Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Appl Environ Microbiol ; 85(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30824455

ABSTRACT

Multidrug-resistant bacterial pathogens are becoming increasingly prevalent, and novel strategies to treat bacterial infections caused by these organisms are desperately needed. Bacterial central metabolism is crucial for catabolic processes and provides precursors for anabolic pathways, such as the biosynthesis of essential biomolecules like amino acids or vitamins. However, most essential pathways are not regarded as good targets for antibiotic therapy since their products might be acquired from the environment. This issue raises doubts about the essentiality of such targets during infection. A putative target in bacterial anabolism is the methionine biosynthesis pathway. In contrast to humans, almost all bacteria carry methionine biosynthesis pathways which have often been suggested as putative targets for novel anti-infectives. While the growth of methionine auxotrophic strains can be stimulated by exogenous methionine, the extracellular concentrations required by most bacterial species are unknown. Furthermore, several phenotypic characteristics of methionine auxotrophs are only partly reversed by exogenous methionine. We investigated methionine auxotrophic mutants of Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli (all differing in methionine biosynthesis enzymes) and found that each needed concentrations of exogenous methionine far exceeding that reported for human serum (∼30 µM). Accordingly, these methionine auxotrophs showed a reduced ability to proliferate in human serum. Additionally, S. aureus and P. aeruginosa methionine auxotrophs were significantly impaired in their ability to form and maintain biofilms. Altogether, our data show intrinsic defects of methionine auxotrophs. This result suggests that the pathway should be considered for further studies validating the therapeutic potential of inhibitors.IMPORTANCE New antibiotics that attack novel targets are needed to circumvent widespread resistance to conventional drugs. Bacterial anabolic pathways, such as the enzymes for biosynthesis of the essential amino acid methionine, have been proposed as potential targets. However, the eligibility of enzymes in these pathways as drug targets is unclear because metabolites might be acquired from the environment to overcome inhibition. We investigated the nutritional needs of methionine auxotrophs of the pathogens Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli We found that each auxotrophic strain retained a growth disadvantage at methionine concentrations mimicking those available in vivo and showed that biofilm biomass was strongly influenced by endogenous methionine biosynthesis. Our experiments suggest that inhibition of the methionine biosynthesis pathway has deleterious effects even in the presence of external methionine. Therefore, additional efforts to validate the effects of methionine biosynthesis inhibitors in vivo are warranted.


Subject(s)
Biofilms/drug effects , Escherichia coli/physiology , Methionine/deficiency , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/physiology , Escherichia coli/drug effects , Escherichia coli/genetics , Mutation , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
2.
Eur J Clin Microbiol Infect Dis ; 36(3): 467-478, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27816993

ABSTRACT

FimH-mediated adhesion of Escherichia coli to bladder epithelium is a prerequisite for urinary tract infections. FimH is also essential for blood-borne bacterial dissemination, but the mechanisms are poorly understood. The purpose of this study was to assess the influence of different FimH mutations on bacterial adhesion using a novel adhesion assay, which models the physiological flow conditions bacteria are exposed to. We introduced 12 different point mutations in the mannose binding pocket of FimH in an E. coli strain expressing type 1 fimbriae only (MSC95-FimH). We compared the bacterial adhesion of each mutant across several commonly used adhesion assays, including agglutination of yeast, adhesion to mono- and tri-mannosylated substrates, and static adhesion to bladder epithelial and endothelial cells. We performed a comparison of these assays to a novel method that we developed to study bacterial adhesion to mammalian cells under flow conditions. We showed that E. coli MSC95-FimH adheres more efficiently to microvascular endothelium than to bladder epithelium, and that only endothelium supports adhesion at physiological shear stress. The results confirmed that mannose binding pocket mutations abrogated adhesion. We demonstrated that FimH residues E50 and T53 are crucial for adhesion under flow conditions. The coating of endothelial cells on biochips and modelling of physiological flow conditions enabled us to identify FimH residues crucial for adhesion. These results provide novel insights into screening methods to determine the effect of FimH mutants and potentially FimH antagonists.


Subject(s)
Adhesins, Escherichia coli/genetics , Bacterial Adhesion , Escherichia coli/genetics , Escherichia coli/physiology , Fimbriae Proteins/genetics , Point Mutation , Binding Sites , Cells, Cultured , Endothelial Cells/microbiology , Epithelial Cells/microbiology , Humans , Mannose-Binding Lectin/genetics
3.
J Bacteriol ; 193(16): 4134-42, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21665977

ABSTRACT

In many bacteria, including Staphylococcus aureus, progression from the logarithmic to the stationary phase is accompanied by conversion of most of bacterial membrane phosphatidylglycerol (PG) to cardiolipin (CL). Phagocytosis of S. aureus by human neutrophils also induces the conversion of most bacterial PG to CL. The genome of all sequenced strains of S. aureus contains two open reading frames (ORFs) predicting proteins encoded with ∼30% identity to the principal CL synthase (cls) of Escherichia coli. To test whether these ORFs (cls1 and cls2) encode cardiolipin synthases and contribute to CL accumulation in S. aureus, we expressed these proteins in a cls strain of E. coli and created isogenic single and double mutants in S. aureus. The expression of either Cls1 or Cls2 in CL-deficient E. coli resulted in CL accumulation in the stationary phase. S. aureus with deletion of both cls1 and cls2 showed no detectable CL accumulation in the stationary phase or after phagocytosis by neutrophils. CL accumulation in the stationary phase was due almost solely to Cls2, whereas both Cls1 and Cls2 contributed to CL accumulation following phagocytosis by neutrophils. Differences in the relative contributions of Cls1 and Cls2 to CL accumulation under different triggering conditions suggest differences in the role and regulation of these two enzymes.


Subject(s)
Cardiolipins/metabolism , Gene Expression Regulation, Bacterial/physiology , Membrane Proteins/metabolism , Phagocytes/metabolism , Staphylococcus aureus/enzymology , Transferases (Other Substituted Phosphate Groups)/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Enzymologic , Genome, Bacterial , Humans , Membrane Proteins/classification , Membrane Proteins/genetics , Molecular Sequence Data , Mutation , Neutrophils/metabolism , Phagocytes/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Transferases (Other Substituted Phosphate Groups)/classification , Transferases (Other Substituted Phosphate Groups)/genetics
4.
Int J Antimicrob Agents ; 34(1): 86-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19201578

ABSTRACT

Dermcidin (DCD) is an antimicrobial peptide constitutively expressed in eccrine sweat glands in human skin. By post-secretory proteolytic processing in sweat, the DCD protein gives rise to anionic and cationic DCD peptides that are able to kill several Gram-positive and Gram-negative bacteria but are only weakly active against Pseudomonas aeruginosa. Here, we questioned whether bacterial resistance to DCD peptides is mediated by proteolytic degradation. It was shown that DCD-derived peptides are degraded by purified bacterial proteases and by extracellular proteases secreted by P. aeruginosa in a concentration-dependent manner. However, protease-deficient mutants of P. aeruginosa PAO1 lacking either lasA, lasB (elastase) or both showed a similar sensitivity towards DCD-derived peptides as the wild-type strain. Finally, inhibition of total protease activity indicated that proteases secreted by P. aeruginosa are not responsible for the poor activity of DCD-derived peptides against P. aeruginosa. These data suggest that the decreased sensitivity of P. aeruginosa to DCD-derived peptides is not mediated by proteolytic degradation under physiological conditions.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Peptides/pharmacology , Pseudomonas aeruginosa/drug effects , Bacterial Proteins/genetics , Gene Deletion , Humans , Metalloendopeptidases/genetics , Microbial Sensitivity Tests , Peptide Hydrolases/genetics
5.
Curr Top Microbiol Immunol ; 306: 231-50, 2006.
Article in English | MEDLINE | ID: mdl-16909924

ABSTRACT

Cationic antimicrobial peptides (CAMPs) are integral compounds of the antimicrobial arsenals in virtually all kinds of organisms, with important roles in microbial ecology and higher organisms' host defense. Many bacteria have developed countermeasures to limit the efficacy of CAMPs such as defensins, cathelicidins, kinocidins, or bacteriocins. The best-studied bacterial CAMP resistance mechanisms involve electrostatic repulsion of CAMPs by modification of cell envelope molecules, proteolytic cleavage of CAMPs, production of CAMP-trapping proteins, or extrusion of CAMPs by energy-dependent efflux pumps. The repertoire of CAMPs produced by a given host organism and the efficiency of microbial CAMP resistance mechanisms appear to be crucial in host-pathogen interactions, governing the composition of commensal microbial communities and the virulence of bacterial pathogens. However, all CAMP resistance mechanisms have limitations and bacteria have never succeeded in becoming fully insensitive to a broad range of CAMPs. CAMPs or conserved CAMP resistance factors are discussed as new mediators and targets, respectively, of novel and sustainable anti-infective strategies.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Bacterial Infections/immunology , Animals , Carrier Proteins/metabolism , Drug Resistance, Bacterial , Humans , Static Electricity
6.
Antimicrob Agents Chemother ; 50(8): 2608-20, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16870749

ABSTRACT

Dermcidin (DCD) is a recently described antimicrobial peptide, which is constitutively expressed in eccrine sweat glands and transported via sweat to the epidermal surface. By postsecretory proteolytic processing in sweat the dermcidin protein gives rise to several truncated DCD peptides which differ in length and net charge. In order to understand the mechanism of antimicrobial activity, we analyzed the spectrum of activity of several naturally processed dermcidin-derived peptides, the secondary structure in different solvents, and the ability of these peptides to interact with or permeabilize the bacterial membrane. Interestingly, although all naturally processed DCD peptides can adopt an alpha-helical conformation in solvents, they have a diverse and partially overlapping spectrum of activity against gram-positive and gram-negative bacteria. This indicates that the net charge and the secondary structure of the peptides are not important for the toxic activity. Furthermore, using carboxyfluorescein-loaded liposomes, membrane permeability studies and electron microscopy we investigated whether DCD peptides are able to permeabilize bacterial membranes. The data convincingly show that irrespective of charge the different DCD peptides are not able to permeabilize bacterial membranes. However, bacterial mutants lacking specific cell envelope modifications exhibited different susceptibilities to killing by DCD peptides than wild-type bacterial strains. Finally, immunoelectron microscopy studies indicated that DCD peptides are able to bind to the bacterial surface; however, signs of membrane perturbation were not observed. These studies indicate that DCD peptides do not exert their activity by permeabilizing bacterial membranes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/analysis , Cell Membrane Permeability/physiology , Peptides/chemistry , Protein Processing, Post-Translational , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli/physiology , Humans , Inhibitory Concentration 50 , Molecular Sequence Data , Mutation , Protein Conformation , Solvents/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/ultrastructure , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism , Sweat/chemistry , Sweat/metabolism
7.
Cell Mol Life Sci ; 62(12): 1297-307, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15971105

ABSTRACT

The innate immune system plays a crucial role in maintaining the integrity of the intestine and protecting the host against a vast number of potential microbial pathogens from resident and transient gut microflora. Mucosal epithelial cells and Paneth cells produce a variety of antimicrobial peptides (defensins, cathelicidins, crytdinrelated sequence peptides, bactericidal/permeabilityincreasing protein, chemokine CCL20) and bacteriolytic enzymes (lysozyme, group IIA phospholipase A2) that protect mucosal surfaces and crypts containing intestinal stem cells against invading microbes. Many of the intestinal antimicrobial molecules have additional roles of attracting leukocytes, alarming the adaptive immune system or neutralizing proinflammatory bacterial molecules. Dysfunction of components of the innate immune system has recently been implicated in chronic inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, illustrating the pivotal role of innate immunity in maintaining the delicate balance between immune tolerance and immune response in the gut.


Subject(s)
Immunity, Innate , Intestinal Mucosa/immunology , Intestines/immunology , Animals , Anti-Infective Agents/pharmacology , Defensins/immunology , Humans , Immune Tolerance , Immunity, Mucosal , Intestines/drug effects , Receptors, Cell Surface/immunology
8.
J Pept Sci ; 7(10): 552-64, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11695650

ABSTRACT

Indolicidin is a cationic 13 amino acid peptide amide produced in the granules of bovine neutrophils with the sequence H-ILPWKWPWWPWRR-NH2. Indolicidin is both antimicrobial and, to a lesser extent, haemolytic. In order to systematically investigate structure-function relationships, the solid-phase synthesis of indolicidin and 48 distinct analogues are reported, as well as the characterization of their respective biological properties. Peptides synthesized and characterized include analogues with modified terminal functions, truncations from either terminus, an alanine scan to determine the role of each individual amino acid, specific amino acid exchanges of aromatic, charged and structural residues and several retro-, inverso- and retroinverso-analogues. Together, characterization of these analogues identifies specific residues involved in antimicrobial or haemolytic activity and suggests a core structure that may form a scaffold for the further development of peptidomimetic analogues of indolicidin.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemical synthesis , Antimicrobial Cationic Peptides/chemical synthesis , Cattle , Escherichia coli/drug effects , Hemolysis/drug effects , Humans , In Vitro Techniques , Staphylococcus aureus/drug effects , Structure-Activity Relationship , Tryptophan/chemistry
9.
Peptides ; 22(10): 1651-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11587793

ABSTRACT

Antimicrobial host defense peptides, such as defensins, protegrins, and platelet microbicidal proteins are deployed by mammalian skin, epithelia, phagocytes, and platelets in response to Staphylococcus aureus infection. In addition, staphylococcal products with similar structures and activities, called bacteriocins, inhibit competing microorganisms. Staphylococci have developed resistance mechanisms, which are either highly specific for certain host defense peptides or bacteriocins or which broadly protect against a range of cationic antimicrobial peptides. Experimental infection models can be used to study the molecular mechanisms of antimicrobial peptides, the peptide resistance strategies of S. aureus, and the therapeutic potential of peptides in staphylococcal diseases.


Subject(s)
Anti-Bacterial Agents/immunology , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/pharmacology , Drug Resistance , Staphylococcus aureus/drug effects , Adjuvants, Immunologic/pharmacology , Animals , Antimicrobial Cationic Peptides/metabolism , Bacteria/metabolism , Humans , Immunity, Innate/immunology , Mammals/metabolism , Molecular Sequence Data , Staphylococcal Infections/immunology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity
10.
J Exp Med ; 193(9): 1067-76, 2001 May 07.
Article in English | MEDLINE | ID: mdl-11342591

ABSTRACT

Defensins, antimicrobial peptides of the innate immune system, protect human mucosal epithelia and skin against microbial infections and are produced in large amounts by neutrophils. The bacterial pathogen Staphylococcus aureus is insensitive to defensins by virtue of an unknown resistance mechanism. We describe a novel staphylococcal gene, mprF, which determines resistance to several host defense peptides such as defensins and protegrins. An mprF mutant strain was killed considerably faster by human neutrophils and exhibited attenuated virulence in mice, indicating a key role for defensin resistance in the pathogenicity of S. aureus. Analysis of membrane lipids demonstrated that the mprF mutant no longer modifies phosphatidylglycerol with l-lysine. As this unusual modification leads to a reduced negative charge of the membrane surface, MprF-mediated peptide resistance is most likely based on repulsion of the cationic peptides. Accordingly, inactivation of mprF led to increased binding of antimicrobial peptides by the bacteria. MprF has no similarity with genes of known function, but related genes were identified in the genomes of several pathogens including Mycobacterium tuberculosis, Pseudomonas aeruginosa, and Enterococcus faecalis. MprF thus constitutes a novel virulence factor, which may be of general relevance for bacterial pathogens and represents a new target for attacking multidrug resistant bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Defensins/pharmacology , Lysine/metabolism , Neutrophils/immunology , Phosphatidylglycerols/metabolism , Staphylococcus aureus/metabolism , Amino Acid Sequence , Aminoacyltransferases , Animals , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Base Sequence , Cell Membrane/metabolism , DNA, Bacterial , Drug Resistance, Microbial , Esterification , Genes, Bacterial , Humans , Molecular Sequence Data , Peptides/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Swine , Virulence , alpha-Defensins/pharmacology
11.
Arch Microbiol ; 175(4): 295-300, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11382225

ABSTRACT

Staphylococcal exoproteins can be divided into two groups. One group comprises proteins bearing only a signal peptide, the other group requires an additional propeptide for secretion. The secretion signals of the propeptide-requiring lipase from Staphylococcus hyicus (Lip) have been frequently used to produce recombinant secretory proteins in the food-grade species Staphylococcus carnosus. However, it has been unclear whether recombinant proteins can be secreted using signal peptides of staphylococcal proteins without propeptide. The human growth hormone protein (hGH) was fused to various staphylococcal secretion signals of proteins without propeptide (Seb, SceA, and SceB) and of proteins requiring a propeptide (lipase, lysostaphin, and glycerol ester hydrolase). Secretory hGH was efficiently produced by S. carnosus after fusion with any propeptide-containing secretion signal, whereas precursor proteins were retained in the cells when only a signal peptide was used. Addition of the first six amino acid residues of mature SceA to the signal peptide did also not lead to secretion of hGH. It was concluded that the properties of the mature protein domains determine whether a propeptide is required for secretion or not. The Lip propeptide could be efficiently removed from hGH after introduction of an enterokinase cleavage site between the two protein domains.


Subject(s)
Human Growth Hormone/metabolism , Protein Sorting Signals , Staphylococcus/metabolism , Amino Acid Sequence , Enteropeptidase/metabolism , Molecular Sequence Data , Recombinant Fusion Proteins/metabolism
12.
Infect Immun ; 69(5): 3423-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11292767

ABSTRACT

Staphylococcus aureus is responsible for a large percentage of infections associated with implanted biomedical devices. The molecular basis of primary adhesion to artificial surfaces is not yet understood. Here, we demonstrate that teichoic acids, highly charged cell wall polymers, play a key role in the first step of biofilm formation. An S. aureus mutant bearing a stronger negative surface charge due to the lack of D-alanine esters in its teichoic acids can no longer colonize polystyrene or glass. The mutation abrogates primary adhesion to plastic while production of the glucosamine-based polymer involved in later steps of biofilm formation is not affected. Our data suggest that repulsive electrostatic forces can lead to reduced staphylococcal biofilm formation, which could have considerable impact on the design of novel implanted materials.


Subject(s)
Alanine/physiology , Biofilms , Staphylococcus aureus/physiology , Teichoic Acids/pharmacology , Bacterial Adhesion , Bacterial Proteins/analysis , Polysaccharides, Bacterial/biosynthesis , Static Electricity , Teichoic Acids/chemistry
13.
Appl Environ Microbiol ; 67(3): 1380-3, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11229936

ABSTRACT

The biosynthetic gene clusters of the staphylococcal lantibiotics epidermin and gallidermin are distinguished by the presence of the unique genes epiH and gdmH, respectively. They encode accessory factors for the ATP-binding cassette transporters that mediate secretion of the antimicrobial peptides. Here, we show that gdmH also contributes to immunity to gallidermin but not to nisin. gdmH alone affected susceptibility to gallidermin only moderately, but it led to a multiplication of the immunity level mediated by the FEG immunity genes when cloned together with the gdmT gene, suggesting a synergistic activity of the H and FEG systems. gdmH-related genes were identified in the genomes of several bacteria, indicating an involvement in further cellular functions.


Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Membrane Proteins/genetics , Peptides , Staphylococcus/drug effects , Amino Acid Sequence , Bacterial Proteins/metabolism , Bacteriocins , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Sequence Data , Nisin/pharmacology , Staphylococcus/genetics , Staphylococcus/metabolism
14.
Antimicrob Agents Chemother ; 44(10): 2845-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10991869

ABSTRACT

Recently, Staphylococcus aureus strains with intermediate resistance to vancomycin, the antibiotic of last resort, have been described. Multiple changes in peptidoglycan turnover and structure contribute to the resistance phenotype. Here, we describe that structural changes of teichoic acids in the cell envelope have a considerable influence on the susceptibility to vancomycin and other glycopeptides. S. aureus cells lacking D-alanine esters in teichoic acids exhibited an at least threefold-increased sensitivity to glycopeptide antibiotics. Furthermore, the autolytic activity of the D-alanine mutant was reduced compared to the wild-type, and the mutant was more susceptible to the staphylolytic enzyme lysostaphin. Vancomycin inhibited autolysis at very high concentrations but neither in the wild-type nor in the mutant was the autolytic activity influenced in the range of the MIC. Mutant cells had a considerably higher capacity to bind vancomycin.


Subject(s)
Alanine/chemistry , Staphylococcus aureus/chemistry , Staphylococcus aureus/drug effects , Teichoic Acids/chemistry , Vancomycin Resistance , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Autolysis , Cell Wall/drug effects , Cell Wall/metabolism , Lysostaphin/pharmacology , Mutation/genetics , Phosphates/metabolism , Staphylococcus aureus/genetics , Vancomycin/metabolism
15.
J Biol Chem ; 274(13): 8405-10, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085071

ABSTRACT

Positively charged antimicrobial peptides with membrane-damaging activity are produced by animals and humans as components of their innate immunity against bacterial infections and also by many bacteria to inhibit competing microorganisms. Staphylococcus aureus and Staphylococcus xylosus, which tolerate high concentrations of several antimicrobial peptides, were mutagenized to identify genes responsible for this insensitivity. Several mutants with increased sensitivity were obtained, which exhibited an altered structure of teichoic acids, major components of the Gram-positive cell wall. The mutant teichoic acids lacked D-alanine, as a result of which the cells carried an increased negative surface charge. The mutant cells bound fewer anionic, but more positively charged proteins. They were sensitive to human defensin HNP1-3, animal-derived protegrins, tachyplesins, and magainin II, and to the bacteria-derived peptides gallidermin and nisin. The mutated genes shared sequence similarity with the dlt genes involved in the transfer of D-alanine into teichoic acids from other Gram-positive bacteria. Wild-type strains bearing additional copies of the dlt operon produced teichoic acids with higher amounts of D-alanine esters, bound cationic proteins less effectively and were less sensitive to antimicrobial peptides. We propose a role of the D-alanine-esterified teichoic acids which occur in many pathogenic bacteria in the protection against human and animal defense systems.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides , Bacterial Proteins , Carrier Proteins/genetics , Membrane Transport Proteins , Operon/genetics , Peptides , Staphylococcus aureus/genetics , alpha-Defensins , Alanine/analysis , Amino Acid Sequence , Bacteriocins , DNA-Binding Proteins/pharmacology , Defensins , Genes, Bacterial/drug effects , Genes, Bacterial/genetics , Molecular Sequence Data , Mutagenesis , Nisin/pharmacology , Peptides, Cyclic/pharmacology , Proteins/pharmacology , Sequence Alignment , Staphylococcus aureus/drug effects , Teichoic Acids/chemistry , Teichoic Acids/genetics
16.
FEMS Microbiol Lett ; 166(2): 203-11, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9770275

ABSTRACT

Self-protection of the epidermin-producing strain Staphylococcus epidermidis Tü3298 against the pore-forming lantibiotic epidermin is mediated by an ABC transporter composed of the EpiF, EpiE, and EpiG proteins. We developed a sensitive assay based on HPLC analysis to investigate the capacity of the EpiFEG transporter to release epidermin and analogues from the cell surface to the external fluid. Our results indicate that the EpiFEG transporter works by expelling the lantibiotic from the cytoplasmic membrane into the surrounding medium. Analysis of transporter efficacy using nisin and gallidermin derivatives as substrates revealed a high substrate specificity. Furthermore, we showed that the activity of the gallidermin derivative L6G is enhanced by the presence of EpiE.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/metabolism , Peptides , Staphylococcus epidermidis/metabolism , ATP-Binding Cassette Transporters/genetics , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriocins , Gene Expression Regulation, Bacterial , Genes, Bacterial , Glucose/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microbial Sensitivity Tests , Nisin/metabolism , Nisin/pharmacology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/genetics , Substrate Specificity
17.
Mol Gen Genet ; 254(3): 312-8, 1997 Apr 16.
Article in English | MEDLINE | ID: mdl-9150266

ABSTRACT

The closely related lantibiotics epidermin and gallidermin are produced by Staphylococcus epidermidis Tu3298 and S. gallinarum Tu3928, respectively. The epidermin biosynthetic genes involved in maturation, regulation, and immunity have been identified previously. How epidermin or gallidermin is secreted, however, has remained unclear. Here, we characterize two additional genes, epiH and epiT, as well as the homologous gallidermin genes gdmH and gdmT. EpiT and GdmT are similar to one-component ABC transporters that are involved in the secretion of proteins or peptides. EpiH and GdmH are hydrophobic proteins without conspicuous similarities to other proteins. Comparison of the gene sequences revealed that epiT is incomplete, having an internal deletion that causes a frame shift and a second deletion at the 3'-end, while gdmT is intact. Introduction of epiT and epiH into the heterologous host S. carnosus (pTepi14) bearing the maturation and regulation genes had no significant effect on the rather low level of epidermin production. The presence of the homologous gdmT and gdmH, however, resulted in a strong increase (seven- to tenfold) in the production level, which is very likely to be due to increased efficiency of epidermin secretion. Both gdmT and gdmH were necessary for this effect, indicating that the two gene products cooperate in some way. In the epidermin-producing wild-type strain Tu3298, which contains epiH and the disrupted epiT, the addition of gdmT alone led to a two-fold increase in epidermin production. Both gdmT and gdmH and the corresponding epi genes were activated by the transcriptional regulator EpiQ; this is in accordance with the presence of putative EpiQ operator sites in the promoter regions.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins , DNA-Binding Proteins , Gene Expression Regulation, Bacterial , Genes, Bacterial , Peptides , Staphylococcus/genetics , Trans-Activators/genetics , Amino Acid Sequence , Bacteriocins , Base Sequence , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Staphylococcus/metabolism
18.
FEMS Microbiol Lett ; 137(2-3): 279-84, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8998998

ABSTRACT

The antimicrobial peptide epidermin is distinguished by thioether amino acids such as meso-lanthionine, 3-methyl-lanthionine, and 2-aminovinylcysteine. The enzyme EpiB, encoded on a plasmid of the producing strain Staphylococcus epidermidis Tü3298, is very likely involved in the formation of these unusual amino acids. In order to obtain high-level production of EpiB, an improved staphylococcal expression vector based on the xylose-inducible xylA promoter of Staphylococcus xylosus was constructed. As shown by the expression of a lipase reporter gene, the new plasmid pTX15 mediated a considerably higher expression level after induction and a lower background expression level in the uninduced state than the previously described vector pCX15. The epiB gene was inserted in pTX15 and expressed in Staphylococcus carnosus. The EpiB protein was detected both in the cytoplasmic and the membrane fraction and was partially purified in three steps.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/biosynthesis , Membrane Proteins/biosynthesis , Peptides , Staphylococcus/genetics , Staphylococcus/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriocins , Base Sequence , Cloning, Molecular , DNA Primers/genetics , DNA, Bacterial/genetics , Gene Expression , Genetic Vectors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism , Subcellular Fractions/metabolism
19.
Antonie Van Leeuwenhoek ; 69(2): 119-127, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8775972

ABSTRACT

Lantibiotics are antibiotic peptides that contain the rare thioether amino acids lanthionine and/or methyllanthionine. Epidermin, Pep5 and epilancin K7 are produced by Staphylococcus epidermidis whereas gallidermin (6L-epidermin) was isolated from the closely related species Staphylococcus gallinarum. The biosynthesis of all four lantibiotics proceeds from structural genes which code for prepeptides that are enzymatically modified to give the mature peptides. The genes involved in biosynthesis, processing, export etc. are found in gene clusters adjacent to the structural genes and code for transporters, immunity functions, regulatory proteins and the modification enzymes LanB, LanC and LanD, which catalyze the biosynthesis of the rare amino acids. LanB and LanC are responsible for the dehydration of the serine and threonine residues to give dehydroalanine and dehydrobutyrine and subsequent addition of cysteine SH-groups to the dehydro amino acids which results in the thioether rings. EpiD, the only LanD enzyme known so far, catalyzes the oxidative decarboxylation of the C-terminal cysteine of epidermin which gives the C-terminal S-aminovinylcysteine after addition of a dehydroalanine residue.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Peptides , Staphylococcus/genetics , ATP-Binding Cassette Transporters , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Bacteriocins , Gene Expression Regulation, Bacterial , Genes, Bacterial , Molecular Sequence Data , Multigene Family , Protein Processing, Post-Translational , Staphylococcus epidermidis/genetics
20.
J Bacteriol ; 178(2): 531-6, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8550476

ABSTRACT

The lantibiotic epidermin is produced by Staphylococcus epidermidis Tü3298. The known genes involved in epidermin biosynthesis and regulation are organized as operons (epiABCD and epiQP) that are encoded on the 54-kb plasmid pTü32. Here we describe the characterization of a DNA region that mediates immunity and increased epidermin production, located upstream of the structural gene epiA. The sequence of a 2.6-kb DNA fragment revealed three open reading frames, epiF, -E, and -G, which may form an operon. In the cloning host Staphylococcus carnosus, the three genes mediated an increased tolerance to epidermin, and the highest level of immunity (sevenfold) was achieved with S. carnosus carrying epiFEG and epiQ. The promoter of the first gene, epiF, responded to the activator protein EpiQ and contained a palindromic sequence similar to the EpiQ binding site of the epiA promoter, which is also activated by EpiQ. Inactivation of epiF, -E, or -G resulted in the complete loss of the immunity phenotype. An epidermin-sensitive S. epidermidis Tü3298 mutant was complemented by a DNA fragment containing all three genes. When the epiFEG genes were cloned together with plasmid pTepi14, containing the biosynthetic genes epiABCDQP, the level of epidermin production was approximately fivefold higher. The proteins EpiF, -E, and -G are similar in deduced sequence and proposed structure to the components of various ABC transporter systems. EpiF is a hydrophilic protein with conserved ATP-binding sites, while EpiE and -G have six alternating hydrophobic regions and very likely constitute the integral membrane domains. When EpiF was overproduced in S. carnosus, it was at least partially associated with the cytoplasmic membrane. A potential mechanism for how EpiFEG mediates immunity is discussed.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Genes, Bacterial/genetics , Peptides , Staphylococcus epidermidis/genetics , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacteriocins , Base Sequence , Cloning, Molecular , Drug Resistance, Microbial/genetics , Genes, Bacterial/physiology , Membrane Proteins/genetics , Microbial Sensitivity Tests , Molecular Sequence Data , Molecular Weight , Operon/genetics , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Staphylococcus/drug effects , Staphylococcus epidermidis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...