Next Article in Journal
Ceramide Synthase 6: Comparative Analysis, Phylogeny and Evolution
Previous Article in Journal
A Marine Diterpenoid Modulates the Proteasome Activity in Murine Macrophages Stimulated with LPS
Previous Article in Special Issue
Critical Assessment of Methods to Quantify Biofilm Growth and Evaluate Antibiofilm Activity of Host Defence Peptides
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Milk-Derived Antibacterial Peptides in Modern Food Biotechnology: Their Synthesis, Applications and Future Perspectives

by
Muhammad Usman Khan
1,2,
Maryam Pirzadeh
3,
Carola Yvette Förster
4,
Sergey Shityakov
4,* and
Mohammad Ali Shariati
5,*
1
Bioproducts Sciences and Engineering Laboratory (BSEL), Washington State University, Richland, WA 99354, USA
2
Department of Energy Systems Engineering, Faculty of Agricultural Engineering and Technology, University of Agriculture, Faisalabad 38000, Pakistan
3
Department of Food Science and Technology, Faculty of Agriculture, Sarvestan Branch, Islamic Azad University, Sarvestan 73451-173, Iran
4
Department of Anesthesia and Critical Care, University of Würzburg, 97080 Würzburg, Germany
5
Laboratory of Biocontrol and Antimicrobial Resistance, Orel State University Named After I.S. Turgenev, 302026 Orel, Russia
*
Authors to whom correspondence should be addressed.
Submission received: 20 August 2018 / Revised: 25 September 2018 / Accepted: 26 September 2018 / Published: 5 October 2018
(This article belongs to the Special Issue Antimicrobial Peptides: Development, Conjugation, and Beyond)

Abstract

:
Milk-derived antibacterial peptides (ABPs) are protein fragments with a positive influence on the functions and conditions of a living organism. Milk-derived ABPs have several useful properties important for human health, comprising a significant antibacterial effect against various pathogens, but contain toxic side-effects. These compounds are mainly produced from milk proteins via fermentation and protein hydrolysis. However, they can also be produced using recombinant DNA techniques or organic synthesis. This review describes the role of milk-derived ABPs in modern food biotechnology with an emphasis on their synthesis and applications. Additionally, we also discuss the mechanisms of action and the main bioproperties of ABPs. Finally, we explore future perspectives for improving ABP physicochemical properties and diminishing their toxic side-effects.

Graphical Abstract

1. Introduction

Milk-derived antibacterial peptides (ABPs) are a plentiful group of biochemical substances produced from milk with a molecular weight below 10 kD [1,2,3,4]. Most of these ABP compounds are produced by organic synthesis, in vitro via enzymatic proteolysis (fermentation or protein hydrolysis) of milk proteins, in vivo by molecular cloning using natural sequences [5].
Milk of all mammalian species as a heterogeneous mixture is produced by lacteal glands [6,7]. It comprises approximately 3.5% of total protein fraction, including 80% of casein and the rest of the whey proteins, which exhibits a variety of biochemical and physiological properties [8,9,10]. In turn, casein and whey fractions have been subdivided into α-, β- and κ-caseins, and whey lactalbumins and lactoglobulins with some additional proteins, such as immunoglobulins, enzymes, and mineral-binding proteins [11,12,13].
The various multifunctional properties of milk-derived ABPs have been extensively studied to investigate their positive impact on human health [14,15]. Primarily, this naturally occurring bioactive peptides are low-density molecules (5–90 amino acids) representing their bioactivity features only if they are separated from the parental proteins [16] being produced in several different forms [17].
All ABPs in this review are mainly divided into four classified groups: (i) milk-derived, such as isracidin αs1 f(1–23) and lactoferricin f(17–41); (ii) whey-derived peptides such as β-lactoglobulin f(15–20); (iii) casein-derived, such as κ-casecidin and its partial peptide fragments, and (iv) lysozyme-derived ABPs.
The degree of ABP antibacterial activities depends on the biophysical features such as negatively and positively charged groups of peptides, molecular size, conformational and hydrophobic properties [18]. Additionally, some milk-derived ABPs may exploit routine regulating activities in the human body as products of proteolytic reactions, such as enzymatic hydrolysis and fermentations [19]. Proteolytic enzymes from the dairy products, such as milk plasmin might hydrolyze proteins to release ABPs during milk processing and storage [20]. Moreover, many types of bacteria, which reside in the gastrointestinal tract of animals and humans, can produce bioactive ABPs from milk during its digestion [21].
In general, milk-derived ABPs have drawn much attention of the scientific community worldwide due to their biological versatility with the ability to formulate them with pharmaceutical ingredients and health-promoting food supplements [22]. Moreover, these peptides are also prone to polymorphism, so they occur in multiple isoforms [23,24,25,26]. Here, we discuss the role of milk-derived ABPs in modern food biotechnology, focusing on their application, production, and future perspectives.

2. Mechanism of Action

Antibacterial activity of ABPs depends on their cationic and hydrophobic amino acid composition [27] with a mild positive charge (+4) under physiological conditions (Table 1). Most of the charged ABPs disrupt lipid membranes altering their permeability and transport properties [27,28,29]. In particular, positively charged amino acids are extremely active against Gram-positive and Gram-negative bacteria [30,31,32,33,34,35,36,37,38,39,40]. In comparison to conventional antibiotics, ABPs have considered interacting with bacterial DNA and RNA [32,41], forming a hydrogen bond with substances such as 3,4-dihydroxyphenylalanine [33,42] or sodium chloride [30,39]. This ABPs antibacterial action would lead to the membrane dissolution or a specific binding to nucleic acids [34,35,43,44].

3. Milk-Derived Antibacterial Peptides

A diversity of peptides comes from the different food protein sources (so-called functional food) with some specific properties such as antibacterial, anti-carcinogenic, hormone-tropic, immunomodulatory and antihypertensive effects [36,37,38,39]. The main source of bioactive peptides is dairy products, such as milk [40,41,42,43].
Milk contains different nutrient as an entire source of various proteins and peptides [44,45,46]. Since milk contains a wide spectrum of peptides, the concept of consideration of milk as a high nutritive source increased its attractions among consumers. The lower size of peptides provides them with the ability of quick diffusion into the cell membrane of pathogens and makes the leaky as direct suppressing and antibacterial actions [47,48,49,50]. Overall, naturally occurring milk peptides might even diminish the time span of disease, which is the result of their antibacterial effect on pathogens [51,52].
Milk is a well-balanced protein source, containing two main fractions, such as casein and whey [53] to satisfy mammalian offspring necessitates. Milk has also immunological peptides as bacterial inhibitors to decrease the growth of pathogens. Furthermore, proteins such as lactoferrin (Lf), lactoperoxidase, and lysozyme are among those protecting ingredients [54,55].
Considering milk as a primary nutritional source presenting of more than 10,000 nutritional compounds in milk has given a perspective of being recognized as a functional food. Beyond basic functional compounds, bioactive peptides are the main group of milk health affecting constituents, which provides an array of functional activities with treating properties such as reducing grade inflammation, antibacterial effects, etc. [56,57,58].
Amino acids and nitrogen constituents are the two main targets of milk consumption [59]. Milk proteins are precipitated in its isoelectric pH 4.6 at 20 °C, following by the fragmentation mainly to β-lactoglobulin-β-LG-(7–12% of total skim milk protein), -lactalbumin-LA-(2–5% of skim milk total protein), serum albumin (SA), Immunoglobulins-Ig, lactotransferrin (lactoferrin-Lf) and β2-microglobulin. Table 2 and Table 3 summarize some ABPs with the corresponding minimal inhibitory concentrations together with peptide production approaches and antibacterial effects.

4. Whey-Derived Antibacterial Peptides

Lactoferrin, is a glycoprotein, which consists of small fractions of milk proteins. This protein produces structural fragments proteolytic treatment than create antibacterial potencies as ABPs against various bacterial pathogens [50,71,72]. The antibacterial properties of Lf are dependent on its charge, hydrophobic properties, or secondary structure as it has is a higher affinity for iron atom than most of the proteins in Streptococcus mutans, Vibrio cholerae, Escherichia coli, and Legionella pneumophila. In fact, Lf follows multiple mechanisms in suppressing pathogens [64,73]. Some of them are based on positively charged arginine and tryptophan residues that facilitate the interaction with negative charges of lipopolysaccharides of the lipid membrane, leading to a bacterial death.
Lactoferrin, itself has pathogenic properties, acting as a chelating agent by capturing the ions (apo-Lf) important for bacteria [31,74,75].
Lactoferrin was identified in two protein variants as human (Lf H) and bovine (Lf B), with antibacterial potentials on both positive and negative bacteria strains. The N1-domain of Lf acts against pathogens such as Bacillus subtilis, Escherichia coli, and Pseudomonas aeruginosa, but effective against the fermentation bacteria [76]. Unlike bovine whey, in which β-lactoglobulin contains 50% of whole protein, it has no presence in human milk. Trypsin digestion method of β-lactoglobulin produces four peptide fragments (f), including f(15–20), f(25–40), f(78–83) and f(92–100) active mainly against gram-positive bacteria. On the other hand, α-lactalbumin produces anti-gram-positive peptides after trypsin or chymotrypsin digestion [76].

