Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1786 2024-02-20 16:46:47

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Santos, I.; Silva, M.; Grácio, M.; Pedroso, L.; Lima, A. Milk-Derived Antiviral Peptides Targeting Zoonotic Viruses. Encyclopedia. Available online: https://encyclopedia.pub/entry/55244 (accessed on 05 May 2024).
Santos I, Silva M, Grácio M, Pedroso L, Lima A. Milk-Derived Antiviral Peptides Targeting Zoonotic Viruses. Encyclopedia. Available at: https://encyclopedia.pub/entry/55244. Accessed May 05, 2024.
Santos, Isabel, Mariana Silva, Madalena Grácio, Laurentina Pedroso, Ana Lima. "Milk-Derived Antiviral Peptides Targeting Zoonotic Viruses" Encyclopedia, https://encyclopedia.pub/entry/55244 (accessed May 05, 2024).
Santos, I., Silva, M., Grácio, M., Pedroso, L., & Lima, A. (2024, February 20). Milk-Derived Antiviral Peptides Targeting Zoonotic Viruses. In Encyclopedia. https://encyclopedia.pub/entry/55244
Santos, Isabel, et al. "Milk-Derived Antiviral Peptides Targeting Zoonotic Viruses." Encyclopedia. Web. 20 February, 2024.
Milk-Derived Antiviral Peptides Targeting Zoonotic Viruses
Edit

Animals often serve as reservoirs for viral zoonoses, diseases transmissible from animals to humans. While zoonotic viral diseases have been present in human populations since the inception of agricultural practices, they have gained increasing prominence as a global public health concern, particularly with recent epidemics like SARS-CoV-2 (COVID-19). Some of these diseases are categorized as “emerging infectious diseases” due to their newfound recognition or significant changes in their range and epidemiology. Notable zoonotic diseases include influenza, Ebola virus, West Nile virus, emerging coronaviruses, monkeypox, rabies, Zika, and Lyme disease. Six out of every ten infectious diseases in humans are zoonotic, with many being viral. Therefore, it is imperative to enhance our capabilities to prevent and respond to these diseases, adopting a One Health approach. Finding new therapies and ways to prevent viral zoonoses is just as important as increasing efforts on surveillance and early detection, and with the rising amount of research demonstrating the potential of bioactive peptides produced from milk as antivirals, a vital opportunity arises to assess their usage in viral diseases with a focus on One Health. Indeed, several peptides derived from milk protein parents have shown potential for zoonotic viral diseases. 

