Next Article in Journal
Mediterranean Alcohol-Drinking Pattern and the Incidence of Cardiovascular Disease and Cardiovascular Mortality: The SUN Project
Next Article in Special Issue
Eggs and Health Special Issue
Previous Article in Journal
Oral Fat Sensing and CD36 Gene Polymorphism in Algerian Lean and Obese Teenagers
Previous Article in Special Issue
Immune-Relevant and Antioxidant Activities of Vitellogenin and Yolk Proteins in Fish
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Nutraceutical Properties of Ovotransferrin and Its Potential Utilization as a Functional Food

by
Francesco Giansanti
1,2,*,
Loris Leboffe
2,3,
Francesco Angelucci
1 and
Giovanni Antonini
2,3
1
Department of Health, Life and Environmental Sciences, University of L’Aquila, L’Aquila I-67100, Italy
2
Interuniversity Consortium INBB Biostructures and Biosystems National Institute, Rome I-00136, Italy
3
Department of Sciences, Roma Tre University, Rome I-00146, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2015, 7(11), 9105-9115; https://doi.org/10.3390/nu7115453
Submission received: 29 September 2015 / Revised: 21 October 2015 / Accepted: 23 October 2015 / Published: 4 November 2015
(This article belongs to the Special Issue Egg Consumption and Human Health)

Abstract

:
Ovotransferrin or conalbumin belong to the transferrin protein family and is endowed with both iron-transfer and protective activities. In addition to its well-known antibacterial properties, ovotransferrin displays other protective roles similar to those already ascertained for the homologous mammalian lactoferrin. These additional functions, in many cases not directly related to iron binding, are also displayed by the peptides derived from partial hydrolysis of ovotransferrin, suggesting a direct relationship between egg consumption and human health.

Graphical Abstract

1. Introduction

Ovotransferrin (Otrf) or conalbumin belongs to the family of transferrin iron-binding glycoproteins. In mammals, two different soluble iron-binding glycoproteins are present: (i) serum transferrin, involved in iron transport and delivery to cells and (ii) lactoferrin, involved in the so-called natural immunity. Differently, Otrf is the only soluble glycoprotein of the transferrin protein family present in avian. Otrf is present both in avian plasma and egg white and possesses both iron-transfer and protective properties [1]. Otrf represents about 12%–13% of total egg white proteins and contributes to promoting the growth and development of the chicken embryo mainly preventing the growth of micro-organisms together with other proteins such as lysozyme [2], cystatin [3,4], ovomacroglobulin [5] and avidin [6]. Galliformes (chicken, Gallus gallus and turkey, Meleagris gallopavo) appear to possess albumens with greater antimicrobial activity than those of the anseriformes (duck, Anas platyrhynchos), possibly due to higher concentrations of ovotransferrin and of the broad active c-type lysozyme [7]. However, recent evidence indicates that Otrf is endowed not only with the antibacterial activity related to iron withholding, but also with other roles related to the protection of the growing embryo, including: regulation of iron absorption; immune response; and anti-bacterial, anti-viral and anti-inflammatory properties. Some of these properties are shared by both the human protein homologues and peptides deriving from its partial enzymatic hydrolysis [8], being in this latter case also increased.
The state of the art hereby described suggests that Otrf and its peptides can be used as functional food ingredients and as important components for nutraceuticals, being characterized both by protective functions and by substantial nutritional benefits; for these reasons, the utilization of Otrf and its peptides in functional foods can present several additional advantages over other natural compounds.

2. Ovotransferrin Synthesis and Structure

Otrf is a monomeric glycoprotein containing 686 amino acids, with a molecular weight of 77.9 kDa and an isoelectric point of 6.0 [9,10].
The avian transferrin gene is transcripted in the liver and the oviduct. In the liver, the transferrin is secreted in the serum, where it is involved in iron transport and storage, while the oviduct transferrin (ovotransferrin) is secreted at high levels in the egg white. In particular, progesterone and oestrogen regulate the expression of Otrf in the oviduct: oestrogen can interact with chromatin through a nuclear receptor protein stimulating transcription and synthesis of the protein precursor [11]. Instead, the transcription of serum Otrf may depend on iron concentration [12]. Although the serum transferrin and Otrf have the same amino acidic sequence, they differ in the glycosylation sites. The glycan of Otrf is constituted by four residues of mannose and four residues of N-acetylglucosamine whereas serum transferrin is composed of two residues of mannose, two residues of galactose, three residues of N-acetylglucosamine, and one or two residues of sialic acid at its C-terminus [13,14,15].
Like the mammalian transferrins, the single chain of Otrf consists of two globular lobes (N- and C-lobes), interconnected by a α-helix of nine amino acidic residues (residues 333–341) that can be released by tryptic digestion. Each lobe contains an iron binding site and is divided in two domains, (domains N1 and N2 in the N-lobe and domains C1 and C2 in the C-lobe, respectively; Figure 1A) [16,17,18].
As shown in Figure 1, the domains are linked by anti-paralleled β-strands. The N- and C-lobes show about 38% sequence homology; indeed, it is has been hypothesized that all members of transferrin family resulted from gene fusion and duplication [19]. Fifteen disulfide bridges stabilize the structure. Six of them are conserved in both lobes, while three are present only in the C-lobe, conferring to the lobes a different metal affinity.
Figure 1. (A) Ribbon representation and the solvent-accessible surface (in transparency) of holo-Otrf (PDB ID:1OVT) [16]. N1, N2 and C1, C2 indicate the subdomains of each lobe. (B) The N-lobe iron binding site of hen’s ovotransferrin (PDB ID:1OVT). The amino acids involved in iron binding are shown in sticks. H-bonds are displayed by purple broken lines. Both in (A) and (B), the iron is indicated as a yellow sphere. (C) Schematic ribbon representation of the OTAP-92 peptide. The three disulfide linkages are represented in yellow, while the hydrophobic residues are highlighted in blue. The peptide is shown with the same conformation displayed in the intact protein. Molecular graphic images were produced using the UCSF chimera package [20].
Figure 1. (A) Ribbon representation and the solvent-accessible surface (in transparency) of holo-Otrf (PDB ID:1OVT) [16]. N1, N2 and C1, C2 indicate the subdomains of each lobe. (B) The N-lobe iron binding site of hen’s ovotransferrin (PDB ID:1OVT). The amino acids involved in iron binding are shown in sticks. H-bonds are displayed by purple broken lines. Both in (A) and (B), the iron is indicated as a yellow sphere. (C) Schematic ribbon representation of the OTAP-92 peptide. The three disulfide linkages are represented in yellow, while the hydrophobic residues are highlighted in blue. The peptide is shown with the same conformation displayed in the intact protein. Molecular graphic images were produced using the UCSF chimera package [20].
Nutrients 07 05453 g001
Each lobe has the capability to reversibly bind one Fe3+ ion along with one CO32− anion. Although each lobe displays a high sequence homology, they show different iron-binding properties. In particular, the (approximate) iron binding affinity is 1.5 × 1018 M−1 for the C-lobe and 1.5 × 1014 M−1 for the N-lobe. As mentioned before, this difference is due to the presence, in the C-lobe, of an extra interdomain disulfide bond, Cys478-Cys671, which confers less flexibility and possibly less affinity towards Fe3+ ion [21].
From a stereochemical point of view, the iron binding pocket is conserved among all members of transferrins [16,17,18,22,23,24]. As shown in Figure 1B, in the N-lobe there are two phenolate oxygens of two tyrosine residues (Tyr92 and Tyr191), a carboxylate oxygen of the aspartic acid (Asp60), and the imidazole nitrogen of the histidine (His250), together with two oxygens of the synergistically bound CO32− anion. In C-lobe, the corresponding amino acids are Asp 395, Tyr524, Tyr431 and His592, respectively. In this latter, Arg460 keeps in place the carbonate moiety while in the N-lobe Arg121 plays the same role. Upon iron binding, each lobe of Otrf undergoes large conformational change; the observed movement is necessary to bury the metal inside the polypeptide chain given that, in its absence, the iron-coordinating amino acidic residues are solvent exposed.