5. Casein-Derived Antibacterial Peptides

Casein, comprising 80% of milk protein, is believed to be the main protein fraction of milk [77]. Although casein itself exhibits no any antibacterial effect, the ABPs, which released from its enzymatic digestion, may exert antibacterial activity as functional oligopeptides [78]. The digesting method of casein by chymosin at neutral pH produces some antibacterial agents, such as casecidin, lactenin, and isracidin, inhibiting the growth of some strains in vitro [79] by the N-terminal fragment of αs1-casein [79]. Additionally, chymosin proteolytic digestion may also generate other fragments, comprising caseicin A and B that inhibit several pathogens, among them Staphylococcus aureus, Sarcina lutea, Bacillus subtilis, Diplococcus pneumoniae, and Streptococcus pyogenes [73,80]. These two fragments from αs1-casein with the ability to inhibit Cronobacter sakazakii, Salmonella and Klebsiella, and Gram-positive Staphylococcus aureus in the powdered food [81,82,83,84]. Furthermore, casecidin-I as a cationic αs2-casein-derived peptide (165–203 amino acids) could suppress the growth of Gram-negative (E. coli) and Gram-positive (Staphylococcus carnosus) bacteria [85]. Some other αS2-casein-derived peptides f(181–207), f(175–207), f(164–207) possess antibacterial properties versus pathogenic bacteria [65,73].
On the other hand, kappacin (nonglycosylated κ-casein), is a peptide of human milk acidification, belonging to the phosphorylated form of ABP. Kappacin shows bactericidal potential against Gram-positive (Streptococcus mutans) and Gram-negative (Porphyromonas gingivals) bacteria, whereas its non-phosphorylated and glycosylated forms might have no effect on some groups of bacteria such as Streptococcus mutans [86]. κ-casecidin and its partial peptide fragments are produced via trypsin digestion method of casein to reduce the growth of S. aureus, E. coli and S. typhimurium. Casein macropeptide (CMP) derived from κ-casecidin can disrupt replication of invasive bacteria through the specific binding to their receptors on the cell wall [85]. In addition, CMP also prevents the propagation of microflora, forming dental plaques caused by Streptococcus mutans [86].

6. Lysozyme-Derived Antibacterial Peptides

There are different types of lysozyme available in milk whose properties differ from each other, depending on their structure, physiochemical properties, and the ability of binding to calcium [87]. In fact, milk-derived lysozyme has a significant potential of being recognized as an antibacterial compound through its lysis reaction. Some lysozyme-derived ABPs, such as RAWVAWR-NH2 and IVSDGNGMNAWVAWR-NH2, were found to exhibit antibacterial activity with the ability to rapidly enter both E. coli and Staphylococcus aureus [88]. These peptides can cause a significant perturbation of membranes due to their strong affinity to the lipids with negatively charged head groups [88]. Additionally, 87–114 and 87–115 amino acid residues of chicken and human lysozyme were tested for bactericidal activity against Gram-positive and Gram-negative bacteria in the attempt to design novel ABPs [89]. However, ion concentration changes of sodium, potassium, ammonium, magnesium, and calcium might influence the lysozyme and probably its ABPs antibacterial activity by a reduction of their minimum inhibitory concentration (MIC) in the experiment [87].

7. Production of Antibacterial Peptides

In general, most ABPs belong to biologically active protein fragments that are mainly being produced with some modifications either by enzymatic (proteinases and peptidase) digestion and fermentation processes, or by lactic acid bacteria (LAB) hydrolysis, not excluding the application of some peptidases from other organisms, such as animals, plants or even fungi, etc. [90]. In this regard, four main strategies were elaborated and optimized for the industrial production of ABPs: fermentation, protein hydrolysis with extracellular enzymes, recombinant DNA method, and organic synthesis (Table 4).

7.1. Fermentation by Lactic Acid Bacteria

Fermentation is a process in which some peptidases are produced by LABs decomposing proteins into their structural fragments. Currently, this methodology is considered outdated for efficient production of functional food [92,93]. Conversely, the LAB usage to produce bioactive peptides such as ABPs from milk proteins is a straightforward process strategy believed to be GRAS, “generally recognized as safe” [93].
Fermentation is used to decompose milk ingredients, producing better taste, smell and color (organoleptic properties) together with bioactive peptides active against Salmonella enteridi and Escherichia coli [94]. Lysozyme, H2O2, lactoferrin and various ABPs are known as substances, which reduce blood pressure and stimulate the innate immune system [95]. Moreover, they also act as food preservatives against Escherichia coli, Listeria monocytogenes, Staphylococcus aureus, and Salmonella typhimurium [93].
On the other hand, ABPs, as bacteriocins (colicins) may refer to antibacterial peptides mainly from the Gram-positive bacteria (LABs), which secrete these components to the surrounded media and suppress pathogen growth [96,97,98,99]. Therefore, the development of a system introducing sufficient production, distribution, and delivery of ABPs is of primary concern for modern food biotechnology to improve the antibacterial activity of naturally manufactured compounds via bacteria [100,101].
In the fermentation process, LABs can produce lantibiotic bacteriocins, among them nisin, helveticin, lactacin, etc., which are secreted (nisin) by some genera including Lactococcus lactis, Pediococcus acidilactici, and P. pentosaceus. Pediocin is another bacteriocin, which inactivates L. monocytogenes, Enterococcus faecalis, S. aureus, and C. perfringens. Furthermore, nisin as a small cationic polypeptide is approved by FAO/WHO to be safe as a food supplement [102,103,104,105,106]. This ABP might prevent pathogenic effects of both types (Gram-positive and negative) of bacteria [20,106].
As protein-based synthetic components, bacteriocin are involved in the suppression of different the Gram-positive and negative bacteria via the interaction with lipid membranes due to their amphiphilic and hydrophobic properties [107]. Natamycin, another bacteriocin, a polyene antifungal agent produced by Streptomyces natalensis, is effective against molds and yeasts, but it has mild or no effect on bacteria or viruses [108]. Natamycin has a very low aqueous solubility, therefore, it needs to be applied at high concentration and is effective at very low levels [109]. In particular, reuterin and reutericyclin made by Lactobacillus reuteri are highly active against L. monocytogenes, E. coli (O157:H7), S. choleraesuis, Yersinia enterocolitica, Aeromonas hydrophila, and Campylobacter jejuni. Therefore, the development of a system introducing sufficient production, distribution, and delivery of ABPs is of primary concern for modern food biotechnology to improve the antibacterial activity of naturally manufactured compounds via bacteria [100,101].

7.2. Protein Hydrolysis with Extracellular Enzymes (Proteases)

Other strategies have been employed in modern food biotechnology to produce more effective ABPs [91]. In particular, the chymosin (rennin) proteolytic reactions of casein have resulted in the formation of some antibacterial peptides, such as isracidin, matching the N-terminal part of αs1-casein [64]. This casein-derived substance remains active against Staphylococcus aureus and Candida albicans [110]. Similarly, another peptide casiocidine is formed from the αs2-casein protein together with (183–207) and (164–179) fragments, via pepsin proteolytic processing [66,73]. Additionally, κ-casein has two antibacterial fragments after pepsin digestion as the (138–158) and (64–117) fragments named kappacine, which could kill cariogenic bacteria [76]. A proteolytic hydrolysis of β-casein by Lactobacillus helveticus PR4 creates the (184–210) fragment with antibacterial properties and the (138–158) fragment active against Str. mutans, E. coli and Porphyromonas gingivalis [76].
Apart from the above-mentioned peptides, other fragments can be formed due to ionic conditions and pH changes, leading to their precipitation without a separation phase [48]. For instance, caseinomacropeptide characterized by the reversed-phase high-performance liquid chromatography coupled with mass spectrometry (HPLC-MS) has antibacterial properties against Streptococcus mutans, Porphyromonas gingivalis and Escherichia coli [68].
Additionally, kappacin as the κ-casein-derived product active against S. mutans, E. coli, and Porphyromonas gingivalis [68] together with chymosin, which was found during proteolytic activities of sodium caseinate [62,111].
Pepsin proteolytic activities have been commonly employed to denature milk proteins and to produce various fragments for further evaluation with HPLC-MS of these oligopeptides, which turned out be active against Bacillus cereus, Staphylococcus aureus, Enterococcus faecalis, and Escherichia coli [77].
Two classes of antibacterial peptides are defined of fungal- and bacterial-origin, characterized by the cyclic or branched composition of ABPs [112]. Another classification subdivides them into (i) cryptic peptides as a product of enzymatic reactions, (ii) lantibiotics, such as nisin, (iii) defensin and cathelicidins related to the immune system [113,114]. Most ABPs are derived from native proteins by enzymatic hydrolysis to generate desired peptides or fragments by screening, fractionation, and purification [115].
Sometimes de novo peptide sequencing is needed by mass spectrometry to obtain the predicted ABP structure [116]. However, some peptides in the active fraction are sometimes not biologically active requiring additional bioinformatics analysis to screen them for their biological and physiological activity [117]. Therefore, the ABP purification method implies the use of the ammonium sulfate, which precipitates protein fragments with 80–100% fractional activity [117]. For a supernatant concentration, there are several approaches such as ammonium sulfate concentration adjustment, absorption-desorption technique, and organic solvent extraction [118].
By applying a salting out technique, one could extract bacteriocins of different microorganisms such as LABs [119], Pediococcus spp. [120], Lactococcus spp. [121], and Leuconostoc spp. [122]. In the previous study, the researchers found that the membrane benzylation followed by the dialysis with a cutoff of 2–3.5 kDa resulted in the highest extraction of smaller size bacterocins [118].
Milk-derived peptides from the pepsin digestion are active against a wide range of pathogens [62]. Caseinate fermentation by L. acidophilus DPC6026 produces caseicins A, B, and C [81]. Fermentation is one of the cheapest methods for the efficient production of ABPs in comparison to the proteinase approach [91].
Rana and co-authors have already used this method to evaluate and characterized ABP-like peptides from milk fermentation products by L. rhamnosus C6 [123]. Additionally, pepsin digestion method might be also useful to ABPs [124], which was confirmed by reversed-phase chromatography and sensitive radial diffusion method to characterize the antibacterial activity of separated fragments present in human milk [124].
Due to the interference in the ABPs identification, a technology named matrix-assisted laser desorption ionization–mass spectrometry (MALDI–MS) can be applied [125]. The technique was successfully implemented to observe a bacteriostatic effect of the human k-casein fragment (63–117) [85]. Additionally, hydrochloric acid can be supplemented as an activation factor for pepsin, trypsin, and chymotrypsin to denature casein with a subsequent release of various ABPs [45,54,126].