milk peptides zoonoses antiviral one health

1. COVID-19

Severe acute respiratory syndrome (SARS) is a viral respiratory disease induced by SARS-associated coronavirus (SARS-CoV) which affected Asia, North America, and Europe in 2002–2003 [1]. In late 2019, a new type of coronavirus called SARS-CoV-2 caused a viral pandemic known as Coronavirus disease (COVID-19), which led to severe respiratory problems. Worldwide, researchers searched earnestly for molecules that could help create therapies for COVID-19 prevention and treatment. Coronaviruses enter target cells by fusing the membranes of the virus and the host cell, which is mediated by a viral spike glycoprotein (S protein) [2]. Earlier in 2020 a study showed that whey proteins from human, goat, and cow milk could inhibit SARS-CoV-2 entry and replication in Vero E6 and A549 cell lines, with an EC50 of about 0.13 mg/mL of total protein [3]. Among these molecules, milk and whey proteins, particularly lactoferrin, have been identified as one of the main antivirals present in milk and have been reviewed [4]. Lactoferrin was shown to present an array of activities against the virus, including a high affinity with the spike domain [5], competing for binding to an ACE2 receptor [6], and blocking the spike protein Furin-cleavage site [7]. It also hinders viral attachment by binding at the level of heparan sulfate proteoglycans [8][9] and binding to sialic acid [6]. Lactoferrin also reduces SARS-CoV-2 infectivity by inhibiting cathepsin L activity [10] and acts as an immune modulator of the antiviral immune response [11]. Lactoferrin was also shown to potentiate the effect of certain treatments towards SARS-CoV-2, such as remdesivir, hypothiocyanite anion, and the oral administration of liposomal Lf and oral zinc solution [9][12][13][14][15]. In addition to lactoferrin, other milk proteins were demonstrated to present antiviral activity against SARS-CoV-2 [16]; in particular, two β-lactoglobulin-derived peptides, produced by trypsin digestion, Ala-Leu-Pro-Met-His-Ile-Arg (ALMPHIR) and Ile-Pro-Ala-Val-Phe-Lys (IPAVFK), exhibited the ability to inactivate the virus via cathepsin inhibition and binding to the spike protein and also the host cell membrane receptor, showing great potential as a treatment for SARS-CoV-2 [10][17].
Interestingly, recent works have shown that milk-derived peptides are effective multi-targeted therapeutic candidates to treat SARS-CoV-2. Pradeep et al. [16] showed an interesting strategy involving the concurrent blockade of diverse pathways in the infectious cycle of SARS-CoV-2 to mitigate the COVID-19 threat. Through a combination of molecular docking, molecular simulation, heat mapping, and manual interpretation, the study developed a new strategy to identify a plethora of peptides capable of impeding the spread of the coronavirus [16].

2. Human Immunodeficiency Virus (HIV)

Human immunodeficiency virus infection and acquired immunodeficiency syndrome (HIV/AIDS) are caused by a retrovirus known as the human immunodeficiency virus (HIV). HIV is a life-threatening infection that has spread globally since the first case was discovered, leading to a significant need for effective antiviral therapies. Milk-derived proteins such as human and bovine lactoferrin, modified β-lactoglobulin, α-lactalbumin, and lactoperoxidase have all demonstrated antiviral properties against human immunodeficiency viruses, mostly via the ability to link themselves to host cell receptors, inhibiting not only viral absorption but also the virus replication cycle [18][19][20][21][22][23].
Bovine lactoferrin and its derived peptide lactoferricin inhibit HIV-1 by acting on CXCR4 and CCR5 receptors [18][23]. Recently Berkhout et al. [18] showed that lactoferricin presents a lower inhibition when compared to lactoferrin, suggesting that other domains in the native protein may also aid in inhibition. Indeed, the apo form of bovine lactoferrin also inhibits HIV-1 replication in a different study [24]. Other whey proteins were also shown to inhibit the HIV virus. Modified β-lactoglobulin and α-lactalbumin were able to block the virus entry in the host cells due to interactions with the gp120 envelope protein [25][26], and Tenascin-C also displayed antiviral activity against HIV by interacting with its envelope domain, effectively neutralizing the retrovirus, and preventing transmission via breast milk [27]. In fact, the potency of Tenascin-C’s inhibition surpasses that of lactoferrin and is comparable to HIV-1-neutralizing monoclonal antibodies [28].

3. Human Cytomegalovirus (HCMV)

Human cytomegalovirus (HCMV), also known as human herpesvirus 5 (HHV-5), is a virus that belongs to the Herpesviridae family. This virus can enter the human body through contact with mucous membranes or through blood components containing cells, as well as through stem cell/organ transplants [29]. In human cytomegalovirus infection, lactoferrin, lactoferricin, and methylated β-lactoglobulin and α-lactalbumin can inhibit virus replication and transcription by interacting with the viral genome [30][31]. Lactoferrin and lactoferricin display different mechanisms of action, being able to both interfere with the virus target cells and up-regulate the immune system, but also exerting a synergistic antiviral effect with cidofovir, an antiviral drug commonly used in patients with human cytomegalovirus [32][33][34][35][36][37]. Methylated β-lactoglobulin and α-lactalbumin, on the other hand, inhibit viral replication and transcription by interacting with the viral genome [4][30].

