Structure-Microbicidal Activity Relationship of Synthetic Fragments Derived from the Antibacterial α-Helix of Human Lactoferrin

ABSTRACT There is a need for new microbicidal agents with therapeutic potential due to antibiotic resistance in bacteria and fungi. In this study, the structure-microbicidal activity relationship of amino acid residues 14 to 31 (sequence 14-31) from the N-terminal end, corresponding to the antibacterial α-helix of human lactoferrin (LF), was investigated by downsizing, alanine scanning, and substitution of amino acids. Microbicidal analysis (99% killing) was performed by a microplate assay using Escherichia coli, Staphylococcus aureus, and Candida albicans as test organisms. Starting from the N-terminal end, downsizing of peptide sequence 14-31 showed that the peptide sequence 19-31 (KCFQWQRNMRKVR, HL9) was the optimal length for antimicrobial activity. Furthermore, HL9 bound to lipid A/lipopolysaccharide, as shown by neutralizing endotoxic activity in a Limulus assay. Alanine scanning of peptide sequence 20-31 showed that Cys20, Trp23, Arg28, Lys29, or Arg31 was important for expressing full killing activity, particularly against C. albicans. Substituting the neutral hydrophilic amino acids Gln24 and Asn26 for Lys and Ala (HLopt2), respectively, enhanced microbicidal activity significantly against all test organisms compared to the amino acids natural counterpart, also, in comparison with HL9, HLopt2 had more than 10-fold-stronger fungicidal activity. Furthermore, HLopt2 was less affected by metallic salts than HL9. The microbicidal activity of HLopt2 was slightly reduced only at pH 7.0, as tested in the pH range of 4.5 to 7.5. The results showed that the microbicidal activity of synthetic peptide sequences, based on the antimicrobial α-helix region of LF, can be significantly enhanced by optimizing the length and substitution of neutral amino acids at specific positions, thus suggesting a sequence lead with therapeutic potential.

[1]  D. Morrison,et al.  Binding of polymyxin B to the lipid A portion of bacterial lipopolysaccharides. , 1976, Immunochemistry.

[2]  R. Ellison,et al.  Damage of the outer membrane of enteric gram-negative bacteria by lactoferrin and transferrin , 1988, Infection and immunity.

[3]  D. Rice,et al.  Structure of human lactoferrin: crystallographic structure analysis and refinement at 2.8 A resolution. , 1989, Journal of molecular biology.

[4]  K. Yamauchi,et al.  Identification of the bactericidal domain of lactoferrin. , 1992, Biochimica et biophysica acta.

[5]  A. S. Naidu,et al.  Relationship between antibacterial activity and porin binding of lactoferrin in Escherichia coli and Salmonella typhimurium , 1993, Antimicrobial Agents and Chemotherapy.

[6]  K. Yamauchi,et al.  Antibacterial activity of lactoferrin and a pepsin-derived lactoferrin peptide fragment , 1993, Infection and immunity.

[7]  B. Appelmelk,et al.  Lactoferrin is a lipid A-binding protein , 1994, Infection and immunity.

[8]  D. Mann,et al.  Delineation of the glycosaminoglycan-binding site in the human inflammatory response protein lactoferrin. , 1994, The Journal of biological chemistry.

[9]  V. Salmon,et al.  Lactoferrin-lipopolysaccharide interaction: involvement of the 28-34 loop region of human lactoferrin in the high-affinity binding to Escherichia coli 055B5 lipopolysaccharide. , 1995, The Biochemical journal.

[10]  R. W. Evans,et al.  Antibacterial activity of peptides homologous to a loop region in human lactoferrin , 1996, FEBS letters.

[11]  E. Valore,et al.  Broad-spectrum antimicrobial activity of human intestinal defensin 5 , 1997, Infection and immunity.

[12]  R. Hancock Peptide antibiotics , 1997, The Lancet.

[13]  James M. Wilson,et al.  Human β-Defensin-1 Is a Salt-Sensitive Antibiotic in Lung That Is Inactivated in Cystic Fibrosis , 1997, Cell.

[14]  D. J. Mason,et al.  A helical region on human lactoferrin. Its role in antibacterial pathogenesis. , 1998, Advances in experimental medicine and biology.

[15]  D. J. Mason,et al.  A Helical Region on Human Lactoferrin , 1998 .

[16]  H. Ostolaza,et al.  Permeabilizing action of an antimicrobial lactoferricin‐derived peptide on bacterial and artificial membranes , 1999, FEBS letters.

[17]  D. Mann,et al.  Neutralization of Endotoxin In Vitro and In Vivo by a Human Lactoferrin-Derived Peptide , 1999, Infection and Immunity.

