via passive diffusion [Citation122]. Whether this is also true for oxidised AAs remains to be determined. Data have been reported on the plasma and urinary levels of CML in breast milk- and formula-fed infants consistent with uptake across the gastrointestinal tract in infants, and subsequent excretion in urine [Citation123]. Furthermore, infants fed on formula have been reported to have significantly increased levels of antibodies against oxidised low-density-lipoproteins, consistent with significant dietary exposure to oxidising species [Citation124].

Overall, these data suggest that modified AAs from IFs may induce mild oxidative stress to cells, as evidenced by an increase in cellular protein carbonyls [Citation113]. However, these products appear to penetrate across Caco2 cell monolayers to only a moderate extent under the conditions examined. The effects of long-term exposure, and potential accumulation [Citation106], have not been examined in great detail, and this may be significant given that infants are typically fed every few hours for many months. Whether this results in long-term adverse health effects is controversial, though some epidemiological studies have suggested that long-term feeding with IF can induce inflammatory responses [Citation125] and predispose to an increased risk of diabetes later in life [Citation82,Citation112,Citation126–128]. Whether this is related to exposure to the modified AAs present in IFs, or from other effects (e.g. immunological responses to bovine proteins [Citation129,Citation130]) remains to be determined.

Conclusions

Human intervention studies have provided evidence that consumption of whey proteins can boost antioxidant markers in blood and various organs. This health benefit is likely to be as a result of the contribution of reactive free thiol groups (Cys residues) for GSH synthesis and the presence of bioavailable bioactive peptides. However, whey proteins undergo considerable processing steps from raw milk to consumed products which makes them vulnerable to oxidation, glycation and racemisation. These modifications may have adverse effects on human health particularly when consumed over a prolonged period of time, though there is a lack of definitive data with regard to the specific effects of known concentrations of specific products. Further work is clearly warranted in this area. Alternative processing maps and storage conditions may be needed to protect whey proteins from oxidation during processing and capitalise on whey as a dietary antioxidant.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was supported by the European Cooperation in Science and Technology (COST) under Grant COST Action CA16112-NutRedOx Network, the Novo Nordisk Foundation [grant NNF13OC0004294 to MJD], Science Foundation Ireland [grant 16/RC/3835 to LG], Irish Department of Agriculture, Food and Fisheries [grant FIRM 15F604 to MC] and Marie Skłodowska-Curie-Career FIT by Enterprise Ireland and European Union’s Horizon 2020 [grant MF2018-0151 to EA].

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.