4. Hepatitis B

Hepatitis B is an often-life-threatening liver infection caused by the hepatitis B virus (HBV). It is a major global health problem that can cause chronic infection and puts people at great risk of death from cirrhosis and liver cancer. The inhibition of hepatitis B (and C) viruses was shown by human and bovine lactoferrin (and peptide derivatives) as well as by α-lactalbumin, β-lactoglobulin, and lysozyme, via interaction between viral and cell proteins that interfere with virus entry and multiplication [38]. Here, lactoferrin, when saturated with zinc or iron, showed antiviral activity against this disease. As for the mechanism of action, lactoferrin, saturated with the positive ions mentioned above, was able to bind to several molecules of the host cell and, therefore, interfere with the virus’s ability to attach itself to the host cell and its ability to enter the host cell [39][40].

5. Influenza

Influenza viruses seriously threaten global health, causing substantial morbidity and mortality, especially among individuals with weakened immune systems [41]. Whilst vaccination remains a cornerstone for infection control, its effectiveness is compromised by the rapid antigenic drift and the emergence of new viral subtypes [41]. Breastfeeding has been acknowledged for its protective role against respiratory and gastrointestinal infections in infants, so it would be expected that milk-derived peptides exert some activity against this virus [42]. Indeed, an early study found that lactoferrin exerts a protective effect against influenza-induced apoptosis by modulating caspase 3 function and impeding the export of viral ribonucleoproteins from the nucleus to the cytoplasm [43]. Subsequent research revealed a lactoferrin-derived peptide, bLf, which was shown to bind to the viral HA, inhibiting both hemagglutination and infection by influenza A viruses, of both group 1 and group 2 subtypes. Notably, bLf demonstrated binding to the HA2 subunit, which harbors the universally conserved HA epitope, explaining the broad-spectrum anti-influenza activity observed [41]. In a more recent study, Scala et al. [44] delved deeper into the inhibitory potential of lactoferrin-derived peptides against influenza virus infection and identified novel sequences derived from the C-lobe of bovine lactoferrin, demonstrating broad anti-influenza activity and the ability to prevent viral hemagglutination and infection at remarkably low concentrations [44]. Some in vitro studies have reported that lactoferrin and lactoperoxidase may also depend on other mechanisms, such as the inhibition of viral shedding, leading to the suppression of influenza virus A (H1N1) [45][46]. Bovine lactoferrin was found to interact with viral haemagglutinin, which resulted in the inhibition of virus-induced haemagglutination for influenza A virus. [41]; a similar mechanism of action was observed for glycomacropeptide in previous studies on influenza virus A [47].

6. Zika and Usutu Virus

Zika virus is a mosquito-borne flavivirus that gained global attention due to its association with serious health concerns. Infection with Zika virus has been linked to neurological complications, including microcephaly in infants born to infected mothers, making it a significant public health issue [48]. A recent study [49] explored the antiviral properties of human milk at different stages of maturation against Zika virus and Usutu virus. The results indicated that human milk exhibited antiviral activity against both Zika virus and Usutu virus across all stages of lactation, and that extracellular vesicles and glycosaminoglycans played a role in the protective effect of milk, with no significant variations observed between colostrum, transitional, or mature milk. Mechanistic studies revealed that the mechanism was not due to the inactivation of the viral particles but was instead due to blocking the binding of both flaviviruses to cells.

