α-Lactalbumin: Comparison
Please note this is a comparison between Version 2 by Rita Xu and Version 1 by Eugene Permyakov.

α-Lactalbumin (α-LA) is a small (Mr 14,200), acidic (pI 4-5), Ca2+-binding protein. α-LA is a regulatory component of lactose synthase enzyme system. α-LA is very important in infant nutrition since it constitutes a large part of the whey and total protein in human milk. The protein possesses a single strong Ca2+-binding site, which can also bind Mg2+, Mn2+, Na+, K+, and some other metal cations. It contains several distinct Zn2+-binding sites. Physical properties of α-LA strongly depend on the occupation of its metal binding sites by metal ions. In the absence of bound metal ions α-LA is in the molten globule-like state. The binding of metal ions, and especially of Ca2+, increases stability of α-LA against action of heat, various denaturing agents, and proteases, while the binding of Zn2+ to the Ca2+-loaded protein decreases its stability and causes its aggregation. The thermal unfolding of apo-α-LA takes place in the temperature region from 10 to 30 °C. The binding of Ca2+ under the conditions of low ionic strength shifts the thermal transition to higher temperatures by more than 40 °C. The binding of Mg2+, Na+, and K+ increases protein stability as well. The stronger an ion binds to the protein, the more pronounced the thermal transition shift. All four classes of surfactants (anionic, cationic, non-ionic, and zwitterionic) denature α-LA and the denaturation involves at least one intermediate. The position of any denaturation transition in α-LA (half-transition temperature, half-transition pressure, half-transition denaturant concentration) depends upon metal ion concentration in solution (especially if this metal ion is Ca2+). Therefore, values of denaturation temperature or urea or guanidine hydrochloride denaturing concentration are relatively meaningless for α-LA without specifying the metal ion content(s) and their solution concentration(s). At a neutral or slightly acidic pH at a physiological temperature, α-LA can associate with membranes. The conformations of the membrane-bound protein range from native-like to molten globule-like states. At a low pH, α-LA penetrates the interior of the negatively charged membranes and exhibits a molten globule conformation. Depending on external conditions, α-LA can form amyloid fibrils, amorphous aggregates, nanoparticles, and nanotubes. At pH 2, α-LA in the classical molten globule conformation can form amyloid fibrils. Some of these aggregated states of α-LA (nanoparticles, nanotubes) can be used in practical applications such as drug delivery to tissues and organs. The structure and self-assembly behavior of α-LA are governed by a subtle balance between hydrophobic and polar interactions and this balance can be finely tuned through the addition of selected substances. Small size nanoparticles of α-LA (100 to 200 nm) can be obtained with the use of various desolvating agents. Partially hydrolyzed α-LA can form nanotubes. α-LA and some of its fragments possess bactericidal and antiviral activities. Complexes of partially unfolded α-LA with oleic acid showed significant cytotoxicity to various tumor and bacterial cells. α-LA in such complexes plays a role of a delivery carrier of cytotoxic fatty acid molecules into tumor cells across the cell membrane. Cytotoxic protein–oleic acid complexes possess a common core-shell structure, where an oily core is made of a micellar oleic acid, whereas a proteinaceous shell, which stabilizes the oleic acid micelle, is formed from the flexible, partially unfolded proteins. These complexes called liprotides (lipids and partially denatured proteins), which are potential novel anti-tumorous drugs, can be considered as molten globular containers filled with the toxic oil.

.

