Natural and Synthetic Halogenated Amino Acids: Comparison
Please note this is a comparison between Version 2 by Bruce Ren and Version 1 by Andrea Caporale.

The 3D structure and surface characteristics of proteins and peptides are crucial for interactions with receptors or ligands and can be modified to some extent to modulate their biological roles and pharmacological activities. The introduction of halogen atoms on the side-chains of amino acids is a powerful tool for effecting this type of tuning, influencing both the physico-chemical and structural properties of the modified polypeptides, helping to first dissect and then rationally modify features that affect their mode of action.

  • antimicrobial peptides (AMPs)
  • structure-activity relationship
  • fluoro amino acids
  • fluoro-proline
  • halogenation
  • bromo-tryptophan
  • peptides
  • α- and β-peptoids
Please wait, diff process is still running!

References

  1. Hruby, V.J.; Li, G.; Haskell-Luevano, C.; Shenderovich, M. Design of peptides, proteins, and peptidomimetics in chi space. Biopolymers 1997, 43, 219–266.
  2. Hruby, V.J.; Balse, P.M. Conformational and topographical considerations in designing agonist peptidomimetics from peptide leads. Curr. Med. Chem. 2000, 7, 945–970.
  3. Hruby, V.J.; Cai, M. Design of peptide and peptidomimetic ligands with novel pharmacological activity profiles. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 557–580.
  4. Cowell, S.M.; Lee, Y.S.; Cain, J.P.; Hruby, V.J. Exploring Ramachandran and chi space: Conformationally constrained amino acids and peptides in the design of bioactive polypeptide ligands. Curr. Med. Chem. 2004, 11, 2785–2798.
  5. Marchand, J.A.; Neugebauer, M.E.; Ing, M.C.; Lin, C.I.; Pelton, J.G.; Chang, M.C.Y. Discovery of a pathway for terminal-alkyne amino acid biosynthesis. Nature 2019, 567, 420–424.
  6. Xu, H.X.; Hong, Y.; Zhang, M.Z.; Wang, Y.L.; Liu, S.S.; Wang, X.W. Transcriptional responses of invasive and indigenous whiteflies to different host plants reveal their disparate capacity of adaptation. Sci. Rep. 2015, 5, 10774.
  7. Zou, H.; Li, L.; Zhang, T.; Shi, M.; Zhang, N.; Huang, J.; Xian, M. Biosynthesis and biotechnological application of non-canonical amino acids: Complex and unclear. Biotechnol. Adv. 2018, 36, 1917–1927.
  8. Rodgers, K.J.; Samardzic, K.; Main, B.J. Toxic Nonprotein Amino Acids. In Plant Toxins; Carlini, C.R., Ligabue-Braun, R., Gopalakrishnakone, P., Eds.; Springer: Dordrecht, The Netherlands, 2017; pp. 263–285.
  9. Jiang, S.; Zhang, L.; Cui, D.; Yao, Z.; Gao, B.; Lin, J.; Wei, D. The Important Role of Halogen Bond in Substrate Selectivity of Enzymatic Catalysis. Sci. Rep. 2016, 6, 34750.
  10. Shah, M.B.; Liu, J.; Zhang, Q.; Stout, C.D.; Halpert, J.R. Halogen-pi Interactions in the Cytochrome P450 Active Site: Structural Insights into Human CYP2B6 Substrate Selectivity. ACS Chem. Biol. 2017, 12, 1204–1210.
  11. Parisini, E.; Metrangolo, P.; Pilati, T.; Resnati, G.; Terraneo, G. Halogen bonding in halocarbon-protein complexes: A structural survey. Chem. Soc. Rev. 2011, 40, 2267–2278.
  12. Huhmann, S.; Koksch, B. Fine-Tuning the Proteolytic Stability of Peptides with Fluorinated Amino Acids. Eur. J. Org. Chem. 2018, 2018, 3667–3679.
  13. Imai, Y.N.; Inoue, Y.; Nakanishi, I.; Kitaura, K. Cl-pi interactions in protein-ligand complexes. Protein Sci. 2008, 17, 1129–1137.
  14. Hayashi, A.; Haruna, K.I.; Sato, H.; Ito, K.; Makino, C.; Ito, T.; Sakamoto, K. Incorporation of Halogenated Amino Acids into Antibody Fragments at Multiple Specific Sites Enhances Antigen Binding. ChemBioChem 2021, 22, 120–123.
  15. Lu, Y.; Wang, Y.; Xu, Z.; Yan, X.; Luo, X.; Jiang, H.; Zhu, W. C-X⋯H contacts in biomolecular systems: How they contribute to protein-ligand binding affinity. J. Phys. Chem. B 2009, 113, 12615–12621.
  16. Borozan, S.Z.; Stojanovic, S.D. Halogen bonding in complexes of proteins and non-natural amino acids. Comput. Biol. Chem. 2013, 47, 231–239.
  17. Cosimi, E.; Engl, O.D.; Saadi, J.; Ebert, M.-O.; Wennemers, H. Stereoselective Organocatalyzed Synthesis of α-Fluorinated β-Amino Thioesters and Their Application in Peptide Synthesis. Angew. Chem. Int. Ed. 2016, 55, 13127–13131.
  18. Kubyshkin, V.; Grage, S.L.; Ulrich, A.S.; Budisa, N. Bilayer thickness determines the alignment of model polyproline helices in lipid membranes. Phys. Chem. Chem. Phys. 2019, 21, 22396–22408.
  19. Mansour, F.; Hunter, L. 9—Synthesis and applications of backbone-fluorinated amino acids. In Fluorine in Life Sciences: Pharmaceuticals, Medicinal Diagnostics, and Agrochemicals; Haufe, G., Leroux, F.R., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 325–347.
  20. Berger, A.A.; Voller, J.S.; Budisa, N.; Koksch, B. Deciphering the Fluorine Code-The Many Hats Fluorine Wears in a Protein Environment. ACC Chem. Res. 2017, 50, 2093–2103.
  21. Welte, H.; Zhou, T.; Mihajlenko, X.; Mayans, O.; Kovermann, M. What does fluorine do to a protein? Thermodynamic, and highly-resolved structural insights into fluorine-labelled variants of the cold shock protein. Sci. Rep. 2020, 10, 2640.
  22. Hunter, L. The C-F bond as a conformational tool in organic and biological chemistry. Beilstein J. Org. Chem. 2010, 6, 38.
  23. Nishi, Y.; Uchiyama, S.; Doi, M.; Nishiuchi, Y.; Nakazawa, T.; Ohkubo, T.; Kobayashi, Y. Different effects of 4-hydroxyproline and 4-fluoroproline on the stability of collagen triple helix. Biochemistry 2005, 44, 6034–6042.
  24. Shoulders, M.D.; Raines, R.T. Modulating collagen triple-helix stability with 4-chloro, 4-fluoro, and 4-methylprolines. Adv. Exp. Med. Biol. 2009, 611, 251–252.
  25. Moroder, L.; Budisa, N. Synthetic biology of protein folding. Chemphyschem 2010, 11, 1181–1187.
  26. Crespo, M.D.; Rubini, M. Rational design of protein stability: Effect of (2S,4R)-4-fluoroproline on the stability and folding pathway of ubiquitin. PLoS ONE 2011, 6, e19425.
  27. Roderer, D.; Glockshuber, R.; Rubini, M. Acceleration of the Rate-Limiting Step of Thioredoxin Folding by Replacement of its Conserved cis-Proline with (4 S)-Fluoroproline. ChemBioChem 2015, 16, 2162–2166.
  28. Newberry, R.W.; Raines, R.T. 4-Fluoroprolines: Conformational Analysis and Effects on the Stability and Folding of Peptides and Proteins. Top. Heterocycl. Chem. 2017, 48, 1–25.
