Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 169(7): 1240-1248.e23, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28622509

ABSTRACT

Drug-resistant bacterial pathogens pose an urgent public-health crisis. Here, we report the discovery, from microbial-extract screening, of a nucleoside-analog inhibitor that inhibits bacterial RNA polymerase (RNAP) and exhibits antibacterial activity against drug-resistant bacterial pathogens: pseudouridimycin (PUM). PUM is a natural product comprising a formamidinylated, N-hydroxylated Gly-Gln dipeptide conjugated to 6'-amino-pseudouridine. PUM potently and selectively inhibits bacterial RNAP in vitro, inhibits bacterial growth in culture, and clears infection in a mouse model of Streptococcus pyogenes peritonitis. PUM inhibits RNAP through a binding site on RNAP (the NTP addition site) and mechanism (competition with UTP for occupancy of the NTP addition site) that differ from those of the RNAP inhibitor and current antibacterial drug rifampin (Rif). PUM exhibits additive antibacterial activity when co-administered with Rif, exhibits no cross-resistance with Rif, and exhibits a spontaneous resistance rate an order-of-magnitude lower than that of Rif. PUM is a highly promising lead for antibacterial therapy.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Streptomyces/chemistry , Animals , Anti-Bacterial Agents/chemistry , Bacteria/classification , Bacteria/drug effects , Bacteria/growth & development , DNA-Directed RNA Polymerases/chemistry , Drug Resistance, Bacterial , Female , HeLa Cells , Humans , Mice , Mice, Inbred ICR , Soil Microbiology , Streptococcal Infections/drug therapy , Streptococcus pyogenes/drug effects , Transcription, Genetic/drug effects
2.
Biochemistry ; 60(39): 2943-2955, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34547893

ABSTRACT

The increasing number of resistant bacteria is a major threat worldwide, leading to the search for new antibiotic agents. One of the leading strategies is the use of antimicrobial peptides (AMPs), cationic and hydrophobic innate immune defense peptides. A major target of AMPs is the bacterial membrane. Notably, accumulating data suggest that AMPs can activate the two-component systems (TCSs) of Gram-negative bacteria. These include PhoP-PhoQ (PhoPQ) and PmrA-PmrB (PmrAB), responsible for remodeling of the bacterial cell surface. To better understand this mechanism, we utilized bacteria deficient either in one system alone or in both and biophysical tools including fluorescence spectroscopy, single-cell atomic force microscopy, electron microscopy, and mass spectrometry (Moskowitz, S. M.; Antimicrob. Agents Chemother. 2012, 56, 1019-1030; Cheng, H. Y.; J. Biomed. Sci. 2010, 17, 60). Our data suggested that the two systems have opposing effects on the properties of Salmonella enterica. The knockout of PhoPQ made the bacteria more susceptible to AMPs by making the surface less rigid, more polarized, and permeable with a slightly more negatively charged cell wall. In addition, the periplasmic space is thinner. In contrast, the knockout of PmrAB did not affect its susceptibility, while it made the bacterial outer layer very rigid, less polarized, and less permeable than the other two mutants, with a negatively charged cell wall similar to the WT. Overall, the data suggest that the coexistence of systems with opposing effects on the biophysical properties of the bacteria contribute to their membrane flexibility, which, on the one hand, is important to accommodate changing environments and, on the other hand, may inhibit the development of meaningful resistance to AMPs.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/metabolism , Cell Wall/drug effects , Salmonella Infections/drug therapy , Salmonella enterica/drug effects , Bacterial Proteins/genetics , Drug Resistance, Bacterial , Periplasm/drug effects , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella enterica/isolation & purification , Salmonella enterica/metabolism , Serogroup
3.
Nat Prod Rep ; 38(1): 130-239, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32935693

ABSTRACT

Covering: up to June 2020Ribosomally-synthesized and post-translationally modified peptides (RiPPs) are a large group of natural products. A community-driven review in 2013 described the emerging commonalities in the biosynthesis of RiPPs and the opportunities they offered for bioengineering and genome mining. Since then, the field has seen tremendous advances in understanding of the mechanisms by which nature assembles these compounds, in engineering their biosynthetic machinery for a wide range of applications, and in the discovery of entirely new RiPP families using bioinformatic tools developed specifically for this compound class. The First International Conference on RiPPs was held in 2019, and the meeting participants assembled the current review describing new developments since 2013. The review discusses the new classes of RiPPs that have been discovered, the advances in our understanding of the installation of both primary and secondary post-translational modifications, and the mechanisms by which the enzymes recognize the leader peptides in their substrates. In addition, genome mining tools used for RiPP discovery are discussed as well as various strategies for RiPP engineering. An outlook section presents directions for future research.