3. Antibacterial Activity of Ovotransferrin and Its Peptides

Among the several protective functions of Otrf, the most important one is likely to be the antibacterial activity, which is directly related to the Otrf’s ability to bind iron (Fe3+), making it unavailable for bacterial growth [25,26]. This bacteriostatic activity is reversed by adding iron ions to the medium and it is blocked by iron saturation [27]; moreover, it can be enhanced by (i) adding carbonate ion [28] which is one of the iron ligands in the Otrf metal binding site [29]; (ii) increasing the pH from 6 to 8 [27]; (iii) and immobilizing Otrf by covalent linkage to Sepharose 4B [30]. An increase of the bacteriostatic activity towards E. coli O157:H7 as iron chelator was demonstrated using a combination of ovotransferrin, NaHCO3, and EDTA [31]. The antibacterial activity was demonstrated also in an in vivo study using newborn guinea pigs orally infected with E. coli 0111 B4 [32]. On the contrary, citrate exerts an antagonistic effect in those bacteria that possess a receptor for iron-citrate complex [32]. The most sensitive species to the iron deprivation effect of Otrf are Pseudomonas spp., Escherichia coli, Streptococcus mutans, while the most resistant ones are Proteus spp., and Klebsiella spp. [32], according to the ability of these latter bacteria to produce molecules (i.e., siderophores) able to compete with Otrf for iron binding. However, it is worth noting that some bacterial species that are also human pathogens, e.g., Neisseria meningitidis, Neisseria gonorrhoeae and Moraxella catarrhalis, have developed a mechanism for acquiring iron directly from transferrin-like proteins through surface receptors capable of specifically binding ovotransferrin [33].
Other studies suggested that part of the antibacterial activity of Otrf is not simply due to the removal of iron from the medium, but also involves more complex mechanisms related to a direct binding of Otrf to the bacterial surface. As a matter of fact, it has been initially demonstrated that the antibacterial activity of Otrf decreased when the protein is separated by dialysis from the bacteria, a condition in which it can exert only the iron-chelating property [34]. Accordingly, it was shown that Otrf is able to permeate the E. coli outer membrane and access the inner membrane, causing both ion leakage inside bacteria and the uncoupling of the respiration-dependent energy production [35]. The antibacterial effect of Otrf towards Salmonella enterica (serovar Choleraesuis) has been also demonstrated to be dependent on culture conditions that either favor or hinder binding Otrf to the bacterial surface [36]. These data suggest that this Otrf function, not related to iron binding, could be due to a cationic bactericidal domain which, as other transferrins, is located in the N-lobe [35].
The isolation of the bactericidal domain of Otrf, was carried out by Zhou and Smith in 1990 by a partial acid proteolysis. OTAP-92 is a peptide of 9.9 kDa, consisting of 92 aminoacidic residues (Leu109-Asp200) showing strong sequence similarity with defensins [37]. This peptide is characterized by three disulfide bridges (Cys115-Cys197, Cys160-174, and Cys171-Cys182) and several positively charged residues (Figure 1, panel C).
It has been suggested that the antibacterial action of OTAP-92 may be due to its relatively high alkalinity and to the cysteine array. Both these features are shared by native antibacterial peptides [38,39] and by insect defensins whose mechanism of action involves the blocking of the voltage-dependent K+ channels [38,39,40,41,42].
As concerning the possible biotechnological applications of the Otrf antibacterial activity, Ko and coworkers [31] showed that a combination of Otrf, NaHCO3, EDTA and/or Lysozyme have a potential growth inhibition effect against E. coli O157:H7 or L. monocytogenes, demonstrating also a potential application of Otrf as a natural preservative for food (i.e., pork chops and commercial hams) [31,43,44].