  29. Choudhary, A.; Gandla, D.; Krow, G.R.; Raines, R.T. Nature of amide carbonyl-carbonyl interactions in proteins. J. Am. Chem. Soc. 2009, 131, 7244–7246.
  30. Shoulders, M.D.; Raines, R.T. Collagen Structure and Stability. Annu. Rev. Biochem. 2009, 78, 929–958.
  31. Renner, C.; Alefelder, S.; Bae, J.H.; Budisa, N.; Huber, R.; Moroder, L. Fluoroprolines as Tools for Protein Design and Engineering. Angew. Chem. Int. Ed. 2001, 40, 923–925.
  32. Kubyshkin, V.; Davis, R.; Budisa, N. Biochemistry of fluoroprolines: The prospect of making fluorine a bioelement. Beilstein J. Org. Chem. 2021, 17, 439–460.
  33. Wedemeyer, W.J.; Welker, E.; Scheraga, H.A. Proline cis-trans isomerization and protein folding. Biochemistry 2002, 41, 14637–14644.
  34. Roderer, D.J.; Scharer, M.A.; Rubini, M.; Glockshuber, R. Acceleration of protein folding by four orders of magnitude through a single amino acid substitution. Sci. Rep. 2015, 5, 11840.
  35. O’Loughlin, J.; Napolitano, S.; Rubini, M. Protein Design with Fluoroprolines: 4,4-Difluoroproline Does Not Eliminate the Rate-Limiting Step of Thioredoxin Folding. ChemBioChem 2021.
  36. Rubini, M.; Scharer, M.A.; Capitani, G.; Glockshuber, R. (4R)- and (4S)-fluoroproline in the conserved cis-prolyl peptide bond of the thioredoxin fold: Tertiary structure context dictates ring puckering. ChemBioChem 2013, 14, 1053–1057.
  37. Golbik, R.; Yu, C.; Weyher-Stingl, E.; Huber, R.; Moroder, L.; Budisa, N.; Schiene-Fischer, C. Peptidyl prolyl cis/trans-isomerases: Comparative reactivities of cyclophilins, FK506-binding proteins, and parvulins with fluorinated oligopeptide and protein substrates. Biochemistry 2005, 44, 16026–16034.
  38. Torbeev, V.Y.; Hilvert, D. Both the cis-trans equilibrium and isomerization dynamics of a single proline amide modulate β2-microglobulin amyloid assembly. Proc. Natl. Acad. Sci. USA 2013, 110, 20051–20056.
  39. Bella, J.; Eaton, M.; Brodsky, B.; Berman, H.M. Crystal and molecular structure of a collagen-like peptide at 1.9 A resolution. Science 1994, 266, 75–81.
  40. Borgogno, A.; Ruzza, P. The impact of either 4-R-hydroxyproline or 4-R-fluoroproline on the conformation and SH3m-cort binding of HPK1 proline-rich peptide. Amino Acids 2013, 44, 607–614.
  41. Dietz, D.; Kubyshkin, V.; Budisa, N. Applying γ-Substituted Prolines in the Foldon Peptide: Polarity Contradicts Preorganization. ChemBioChem 2015, 16, 403–406.
  42. Costantini, N.V.; Ganguly, H.K.; Martin, M.I.; Wenzell, N.A.; Yap, G.P.A.; Zondlo, N.J. The Distinct Conformational Landscapes of 4S-Substituted Prolines That Promote an endo Ring Pucker. Chemistry 2019, 25, 11356–11364.
  43. Ruzza, P.; Siligardi, G.; Donella-Deana, A.; Calderan, A.; Hussain, R.; Rubini, C.; Cesaro, L.; Osler, A.; Guiotto, A.; Pinna, L.A.; et al. 4-Fluoroproline derivative peptides: Effect on PPII conformation and SH3 affinity. J. Pept. Sci. 2006, 12, 462–471.
  44. Lin, Y.J.; Horng, J.C. Impacts of terminal (4R)-fluoroproline and (4S)-fluoroproline residues on polyproline conformation. Amino Acids 2014, 46, 2317–2324.
  45. D’Annessa, I.; Di Leva, F.S.; La Teana, A.; Novellino, E.; Limongelli, V.; Di Marino, D. Bioinformatics and Biosimulations as Toolbox for Peptides and Peptidomimetics Design: Where Are We? Front. Mol. Biol. Sci. 2020, 7, 66.
  46. Lupia, A.; Mimmi, S.; Iaccino, E.; Maisano, D.; Moraca, F.; Talarico, C.; Vecchio, E.; Fiume, G.; Ortuso, F.; Scala, G.; et al. Molecular modelling of epitopes recognized by neoplastic B lymphocytes in Chronic Lymphocytic Leukemia. Eur. J. Med. Chem. 2020, 185, 111838.
  47. Perricone, U.; Gulotta, M.R.; Lombino, J.; Parrino, B.; Cascioferro, S.; Diana, P.; Cirrincione, G.; Padova, A. An overview of recent molecular dynamics applications as medicinal chemistry tools for the undruggable site challenge. MedChemComm 2018, 9, 920–936.
  48. Autiero, I.; Langella, E.; Saviano, M. Insights into the mechanism of interaction between trehalose-conjugated beta-sheet breaker peptides and Aβ(1–42) fibrils by molecular dynamics simulations. Mol. BioSystems 2013, 9, 2835–2841.
  49. Georgoulia, P.S.; Glykos, N.M. Molecular simulation of peptides coming of age: Accurate prediction of folding, dynamics and structures. Arch. Biochem. Biophys. 2019, 664, 76–88.
  50. Kurczab, R.; Canale, V.; Satała, G.; Zajdel, P.; Bojarski, A.J. Amino Acid Hot Spots of Halogen Bonding: A Combined Theoretical and Experimental Case Study of the 5-HT7 Receptor. J. Med. Chem. 2018, 61, 8717–8733.
  51. Carter, M.; Rappe, A.K.; Ho, P.S. Scalable Anisotropic Shape and Electrostatic Models for Biological Bromine Halogen Bonds. J. Chem. Theory Comput. 2012, 8, 2461–2473.
  52. Scholfield, M.R.; Ford, M.C.; Vander Zanden, C.M.; Billman, M.M.; Ho, P.S.; Rappe, A.K. Force Field Model of Periodic Trends in Biomolecular Halogen Bonds. J. Phys. Chem. B 2015, 119, 9140–9149.
  53. Ford, M.C.; Rappé, A.K.; Ho, P.S. A Reduced Generalized Force Field for Biological Halogen Bonds. J. Chem. Theory Comput. 2021, 17, 5369–5378.
  54. Robalo, J.R.; Vila Verde, A. Unexpected trends in the hydrophobicity of fluorinated amino acids reflect competing changes in polarity and conformation. Phys. Chem. Chem. Phys. 2019, 21, 2029–2038.
  55. Peng, J.; Li, J.; Hamann, M.T. The marine bromotyrosine derivatives. Alkaloids Chem. Biol. 2005, 61, 59–262.
  56. Ueberlein, S.; Machill, S.; Niemann, H.; Proksch, P.; Brunner, E. The skeletal amino acid composition of the marine demosponge Aplysina cavernicola. Mar. Drugs 2014, 12, 4417–4438.
  57. Ueberlein, S.; Machill, S.; Schupp, P.J.; Brunner, E. Determination of the Halogenated Skeleton Constituents of the Marine Demosponge Ianthella basta. Mar. Drugs 2017, 15, 34.
  58. Hunt, S.; Breuer, S.W. Isolation of a new naturally occurring halogenated amino acid: Monochloromonobromotyrosine. Biochim. Biophys. Acta 1971, 252, 401–404.
  59. Carroll, A.; Bowden, B.; Coll, J. Studies of Australian Ascidians. II. Novel Cytotoxic Iodotyrosine-Based Alkaloids from Colonial Ascidians, Aplidium sp. Aust. J. Chem. 1993, 46, 825–832.