[18]  T. Gutteberg,et al.  Antibacterial effects of lactoferricin B. , 1999, Scandinavian journal of infectious diseases.

[19]  L. Vorland Lactoferrin: A multifunctional glycoprotein , 1999, APMIS : acta pathologica, microbiologica, et immunologica Scandinavica.

[20]  I. Mattsby‐Baltzer,et al.  Human Lactoferrin and Peptides Derived from a Surface-Exposed Helical Region Reduce Experimental Escherichia coli Urinary Tract Infection in Mice , 2000, Infection and Immunity.

[21]  P. Nibbering,et al.  Candidacidal Activities of Human Lactoferrin Peptides Derived from the N Terminus , 2000, Antimicrobial Agents and Chemotherapy.

[22]  E. Pauwels,et al.  Human Lactoferrin and Peptides Derived from Its N Terminus Are Highly Effective against Infections with Antibiotic-Resistant Bacteria , 2001, Infection and Immunity.

[23]  B. Ohlsson,et al.  Lactoferrin down-regulates the LPS-induced cytokine production in monocytic cells via NF-κB , 2002 .

[24]  B. Ohlsson,et al.  Lactoferrin down-regulates the LPS-induced cytokine production in monocytic cells via NF-kappa B. , 2002, Cellular immunology.

[25]  H. Vogel,et al.  Tryptophan-rich antimicrobial peptides: comparative properties and membrane interactions. , 2002, Biochemistry and cell biology = Biochimie et biologie cellulaire.

[26]  H. Vogel,et al.  Towards a structure-function analysis of bovine lactoferricin and related tryptophan- and arginine-containing peptides. , 2002, Biochemistry and cell biology = Biochimie et biologie cellulaire.

[27]  Lorna J. Smith,et al.  Long-Range Interactions Within a Nonnative Protein , 2002, Science.

[28]  Po-Wen Chen,et al.  Antibacterial activity of short hydrophobic and basic-rich peptides. , 2003, American journal of veterinary research.

[29]  I. Mattsby‐Baltzer,et al.  Anti‐Inflammatory Activities of Human Lactoferrin in Acute Dextran Sulphate‐Induced Colitis in Mice , 2003, Scandinavian journal of immunology.

[30]  K. Crowhurst,et al.  Aromatic and methyl NOEs highlight hydrophobic clustering in the unfolded state of an SH3 domain. , 2003, Biochemistry.

[31]  R. W. Evans,et al.  Variation in antimicrobial activity of lactoferricin-derived peptides explained by structure modelling. , 2004, FEMS microbiology letters.

[32]  V. Gant,et al.  Interactions of lactoferricin-derived peptides with LPS and antimicrobial activity. , 2004, FEMS microbiology letters.

[33]  M. Dathe,et al.  Antimicrobial activity of arginine- and tryptophan-rich hexapeptides: the effects of aromatic clusters, D-amino acid substitution and cyclization. , 2004, The journal of peptide research : official journal of the American Peptide Society.

[34]  Robert E. W. Hancock,et al.  Structure and Association of Human Lactoferrin Peptides with Escherichia coli Lipopolysaccharide , 2004, Antimicrobial Agents and Chemotherapy.

[35]  J. Pérez-Gil,et al.  Potassium Efflux Induced by a New Lactoferrin-Derived Peptide Mimicking the Effect of Native Human Lactoferrin on the Bacterial Cytoplasmic Membrane , 2003, Biochemistry (Moscow).

[36]  H. Vogel,et al.  Human Lactoferricin Is Partially Folded in Aqueous Solution and Is Better Stabilized in a Membrane Mimetic Solvent , 2005, Antimicrobial Agents and Chemotherapy.

[37]  U. Joosten,et al.  Comparable Efficacies of the Antimicrobial Peptide Human Lactoferrin 1-11 and Gentamicin in a Chronic Methicillin-Resistant Staphylococcus aureus Osteomyelitis Model , 2005, Antimicrobial Agents and Chemotherapy.

[38]  Andreja Majerle,et al.  Structural Origin of Endotoxin Neutralization and Antimicrobial Activity of a Lactoferrin-based Peptide* , 2005, Journal of Biological Chemistry.

[39]  A. V. Amerongen,et al.  Histatin and lactoferrin derived peptides: Antimicrobial properties and effects on mammalian cells , 2005, Peptides.

[40]  T. Osaki,et al.  Antimicrobial peptides enhance the candidacidal activity of antifungal drugs by promoting the efflux of ATP from Candida cells. , 2006, The Journal of antimicrobial chemotherapy.