7. Rotavirus

Rotavirus is a global pathogen that is the major cause of severe diarrhea in infant mammals. In vitro tests showed the ability of human milk fractions to inhibit rotavirus replication, [50] particularly a mucin complex fraction containing the milk-fat globule membrane proteins MUC, lactadherin, and an unidentified 80-kDa whey protein [51][52]. While it was suggested that lactadherin might be responsible for the action of the mucin complex [51][52], MUC1 showed antiviral activity by inhibiting the replication of 3 human rotavirus strains [50]. Interestingly, only the human form of lactadherin could inhibit Wa rotavirus infection in vitro, apparently through a mechanism involving protein–virus interactions, which is dependent on the protein structure or the attached oligosaccharides [53]. According to Yolken et al. [51], the sialic acid present in lactadherin also plays a vital role in its antiviral action. Also, several whey proteins, including apo-lactoferrin (iron-free), homo-lactoferrin (carrying Fe3+), α-lactalbumin, and β-lactoglobulin, have demonstrated the capacity to hinder the attachment of rotavirus viral particles to host cellular receptors [54].

8. Dengue Virus

Dengue is a significant mosquito-borne viral disease in tropical and subtropical regions. The responsible pathogen, dengue virus (DENV), consists of four distinctive serotypes: DENV-1, -2, -3, and -4. The infection can be mild or can result in clinically severe presentations, causing mild dengue fever, the more serious dengue hemorrhagic fever, or dengue shock syndrome [55]. A study in 2017 [56] reported the antiviral effect of bovine lactoferrin against DENV infection both in vivo and in vitro. Lactoferrin significantly inhibited the infection of the four serotypes of DENV and blocked binding between DENV-2 and the cellular membrane by interacting with heparan sulfate (HS), dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN), and low-density lipoprotein receptors (LDLR) [56].

