Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 33(4): 555-559, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35319881

ABSTRACT

Endogenous Staphylococcus aureus sortase A (SrtA) covalently incorporates cell wall anchored proteins equipped with a SrtA recognition motif (LPXTG) via a lipid II-dependent pathway into the staphylococcal peptidoglycan layer. Previously, we found that the endogenous S. aureus SrtA is able to recognize and process a variety of exogenously added synthetic SrtA substrates, including K(FITC)LPMTG-amide and K(FITC)-K-vancomycin-LPMTG-amide. These synthetic substrates are covalently incorporated into the bacterial peptidoglycan (PG) of S. aureus with varying efficiencies. In this study, we examined if native and synthetic substrates are processed by SrtA via the same pathway. Therefore, the effect of the lipid II inhibiting antibiotic bacitracin on the incorporation of native and synthetic SrtA substrates was assessed. Treatment of S. aureus with bacitracin resulted in a decreased incorporation of protein A in the bacterial cell wall, whereas incorporation of exogenous synthetic substrates was increased. These results suggest that natural and exogenous synthetic substrates are processed by S. aureus via different pathways.


Subject(s)
Peptidoglycan , Staphylococcus aureus , Amides , Aminoacyltransferases , Bacitracin/metabolism , Bacitracin/pharmacology , Bacterial Proteins/metabolism , Cysteine Endopeptidases , Fluorescein-5-isothiocyanate , Peptidoglycan/metabolism
2.
PLoS One ; 11(1): e0147401, 2016.
Article in English | MEDLINE | ID: mdl-26799839

ABSTRACT

The endogenous Staphylococcus aureus sortase A (SrtA) transpeptidase covalently anchors cell wall-anchored (CWA) proteins equipped with a specific recognition motif (LPXTG) into the peptidoglycan layer of the staphylococcal cell wall. Previous in situ experiments have shown that SrtA is also able to incorporate exogenous, fluorescently labelled, synthetic substrates equipped with the LPXTG motif (K(FITC)LPETG-amide) into the bacterial cell wall, albeit at high concentrations of 500 µM to 1 mM. In the present study, we have evaluated the effect of substrate modification on the incorporation efficiency. This revealed that (i) by elongation of LPETG-amide with a sequence of positively charged amino acids, derived from the C-terminal domain of physiological SrtA substrates, the incorporation efficiency was increased by 20-fold at 10 µM, 100 µM and 250 µM; (ii) Substituting aspartic acid (E) for methionine increased the incorporation of the resulting K(FITC)LPMTG-amide approximately three times at all concentrations tested; (iii) conjugation of the lipid II binding antibiotic vancomycin to K(FITC)LPMTG-amide resulted in the same incorporation levels as K(FITC)LPETG-amide, but much more efficient at an impressive 500-fold lower substrate concentration. These newly developed synthetic substrates can potentially find broad applications in for example the in situ imaging of bacteria; the incorporation of antibody recruiting moieties; the targeted delivery and covalent incorporation of antimicrobial compounds into the bacterial cell wall.


Subject(s)
Aminoacyltransferases/physiology , Bacterial Proteins/physiology , Cell Wall/metabolism , Cysteine Endopeptidases/physiology , Peptides/metabolism , Staphylococcus aureus/metabolism , Aminoacyltransferases/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Cell Wall/drug effects , Cysteine Endopeptidases/metabolism , Flow Cytometry , Microbial Sensitivity Tests , RNA Recognition Motif Proteins/metabolism , RNA Recognition Motif Proteins/physiology , Staphylococcus aureus/drug effects , Substrate Specificity , Vancomycin/pharmacology
3.
FEMS Yeast Res ; 15(8)2015 Dec.
Article in English | MEDLINE | ID: mdl-26392045

ABSTRACT

Saliva plays a key role in the maintenance of a stable oral microflora. It contains antimicrobial compounds but also functions as a substrate for growth of bacteria under conditions of low external nutrient supply. Besides bacteria, yeasts, in particular Candida albicans, commonly inhabit the oral cavity. Under immunocompromised conditions, instantaneous outgrowth of this yeast occurs in oral carriers of C. albicans, suggesting that this yeast is able to survive in the oral cavity with saliva as sole source of growth substrate. The aim of the present study was to identify the salivary constituents that are used by C. albicans for growth and survival in saliva. In addition, we have explored the effect of growth in saliva on the susceptibility of C. albicans to histatin 5, a salivary antifungal peptide. It was found that C. albicans was able to grow in human saliva without addition of glucose, and in the stationary phase could survive for more than 400 h. Candida albicans grown in saliva was more than 10 times less susceptible for salivary histatin 5 than C. albicans cultured in Sabouraud medium.