4. Antiviral Activity of Ovotransferrin and Its Peptides

The antiviral activity of Otrf was firstly demonstrated towards the avian herpesvirus Marek’s disease virus (MDV), and no correlation between antiviral efficacy and iron saturation was found [45]. It has been postulated that the ovotransferrin antiviral activity towards MDV is associated with two Otrf fragments: DQKDEYELL (hOtrf219-27) and KDLLFK (hOtrf269-301 and hOtrf633-638) capable of blocking Marek’s disease virus infection in chicken embryo fibroblasts (CEF), even though the infection blocking efficiency of the isolated peptides is lower than that of the intact protein [46]. Interestingly, from an evolutionary point of view, these two Otrf peptides share sequence homology with two protein fragments, derived from human and bovine lactoferrin, known to be effective against Herpes simplex Virus (HSV-1) [47].

5. Antioxidant Activity of Ovotransferrin and Its Peptides

Ovotransferrin is a superoxide dismutase-mimicking protein exhibiting a superoxide radical (O2•−)-scavenging activity. Furthermore, self-cleaved Otrf exhibited O2•− scavenging capacity greater than intact protein [48,49]. Accordingly, it has been shown that, after digestion by thermolysin and pepsin, the resulting Otrf hydrolysates possessed significantly higher oxygen radical absorption capacity (1.69 μmol Trolox equivalent mg−1) than natural ovotransferrin [50].
Kim and coworkers [51] demonstrated that the antioxidant effects of hen’s ovotransferrin and of its hydrolyzed peptides is approximately 3.2–13.5 times higher than superoxide anion scavenging activity than Otrf, with the maximum activity displayed by octapeptides. Similar results were obtained for oxygen radical absorbance capacity assay and against the oxidative stress-induced DNA damage in human leukocytes [51]. In addition, Otrf-derived peptides showed synergistic antioxidant effects with Vitamin C, epigallocatechin gallate (EGCG), and caffeic acid [52]. Otrf hydrolyzate (obtained using enzymes such as protamex, alkalase, trypsin, and α-chymotrypsin) showed protective effects against oxidative stress including DNA damage in human leukocytes [53].
The conjugation of ovotransferrin with catechin (a polyphenol antioxidant found in tea, wine, fruits, with high affinity to bind protein) improved the oxygen radical absorbance capacity of the protein. The ovotransferrin-catechin conjugates were prepared using a hydrogen peroxide–ascorbic acid pair as radical initiator system and alkaline method [54]. Moreover, it has been also shown that catechin, after the conjugation reaction and after UPLC (Ultra-Performance Liquid Chromatography), MALDI-TOF (Matrix Assisted Laser Desorption Ionization Time-of-Flight) and Liquid chromatography-tandem mass spectrometry (LC-MS-MS) analysis, was bound to lysine (residues 327) of the Otrf peptide DLLFKDSAIMLK (residues 316–327) and to glutamic acid (residues 186) of the Otrf peptide FFSASCVPGATIE (residues 174–186) present in ovotransferrin N-lobe [54].
Autocleaved Otrf was shown to (i) hinder effectively the discoloration of ß-carotene (used as radical target in a bleaching test); (ii) prevent the oxidation of linoleic acid during five days of storage at 4 °C; and (iii) show strong Cu2+- and Ca2+-binding capacities, suggesting that it could be a good source of natural antioxidants. Once again, its metal-chelating activity could be at least partly responsible for the observed antioxidant mechanisms [55].
In conclusion, the use of Otrf-derived conjugates as a novel proteinaceous antioxidants as ingredient in the nutraceutical and functional food is desirable.

6. Anti-Inflammatory Activities of Ovotransferrin and Its Peptides

During inflammation in avians, Otrf, as well as the positive acute phase protein (APP), is up-regulated both in vivo and in vitro [56,57,58,59,60,61,62,63]; its levels in blood remain elevated as long as the inflammation persists [64,65]. Indeed, the use of blood Otrf concentration as an infection and inflammation marker in chickens has been hypothesized [66]. Otrf may act directly as an immunomodulator [63], even though its role in inflammation preventing microbial growth [67] and acting as an antioxidant against Fenton reaction products [36] cannot be ruled out.
More recently, two tripeptides, IRW and IQW, both derived from Otrf hydrolysis, were found to attenuate TNF-α-induced inflammatory responses and oxidative stress in vascular endothelial cells [68]. Furthermore, other peptides derived from ovomucoid showed an immunomodulating activity against T-cells and macrophage-stimulating activities in vitro [69], indicating that they also can be good candidates for pharmaceutical use in humans [70].