References

  1. Hollmann, A.; Cardoso, N.P.; Espeche, J.C.; Maffía, P.C. Review of Antiviral Peptides for Use against Zoonotic and Selected Non-Zoonotic Viruses. Peptides 2021, 142, 170570.
  2. Gallagher, T.M.; Buchmeier, M.J. Coronavirus Spike Proteins in Viral Entry and Pathogenesis. Virology 2001, 279, 371–374.
  3. Fan, H.; Hong, B.; Luo, Y.; Peng, Q.; Wang, L.; Jin, X.; Chen, Y.; Hu, Y.; Shi, Y.; Li, T.; et al. The Effect of Whey Protein on Viral Infection and Replication of SARS-CoV-2 and Pangolin Coronavirus in Vitro. Signal Transduct. Target. Ther. 2020, 5, 275.
  4. Gallo, V.; Giansanti, F.; Arienzo, A.; Antonini, G. Antiviral Properties of Whey Proteins and Their Activity against SARS-CoV-2 Infection. J. Funct. Foods 2022, 89, 104932.
  5. Campione, E.; Lanna, C.; Cosio, T.; Rosa, L.; Conte, M.P.; Iacovelli, F.; Romeo, A.; Falconi, M.; Del Vecchio, C.; Franchin, E.; et al. Lactoferrin against SARS-CoV-2: In Vitro and In Silico Evidences. Front. Pharmacol 2021, 12, 666600.
  6. Miotto, M.; Di Rienzo, L.; Bò, L.; Boffi, A.; Ruocco, G.; Milanetti, E. Molecular Mechanisms behind Anti SARS-CoV-2 Action of Lactoferrin. Front. Mol. Biosci. 2021, 8, 607443.
  7. Naidu, S.A.G.; Clemens, R.A.; Pressman, P.; Zaigham, M.; Davies, K.J.A.; Naidu, A.S. COVID-19 during Pregnancy and Postpartum. J. Diet. Suppl. 2022, 19, 78–114.
  8. Lang, J.; Yang, N.; Deng, J.; Liu, K.; Yang, P.; Zhang, G.; Jiang, C. Inhibition of SARS Pseudovirus Cell Entry by Lactoferrin Binding to Heparan Sulfate Proteoglycans. PLoS ONE 2011, 6, e23710.
  9. Hu, Y.; Meng, X.; Zhang, F.; Xiang, Y.; Wang, J. The in Vitro Antiviral Activity of Lactoferrin against Common Human Coronaviruses and SARS-CoV-2 Is Mediated by Targeting the Heparan Sulfate Co-Receptor. Emerg. Microbes Infect. 2021, 10, 317–330.
  10. Madadlou, A. Food Proteins Are a Potential Resource for Mining Cathepsin L Inhibitory Drugs to Combat SARS-CoV-2. Eur. J. Pharmacol. 2020, 885, 173499.
  11. Salaris, C.; Scarpa, M.; Elli, M.; Bertolini, A.; Guglielmetti, S.; Pregliasco, F.; Blandizzi, C.; Brun, P.; Castagliuolo, I. Protective Effects of Lactoferrin against SARS-CoV-2 Infection In Vitro. Nutrients 2021, 13, 328.
  12. Mirabelli, C.; Wotring, J.W.; Zhang, C.J.; McCarty, S.M.; Fursmidt, R.; Pretto, C.D.; Qiao, Y.; Zhang, Y.; Frum, T.; Kadambi, N.S.; et al. Morphological Cell Profiling of SARS-CoV-2 Infection Identifies Drug Repurposing Candidates for COVID-19. Proc. Natl. Acad. Sci. USA 2021, 118, e2105815118.
  13. Cegolon, L.; Javanbakht, M.; Mastrangelo, G. Nasal Disinfection for the Prevention and Control of COVID-19: A Scoping Review on Potential Chemo-Preventive Agents. Int. J. Hyg. Environ. Health 2020, 230, 113605.
  14. Cegolon, L.; Mirandola, M.; Salaris, C.; Salvati, M.V.; Mastrangelo, G.; Salata, C. Hypothiocyanite and Hypothiocyanite/Lactoferrin Mixture Exhibit Virucidal Activity In Vitro against SARS-CoV-2. Pathogens 2021, 10, 233.
  15. Cegolon, L.; Mastrangelo, G. Hypothiocyanite for the Prevention and Control of COVID-19. Soc. Sci. Res. 2020.