Subject(s)
Candida albicans/growth & development , Saliva/microbiology , Antifungal Agents/metabolism , Candida albicans/drug effects , Candida albicans/metabolism , Candida albicans/physiology , Histatins/metabolism , Humans , Microbial Viability , Time Factors
4.
Biol Chem ; 396(4): 283-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25581753

ABSTRACT

Backbone cyclization has a profound impact on the biological activity and thermal and proteolytic stability of proteins and peptides. Chemical methods for cyclization are not always feasible, especially for large peptides or proteins. Recombinant Staphylococcus aureus sortase A shows potential as a new tool for the cyclization of both proteins and peptides. In this review, the scope and background of the sortase-mediated cyclization are discussed. High efficiency, versatility, and easy access make sortase A a promising cyclization tool, both for recombinant and chemo-enzymatic production methods.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Peptides, Cyclic/metabolism , Peptides/metabolism , Proteins/metabolism , Staphylococcus aureus/enzymology , Amino Acid Sequence , Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Carbohydrate Sequence , Cyclization , Cysteine Endopeptidases/chemistry , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides, Cyclic/chemistry , Peptidoglycan/chemistry , Peptidoglycan/metabolism , Protein Conformation , Protein Engineering , Proteins/chemistry , Recombinant Proteins/metabolism , Staphylococcus aureus/metabolism
5.
Caries Res ; 49(1): 9-17, 2015.
Article in English | MEDLINE | ID: mdl-25300299

ABSTRACT

Calcium hydroxyapatite (HAp), the main constituent of dental enamel, is inherently susceptible to the etching and dissolving action of acids, resulting in tooth decay such as dental caries and dental erosion. Since the prevalence of erosive wear is gradually increasing, there is urgent need for agents that protect the enamel against erosive attacks. In the present study we studied in vitro the anti-erosive effects of a number of sphingolipids and sphingoid bases, which form the backbone of sphingolipids. Pretreatment of HAp discs with sphingosine, phytosphingosine (PHS), PHS phosphate and sphinganine significantly protected these against acid-induced demineralization by 80 ± 17%, 78 ± 17%, 78 ± 7% and 81 ± 8%, respectively (p < 0.001). On the other hand, sphingomyelin, acetyl PHS, octanoyl PHS and stearoyl PHS had no anti-erosive effects. Atomic force measurement revealed that HAp discs treated with PHS were almost completely and homogeneously covered by patches of PHS. This suggests that PHS and other sphingoid bases form layers on the surface of HAp, which act as diffusion barriers against H(+) ions. In principle, these anti-erosive properties make PHS and related sphingosines promising and attractive candidates as ingredients in oral care products.


Subject(s)
Durapatite/chemistry , Protective Agents/chemistry , Sphingolipids/chemistry , Tooth Erosion/metabolism , Adsorption , Citric Acid/chemistry , Dental Pellicle/chemistry , Diffusion , Edetic Acid/chemistry , Humans , Hydrogen-Ion Concentration , Materials Testing , Microscopy, Atomic Force , Protein Kinase C/antagonists & inhibitors , Sphingomyelins/chemistry , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Surface Properties , Time Factors
6.
Monogr Oral Sci ; 24: 40-51, 2014.
Article in English | MEDLINE | ID: mdl-24862593

