Skip to main content
Log in

Enhancing bioactive peptide release and identification using targeted enzymatic hydrolysis of milk proteins

  • Review
  • Published:
Analytical and Bioanalytical Chemistry Aims and scope Submit manuscript

Abstract

Milk proteins have been extensively studied for their ability to yield a range of bioactive peptides following enzymatic hydrolysis/digestion. However, many hurdles still exist regarding the widespread utilization of milk protein-derived bioactive peptides as health enhancing agents for humans. These mostly arise from the fact that most milk protein-derived bioactive peptides are not highly potent. In addition, they may be degraded during gastrointestinal digestion and/or have a low intestinal permeability. The targeted release of bioactive peptides during the enzymatic hydrolysis of milk proteins may allow the generation of particularly potent bioactive hydrolysates and peptides. Therefore, the development of milk protein hydrolysates capable of improving human health requires, in the first instance, optimized targeted release of specific bioactive peptides. The targeted hydrolysis of milk proteins has been aided by a range of in silico tools. These include peptide cutters and predictive modeling linking bioactivity to peptide structure [i.e., molecular docking, quantitative structure activity relationship (QSAR)], or hydrolysis parameters [design of experiments (DOE)]. Different targeted enzymatic release strategies employed during the generation of milk protein hydrolysates are reviewed herein and their limitations are outlined. In addition, specific examples are provided to demonstrate how in silico tools may help in the identification and discovery of potent milk protein-derived peptides. It is anticipated that the development of novel strategies employing a range of in silico tools may help in the generation of milk protein hydrolysates containing potent and bioavailable peptides, which in turn may be used to validate their health promoting effects in humans.

The targeted enzymatic hydrolysis of milk proteins may allow the generation of highly potent and bioavailable bioactive peptides.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Korhonen H, Pihlanto A (2006) Bioactive peptides: production and functionality. Int Dairy J 16:945–160

  2. Anusha R, Bindhu OS. Bioactive peptides from milk. In: Gigli I, Ed. Milk Proteins - From Structure to Biological Properties and Health Aspects. Rijeka: InTech. DOI: 10.5772/62993. Available from: https://www.intechopen.com/books/milk-proteins-from-structure-to-biological-properties-and-health-aspects/bioactive-peptides-from-milk; 2016.

  3. Nongonierma AB, O’Keeffe MB, RJ FG. Milk Protein Hydrolysates and Bioactive Peptides. Advanced Dairy Chemistry. New York: Springer; 2016. p. 417–82.

    Google Scholar 

  4. Nongonierma AB, FitzGerald RJ. Biofunctional properties of caseinophosphopeptides in the oral cavity. Caries Res. 2012;46:234–67.

    Article  CAS  PubMed  Google Scholar 

  5. Nongonierma AB, FitzGerald RJ. The scientific evidence for the role of milk protein-derived bioactive peptides in humans: A review. J Funct Foods. 2015;640:640–56.

    Article  CAS  Google Scholar 

  6. Nongonierma AB, FitzGerald RJ. Bioactive properties of milk proteins in humans: A review. Peptides. 2015;73:20–34.

    Article  CAS  PubMed  Google Scholar 

  7. Boutrou R, Henry G, Sanchez-Rivera L. On the trail of milk bioactive peptides in human and animal intestinal tracts during digestion: A review. Dairy Sci Technol. 2015:1–15.

  8. Egger L, Ménard O. Update on bioactive peptides after milk and cheese digestion. Curr Opin Food Sci. 2017;14:116–21.

    Article  Google Scholar 

  9. Kaiser S, Martin M, Lunow D, Rudolph S, Mertten S, Möckel U, et al. Tryptophan-containing dipeptides are bioavailable and inhibit plasma human angiotensin-converting enzyme in vivo. Int Dairy J. 2016;52:107–14.

    Article  CAS  Google Scholar 

  10. Foltz M, Meynen EE, Bianco V, van Platerink C, Koning TMMG, Kloek J. Angiotensin converting enzyme inhibitory peptides from a lactotripeptide-enriched milk beverage are absorbed intact into the circulation. J Nutr. 2007;137:953–8.

    Article  CAS  PubMed  Google Scholar 

  11. Caira S, Pinto G, Vitaglione P, Dal Piaz F, Ferranti P, Addeo F. Identification of casein peptides in plasma of subjects after a cheese-enriched diet. Food Res Int. 2016;84:108–12.

    Article  CAS  Google Scholar 

  12. Boutrou R, Gaudichon C, Dupont D, Jardin J, Airinei G, Marsset-Baglieri A, et al. Sequential release of milk protein-derived bioactive peptides in the jejunum in healthy humans. Am J Clin Nutr. 2013;97:1314–23.

    Article  CAS  PubMed  Google Scholar 

  13. Pripp AH. Initial proteolysis of milk proteins and its effect on formation of ACE-inhibitory peptides during gastrointestinal proteolysis: a bioinformatic, in silico, approach. Eur Food Res Technol. 2005;221:712–6.