Subject(s)
Computational Biology/methods , Enzymes/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Engineering/methods , Biological Products/chemistry , Biological Products/classification , Biological Products/metabolism , Enzymes/chemistry , Hydroxylation , Methylation , Peptides/classification , Peptides/genetics , Phosphorylation , Protein Processing, Post-Translational , Protein Sorting Signals/physiology , Ribosomes/metabolism
4.
J Nat Prod ; 82(2): 301-308, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30666877

ABSTRACT

Genome mining of the Gram-negative bacterium Pseudomonas fluorescens Pf0-1 showed that the strain possesses a silent NRPS-based biosynthetic gene cluster encoding a new lipopeptide; its activation required the repair of the global regulator system. In this paper, we describe the genomics-driven discovery and characterization of the associated secondary metabolite gacamide A, a lipodepsipeptide that forms a new family of Pseudomonas lipopeptides. The compound has a moderate, narrow-spectrum antibiotic activity and facilitates bacterial surface motility.


Subject(s)
Bacterial Proteins/genetics , Drug Discovery , Lipopeptides/genetics , Peptides, Cyclic/genetics , Pseudomonas fluorescens/genetics , Lipopeptides/pharmacology , Multigene Family , Peptides, Cyclic/pharmacology
5.
Proc Natl Acad Sci U S A ; 113(45): E7077-E7086, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27791134

ABSTRACT

Daptomycin is a highly efficient last-resort antibiotic that targets the bacterial cell membrane. Despite its clinical importance, the exact mechanism by which daptomycin kills bacteria is not fully understood. Different experiments have led to different models, including (i) blockage of cell wall synthesis, (ii) membrane pore formation, and (iii) the generation of altered membrane curvature leading to aberrant recruitment of proteins. To determine which model is correct, we carried out a comprehensive mode-of-action study using the model organism Bacillus subtilis and different assays, including proteomics, ionomics, and fluorescence light microscopy. We found that daptomycin causes a gradual decrease in membrane potential but does not form discrete membrane pores. Although we found no evidence for altered membrane curvature, we confirmed that daptomycin inhibits cell wall synthesis. Interestingly, using different fluorescent lipid probes, we showed that binding of daptomycin led to a drastic rearrangement of fluid lipid domains, affecting overall membrane fluidity. Importantly, these changes resulted in the rapid detachment of the membrane-associated lipid II synthase MurG and the phospholipid synthase PlsX. Both proteins preferentially colocalize with fluid membrane microdomains. Delocalization of these proteins presumably is a key reason why daptomycin blocks cell wall synthesis. Finally, clustering of fluid lipids by daptomycin likely causes hydrophobic mismatches between fluid and more rigid membrane areas. This mismatch can facilitate proton leakage and may explain the gradual membrane depolarization observed with daptomycin. Targeting of fluid lipid domains has not been described before for antibiotics and adds another dimension to our understanding of membrane-active antibiotics.