ABSTRACT

The oral cavity is one of the most heavily colonized parts of our body. The warm, nutrient-rich and moist environment promotes the growth of a diverse microflora. One of the factors responsible for the ecological equilibrium in the mouth is saliva, which in several ways affects the colonization and growth of bacteria. In this paper, we discuss the various mechanisms by which the composition of the oral microflora is modulated by saliva. Saliva covers the oral hard and soft tissues with a conditioning film which governs the initial attachment of microorganisms, a crucial step in the setup of the oral microflora. It furthermore contains proteins which in the soluble phase bind to bacteria, blocking their adherence to surfaces. When the supply of nutrients is diminished, bacteria use salivary glycoproteins, especially high-molecular-weight mucins, as a source of complex carbohydrates, requiring a consortium of microorganisms for breakdown. In this way saliva promotes the complexity of the oral microflora, which in itself protects against overgrowth by few pathogenic species. Finally, saliva harbors a large panel of antimicrobial proteins which directly and indirectly inhibit uncontrolled outgrowth of bacteria. These include lactoferrin, lactoperoxidase, lysozyme and antimicrobial peptides. Under pathological conditions serum leakage occurs, and saliva mobilizes the humoral and cellular defense mechanisms in the blood. In sum, saliva favors the establishment of a highly diverse microflora, rather than a semisterile environment.


Subject(s)
Saliva/physiology , Antimicrobial Cationic Peptides/physiology , Bacteria/growth & development , Bacteria/metabolism , Bacterial Adhesion/physiology , Humans , Microbial Consortia/physiology , Mouth/microbiology , Saliva/microbiology , Salivary Proteins and Peptides/metabolism , Salivary Proteins and Peptides/physiology
7.
Biochem Cell Biol ; 90(3): 378-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22364313

ABSTRACT

Bovine lactoferrin harbors 2 antimicrobial sequences (LFcin and LFampin), situated in close proximity in the N1-domain. To mimic their semi parallel configuration we have synthesized a chimeric peptide (LFchimera) in which these sequences are linked in a head-to-head fashion to the α- and ε-amino group, respectively, of a single lysine. In line with previously described bactericidal effects, this peptide was also a stronger candidacidal agent than the antimicrobial peptides LFcin17-30 and LFampin265-284, or a combination of these 2. Conditions that strongly reduced the candidacidal activities of LFcin17-30 and LFampin265-284, such as high ionic strength and energy depletion, had little influence on the activity of LFchimera. Freeze-fracture electron microscopy showed that LFchimera severely affected the membrane morphology, resulting in disintegration of the membrane bilayer and in an efflux of small and high molecular weight molecules such as ATP and proteins. The differential effects displayed by the chimeric peptide and a mixture of its constituent peptides clearly demonstrate the synergistic effect of linking these peptides in a fashion that allows a similar spatial arrangement as in the parent protein, suggesting that in bovine lactoferrrin the corresponding fragments act in concert in its candidacidal activity.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Lactoferrin/pharmacology , Peptide Fragments/pharmacology , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Candida albicans/metabolism , Candida albicans/ultrastructure , Cattle , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Fluorescent Dyes/metabolism , Fungal Proteins/metabolism , Molecular Sequence Data , Permeability , Propidium/metabolism , Protein Structure, Secondary
8.
FASEB J ; 25(8): 2650-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21525488

ABSTRACT

Cyclic peptides are highly valued tools in biomedical research. In many cases, they show higher receptor affinity, enhanced biological activity, and improved serum stability. Technical difficulties in producing cyclic peptides, especially larger ones, in appreciable yields have precluded a prolific use in biomedical research. Here, we describe a novel and efficient cyclization method that uses the peptidyl-transferase activity of the Staphylococcus aureus enzyme sortase A to cyclize linear synthetic precursor peptides. As a model, we used histatin 1, a 38-mer salivary peptide with motogenic activity. Chemical cyclization of histatin 1 resulted in ≤ 3% yields, whereas sortase-mediated cyclization provided a yield of >90%. The sortase-cyclized peptide displayed a maximum wound closure activity at 10 nM, whereas the linear peptide displayed maximal activity at 10 µM. Circular dichroism and NMR spectroscopic analysis of the linear and cyclic peptide in solution showed no evidence for conformational changes, suggesting that structural differences due to cyclization only became manifest when these peptides were located in the binding domain of the receptor. The sortase-based cyclization technology provides a general method for easy and efficient manufacturing of large cyclic peptides.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Histatins/biosynthesis , Peptides, Cyclic/biosynthesis , Amino Acid Sequence , Aminoacyltransferases/genetics , Bacterial Proteins/genetics , Cell Line , Circular Dichroism , Cysteine Endopeptidases/genetics , Histatins/chemistry , Histatins/genetics , Histatins/pharmacology , Humans , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptides, Cyclic/chemistry , Peptides, Cyclic/genetics , Peptides, Cyclic/pharmacology , Protein Conformation , Protein Engineering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Wound Healing/drug effects
9.
Biol Chem ; 391(1): 65-71, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19919184