7.3. Antibacterial Peptides Synthesis by Recombinant DNA Method

Recombinant DNA technology has been widely used as an alternative to the aforementioned techniques to produce ABPs in high amounts [127,128]. This procedure is particularly useful for the synthesis of large ABPs (>150 amino acids) and proteins [129,130,131,132]. The overall strategy relies upon the construction of the ABP coding region with its subsequent cloning into a prokaryotic expression vector, allowing the production of ABP or several peptides, simultaneously. To achieve this goal, E. coli cells—the most widely used host—might be implemented as the expression system [133].
Since most ABPs represent a strong antibacterial activity against the expression vector cells and relative sensitivity to proteolytic enzymes, these peptides are usually expressed as fusion proteins to neutralize their inherent toxic properties and improve their expression levels [133]. Compared with isolation from natural sources and organic synthesis methods, the recombinant DNA approach provides the most cost-effective alternative for industrial (large-scale) ABP production. Table 5 summarizes the synthesis of milk-derived ABPs by using recombinant DNA technology.

8. Summary and Future Perspectives

In this review, we discuss the role of milk-derived ABPs in modern food biotechnology, focusing on their application and production. Although different methods (fermentation, protein hydrolysis, recombinant DNA technology, and organic synthesis) have been successfully applied to produce various ABPs from milk, their stability and solubility should to be considered. To enhance these features, some formulated excipients, such as amphiphilic cyclodextrins, might be used.
Cyclodextrins (CDs) are starch by-products of converting enzymes that are composed of a (1, 4)-linked glucopyranose and defined as α, β, γ according to the number of the glucose units (6, 7, and 8) in the molecule [138]. These amphiphilic molecules possess a lipophilic binding cavity that could mediate complexation with ABPs (Figure 1).
In some cases, the antibacterial activity of ABPs might be inhibited when these peptides are exposed to cholesterol [139]. Therefore, any addition of cyclodextrins may diminish this effect due to the cholesterol absorption by CDs [139].
Conversely, CDs are well known for increasing drug-like molecule solubility upon their complexation with the former molecules [140,141]. For instance, in the study of colicin, the β-CD (β-cyclodextrin) addition to the oleic acid (OA) solution provided better OA delivery and insertion into the lipid membrane [142].
The interaction of CDs with the cellular membranes causes its structural change, allowing ABPs to enter the cell [143]. The presence of hydroxyl groups and carbon core in the CD structure divides the molecule into a hydrophilic exterior and a hydrophobic interior as binding cavity [138,144]. Hydrophobic amino acids (mainly tyrosine and tryptophan) with aromatic rings are the driving force of interaction between amphiphilic cyclodextrins and ABPs due to the steric effects (Figure 1).
Another important approach alleviate ABP toxicity can be found in the replacement of highly toxic dimethylformamide and methylpyrrolidone organic solvents via hydrophilic cyclodextrin complexation/formulation of lipophilic peptides or using less toxic solvent analogs, such as 3-methoxy-3-methyl-1-butanol (MMB), PEG-400, glycerol and propylene carbonate.
The other strategies might be employed to increase the ABPs efficiency against pathogenic bacteria is to use them in combination with other antibacterials and prebiotics, such as milk oligosaccharides (MOs) [145]. It is well known that some MOs may attenuate pathogens because of their stimulation of the lactic acid and bifidobacteria growth in the gut [146]. Moreover, MOs may defend the human body against pathogens via the creation of entrapment system to inhibit their binding to epithelial cells [145]. This could be achieved by the hypothetical synergistic effect of dietary monosaccharides (DMs) and MOs where DMs might be taken up by the intestinal cell and used for the synthesis of modified cell surface glycoconjugates [145]. These surface glycoconjugates together with MOs might interfere with the adhesion of bacterial pathogens to the cell wall by the inhibition of this process (Figure 2).
Additional research is needed to investigate the pharmacokinetic/pharmacodynamic parameters of complexed ABPs and their ability to permeate different biological barriers, such as the blood-brain barrier (logBB determination) for more effective treatment of infectious diseases. Finally, the advent of nanobiotechnology allows for the design of highly effective hybrid nanomaterials with synergistic effects of ABPs and nanoparticles, such as metals and their oxides, metal-organic frameworks, and nanoclays, to enhance the biodistributional and barrier properties of ABP formulations.

Author Contributions

Conceptualization, S.S., M.A.S.; Writing-original draft, M.U.K., M.P., C.F., S.S., M.A.S.; Final editing, S.S., M.A.S.