  60. Pettit, G.R.; Butler, M.S.; Williams, M.D.; Filiatrault, M.J.; Pettit, R.K. Isolation and structure of hemibastadinols 1-3 from the Papua New Guinea marine sponge Ianthella basta. J. Nat. Prod. 1996, 59, 927–934.
  61. Aiello, A.; Fattorusso, E.; Imperatore, C.; Menna, M.; Muller, W.E. Iodocionin, a cytotoxic iodinated metabolite from the Mediterranean ascidian Ciona edwardsii. Mar. Drugs 2010, 8, 285–291.
  62. Galeano, E.; Thomas, O.P.; Robledo, S.; Munoz, D.; Martinez, A. Antiparasitic bromotyrosine derivatives from the marine sponge Verongula rigida. Mar. Drugs 2011, 9, 1902–1913.
  63. Won, T.H.; Kim, C.K.; Lee, S.H.; Rho, B.J.; Lee, S.K.; Oh, D.C.; Oh, K.B.; Shin, J. Amino Acid-Derived Metabolites from the Ascidian Aplidium sp. Mar. Drugs 2015, 13, 3836–3848.
  64. Tian, D.; Moe, B.; Huang, G.; Jiang, P.; Ling, Z.C.; Li, X.F. Cytotoxicity of Halogenated Tyrosyl Compounds, an Emerging Class of Disinfection Byproducts. Chem. Res. Toxicol. 2020, 33, 1028–1035.
  65. Guillen, P.O.; Jaramillo, K.B.; Jennings, L.; Genta-Jouve, G.; de la Cruz, M.; Cautain, B.; Reyes, F.; Rodriguez, J.; Thomas, O.P. Halogenated Tyrosine Derivatives from the Tropical Eastern Pacific Zoantharians Antipathozoanthus hickmani and Parazoanthus darwini. J. Nat. Prod. 2019, 82, 1354–1360.
  66. Song, G.; Delroisse, J.; Schoenaers, D.; Kim, H.; Nguyen, T.C.; Horbelt, N.; Leclere, P.; Hwang, D.S.; Harrington, M.J.; Flammang, P. Structure and composition of the tunic in the sea pineapple Halocynthia roretzi: A complex cellulosic composite biomaterial. Acta Biomater 2020, 111, 290–301.
  67. Jimenez, E.C. Bromotryptophan and its Analogs in Peptides from Marine Animals. Protein Pept. Lett. 2019, 26, 251–260.
  68. Fusetani, N.; Sugawara, T.; Matsunaga, S.; Hirota, H. Orbiculamide A: A novel cytotoxic cyclic peptide from a marine sponge Theonella sp. J. Am. Chem. Soc. 1991, 113, 7811–7812.
  69. Kobayashi, J.i.; Itagaki, F.; Shigemori, I.; Takao, T.; Shimonishi, Y. Keramamides E, G, H, and J, new cyclic peptides containing an oxazole or a thiazole ring from a Theonella sponge. Tetrahedron 1995, 51, 2525–2532.
  70. Kobayashi, J.i.; Sato, M.; Murayama, T.; Ishibashi, M.; Wälchi, M.R.; Kanai, M.; Shoji, J.; Ohizumi, Y. Konbamide, a novel peptide with calmoduiin antagonistic activity from the Okinawan marine sponge Theonella sp. J. Chem. Soc. Chem. Commun. 1991, 15, 1050–1052.
  71. Zabriskie, T.M.; Klocke, J.A.; Ireland, C.M.; Marcus, A.H.; Molinski, T.F.; Faulkner, D.J.; Xu, C.; Clardy, J. Jaspamide, a modified peptide from a Jaspis sponge, with insecticidal and antifungal activity. J. Am. Chem. Soc. 1986, 108, 3123–3124.
  72. Ebada, S.S.; Wray, V.; de Voogd, N.J.; Deng, Z.W.; Lin, W.H.; Proksch, P. Two New Jaspamide Derivatives from the Marine Sponge Jaspis splendens. Mar. Drugs 2009, 7, 435–444.
  73. Watts, K.R.; Morinaka, B.I.; Arnagata, T.; Robinson, S.J.; Tenney, K.; Bray, W.M.; Gassner, N.C.; Lokey, R.S.; Media, J.; Valeriote, F.A.; et al. Biostructural Features of Additional Jasplakinolide (Jaspamide) Analogues. J. Nat. Prod. 2011, 74, 341–351.
  74. Clark, W.D.; Corbett, T.; Valeriote, F.; Crews, P. Cyclocinamide, A. An unusual cytotoxic halogenated hexapeptide from the marine sponge Psammocinia. J. Am. Chem. Soc. 1997, 119, 9285–9286.
  75. Sjogren, M.; Goransson, U.; Johnson, A.L.; Dahlstrom, M.; Andersson, R.; Bergman, J.; Jonsson, P.R.; Bohlin, L. Antifouling activity of brominated cyclopeptides from the marine sponge Geodia barretti. J. Nat. Prod. 2004, 67, 368–372.
  76. Hedner, E.; Sjogren, M.; Hodzic, S.; Andersson, R.; Goransson, U.; Jonsson, P.R.; Bohlin, L. Antifouling activity of a dibrominated cyclopeptide from the marine sponge Geodia barretti. J. Nat. Prod. 2008, 71, 330–333.
  77. Azumi, K.; Yokosawa, H.; Ishii, S. Halocyamines—Novel Antimicrobial Tetrapeptide-Like Substances Isolated from the Hemocytes of the Solitary Ascidian Halocynthia-Roretzi. Biochemistry 1990, 29, 159–165.
  78. Bittner, S.; Scherzer, R.; Harlev, E. The five bromotryptophans. Amino Acids 2007, 33, 19–42.
  79. Lee, I.H.; Cho, Y.; Lehrer, R.I. Styelins, Broad-Spectrum Antimicrobial Peptides from the Solitary Tunicate, Styela clava. Comp. Biochem. Physiol. Part B Biochem. Mol. Biol. 1997, 118, 515–521.
  80. Craig, A.G.; Jimenez, E.C.; Dykert, J.; Nielsen, D.B.; Gulyas, J.; Abogadie, F.C.; Porter, J.; Rivier, J.E.; Cruz, L.J.; Olivera, B.M.; et al. A novel post-translational modification involving bromination of tryptophan. Identification of the residue, L-6-bromotryptophan, in peptides from Conus imperialis and Conus radiatus venom. J. Biol. Chem. 1997, 272, 4689–4698.
  81. Jin, A.-H.; Muttenthaler, M.; Dutertre, S.; Himaya, S.W.A.; Kaas, Q.; Craik, D.J.; Lewis, R.J.; Alewood, P.F. Conotoxins: Chemistry and Biology. Chem. Rev. 2019, 119, 11510–11549.
  82. Payne, J.T.; Andorfer, M.C.; Lewis, J.C. Regioselective arene halogenation using the FAD-dependent halogenase RebH. Angew Chem. Int. Ed. Engl. 2013, 52, 5271–5274.
  83. Flecks, S.; Patallo, E.P.; Zhu, X.; Ernyei, A.J.; Seifert, G.; Schneider, A.; Dong, C.; Naismith, J.H.; van Pee, K.H. New insights into the mechanism of enzymatic chlorination of tryptophan. Angew. Chem. Int. Ed. Engl. 2008, 47, 9533–9536.
  84. Agostini, F.; Völler, J.-S.; Koksch, B.; Acevedo-Rocha, C.G.; Kubyshkin, V.; Budisa, N. Biocatalysis with Unnatural Amino Acids: Enzymology Meets Xenobiology. Angew. Chem. Int. Ed. 2017, 56, 9680–9703.
  85. Baumann, T.; Nickling, J.H.; Bartholomae, M.; Buivydas, A.; Kuipers, O.P.; Budisa, N. Prospects of In vivo Incorporation of Non-canonical Amino Acids for the Chemical Diversification of Antimicrobial Peptides. Front. Microbiol. 2017, 8, 124.