7. Other Protective Activities of Ovotransferrin and Its Peptides

In the last years, it has been reported that Otrf underwent thiol-linked auto-cleavage after reduction, and produced partially hydrolyzed products with very strong anticancer effects against colon (HCT-116) and breast cancer (MCF-7) by inducting apoptosis [71].
An antihypertensive subsidiary function of the Otrf was detected in the hen Otrf’s peptide KVREGT, showing an IC50 value of 9.1 μM towards angiotensin Ι-converting enzyme [72]. Moreover, the same peptide displayed both a strong ACE-inhibitory and a vasodilatory activities [73].
Ovotransferrin has also shown both in vitro and in vivo development promoting activity, which has been associated to its iron binding/transport capabilities. Ovotransferrin is transiently expressed and secreted in large amounts during the in vitro differentiation of hypertrophic chondrocytes into osteoblast-like cells. Cells expressing ovotransferrin also co-express ovotransferrin receptors, suggesting a self-regulatory mechanism in the control of chondrocyte differentiation to osteoblast-like cells [74,75,76,77,78,79].
Otrf shares with human and bovine lactoferrin a proteolytic activity catalyzing the hydrolysis of several synthetic substrates [80]. Serine protease inhibitors PMSF (phenylmethylsulfonyl fluoride), LPS (lipopolysaccharide) and Pefabloc impair this proteolytic activity suggesting that it is similar to that of serine proteases [81,82]. This function is conserved in several mammalian lactoferrins but not in serum transferrins, and thus it is plausible that it belongs to the protective functions of Otrf, although the physiological target has not been identified, yet.
Lastly, Ibrahim et al. [83] demonstrated the efficiency of Otrf to serve as a drug carrier to improve the solubility of three water-insoluble antibiotics and to facilitate their specific delivery into microbial or infected cells. For a complete overview of the Otrf properties, see Table 1.
Table 1. Physiological and pharmacological activities of Ovotransferrin (Otrf) and its peptides identified to-date.
Table 1. Physiological and pharmacological activities of Ovotransferrin (Otrf) and its peptides identified to-date.
PROTEIN OR PEPTIDEACTIVITYMECHANISM/SREFERENCES
Ovotransferrin (whole molecule)ANTIMICROBIAL
BACTERIAL SENSITIVITY (BACTERIOSTATIC)
  • Otrf iron Binding (Iron withholding)
[25,27,30,32]
BACTERIAL SENSITIVITY (BACTERICIDAL)
  • Membrane damage
[7,25,32]
BACTERIAL RESISTANCE
  • Bacterial production of Iron chelators or Tbp1 and 2
[33]
ANTIBACTERIAL (FOOD PRESERVATIVE)
  • In combination with EDTA and/or Lysozyme prevents E. coli O157:H7 or L. monocytogenes, proliferation
[43]
ANTIVIRAL
  • Viral adsorption inhibition
[45]
ANTIOXIDANT
  • Iron Binding
  • SuperOxide Dismutase (SOD)-like activity
  • Fenton’s reaction inhibition
  • Catechin conjugation
[49,50]
FLOGOSIS MARKER
  • Otrf Belongs to Acute Phase Proteins (APP).
  • Recognition and protection against invading pathogens, and restoration of the physiological homeostasis.
  • Otrf upregulation of interleukin-6, nitric oxide, and matrix metalloproteinase.
[61,63,64,66]
IMMUNE/ANTI-INFLAMMATION
  • Modulation of macrophages and heterophils functions.
  • SOD-Like Activity
[49,63,64]
PROTEOLYTIC
  • Hydrolysis of Haemophilus colonization factors and of several synthetic substrates
[80]
GROWTH FACTOR:
CARTILAGE NEOVASCULARIZATION
  • Chemotactic factor for endothelial cells.
  • Iron delivery
[59]
CHONDROGENESIS AND OSTEOGNESIS REGULATION
  • Iron delivery
[77,78]
MYOTROPHIC
  • Iron delivery
[74]
NEUROTROPHYC
  • Iron delivery
[79]
CARRIER FOR DRUG DELIVERY
  • Binding and delivery of water insoluble antibiotics (sulphantibiotics)
[83]
Otrf peptide OTAP-92ANTIMICROBIAL
  • Bacterial Membrane damage
[41]
Otrf peptides219–227; 269–301; 633–638ANTIVIRAL
  • Viral adsorption inhibition
[46]
Reduced autocleaved Otrf (rac-Otrf)ANTICANCER/ANTIPROLIFERATION
  • Apoptosis induction
[71]
ANTIOXIDANT
  • Autocleaved Otrf as preservative to prevent ß-carotene discoloration
[55]
Otrf Peptides (IRW or IQW)ANTINFLAMMATORY
  • Attenuate TNF-α-induced inflammatory responses
[68]
Otrf peptide (KVREGT)ANTIHYPERTENSIVE
  • Inhibition of Angiotensin Ι-Converting Enzyme.
[72,73]
Otrf Peptides (DLLFKDSAIMLK) (FFSASCVPGATIE)ANTIOXIDANT
  • Catechin conjugation
[54]
Otrf peptides (mix obtained using: protamex, or alkalase, or trypsin, or α-chymotrypsin)ANTIOXIDANT
  • Synergistic antioxidant effects with vitamin C, epigallocatechin gallate (EGCG), and caffeic acid.
[52,53]

8. Conclusions

Many of the Otrf’s protective properties, described in this review and outlined in Table 1, contribute, in addition to the well-known iron transfer activity, to the proper development of the chicken embryo. Moreover, the demonstration that several defensive properties of Otrf are also possessed by its proteolytic fragments and that these properties may be of importance for human wellness strongly support the use of egg white (preferably raw or cooked at low temperature to preserve ovotransferrin properties) and its derivatives as dietary additives in normal and pathological human conditions.

Author Contributions

The authors warrant that all of the authors have contributed substantially to the manuscript and approved the final submission.

Conflicts of Interests

The authors warrant the absence of any real or perceived conflicts of interest.