Funding

This work was supported in the intramural funding by the State University of Orel.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mohanty, D.P.; Mohapatra, S.; Misra, S.; Sahu, P.S. Milk derived bioactive peptides and their impact on human health—A review. Saudi J. Biol. Sci. 2016, 23, 577–583. [Google Scholar] [CrossRef] [PubMed]
  2. Memarpoor-Yazdi, M.; Asoodeh, A.; Chamani, J. A novel antioxidant and antimicrobial peptide from hen egg white lysozyme hydrolysates. J. Funct. Foods 2012, 4, 278–286. [Google Scholar] [CrossRef]
  3. Tomioka, H.; Nakagami, H.; Tenma, A.; Saito, Y.; Kaga, T.; Kanamori, T.; Tamura, N.; Tomono, K.; Kaneda, Y.; Morishita, R. Novel anti-microbial peptide SR-0379 accelerates wound healing via the PI3 Kinase/Akt/mTOR pathway. PLoS ONE 2014, 9, e92597. [Google Scholar] [CrossRef] [PubMed]
  4. Mansour, S.C.; Pena, O.M.; Hancock, R.E. Host defense peptides: Front-line immunomodulators. Trends Immunol. 2014, 35, 443–450. [Google Scholar] [CrossRef] [PubMed]
  5. Kim, S.K.; Wijesekara, I. Development and biological activities of marine-derived bioactive peptides: A review. J. Funct. Foods 2010, 2, 1–9. [Google Scholar] [CrossRef]
  6. Moylan, D.C.; Pati, S.K.; Ross, S.A.; Fowler, K.B.; Boppana, S.B.; Sabbaj, S. Breast Milk Human Cytomegalovirus (CMV) Viral Load and the Establishment of Breast Milk CMV-pp65-Specific CD8 T Cells in Human CMV Infected Mothers. J. Infect. Dis. 2017, 216, 1176–1179. [Google Scholar] [CrossRef] [PubMed]
  7. Witkowska-Zimny, M.; Kaminska-El-Hassan, E. Cells of human breast milk. Cell. Mol. Biol. Lett. 2017, 22, 11. [Google Scholar] [CrossRef] [PubMed]
  8. Sindayikengera, S.; Xia, W.-S. Nutritional evaluation of caseins and whey proteins and their hydrolysates from Protamex. J. Zhejiang Univ. Sci. B 2006, 7, 90–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Mignone, L.E.; Wu, T.; Horowitz, M.; Rayner, C.K. Whey protein: The “whey” forward for treatment of type 2 diabetes? World J. Diabetes 2015, 6, 1274–1284. [Google Scholar] [CrossRef] [PubMed]
  10. Tacoma, R.; Fields, J.; Ebenstein, D.B.; Lam, Y.-W.; Greenwood, S.L. Characterization of the bovine milk proteome in early-lactation Holstein and Jersey breeds of dairy cows. J. Proteom. 2016, 130, 200–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Jakala, P.; Vapaatalo, H. Antihypertensive Peptides from Milk Proteins. Pharmaceuticals 2010, 3, 251–272. [Google Scholar] [CrossRef] [PubMed]
  12. Aleixandre, A.; Miguel, M.; Muguerza, B. Peptides with antihypertensive activity from milk and egg proteins. Peptidos antihipertensivos derivados de proteinas de leche y huevo. Nutr. Hosp. 2008, 23, 313–318. [Google Scholar] [PubMed]
  13. Yamamoto, N.; Takano, T. Antihypertensive peptides derived from milk proteins. Nahrung 1999, 43, 159–164. [Google Scholar] [CrossRef]
  14. Davoodi, S.H.; Shahbazi, R.; Esmaeili, S.; Sohrabvandi, S.; Mortazavian, A.; Jazayeri, S.; Taslimi, A. Health-Related Aspects of Milk Proteins. Iran. J. Pharm. Res. 2016, 15, 573–591. [Google Scholar] [PubMed]
  15. Mohanty, D.; Jena, R.; Choudhury, P.K.; Pattnaik, R.; Mohapatra, S.; Saini, M.R. Milk Derived Antimicrobial Bioactive Peptides: A Review. Int. J. Food Prop. 2016, 19, 837–846. [Google Scholar] [CrossRef]
  16. Davidson, P.M.; Critzer, F.J.; Taylor, T.M. Naturally occurring antimicrobials for minimally processed foods. Annu. Rev. Food Sci. Technol. 2013, 4, 163–190. [Google Scholar] [CrossRef] [PubMed]
  17. Berge, G.; Eliassen, L.T.; Camilio, K.A.; Bartnes, K.; Sveinbjrnsson, B.; Rekdal, O. Therapeutic vaccination against a murine lymphoma by intratumoral injection of a cationic anticancer peptide. Cancer Immunol. Immunother. 2010, 59, 1285–1294. [Google Scholar] [CrossRef] [PubMed]
  18. Gould, G.W.; Russell, N.J. Food Preservatives, 2nd ed.; Kluwer Academic/Plenum Publishers: New York, NY, USA, 2003. [Google Scholar]
  19. Cleveland, J.; Montville, T.J.; Nes, I.F.; Chikindas, M.L. Bacteriocins: Safe, natural antimicrobials for food preservation. Int. J. Food Microbiol. 2001, 71, 1–20. [Google Scholar] [CrossRef]
  20. Albenzio, M.; Santillo, A.; Caroprese, M.; Della Malva, A.; Marino, R. Bioactive Peptides in Animal Food Products. Foods 2017, 6, 35. [Google Scholar] [CrossRef] [PubMed]
  21. Bhat, Z.F.; Kumar, S.; Bhat, H.F. Bioactive peptides of animal origin: A review. J. Food Sci. Technol. 2015, 52, 5377–5392. [Google Scholar] [CrossRef] [PubMed]
  22. Park, Y.W.; Nam, M.S. Bioactive Peptides in Milk and Dairy Products: A Review. Korean J. Food Sci. Anim. Resour. 2015, 35, 831–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mokoena, M.P. Lactic Acid Bacteria and Their Bacteriocins: Classification, Biosynthesis and Applications against Uropathogens: A Mini-Review. Molecules 2017, 22, E1255. [Google Scholar] [CrossRef] [PubMed]
  24. Deegan, L.H.; Cotter, P.D.; Hill, C.; Ross, P. Bacteriocins: Biological tools for bio-preservation and shelf-life extension. Int. Dairy J. 2006, 16, 1058–1071. [Google Scholar] [CrossRef]
  25. Gálvez, A.; Abriouel, H.; Lucas, L.R.; Omar, N.B. Bacteriocin-based strategies for food biopreservation. Int. J. Food Microbiol. 2007, 120, 51–70. [Google Scholar] [CrossRef] [PubMed]
  26. Rodríguez, J.M.; Martínez, M.I.; Kok, J. Pediocin PA-1, a wide-spectrum bacteriocin from lactic acid bacteria. Crit. Rev. Food Sci. Nutr. 2002, 42, 91–121. [Google Scholar] [CrossRef] [PubMed]
  27. Maria-Neto, S.; de Almeida, K.C.; Macedo, M.L.R.; Franco, O.L. Understanding bacterial resistance to antimicrobial peptides: From the surface to deep inside. Biochim. Biophys. Acta 2015, 848, 3078–3088. [Google Scholar] [CrossRef] [PubMed]
  28. Wimley, W.C. Describing the Mechanism of Antimicrobial Peptide Action with the Interfacial Activity Model. ACS Chem. Biol. 2010, 5, 905–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Nguyen, L.T.; Haney, E.F.; Vogel, H.J. The expanding scope of antimicrobial peptide structures and their modes of action. Trends Biotechnol. 2011, 29, 464–472. [Google Scholar] [CrossRef] [PubMed]
  30. Guilhelmelli, F.; Vilela, N.; Albuquerque, P.; Derengowski, L.D.S.; Silva-Pereira, I.; Kyaw, C.M. Antibiotic development challenges: The various mechanisms of action of antimicrobial peptides and of bacterial resistance. Front. Microbiol. 2013, 4, 63–74. [Google Scholar] [CrossRef] [PubMed]
  31. Shukla, A.; Fleming, K.E.; Chuang, H.F.; Chau, T.M.; Loose, C.R.; Stephanopoulos, G.N. Controlling the release of peptide antimicrobial agents from surfaces. Biomaterials 2010, 31, 2348–2357. [Google Scholar] [CrossRef] [PubMed]
  32. Zasloff, M. Antimicrobial peptides of multicellular organisms. Nature 2002, 415, 389–395. [Google Scholar] [CrossRef] [PubMed]
  33. Corrales-Urena, Y.R.; Sanchez, A.; Pereira, R.; Rischka, K.; Kowalik, T.; Vega-Baudrit, J. Extracellular micro and nanostructures forming the velvet worm solidified adhesive secretion. Mater. Res. Express 2017, 4, 125013. [Google Scholar] [CrossRef] [Green Version]
  34. Bechinger, B.; Lohner, K. Detergent-like actions of linear amphipathic cationic antimicrobial peptides. Biochim. Biophys. Acta 2006, 1758, 1529–1539. [Google Scholar] [CrossRef] [PubMed]
  35. Hancock, R.E.; Sahl, H.G. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat. Biotechnol. 2006, 24, 1551–1557. [Google Scholar] [CrossRef] [PubMed]
  36. Pellegrini, A. Antimicrobial peptides from food proteins. Curr. Pharm. Des. 2003, 9, 1225–1238. [Google Scholar] [CrossRef] [PubMed]