    Article  CAS  Google Scholar 

  14. Udenigwe CC. Bioinformatics approaches, prospects and challenges of food bioactive peptide research. Trends Food Sci Technol. 2014;36:137–43.

    Article  CAS  Google Scholar 

  15. Nongonierma AB, FitzGerald RJ. Strategies for the discovery, identification and validation of milk protein-derived bioactive peptides. Trends Food Sci Technol. 2016;50:26–43.

    Article  CAS  Google Scholar 

  16. Li-Chan EC. Bioactive peptides and protein hydrolysates: Research trends and challenges for application as nutraceuticals and functional food ingredients. Curr Opin Food Sci. 2015;1:28–37.

    Article  Google Scholar 

  17. Contreras MM. Hernández-Ledesma B, Amigo L, Martín-Álvarez PJ, Recio I. Production of antioxidant hydrolyzates from a whey protein concentrate with thermolysin: Optimization by response surface methodology. LWT Food Sci Technol. 2011;44:9–15.

    Article  CAS  Google Scholar 

  18. van der Ven C, Gruppen H, de Bont DBA, Voragen AGJ. Optimization of the angiotensin converting enzyme inhibition by whey protein hydrolysates using response surface methodology. Int Dairy J. 2002;12:813–20.

    Article  Google Scholar 

  19. Iwaniak A, Minkiewicz P, Darewicz M, Protasiewicz M, Mogut D. Chemometrics and cheminformatics in the analysis of biologically active peptides from food sources. J Funct Foods. 2015;16:334–51.

    Article  CAS  Google Scholar 

  20. Udenigwe CC, Gong M, Wu S. In silico analysis of the large and small subunits of cereal RuBisCO as precursors of cryptic bioactive peptides. Process Biochem. 2013;48:1794–9.

    Article  CAS  Google Scholar 

  21. Udenigwe CC. Towards rice bran protein utilization: In silico insight on the role of oryzacystatins in biologically-active peptide production. Food Chem. 2016;191:135–8.

    Article  CAS  PubMed  Google Scholar 

  22. Wang T-Y, Hsieh C-H, Hung C-C, Jao C-L, Lin P-Y, Hsieh Y-L, et al. A study to evaluate the potential of an in silico approach for predicting dipeptidyl peptidase-IV inhibitory activity in vitro of protein hydrolysates. Food Chem. 2017;234:431–8.

    Article  CAS  PubMed  Google Scholar 

  23. Lacroix IME, Li-Chan ECY. Evaluation of the potential of dietary proteins as precursors of dipeptidyl peptidase (DPP)-IV inhibitors by an in silico approach. J Funct Foods. 2012;4:403–22.

    Article  CAS  Google Scholar 

  24. Minkiewicz P, Dziuba J, Michalska J. Bovine meat proteins as potential precursors of biologically active peptides - a computational study based on the BIOPEP database. Food Sci Technol Int. 2011;17:39–45.

    Article  CAS  PubMed  Google Scholar 

  25. Vercruysse L, Van Camp J, Smagghe G. ACE inhibitory peptides derived from enzymatic hydrolysates of animal muscle protein: a review. J Agric Food Chem. 2005;53:8106–15.

    Article  CAS  PubMed  Google Scholar 

  26. Gu Y, Majumder K, Wu J. QSAR-aided in silico approach in evaluation of food proteins as precursors of ACE inhibitory peptides. Food Res Int. 2011;44:2465–74.

    Article  CAS  Google Scholar 

  27. Fu Y, Wu W, Zhu M, Xiao Z. In silico assessment of the potential of patatin as a precursor of bioactive peptides. J Food Biochem. 2016;40:366–70.

    Article  CAS  Google Scholar 

  28. Rajendran SRCK, Mason B, Udenigwe CC. Peptidomics of peptic digest of selected potato tuber proteins: Post-translational modifications and limited cleavage specificity. J Agric Food Chem. 2016;64:2432–7.

    Article  CAS  Google Scholar 

  29. Nongonierma AB, FitzGerald RJ. Strategies for the discovery and identification of food protein-derived biologically active peptides. Trends Food Sci Technol. 2017;69:289-305.

  30. Capriotti AL, Cavaliere C, Piovesana S, Samperi R, Laganà A. Recent trends in the analysis of bioactive peptides in milk and dairy products. Anal Bioanal Chem. 2016;408:2677–85.

    Article  CAS  PubMed  Google Scholar 

  31. El-Salam MA, El-Shibiny S. Bioactive peptides of buffalo, camel, goat, sheep, mare, and yak milks and milk products. Food Rev Int. 2013;29:1–23.

    Article  CAS  Google Scholar 

  32. Mati A, Senoussi-Ghezali C, Zennia SSA, Almi-Sebbane D, El-Hatmi H, Girardet J-M. Dromedary camel milk proteins, a source of peptides having biological activities–A review. Int Dairy J. 2017;73:25–37.

    Article  CAS  Google Scholar 

  33. Vincenzetti S, Pucciarelli S, Polzonetti V, Polidori P. Role of proteins and of some bioactive peptides on the nutritional quality of donkey milk and their impact on human health. Beverages. 2017;3:34.

    Article  Google Scholar 

  34. Agarwal S, Beausire RLW, Patel S, Patel H. Innovative uses of milk protein concentrates in product development. J Food Sci. 2015;80:A23–A9.

    Article  CAS  PubMed  Google Scholar 

  35. de la Fuente MA, Hemar Y, Tamehana M, Munro PA, Singh H. Process-induced changes in whey proteins during the manufacture of whey protein concentrates. Int Dairy J. 2002;12:361–9.

    Article  Google Scholar 

  36. Le Maux S, Nongonierma AB, FitzGerald RJ. Peptide composition and dipeptidyl peptidase IV inhibitory properties of β-lactoglobulin hydrolysates having similar extents of hydrolysis while generated using different enzyme-to-substrate ratios. Food Res Int. 2017;99:84–90.

    Article  CAS  PubMed  Google Scholar 

  37. Nongonierma AB, FitzGerald RJ. Tryptophan-containing milk protein-derived dipeptides inhibit xanthine oxidase. Peptides. 2012;37:263–72.

    Article  CAS  PubMed  Google Scholar 

  38. Kalyankar P, Zhu Y, O’Keeffe M, O’Cuinn G, RJ FG. Substrate specificity of glutamyl endopeptidase (GE): Hydrolysis studies with a bovine α-casein preparation. Food Chem. 2013;136:501–12.

    Article  CAS  PubMed  Google Scholar 

  39. Lacroix IM, Li-Chan ECY. Inhibition of dipeptidyl peptidase (DPP)-IV and α-glucosidase activities by pepsin-treated whey proteins. J Agric Food Chem. 2013;61:7500–6.

    Article  CAS  PubMed  Google Scholar 

  40. Fan F, Tu M, Liu M, Shi P, Wang Y, Wu D, et al. Isolation and characterization of lactoferrin peptides with stimulatory effect on osteoblast proliferation. J Agric Food Chem. 2017;65:7179–85.

    Article  CAS  PubMed  Google Scholar 

  41. Cheison SC, Schmitt M, Leeb E, Letzel T, Kulozik U. Influence of temperature and degree of hydrolysis on the peptide composition of trypsin hydrolysates of β-lactoglobulin: Analysis by LC–ESI-TOF/MS. Food Chem. 2010;121:457–67.

    Article  CAS  Google Scholar 

  42. Nongonierma AB, FitzGerald RJ. Enzymes exogenous to milk in dairy technology | Proteinases. In: Fuquay JW, editor. Encyclopedia of Dairy Sciences. 2nd ed. San Diego: Academic Press; 2011. p. 289–96.

    Chapter  Google Scholar 

  43. Tavano OL. Protein hydrolysis using proteases: an important tool for food biotechnology. J Mol Catal B Enzym. 2013;90:1–11.

    Article  CAS  Google Scholar 

  44. Stepaniak L. Dairy enzymology. Int J Dairy Technol. 2004;57:153–71.

    Article  CAS  Google Scholar 

  45. Sanchón J, Fernández-Tomé S, Miralles B, Hernández-Ledesma B, Tomé D, Gaudichon C, et al. Protein degradation and peptide release from milk proteins in human jejunum. Comparison with in vitro gastrointestinal simulation. Food Chem. 2018;239:486–94.

    Article  CAS  PubMed  Google Scholar 

  46. Lemes AC, Sala L, Ores JDC, ARC B, Egea MB, Fernandes KF. A review of the latest advances in encrypted bioactive peptides from protein-rich waste. Int J Mol Sci. 2016;17:950.

    Article  CAS  PubMed Central  Google Scholar 

  47. Toldrá F, Reig M, Aristoy MC, Mora L. Generation of bioactive peptides during food processing. Food Chem. 2017.

  48. Mullally MM, O'Callaghan DM, FitzGerald RJ, Donnelly W, Dalton JP. Proteolytic and peptidolytic activities in commercial pancreatic protease preparations and their relationship to some whey protein hydrolyzate characteristics. J Agric Food Chem. 1994;42:2973–81.

    Article  CAS  Google Scholar 

  49. Spellman D, Kenny P, O'Cuinn G, FitzGerald RJ. Aggregation properties of whey protein hydrolysates generated with Bacillus licheniformis proteinase activities. J Agric Food Chem. 2005;53:1258–65.

    Article  CAS  PubMed  Google Scholar 

  50. Kilcawley KN, Wilkinson MG, Fox PF. Determination of key enzyme activities in commercial peptidase and lipase preparations from microbial or animal sources. Enzym Microb Technol. 2002;31:310–20.

    Article  CAS  Google Scholar 

  51. Merz M, Eisele T, Berends P, Appel D, Rabe S, Blank I, et al. Flavourzyme, an enzyme preparation with industrial relevance: Automated nine-step purification and partial characterization of eight enzymes. J Agric Food Chem. 2015;63:5682–93.

    Article  CAS  PubMed  Google Scholar 

  52. Merz M, Appel D, Berends P, Rabe S, Blank I, Stressler T, et al. Batch-to-batch variation and storage stability of the commercial peptidase preparation Flavourzyme in respect of key enzyme activities and its influence on process reproducibility. Eur Food Res Technol. 2016;242:1005–12.

    Article  CAS  Google Scholar 

  53. Butré CI, Sforza S, Gruppen H, Wierenga PA. Determination of the influence of substrate concentration on enzyme selectivity using whey protein isolate and Bacillus licheniformis protease. J Agric Food Chem. 2014;62:10230–9.

    Article  CAS  PubMed  Google Scholar 

  54. Butré CI, Sforza S, Wierenga PA, Gruppen H. Determination of the influence of the pH of hydrolysis on enzyme selectivity of Bacillus licheniformis protease towards whey protein isolate. Int Dairy J. 2015;44:44–53.

    Article  CAS  Google Scholar 

  55. NCBI. https://www.ncbi.nlm.nih.gov/protein/. Accessed 19 Sept 2017.

  56. UniProt. www.uniprot.org/. Accessed 19 Sept 2017.

  57. Dziuba J, Minkiewicz P, Nałecz D, Iwaniak A. Database of biologically active peptide sequences. Food/Nahrung. 1999;43:190–5.

    Article  CAS  PubMed  Google Scholar 

  58. BIOPEP. http://www.uwm.edu.pl/biochemia/index.php/en/biopep. Accessed 18 Sept 2017.

  59. MBPDB. http://mbpdb.nws.oregonstate.edu. Accessed 19 Sept 2017.

  60. Nielsen SD, Beverly RL, Qu Y, Dallas DC. Milk bioactive peptide database: A comprehensive database of milk protein-derived bioactive peptides and novel visualization. Food Chem. 2017;232:673–82.

    Article  CAS  PubMed  Google Scholar 

  61. Vermeirssen V, van der Bent A, Van Camp J, van Amerongen A, Verstraete W. A quantitative in silico analysis calculates the angiotensin I converting enzyme (ACE) inhibitory activity in pea and whey protein digests. Biochimie. 2004;86:231–9.

    Article  CAS  PubMed  Google Scholar 

  62. Dziuba M, Dziuba B, Iwaniak A. Milk proteins as precursors of bioactive peptides. Acta Sci Pol Technol Aliment. 2009;8:71–90.

    CAS  Google Scholar 

  63. Nongonierma AB, FitzGerald RJ. An in silico model to predict the potential of dietary proteins as sources of dipeptidyl peptidase IV (DPP-IV) inhibitory peptides. Food Chem. 2014;165:489–98.

    Article  CAS  PubMed  Google Scholar 

  64. Dziuba J, Iwaniak A, Minkiewicz P. Computer-aided characteristics of proteins as potential precursors of bioactive peptides. Polimery. 2003;48:50–3.

    CAS  Google Scholar 

  65. Tulipano G, Sibilia V, Caroli AM, Cocchi D. Whey proteins as source of dipeptidyl dipeptidase IV (dipeptidyl peptidase-4) inhibitors. Peptides. 2011;32:835–8.

    Article  CAS  PubMed  Google Scholar 

  66. Dziuba B, Dziuba M. New milk protein-derived peptides with potential antimicrobial activity: An approach based on bioinformatic studies. Int J Mol Sci. 2014;15:14531–45.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Nongonierma AB, FitzGerald RJ. Dipeptidyl peptidase IV inhibitory and antioxidative properties of milk-derived dipeptides and hydrolysates. Peptides. 2013;39:157–63.

    Article  CAS  PubMed  Google Scholar 

  68. Nongonierma AB, FitzGerald RJ. Inhibition of dipeptidyl peptidase IV (DPP-IV) by proline containing peptides. J Funct Foods. 2013;5:1909–17.

    Article  CAS  Google Scholar 

  69. ExPASy. http://web.expasy.org/peptide_cutter/peptidecutter_enzymes.html#Peps. Accessed 18 Sept 2017.

  70. MEROPS. https://www.ebi.ac.uk/merops/. Accessed 18 Sept 2017.

  71. O’Loughlin IB, Murray BA, Kelly PM, FitzGerald RJ, Brodkorb A. Enzymatic hydrolysis of heat-induced aggregates of whey protein isolate. J Agric Food Chem. 2012;60:4895–904.

    Article  CAS  PubMed  Google Scholar 

  72. Vorob’ev MM, Butré CI, Sforza S, Wierenga PA, Gruppen H. Demasking kinetics of peptide bond cleavage for whey protein isolate hydrolyzed by Bacillus licheniformis protease. J Mol Catal B: Enzym. 2017.

  73. Panchaud A, Affolter M, Kussmann M. Mass spectrometry for nutritional peptidomics: How to analyze food bioactives and their health effects. J Proteome. 2012;75:3546–59.

    Article  CAS  Google Scholar 

  74. Sánchez-Rivera L, Martínez-Maqueda D, Cruz-Huerta E, Miralles B, Recio I. Peptidomics for discovery, bioavailability and monitoring of dairy bioactive peptides. Food Res Int. 2014;63(Part B):170–81.

    Article  CAS  Google Scholar 

  75. Nongonierma AB, Le Maux S, Hamayon J, FitzGerald RJ. Strategies for the release of dipeptidyl peptidase IV (DPP-IV) inhibitory peptides in an enzymatic hydrolyzate of α-lactalbumin. Food Funct. 2016;7:3437–43.

    Article  CAS  PubMed  Google Scholar 

  76. Kalyankar P, Zhu Y, O’Cuinn G, FitzGerald RJ. Investigation of the substrate specificity of glutamyl endopeptidase using purified bovine β-casein and synthetic peptides. J Agric Food Chem. 2013;61:3193–204.

    Article  CAS  PubMed  Google Scholar 

  77. Norris R, Poyarkov A, O’Keeffe MB, FitzGerald RJ. Characterization of the hydrolytic specificity of Aspergillus niger derived prolyl endoproteinase on bovine β-casein and determination of ACE inhibitory activity. Food Chem. 2014;156:29–36.

    Article  CAS  PubMed  Google Scholar 

  78. Norris R, O’Keeffe MB, Poyarkov A, FitzGerald RJ. Peptide identification and angiotensin converting enzyme (ACE) inhibitory activity in prolyl endoproteinase digests of bovine αs-casein. Food Chem. 2015;188:210–7.

    Article  CAS  PubMed  Google Scholar 

  79. Tulipano G, Faggi L, Nardone A, Cocchi D, Caroli AM. Characterization of the potential of β-lactoglobulin and α-lactalbumin as sources of bioactive peptides affecting incretin function: in silico and in vitro comparative studies. Int Dairy J. 2015;48:66–72.

    Article  CAS  Google Scholar 

  80. Guinane CM, Kent RM, Norberg S, O'Connor PM, Cotter PD, Hill C, et al. Generation of the antimicrobial peptide caseicin A from casein by hydrolysis with thermolysin enzymes. Int Dairy J. 2015;49:1–7.

    Article  CAS  Google Scholar 

  81. Zhang Y, Chen R, Zuo F, Ma H, Zhang Y, Chen S. Comparison of dipeptidyl peptidase IV-inhibitory activity of peptides from bovine and caprine milk casein by in silico and in vitro analyses. Int Dairy J. 2016;53:37–44.

    Article  CAS  Google Scholar 

  82. Nongonierma AB, Paolella S, Mudgil P, Maqsood S, FitzGerald RJ. Dipeptidyl peptidase IV (DPP-IV) inhibitory properties of camel milk protein hydrolysates generated with trypsin. J Funct Foods. 2017;34:49–58.

    Article  CAS  Google Scholar 

  83. Nongonierma AB, FitzGerald RJ. Susceptibility of milk protein-derived peptides to dipeptidyl peptidase IV (DPP-IV) hydrolysis. Food Chem. 2014;145:845–52.

    Article  CAS  PubMed  Google Scholar 

  84. Butré CI, Buhler S, Sforza S, Gruppen H, Wierenga PA. Spontaneous, non-enzymatic breakdown of peptides during enzymatic protein hydrolysis. Biochim Biophysica Acta (BBA) - Proteins Proteomics. 2015;1854:987–94.

    Article  CAS  Google Scholar 

  85. Pina AS, Roque ACA. Studies on the molecular recognition between bioactive peptides and angiotensin-converting enzyme. J Mol Recognit. 2009;22:162–8.

    Article  CAS  PubMed  Google Scholar 

  86. Norris R, Casey F, FitzGerald RJ, Shields DC, Mooney C. Predictive modeling of angiotensin converting enzyme inhibitory dipeptides. Food Chem. 2012;133:1349–54.

    Article  CAS  Google Scholar 

  87. Pripp AH. Docking and virtual screening of ACE inhibitory dipeptides. Eur Food Res Technol. 2007;225:589–92.

    Article  CAS  Google Scholar 

  88. Nongonierma AB, Mooney C, Shields DC, FitzGerald RJ. Inhibition of dipeptidyl peptidase IV and xanthine oxidase by amino acids and dipeptides. Food Chem. 2013;141:644–53.

    Article  CAS  PubMed  Google Scholar 

  89. Vukic VR, Vukic DV, Milanovic SD, Ilicic MD, Kanuric KG, Johnson MS. In silico identification of milk antihypertensive di- and tripeptides involved in angiotensin I–converting enzyme inhibitory activity. Nutr Res. 2017;46:22–30.

    Article  CAS  PubMed  Google Scholar 

  90. Nongonierma AB, Mooney C, Shields DC, FitzGerald RJ. In silico approaches to predict the potential of milk protein-derived peptides as dipeptidyl peptidase IV (DPP-IV) inhibitors. Peptides. 2014;57:43–51.

    Article  CAS  PubMed  Google Scholar 

  91. Fernández-Tomé S, Martínez-Maqueda D, Girón R, Goicoechea C, Miralles B, Recio I. Novel peptides derived from αs1-casein with opioid activity and mucin stimulatory effect on HT29-MTX cells. J Funct Foods. 2016;25:466–76.

    Article  CAS  Google Scholar 

  92. Nongonierma AB, FitzGerald RJ. Learnings from quantitative structure-activity relationship (QSAR) studies with respect to food protein-derived bioactive peptides: A review. RSC Adv. 2016;6:75400–13.

    Article  CAS  Google Scholar 

  93. Udenigwe CC, Li H, Aluko RE. Quantitative structure–activity relationship modeling of renin-inhibiting dipeptides. Amino Acids. 2011;42:1379–86.

    Article  CAS  PubMed  Google Scholar 

  94. Wu J, Aluko RE. Quantitative structure-activity relationship study of bitter di- and tri-peptides including relationship with angiotensin I-converting enzyme inhibitory activity. J Pept Sci. 2007;13:63–9.

    Article  CAS  PubMed  Google Scholar 

  95. Wu J, Aluko RE, Nakai S. Structural requirements of angiotensin I-converting enzyme inhibitory peptides: Quantitative structure-activity relationship study of di-and tripeptides. J Agric Food Chem. 2006;54:732–8.

    Article  CAS  PubMed  Google Scholar 

  96. Wang J-H, Liu Y-L, Ning J-H, Yu J, Li X-H, Wang F-X. Is the structural diversity of tripeptides sufficient for developing functional food additives with satisfactory multiple bioactivities? J Mol Struct. 2013;1040:164–70.

    Article  CAS  Google Scholar 

  97. Pripp AH, Isaksson T, Stepaniak L, Sørhaug T. Quantitative structure-activity relationship modeling of ACE-inhibitory peptides derived from milk proteins. Eur Food Res Technol. 2004;219:579–83.

    Article  CAS  Google Scholar 

  98. Nongonierma AB, FitzGerald RJ. Structure activity relationship modeling of milk protein-derived peptides with dipeptidyl peptidase IV (DPP-IV) inhibitory activity. Peptides. 2016;79:1–7.

    Article  CAS  PubMed  Google Scholar 

  99. Maruyama S, Miyoshi S, Kaneko T, Tanaka H. Angiotensin I-converting enzyme inhibitory activities of synthetic peptides related to the tandem repeated sequence of a maize endosperm protein. Agric Biol Chem. 1989;53:1077–81.

    CAS  Google Scholar 

  100. Mooney C, Haslam NJ, Pollastri G, Shields DC. Towards the improved discovery and design of functional peptides: common features of diverse classes permit generalized prediction of bioactivity. PLoS One. 2012;7:e45012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Zenezini Chiozzi R, Capriotti AL, Cavaliere C, La Barbera G, Piovesana S, Samperi R, et al. Purification and identification of endogenous antioxidant and ACE-inhibitory peptides from donkey milk by multidimensional liquid chromatography and nanoHPLC-high resolution mass spectrometry. Anal Bioanal Chem. 2016;408:5657–66.

    Article  CAS  PubMed  Google Scholar 

  102. Rani S, Pooja K, Pal GK. Exploration of potential angiotensin converting enzyme inhibitory peptides generated from enzymatic hydrolysis of goat milk proteins. Biocatalysis Agric Biotechnol. 2017;11:83–8.

    Google Scholar 

  103. Nongonierma AB, Le Maux S, Esteveny C, FitzGerald RJ. Response surface methodology (RSM) applied to the generation of casein hydrolysates with antioxidant and dipeptidyl peptidase IV (DPP-IV) inhibitory properties. J Sci Food Agric. 2017;97:1093–101.

    Article  CAS  PubMed  Google Scholar 

  104. Naik L, Mann B, Bajaj R, Sangwan R, Sharma R. Process optimization for the production of bio-functional whey protein hydrolysates: adopting response surface methodology. Int J Peptide Res Therapeut. 2013;19:231–7.

    Article  CAS  Google Scholar 

  105. Zhao XH, Wu D, Li TJ. Preparation and radical scavenging activity of papain-catalyzed casein plasteins. Dairy Sci Technol. 2010;90:521–35.

    Article  CAS  Google Scholar 

  106. Zheng H, Shen X, Bu G, Luo Y. Effects of pH, temperature and enzyme-to-substrate ratio on the antigenicity of whey protein hydrolysates prepared by Alcalase. Int Dairy J. 2008;18:1028–33.

    Article  CAS  Google Scholar 

  107. Liu X, Luo Y, Li Z. Effects of pH, temperature, enzyme-to-substrate ratio and reaction time on the antigenicity of casein hydrolysates prepared by papain. Food Agric Immunol. 2012;23:69–82.

    Article  CAS  Google Scholar 

  108. Guo Y, Pan D, Tanokura M. Optimization of hydrolysis conditions for the production of the angiotensin-I converting enzyme (ACE) inhibitory peptides from whey protein using response surface methodology. Food Chem. 2009;114:328–33.

    Article  CAS  Google Scholar 

  109. Tavares TG, Contreras MM, Amorim M, Martín-Álvarez PJ, Pintado ME, Recio I, et al. Optimization, by response surface methodology, of degree of hydrolysis and antioxidant and ACE-inhibitory activities of whey protein hydrolysates obtained with cardoon extract. Int Dairy J. 2011;21:926–33.

    Article  CAS  Google Scholar 

  110. Vermeirssen V, Van Camp J, Devos L, Verstraete W. Release of angiotensin I converting enzyme (ACE) inhibitory activity during in vitro gastrointestinal digestion: from batch experiment to semicontinuous model. J Agric Food Chem. 2003;51:5680–7.

    Article  CAS  PubMed  Google Scholar 

  111. Pan D, Guo Y. Optimization of sour milk fermentation for the production of ACE-inhibitory peptides and purification of a novel peptide from whey protein hydrolysate. Int Dairy J. 2010;20:472–9.

    Article  CAS  Google Scholar 

  112. Zhao X-H, Li Y-Y. An approach to improve ACE-inhibitory activity of casein hydrolysates with plastein reaction catalyzed by Alcalase. Eur Food Res Technol. 2009;229:795–805.

    Article  CAS  Google Scholar 

  113. Uluko H, Li H, Cui W, Zhang S, Liu L, Chen J, et al. Response surface optimization of angiotensin converting enzyme inhibition of milk protein concentrate hydrolysates in vitro after ultrasound pretreatment. Innovative Food Sci Emerg Technol. 2013;20:133–9.

    Article  CAS  Google Scholar 

  114. Quirós A, Hernández-Ledesma B, Ramos M, Martín-Álvarez PJ, Recio I. Short communication: Production of antihypertensive peptide HLPLP by enzymatic hydrolysis: Optimization by response surface methodology. J Dairy Sci. 2012;95:4280–5.

    Article  CAS  PubMed  Google Scholar 

  115. Nongonierma AB, Lalmahomed M, Paolella S, FitzGerald RJ. Milk protein isolate (MPI) as a source of dipeptidyl peptidase IV (DPP-IV) inhibitory peptides. Food Chem. 2017;231:202–11.

    Article  CAS  PubMed  Google Scholar 

  116. Nongonierma AB, Mazzocchi C, Paolella S, FitzGerald RJ. Release of dipeptidyl peptidase IV (DPP-IV) inhibitory peptides from milk protein isolate (MPI) during enzymatic hydrolysis. Food Res Int. 2017;94:79–89.

    Article  CAS  PubMed  Google Scholar 

  117. Asledottir T, Le TT, Petrat-Melin B, Devold TG, Larsen LB, Vegarud GE. Identification of bioactive peptides and quantification of β-casomorphin-7 from bovine β-casein A1, A2 and I after ex vivo gastrointestinal digestion. Int Dairy J. 2017;71:98–106.

    Article  CAS  Google Scholar 

  118. Cunsolo V, Muccilli V, Saletti R, Foti S. Applications of mass spectrometry techniques in the investigation of milk proteome. Eur J Mass Spectrom. 2011;17:305–20.

    Article  CAS  Google Scholar 

  119. Le TT, Deeth HC, Larsen LB. Proteomics of major bovine milk proteins: Novel insights. Int Dairy J. 2017;67:2–15.

    Article  CAS  Google Scholar 

  120. Giacometti J, Buretić-Tomljanović A. Peptidomics as a tool for characterizing bioactive milk peptides. Food Chem. 2017;230:91–8.

    Article  CAS  PubMed  Google Scholar 

  121. Nongonierma AB, Gaudel C, Murray BA, Flynn S, Kelly PM, Newsholme P, et al. Insulinotropic properties of whey protein hydrolysates and impact of peptide fractionation on insulinotropic response. Int Dairy J. 2013;32:163–8.

    Article  CAS  Google Scholar 

  122. Lahrichi SL, Affolter M, Zolezzi IS, Panchaud A. Food Peptidomics: Large scale analysis of small bioactive peptides - A pilot study. J Proteome. 2013;88:83–91.

    Article  CAS  Google Scholar 

  123. Morifuji M, Koga J, Kawanaka K, Higuchi M. Branched-chain amino acid-containing dipeptides, identified from whey protein hydrolysates, stimulate glucose uptake rate in L6 myotubes and isolated skeletal muscles. J Nutr Sci Vitaminol. 2009;55:81–6.

    Article  CAS  PubMed  Google Scholar 

  124. Dallas DC, Guerrero A, Parker EA, Robinson RC, Gan J, German JB, et al. Current peptidomics: Applications, purification, identification, quantification, and functional analysis. Proteomics. 2015;15:1026–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Le Maux S, Nongonierma AB, FitzGerald RJ. Improved short peptide identification using HILIC-MS/MS: Retention time prediction model based on the impact of amino acid position in the peptide sequence. Food Chem. 2015;175:847–54.

    Article  CAS  Google Scholar 

  126. Harscoat-Schiavo C, Nioi C, Ronat-Heit E, Paris C, Vanderesse R, Fournier F, et al. Hydrophilic properties as a new contribution for computer-aided identification of short peptides in complex mixtures. Anal Bioanal Chem. 2012;403:1939–49.

    Article  CAS  PubMed  Google Scholar 

  127. Le Maux S, Nongonierma AB, Murray B, Kelly PM, FitzGerald RJ. Identification of short peptide sequences in the nanofiltration permeate of a bioactive whey protein hydrolysate. Food Res Int. 2015;77:534–9.

    Article  CAS  Google Scholar 

  128. Sagardia I, Iloro I, Elortza F, Bald C. Quantitative structure–activity relationship based screening of bioactive peptides identified in ripened cheese. Int Dairy J. 2013;33:184–90.

    Article  CAS  Google Scholar 

  129. Nongonierma AB, Paolella S, Mudgil P, Maqsood S, Fitzgerald RJ. Identification of novel dipeptidyl peptidase IV (DPP-IV) inhibitory peptides in camel milk protein hydrolysates. Food Chem. 2017;Submitted.

  130. Murray BA, Walsh DJ, FitzGerald RJ. Modification of the furanacryloyl-L-phenylalanylglycylglycine assay for determination of angiotensin-I-converting enzyme inhibitory activity. J Biochem Biophys Methods. 2004;59:127–37.

    Article  CAS  PubMed  Google Scholar 

  131. Shan L, Marti T, Sollid LM, Khosla C. Comparative biochemical analysis of three bacterial prolyl endopeptidases: Implications for coeliac sprue. Biochem J. 2004;383:311–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Lacroix IM, Li-Chan EC. Comparison of the susceptibility of porcine and human dipeptidyl-peptidase IV to inhibition by protein-derived peptides. Peptides. 2015;69:19–25.

    Article  CAS  PubMed  Google Scholar 

  133. Devabhaktuni A, Elias JE. Application of de novo sequencing to large-scale complex proteomics data sets. J Proteome Res. 2016;15:732–42.

    Article  CAS  PubMed  Google Scholar 

  134. Tagliazucchi D, Shamsia S, Helal A, Conte A. Angiotensin-converting enzyme inhibitory peptides from goats' milk released by in vitro gastro-intestinal digestion. Int Dairy J. 2017;71:6–16.

    Article  CAS  Google Scholar 

  135. Zhu Y-s. Kalyankar P, FitzGerald RJ. Relative quantitation analysis of the substrate specificity of glutamyl endopeptidase with bovine α-caseins. Food Chem. 2015;167:463–7.

    Article  CAS  PubMed  Google Scholar 

  136. Rocha-Martin J, Fernández-Lorente G, Guisan JM. Sequential hydrolysis of commercial casein hydrolysate by immobilized trypsin and thermolysin to produce bioactive phosphopeptides. Biocatal Biotransform. 2017:1–16.

  137. Cheung LK, Aluko RE, Cliff MA, Li-Chan EC. Effects of exopeptidase treatment on antihypertensive activity and taste attributes of enzymatic whey protein hydrolysates. J Funct Foods. 2015;13:262–75.

    Article  CAS  Google Scholar 

  138. Barry C, O'Cuinn G, Harrington D, O'Callaghan D, Fitzgerald R. Debittering of a tryptic digest of bovine b-casein using porcine kidney general aminopeptidase and X-prolydipeptidyl aminopeptidase from Lactococcus lactis subsp. cremoris AM2. J Food Sci. 2000;65:1145–50.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The work described herein was supported by Enterprise Ireland under grant number TC2013-0001. The authors thank Dr. Sara Paolella for her insightful discussions on peptide bond selectivity during enzymatic hydrolysis.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Richard J. FitzGerald.

Ethics declarations

Conflicts of interests

The authors declare that they have no conflict of interest.

Additional information

Published in the topical collection Discovery of Bioactive Compounds with guest editors Aldo Laganà, Anna Laura Capriotti, and Chiara Cavaliere.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nongonierma, A.B., FitzGerald, R.J. Enhancing bioactive peptide release and identification using targeted enzymatic hydrolysis of milk proteins. Anal Bioanal Chem 410, 3407–3423 (2018). https://doi.org/10.1007/s00216-017-0793-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00216-017-0793-9

Keywords

Navigation