  86. Mäde, V.; Els-Heindl, S.; Beck-Sickinger, A.G. Automated solid-phase peptide synthesis to obtain therapeutic peptides. Beilstein J. Org. Chem. 2014, 10, 1197–1212.
  87. Frese, M.; Schnepel, C.; Minges, H.; Voss, H.; Feiner, R.; Sewald, N. Modular Combination of Enzymatic Halogenation of Tryptophan with Suzuki-Miyaura Cross-Coupling Reactions. ChemCatChem 2016, 8, 1799–1803.
  88. Schnepel, C.; Kemker, I.; Sewald, N. One-Pot Synthesis of d-Halotryptophans by Dynamic Stereoinversion Using a Specific l-Amino Acid Oxidase. ACS Catal. 2019, 9, 1149–1158.
  89. Narayanan, S.; Iyengar, M.R.; Ganju, P.L.; Rengaraju, S.; Shomura, T.; Tsuruoka, T.; Inouye, S.; Niida, T. γ-Chloronorvaline, a leucine analog from Streptomyces. J. Antibiot. 1980, 33, 1249–1255.
  90. Chaiet, L.; Arison, B.H.; Monaghan, R.L.; Springer, J.P.; Smith, J.L.; Zimmerman, S.B. -4-amino-3-chloro-2-pentenedioic acid, a new antibiotic. Fermentation, isolation and characterization. J. Antibiot. 1984, 37, 207–210.
  91. Kuroda, Y.; Okuhara, M.; Goto, T.; Yamashita, M.; Iguchi, E.; Kohsaka, M.; Aoki, H.; Imanaka, H. FR-900148, a new antibiotic. I. Taxonomy, fermentation, isolation and characterization. J. Antibiot. 1980, 33, 259–266.
  92. Kuroda, Y.; Okuhara, M.; Goto, T.; Okamoto, M.; Yamashita, M.; Kohsaka, M.; Aoki, H.; Imanaka, H. FR-900148, a new antibiotic. II. Structure determination of FR-900148. J. Antibiot. 1980, 33, 267–271.
  93. Martin, D.G.; Chidester, C.G.; Mizsak, S.A.; Duchamp, D.J.; Baczysnkyj, L. The isolation, structure, and absolute configuration of U-43,795, a new antitumor agent. J. Antibiot. 1975, 28, 91–93.
  94. Hatanaka, S.-I. Amino Acids from Mushrooms. In Fortschritte der Chemie organischer Naturstoffe / Progress in the Chemistry of Organic Natural Products; Herz, W., Kirby, G.W., Moore, R.E., Steglich, W., Tamm, C., Eds.; Springer: Vienna, Austria, 1992; pp. 1–140.
  95. Chilton, W.S.; Tsou, G. A chloro amino acid from Amanita solitaria. Phytochemistry 1972, 11, 2853–2857.
  96. Ohta, T.; Nakajima, S.; Hatanaka, S.-I.; Yamamoto, M.; Shimmen, Y.; Nishimura, S.; Yamaizumi, Z.; Nozoe, S. A chlorohydrin amino acid from Amanita abrupta. Phytochemistry 1987, 26, 565–566.
  97. Sanada, M.; Miyano, T.; Iwadare, S.; Williamson, J.M.; Arison, B.H.; Smith, J.L.; Douglas, A.W.; Liesch, J.M.; Inamine, E. Biosynthesis of fluorothreonine and fluoroacetic acid by the thienamycin producer, Streptomyces cattleya. J. Antibiot. 1986, 39, 259–265.
  98. Moravej, H.; Moravej, Z.; Yazdanparast, M.; Heiat, M.; Mirhosseini, A.; Moosazadeh Moghaddam, M.; Mirnejad, R. Antimicrobial Peptides: Features, Action, and Their Resistance Mechanisms in Bacteria. Microb. Drug Resist. 2018, 24, 747–767.
  99. Gribble, G.W. Biohalogenation. Fortschr. Chem. Org. Naturst. 2010, 91, 349–365.
  100. Agarwal, V.; Miles, Z.D.; Winter, J.M.; Eustaquio, A.S.; El Gamal, A.A.; Moore, B.S. Enzymatic Halogenation and Dehalogenation Reactions: Pervasive and Mechanistically Diverse. Chem. Rev. 2017, 117, 5619–5674.
  101. Lohman, D.C.; Edwards, D.R.; Wolfenden, R. Catalysis by Desolvation: The Catalytic Prowess of SAM-Dependent Halide-Alkylating Enzymes. J. Am. Chem. Soc. 2013, 135, 14473–14475.
  102. O’Hagan, D.; Schaffrath, C.; Cobb, S.L.; Hamilton, J.T.G.; Murphy, C.D. Biosynthesis of an organofluorine molecule. Nature 2002, 416, 279.
  103. Fang, J.; Hait, D.; Head-Gordon, M.; Chang, M.C.Y. Chemoenzymatic Platform for Synthesis of Chiral Organofluorines Based on Type II Aldolases. Angew. Chem. Int. Ed. 2019, 58, 11841–11845.
  104. Strickland, M.; Willis, C.L. Synthesis of Halogenated α-Amino Acids. In Amino Acids, Peptides and Proteins in Organic Chemistry; Wiley: Hoboken, NJ, USA, 2010; pp. 441–471.
  105. Veldmann, K.H.; Minges, H.; Sewald, N.; Lee, J.H.; Wendisch, V.F. Metabolic engineering of Corynebacterium glutamicum for the fermentative production of halogenated tryptophan. J. Biotechnol. 2019, 291, 7–16.
  106. Chen, B.; Feng, Z. Asymmetric Synthesis Method for Natural Product Chlorinated Gamma-Norvaline Hydrochloride. Patent CN103145572A, 12 June 2013.
  107. Bencivenni, G.; Salazar Illera, D.; Moccia, M.; Houk, K.N.; Izzo, J.A.; Novacek, J.; Grieco, P.; Vetticatt, M.J.; Waser, M.; Adamo, M.F.A. Study of Ground State Interactions of Enantiopure Chiral Quaternary Ammonium Salts and Amides, Nitroalkanes, Nitroalkenes, Esters, Heterocycles, Ketones and Fluoroamides. Chem. Eur. J. 2021, 27, 11352–11366.
  108. Cortigiani, M.; Mereu, A.; Gillick Healy, M.; Adamo, M.F.A. Enantioselective Desymmetrization of cis-3,5-O-Arylidenecyclohexanones Catalyzed by Cinchona-Derived Quaternary Ammonium Salts. J. Org. Chem. 2019, 84, 4112–4119.
  109. Baschieri, A.; Bernardi, L.; Ricci, A.; Suresh, S.; Adamo, M.F.A. Catalytic Asymmetric Conjugate Addition of Nitroalkanes to 4-Nitro-5-styrylisoxazoles. Angew. Chem. Int. Ed. 2009, 48, 9342–9345.
  110. Fiandra, C.D.; Piras, L.; Fini, F.; Disetti, P.; Moccia, M.; Adamo, M.F.A. Phase transfer catalyzed enantioselective cyclopropanation of 4-nitro-5-styrylisoxazoles. Chem. Commun. 2012, 48, 3863–3865.
  111. Moccia, M.; Wells, R.J.; Adamo, M.F.A. An improved procedure to prepare 3-methyl-4-nitroalkylenethylisoxazoles and their reaction under catalytic enantioselective Michael addition with nitromethane. Org. Biomol. Chem. 2015, 13, 2192–2195.
  112. Piras, L.; Moccia, M.; Cortigiani, M.; Adamo, M.F.A. Cyclopropanation of 5-(1-Bromo-2-phenyl- vinyl)-3-methyl-4-nitro-isoxazoles under Phase Transfer Catalysis (PTC) Conditions. Catalysts 2015, 5, 595–605.