  16. Pradeep, H.; Najma, U.; Aparna, H.S. Milk Peptides as Novel Multi-Targeted Therapeutic Candidates for SARS-CoV-2. Protein J. 2021, 40, 310–327.
  17. Çakır, B.; Okuyan, B.; Şener, G.; Tunali-Akbay, T. Investigation of Beta-Lactoglobulin Derived Bioactive Peptides against SARS-CoV-2 (COVID-19): In Silico Analysis. Eur. J. Pharmacol. 2021, 891, 173781.
  18. Berkhout, B.; Van Wamel, J.L.B.; Beljaars, L.; Meijer, D.K.F.; Visser, S.; Floris, R. Characterization of the Anti-HIV Effects of Native Lactoferrin and Other Milk Proteins and Protein-Derived Peptides. Antivir. Res. 2002, 55, 341–355.
  19. Saidi, H.; Eslaphazir, J.; Carbonneil, C.; Carthagena, L.; Requena, M.; Nassreddine, N.; Belec, L. Differential Modulation of Human Lactoferrin Activity against Both R5 and X4-HIV-1 Adsorption on Epithelial Cells and Dendritic Cells by Natural Antibodies. J. Immunol. 2006, 177, 5540–5549.
  20. Legrand, D.; Vigié, K.; Said, E.A.; Elass, E.; Masson, M.; Slomianny, M.C.; Carpentier, M.; Briand, J.P.; Mazurier, J.; Hovanessian, A.G. Surface Nucleolin Participates in Both the Binding and Endocytosis of Lactoferrin in Target Cells. Eur. J. Biochem. 2004, 271, 303–317.
  21. Carthagena, L. Modulation of HIV Binding to Epithelial Cells and HIV Transfer from Immature Dendritic Cells to CD4 T Lymphocytes by Human Lactoferrin and Its Major Exposed LF-33 Peptide. Virol. J. 2011, 5, 27–34.
  22. Groot, F.; Geijtenbeek, T.B.H.; Sanders, R.W.; Baldwin, C.E.; Sanchez-Hernandez, M.; Floris, R.; van Kooyk, Y.; de Jong, E.C.; Berkhout, B. Lactoferrin Prevents Dendritic Cell-Mediated Human Immunodeficiency Virus Type 1 Transmission by Blocking the DC-SIGN—Gp120 Interaction. Virol. J. 2005, 79, 3009–3015.
  23. Berkhout, B.; Floris, R.; Recio, I.; Visser, S. The Antiviral Activity of the Milk Protein Lactoferrin against the Human Immunodeficiency Virus Type 1. BioMetals 2004, 17, 291–294.
  24. Puddu, P.; Borghi, P.; Gessani, S.; Valenti, P.; Belardelli, F.; Seganti, L. Antiviral Effect of Bovine Lactoferrin Saturated with Metal Ions on Early Steps of Human Immunodeficiency Virus Type 1 Infection. Int. J. Biochem 1998, 30, 1055–1063.
  25. Neurath, A.R.; Debnath, A.K.; Strick, N.; Li, Y.; Lin, K.; Jiang, S. Blocking of CD4 Cell Receptors for the Human Immunodeficiency Virus Type 1 (HIV-1) by Chemically Modified Bovine Milk Proteins: Potential for AIDS Prophylaxis. J. Mol. Recognit. 1995, 8, 304–316.
  26. Neurath, A.R.; Jiang, S.; Strick, N.; Lin, K.; Li, Y.-Y.; Debnath, A.K. Bovine β–Lactoglobulin Modified by 3–Hydroxyphthalic Anhydride Blocks the CD4 Cell Receptor for HIV. Nat. Med. 1996, 2, 230–234.
  27. Mangan, R.J.; Stamper, L.; Ohashi, T.; Eudailey, J.A.; Go, E.P.; Jaeger, F.H.; Itell, H.L.; Watts, B.E.; Fouda, G.G.; Erickson, H.P.; et al. Determinants of Tenascin-C and HIV-1 Envelope Binding and Neutralization. Mucosal Immunol. 2019, 12, 1004–1012.
  28. Fouda, G.G.; Jaeger, F.H.; Amos, J.D.; Ho, C.; Kunz, E.L.; Anasti, K.; Stamper, L.W.; Liebl, B.E.; Barbas, K.H.; Ohashi, T.; et al. Tenascin-C Is an Innate Broad-Spectrum, HIV-1-Neutralizing Protein in Breast Milk. Proc. Natl. Acad. Sci. USA 2013, 110, 18220–18225.