References

  1. Giansanti, F.; Leboffe, L.; Pitari, G.; Ippoliti, R.; Antonini, G. Physiological roles of ovotransferrin. Biochim. Biophys. Acta 2012, 1820, 218–225. [Google Scholar] [CrossRef] [PubMed]
  2. Deeming, D.C. Behavior patterns during incubation. In Avian Incubation: Behaviour, Environment, and Evolution; Deeming, D.C., Ed.; Oxford University Press: Oxford, UK, 2002; pp. 63–87. [Google Scholar]
  3. Saxena, I.; Tayyab, S. Protein proteinase inhibitors from avian egg whites. Cell. Mol. Life Sci. 1997, 53, 13–23. [Google Scholar] [CrossRef] [PubMed]
  4. Wesierska, E.; Saleh, Y.; Trziska, T.; Kopec, W.; Sierwinski, M.; Korzekwa, K. Antimicrobial activity of chicken egg white cystatin. World J. Microbiol. Biotechnol. 2005, 21, 59–64. [Google Scholar] [CrossRef]
  5. Miyagawa, S.; Matsumaoto, K.; Kamata, R.; Okamura, R.; Maeda, H. Spreading of Serratia marcescens in experimental keratitis and growth suppression by chicken egg white ovomacroglobulin. Jpn. J. Ophthalmol. 1991, 35, 402–410. [Google Scholar] [PubMed]
  6. Board, P.A.; Fuller, R. Non-specific antimicrobial defences of the avian egg, embryo and neonate. Biol. Rev. Camb. Philos. Soc. 1974, 49, 15–49. [Google Scholar] [CrossRef] [PubMed]
  7. Wellman-Labadie, O.; Picman, J.; Hinke, M.T. Comparative antibacterial activity of avian egg white protein extracts. Br. Poult. Sci. 2008, 49, 125–132. [Google Scholar] [CrossRef] [PubMed]
  8. Walther, B.; Sieber, R. Bioactive proteins and peptides in foods. Int. J. Vitam. Nutr. Res. 2011, 81, 181–192. [Google Scholar] [CrossRef] [PubMed]
  9. Jeltsch, J.M.; Chambon, P. The complete nucleotide sequence of the chicken ovotransferrin mRNA. Eur. J. Biochem. 1982, 122, 291–295. [Google Scholar] [CrossRef] [PubMed]
  10. Williams, J.; Elleman, T.C.; Kingston, I.B.; Wilkins, A.G.; Kuhn, K.A. The primary structure of hen ovotransferrin. Eur. J. Biochem. 1982, 122, 297–303. [Google Scholar] [CrossRef] [PubMed]
  11. Sutherland, R.L.; Geynet, C.; Binart, N.; Catelli, M.G.; Schmelck, P.H.; Mester, J.; Lebeau, M.C.; Baulieu, E.E. Steroid receptors and effects of oestradiol and progesterone on chick oviduct proteins. Eur. J. Biochem. 1980, 107, 155–164. [Google Scholar] [CrossRef] [PubMed]
  12. Dierich, A.; Gaub, M.P.; LePennec, J.P.; Astinotti, D.; Chambon, P. Cell-specificity of the chicken ovalbumin and conalbumin promoters. EMBO J. 1987, 6, 2305–2312. [Google Scholar] [PubMed]
  13. Williams, J. A comparison of glycopeptides from the ovotransferrin and serum transferrin of the hen. Biochem. J. 1968, 108, 57–67. [Google Scholar] [CrossRef] [PubMed]
  14. Iwase, H.; Hotta, K. Ovotransferrin subfractionation dependent upon chain differences. J. Biol. Chem. 1977, 252, 5437–5443. [Google Scholar] [PubMed]
  15. Jacquinot, P.M.; Leger, D.; Wieruszeski, J.M.; Coddeville, B.; Montreuil, J.; Spik, G. Change in glycosylation of chicken transferrin glycans biosynthesized during embryogenesis and primary culture of embryo hepatocytes. Glycobiology 1994, 4, 617–624. [Google Scholar] [CrossRef] [PubMed]
  16. Kurokawa, H.; Mikami, B.; Hirose, M. Crystal structure of diferric hen ovotransferrin at 2.4 Å resolution. J. Mol. Biol. 1995, 254, 196–207. [Google Scholar] [CrossRef] [PubMed]
  17. Kurokawa, H.; Dewan, J.C.; Mikami, B.; Sacchettini, J.C.; Hirose, M. Crystal structure of hen apo-ovotransferrin: Both lobes adopt an open conformation upon loss of iron. J. Biol. Chem. 1999, 274, 28445–28452. [Google Scholar] [CrossRef] [PubMed]
  18. Thakurta, G.P.; Choudhury, D.; Dasgupta, R.; Dattagupta, J.K. Structure of diferric hen serum transferrin at 2.8 Å resolution. Acta Crystallogr. Sect. D 2003, 59, 1773–1781. [Google Scholar] [CrossRef]
  19. Williams, J. The evolution of transferrins. Trends Biochem. Sci. 1982, 7, 394–397. [Google Scholar] [CrossRef]
  20. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef] [PubMed]
  21. Williams, J.; Moreton, K.; Goodearl, A.D. Selective reduction of a disulphide bridge in hen ovotransferrin. Biochem. J. 1985, 228, 661–665. [Google Scholar] [CrossRef] [PubMed]
  22. Mizutani, K.; Yamashita, H.; Kurokawa, H.; Mikami, B.; Mikami, B. Alternative structural state of transferrin. The crystallographic analysis of iron-loaded but domain-opened ovotransferrin N-lobe. J. Biol. Chem. 1999, 274, 10190–10194. [Google Scholar] [CrossRef] [PubMed]
  23. Lindley, P.F.; Bajaj, M.; Evans, R.W.; Garatt, R.C.; Hasnain, S.S.; Jhoti, H.; Kuser, P.; Neu, M.; Patel, K.; Sarra, R.; et al. The mechanism of iron uptake by transferrins: The structure of an 18 kDa NII-domain fragment from duck ovotransferrin at 2.3 Å resolution. Acta Crystallogr. Sect. D 1993, 49, 292–304. [Google Scholar] [CrossRef] [PubMed]
  24. Kuser, P.; Hall, D.R.; Haw, M.L.; Neu, M.; Evans, R.W.; Lindley, P.F. The mechanism of iron uptake by transferrins: The X-ray structures of the 18 kDa NII domain fragment of duck ovotransferrin and its nitrilotriacetate complex. Acta Crystallogr. Sect. D 2002, 58, 777–783. [Google Scholar] [CrossRef]