ABSTRACT

The mechanism of action of phytosphingosine (PHS), a member of the sphingosine family which has candidacidal activity when added externally, was investigated. Previously, it has been reported that the fungicidal activity of PHS is based on the induction of caspase-independent apoptosis. In contrast, we found that addition of PHS causes a direct permeabilization of the plasma membrane of yeast, highlighted by the influx of the membrane probe propidium iodide, and the efflux of small molecules (i.e., adenine nucleotides) as well as large cellular constituents such as proteins. Freeze-fracture electron microscopy revealed that PHS treatment causes severe damage of the plasma membrane of the cell, which seems to have lost its integrity completely. We also found that PHS reverts the azide-induced insensitivity to histatin 5 (Hst5) of Candida albicans. In a previous study, we had found that the decreased sensitivity to Hst5 of energy-depleted cells is due to rigidification of the plasma membrane, which could be reverted by the membrane fluidizer benzyl alcohol. In line with the increased membrane permeabilization and ultrastructural damage, this reversal of the azide-induced insensitivity by PHS also points to a direct interaction between PHS and the cytoplasmic membrane of C. albicans.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Cell Membrane/drug effects , Sphingosine/analogs & derivatives , Candida albicans/ultrastructure , Cell Membrane Permeability/drug effects , Histatins/pharmacology , Sodium Azide/pharmacology , Sphingosine/pharmacology
10.
FASEB J ; 23(11): 3928-35, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19652025

ABSTRACT

Wounds in the mouth heal faster and with less scarification and inflammation than those in the skin. Saliva is thought to be essential for the superior oral wound healing, but the involved mechanism is still unclear. We have previously discovered that a human-specific peptide, histatin, might be implicated in the wound-healing properties of saliva. Here we report that histatin enhances reepithelialization in a human full-skin wound model closely resembling normal skin. The peptide does not stimulate proliferation but induces cell spreading and migration, two key initiating steps in reepithelialization. Activation of cells by histatin requires a G-protein-coupled receptor that activates the ERK1/2 pathway. Using a stepwise-truncation method, we determined the minimal domain (SHREFPFYGDYGS) of the 38-mer-parent peptide that is required for activity. Strikingly, N- to C-terminal cyclization of histatin-1 potentiates the molar activity approximately 1000-fold, indicating that the recognition of histatin by its cognate receptor requires a specific spatial conformation of the peptide. Our results emphasize the importance of histatin in human saliva for tissue protection and recovery and establish the experimental basis for the development of synthetic histatins as novel skin wound-healing agents.


Subject(s)
Histatins/physiology , Wound Healing/physiology , Amino Acid Sequence , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclization , Drug Synergism , Humans , Infant, Newborn , Keratinocytes/drug effects , Male , Models, Biological , Saliva/chemistry , Structure-Activity Relationship , Wound Healing/drug effects
11.
Biochimie ; 91(1): 123-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18573310

ABSTRACT

The innate immunity factor lactoferrin harbours two antimicrobial moieties, lactoferricin and lactoferrampin, situated in close proximity in the N1 domain of the molecule. Most likely they cooperate in many of the beneficial activities of lactoferrin. To investigate whether chimerization of both peptides forms a functional unit we designed a chimerical structure containing lactoferricin amino acids 17-30 and lactoferrampin amino acids 265-284. The bactericidal activity of this LFchimera was found to be drastically stronger than that of the constituent peptides, as was demonstrated by the need for lower dose, shorter incubation time and less ionic strength dependency. Likewise, strongly enhanced interaction with negatively charged model membranes was found for the LFchimera relative to the constituent peptides. Thus, chimerization of the two antimicrobial peptides resembling their structural orientation in the native molecule strikingly improves their biological activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Lactoferrin/pharmacology , Lactoglobulins/pharmacology , Peptide Fragments/pharmacology , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Calorimetry, Differential Scanning , Circular Dichroism , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Lactoferrin/chemistry , Lactoferrin/genetics , Lactoglobulins/chemistry , Lactoglobulins/genetics , Microbial Sensitivity Tests , Osmolar Concentration , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptides/chemistry , Peptides/genetics , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
12.
FASEB J ; 22(11): 3805-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18650243

ABSTRACT

Wounds in the oral cavity heal much faster than skin lesions. Among other factors, saliva is generally assumed to be of relevance to this feature. Rodent saliva contains large amounts of growth factors such as epidermal growth factor (EGF) and nerve growth factor (NGF). In humans, however, the identity of the involved compounds has remained elusive, especially since EGF and NGF concentrations are approximately 100,000 times lower than those in rodent saliva. Using an in vitro model for wound closure, we examined the properties of human saliva and the fractions that were obtained from saliva by high-performance liquid chromotography (HPLC) separation. We identified histatin 1 (Hst1) and histatin 2 (Hst2) as major wound-closing factors in human saliva. In contrast, the d-enantiomer of Hst2 did not induce wound closure, indicating stereospecific activation. Furthermore, histatins were actively internalized by epithelial cells and specifically used the extracellular signal-regulated kinases 1/2 (ERK1/2) pathway, thereby enhancing epithelial migration. This study demonstrates that members of the histatin family, which up to now were implicated in the antifungal weaponry of saliva, exert a novel function that likely is relevant for oral wound healing.


Subject(s)
Histatins/metabolism , Mouth/metabolism , Saliva/metabolism , Salivary Proteins and Peptides/metabolism , Wound Healing/physiology , Animals , Biological Assay/methods , Cell Line , Histatins/chemistry , Histatins/pharmacology , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mouth/chemistry , Rodentia , Saliva/chemistry , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/isolation & purification , Salivary Proteins and Peptides/pharmacology , Species Specificity , Wound Healing/drug effects
13.
FASEB J ; 22(6): 1817-28, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18230684

ABSTRACT

Histatin 5 (Hst5) is a human salivary antimicrobial peptide that targets fungal mitochondria. In the human parasitic protozoa Leishmania, the mitochondrial ATP production is essential, as it lacks the bioenergetic switch between glycolysis and oxidative phosphorylation described in some yeasts. On these premises, Hst5 activity was assayed on both stages of its life cycle, promastigotes and amastigotes (LC(50)=7.3 and 14.4 microM, respectively). In a further step, its lethal mechanism was studied. The main conclusions drawn were as follows: 1) Hst5 causes limited and temporary damage to the plasma membrane of the parasites, as assessed by electron microscopy, depolarization, and entrance of the vital dye SYTOX Green; 2) Hst5 translocates into the cytoplasm of Leishmania in an achiral receptor-independent manner with accumulation into the mitochondrion, as shown by confocal microscopy; and 3) Hst5 produces a bioenergetic collapse of the parasite, caused essentially by the decrease of mitochondrial ATP synthesis through inhibition of F(1)F(0)-ATPase, with subsequent fast ATP exhaustion. By using the Hst5 enantiomer, it was found that the key steps of its lethal mechanism involved no chiral recognition. Hst5 thus constitutes the first leishmanicidal peptide with a defined nonstereospecific intracellular target. The prospects of its development, by its own or as a carrier molecule for other leishmanicidal molecules, into a novel anti-Leishmania drug with a preferential subcellular accumulation are discussed.


Subject(s)
Adenosine Triphosphate/biosynthesis , Histatins/pharmacology , Leishmania/drug effects , Proton-Translocating ATPases/antagonists & inhibitors , Animals , Antimicrobial Cationic Peptides , Antiprotozoal Agents , Cell Membrane Permeability , Humans , Leishmania/metabolism , Mitochondria/metabolism
14.
Antimicrob Agents Chemother ; 51(9): 3416-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17620386

ABSTRACT

Homodimerization of histatin-derived peptides generally led to improved bactericidal activity against Staphylococcus aureus in vitro. In vivo, monomers and dimers were equally active in killing bacteria in mice with a soft tissue infection. Altogether, these peptides are promising compounds for the development of novel therapeutics against infections with drug-resistant bacteria.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/pharmacokinetics , Drug Resistance, Multiple, Bacterial , Methicillin Resistance/drug effects , Mice , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Tissue Distribution
15.
J Biol Chem ; 282(26): 18831-41, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17485465