  37. Vogel, H.J.; Schibli, D.J.; Weiguo, J.; Lohmeier-Vogel, E.M.; Epand, R.F.; Epand, R.M. Towards a structure-function analysis of bovine lactoferricin and related tryptophan and arginine containing peptides. Biochem. Cell Biol. 2002, 80, 49–63. [Google Scholar] [CrossRef] [PubMed]
  38. Benkerroum, N. Antimicrobial peptides generated from milk proteins: A survey and prospects for application in the food industry. A review. Int. J. Dairy Technol. 2010, 63(3), 320–338. [Google Scholar] [CrossRef]
  39. Hayes, M. Food Proteins and Bioactive Peptides: New and Novel Sources, Characterisation Strategies and Applications. Foods 2018, 7, 38. [Google Scholar] [CrossRef] [PubMed]
  40. El-Salam, M.H.A.; El-Shibiny, S. Bioactive peptides of buffalo, camel, goat, sheep, mare, and yak milks and milk products. Food Rev. Int. 2013, 29, 1–23. [Google Scholar] [CrossRef]
  41. Lemes, A.C.; Sala, L.; Ores, J.D.C.; Braga, A.R.C.; Egea, M.B.; Fernandes, K.F. A review of the latest advances in encrypted bioactive peptides from protein-rich waste. Int. J. Mol. Sci. 2016, 17, 950. [Google Scholar] [CrossRef] [PubMed]
  42. Xiang, N.; Lyu, Y.; Bhunia, A.; Narsimhan, G. Antimicrobial peptide segments from soy protein for use in food safety. Abstr. Pap. Am. Chem. S. 2015, 250. [Google Scholar]
  43. Wessolowski, A.; Bienert, M.; Dathe, M. Antimicrobial activity of arginine- and tryptophan-rich hexapeptides: The effects of aromatic clusters, D-amino acid substitution and cyclization. J. Pept. Res. 2004, 64, 159–169. [Google Scholar] [CrossRef] [PubMed]
  44. Naidu, A.S.; Fowler, R.S.; Martinez, C.; Chen, J.; Tulpinski, J. Activated lactoferrin and fluconazole synergism against Candida albicans and Candida glabrata vaginal isolates. J. Reprod. Med. 2004, 49, 800–807. [Google Scholar] [PubMed]
  45. Nehete, J.Y.; Bhambar, R.S.; Narkhede, M.R.; Gawali, S.R. Natural proteins: Sources, isolation, characterization and applications. Pharmacogn. Rev. 2013, 7, 107–116. [Google Scholar] [CrossRef] [PubMed]
  46. Hsieh, C.-C.; Hernandez-Ledesma, B.; Fernandez-Tome, S.; Weinborn, V.; Barile, D.; de Moura Bell, J.M.L.N. Milk proteins, peptides, and oligosaccharides: Effects against the 21st century disorders. Biomed. Res. Int. 2015, 2015, 146840. [Google Scholar] [CrossRef] [PubMed]
  47. Bechara, C.; Sagan, S. Cell-penetrating peptides: 20 years later, where do we stand? FEBS Lett. 2013, 587, 1693–1702. [Google Scholar] [CrossRef] [PubMed]
  48. Nagarajan, K.; Marimuthu, S.K.; Palanisamy, S.; Subbiah, L. Peptide Therapeutics Versus Superbugs: Highlight on Current Research and Advancements. Int. J. Pept. Res. Ther. 2018, 24(1), 19–33. [Google Scholar] [CrossRef]
  49. Madani, F.; Lindberg, S.; Langel, U.; Futaki, S.; Graslund, A. Mechanisms of cellular uptake of cell-penetrating peptides. J. Biophys. 2011, 2011, 414729. [Google Scholar] [CrossRef] [PubMed]
  50. Trabulo, S.; Cardoso, A.L.; Mano, M.; De Lima, M.C.P. Cell-Penetrating Peptides-Mechanisms of Cellular Uptake and Generation of Delivery Systems. Pharmaceuticals 2010, 3, 961–993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Anderson, G.H.; Moore, S.E. Dietary proteins in the regulation of food intake and body weight in humans. J. Nutr. 2004, 134, 974–979. [Google Scholar] [CrossRef] [PubMed]
  52. Kittis, D.; Weiler, K. Bioactive Proteins and Peptides from Food Sources. Applications of Bioprocesses used in Isolation and Recovery. Curr. Pharm. Des. 2003, 9, 1309–1323. [Google Scholar] [CrossRef]
  53. Lamberti, C.; Purrotti, M.; Mazzoli, R.; Fattori, P.; Barello, C.; Daniel Coïsson, J.; Giunta, C.; Pessione, E. ADI pathway and histidine decarboxylation are reciprocally regulated in Lactobacillus hilgardii ISE 5211: Proteomic evidence. Amino Acids 2011, 41, 517–527. [Google Scholar] [CrossRef] [PubMed]
  54. FitzGerald, R.J.; Murray, B.A.; Walsh, D.J. Hypotensive peptides from milk proteins. J. Nutr. 2004, 134, 980–988. [Google Scholar] [CrossRef] [PubMed]
  55. Haque, E.; Chand, R.; Kapila, S. Biofunctional Properties of Bioactive Peptides of Milk Origin. Food Rev. Int. 2009, 25, 28–43. [Google Scholar] [CrossRef]
  56. Udenigwe, C.C.; Aluko, R.E. Food protein-derived bioactive peptides: Production, processing, and potential health benefits. J. Food Sci. 2012, 77, 11–24. [Google Scholar] [CrossRef] [PubMed]
  57. Nongonierma, A.B.; FitzGerald, R.J. Biofunctional properties of caseinophosphopeptides in the oral cavity. Caries Res. 2012, 46, 234–267. [Google Scholar] [CrossRef] [PubMed]
  58. Power, O.; Jakeman, P.; FitzGerald, R.J. Antioxidative peptides: Enzymatic production, in vitro and in vivo antioxidant activity and potential applications of milk-derived antioxidative peptides. Amino Acids 2013, 44, 797–820. [Google Scholar] [CrossRef] [PubMed]
  59. Sharma, S.; Singh, R.; Rana, S. Bioactive peptides: A review. Int. J. Bioautom. 2011, 15, 223–250. [Google Scholar]
  60. Beltrán-Barrientos, L.M.; Hernández-Mendoza, A.; Torres-Llanez, M.J.; González-Córdova, A.F.; Vallejo-Córdoba, B. Invited review: Fermented milk as antihypertensive functional food. J. Dairy. Sci. 2016, 99, 4099–4110. [Google Scholar] [CrossRef] [PubMed]
  61. Liu, Y.; Eichler, J.; Pischetsrieder, M. Virtual screening of a milk peptide database for the identification of food-derived antimicrobial peptides. Mol. Nutr. Food Res. 2015, 59, 2243–2254. [Google Scholar] [CrossRef] [PubMed]
  62. McCann, K.B.; Shiell, B.J.; Michalski, W.P.; Lee, A.; Wan, J.; Roginski, H. Isolation and characterisation of a novel antibacterial peptide from bovine αs1-casein. Int. Dairy J. 2006, 16, 316–323. [Google Scholar] [CrossRef]
  63. Lupetti, A.; Paulusma-Annema, A.; Welling, M.M.; Dogterom-Ballering, H.; Brouwer, C.P.J.M.; Senesi, S.; van Dissel, J.T.; Nibbering, P.H. Synergistic activity of the N-terminal peptide of human lactoferrin and fluconazole against Candida species. Antimicrob. Agents. Ch. 2003, 47(1), 262–267. [Google Scholar] [CrossRef]
  64. Hill, R.D.; Lahov, E.; Givol, D. A rennin-sensitive bond in alpha-s1 b-casein. J. Dairy Res. 1974, 41, 147–153. [Google Scholar] [CrossRef] [PubMed]
  65. Bellamy, W.; Takase, M.; Yamauchi, K.; Wakabayashi, H.; Kawase, K.; Tomita, M. Identification of the bactericidal domain of lactoferrin. Biochim. Biophys. Acta 1992, 1121, 130–136. [Google Scholar] [CrossRef]
  66. Recio, I.; Visser, S. Identification of two distinct antibacterial domains within the sequence of bovine αS2-casein. Biochim. Biophys. Acta 1999, 1428, 314–326. [Google Scholar] [CrossRef]
  67. Wakabayashi, H.; Hiratani, T.; Uchida, K.; Yamaguchi, H. Antifungal spectrum and fungicidal mechanism of an N-terminal peptide of bovine lactoferrin. J. Infect. Chemother. 1996, 1, 185–189. [Google Scholar] [CrossRef] [PubMed]
  68. Van der Kraan, M.I.A.; Groenink, J.; Nazmi, K.; Veerman, E.C.I.; Bolscher, J.G.M.; Nieuw Amerongen, A.V. Lactoferrampin: A novel antimicrobial peptide in the N1-domain of bovine lactoferrin. Peptides 2004, 25, 177–183. [Google Scholar] [CrossRef] [PubMed]
  69. Malkoski, M.; Dashper, S.G.; O’Brien-Simpson, N.M.; Talbo, G.H.; Macris, M.; Cross, K.J. Kappacin a novel antimicrobial peptide from bovine milk. Antimicrob. Agents Chemother. 2001, 45, 2309–2315. [Google Scholar] [CrossRef] [PubMed]
  70. Lopez-Exposito, I.; Quiros, A.; Amigo, L.; Recio, I. Casein hydrolysates as a source of antimicrobial, antioxidative and antihypertensive peptides. Le Lait 2007, 87, 241–249. [Google Scholar] [CrossRef]
  71. Wakabayashi, H.; Takase, M.; Tomita, M. Lactoferricin derived from milk protein lactoferrin. Curr. Pharm. Des. 2003, 9, 1277–1287. [Google Scholar] [CrossRef] [PubMed]