  25. Alderton, G.; Ward, W.H.; Fevold, H.L. Identification of the bacteria-inhibiting iron-binding protein of egg white as conalbumin. Arch. Biochem. 1946, 11, 9–13. [Google Scholar] [PubMed]
  26. Bullen, J.J.; Rogers, H.J.; Griffiths, E. Role of iron in bacterial infection. Curr. Top. Microbiol. Immunol. 1978, 80, 1–35. [Google Scholar] [PubMed]
  27. Antonini, E.; Orsi, N.; Valenti, P. Effetto delle transferrine sulla patogenicità delle Enterobacteriaceae. G. Mal. Infett. Parassit. 1977, 29, 481–489. [Google Scholar]
  28. Valenti, P.; de Stasio, A.; Mastromarino, P.; Seganti, L.; Sinibaldi, L.; Orsi, N. Influence of bicarbonate and citrate on the bacteriostatic action of ovotransferrin towards staphylococci. FEMS Microbiol. Lett. 1981, 10, 77–79. [Google Scholar] [CrossRef]
  29. MacGillivray, R.T.; Moore, S.A.; Chen, J.; Anderson, B.F.; Baker, H.; Luo, Y.; Bewley, M.; Smith, C.A.; Murphy, M.E.; Wang, Y.; et al. Two high-resolution crystal structures of the recombinant N-lobe of human transferrin reveal a structural change implicated in iron release. Biochemistry 1998, 37, 7919–7928. [Google Scholar] [CrossRef] [PubMed]
  30. Valenti, P.; Antonini, G.; Fanelli, M.R.; Orsi, N.; Antonini, E. Antibacterial activity of matrixbound ovotransferrin. Antimicrob. Agents Chemother. 1982, 21, 840–841. [Google Scholar] [CrossRef] [PubMed]
  31. Ko, K.Y.; Mendonca, A.F.; Ahn, D.U. Effect of ethylenediaminetetraacetate and lysozyme on the antimicrobial activity of ovotransferrin against Listeria monocytogenes. Poult. Sci. 2008, 87, 1649–1658. [Google Scholar] [CrossRef] [PubMed]
  32. Valenti, P.; Antonini, G.; von Hunolstein, C.; Visca, P.; Orsi, N.; Antonini, E. Studies of the antimicrobial activity of ovotransferrin. Int. J. Tissue React. 1983, 5, 97–105. [Google Scholar] [PubMed]
  33. Alcantara, J.; Schryvers, A.B. Transferrin binding protein two interacts with both the N-lobe and C-lobe of ovotransferrin. Microb. Pathog. 1996, 20, 73–85. [Google Scholar] [CrossRef] [PubMed]
  34. Valenti, P.; Visca, P.; Antonini, G.; Orsi, N. Antifungal activity of ovotransferrin toward genus Candida. Mycopathologia 1985, 89, 169–175. [Google Scholar] [CrossRef] [PubMed]
  35. Aguilera, O.; Quiros, L.M.; Fierro, J.F. Transferrins selectively cause ion efflux through bacterial and artificial membranes. FEBS Lett. 2003, 548, 5–10. [Google Scholar] [CrossRef]
  36. Superti, F.; Ammendolia, M.G.; Berlutti, F.; Valenti, P. Ovotransferrin. In Bioactive Egg Compounds; Huopalahti, R., Lopez-Fandino, R., Eds.; Springer-Verlag: Berlin, Germany, 2007; pp. 43–48. [Google Scholar]
  37. Zhou, Z.R.; Smith, D.L. Assignment of disulfide bonds in proteins by partial acid hydrolysis and mass spectrometry. J. Protein Chem. 1990, 9, 523–532. [Google Scholar] [CrossRef] [PubMed]
  38. Strahilevitz, J.; Mor, A.; Nicolas, P.; Shai, Y. Spectrum of antimicrobial activity and assembly of dermaseptin-β and its precursor form in phospholipid membranes. Biochemistry 1994, 33, 10951–10960. [Google Scholar] [CrossRef] [PubMed]
  39. Ehret-Sabatier, L.; Loew, D.; Goyffon, M.; Fehlbaum, P.; Hoffmann, J.A.; Dorsselaer, A.V.; Bulet, P. Characterization of novel cysteine-rich antimicrobial peptides from scorpion blood. J. Biol. Chem. 1996, 271, 29537–29544. [Google Scholar] [CrossRef] [PubMed]
  40. Ibrahim, H.R.; Iwamori, E.; Sugimoto, Y.; Aoki, T. Identification of a distinct antibacterial domain within the N-lobe of Ovotransferrin. Biochim. Biophys. Acta 1998, 1401, 289–303. [Google Scholar] [CrossRef]
  41. Ibrahim, H.R.; Sugimoto, Y.; Aoki, T. Ovotransferrin antimicrobial peptide (OTAP-92) kill bacteria through a membrane damage mechanism. Biochim. Biophys. Acta 2000, 1523, 196–205. [Google Scholar] [CrossRef]
  42. Galvez, A.; Gimenez-Gallego, G.; Reuben, J.P.; Roy-Contancin, L.; Feigenbaum, P.; Kaczorowski, G.J.; Garcia, M.L. Purification and characterization of a unique, potent, peptidyl probe for the high conductance calcium-activated potassium channel from venom of the scorpion Buthus tamulus. J. Biol. Chem. 1990, 265, 11083–11090. [Google Scholar] [PubMed]
  43. Ko, K.Y.; Mendoncam, A.F.; Ismail, H.; Ahn, D.U. Ethylenediaminetetraacetate and lysozyme improves antimicrobial activities of ovotransferrin against Escherichia coli O157:H7. Poult. Sci. 2009, 88, 406–414. [Google Scholar] [CrossRef] [PubMed]
  44. Seol, K.H.; Lim, D.G.; Jang, A.; Jo, C.; Lee, M. Antimicrobial effect of kappa carrageenan-based edible film containing ovotransferrin in fresh chicken breast stored at 5 °C. Meat Sci. 2009, 83, 479–483. [Google Scholar] [CrossRef] [PubMed]
  45. Giansanti, F.; Rossi, P.; Massucci, M.T.; Botti, D.; Antonini, G.; Valenti, P.; Seganti, L. Antiviral activity of ovotransferrin discloses an evolutionary strategy for the defensive activities of lactoferrin. Biochem. Cell Biol. 2002, 80, 125–130. [Google Scholar] [CrossRef] [PubMed]
  46. Giansanti, F.; Massucci, M.T.; Giardi, M.F.; Nozza, F.; Pulsinelli, E.; Nicolini, C.; Botti, D.; Antonini, G. Antiviral activity of ovotransferrin derived peptides. Biochem. Biophys. Res. Commun. 2005, 331, 69–73. [Google Scholar] [CrossRef] [PubMed]
  47. Siciliano, R.; Rega, B.; Marchetti, M.; Seganti, L.; Antonini, G.; Valenti, P. Bovine lactoferrin peptidic fragments involved in inhibition of Herpes simplex virus type 1 infection. Biochem. Biophys. Res. Commun. 1999, 264, 19–23. [Google Scholar] [CrossRef] [PubMed]
  48. Ibrahim, H.R.; Haraguchi, T.; Aoki, T. Ovotransferrin is a redox-dependent autoprocessing protein incorporating four consensus self-cleaving motifs flanking the two kringles. Biochim. Biophys. Acta 2006, 1760, 347–355. [Google Scholar] [CrossRef] [PubMed]
  49. Ibrahim, H.R.; Hoq, M.I.; Aoki, T. Ovotransferrin possesses SOD-like superoxide anion scavenging activity that is promoted by copper and manganese binding. Int. J. Biol. Macromol. 2007, 41, 631–640. [Google Scholar] [CrossRef] [PubMed]
  50. Huang, W.Y.; Majumder, K.; Wu, J. Oxygen radical absorbance capacity of peptides from egg white protein ovotransferrin and their interaction with phytochemicals. Food Chem. 2010, 123, 635–641. [Google Scholar] [CrossRef]
  51. Kim, J.; Moon, S.H.; Ahn, D.U.; Paik, H.D.; Park, E. Antioxidant effects of ovotransferrin and its hydrolysates. Poult. Sci. 2012, 91, 2747–2754. [Google Scholar] [CrossRef] [PubMed]
  52. Huang, W.; Shen, S.; Nimalaratne, C.; Li, S.; Majumder, K.; Wu, J. Effects of addition of egg ovotransferrin-derived peptides on the oxygen Radical absorbance capacity of different teas. Food Chem. 2012, 135, 1600–1607. [Google Scholar] [CrossRef] [PubMed]
  53. Moon, S.H.; Lee, J.H.; Lee, Y.J.; Chang, K.H.; Paik, J.Y.; Ahn, D.U.; Paik, H.D. Screening for cytotoxic activity of ovotransferrin and its enzyme hydrolysates. Poult. Sci. 2013, 92, 424–434. [Google Scholar] [CrossRef] [PubMed]
  54. You, J.; Luo, Y.; Wu, J. Conjugation of ovotransferrin with catechin shows improved antioxidant activity. J. Agric. Food Chem. 2014, 62, 2581–2587. [Google Scholar] [CrossRef] [PubMed]
  55. Moon, S.H.; Lee, J.H.; Ahnb, D.U.; Paika, H.D. In vitro antioxidant and mineral-chelating properties of natural and autocleaved ovotransferrin. J. Sci. Food Agric. 2015, 95, 2065–2070. [Google Scholar] [CrossRef] [PubMed]
  56. Morgan, R.W.; Sofer, L.; Anderson, A.S.; Berneberg, E.L.; Cui, J.; Burnside, J. Induction of host gene expression following infection of chicken embryo fibroblasts with oncogenic Marek’s disease virus. J. Virol. 2001, 75, 533–539. [Google Scholar] [CrossRef] [PubMed]
  57. Kushner, I.; Mackiewicz, A. The acute phase response: An overview. In Acute Phase Proteins: Molecular Biology, Biochemistry, and Clinical Applications; Kushner, I., Baumann, H., Mackiewicz, A., Eds.; CRC Press: Boca Raton, FL, USA, 1993; pp. 3–19. [Google Scholar]
  58. Gabay, C.; Kushner, I. Acute phase proteins and other systemic response to inflammation. N. Engl. J. Med. 1999, 340, 448–454. [Google Scholar] [PubMed]
  59. Carlevaro, M.F.; Albini, A.; Ribatti, D.; Gentili, C.; Benelli, R.; Cermelli, S.; Cancedda, R.; Cancedda, F.D. Transferrin promotes endothelial cell migration and invasion: Implication in cartilage neovascularisation. J. Cell Biol. 1997, 136, 1375–1384. [Google Scholar] [CrossRef] [PubMed]
  60. Hallquist, N.A.; Klasing, K.C. Serotransferrin, ovotransferrin and metallothionein levels during an immune response in chickens. Comp. Biochem. Physiol. B 1994, 108, 375–384. [Google Scholar] [CrossRef]
  61. Tohjo, H.; Miyoshi, F.; Uchida, E.; Niiyama, M. Polyacrylamide gel electrophoretic patterns of chicken serum in acute inflammation induced by intramuscular injection of turpentine. Poult. Sci. 1995, 74, 648–655. [Google Scholar] [CrossRef] [PubMed]
  62. Chamanza, R.; Toussaint, M.J.M.; van Ederen, A.M.; van Veen, L.; Hulskamp-Koch, C. Serum amyloid A and transferrin in chicken. A preliminary investigation of using acute phase variables to assess diseases in chickens. Vet. Q. 1999, 21, 158–162. [Google Scholar] [CrossRef] [PubMed]
  63. Xie, H.; Huff, G.R.; Huff, W.E.; Balog, J.M.; Rath, N.C. Effects of ovotransferrin on chicken macrophages and heterophil-granulocytes. Dev. Comp. Immunol. 2002, 26, 805–815. [Google Scholar] [CrossRef]
  64. Xie, H.; Huff, G.R.; Huff, W.E.; Balog, J.M.; Holt, P.; Rath, N.C. Identification of ovotransferrin as an acute phase protein in chickens. Poult. Sci. 2002, 81, 112–120. [Google Scholar] [CrossRef] [PubMed]
  65. Rath, N.C.; Xie, H.; Huff, W.E.; Huff, G.R. Avian acute phase protein ovotransferrin modulates phagocyte function. In New Immunology Research Development; Muller, G.V., Ed.; Nova Science Publishers: New York, NY, USA, 2008; pp. 95–108. [Google Scholar]
  66. Rath, N.C.; Anthony, N.B.; Kannan, L.; Huff, W.E.; Huff, G.R.; Chapman, H.D.; Erf, G.F.; Wakenell, P. Serum ovotransferrin as a biomarker of inflammatory diseases in chickens. Poult. Sci. 2009, 88, 2069–2074. [Google Scholar] [CrossRef] [PubMed]
  67. Giansanti, F.; Giardi, M.F.; Massucci, M.T.; Botti, D.; Antonini, G. Ovotransferrin expression and release by chicken cell lines infected with Marek’s disease virus. Biochem. Cell Biol. 2007, 85, 150–155. [Google Scholar] [CrossRef] [PubMed]
  68. Majumder, K.; Chakrabarti, S.; Davidge, S.T.; Wu, J. Structure and activity study of egg protein ovotransferrin derived peptides (IRW and IQW) on endothelial inflammatory response and oxidative stress. J. Agric. Food Chem. 2013, 61, 2120–2129. [Google Scholar] [CrossRef] [PubMed]
  69. Kovacs-Nolan, J.; Phillips, M.; Mine, Y. Advances in the value of eggs and egg components for human health. J. Agric. Food Chem. 2005, 53, 8421–8431. [Google Scholar] [CrossRef] [PubMed]
  70. Abeyrathne, E.D.N.S.; Lee, H.Y.; Ahn, D.U. Egg white proteins and their potential use in food processing or as nutraceutical and pharmaceutical agents—A review. Poult. Sci. 2013, 92, 3292–3299. [Google Scholar] [CrossRef] [PubMed]
  71. Ibrahim, H.R.; Kiyono, T. Novel anticancer activity of the autocleaved ovotransferrin against human colon and breast cancer cells. J. Agric. Food Chem. 2009, 57, 11383–11390. [Google Scholar] [CrossRef] [PubMed]
  72. Lee, N.Y.; Cheng, J.T.; Enomoto, T.; Nakano, Y. One peptide derived from hen ovotransferrin as pro-drug to inhibit angiotensin converting enzyme. J. Food Drug Anal. 2006, 14, 31–35. [Google Scholar] [CrossRef]
  73. Wu, J.; Acero-Lopez, A. Ovotransferrin: Structure, bioactivities and preparation. Food Res. Int. 2012, 46, 480–487. [Google Scholar] [CrossRef]
  74. Shimo-Oka, T.; Hagiwara, Y.; Ozawa, E. Class specificity of transferrin as a muscle trophic factor. J. Cell. Physiol. 1986, 126, 341–351. [Google Scholar] [CrossRef] [PubMed]
  75. Leitner, D.F.; Connor, J.R. Functional roles of transferrin in the brain. Biochim. Biophys. Acta 2012, 1820, 393–402. [Google Scholar] [CrossRef] [PubMed]
  76. Paek, S.H.; Shin, H.Y.; Kim, J.W.; Park, S.H.; Son, J.H.; Kim, D.G. Primary culture of central neurocytoma: A case report. J. Korean Med. Sci. 2010, 25, 798–803. [Google Scholar] [CrossRef] [PubMed]
  77. Cancedda, R.; Castagnola, P.; Cancedda, F.D.; Dozin, B.; Quarto, R. Developmental control of chondrogenesis and osteogenesis. Int. J. Dev. Biol. 2000, 44, 707–714. [Google Scholar] [PubMed]
  78. Gentili, C.; Doliana, R.; Bet, P.; Campanile, G.; Colombatti, A.; Cancedda, F.D.; Cancedda, R. Ovotransferrin and ovotransferrin receptor expression during chondrogenesis and endochondral bone formation in developing chick embryo. J. Cell Biol. 1994, 124, 579–588. [Google Scholar] [CrossRef] [PubMed]
  79. Bruinink, A.; Sidler, C.; Birchler, F. Neurotrophic effects of transferrin on embryonic chick brain and neural retinal cell cultures, Int. J. Dev. Neurosci. 1996, 14, 785–795. [Google Scholar] [CrossRef]
  80. Leboffe, L.; Giansanti, F.; Antonini, G. Antifungal and antiparasitic activities of lactoferrin. Anti-Infect. Agents Med. Chem. 2009, 8, 114–127. [Google Scholar] [CrossRef]
  81. Massucci, M.T.; Giansanti, F.; di Nino, G.; Turacchio, M.; Giardi, M.F.; Botti, D.; Ippoliti, R.; de Giulio, B.; Siciliano, R.A.; Donnarumma, G.; et al. Proteolytic activity of bovine lactoferrin. Biometals 2004, 17, 249–255. [Google Scholar] [CrossRef] [PubMed]
  82. Hendrixson, D.R.; Qiu, J.; Shewry, S.C.; Fink, D.L.; Petty, S.; Baker, E.N.; Plaut, A.G.; St Geme, J.W., 3rd. Human milk lactoferrin is a serine protease that cleaves Haemophilus surface proteins at arginine-rich sites. Mol. Microbiol. 2003, 47, 607–617. [Google Scholar] [CrossRef] [PubMed]
  83. Ibrahim, H.R.; Tatsumoto, S.; Ono, H.; van Immerseel, F.; Raspoet, R.; Miyata, T. A novel antibiotic-delivery system by using ovotransferrin as targeting Molecule. Eur. J. Pharm. Sci. 2015, 66, 59–69. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Giansanti, F.; Leboffe, L.; Angelucci, F.; Antonini, G. The Nutraceutical Properties of Ovotransferrin and Its Potential Utilization as a Functional Food. Nutrients 2015, 7, 9105-9115. https://doi.org/10.3390/nu7115453

AMA Style

Giansanti F, Leboffe L, Angelucci F, Antonini G. The Nutraceutical Properties of Ovotransferrin and Its Potential Utilization as a Functional Food. Nutrients. 2015; 7(11):9105-9115. https://doi.org/10.3390/nu7115453

Chicago/Turabian Style

Giansanti, Francesco, Loris Leboffe, Francesco Angelucci, and Giovanni Antonini. 2015. "The Nutraceutical Properties of Ovotransferrin and Its Potential Utilization as a Functional Food" Nutrients 7, no. 11: 9105-9115. https://doi.org/10.3390/nu7115453

Article Metrics

Back to TopTop