ABSTRACT

Inhibitors of the energy metabolism, such as sodium azide and valinomycin, render yeast cells completely resistant against the killing action of a number of cationic antimicrobial peptides, including the salivary antimicrobial peptide Histatin 5. In this study the Histatin 5-mediated killing of the opportunistic yeast Candida albicans was used as a model system to comprehensively investigate the molecular basis underlying this phenomenon. Using confocal and electron microscopy it was demonstrated that the energy poison azide reversibly blocked the entry of Histatin 5 at the level of the yeast cell wall. Azide treatment hardly induced depolarization of the yeast cell membrane potential, excluding it as a cause of the lowered sensitivity. In contrast, the diminished sensitivity to Histatin 5 of energy-depleted C. albicans was restored by increasing the fluidity of the membrane using the membrane fluidizer benzyl alcohol. Furthermore, rigidification of the membrane by incubation at low temperature or in the presence of the membrane rigidifier Me(2)SO increased the resistance against Histatin 5, while not affecting the energy charge of the cell. In line, azide induced alterations in the physical state of the interior of the lipid bilayer. These data demonstrate that changes in the physical state of the membrane underlie the increased resistance to antimicrobial peptides.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Candida albicans/drug effects , Candida albicans/metabolism , Energy Metabolism/physiology , Antifungal Agents/pharmacology , Azides/pharmacology , Benzyl Alcohol/pharmacology , Candida albicans/ultrastructure , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cytoskeleton/drug effects , Depsipeptides/pharmacology , Drug Resistance, Fungal , Energy Metabolism/drug effects , Histatins , Membrane Fluidity/drug effects , Membrane Fluidity/physiology , Microscopy, Confocal , Microscopy, Electron , Salivary Proteins and Peptides/metabolism , Salivary Proteins and Peptides/pharmacology
16.
Biol Chem ; 387(10-11): 1495-502, 2006.
Article in English | MEDLINE | ID: mdl-17081124

ABSTRACT

The human cathelicidin peptide LL-37 and several truncated variants differ in their capability to transmigrate over the plasma membrane of Candida albicans. We investigated whether retention at the cell perimeter or membrane transmigration affects their membrane-disrupting activities and candidacidal properties. Using fluorescein-labeled peptides, we demonstrate that LL-37 and its C-terminally truncated peptide LL-31 remain permanently associated with the perimeter of the cell. The N-terminally truncated peptide RK-31 initially accumulated at the cell boundary, but transmigrated into the cytoplasm within 30 min. The C-terminally truncated peptide LL-25 transmigrated instantaneously into the cytoplasm. The ultrastructural effects on the plasma membrane were studied by freeze-fracture electron microscopy combined with filipin cytochemistry. All peptides, whether they transmigrated over the plasma membrane or not, induced phase separation in the plasma membrane. All peptides induced leakage of cell components, including nucleotides and proteins. Proteins were identified by SDS-PAGE in combination with mass spectrometry, which revealed that predominantly proteins smaller than 50 kDa had leaked out of C. albicans.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Candida albicans/metabolism , Cell Membrane/metabolism , Genetic Variation/genetics , Lipid Metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Candida albicans/ultrastructure , Cell Membrane/ultrastructure , Cell Membrane Permeability , Humans , Microscopy, Electron, Transmission , Molecular Sequence Data , Nucleotides/metabolism , Sequence Alignment , Cathelicidins
17.
Biochem Cell Biol ; 84(3): 358-62, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16936807

ABSTRACT

Two lactoferrampin (LFampin) peptides derived from bovine lactoferrin were compared with respect to their bactericidal activities. LFampin 265-284 killed a set of Gram-positive bacteria that were resistant to LFampin 268-284. The presence of 265Asp-Leu-267Ile did not simply lead to an overall increased potency, since higher concentrations of LFampin 265-284 than LFampin 268-284 were needed to kill the Gram-negative bacteria that were tested. The Asp-Leu-Ile sequence enhances the propensity of LFampin to adopt an alpha-helix, as shown by circular dichroism spectroscopy. These results suggest that the helical conformation of the peptide is an important determinant of the susceptibility of Gram-positive bacteria.


Subject(s)
Anti-Bacterial Agents/metabolism , Lactoferrin/metabolism , Lactoglobulins/metabolism , Peptide Fragments/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , Cattle , Circular Dichroism , Lactoferrin/chemistry , Lactoglobulins/chemistry , Microbial Sensitivity Tests , Molecular Sequence Data , Peptide Fragments/chemistry , Peptides/chemistry
18.
Peptides ; 27(1): 1-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16087276

ABSTRACT

Antimicrobial peptides have been found throughout living nature, yet antimicrobial sequences may still lie hidden within a wide variety of proteins. A rational strategy was developed to select interesting domains, based on the presumed common features of antimicrobial peptides, and to release these from accessible and safe proteins. In silico proteolysis simulations of bovine lactoferrin (bLF) with selected endoproteinases predicted the liberation of peptides that encompasses a cationic amphipathic alpha-helix. Three predicted peptides were synthesized and tested for their biological activity, demonstrating that one single enzyme was sufficient to obtain an antimicrobial peptide. The proof of principle demonstrated that a 32-mer fragment isolated from the endoproteinase AspN digestion of bLF possessed strong antimicrobial activity. Moreover, desalted crude digest had improved activity over native bLF. Hence, selective digestion of bLF increases its antimicrobial activity by release of antimicrobial stretches.


Subject(s)
Anti-Infective Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Lactoferrin/metabolism , Lactoglobulins/metabolism , Metalloendopeptidases/physiology , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Candida albicans , Cattle , Computational Biology , Escherichia coli , Lactoferrin/chemistry , Lactoglobulins/chemistry , Metalloendopeptidases/chemistry , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Structure, Tertiary
19.
Peptides ; 26(9): 1537-42, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16112390

ABSTRACT

Antimicrobial peptides allegedly exert their action on microbial membranes. Bovine lactoferrin enfold two antimicrobial domains, lactoferricin B (LFcin B) and lactoferrampin (LFampin). Effects of representative peptides thereof on the membranes of Candida albicans and Escherichia coli were investigated. Confocal laser scanning microscopy revealed that these peptides were internalized within a few minutes, concurrently with disrupting membrane integrity as indicated by freeze-fracture transmission electron microscopy. The most striking findings were induction of distinct vesicle-like structures in the membrane of C. albicans by the LFampin peptide, and detachment of the outer membrane and surface protrusions in E. coli by the LFcin B peptide.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Candida albicans/drug effects , Cell Membrane/drug effects , Escherichia coli/drug effects , Lactoferrin/chemistry , Animals , Candida albicans/cytology , Candida albicans/ultrastructure , Cattle , Cell Division/drug effects , Cell Membrane/ultrastructure , Escherichia coli/cytology , Escherichia coli/ultrastructure , Fluorescein-5-isothiocyanate/chemistry , Freeze Fracturing , Lactoglobulins/chemistry , Microscopy, Confocal , Microscopy, Electron/methods , Peptide Fragments/chemistry , Peptide Fragments/pharmacology
20.
Peptides ; 26(11): 2093-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15946771

ABSTRACT

LFampin 265-284, derived from bovine lactoferrin, has broad-spectrum antimicrobial activity against the yeast Candida albicans and several Gram-positive and Gram-negative bacteria. A glycine substitution scan was used to identify residues that are important for its candidacidal activity. Each single substitution of a positively charged residue led to considerable reduction in candidacidal activity, for each residue to a different extent. Substitution within the helix-facilitating N-terminal sequence DLIW had less severe effect; substitution of Ile and Trp led to a somewhat reduced potency. No substantial effects were found on the propensity to adopt a helical structure or to bind to C. albicans cells.


Subject(s)
Amino Acid Substitution , Antifungal Agents , Candida albicans/drug effects , Lactoferrin/genetics , Peptide Fragments/genetics , Peptides/genetics , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Candida albicans/growth & development , Cattle , Lactoferrin/chemistry , Lactoferrin/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/chemistry , Peptides/pharmacology , Protein Structure, Secondary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...