  72. Haney, E.F.; Nazmi, K.; Lau, F.; Bolscher, J.G.M.; Vogel, H.J. Novel lactoferrampin antimicrobial peptides derived from human lactoferrin. Biochimie 2009, 91, 141–154. [Google Scholar] [CrossRef] [PubMed]
  73. Zucht, H.D.; Raida, M.; Adermann, K.; Magert, H.J.; Forssman, W.G. Casocidin-I: A casein αS2-derived peptide exhibits antibacterial activity. FEBS Lett. 1995, 372, 185–188. [Google Scholar] [CrossRef]
  74. Reiter, B.; Oram, J.D. Bacterial inhibitors in milk and other biological fluids. Nature 1967, 216, 328–330. [Google Scholar] [CrossRef]
  75. Arnold, R.R.; Brewer, M.; Gauthier, J.J. Bactericidal activity of human lactoferrin: Sensitivity of a variety of microorganisms. Infect. Immun. 1980, 28, 893–898. [Google Scholar] [PubMed]
  76. Hettiarachchy, S.N.; Sato, K.; Marshall, M.R.; Kannan, A. Bioactive Food Proteins and Peptides: Applications in Human Health; CRC Press: Boca Raton, FL, USA, 2016. [Google Scholar]
  77. Gigli, I. Milk Proteins from Structure to Biological Properties and Health Aspects; INTech Open Publications: Rijeka, Croatia, 2016. [Google Scholar]
  78. Holt, C. The milk salts and their interaction with casein. In Advanced Dairy Chemistry; Fox, P.F., Ed.; Chapman & Hall: London, UK, 1997; pp. 233–256. [Google Scholar]
  79. Jabbari, A.; Suárez-Fariñas, M.; Dewell, S.; Krueger, J.G. Transcriptional profiling of psoriasis using RNA-seq reveals previously unidentified differentially expressed genes. J. Investig. Dermatol. 2012, 132, 246–249. [Google Scholar] [CrossRef] [PubMed]
  80. Tomita, M.; Takase, M.; Bellami, W.; Shimamura, S. A review: The active peptide of lactoferrin. Acta Paediatr. Jpn. 1994, 36, 585–591. [Google Scholar] [CrossRef] [PubMed]
  81. Hayes, M.; Ross, R.P.; Fitzgerald, G.F.; Hill, C.; Stanton, C. Casein-derived antimicrobial peptides generated by Lactobacillus acidophilus DPC6026. Appl. Environ. Microbiol. 2006, 72, 2260–2264. [Google Scholar] [CrossRef] [PubMed]
  82. Hayes, M.; Barrett, E.; Ross, R.P.; Fitzgerald, G.F.; Hill, C.; Stanton, C. Evaluation of an antimicrobial ingredient prepared from a Lactobacillus acidophilus caseinfermentate against Enterobacter sakazakii. J. Food Prot. 2009, 72, 340–346. [Google Scholar] [CrossRef] [PubMed]
  83. McDonnell, M.J.; Rivas, L.; Burgess, C.M.; Fanning, S.; Duffy, G. Inhibition of verocytotoxigenic Escherichia coli by antimicrobial peptides caseicin A and B and the factors affecting their antimicrobial activities. Int. J. Food Microbiol. 2012, 153, 260–268. [Google Scholar] [CrossRef] [PubMed]
  84. Norberg, S.; O’Connor, P.M.; Stanton, C.; Ross, R.P.; Hill, C.; Fitzgerald, G.F. Altering the composition of caseicins A and B as a means of determining the contribution of specific residues to antimicrobial activity. Appl. Environ. Microbiol. 2011, 77, 2496–2501. [Google Scholar] [CrossRef] [PubMed]
  85. Fadaei, V. Milk Proteins-derived antibacterial peptides as novel functional food ingredients. Ann. Biol. Res. 2012, 3, 2520–2526. [Google Scholar]
  86. Tidona, F.; Criscione, A.; Guastella, A.N.; Zuccaro, A.; Bordonaro, S.; Marletta, D. Bioactive peptides in dairy products. Ital. J. Anim. Sci. 2009, 8, 315–340. [Google Scholar] [CrossRef]
  87. Priyadarshini, S.; Kansal, V.K. Purification, characterization, antibacterial activity and N-terminal sequencing of buffalo-milk lysozyme. J Dairy Res. 2002, 69, 419–431. [Google Scholar] [CrossRef] [PubMed]
  88. Hunter, H.N.; Jing, W.; Schibli, D.J.; Trinh, T.; Park, I.Y.; Kim, S.C.; Vogel, H.J. The interactions of antimicrobial peptides derived from lysozyme with model membrane systems. Biochim. Biophys. Acta 2005, 1668, 175–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Lopez-Exposito, I.; Minervini, F.; Amigo, L.; Recio, I. Identification of antibacterial peptides from bovine kappa-casein. J. Food Prot. 2006, 69, 2992–2997. [Google Scholar] [CrossRef] [PubMed]
  90. Korhonen, H.; Pihlanto, A. Food-derived bioactive peptides—Opportunities for designing future foods. Curr. Pharm. Des. 2003, 9, 1297–1308. [Google Scholar] [CrossRef] [PubMed]
  91. Zambrowicz, A.; Timmer, M.; Polanowski, A.; Lubec, G.; Trziszka, T. Manufacturing of peptides exhibiting biological activity. Amino Acids 2013, 44, 315–320. [Google Scholar] [CrossRef] [PubMed]
  92. Kunji, E.R.; Mierau, I.; Hagting, A.; Poolman, B.; Konings, W.N. The proteotytic systems of lactic acid bacteria. Antonie Van Leeuwenhoek 1996, 70, 187–221. [Google Scholar] [CrossRef] [PubMed]
  93. Benkerroum, N. Antimicrobial peptides generated from milk proteins: A survey and prospects for application in the food industry—A review. Int. J. Dairy Technol. 2010, 63, 320–338. [Google Scholar] [CrossRef]
  94. Martinenko, N.I.; Yagodinskaya, S.G.; Adhundov, A.A.; Charyev, K.C.; Khumedov, O. Contet of trace element, copper, manganese molybdenum in culture of chal and camel milk and their clinical significance. Dairy Sci. 1977, 40, 824. [Google Scholar]
  95. Muhialdin, B.J.; Hassan, Z.; Abu Bakar, F.; Saari, N. Identification of Antifungal Peptides Produced by Lactobacillus plantarum IS10 Grown in the MRS broth. Food Control 2016, 59, 27–30. [Google Scholar] [CrossRef]
  96. Anastasiadou, S.; Papagianni, M.; Filiousis, G.; Ambrosiadis, I.; Koidis, P. Pediocin SA-1, an antimicrobial peptide from Pediococcus acidilactici NRRL B5627: Production conditions, purification and characterization. Bioresour. Technol. 2008, 99, 5384–5390. [Google Scholar] [CrossRef] [PubMed]
  97. Riley, M.A.; Wertz, J.E. Bacteriocins: Evolution, ecology, and application. Annu. Rev. Microbiol. 2002, 56, 117–137. [Google Scholar] [CrossRef] [PubMed]
  98. Milioni, C.; Martínez, B.; Degl’Innocenti, S.; Turchi, B.; Fratini, F.; Cerri, D.; Fischetti, R. A novel bacteriocin produced by Lactobacillus plantarum LpU4 as a valuable candidate for biopreservation in artisanal raw milk cheese. Dairy Sci. Technol. 2015. [Google Scholar] [CrossRef]
  99. Sonsa-Ard, N.; Rodtong, S.; Chikindas, M.L.; Yongsawatdigul, J. Characterization of bacteriocin produced by Enterococcus faecium CN-25 isolated from traditionally Thai fermented fish roe. Food Control 2015, 54, 308–316. [Google Scholar] [CrossRef]
  100. Silva, C.C.G.; Silva, S.P.M.; Ribeiro, S.C. Application of Bacteriocins and Protective Cultures in Dairy Food Preservation. Front. Microbiol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  101. Khaksar, R.; Carlson, T.; Schaffner, D.W. Unmasking seafood mislabeling in US markets: DNA barcoding as a unique technology for food authentication and quality control. Food Control 2015, 56, 71–76. [Google Scholar] [CrossRef]
  102. Théolier, J.; Fliss, I.; Jean, J.; Hammami, R. Antimicrobial peptides of dairy proteins: From fundamental to applications. Food Rev. Int. 2014, 30, 134–154. [Google Scholar] [CrossRef]
  103. Cheigh, C.; Pyun, Y. Nisin biosynthesis and its properties. Biotechnol. Lett. 2005, 27, 1641–1648. [Google Scholar] [CrossRef] [PubMed]
  104. De Arauz, L.J.; Jozala, A.F.; Mazzola, P.G.; Vessoni Penna, T.C. Nisin biotechnological production and application: A review. Trends Food Sci. Technol. 2009, 20, 146–154. [Google Scholar] [CrossRef]
  105. Delves-Broughton, J.; Blackburn, P.; Evans, R.J.; Hugenholtz, J. Applications of the bacteriocin, nisin. Antonie Van Leeuwenhoek 1996, 69, 193–202. [Google Scholar] [CrossRef] [PubMed]
  106. Sobrino-López, A.; Martín-Belloso, O. Use of nisin and other bacteriocins for preservation of dairy products. Int. Dairy J. 2008, 18, 329–343. [Google Scholar] [CrossRef]