6.
Biochemistry ; 57(13): 2024-2034, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29518324

ABSTRACT

Multidrug resistant bacteria possess various mechanisms that can sense environmental stresses such as antibiotics and antimicrobial peptides and rapidly respond to defend themselves. Two known defense strategies are biofilm formation and lipopolysaccharide (LPS) modification. Though LPS modifications are observed in biofilm-embedded bacteria, their effect on biofilm formation is unknown. Using biochemical and biophysical methods coupled with confocal microscopy, atomic force microscopy, and transmission electron microscopy, we show that biofilm formation is promoted in a Pseudomonas aeruginosa PAO1 strain with a loss of function mutation in the arnB gene. This loss of function prevents the addition of the positively charged sugar 4-amino-4-deoxy-l-arabinose to lipid A of LPS under restrictive magnesium conditions. The data reveal that the arnB mutant, which is susceptible to antimicrobial peptides, forms a biofilm that is more robust than that of the wild type. This is in line with the observations that the arnB mutant exhibits outer surface properties such as hydrophobicity and net negative charge that promote the formation of biofilms. Moreover, when grown under Mg2+ limitation, both the wild type and the arnB mutant exhibited a reduction in the level of membrane-bound polysaccharides. The data suggest that the loss of polysaccharides exposes the membrane and alters its biophysical properties, which in turn leads to more biofilm formation. In summary, we show for the first time that blocking a specific lipid A modification promotes biofilm formation, suggesting a trade-off between LPS remodeling and resistance mechanisms of biofilm formation.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins , Biofilms/drug effects , Lipid A , Polysaccharides, Bacterial , Pseudomonas aeruginosa/physiology , Antimicrobial Cationic Peptides/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , Lipid A/genetics , Lipid A/metabolism , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/metabolism
7.
Article in English | MEDLINE | ID: mdl-30061290

ABSTRACT

Sulfide production has been proposed to be a universal defense mechanism against antibiotics in bacteria (K. Shatalin, E. Shatalina, A. Mironov, and E. Nudler, Science 334:986-990, 2011, doi:10.1126/science.1209855). To gain insight into the mechanism underlying sulfide protection, we systematically and comparatively addressed the interference of sulfide with antibiotic activity against Staphylococcus aureus, as a model organism. The impact of sulfide and sulfide precursors on the antibiotic susceptibility of S. aureus to the most important classes of antibiotics was analyzed using modified disk diffusion assays, killing kinetic assays, and drug uptake studies. In addition, sulfide production and the impact of exogenously added sulfide on the physiology of S. aureus were analyzed. Sulfide protection was found to be limited to aminoglycoside antibiotics, which are known to be taken up by bacterial cells in an energy-dependent process. The protective mechanism was found to rely on an inhibitory effect of sulfide on the bacterial respiratory chain, leading to reduced drug uptake. S. aureus was found to be incapable of producing substantial amounts of sulfide. We propose that bacterial sulfide production should not be regarded as a general defense mechanism against antibiotics, since (i) it is limited to aminoglycosides and (ii) production levels vary considerably among species and, as for S. aureus, may be too low for protection.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Sulfides/metabolism , Drug Resistance, Bacterial , Microbial Sensitivity Tests
8.
Int J Med Microbiol ; 308(3): 335-348, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29429584

ABSTRACT

The first-in-class lipopeptide antibiotic daptomycin (DAP) is highly active against Gram-positive pathogens including ß-lactam and glycopeptide resistant strains. Its molecular mode of action remains enigmatic, since a defined target has not been identified so far and multiple effects, primarily on the cell envelope have been observed. Reduced DAP susceptibility has been described in S. aureus and enterococci after prolonged treatment courses. In line with its pleiotropic antibiotic activities, a unique, defined molecular mechanism of resistance has not emerged, instead non-susceptibility appears often accompanied by alterations in membrane composition and changes in cell wall homeostasis. We compared S. aureus strains HG001 and SG511, which differ primarily in the functionality of the histidine kinase GraS, to evaluate the impact of the GraRS regulatory system on the development of DAP non-susceptibility. After extensive serial passing, both DAPR variants reached a minimal inhibitory concentration of 31 µg/ml and shared some phenotypic characteristics (e.g. thicker cell wall, reduced autolysis). However, based on comprehensive analysis of the underlying genetic, transcriptomic and proteomic changes, we found that both strains took different routes to achieve DAP resistance. Our study highlights the impressive genetic and physiological capacity of S. aureus to counteract pleiotropic activities of cell wall- and membrane-active compounds even when a major cell wall regulatory system is dysfunctional.


Subject(s)
Bacterial Proteins/genetics , Daptomycin/pharmacology , Gene Expression Regulation, Bacterial , Mutation , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Cell Membrane/metabolism , Drug Resistance, Bacterial/genetics , Genotype , Histidine Kinase/genetics , Microbial Sensitivity Tests , Proteomics , Staphylococcal Infections/microbiology , Staphylococcus aureus/enzymology , Staphylococcus aureus/physiology
9.
J Biol Chem ; 291(5): 2535-46, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26620564

ABSTRACT

Screening of new compounds directed against key protein targets must continually keep pace with emerging antibiotic resistances. Although periplasmic enzymes of bacterial cell wall biosynthesis have been among the first drug targets, compounds directed against the membrane-integrated catalysts are hardly available. A promising future target is the integral membrane protein MraY catalyzing the first membrane associated step within the cytoplasmic pathway of bacterial peptidoglycan biosynthesis. However, the expression of most MraY homologues in cellular expression systems is challenging and limits biochemical analysis. We report the efficient production of MraY homologues from various human pathogens by synthetic cell-free expression approaches and their subsequent characterization. MraY homologues originating from Bordetella pertussis, Helicobacter pylori, Chlamydia pneumoniae, Borrelia burgdorferi, and Escherichia coli as well as Bacillus subtilis were co-translationally solubilized using either detergent micelles or preformed nanodiscs assembled with defined membranes. All MraY enzymes originating from Gram-negative bacteria were sensitive to detergents and required nanodiscs containing negatively charged lipids for obtaining a stable and functionally folded conformation. In contrast, the Gram-positive B. subtilis MraY not only tolerates detergent but is also less specific for its lipid environment. The MraY·nanodisc complexes were able to reconstitute a complete in vitro lipid I and lipid II forming pipeline in combination with the cell-free expressed soluble enzymes MurA-F and with the membrane-associated protein MurG. As a proof of principle for future screening platforms, we demonstrate the inhibition of the in vitro lipid II biosynthesis with the specific inhibitors fosfomycin, feglymycin, and tunicamycin.


Subject(s)
Bacterial Proteins/chemistry , Monosaccharides/biosynthesis , Oligopeptides/biosynthesis , Transferases/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Bacillus subtilis/enzymology , Biosynthetic Pathways , Bordetella pertussis/enzymology , Borrelia burgdorferi/enzymology , Cell Wall/chemistry , Cell-Free System , Chlamydophila pneumoniae/enzymology , Cytoplasm/chemistry , DNA/chemistry , Detergents/chemistry , Escherichia coli/enzymology , Fosfomycin/chemistry , Helicobacter pylori/enzymology , Micelles , Peptides/chemistry , Peptidoglycan/chemistry , Proteins/chemistry , Recombinant Proteins/chemistry , Transferases (Other Substituted Phosphate Groups) , Tunicamycin/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/biosynthesis
10.
Int J Med Microbiol ; 307(1): 1-10, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27989665

ABSTRACT

The assembly of the bacterial cell wall requires synchronization of a multitude of biosynthetic machineries and regulatory networks. The eukaryotic-like serine/threonine kinase PknB has been implicated in coordinating cross-wall formation, autolysis and cell division in Staphylococcus aureus. However, the signal molecule sensed by this kinase remained elusive so far. Here, we provide compelling biochemical evidence that PknB interacts with the ultimate cell wall precursor lipid II, triggering kinase activity. Moreover, we observed crosstalk of PknB with the two component system WalKR and identified the early cell division protein FtsZ as another PknB phosphorylation substrate in S. aureus. In agreement with the implied role in regulation of cell envelope metabolism, we found PknB to preferentially localize to the septum of S. aureus and the PASTA domains to be crucial for recruitment to this site. The data provide a model for the contribution of PknB to control cell wall metabolism and cell division.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/metabolism , Protein Serine-Threonine Kinases/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Cytoskeletal Proteins/metabolism , Protein Binding , Protein Interaction Maps , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
11.
Int J Med Microbiol ; 307(1): 57-63, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27931949

ABSTRACT

The species Staphylococcus argenteus was separated recently from Staphylococcus aureus (Tong S.Y., F. Schaumburg, M.J. Ellington, J. Corander, B. Pichon, F. Leendertz, S.D. Bentley, J. Parkhill, D.C. Holt, G. Peters, and P.M. Giffard, 2015). The objective of this work was to characterise the genome of a non-human S. argenteus strain, which had been isolated from the faeces of a wild-living western lowland gorilla in Gabon, and analyse the spectrum of this species in matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS). The full genome sequence revealed a scarcity of virulence genes and absence of resistance genes, indicating a decreased virulence potential compared to S. aureus and the human methicillin-resistant S. argenteus isolate MSHR1132T. Spectra obtained by MALDI-TOF MS and the analysis of available sequences in the genome databases identified several MALDI-TOF MS signals that clearly differentiate S. argenteus, the closely related Staphylococcus schweitzeri and S. aureus. In conclusion, in the absence of biochemical tests that identify the three species, mass spectrometry should be employed as method of choice.


Subject(s)
Bacteriological Techniques/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Staphylococcus/chemistry , Staphylococcus/classification , Animals , Carrier State/veterinary , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Drug Resistance, Bacterial/genetics , Gabon , Gorilla gorilla , Sequence Analysis, DNA , Staphylococcal Infections/veterinary , Staphylococcus/isolation & purification , Virulence Factors/genetics
12.
Proc Natl Acad Sci U S A ; 111(14): E1409-18, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706874

ABSTRACT

Short antimicrobial peptides rich in arginine (R) and tryptophan (W) interact with membranes. To learn how this interaction leads to bacterial death, we characterized the effects of the minimal pharmacophore RWRWRW-NH2. A ruthenium-substituted derivative of this peptide localized to the membrane in vivo, and the peptide also integrated readily into mixed phospholipid bilayers that resemble Gram-positive membranes. Proteome and Western blot analyses showed that integration of the peptide caused delocalization of peripheral membrane proteins essential for respiration and cell-wall biosynthesis, limiting cellular energy and undermining cell-wall integrity. This delocalization phenomenon also was observed with the cyclic peptide gramicidin S, indicating the generality of the mechanism. Exogenous glutamate increases tolerance to the peptide, indicating that osmotic destabilization also contributes to antibacterial efficacy. Bacillus subtilis responds to peptide stress by releasing osmoprotective amino acids, in part via mechanosensitive channels. This response is triggered by membrane-targeting bacteriolytic peptides of different structural classes as well as by hypoosmotic conditions.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Membrane Proteins/metabolism , Bacillus subtilis/metabolism , Binding Sites , Cytochromes c/metabolism , Homeostasis , Lipid Bilayers , Phospholipids/metabolism
13.
Biochim Biophys Acta ; 1848(11 Pt B): 3062-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25934055

ABSTRACT

Antimicrobial peptides (AMPs) are natural antibiotics produced by virtually all living organisms. Typically, AMPs are cationic and amphiphilic and first contacts with target microbes involve interactions with negatively charged components of the cell envelope such as lipopolysaccharide (LPS), and wall- or lipoteichoic acids (WTA, LTA). The importance of charge-mediated interactions of AMPs with the cell envelope is reflected by effective microbial resistance mechanisms which are based on reduction of the overall charge of these polymers. The anionic polymers are linked in various ways to the stress-bearing polymer of the cell envelope, the peptidoglycan, which is made of a highly conserved building block, a disaccharide-pentapeptide moiety that also contains charged residues. This structural element, in spite of its conservation throughout the bacterial world, can undergo genus- and species-specific modifications that also impact significantly on the overall charge of the cell envelope and on the binding affinity of AMPs. The modification reactions involved largely occur on the membrane-bound peptidoglycan building block, the so-called lipid II, which is a most prominent target for AMPs. In this review, we focus on modifications of lipid II and peptidoglycan and discuss their consequences for the interactions with various classes of AMPs, such as defensins, lantibiotics and glyco-(lipo)-peptide antibiotics. This article is part of a Special Issue entitled: Bacterial Resistance to Antimicrobial Peptides.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Cell Wall/metabolism , Drug Resistance, Bacterial , Gram-Positive Bacteria/metabolism , Gram-Positive Bacterial Infections/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Animals , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/therapeutic use , Gram-Positive Bacteria/drug effects , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/prevention & control , Host-Pathogen Interactions , Humans , Microbial Viability , Models, Molecular , Molecular Structure , Protein Binding , Signal Transduction , Structure-Activity Relationship , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
14.
J Nat Prod ; 79(4): 685-90, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27057690

ABSTRACT

Three new epithiodiketopiperazine natural products [outovirin A (1), outovirin B (2), and outovirin C (3)] resembling the antifungal natural product gliovirin have been identified in extracts of Penicillium raciborskii, an endophytic fungus isolated from Rhododendron tomentosum. The compounds are unusual for their class in that they possess sulfide bridges between α- and ß-carbons rather than the typical α-α bridging. To our knowledge, outovirin A represents the first reported naturally produced epimonothiodiketopiperazine, and antifungal outovirin C is the first reported trisulfide gliovirin-like compound. This report describes the identification and structural elucidation of the compounds by LC-MS/MS and NMR.


Subject(s)
Antifungal Agents/isolation & purification , Penicillium/chemistry , Piperazines/isolation & purification , Rhododendron/microbiology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Piperazines/chemistry , Piperazines/pharmacology
15.
Biophys J ; 108(5): 1114-24, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25762323

ABSTRACT

The antimicrobial peptide nisin exerts its activity by a unique dual mechanism. It permeates the cell membranes of Gram-positive bacteria by binding to the cell wall precursor Lipid II and inhibits cell wall synthesis. Binding of nisin to Lipid II induces the formation of large nisin-Lipid II aggregates in the membrane of bacteria as well as in Lipid II-doped model membranes. Mechanistic details of the aggregation process and its impact on membrane permeation are still unresolved. In our experiments, we found that fluorescently labeled nisin bound very inhomogeneously to bacterial membranes as a consequence of the strong aggregation due to Lipid II binding. A correlation between cell membrane damage and nisin aggregation was observed in vivo. To further investigate the aggregation process of Lipid II and nisin, we assessed its dynamics by single-molecule microscopy of fluorescently labeled Lipid II molecules in giant unilamellar vesicles using light-sheet illumination. We observed a continuous reduction of Lipid II mobility due to a steady growth of nisin-Lipid II aggregates as a function of time and nisin concentration. From the measured diffusion constants of Lipid II, we estimated that the largest aggregates contained tens of thousands of Lipid II molecules. Furthermore, we observed that the formation of large nisin-Lipid II aggregates induced vesicle budding in giant unilamellar vesicles. Thus, we propose a membrane permeation mechanism that is dependent on the continuous growth of nisin-Lipid II aggregation and probably involves curvature effects on the membrane.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Cytoplasmic Vesicles/drug effects , Nisin/pharmacology , Unilamellar Liposomes/metabolism , Bacillus subtilis/drug effects , Bacillus subtilis/metabolism , Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Unilamellar Liposomes/chemistry
16.
J Biol Chem ; 289(18): 12390-403, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24648525

ABSTRACT

The formation of periplasmic sulfur globules is an intermediate step during the oxidation of reduced sulfur compounds in various sulfur-oxidizing microorganisms. The mechanism of how this sulfur is activated and crosses the cytoplasmic membrane for further oxidation to sulfite by the dissimilatory reductase DsrAB is incompletely understood, but it has been well documented that the pathway involves sulfur trafficking mediated by sulfur-carrying proteins. So far sulfur transfer from DsrEFH to DsrC has been established. Persulfurated DsrC very probably serves as a direct substrate for DsrAB. Here, we introduce further important players in oxidative sulfur metabolism; the proteins Rhd_2599, TusA, and DsrE2 are strictly conserved in the Chromatiaceae, Chlorobiaceae, and Acidithiobacillaceae families of sulfur-oxidizing bacteria and are linked to genes encoding complexes involved in sulfur oxidation (Dsr or Hdr) in the latter two. Here we show via relative quantitative real-time PCR and microarray analysis an increase of mRNA levels under sulfur-oxidizing conditions for rhd_2599, tusA, and dsrE2 in Allochromatium vinosum. Transcriptomic patterns for the three genes match those of major genes for the sulfur-oxidizing machinery rather than those involved in biosynthesis of sulfur-containing biomolecules. TusA appears to be one of the major proteins in A. vinosum. A rhd_2599-tusA-dsrE2-deficient mutant strain, although not viable in liquid culture, was clearly sulfur oxidation negative upon growth on solid media containing sulfide. Rhd_2599, TusA, and DsrE2 bind sulfur atoms via conserved cysteine residues, and experimental evidence is provided for the transfer of sulfur between these proteins as well as to DsrEFH and DsrC.


Subject(s)
Bacterial Proteins/metabolism , Chromatiaceae/metabolism , Cytoplasm/metabolism , Sulfur/metabolism , Bacterial Proteins/genetics , Binding Sites/genetics , Biological Transport , Chromatiaceae/genetics , Cysteine/genetics , Cysteine/metabolism , Electrophoresis , Gene Expression Regulation, Bacterial , Gene Order , Microbial Viability/genetics , Mutation , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sulfurtransferases/genetics , Sulfurtransferases/metabolism , Transcriptome/genetics
17.
J Biol Chem ; 289(17): 12063-12076, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24627484

ABSTRACT

The lantibiotic NAI-107 is active against Gram-positive bacteria including vancomycin-resistant enterococci and methicillin-resistant Staphylococcus aureus. To identify the molecular basis of its potency, we studied the mode of action in a series of whole cell and in vitro assays and analyzed structural features by nuclear magnetic resonance (NMR). The lantibiotic efficiently interfered with late stages of cell wall biosynthesis and induced accumulation of the soluble peptidoglycan precursor UDP-N-acetylmuramic acid-pentapeptide (UDP-MurNAc-pentapeptide) in the cytoplasm. Using membrane preparations and a complete cascade of purified, recombinant late stage peptidoglycan biosynthetic enzymes (MraY, MurG, FemX, PBP2) and their respective purified substrates, we showed that NAI-107 forms complexes with bactoprenol-pyrophosphate-coupled precursors of the bacterial cell wall. Titration experiments indicate that first a 1:1 stoichiometric complex occurs, which then transforms into a 2:1 (peptide: lipid II) complex, when excess peptide is added. Furthermore, lipid II and related molecules obviously could not serve as anchor molecules for the formation of defined and stable nisin-like pores, however, slow membrane depolarization was observed after NAI-107 treatment, which could contribute to killing of the bacterial cell.


Subject(s)
Bacteriocins/metabolism , Cell Wall/metabolism , Terpenes/metabolism , Amino Acid Sequence , Bacteriocins/chemistry , Bacteriocins/pharmacology , Cell Wall/drug effects , Cell Wall/physiology , Electrophoresis, Polyacrylamide Gel , Microbial Sensitivity Tests , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Proteomics , Staphylococcus aureus/drug effects
18.
J Biol Chem ; 289(50): 34953-64, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25342741

ABSTRACT

Fungi and bacteria compete with an arsenal of secreted molecules for their ecological niche. This repertoire represents a rich and inexhaustible source for antibiotics and fungicides. Antimicrobial peptides are an emerging class of fungal defense molecules that are promising candidates for pharmaceutical applications. Based on a co-cultivation system, we studied the interaction of the coprophilous basidiomycete Coprinopsis cinerea with different bacterial species and identified a novel defensin, copsin. The polypeptide was recombinantly produced in Pichia pastoris, and the three-dimensional structure was solved by NMR. The cysteine stabilized α/ß-fold with a unique disulfide connectivity, and an N-terminal pyroglutamate rendered copsin extremely stable against high temperatures and protease digestion. Copsin was bactericidal against a diversity of Gram-positive bacteria, including human pathogens such as Enterococcus faecium and Listeria monocytogenes. Characterization of the antibacterial activity revealed that copsin bound specifically to the peptidoglycan precursor lipid II and therefore interfered with the cell wall biosynthesis. In particular, and unlike lantibiotics and other defensins, the third position of the lipid II pentapeptide is essential for effective copsin binding. The unique structural properties of copsin make it a possible scaffold for new antibiotics.


Subject(s)
Agaricales/metabolism , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/metabolism , Defensins/pharmacology , Fungal Proteins/pharmacology , Peptidoglycan/biosynthesis , Agaricales/growth & development , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacteria/growth & development , Coculture Techniques , Defensins/chemistry , Defensins/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation
19.
J Biol Chem ; 289(39): 26949-26959, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25122768

ABSTRACT

Conserved clusters of genes encoding DsrE and TusA homologs occur in many archaeal and bacterial sulfur oxidizers. TusA has a well documented function as a sulfurtransferase in tRNA modification and molybdenum cofactor biosynthesis in Escherichia coli, and DsrE is an active site subunit of the DsrEFH complex that is essential for sulfur trafficking in the phototrophic sulfur-oxidizing Allochromatium vinosum. In the acidothermophilic sulfur (S(0))- and tetrathionate (S4O6(2-))-oxidizing Metallosphaera cuprina Ar-4, a dsrE3A-dsrE2B-tusA arrangement is situated immediately between genes encoding dihydrolipoamide dehydrogenase and a heterodisulfide reductase-like complex. In this study, the biochemical features and sulfur transferring abilities of the DsrE2B, DsrE3A, and TusA proteins were investigated. DsrE3A and TusA proved to react with tetrathionate but not with NaSH, glutathione persulfide, polysulfide, thiosulfate, or sulfite. The products were identified as protein-Cys-S-thiosulfonates. DsrE3A was also able to cleave the thiosulfate group from TusA-Cys(18)-S-thiosulfonate. DsrE2B did not react with any of the sulfur compounds tested. DsrE3A and TusA interacted physically with each other and formed a heterocomplex. The cysteine residue (Cys(18)) of TusA is crucial for this interaction. The single cysteine mutants DsrE3A-C(93)S and DsrE3A-C(101)S retained the ability to transfer the thiosulfonate group to TusA. TusA-C(18)S neither reacted with tetrathionate nor was it loaded with thiosulfate with DsrE3A-Cys-S-thiosulfonate as the donor. The transfer of thiosulfate, mediated by a DsrE-like protein and TusA, is unprecedented not only in M. cuprina but also in other sulfur-oxidizing prokaryotes. The results of this study provide new knowledge on oxidative microbial sulfur metabolism.


Subject(s)
Archaeal Proteins/metabolism , Carrier Proteins/metabolism , Multiprotein Complexes/metabolism , Sulfolobaceae/metabolism , Sulfur/metabolism , Sulfurtransferases/metabolism , Amino Acid Substitution , Archaeal Proteins/genetics , Carrier Proteins/genetics , Multiprotein Complexes/genetics , Mutation, Missense , Oxidation-Reduction , Sequence Homology, Amino Acid , Sulfolobaceae/genetics , Sulfurtransferases/genetics
20.
Biochim Biophys Acta ; 1838(12): 3061-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25128154

ABSTRACT

Representing a physiological "Achilles' heel", the cell wall precursor lipid II (LII) is a prime target for various classes of antibiotics. Over the years LII-binding agents have been recognized as promising candidates and templates in the search for new antibacterial compounds to complement or replace existing drugs. To elucidate the molecular structural basis underlying LII functional mechanism and to better understand if and how lantibiotic binding alters the molecular behavior of LII, we performed molecular dynamics (MD) simulations of phospholipid membrane-embedded LII in the absence and presence of the LII-binding lantibiotic nisin. In a series of 2×4 independent, unbiased 100ns MD simulations we sampled the conformational dynamics of nine LII as well as nine LII-nisin complexes embedded in an aqueous 150mM NaCl/POPC phospholipid membrane environment. We found that nisin binding to LII induces a reduction of LII mobility and flexibility, an outward shift of the LII pentapeptide, an inward movement of the LII disaccharide section, and an overall deeper insertion of the LII tail group into the membrane. The latter effect might indicate an initial step in adopting a stabilizing, scaffold-like structure in the process of nisin-induced membrane leakage. At the same time nisin conformation and LII interaction remain similar to the 1WCO LII-nisin NMR solution structure.

SELECTION OF CITATIONS
SEARCH DETAIL