  29. Griffiths, P.; Baraniak, I.; Reeves, M. The Pathogenesis of Human Cytomegalovirus. J. Pathol. 2015, 235, 288–297.
  30. Chobert, J.M.; Sitohy, M.; Billaudel, S.; Dalgalarrondo, M.; Haertlé, T. Anticytomegaloviral Activity of Esterified Milk Proteins and L-Polylysines. J. Mol. Microbiol. Biotechnol. 2007, 13, 255–258.
  31. Swart, P.J.; Kuipers, E.M.; Smit, C.; Van Der Strate, B.W.; Harmsen, M.C.; Meijer, D.K. Lactoferrin. Antiviral Activity of Lactoferrin. Adv. Exp. Med. Biol. 1998, 443, 205–213.
  32. Florisa, R.; Recio, I.; Berkhout, B.; Visser, S. Antibacterial and Antiviral Effects of Milk Proteins and Derivatives Thereof. Curr. Pharm. Des. 2003, 9, 1257–1275.
  33. Shimizu, K.; Matsuzawa, H.; Okada, K.; Tazume, S.; Dosako, S.; Kawasaki, Y.; Hashimoto, K.; Koga, Y. Lactoferrin-Mediated Protection of the Host from Murine Cytomegalovirus Infection by a T-Cell-Dependent Augmentation of Natural Killer Cell Activity. Arch. Virol. 1996, 141, 1875–1889.
  34. Andersen, J.H.; Osbakk, S.A.; Vorland, L.H.; Traavik, T.; Gutteberg, T.J. Lactoferrin and Cyclic Lactoferricin Inhibit the Entry of Human Cytomegalovirus into Human Fibroblasts. Antivir. Res. 2001, 51, 141–149.
  35. Beljaars, L.; Van Der Strate, B.W.A.; Bakker, H.I.; Reker-Smit, C.; Van Loenen-Weemaes, A.M.; Wiegmans, F.C.; Harmsen, M.C.; Molema, G.; Meijer, D.K.F. Inhibition of Cytomegalovirus Infection by Lactoferrin in Vitro and in Vivo. Antivir. Res. 2004, 63, 197–208.
  36. Van Der Strate, B.W.A.; De Boer, F.M.; Bakker, H.I.; Meijer, D.K.F.; Molema, G.; Harmsen, M.C. Synergy of Bovine Lactoferrin with the Anti-Cytomegalovirus Drug Cidofovir in Vitro. Antivir. Res. 2003, 58, 159–165.
  37. Hasegawa, K.; Motsuchi, W.; Tanaka, S.; Dosako1, S.-I. Inhibition with Lactoferrin of in Vitro Infection with Human Herpes Virus. Jpn. J. Med. Sci. Biol. 1994, 47, 73–85.
  38. Sitohy, M.; Billaudel, S.; Haertlé, T.; Chobert, J.M. Antiviral Activity of Esterified α-Lactalbumin and β-Lactoglobulin against Herpes Simplex Virus Type 1. Comparison with the Effect of Acyclovir and L-Polylysines. J. Agric. Food Chem. 2007, 55, 10214–10220.
  39. Hara, K.; Ikeda, M.; Saito, S.; Matsumoto, S.; Numata, K.; Kato, N.; Tanaka, K.; Sekihara, H. Lactoferrin Inhibits Hepatitis B Virus Infection in Cultured Human Hepatocytes. Hepatol. Res. 2002, 24, 228–235.
  40. Li, S.; Zhou, H.; Huang, G.; Liu, N. Inhibition of HBV Infection by Bovine Lactoferrin and Iron-, Zinc-Saturated Lactoferrin. Med. Microbiol. Immunol. 2009, 198, 19–25.
  41. Ammendolia, M.G.; Agamennone, M.; Pietrantoni, A.; Lannutti, F.; Siciliano, R.A.; de Giulio, B.; Amici, C.; Superti, F. Bovine Lactoferrin-Derived Peptides as Novel Broad-Spectrum Inhibitors of Influenza Virus. Pathog. Glob. Health 2012, 106, 12–19.
  42. May, J.T. Microbial Contaminants and Antimicrobial Properties of Human Milk. Microbiol. Sci. 1988, 5, 42–46.
  43. Pietrantoni, A.; Dofrelli, E.; Tinari, A.; Ammendolia, M.G.; Puzelli, S.; Fabiani, C.; Donatelli, I.; Superti, F. Bovine Lactoferrin Inhibits Influenza A Virus Induced Programmed Cell Death in Vitro. BioMetals 2010, 23, 465–475.
  44. Scala, M.C.; Sala, M.; Pietrantoni, A.; Spensiero, A.; Di Micco, S.; Agamennone, M.; Bertamino, A.; Novellino, E.; Bifulco, G.; Gomez-Monterrey, I.M.; et al. Lactoferrin-Derived Peptides Active towards Influenza: Identification of Three Potent Tetrapeptide Inhibitors. Sci. Rep. 2017, 7, 10593.
  45. Shin, K.; Wakabayashi, H.; Yamauchi, K.; Teraguchi, S.; Tamura, Y.; Kurokawa, M.; Shiraki, K. Effects of Orally Administered Bovine Lactoferrin and Lactoperoxidase on Influenza Virus Infection in Mice. J. Med. Microbiol. 2005, 54, 717–723.
  46. Pietrantoni, A.; Ammendolia, M.G.; Tinari, A.; Siciliano, R.; Valenti, P.; Superti, F. Bovine Lactoferrin Peptidic Fragments Involved in Inhibition of Echovirus 6 in Vitro Infection. Antivir. Res. 2006, 69, 98–106.
  47. Kawasaki, Y.; Isoda, H.; Shinmoto, H.; Tanimoto, M.; Dosako, S.; Idota, T.; Nakajima, I. Inhibition by κ-Casein Glycomacropeptide and Lactoferrin of Influenza Virus Hemagglutination. Biosci. Biotechnol. Biochem. 1993, 57, 1214–1215.
  48. Freitas, D.A.; Souza-Santos, R.; Carvalho, L.M.A.; Barros, W.B.; Neves, L.M.; Brasil, P.; Wakimoto, M.D. Congenital Zika Syndrome: A Systematic Review. PLoS ONE 2020, 15, e0242367.
  49. Francese, R.; Civra, A.; Donalisio, M.; Volpi, N.; Capitani, F.; Sottemano, S.; Tonetto, P.; Coscia, A.; Maiocco, G.; Moro, G.E.; et al. Anti-Zika Virus and Anti-Usutu Virus Activity of Human Milk and Its Components. PLoS Negl. Trop. Dis. 2020, 14, e0008713.
  50. Kanamaru, Y.; Etoh, M.; Song, X.-G.; Mikogami, T.; Hayasawa, H.; Ebina, T.; Minamoto, N. A High-Mr Glycoprotein Fraction from Cow’s Milk Potent in Inhibiting Replication of Human Rotavirus in Vitro. Biosci. Biotechnol. Biochem. 1999, 63, 246–249.
  51. Yolken, R.H.; Peterson, J.A.; Vonderfecht, S.L.; Fouts, E.T.; Midthun, K.; Newburg, D.S. Human Milk Mucin Inhibits Rotavirus Replication and Prevents Experimental Gastroenteritis. J. Clin. Investig. 1992, 90, 1984–1991.
  52. Newburg, D.S.; Peterson, J.A.; Ruiz-Palacios, G.M.; Matson, D.O.; Morrow, A.L.; Shults, J.; Guerrero, M.d.L.; Chaturvedi, P.; Newburg, S.O.; Scallan, C.D.; et al. Role of Human-Milk Lactadherin in Protectoin against Symptomatic Rotavirus Infection. Lancet 1998, 351, 1160–1164.
  53. Nousiainen, J.; Shingfield, K.J.; Huhtanen, P. Evaluation of Milk Urea Nitrogen as a Diagnostic of Protein Feeding. J. Dairy Sci. 2004, 87, 386–398.
  54. Superti, F.; Ammendolia, M.G.; Valenti, P.; Seganti, L. Antirotaviral Activity of Milk Proteins: Lactoferrin Prevents Rotavirus Infection in the Enterocyte-like Cell Line HT-29. Med. Microbiol. Immunol. 1997, 186, 83–91.
  55. Halstead, S.B. Antibody, Macrophages, Dengue Virus Infection, Shock, and Hemorrhage: A Pathogenetic Cascade. Clin. Infect. Dis. 1989, 11, S830–S839.
  56. Chen, J.-M.; Fan, Y.-C.; Lin, J.-W.; Chen, Y.-Y.; Hsu, W.-L.; Chiou, S.-S. Bovine Lactoferrin Inhibits Dengue Virus Infectivity by Interacting with Heparan Sulfate, Low-Density Lipoprotein Receptor, and DC-SIGN. Int. J. Mol. Sci 2017, 18, 1957.
More
Information
Subjects: Others
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 57
Revision: 1 time (View History)
Update Date: 20 Feb 2024
1000/1000