  107. Kjos, M.; Nes, I.F.; Diep, D.B. Class II one-peptide bacteriocins target a phylogenetically defined subgroup of mannose phosphotransferase systems on sensitive cells. Microbiology 2009, 155, 2949–2961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Ahmed Elsayed, E.; Abdel Fattah Farid, M.; Ali El Enshasy, H. Improvement in natamycin production by Streptomyces natalensis with the addition of short-chain carboxylic acids. Process Biochem. 2013, 48, 1831–1838. [Google Scholar] [CrossRef]
  109. Scientific Opinion on the use of natamycin (E 235) as a food additive; Natamycin, Pimaricin, Antibiotics, E 235, CAS 7681-93-8, Antibiotic Resistance; EFSA: Parma, Italy, 2009.
  110. Lahov, E.; Regelson, W. Antibacterial and immunostimulating casein-derived substances from milk: Casecidin, isracidin peptides. Food Chem. Toxicol. 1996, 34, 131–145. [Google Scholar] [CrossRef]
  111. Chantaysakorn, P.; Richter, R.L. Antimicrobial properties of pepsin-digested lactoferrin added to carrot juice and filtrate of carrot juice. J. Food Prot. 2000, 63, 376–380. [Google Scholar] [CrossRef] [PubMed]
  112. Wiesner, J.; Vilcinskas, A. Antimicrobial Peptides: The Ancient Arm of the Human Immune System. Virulence 2010, 5, 440–464. [Google Scholar] [CrossRef] [PubMed]
  113. Ibeagha-Awemu, E.M.; Zhao, X. Epigenetic marks: Regulators of livestock phenotypes and conceivable sources of missing variation in livestock improvement programs. Front. Genet. 2015, 28, 302. [Google Scholar] [CrossRef] [PubMed]
  114. Addis, D.R.; Musicaro, R.; Pan, L.; Schacter, D.L. Episodic simulation of past and future events in older adults: Evidence from an experimental recombination task. Psychol. Aging 2010, 25, 369–376. [Google Scholar] [CrossRef] [PubMed]
  115. Szwajkowska, M.; Wolanciuk, A.; Wolanciuk, J. Bovine milk protein as the source of bioactive peptides influencing the consumers’ immune system. Anim. Sci. Pap. Rep. 2011, 29, 269–280. [Google Scholar]
  116. Waqhu, F.H.; Gopi, L.; Ramteke, P.; Nizami, B.; Idicula-Thomas, S. CAMP: Collection of sequences and structures of antimicrobial peptide. Nucleic Acids Res. 2014, 42, 1154–1158. [Google Scholar] [CrossRef] [PubMed]
  117. Geetha, R.; Sathian, C.T.; Prasad, V.; Gleeja, V.L. Efficacy of purified antimicrobial peptides from lactic acid bacteria against bovine mastitis pathogens. Asian J. Dairy Food Res. 2015, 34, 259–264. [Google Scholar] [CrossRef]
  118. Pingitore, E.; Salvucci, V.E.; Sesma, F.; Macías, N.M.E. Different strategies for purification of antimicrobial peptides from Lactic Acid Bacteria (LAB). Trends Appl. Microbiol. 2007, 21, 557–568. [Google Scholar]
  119. Kozak, W.; Trzpil, M.R.; Dobrzanski, W.T. Preliminary observations on the influence of proflavin, ethidium bromide, and elevated temperature on the production of the antibiotic nisin by Streptococcus lactis strains. Bull. Acad. Pol. Sci. 1973, 21, 811–817. [Google Scholar]
  120. Chassy, B.M. A gentle method for the lysis of oral Streptococci. Biochem. Biophys. Res. Commun. 1976, 68, 603–608. [Google Scholar] [CrossRef]
  121. Daveyand, G.P.; Richardson, B.C. Purification and Some Properties of Diplococcin from Streptococcus cremoris 346. Appl. Environ. Microbiol. 1981, 41, 84–89. [Google Scholar]
  122. Hastings, J.W.; Sailer, M.; Johnson, K.; Roy, K.L.; Vederas, J.C.; Stiles, M.E. Characterization of leucocin A-UAL187 and cloning of the bacteriocin gene from Leuconostoc gelidum. J. Bacteriol. 1991, 173, 7491–7500. [Google Scholar] [CrossRef] [PubMed]
  123. Rana, S.; Bajaj, R.; Mann, B. Characterization of Antimicrobial and Antioxidative Peptides Synthesized by L. rhamnosus C6 Fermentation of Milk. Int. J. Pept. Res. Ther. 2018, 24, 309–321. [Google Scholar] [CrossRef]
  124. Liepke, C.; Zucht, H.D.; Forssmann, W.G.; Standker, L. Purification of novel peptide antibiotics from human milk. J. Chromatogr. B 2001, 752, 369–377. [Google Scholar] [CrossRef]
  125. Kussmann, M.; Nordhoff, E.; Rahbek-Nielsen, H.; Haebel, S.; Rossel-Larsen, M.; Jakobsen, L.; Gobom, J.; Mirgorodskaya, E.; Kroll-Kristensen, A.; Palm, L.; et al. Matrix-assisted Laser Desorption/Ionization Mass Spectrometry Sample Preparation Techniques Designed for Various Peptide and Protein Analytes. J. Mass Spect. 1997, 32, 593–601. [Google Scholar] [CrossRef]
  126. Gobbetti, M.; Stepaniak, L.; De Angelis, M.; Corsetti, A.; Di Cagno, R. Latent bioactive peptides in milk proteins: Proteolytic activation and significance in dairy processing. Crit. Rev. Food Sci. Nutr. 2002, 42, 223–239. [Google Scholar] [CrossRef] [PubMed]
  127. Schrimpf, A.; Hempel, F.; Li, A.; Linne, U.; Maier, U.G.; Reetz, M.T.; Geyer, A. Hinge-Type Dimerization of Proteins by a Tetracysteine Peptide of High Pairing Specificity. Biochemistry 2018, 57, 3658–3664. [Google Scholar] [CrossRef] [PubMed]
  128. De Brito, R.C.F.; Cardoso, J.M.D.O.; Reis, L.E.S.; Vieira, J.F.; Mathias, F.A.S.; Roatt, B.M.; Aguiar-Soares, R.D.D.O.; Ruiz, J.C.; Resende, D.D.M.; Reis, A.B. Peptide Vaccines for Leishmaniasis. Front. Immunol. 2018, 9, 1043. [Google Scholar] [CrossRef] [PubMed]
  129. Soundrarajan, N.; Cho, H.-S.; Ahn, B.; Choi, M.; Thong, L.M.; Choi, H.; Cha, S.-Y.; Kim, J.-H.; Park, C.-K.; Seo, K.; et al. Green fluorescent protein as a scaffold for high efficiency production of functional bacteriotoxic proteins in Escherichia coli. Sci. Rep. 2016, 6, 20661. [Google Scholar] [CrossRef] [PubMed]
  130. Chahardoli, M.; Fazeli, A.; Niazi, A.; Ghabooli, M. Recombinant expression of LFchimera antimicrobial peptide in a plant-based expression system and its antimicrobial activity against clinical and phytopathogenic bacteria. Biotechnol. Biotechnol. Equip. 2018, 32, 714–723. [Google Scholar] [CrossRef] [Green Version]
  131. Boga, S.; Bouzada, D.; Pena, D.G.; Lopez, M.V.; Vazquez, M.E. Sequence-Specific DNA Recognition with Designed Peptides. Eur. J. Org. Chem. 2018, 249–261. [Google Scholar] [CrossRef]
  132. Lepage, P.; Heckel, C.; Humbert, S.; Stahl, S.; Rautmann, G. Recombinant Technology as an Alternative to Chemical Peptide-Synthesis—Expression and Characterization of HIV-1 Rev Recombinant Peptides. Anal. Biochem. 1993, 213, 40–48. [Google Scholar] [CrossRef] [PubMed]
  133. Espita, P.J.P.; De Fátima, N.F.S.; Coimbra, J.S.R.; Andrade, N.J.; Cruz, R.S.; Medeiros, E.A.A. Zinc Oxide Nanoparticles: Synthesis, Antimicrobial Activity and Food Packaging Applications. Food Bioprocess Technol. 2009, 5, 1447–1464. [Google Scholar] [CrossRef]
  134. Feng, J. Tyrosine phosphorylation by Src within the cavity of the adenine nucleotide translocase 1 regulates ADP/ATP exchange in mitochondria. Am. J. Physiol. Cell Physiol. 2010, 298, 740–748. [Google Scholar] [CrossRef] [PubMed]
  135. Tian, L.; Stefanidakis, M.; Ning, L.; Van Lint, P.; Nyman-Huttunen, H.; Libert, C.; Itohara, S.; Mishina, M.; Rauvala, H.; Gahmberg, C.G. Activation of NMDA receptors promotes dendritic spine development through MMP-mediated ICAM-5 cleavage. J. Cell Biol. 2007, 178, 687–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Feng, X. Identification of a novel nuclear-localized adenylate kinase 6 from Arabidopsis thaliana as an essential stem growth factor. Plant Physiol. Biochem. 2012, 61, 180–186. [Google Scholar] [CrossRef] [PubMed]
  137. Prak, K.; Utsumi, S. Production of a bioactive peptide (IIAEK) in Escherichia coli using soybean proglycinin A1ab1b as a carrier. J. Agric. Food Chem. 2009, 13, 3792–3799. [Google Scholar] [CrossRef] [PubMed]
  138. Shityakov, S.; Salmas, R.E.; Durdagi, S.; Salvador, E.; Papai, K.; Yanez-Gascon, M.J.; Perez-Sanchez, H.; Puskas, I.; Roewer, N.; Forster, C.; et al. Characterization, in vivo Evaluation, and Molecular Modeling of Different Propofol-Cyclodextrin Complexes to Assess Their Drug Delivery Potential at the Blood-Brain Barrier Level. J. Chem. Inf. Model. 2016, 56, 1914–1922. [Google Scholar] [CrossRef] [PubMed]