α-Lactalbumin (α-LA) is a small (Mr 14,200), acidic (pI 4-5), Ca2+-binding protein. α-LA is a regulatory component of lactose synthase enzyme system. α-LA is very important in infant nutrition since it constitutes a large part of the whey and total protein in human milk. The protein possesses a single strong Ca2+-binding site, which can also bind Mg2+, Mn2+, Na+, K+, and some other metal cations. It contains several distinct Zn2+-binding sites. Physical properties of α-LA strongly depend on the occupation of its metal binding sites by metal ions. In the absence of bound metal ions α-LA is in the molten globule-like state. The binding of metal ions, and especially of Ca2+, increases stability of α-LA against action of heat, various denaturing agents, and proteases, while the binding of Zn2+ to the Ca2+-loaded protein decreases its stability and causes its aggregation. The thermal unfolding of apo-α-LA takes place in the temperature region from 10 to 30 °C. The binding of Ca2+ under the conditions of low ionic strength shifts the thermal transition to higher temperatures by more than 40 °C. The binding of Mg2+, Na+, and K+ increases protein stability as well. The stronger an ion binds to the protein, the more pronounced the thermal transition shift. All four classes of surfactants (anionic, cationic, non-ionic, and zwitterionic) denature α-LA and the denaturation involves at least one intermediate. The position of any denaturation transition in α-LA (half-transition temperature, half-transition pressure, half-transition denaturant concentration) depends upon metal ion concentration in solution (especially if this metal ion is Ca2+). Therefore, values of denaturation temperature or urea or guanidine hydrochloride denaturing concentration are relatively meaningless for α-LA without specifying the metal ion content(s) and their solution concentration(s). At a neutral or slightly acidic pH at a physiological temperature, α-LA can associate with membranes. The conformations of the membrane-bound protein range from native-like to molten globule-like states. At a low pH, α-LA penetrates the interior of the negatively charged membranes and exhibits a molten globule conformation. Depending on external conditions, α-LA can form amyloid fibrils, amorphous aggregates, nanoparticles, and nanotubes. At pH 2, α-LA in the classical molten globule conformation can form amyloid fibrils. Some of these aggregated states of α-LA (nanoparticles, nanotubes) can be used in practical applications such as drug delivery to tissues and organs. The structure and self-assembly behavior of α-LA are governed by a subtle balance between hydrophobic and polar interactions and this balance can be finely tuned through the addition of selected substances. Small size nanoparticles of α-LA (100 to 200 nm) can be obtained with the use of various desolvating agents. Partially hydrolyzed α-LA can form nanotubes. α-LA and some of its fragments possess bactericidal and antiviral activities. Complexes of partially unfolded α-LA with oleic acid showed significant cytotoxicity to various tumor and bacterial cells. α-LA in such complexes plays a role of a delivery carrier of cytotoxic fatty acid molecules into tumor cells across the cell membrane. Cytotoxic protein–oleic acid complexes possess a common core-shell structure, where an oily core is made of a micellar oleic acid, whereas a proteinaceous shell, which stabilizes the oleic acid micelle, is formed from the flexible, partially unfolded proteins. These complexes called liprotides (lipids and partially denatured proteins), which are potential novel anti-tumorous drugs, can be considered as molten globular containers filled with the toxic oil.

  • alpha-lactalbumin
  • structure
  • calcium binding
  • folding
  • molten globule
  • cytotoxicity
Please wait, diff process is still running!

References

  1. Jackson, J.; Janszen, D.; Lönnerdal, B.; Lien, E.; Pramuk, K.; Kuhlman, C. A multinational study of α-lactalbumin concentrations in human milk. J. Nutr. Biochem. 2004, 15, 517–521.
  2. Renner, E. Milk and dairy products in human nutrition. In Milk and Dairy Products in Human Nutrition; John Wiley & Sons: Hoboken, NJ, USA, 1983.
  3. Montagne, P.; Cuilliere, M.L.; Mole, C.; Bene, M.C.; Faure, G. Immunological and nutritional composition of human milk in relation to prematurity and mother’s parity during the first 2 weeks of lactation. J. Pediatric Gastroenterol. Nutr. 1999, 29, 75–80.
  4. Hill, R.L.; Brew, K. Lactose synthetase. Adv. Enzymol. Relat. Areas Mol. Biol. 1975, 43, 411–490.
  5. Hiraoka, Y.; Segawa, T.; Kuwajima, K.; Sugai, S.; Murai, N. α-Lactalbumin—A calcium metalloprotein. Biochem. Biophys. Res. Commun. 1980, 95, 1098–1104.
  6. Permyakov, E.A.; Yarmolenko, V.V.; Kalinichenko, L.P.; Morozova, L.A.; Burstein, E.A. Calcium binding to α-lactalbumin: Structural rearrangement and association constant evaluation by means of intrinsic protein fluorescence changes. Biochem. Biophys. Res. Commun. 1981, 100, 191–197.
  7. Dolgikh, D.A.; Gilmanshin, R.I.; Brazhnikov, E.V.; Bychkova, V.E.; Semisotnov, G.V.; Venyaminov, S.Y.; Ptitsyn, O.B. α-Lactalbumin—Compact state with fluctuating tertiary structure. FEBS Lett. 1981, 136, 311–315.
  8. Kuwajima, K. The molten globule state of α-lactalbumin. FASEB J. 1996, 10, 102–109.
  9. Hakansson, A.; Zhivotovsky, B.; Orrenius, S.; Sabharwal, H.; Svanborg, C. Apoptosis Induced by a human-milk protein. Proc. Natl. Acad. Sci. USA 1995, 92, 8064–8068.
  10. Pellegrini, A.; Thomas, U.; Bramaz, N.; Hunziker, P.; von Fellenberg, R. Isolation and identification of three bactericidal domains in the bovine α-lactalbumin molecule. Biochim. Biophys. Acta 1999, 1426, 439–448.
  11. Permyakov, E.A.; Berliner, L.J. α-Lactalbumin: Structure and function. FEBS Lett. 2000, 473, 269–274.
  12. Permyakov, E.A. α-Lactalbumin; Nova Science Publishers: New York, NY, USA, 2005.
  13. Permyakov, E.A. Metalloproteomics; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2009.
  14. Permyakov, E.A.; Kretsinger, R.H. Calcium Binding Proteins; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2011.
  15. Permyakov, E.A.; Permyakov, S.E.; Breydo, L.; Redwan, E.M.; Almehdar, H.A.; Uversky, V.N. Disorder in milk proteins: α-lactalbumin. Part A. Structural properties and conformational behavior. Curr. Protein Pept. Sci. 2016, 17, 352–367.
  16. Permyakov, E.A.; Permyakov, S.E.; Breydo, L.; Redwan, E.M.; Almehdar, H.A.; Uversky, V.N. Disorder in milk proteins: α-lactalbumin. Part C. Pecularities of metal binding. Curr. Protein Pept. Sci. 2016, 17, 735–745.
  17. Uversky, V.N.; Permyakov, S.E.; Breydo, L.; Redwan, E.M.; Almehdar, H.A.; Permyakov, E.A. Disorder in milk proteins: α-lactalbumin. Part B. A multifunctional whey protein acting as an oligomeric molten globule “oil container” in the anti-tumorogenic drugs, liprotides. Curr. Protein Pept. Sci. 2016, 17, 612–628.
  18. Dautigny, A.; Prager, E.M.; Phamdinh, D.; Jolles, J.; Pakdel, F.; Grinde, B.; Jolles, P. cDNA and amino-acid-sequences of rainbow-trout (Oncorhynchus-Mykiss) lysozymes and their implications for the evolution of lysozyme and lactalbumin. J. Mol. Evol. 1991, 32, 187–198.
  19. Grobler, J.A.; Rao, K.R.; Pervaiz, S.; Brew, K. Sequences of two highly divergent canine type c lysozymes: Implications for the evolutionary origins of the lysozyme/α-lactalbumin superfamily. Arch. Biochem. Biophys. 1994, 313, 360–366.
  20. Nitta, K.; Sugai, S. The Evolution of lysozyme and α-lactalbumin. Eur. J. Biochem. 1989, 182, 111–118.
  21. Prager, E.M.; Wilson, A.C. Ancient origin of lactalbumin from lysozyme—Analysis of DNA and amino acid sequences. J. Mol. Evol. 1988, 27, 326–335.
  22. Threadgill, D.W.; Womack, J.E. Genomic analysis of the major bovine-milk protein genes. Nucleic Acids Res. 1990, 18, 6935–6942.
  23. Gallagher, D.S.; Threadgill, D.W.; Ryan, A.M.; Womack, J.E.; Irwin, D.M. Physical mapping of the lysozyme gene family in cattle. Mamm. Genome 1993, 4, 368–373.
  24. Davies, M.S.; West, L.F.; Davis, M.B.; Povey, S.; Craig, R.K. The gene for human α-lactalbumin is assigned to chromosome-12q13. Ann. Hum. Genet. 1987, 51, 183–188.
  25. Xue, Y.M.; Liu, J.N.; Sun, Z.Y.; Ma, Z.; Wu, C.L.; Zhu, D.X. α-Lactalbumin mutant acting as lysozyme. Proteins 2001, 42, 17–22.
  26. Chang, J.Y.; Li, L. Pathway of oxidative folding of α-lactalbumin: A model for illustrating the diversity of disulfide folding pathways. Biochemistry 2002, 41, 8405–8413.
  27. Barman, T.E. Purification and properties of bovine milk glycol-α-lactalbumin. Biochim. Biophys. Acta 1970, 214, 242–244.
  28. Brown, R.C.; Fish, W.W.; Hudson, B.G.; Ebner, K.E. Isolation and characterization of rat α-lactalbumin—Glycoprotein. Biochim. Biophys. Acta 1977, 491, 82–92.
  29. Pike, A.C.W.; Brew, K.; Acharya, K.R. Crystal structures of guinea-pig, goat and bovine α-lactalbumin highlight the enhanced conformational flexibility of regions that are significant for its action in lactose synthase. Structure 1996, 4, 691–703.
  30. Horii, K.; Saito, M.; Yoda, T.; Tsumoto, K.; Matsushima, M.; Kuwajima, K.; Kumagai, I. Contribution of Thr29 to the thermodynamic stability of goat α-lactalbumin as determined by experimental and theoretical approaches. Proteins 2001, 45, 16–29.
  31. Calderone, V.; Giuffrida, M.G.; Viterbo, D.; Napolitano, L.; Fortunato, D.; Conti, A.; Acharya, K.R. Amino acid sequence and crystal structure of buffalo α-lactalbumin. FEBS Lett. 1996, 394, 91–95.
  32. Acharya, K.R.; Ren, J.S.; Stuart, D.I.; Phillips, D.C.; Fenna, R.E. Crystal structure of human α-lactalbumin at 1.7 Å resolution. J. Mol. Biol. 1991, 221, 571–581.
  33. Acharya, K.R.; Stuart, D.I.; Walker, N.P.; Lewis, M.; Phillips, D.C. Refined structure of baboon α-lactalbumin at 1.7 Å resolution. Comparison with C-type lysozyme. J. Mol. Biol. 1989, 208, 99–127.
  34. Dolgikh, D.A.; Abaturov, L.V.; Bolotina, I.A.; Brazhnikov, E.V.; Bychkova, V.E.; Gilmanshin, R.I.; Lebedev, Y.O.; Semisotnov, G.V.; Tiktopulo, E.I.; Ptitsyn, O.B. Compact state of a protein molecule with pronounced small-scale mobility—Bovine α-lactalbumin. Eur. Biophys. J. 1985, 13, 109–121.
  35. Kataoka, M.; Kuwajima, K.; Tokunaga, F.; Goto, Y. Structural characterization of the molten globule of α-lactalbumin by solution X-ray scattering. Protein Sci. 1997, 6, 422–430.
  36. Polverino de Laureto, P.; Frare, E.; Gottardo, R.; Fontana, A. Molten globule of bovine α-lactalbumin at neutral pH induced by heat, trifluoroethanol, and oleic acid: A comparative analysis by circular dichroism spectroscopy and limited proteolysis. Proteins 2002, 49, 385–397.
  37. Permyakov, E.A.; Morozova, L.A.; Burstein, E.A. Cation binding effects on the pH, thermal and urea denaturation transitions in α-lactalbumin. Biophys. Chem. 1985, 21, 21–31.
  38. Rosner, H.I.; Redfield, C. The human α-lactalbumin molten globule: Comparison of structural preferences at pH 2 and pH 7. J. Mol. Biol. 2009, 394, 351–362.
  39. Bai, P.; Luo, L.; Peng, Z.Y. Side chain accessibility and dynamics in the molten globule state of α-lactalbumin: A F-19-NMR study. Biochemistry 2000, 39, 372–380.
  40. Redfield, C.; Schulman, B.A.; Milhollen, M.A.; Kim, P.S.; Dobson, C.M. α-Lactalbumin forms a compact molten globule in the absence of disulfide bonds. Nat. Struct. Biol. 1999, 6, 948–952.
  41. Permyakov, S.E.; Permyakov, E.A.; The use of the free metal—Temperature ‘phase diagrams’ for studies of single site metal binding proteins. Protein J 2007, 26, 1-12.
  42. Permyakov, S.E.; Khokhlova, T.I.; Uversky, V.N.; Permyakov, E.A. Analysis of Ca2+/Mg2+ selectivity in α-lactalbumin and Ca2+-binding lysozyme reveals a distinct Mg2+-specific site in lysozyme. Proteins 2010, 78, 2609–2624.
  43. Permyakov, A.; Ostrovsky, A.V.; Kalinichenko, L.P. Stopped flow kinetic studies of Ca(II) and Mg(II) dissociation in cod parvalbumin and bovine α-lactalbumin. Biophys. Chem. 1987, 28, 225–233.
  44. Murakami, ; Berliner, L.J. A distinct zinc binding site in the α-lactalbumins regulates calcium binding. Is there a physiological role for this control? Biochemistry 1983, 22, 3370–3374.
  45. Permyakov, E.A.; Shnyrov, V.L.; Kalinichenko, L.P.; Kuchar, A.; Reyzer, I.L.; Berliner, L.J. Binding of Zn(Ii) ions to α-lactalbumin. J. Protein Chem. 1991, 10, 577–584.
  46. Ren, J.S.; Stuart, D.I.; Acharya, K.R. α-Lactalbumin possesses a distinct zinc-binding site. J. Biol. Chem. 1993, 268, 19292–19298.
  47. Permyakov, S.E.; Veprintsev, D.B.; Brooks, C.L.; Permyakov, E.A.; Berliner, L.J. Zinc binding in bovine α-lactalbumin: Sequence homology may not be a predictor of subtle functional features. Proteins 2000, 40, 106–111.
  48. Saeki, K.; Arai, M.; Yoda, T.; Nakao, M.; Kuwajima, K. Localized nature of the transition-state structure in goat α-lactalbumin folding. J. Mol. Biol. 2004, 341, 589–604.
  49. Arai, M.; Ito, K.; Inobe, T.; Nakao, M.; Maki, K.; Kamagata, K.; Kihara, H.; Amemiya, Y.; Kuwajima, K. Fast compaction of α-lactalbumin during folding studied by stopped-flow X-ray scattering. J. Mol. Biol. 2002, 321, 121–132.
  50. Bushmarina, N.A.; Blanchet, C.E.; Vernier, G.; Forge, V. Cofactor effects on the protein folding reaction: Acceleration of α-lactalbumin refolding by metal ions. Protein Sci. 2006, 15, 659–671.
  51. Bu, Z.M.; Cook, J.; Callaway, D.J.E. Dynamic regimes and correlated structural dynamics in native and denatured α-lactalbumin. J. Mol. Biol. 2001, 312, 865–873.
  52. Otzen, E.; Sehgal, P.; Westh, P. α-Lactalbumin is unfolded by all classes of surfactants but by different mechanisms. J. Colloid Interface Sci. 2009, 329, 273–283.
  53. Singh, ; Hassan, M.I.; Islam, A.; Ahmad, F. Cooperative unfolding of residual structure in heat denatured proteins by urea and guanidinum chloride. PLoS ONE 2015, 10, e0128740.
  54. Goers, J.; Permyakov, S.E.; Permyakov, E.A.; Uversky, V.N.; Fink, A.L. Conformational prerequisites for α-lactalbumin fibrillation. Biochemistry 2002, 41, 12546–12551.
  55. Polverino de Laureto, P.; Frare, E.; Battaglia, F.; Mossuto, M.F.; Uversky, V.N.; Fontana, A. Protein dissection enhances the amyloidogenic properties of α-lactalbumin. FEBS J. 2005, 272, 2176–2188.
  56. Buccarielli, S.; Sayedi, E.S.; Osella, S.; Trzaskowsli, B.; Vissing, K.J.; Vestergaard, B.; Fodera, V. Disentangling the role of solvent polarity and protein solvation in folding and self-assembly of α-lactalbumin. J. Colloid Interface Sci. 2020, 561, 749–761.
  57. Arroyo-Maya, I.J.; Rodiles-López, J.O.; Cornejo-Mazón, M.; Gutiérrez-López, G.F.; Hernández-Arana, A.; Toledo-Núñez, C.; Barbosa-Cánovas, G.V.; Flores-Flores, J.O.; Hernández-Sánchez, H.J. Effect of different treatments on the ability of α-lactalbumin to form nanoparticles. Dairy Sci. 2012, 95, 6204–6214.
  58. Zhang, B.; Luo, Y.; Wang, Q. Development of silver/α-lactalbumin nano-composites: A new approach to reduce silver toxicity. Int. J. Antimicrob. Agents 2011, 38, 502–509.
  59. Ipsen, R.; Otte, J.; Qvist, K.B. Molecular self-assembly of partially hydrolysed α-lactalbumin resulting in strong gels with a novel microstructure. J. Dairy Res. 2001, 68, 277–286.
  60. Geng, X.L.; Kain Kirkensgaard, J.J.; Arleth, L.; Otte, J.; Ipsen. R. The influence of pH, protein concentration and calcium ratio on the formation and structure of nanotubes from partially hydrolyzed bovine α-lactalbumin. Soft Matter 2019, 15, 4787–4796.
  61. Permyakov, E.A.; Kreimer, D.I. Effects of pH, Temperature and Ca2+ content on the conformation of α-lactalbumin in a medium modeling physiological conditions. Gen. Physiol. Biophys. 1986, 5, 377–390.
  62. Vanderheeren, G.; Hanssens, I.; Noyelle, K.; Van Dael, H.; Joniau, M. The perturbations of the native state of goat α-lactalbumin induced by 1,1’-bis(4-anilino-5-naphthalenesulfonate) are Ca2+-dependent. Biophys. J. 1998, 75, 2195–2204.
  63. Singh, S.K.; Kishore, N. Elucidating the binding thermodynamics of 8-anilino-1-naphthalene sulfonic acid with the A-state of α-lactalbumin: An isothermal titration calorimetric investigation. Biopolymers 2006, 83, 205–212.
  64. Permyakov, E.A.; Grishchenko, V.M.; Kalinichenko, L.P.; Orlov, N.Y.; Kuwajima, K.; Sugai, S. Calcium-regulated interactions of human α-lactalbumin with bee venom melittin. Biophys. Chem. 1991, 39, 111–117.
  65. Cawthern, K.M.; Permyakov, E.; Berliner, L.J. Membrane-bound states of α-lactalbumin: Implications for protein stability and conformation. Protein Sci. 1996, 5, 1394–1405.
  66. Cawthern, K.M.; Narayan, M.; Chaudhuri, D.; Permyakov, E.A.; Berliner, L.J. Interactions of α-lactalbumin with fatty acids and spin label analogs. J. Biol. Chem. 1997, 272, 30812–30816.
  67. Nigen, M.; Croguennec, T.; Renard, D.; Bouhallab, S. Temperature affects the supramolecular structures resulting from α-lactalbumin-lysozyme interaction. Biochemistry 2007, 46, 1248–1255.
  68. Nigen, M.; Gaillard, C.; Croguennec, T.; Madec, N.; Bouhallab, S. Dynamic and supramolecular organisation of α-lactalbumin/lysozyme microspheres: A microscopic study. Biophys. Chem. 2010, 146, 30–35.
  69. Berliner, L.J.; Koga, K. α-Lactalbumin binding to membranes—Evidence for a partially buried protein. Biochemistry 1987, 26, 3006–3009.
  70. Permyakov, A.; Kreimer, D.I.; Kalinichenko, L.P.; Shnyrov, V.L. Effects of calcium and magnesium-ions on the interactions of calcium-binding proteins parvalbumin and α-lactalbumin with dipalmitoylphosphatidylcholine vesicles. Biofizika 1988, 33, 465–470.
  71. Permyakov, A.; Kreimer, D.I.; Kalinichenko, L.P.; Shnyrov, V.L. Interactions of calcium-binding proteins, parvalbumin and α-lactalbumin, with dipalmitoyl phosphatidyl choline vesicles. Gen. Physiol. Biophys. 1988, 7, 95–107.
  72. Grishchenko, M.; Kalinichenko, L.P.; Deikus, G.Y.; Veprintsev, D.B.; Cawthern, K.M.; Berliner, L.J.; Permyakov, E.A. Interactions of α-lactalbumins with lipid vesicles studied by tryptophan fluorescence. Biochem. Mol. Biol. Int. 1996, 38, 453–466.
  73. Bañuelos, S.; Muga, A. Binding of molten globule-like conformations to lipid bilayers—Structure of native and partially folded α-lactalbumin bound to model membranes. J. Biol. Chem. 1995, 270, 29910–29915.
  74. Bañuelos, ; Muga, A. Structural requirements for the association of native and partially folded conformations of α-lactalbumin with model membranes. Biochemistry 1996, 35, 3892–3898.
  75. Bañuelos, S.; Muga, A. Interaction of native and partially folded conformations of α-lactalbumin with lipid bilayers: Characterization of two membrane-bound states. FEBS Lett. 1996, 386, 21–25.
  76. Rødland, I.; Halskau, Ø.; Martínez, ; Holmsen, H. α-Lactalbumin binding and membrane integrity—Effect of charge and degree of unsaturation of glycerophospholipids. Biochim. Biophys. Acta 2005, 1717, 11–20.
  77. Pellegrini, Antimicrobial peptides from food proteins. Curr. Pharm. Des. 2003, 9, 1225–1238.
  78. Sipola, M.; Finckenberg, P.; Vapaatalo, H.; Pihlanto-Leppala, A.; Korhonen, H.; Korpela, R.; Nurminen, M.L. α-Lactorphin and β-lactorphin improve arterial function in spontaneously hypertensive rats. Life Sci. 2002, 71, 1245–1253.
  79. Kanda, Y.; Hisayasu, S.; Abe, Y.; Katsura, K.; Mashimo, K. Growth-active peptides are produced from α-lactalbumin and lysozyme. Life Sci. 2007, 81, 449–457.
  80. Tavares, T.; Contreras, M.D.; Amorim, M.; Pintado, M.; Recio, I.; Malcata, F.X. Novel whey-derived peptides with inhibitory effect against angiotensin-converting enzyme: In vitro effect and stability to gastrointestinal enzymes. Peptides 2011, 32, 1013–1019.
  81. Hakansson, A.; Svensson, M.; Mossberg, A.K.; Sabharwal, H.; Linse, S.; Lazou, I.; Lonnerdal, S.; Svanborg, C. A folding variant of α-lactalbumin with bactericidal activity against Streptococcus pneumoniae. Mol. Microbiol. 2000, 35, 589–600.
  82. Matsumoto, H.; Shimokawa, Y.; Ushida, Y.; Toida, T.; Hayasawa, H. New biological function of bovine α-lactalbumin: Protective effect against ethanol- and stress-induced gastric mucosal injury in rats. Biosci. Biotechnol. Biochem. 2001, 65, 1104–1111.
  83. Mezzaroba, L.F.H.; Carvalho, J.E.; Ponezi, A.N.; Antônio, M.A.; Monteiro, K.M.; Possenti, A.; Sgarbieri, V.C. Antiulcerative properties of bovine α-lactalbumin. Int. Dairy J. 2006, 16, 1005–1012.
  84. Sachdeva, A; Rawat, S.; Nagpal, J. Efficacy of fermented milk and whey proteins in Helicobacter pylori eradication: A review. World J. Gastroenterol. 2014, 20, 724–737.
  85. Sitohy, M.; Chobert, J.M.; Karwowska, U.; Gozdzicka-Jozefiak, A.; Haertle, T. Inhibition of bacteriophage m13 replication with esterified milk proteins. J. Agric. Food Chem. 2006, 54, 3800–3806.
  86. Yarramala, D.S.; Prakash, P.; Ranade, D.S.; Doshi, S.; Kulkarni, P.P.; Bhaumik, P.; Rao, C.P. Cytotoxicity of apo bovine α-lactalbumin complexed with La3+ on cancer cells supported by its high resolution crystal structure. Sci. Rep. 2019, 9, 1780.
  87. Hakansson, A.; Andreasson, J.; Zhivotovsky, B.; Karpman, D.; Orrenius, S.; Svanborg, C. Multimeric α-lactalbumin from human milk induces apoptosis through a direct effect on cell nuclei. Exp. Cell Res. 1999, 246, 451–460.
  88. Svensson, M.; Sabharwal, H.; Hakansson, A.; Mossberg, A.K.; Lipniunas, P.; Leffler, H.; Svanborg, C.; Linse, S. Molecular characterization of α-lactalbumin folding variants that induce apoptosis in tumor cells. J. Biol. Chem. 1999, 274, 6388–6396.
  89. Kohler, C.; Hakansson, A.; Svanborg, C.; Orrenius, S.; Zhivotovsky, B. Protease activation in apoptosis induced by MAL. Exp. Cell Res. 1999, 249, 260–268.
  90. Svensson, M.; Hakansson, A.; Mossberg, A.K.; Linse, S.; Svanborg, C. Conversion of α-lactalbumin to a protein inducing apoptosis. Proc. Natl. Acad. Sci. USA 2000, 97, 4221–4226.
  91. Svanborg, C.; Agerstam, H.; Aronson, A.; Bjerkvig, R.; Duringer, C.; Fischer, W.; Gustafsson, L.; Hallgren, O.; Leijonhuvud, I.; Linse, S.; et al. HAMLET kills tumor cells by an apoptosis-like mechanism—Cellular, molecular, and therapeutic aspects. Adv. Cancer Res. 2003, 88, 1–29.
  92. Gustafsson, L; Hallgren, O.; Mossberg, A.K.; Pettersson, J.; Fischer, W.; Aronsson, A.; Svanborg, C. HAMLET kills tumor cells by apoptosis: Structure, cellular mechanisms, and therapy. J. Nutr. 2005, 135, 1299–1303.
  93. Svensson, M.; Mossberg, A.K.; Pettersson, J.; Linse, S.; Svanborg, C. Lipids as cofactors in protein folding: Stereo-specific lipidprotein interactions are required to form HAMLET human α-lactalbumin made lethal to tumor cells. Protein Sci. 2003, 12, 2805–2814.
  94. Knyazeva, E.L.; Grishchenko, V.M.; Fadeev, R.S.; Akatov, V.S.; Permyakov, S.E.; Permyakov, E.A. Who is Mr. HAMLET? Interaction of human α-lactalbumin with monomeric oleic acid. Biochemistry 2008, 47, 13127–13137.
  95. Duringer, C; Hamiche, A.; Gustafsson, L.; Kimura, H.; Svanborg, C. HAMLET interacts with histones and chromatin in tumor cell nuclei. J. Biol. Chem. 2003, 278, 42131–42135.
  96. Brest, P; Gustafsson, M.; Mossberg, A.K.; Gustafsson, L.; Duringer, C.; Hamiche, A.; Svanborg, C. Histone deacetylase inhibitors promote the tumoricidal effect of HAMLET. Cancer Res. 2007, 67, 11327–11334.
  97. Mercer, N.; Ramakrishnan, B.; Boeggeman, E.; Qasba, P.K. Applications of site-specific labeling to study HAMLET, a tumoricidal complex of α-lactalbumin and oleic acid. PLoS ONE 2011, 6, e26093.
  98. Permyakov, E.; Pershikova, I.V.; Khokhlova, T.I.; Uversky, V.N.; Permyakov, E.A. No need to be HAMLET or BAMLET to interact with histories: Binding of monomeric α-lactalbumin to histories and basic poly-amino acids. Biochemistry 2004, 43, 5575–5582.
  99. Permyakov, S.E.; Pershikova, I.V.; Zhadan, A.P.; Goers, J.; Bakunts, A.G.; Uversky, V.N.; Berliner, L.J.; Permyakov, E.A. Conversion of human α-lactalbumin to an apo-like state in the complexes with basic poly-amino acids: Toward understanding of the molecular mechanism of antitumor action of HAMLET. J. Proteome Res. 2005, 4, 564–569.
  100. Zherelova, O.M.; Kataev, A.A.; Grishchenko, V.M.; Knyazeva, E.L.; Permyakov, S.E.; Permyakov, E.A. Interaction of antitumor α-lactalbumin-oleic acid complexes with artificial and natural membranes. J. Bioenerg. Biomembr. 2009, 41, 229–237.
  101. Hallgren, O.; Gustafsson, L.; Irjala, H.; Selivanova, G.; Orrenius, S.; Svanborg, C. HAMLET triggers apoptosis but tumor cell death is independent of caspases, Bcl-2 and p53. Apoptosis 2006, 11, 221–233.
  102. Hakansson, P.; Roche-Hakansson, H.; Mossberg, A.K.; Svanborg, C. Apoptosis-like death in bacteria induced by HAMLET, a human milk lipid-protein complex. PLoS ONE 2011, 6, e17717.
  103. Permyakov, S.E.; Knyazeva, E.L.; Khasanova, L.M.; Fadeev, R.S.; Zhadan, A.P.; Roche-Hakansson, H.; Hakansson, A.P.; Akatov, V.S.; Permyakov, E.A. Oleic acid is a key cytotoxic component of HAMLET-like complexes. Biol. Chem. 2012, 393, 85–92.
  104. Nakamura, T.; Aizawa, T.; Kariya, R.; Okada, S.; Demura, M.; Kawano, K.; Makabe, K.; Kuwajima, K. Molecular mechanisms of the cytotoxicity of human α-lactalbumin made lethal to tumor cells (HAMLET) and other protein-oleic acid complexes. J. Biol. Chem. 2013, 288, 14408–14416.
  105. Fontana, A.; Spolaore, B.; Polverino de Laureto, P. The biological activities of protein/oleic acid complexes reside in the fatty acid. Biochim. Biophys. Acta 2013, 1834, 1125–1143.
  106. Brinkmann, R.; Thiel, S.; Otzen, D.E. Protein-fatty acid complexes: Biochemistry, biophysics and function. FEBS J. 2013, 280, 1733–1749.
  107. Hoque, M.; Dave, S.; Gupta, P.; Saleemuddin, M. Oleic acid may be the key contributor in the BAMLET-induced erythrocyte hemolysis and tumoricidal action. PLoS ONE 2013, 8, e68390.
  108. Rath, E.M.; Duff, A.P.; Håkansson, A.P.; Vacher, C.S.; Liu, G.J.; Knott, R.B.; Church, W.B. Structure and potential cellular targets of HAMLET-like anti-cancer compounds made from milk components. J. Pharm. Pharm. Sci. 2015, 18, 773–824.
  109. Ho, C.S.; Rydstrom, A.; Manimekalai, M.S.; Svanborg, C.; Grüber, G. Low resolution solution structure of HAMLET and the importance of its α-domains in tumoricidal activity. PLoS ONE 2012, 7, e53051.
  110. Kaspersen, D.; Pedersen, J.N.; Hansted, J.G.; Nielsen, S.B.; Sakthivel, S.; Wilhelm, K.; Nemashkalova, E.L.; Permyakov, S.E.; Permyakov, E.A.; Pinto Oliveira, C.L.; et al. Generic structures of cytotoxic liprotides: Nano-sized complexes with oleic acid cores and shells of disordered proteins. ChemBioChem 2014, 15, 2693–2702.
  111. Frislev, H.S.; Boye, T.L.; Nylandsted, J.; Otzen, D. Liprotides kill cancer cells by disrupting the plasma membrane. Sci. Rep. 2017, 7, 15129.
  112. Pedersen, N.; Frislev, H.K.S.; Pedersen, J.S.; Otzen, D. Structures and mechanisms of formation of liprotides. Biochim. Biophys. Acta Proteins Proteom. 2020, 1868, 140505, doi:10.1016/ j.bbapap.2020.140505.
More