  113. Destro, D.; Sanchez, S.; Cortigiani, M.; Adamo, M.F.A. Reaction of azides and enolisable aldehydes under the catalysis of organic bases and Cinchona based quaternary ammonium salts. Org. Biomol. Chem. 2017, 15, 5227–5235.
  114. Del Fiandra, C.; Moccia, M.; Adamo, M.F.A. Enantioselective cyclopropanation of (Z)-3-substituted-2-(4-pyridyl)-acrylonitriles catalyzed by Cinchona ammonium salts. Org. Biomol. Chem. 2016, 14, 3105–3111.
  115. Deng, H.; Ma, L.; Bandaranayaka, N.; Qin, Z.; Mann, G.; Kyeremeh, K.; Yu, Y.; Shepherd, T.; Naismith, J.H.; O’Hagan, D. Identification of Fluorinases from Streptomyces sp. MA37, Norcardia brasiliensis, and Actinoplanes sp. N902-109 by Genome Mining. ChemBioChem 2014, 15, 364–368.
  116. Scolastico, C.; Conca, E.; Prati, L.; Guanti, G.; Banfi, L.; Berti, A.; Farina, P.; Valcavi, U. Diastereo- and Enantioselective Synthesis of Fluorinated Threonines. Synthesis 1985, 850–855.
  117. Potenti, S.; Spada, L.; Fuse, M.; Mancini, G.; Gualandi, A.; Leonardi, C.; Cozzi, P.G.; Puzzarini, C.; Barone, V. 4-Fluoro-Threonine: From Diastereoselective Synthesis to pH-Dependent Conformational Equilibrium in Aqueous Solution. ACS Omega 2021, 6, 13170–13181.
  118. Merkx, R.; de Bruin, G.; Kruithof, A.; van den Bergh, T.; Snip, E.; Lutz, M.; El Oualid, F.; Ovaa, H. Scalable synthesis of γ-thiolysine starting from lysine and a side by side comparison with δ-thiolysine in non-enzymatic ubiquitination. Chem. Sci. 2013, 4, 4494–4498.
  119. Mei, H.; Han, J.; Klika, K.D.; Izawa, K.; Sato, T.; Meanwell, N.A.; Soloshonok, V.A. Applications of fluorine-containing amino acids for drug design. Eur. J. Med. Chem. 2020, 186, 111826.
  120. Moschner, J.; Stulberg, V.; Fernandes, R.; Huhmann, S.; Leppkes, J.; Koksch, B. Approaches to Obtaining Fluorinated α-Amino Acids. Chem. Rev. 2019, 119, 10718–10801.
  121. Brittain, W.D.G.; Lloyd, C.M.; Cobb, S.L. Synthesis of complex unnatural fluorine-containing amino acids. J. Fluor. Chem. 2020, 239, 109630.
  122. Ghosh, C.; Sarkar, P.; Issa, R.; Haldar, J. Alternatives to Conventional Antibiotics in the Era of Antimicrobial Resistance. Trends Microbiol. 2019, 27, 323–338.
  123. Nuti, R.; Goud, N.S.; Saraswati, A.P.; Alvala, R.; Alvala, M. Antimicrobial Peptides: A Promising Therapeutic Strategy in Tackling Antimicrobial Resistance. Curr. Med. Chem. 2017, 24, 4303–4314.
  124. Kang, H.K.; Kim, C.; Seo, C.H.; Park, Y. The therapeutic applications of antimicrobial peptides (AMPs): A patent review. J. Microbiol. 2017, 55, 1–12.
  125. Crabtree, D.P.E.; Herrera, B.J.; Kang, S. The response of human bacteria to static magnetic field and radiofrequency electromagnetic field. J. Microbiol. 2017, 55, 809–815.
  126. Sharma, K.; Aaghaz, S.; Shenmar, K.; Jain, R. Short Antimicrobial Peptides. Recent. Pat. Antiinfect. Drug Discov. 2018, 13, 12–52.
  127. Sierra, J.M.; Fuste, E.; Rabanal, F.; Vinuesa, T.; Vinas, M. An overview of antimicrobial peptides and the latest advances in their development. Expert. Opin. Biol. Ther. 2017, 17, 663–676.
  128. Munshi, T.; Sparrow, A.; Wren, B.W.; Reljic, R.; Willcocks, S.J. The Antimicrobial Peptide, Bactenecin 5, Supports Cell-Mediated but Not Humoral Immunity in the Context of a Mycobacterial Antigen Vaccine Model. Antibiotics 2020, 9, 926.
  129. Malik, E.; Dennison, S.R.; Harris, F.; Phoenix, D.A. pH Dependent Antimicrobial Peptides and Proteins, Their Mechanisms of Action and Potential as Therapeutic Agents. Pharmaceuticals 2016, 9, 67.
  130. Ahmed, T.A.E.; Hammami, R. Recent insights into structure-function relationships of antimicrobial peptides. J. Food Biochem. 2019, 43, e12546.
  131. Harris, F.; Dennison, S.R.; Phoenix, D.A. Anionic antimicrobial peptides from eukaryotic organisms. Curr. Protein Pept. Sci. 2009, 10, 585–606.
  132. Price, R.L.; Bugeon, L.; Mostowy, S.; Makendi, C.; Wren, B.W.; Williams, H.D.; Willcocks, S.J. In vitro and in vivo properties of the bovine antimicrobial peptide, Bactenecin 5. PLoS ONE 2019, 14, e0210508.
  133. Mahlapuu, M.; Håkansson, J.; Björn, L.R.C. Antimicrobial Peptides: An Emerging Category of Therapeutic Agents. Front. Cell. Infect. Microbiol. 2016, 6, 12.
  134. Tossi, A.; Sandri, L.; Giangaspero, A. Amphipathic, α-helical antimicrobial peptides. Biopolymers 2000, 55, 4–30.
  135. Bahar, A.A.; Ren, D. Antimicrobial peptides. Pharmaceuticals 2013, 6, 1543–1575.
  136. Pasupuleti, M.; Schmidtchen, A.; Malmsten, M. Antimicrobial peptides: Key components of the innate immune system. Crit. Rev. Biotechnol. 2012, 32, 143–171.
  137. Romeo, D.; Skerlavaj, B.; Bolognesi, M.; Gennaro, R. Structure and bactericidal activity of an antibiotic dodecapeptide purified from bovine neutrophils. J. Biol. Chem. 1988, 263, 9573–9575.
  138. Tossi, A.; Sandri, L. Molecular diversity in gene-encoded, cationic antimicrobial polypeptides. Curr. Pharm. Des. 2002, 8, 743–761.
  139. Antcheva, N.; Guida, F.; Tossi, A. Chapter 18—Defensins. In Handbook of Biologically Active Peptides, 2nd ed.; Kastin, A.J., Ed.; Academic Press: Boston, MA, USA, 2013; pp. 101–118.
  140. Lewies, A.; Wentzel, J.F.; Jacobs, G.; Du Plessis, L.H. The Potential Use of Natural and Structural Analogues of Antimicrobial Peptides in the Fight against Neglected Tropical Diseases. Molecules 2015, 20, 15392–15433.
  141. Powers, J.P.; Hancock, R.E. The relationship between peptide structure and antibacterial activity. Peptides 2003, 24, 1681–1691.
  142. Wang, G. Improved methods for classification, prediction, and design of antimicrobial peptides. Methods Mol. Biol. 2015, 1268, 43–66.
  143. Nguyen, L.T.; Haney, E.F.; Vogel, H.J. The expanding scope of antimicrobial peptide structures and their modes of action. Trends Biotechnol. 2011, 29, 464–472.
  144. Scocchi, M.; Tossi, A.; Gennaro, R. Proline-rich antimicrobial peptides: Converging to a non-lytic mechanism of action. Cell Mol. Life Sci. 2011, 68, 2317–2330.
  145. Vergis, J.; Malik, S.S.; Pathak, R.; Kumar, M.; Ramanjaneya, S.; Kurkure, N.V.; Barbuddhe, S.B.; Rawool, D.B. Antimicrobial Efficacy of Indolicidin Against Multi-Drug Resistant Enteroaggregative Escherichia coli in a Galleria mellonella Model. Front. Microbiol. 2019, 10, 2723.
  146. Smirnova, M.P.; Kolodkin, N.I.; Kolobov, A.A.; Afonin, V.G.; Afonina, I.V.; Stefanenko, L.I.; Shpen, V.M.; Shamova, O.V.; Kolobov, A.A. Indolicidin analogs with broad-spectrum antimicrobial activity and low hemolytic activity. Peptides 2020, 132, 170356.
  147. Tokunaga, Y.; Niidome, T.; Hatakeyama, T.; Aoyagi, H. Antibacterial activity of bactenecin 5 fragments and their interaction with phospholipid membranes. J. Pept. Sci. 2001, 7, 297–304.
  148. Lee, T.H.; Hall, K.N.; Aguilar, M.I. Antimicrobial Peptide Structure and Mechanism of Action: A Focus on the Role of Membrane Structure. Curr. Top. Med. Chem. 2016, 16, 25–39.
  149. Melo, M.N.; Ferre, R.; Castanho, M.A. Antimicrobial peptides: Linking partition, activity and high membrane-bound concentrations. Nat. Rev. Microbiol. 2009, 7, 245–250.
  150. Mardirossian, M.; Grzela, R.; Giglione, C.; Meinnel, T.; Gennaro, R.; Mergaert, P.; Scocchi, M. The host antimicrobial peptide Bac71-35 binds to bacterial ribosomal proteins and inhibits protein synthesis. Chem. Biol. 2014, 21, 1639–1647.
  151. Scocchi, M.; Mardirossian, M.; Runti, G.; Benincasa, M. Non-Membrane Permeabilizing Modes of Action of Antimicrobial Peptides on Bacteria. Curr. Top. Med. Chem. 2016, 16, 76–88.
  152. Scocchi, M.L.C.; Decarli, P.; Mignogna, G.; Christen, P.; Gennaro, R. The proline-rich antibacterial peptide Bac7 binds to and inhibits in vitro the molecular chaperone DnaK. Int. J. Pept. Res. Ther. 2009, 147–155.
  153. Mardirossian, M.; Sola, R.; Beckert, B.; Collis, D.W.P.; Di Stasi, A.; Armas, F.; Hilpert, K.; Wilson, D.N.; Scocchi, M. Proline-Rich Peptides with Improved Antimicrobial Activity against E. coli, K. pneumoniae, and A. baumannii. ChemMedChem 2019, 14, 2025–2033.
  154. Padovan, L.; Scocchi, M.; Tossi, A. Structural aspects of plant antimicrobial peptides. Curr. Protein Pept. Sci. 2010, 11, 210–219.
  155. Orlov, D.S.; Shamova, O.V.; Eliseev, I.E.; Zharkova, M.S.; Chakchir, O.B.; Antcheva, N.; Zachariev, S.; Panteleev, P.V.; Kokryakov, V.N.; Ovchinnikova, T.V.; et al. Redesigning Arenicin-1, an Antimicrobial Peptide from the Marine Polychaeta Arenicola marina, by Strand Rearrangement or Branching, Substitution of Specific Residues, and Backbone Linearization or Cyclization. Mar. Drugs 2019, 17, 376.
  156. Walkenhorst, W.F.; Klein, J.W.; Vo, P.; Wimley, W.C. pH Dependence of microbe sterilization by cationic antimicrobial peptides. Antimicrob. Agents Chemother. 2013, 57, 3312–3320.
  157. Brogden, K.A. Antimicrobial peptides: Pore formers or metabolic inhibitors in bacteria? Nat. Rev. Microbiol. 2005, 3, 238–250.
  158. Molchanova, N.; Hansen, P.R.; Franzyk, H. Advances in Development of Antimicrobial Peptidomimetics as Potential Drugs. Molecules 2017, 22, 1430.
  159. Li, H.; Fu, S.; Wang, Y.; Yuan, X.; Liu, L.; Dong, H.; Wang, Q.; Zhang, Z. Antimicrobial and antitumor activity of peptidomimetics synthesized from amino acids. Bioorg. Chem. 2021, 106, 104506.
  160. Tian, L.; Zhang, D.; Su, P.; Wei, Y.; Wang, Z.; Wang, P.X.; Dai, C.J.; Gong, G.L. Design, recombinant expression, and antibacterial activity of a novel hybrid magainin-thanatin antimicrobial peptide. Prep. Biochem. Biotechnol. 2019, 49, 427–434.
  161. Xu, H.; Tie, K.; Zhang, Y.; Feng, X.; Cao, Y.; Han, W. Design, expression, and characterization of the hybrid antimicrobial peptide T-catesbeianin-1 based on FyuA. J. Pept. Sci. 2018, 24.
  162. Wade, H.M.; Darling, L.E.O.; Elmore, D.E. Hybrids made from antimicrobial peptides with different mechanisms of action show enhanced membrane permeabilization. Biochim. Biophys. Acta Biomembr. 2019, 1861, 182980.
  163. Avan, I.; Hall, C.D.; Katritzky, A.R. Peptidomimetics via modifications of amino acids and peptide bonds. Chem. Soc. Rev. 2014, 43, 3575–3594.
  164. Skovbakke, S.L.; Holdfeldt, A.; Forsman, H.; Bylund, J.; Franzyk, H. The Role of Formyl Peptide Receptors for Immunomodulatory Activities of Antimicrobial Peptides and Peptidomimetics. Curr. Pharm. Des. 2018, 24, 1100–1120.
  165. Uzzell, T.; Stolzenberg, E.D.; Shinnar, A.E.; Zasloff, M. Hagfish intestinal antimicrobial peptides are ancient cathelicidins. Peptides 2003, 24, 1655–1667.
  166. Taylor, S.W.; Craig, A.G.; Fischer, W.H.; Park, M.; Lehrer, R.I. Styelin D, an extensively modified antimicrobial peptide from ascidian hemocytes. J. Biol. Chem. 2000, 275, 38417–38426.
  167. Tasiemski, A.; Schikorski, D.; Le Marrec-Croq, F.; Pontoire-Van Camp, C.; Boidin-Wichlacz, C.; Sautiere, P.E. Hedistin: A novel antimicrobial peptide containing bromotryptophan constitutively expressed in the NK cells-like of the marine annelid, Nereis diversicolor. Dev. Comp. Immunol. 2007, 31, 749–762.
  168. Li, C.; Haug, T.; Styrvold, O.B.; Jorgensen, T.O.; Stensvag, K. Strongylocins, novel antimicrobial peptides from the green sea urchin, Strongylocentrotus droebachiensis. Dev. Comp. Immunol. 2008, 32, 1430–1440.
  169. Solstad, R.G.; Li, C.; Isaksson, J.; Johansen, J.; Svenson, J.; Stensvag, K.; Haug, T. Novel Antimicrobial Peptides EeCentrocins 1, 2 and EeStrongylocin 2 from the Edible Sea Urchin Echinus esculentus Have 6-Br-Trp Post-Translational Modifications. PLoS ONE 2016, 11, e0151820.
  170. Li, C.; Haug, T.; Moe, M.K.; Styrvold, O.B.; Stensvag, K. Centrocins: Isolation and characterization of novel dimeric antimicrobial peptides from the green sea urchin, Strongylocentrotus droebachiensis. Dev. Comp. Immunol. 2010, 34, 959–968.
  171. Shinnar, A.E.; Uzzell, T.; Rao, M.N.; Spooner, E.; Lane, W.S.; Zasloff, M.A. New family of linear antimicrobial peptides from hagfish intestine contains bromo-tryptophan as novel amino acid. In Proceedings of the 14th Peptides—American Symposium, Columbus, Ohio, 18–23 June 1996; pp. 189–191.
  172. Tossi, A.; D’Este, F.; Skerlavaj, B.; Gennaro, R. Structural and Functional Diversity of Cathelicidins. In Antimicrobial Peptides, 2nd ed.; Wang, G., Ed.; CABI: Wallingford, UK, 2017; pp. 20–48.
  173. Zhao, C.; Liaw, L.; Lee, I.H.; Lehrer, R.I. cDNA cloning of three cecropin-like antimicrobial peptides (Styelins) from the tunicate, Styela clava. FEBS Lett. 1997, 412, 144–148.
  174. Castiglione, F.; Lazzarini, A.; Carrano, L.; Corti, E.; Ciciliato, I.; Gastaldo, L.; Candiani, P.; Losi, D.; Marinelli, F.; Selva, E.; et al. Determining the structure and mode of action of microbisporicin, a potent lantibiotic active against multiresistant pathogens. Chem. Biol. 2008, 15, 22–31.
  175. Cruz, J.C.; Iorio, M.; Monciardini, P.; Simone, M.; Brunati, C.; Gaspari, E.; Maffioli, S.I.; Wellington, E.; Sosio, M.; Donadio, S. Brominated Variant of the Lantibiotic NAI-107 with Enhanced Antibacterial Potency. J. Nat. Prod. 2015, 78, 2642–2647.
  176. Jabes, D.; Brunati, C.; Candiani, G.; Riva, S.; Romano, G.; Donadio, S. Efficacy of the new lantibiotic NAI-107 in experimental infections induced by multidrug-resistant Gram-positive pathogens. Antimicrob. Agents Chemother. 2011, 55, 1671–1676.
  177. Thomsen, T.T.; Mojsoska, B.; Cruz, J.C.; Donadio, S.; Jenssen, H.; Lobner-Olesen, A.; Rewitz, K. The Lantibiotic NAI-107 Efficiently Rescues Drosophila melanogaster from Infection with Methicillin-Resistant Staphylococcus aureus USA300. Antimicrob. Agents Chemother. 2016, 60, 5427–5436.
  178. Willey, J.M.; van der Donk, W.A. Lantibiotics: Peptides of diverse structure and function. Annu. Rev. Microbiol. 2007, 61, 477–501.
  179. Bierbaum, G.; Sahl, H.G. Lantibiotics: Mode of action, biosynthesis and bioengineering. Curr. Pharm. Biotechnol. 2009, 10, 2–18.
  180. Maffioli, S.I.; Iorio, M.; Sosio, M.; Monciardini, P.; Gaspari, E.; Donadio, S. Characterization of the congeners in the lantibiotic NAI-107 complex. J. Nat. Prod. 2014, 77, 79–84.
  181. Midura-Nowaczek, K.; Markowska, A. Antimicrobial peptides and their analogs: Searching for new potential therapeutics. Perspect. Med. Chem. 2014, 6, 73–80.
  182. Rotem, S.; Mor, A. Antimicrobial peptide mimics for improved therapeutic properties. Biochim. Biophys. Acta 2009, 1788, 1582–1592.
  183. Marr, A.K.; Gooderham, W.J.; Hancock, R.E. Antibacterial peptides for therapeutic use: Obstacles and realistic outlook. Curr. Opin. Pharmacol. 2006, 6, 468–472.
  184. Chongsiriwatana, N.P.; Patch, J.A.; Czyzewski, A.M.; Dohm, M.T.; Ivankin, A.; Gidalevitz, D.; Zuckermann, R.N.; Barron, A.E. Peptoids that mimic the structure, function, and mechanism of helical antimicrobial peptides. Proc. Natl. Acad. Sci. USA 2008, 105, 2794–2799.
  185. Shuey, S.W.; Delaney, W.J.; Shah, M.C.; Scialdone, M.A. Antimicrobial β-peptoids by a block synthesis approach. Bioorg. Med. Chem. Lett. 2006, 16, 1245–1248.
  186. Liu, D.; DeGrado, W.F. De novo design, synthesis, and characterization of antimicrobial β-peptides. J. Am. Chem. Soc. 2001, 123, 7553–7559.
  187. Schmitt, M.A.; Weisblum, B.; Gellman, S.H. Interplay among folding, sequence, and lipophilicity in the antibacterial and hemolytic activities of α/β-peptides. J. Am. Chem. Soc. 2007, 129, 417–428.
  188. Olsen, C.A. Peptoid-Peptide hybrid backbone architectures. ChemBioChem 2010, 11, 152–160.
  189. Jahnsen, R.D.; Frimodt-Moller, N.; Franzyk, H. Antimicrobial activity of peptidomimetics against multidrug-resistant Escherichia coli: A comparative study of different backbones. J. Med. Chem. 2012, 55, 7253–7261.
  190. Niu, Y.; Wu, H.; Li, Y.; Hu, Y.; Padhee, S.; Li, Q.; Cao, C.; Cai, J. AApeptides as a new class of antimicrobial agents. Org. Biomol. Chem. 2013, 11, 4283–4290.
  191. Gan, B.H.; Gaynord, J.; Rowe, S.M.; Deingruber, T.; Spring, D.R. The multifaceted nature of antimicrobial peptides: Current synthetic chemistry approaches and future directions. Chem. Soc. Rev. 2021, 50, 7820–7880.
  192. Gottler, L.M.; de la Salud-Bea, R.; Marsh, E.N. The fluorous effect in proteins: Properties of α4F6, a 4-α-helix bundle protein with a fluorocarbon core. Biochemistry 2008, 47, 4484–4490.
  193. Anderson, D.G.; Shirts, R.B.; Cross, T.A.; Busath, D.D. Noncontact dipole effects on channel permeation. V. Computed potentials for fluorinated gramicidin. Biophys. J. 2001, 81, 1255–1264.
  194. Salwiczek, M.; Nyakatura, E.K.; Gerling, U.I.; Ye, S.; Koksch, B. Fluorinated amino acids: Compatibility with native protein structures and effects on protein-protein interactions. Chem. Soc. Rev. 2012, 41, 2135–2171.
  195. Kitevski-LeBlanc, J.L.; Prosser, R.S. Current applications of 19F NMR to studies of protein structure and dynamics. Prog. Nucl. Magn. Reson. Spectrosc 2012, 62, 1–33.
  196. Kubyshkin, V.; Afonin, S.; Kara, S.; Budisa, N.; Mykhailiuk, P.K.; Ulrich, A.S. γ-(S)-Trifluoromethyl proline: Evaluation as a structural substitute of proline for solid state (19)F-NMR peptide studies. Org. Biomol. Chem. 2015, 13, 3171–3181.
  197. Gottler, L.M.; Lee, H.Y.; Shelburne, C.E.; Ramamoorthy, A.; Marsh, E.N. Using fluorous amino acids to modulate the biological activity of an antimicrobial peptide. ChemBioChem 2008, 9, 370–373.
  198. Marsh, E.N.; Buer, B.C.; Ramamoorthy, A. Fluorine—A new element in the design of membrane-active peptides. Mol. Biosyst. 2009, 5, 1143–1147.
  199. Niemz, A.; Tirrell, D.A. Self-association and membrane-binding behavior of melittins containing trifluoroleucine. J. Am. Chem. Soc. 2001, 123, 7407–7413.
  200. Meng, H.; Kumar, K. Antimicrobial activity and protease stability of peptides containing fluorinated amino acids. J. Am. Chem. Soc. 2007, 129, 15615–15622.
  201. Jia, F.; Zhang, Y.; Wang, J.; Peng, J.; Zhao, P.; Zhang, L.; Yao, H.; Ni, J.; Wang, K. The effect of halogenation on the antimicrobial activity, antibiofilm activity, cytotoxicity and proteolytic stability of the antimicrobial peptide Jelleine-I. Peptides 2019, 112, 56–66.
  202. Arias, M.; Aramini, J.M.; Riopel, N.D.; Vogel, H.J. Fluorine-19 NMR spectroscopy of fluorinated analogs of tritrpticin highlights a distinct role for Tyr residues in antimicrobial peptides. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183260.
  203. Lawyer, C.; Pai, S.; Watabe, M.; Borgia, P.; Mashimo, T.; Eagleton, L.; Watabe, K. Antimicrobial activity of a 13 amino acid tryptophan-rich peptide derived from a putative porcine precursor protein of a novel family of antibacterial peptides. FEBS Lett. 1996, 390, 95–98.
  204. Arias, M.; Hoffarth, E.R.; Ishida, H.; Aramini, J.M.; Vogel, H.J. Recombinant expression, antimicrobial activity and mechanism of action of tritrpticin analogs containing fluoro-tryptophan residues. Biochim. Biophys. Acta 2016, 1858, 1012–1023.
  205. Wong, C.Y.; Eftink, M.R. Incorporation of tryptophan analogues into staphylococcal nuclease, its V66W mutant, and Δ137-149 fragment: Spectroscopic studies. Biochemistry 1998, 37, 8938–8946.
  206. Xu, Z.J.; Love, M.L.; Ma, L.Y.; Blum, M.; Bronskill, P.M.; Bernstein, J.; Grey, A.A.; Hofmann, T.; Camerman, N.; Wong, J.T. Tryptophanyl-tRNA synthetase from Bacillus subtilis. Characterization and role of hydrophobicity in substrate recognition. J. Biol. Chem. 1989, 264, 4304–4311.
  207. Cotten, M.; Tian, C.; Busath, D.D.; Shirts, R.B.; Cross, T.A. Modulating dipoles for structure-function correlations in the gramicidin A channel. Biochemistry 1999, 38, 9185–9197.
  208. Glossop, H.D.; De Zoysa, G.H.; Pilkington, L.I.; Barker, D.; Sarojini, V. Fluorinated O-phenylserine residues enhance the broad-spectrum antimicrobial activity of ultrashort cationic lipopeptides. J. Fluor. Chem. 2021, 241, 109685.
  209. De Zoysa, G.H.; Cameron, A.J.; Hegde, V.V.; Raghothama, S.; Sarojini, V. Antimicrobial peptides with potential for biofilm eradication: Synthesis and structure activity relationship studies of battacin peptides. J. Med. Chem. 2015, 58, 625–639.
  210. Krenk, O.; Kratochvil, J.; Spulak, M.; Buchta, V.; Kunes, J.; Novakova, L.; Ghavre, M.; Pour, M. Methodology for Synthesis of Enantiopure 3,5-Disubstituted Pyrrol-2-ones. Eur. J. Org. Chem. 2015, 2015, 5414–5423.
  211. Molchanova, N.; Hansen, P.R.; Damborg, P.; Nielsen, H.M.; Franzyk, H. Lysine-Based α-Peptide/β-Peptoid Peptidomimetics: Influence of Hydrophobicity, Fluorination, and Distribution of Cationic Charge on Antimicrobial Activity and Cytotoxicity. ChemMedChem 2017, 12, 312–318.
  212. Molchanova, N.; Hansen, P.R.; Damborg, P.; Franzyk, H. Fluorinated antimicrobial lysine-based peptidomimetics with activity against methicillin-resistant Staphylococcus pseudintermedius. J. Pept. Sci. 2018, 24, e3098.
  213. Zuckermann, R.N.; Kerr, J.M.; Kent, S.B.H.; Moos, W.H. Efficient Method for the Preparation of Peptoids by Submonomer Solid-Phase Synthesis. J. Am. Chem. Soc. 1992, 114, 10646–10647.
  214. Patch, J.A.; Barron, A.E. Helical peptoid mimics of magainin-2 amide. J. Am. Chem. Soc. 2003, 125, 12092–12093.
  215. Ghosh, C.; Manjunath, G.B.; Akkapeddi, P.; Yarlagadda, V.; Hoque, J.; Uppu, D.S.S.M.; Konai, M.M.; Haldar, J. Small Molecular Antibacterial Peptoid Mimics: The Simpler the Better! J. Med. Chem. 2014, 57, 1428–1436.
  216. Bolt, H.L.; Eggimann, G.A.; Jahoda, C.A.B.; Zuckermann, R.N.; Sharples, G.J.; Cobb, S.L. Exploring the links between peptoid antibacterial activity and toxicity. Medchemcomm 2017, 8, 886–896.
  217. Molchanova, N.; Nielsen, J.E.; Sorensen, K.B.; Prabhala, B.K.; Hansen, P.R.; Lund, R.; Barron, A.E.; Jenssen, H. Halogenation as a tool to tune antimicrobial activity of peptoids. Sci. Rep. 2020, 10, 14805.
  218. Paulsen, M.H.; Engqvist, M.; Ausbacher, D.; Strom, M.B.; Bayer, A. Efficient and scalable synthesis of α,α-disubstituted β-amino amides. Org. Biomol. Chem. 2016, 14, 7570–7578.
  219. Paulsen, M.H.; Karlsen, E.A.; Ausbacher, D.; Anderssen, T.; Bayer, A.; Ochtrop, P.; Hedberg, C.; Haug, T.; Ericson Sollid, J.U.; Strom, M.B. An amphipathic cyclic tetrapeptide scaffold containing halogenated β(2,2) -amino acids with activity against multiresistant bacteria. J. Pept. Sci. 2018, 24, e3117.
  220. Paulsen, M.H.; Ausbacher, D.; Bayer, A.; Engqvist, M.; Hansen, T.; Haug, T.; Anderssen, T.; Andersen, J.H.; Sollid, J.U.E.; Strom, M.B. Antimicrobial activity of amphipathic α,α-disubstituted β-amino amide derivatives against ESBL—CARBA producing multi-resistant bacteria; effect of halogenation, lipophilicity and cationic character. Eur. J. Med. Chem. 2019, 183, 111671.
  221. Kasim, J.K.; Kavianinia, I.; Bull, M.; Harris, P.W.; Smaill, J.B.; Patterson, A.V.; Brimble, M.A. Fourth-Generation Analogues of the Anticancer Peptaibol Culicinin D: Probing the Effects of Hydrophobicity and Halogenation on Cytotoxicity. Synthesis 2021, 53, 4239–4245.
  222. Koch, K.; Afonin, S.; Ieronimo, M.; Berditsch, M.; Ulrich, A.S. Solid-state (19)F-NMR of peptides in native membranes. Top. Curr. Chem. 2012, 306, 89–118.
  223. Yu, T.T.; Kuppusamy, R.; Yasir, M.; Hassan, M.M.; Alghalayini, A.; Gadde, S.; Deplazes, E.; Cranfield, C.; Willcox, M.D.P.; Black, D.S.; et al. Design, Synthesis and Biological Evaluation of Biphenylglyoxamide-Based Small Molecular Antimicrobial Peptide Mimics as Antibacterial Agents. Int. J. Mol. Sci. 2020, 21, 6789.
  224. Gunasekaran, P.; Rajasekaran, G.; Han, E.H.; Chung, Y.H.; Choi, Y.J.; Yang, Y.J.; Lee, J.E.; Kim, H.N.; Lee, K.; Kim, J.S.; et al. Cationic Amphipathic Triazines with Potent Anti-bacterial, Anti-inflammatory and Anti-atopic Dermatitis Properties. Sci. Rep. 2019, 9, 1292.
  225. Gruss, H.; Sewald, N. Late-Stage Diversification of Tryptophan-Derived Biomolecules. Chemistry 2020, 26, 5328–5340.
  226. Kemker, I.; Schroder, D.C.; Feiner, R.C.; Muller, K.M.; Marion, A.; Sewald, N. Tuning the Biological Activity of RGD Peptides with Halotryptophansdagger. J. Med. Chem. 2021, 64, 586–601.
More