  139. Wojcik, C.; Sawicki, W.; Marianowski, P.; Benchaib, M.; Czyba, J.C.; Guerin, J.F. Cyclodextrin enhances spermicidal effects of magainin-2-amide. Contraception 2000, 62, 99–103. [Google Scholar] [CrossRef]
  140. Shityakov, S.; Forster, C. Pharmacokinetic delivery and metabolizing rate of nicardipine incorporated in hydrophilic and hydrophobic cyclodextrins using two-compartment mathematical model. Sci. World J. 2013, 2013, 131358. [Google Scholar] [CrossRef] [PubMed]
  141. Shityakov, S.; Broscheit, J.; Forster, C. Alpha-Cyclodextrin dimer complexes of dopamine and levodopa derivatives to assess drug delivery to the central nervous system: ADME and molecular docking studies. Int. J. Nanomed. 2012, 7, 3211–3219. [Google Scholar] [CrossRef] [PubMed]
  142. Sobkoa, A.A.; Kotovaa, E.A.; Antonenkoa, Y.N.; Zakharov, S.D.; Cramerb, W.A. Effect of lipids with different spontaneous curvature on the channel activity of colicin E1: Evidence in favor of a toroidal pore. FEBS Lett. 2004, 576, 205–210. [Google Scholar] [CrossRef] [PubMed]
  143. Niu, S.L.; Mitchell, D.C.; Litman, B.J. Manipulation of cholesterol levels in rod disk membranes by methyl-β-cyclodextrin. Effects on receptor activation. J. Biol. Chem. 2002, 277, 20139–20145. [Google Scholar] [CrossRef] [PubMed]
  144. Sauer, R.-S.; Rittner, H.L.; Roewer, N.; Sohajda, T.; Shityakov, S.; Brack, A.; Broscheit, J.-A. A Novel Approach for the Control of Inflammatory Pain: Prostaglandin E2 Complexation by Randomly Methylated beta-Cyclodextrins. Anesth. Analg. 2017, 124, 675–685. [Google Scholar] [CrossRef] [PubMed]
  145. Martínez-Moreno, R. New insights into the advantages of ammonium as a winemaking nutrient. Int. J. Food Microbiol. 2014, 177, 128–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Deng, P.; Zhongtang, Y. Intestinal microbiome of poultry and its interaction with host and diet. Gut Microbes 2014, 5, 108–119. [Google Scholar]
Figure 1. Hypothetical ABP/β-CD (anti-bacterial peptide/β-cyclodextrin) complexation/formulation process via the molecular docking of peptide lipophilic side chains into the hydrophobic β-CD binding cavity.
Figure 1. Hypothetical ABP/β-CD (anti-bacterial peptide/β-cyclodextrin) complexation/formulation process via the molecular docking of peptide lipophilic side chains into the hydrophobic β-CD binding cavity.
Biomolecules 08 00110 g001
Figure 2. Hypothetical model indicating that dietary monosaccharides (DMs) might be taken up by the intestinal cell and used for the synthesis of cell surface glycoconjugates [145] with modifications). These glycoconjugates and milk oligosaccharides (MOs) might inhibit the adhesion to the cell of bacterial pathogens.
Figure 2. Hypothetical model indicating that dietary monosaccharides (DMs) might be taken up by the intestinal cell and used for the synthesis of cell surface glycoconjugates [145] with modifications). These glycoconjugates and milk oligosaccharides (MOs) might inhibit the adhesion to the cell of bacterial pathogens.
Biomolecules 08 00110 g002
Table 1. Functional activity of milk-derived antibacterial peptides (ABPs) (adopted from [36,45]).
Table 1. Functional activity of milk-derived antibacterial peptides (ABPs) (adopted from [36,45]).
ABPsCirculatory SystemNervous SystemImmune SystemGastrointestinal TractFunctional Peptide
Antihypertensive peptidesOpioid peptidesImmunomodulation peptidesRegulatory and enzyme inhibitorsSensory peptides
Antithrombotic peptidesAntibacterial peptidesCeliac toxicityAntioxidative peptides
Microelement-binding peptidesSurface active peptides
Table 2. Minimum inhibitory concentration (MIC) for different fragments of milk-derived ABPs (adopted from [60,61,62,63]).
Table 2. Minimum inhibitory concentration (MIC) for different fragments of milk-derived ABPs (adopted from [60,61,62,63]).
ABP MIC Pathogen
αs2-casein f(151–181)15.6 μg/mLBacillus subtilis ATCC6051,
16.2 μM (62.5 μg/mL)Escherichia coli NEB5α and E. coli, ATCC25922
αs2-casein f(182–207)2.7 μM (8.6 μg/mL)B. subtilis ATCC6051,
21.4 μM (68.8 μg/mL)E. coli NEB5α,
Lactoferrin 125 mg/mLE. coli,
250 mg/mLSalmonella typhimurium,
125 mg/mLSalmonella enteritidis,
500 mg/mLCitrobacter freundii,
2.5 mg/mLCandida albicans
Table 3. Summary of milk-derived ABPs and their antibacterial effect (adopted from [60]).
Table 3. Summary of milk-derived ABPs and their antibacterial effect (adopted from [60]).
ABPProductionInhibitionReferences
Isracidin αs1 f(1–23)Chymosin digestion Several microorganisms
in vivo and in vitro
[64]
Lactoferrin B f(18–36)
and f(17–41/42)
Enzymatic digestion (pepsin and chymosin)Some Gram (+) and Gram (−) bacteria [65,66]
Lactoferricin f(17–41) Enzymatic digestion (pepsin and chymosin)Some Gram (+) and Gram (−) bacteria, viruses, fungi, and parasites[65,67]
Lf f(268–284) Enzymatic digestion (pepsin and chymosin)B. subtilis, E. coli, P. aeruginosa[68]
αs2 casein f(183–207)Digestion with pepsinSome Gram (+) and Gram (−) bacteria[66]
κ-casein f(106–169) (kappacin)Digestion with chymosin S. mutans, E. coli[69]
κ-casein f(18–24) and f(30–32) and f(139–146)Digestion with pepsin Some Gram (+) and Gram (−) bacteria[70]
Lf f(1–48) and f(1–47) Digestion with pepsin M. flavus[70]
α–La f(1–5) and f(17–31)
and f(61–68)
Digestion with chymotrypsin Some Gram (+) Gram (−) bacteria[71]
B–Lg f(15–20), f(25–40),
f(78–83) and f(92–100)
Digestion with trypsinSome Gram (+) and Gram (−) bacteria[71]
Table 4. Industrial production of ABPs [91].
Table 4. Industrial production of ABPs [91].
Production MethodProductionScale
FermentationNot preciseLaboratory and industrial
Protein hydrolysisNot preciseLimited to laboratory
Recombinant DNA Large ABPs (>150 amino acids)Laboratory and industrial
Organic synthesisMedium-size ABPsLaboratory and industrial
Table 5. Synthesis of milk-derived ABPs by recombinant DNA technology.
Table 5. Synthesis of milk-derived ABPs by recombinant DNA technology.
Derivative Antibacterial PeptidesParental CompoundExpression SystemInhibited Growth Reference
Lactoferricin B-W10 (LfcinB-W10),Lactoferricin Lf-(f17–41)E. coli BL21 (DE3).S. aureus ATCC25923[134]
Lfcin B15-W4,10Lactoferricin Lf-(f17–31)E. coli BL21 (DE3).S. aureus ATTC25923[135]
LFT33Bovine lactoferricin and thanatin (an inducible insect antibacterial peptide)E. coli BL21Significant antibacterial activity compared to parental compound[136]
LactophoricinResidues 113–135 of proteose-peptone (component 3)E. coli C41 (DE3)Not mentioned [137]

Share and Cite

MDPI and ACS Style

Khan, M.U.; Pirzadeh, M.; Förster, C.Y.; Shityakov, S.; Shariati, M.A. Role of Milk-Derived Antibacterial Peptides in Modern Food Biotechnology: Their Synthesis, Applications and Future Perspectives. Biomolecules 2018, 8, 110. https://0-doi-org.brum.beds.ac.uk/10.3390/biom8040110

AMA Style

Khan MU, Pirzadeh M, Förster CY, Shityakov S, Shariati MA. Role of Milk-Derived Antibacterial Peptides in Modern Food Biotechnology: Their Synthesis, Applications and Future Perspectives. Biomolecules. 2018; 8(4):110. https://0-doi-org.brum.beds.ac.uk/10.3390/biom8040110

Chicago/Turabian Style

Khan, Muhammad Usman, Maryam Pirzadeh, Carola Yvette Förster, Sergey Shityakov, and Mohammad Ali Shariati. 2018. "Role of Milk-Derived Antibacterial Peptides in Modern Food Biotechnology: Their Synthesis, Applications and Future Perspectives" Biomolecules 8, no. 4: 110. https://0-doi-org.brum.beds.ac.uk/10.3390/biom8040110

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop