Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
J Am Chem Soc ; 146(36): 24855-24862, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39197836

ABSTRACT

The synthetic small molecule DCAP is a chemically well-characterized compound with antibiotic activity against Gram-positive and Gram-negative bacteria, including drug-resistant pathogens. Until now, its mechanism of action was proposed to rely exclusively on targeting the bacterial membrane, thereby causing membrane depolarization, and increasing membrane permeability (Eun et al. 2012, J. Am. Chem. Soc. 134 (28), 11322-11325; Hurley et al. 2015, ACS Med. Chem. Lett. 6, 466-471). Here, we show that the antibiotic activity of DCAP results from a dual mode of action that is more targeted and multifaceted than previously anticipated. Using microbiological and biochemical assays in combination with fluorescence microscopy, we provide evidence that DCAP interacts with undecaprenyl pyrophosphate-coupled cell envelope precursors, thereby blocking peptidoglycan biosynthesis and impairing cell division site organization. Our work discloses a concise model for the mode of action of DCAP which involves the binding to a specific target molecule to exert pleiotropic effects on cell wall biosynthetic and divisome machineries.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Uridine Diphosphate N-Acetylmuramic Acid , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Molecular Structure , Cell Wall/drug effects , Cell Wall/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis
2.
Int J Mol Sci ; 24(2)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36674846

ABSTRACT

To date, a number of lantibiotics have been shown to use lipid II-a highly conserved peptidoglycan precursor in the cytoplasmic membrane of bacteria-as their molecular target. The α-component (Lchα) of the two-component lantibiotic lichenicidin, previously isolated from the Bacillus licheniformis VK21 strain, seems to contain two putative lipid II binding sites in its N-terminal and C-terminal domains. Using NMR spectroscopy in DPC micelles, we obtained convincing evidence that the C-terminal mersacidin-like site is involved in the interaction with lipid II. These data were confirmed by the MD simulations. The contact area of lipid II includes pyrophosphate and disaccharide residues along with the first isoprene units of bactoprenol. MD also showed the potential for the formation of a stable N-terminal nisin-like complex; however, the conditions necessary for its implementation in vitro remain unknown. Overall, our results clarify the picture of two component lantibiotics mechanism of antimicrobial action.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Anti-Bacterial Agents/chemistry , Peptidoglycan/metabolism , Bacteriocins/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
3.
Biochemistry ; 59(38): 3523-3528, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32885950

ABSTRACT

A pathogenic bacterium has its own mechanisms for not only pathogenic attack but also exogenous invasion defense, in which the bacterial cell wall is the front line of attack and defense. We developed a biochemical lanthanide-encoding approach to quantify the uncanonical d-amino acid (d-X) that was edited in a small proportion into the terminal acyl-d-Ala-d-X of nascent peptidoglycan UDP-MurNAc-pentapeptides in the bacterial cell wall. This approach overcomes the difficulties regarding quantification and accuracy issues encountered by the popular optical imaging and traditional high-performance liquid chromatography-based methods. Newly synthesized azide-d-Leu and ketone-d-Met were used together with alkynyl-d-Ala for their metabolic assembly and then bioorthogonally encoded by the correspondingly fabricated DBCO-DOTA-Gd, H2NO-DOTA-Eu, and azide-DOTA-Sm tags. This approach allows direct quantification of the d-X in situ in the cell wall using 158Gd, 153Eu, and 154Sm species-unspecific isotope dilution inductively coupled plasma mass spectrometry, avoiding any tedious and complex "cell-broken" pretreatment procedures that might induce racemization of the d-X. The obtained site-specific and accurate in situ information about the d-X enables quantitative monitoring of the bacterial response when Staphylococcus aureus meets vancomycin, showing that the amounts of azide-d-Leu and ketone-d-Met assembled are more important after determining the structure- and composition-dependent bacterial antibiotic resistance mechanisms. In addition, we found that the combined use of vancomycin and d-Ala restores the efficacy of vancomycin and might be a wise and simple way to combat vancomycin intermediate-resistant S. aureus.


Subject(s)
Anti-Bacterial Agents/pharmacology , Isotope Labeling/methods , Lanthanoid Series Elements/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Vancomycin/pharmacology , Alanine/analogs & derivatives , Alanine/analysis , Alanine/pharmacology , Europium/chemistry , Gadolinium/chemistry , Leucine/analogs & derivatives , Leucine/analysis , Methionine/analogs & derivatives , Methionine/analysis , Microbial Viability/drug effects , Peptidoglycan/chemistry , Peptidoglycan/metabolism , Samarium/chemistry , Stereoisomerism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
4.
Nat Chem Biol ; 14(6): 601-608, 2018 06.
Article in English | MEDLINE | ID: mdl-29662210

ABSTRACT

Identifying targets of antibacterial compounds remains a challenging step in the development of antibiotics. We have developed a two-pronged functional genomics approach to predict mechanism of action that uses mutant fitness data from antibiotic-treated transposon libraries containing both upregulation and inactivation mutants. We treated a Staphylococcus aureus transposon library containing 690,000 unique insertions with 32 antibiotics. Upregulation signatures identified from directional biases in insertions revealed known molecular targets and resistance mechanisms for the majority of these. Because single-gene upregulation does not always confer resistance, we used a complementary machine-learning approach to predict the mechanism from inactivation mutant fitness profiles. This approach suggested the cell wall precursor Lipid II as the molecular target of the lysocins, a mechanism we have confirmed. We conclude that docking to membrane-anchored Lipid II precedes the selective bacteriolysis that distinguishes these lytic natural products, showing the utility of our approach for nominating the antibiotic mechanism of action.


Subject(s)
Anti-Bacterial Agents/chemistry , Gene Library , Lipids/chemistry , Staphylococcus aureus/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Cell Wall , Computational Biology , DNA Transposable Elements , Genome, Bacterial , Lysobacter , Machine Learning , Mutation , Peptides, Cyclic/chemistry , Up-Regulation , Uridine Diphosphate N-Acetylmuramic Acid/chemistry
5.
Chemistry ; 25(64): 14572-14582, 2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31599485

ABSTRACT

Natural products that target lipid II, such as the lantibiotic nisin, are strategically important in the development of new antibacterial agents to combat the rise of antimicrobial resistance. Understanding the structural factors that govern the highly selective molecular recognition of lipid II by the N-terminal region of nisin, nisin(1-12), is a crucial step in exploiting the potential of such compounds. In order to elucidate the relationships between amino acid sequence and conformation of this bicyclic peptide fragment, we have used solid-phase peptide synthesis to prepare two novel analogues of nisin(1-12) in which the dehydro residues have been replaced. We have carried out an NMR ensemble analysis of one of these analogues and of the wild-type nisin(1-12) peptide in order to compare the conformations of these two bicyclic peptides. Our analysis has shown the effects of residue mutation on ring conformation. We have also demonstrated that the individual rings of nisin(1-12) are pre-organised to an extent for binding to the pyrophosphate group of lipid II, with a high degree of flexibility exhibited in the central amide bond joining the two rings.


Subject(s)
Nisin/analogs & derivatives , Peptides/chemical synthesis , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Amino Acid Sequence , Hydrogen Bonding , Nisin/metabolism , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Peptides/metabolism , Protein Conformation , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
6.
Bioorg Med Chem Lett ; 29(14): 1849-1853, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31126852

ABSTRACT

Lipid II is an essential precursor of bacterial cell wall biosynthesis and an attractive target for antibiotics. Lipid II is comprised of specialized lipid (bactoprenol) linked to a hydrophilic head group consisting of a peptidoglycan subunit (N-acetylglucosamine (GlcNAc)-N-acetylmuramic acid (MurNAc) disaccharide coupled to a short pentapeptide moiety) via a pyrophosphate. We previously identified a (E)-2,4-bis(4-bromophenyl)-6-(4-(dimethylamino)styryl)pyrylium boron tetrafluoride salt, termed 6jc48-1, that interacts with the MurNAc moiety, the phosphate cage and the isoprenyl tail of Lipid II. Here, we report on the structure-activity relationship of 6jc48-1 derivatives obtained by de novo chemical synthesis. Our results indicate that bacterial killing is positively driven by bi-phenyl stacking with peptidoglycan units. Replacement of bromides by fluorides resulted in activity against S. aureus without affecting Lipid II binding and cytotoxicity. Antibacterial activity was affected negatively by extended interaction of the scaffold with Lipid II isoprenyl units.


Subject(s)
Drug Development/methods , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Structure-Activity Relationship , Uridine Diphosphate N-Acetylmuramic Acid/chemistry
7.
Molecules ; 24(11)2019 Jun 06.
Article in English | MEDLINE | ID: mdl-31174247

ABSTRACT

Mammalian protein N-glycosylation requires the transfer of an oligosaccharide containing 2 residues of N-acetylglucosamine, 9 residues of mannose and 3 residues of glucose (Glc3Man9 GlcNAc2) from Glc3Man9GlcNAc2-diphospho (PP)-dolichol (DLO) onto proteins in the endoplasmic reticulum (ER). Under some pathophysiological conditions, DLO biosynthesis is perturbed, and truncated DLO is hydrolyzed to yield oligosaccharyl phosphates (OSP) via unidentified mechanisms. DLO diphosphatase activity (DLODP) was described in vitro, but its characterization is hampered by a lack of convenient non-radioactive substrates. Our objective was to develop a fluorescence-based assay for DLO hydrolysis. Using a vancomycin-based solid-phase extraction procedure coupled with thin layer chromatography (TLC) and mass spectrometry, we demonstrate that mouse liver membrane extracts hydrolyze fluorescent bacterial lipid II (LII: GlcNAc-MurNAc(dansyl-pentapeptide)-PP-undecaprenol) to yield GlcNAc-MurNAc(dansyl-pentapeptide)-P (GM5P). GM5P production by solubilized liver microsomal proteins shows similar biochemical characteristics to those reported for human hepatocellular carcinoma HepG2 cell DLODP activity. To conclude, we show, for the first time, hydrolysis of lipid II by a eukaryotic enzyme. As LII and DLO are hydrolyzed by the same, or closely related, enzymes, fluorescent lipid II analogs are convenient non-radioactive substrates for investigating DLODP and DLODP-like activities.


Subject(s)
Acetylglucosamine/chemistry , Endoplasmic Reticulum/chemistry , Liver/chemistry , Oligosaccharides/chemistry , Animals , Bacteria/chemistry , Endoplasmic Reticulum/metabolism , Eukaryotic Cells/chemistry , Eukaryotic Cells/metabolism , Glucose/chemistry , Glycosylation , Hep G2 Cells , Humans , Hydrolysis , Lipids/chemistry , Liver/metabolism , Mannose/chemistry , Mice , Oligosaccharides/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/chemistry
8.
Proteins ; 86(12): 1306-1312, 2018 12.
Article in English | MEDLINE | ID: mdl-30242905

ABSTRACT

The crystal structure of UDP-N-acetylglucosamine 4-epimerase (UDP-GlcNAc 4-epimerase; WbpP; EC 5.1.3.7), from the archaeal methanogen Methanobrevibacter ruminantium strain M1, was determined to a resolution of 1.65 Å. The structure, with a single monomer in the crystallographic asymmetric unit, contained a conserved N-terminal Rossmann-fold for nucleotide binding and an active site positioned in the C-terminus. UDP-GlcNAc 4-epimerase is a member of the short-chain dehydrogenases/reductases superfamily, sharing sequence motifs and structural elements characteristic of this family of oxidoreductases and bacterial 4-epimerases. The protein was co-crystallized with coenzyme NADH and UDP-N-acetylmuramic acid, the latter an unintended inclusion and well known product of the bacterial enzyme MurB and a critical intermediate for bacterial cell wall synthesis. This is a non-native UDP sugar amongst archaea and was most likely incorporated from the E. coli expression host during purification of the recombinant enzyme.


Subject(s)
Archaeal Proteins/chemistry , Carbohydrate Epimerases/chemistry , Methanobrevibacter/enzymology , Models, Molecular , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Archaeal Proteins/genetics , Carbohydrate Epimerases/genetics , Catalytic Domain , Crystallography, X-Ray , Escherichia coli/metabolism , NAD/chemistry , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
9.
Bioorg Med Chem Lett ; 28(16): 2708-2712, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29602680

ABSTRACT

Described here is the asymmetric synthesis of iminosugar 2b, a Lipid II analog, designed to mimic the transition state of transglycosylation catalyzed by the bacterial transglycosylase. The high density of functional groups, together with a rich stereochemistry, represents an extraordinary challenge for chemical synthesis. The key 2,6-anti- stereochemistry of the iminosugar ring was established through an iridium-catalyzed asymmetric allylic amination. The developed synthetic route is suitable for the synthesis of focused libraries to enable the structure-activity relationship study and late-stage modification of iminosugar scaffold with variable lipid, peptide and sugar substituents. Compound 2b showed 70% inhibition of transglycosylase from Acinetobacter baumannii, providing a basis for further improvement.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Glycosyltransferases/antagonists & inhibitors , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Acinetobacter baumannii/enzymology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Glycosyltransferases/metabolism , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship , Uridine Diphosphate N-Acetylmuramic Acid/chemical synthesis , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/pharmacology
10.
Chem Pharm Bull (Tokyo) ; 66(1): 84-95, 2018.
Article in English | MEDLINE | ID: mdl-29311516

ABSTRACT

A solid-phase synthesis of Park nucleotide as well as lipids I and II analogues, which is applicable to the synthesis of a range of analogues, is described in this work. This technique allows highly functionalized macromolecules to be modularly labeled. Multiple steps are used in a short time (4 d) with a single purification step to synthesize the molecules by solid-phase synthesis.


Subject(s)
Monosaccharides/chemical synthesis , Nucleotides/chemical synthesis , Oligopeptides/chemical synthesis , Solid-Phase Synthesis Techniques , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Molecular Conformation , Monosaccharides/chemistry , Nucleotides/chemistry , Oligopeptides/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/chemical synthesis , Uridine Diphosphate N-Acetylmuramic Acid/chemistry
11.
Biochemistry ; 56(4): 612-622, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28040891

ABSTRACT

Induction of vancomycin resistance in vancomycin-resistant enterococci (VRE) involves replacement of the d-Ala-d-Ala terminus of peptidoglycan (PG) stems with d-Ala-d-Lac, dramatically reducing the binding affinity of vancomycin for lipid II. Effects from vancomycin resistance induction in Enterococcus faecalis (ATCC 51299) were characterized using a combined solid-state nuclear magnetic resonance (NMR) and liquid chromatography-mass spectrometry (LC-MS) analysis. Solid-state NMR directly measured the total amounts of d-Lac and l,d-Ala metabolized from [2-13C]pyruvate, accumulated Park's nucleotide, and changes to the PG bridge-linking density during the early exponential growth phase (OD660 = 0.4) in intact whole cells of VRE. A high level of accumulation of depsipeptide-substituted Park's nucleotide consistent with the inhibition of the transglycosylation step of PG biosynthesis during the initial phase of vancomycin resistance was observed, while no changes to the PG bridge-linking density following the induction of vancomycin resistance were detected. This indicated that the attachment of the PG bridge to lipid II by the peptidyl transferases was not inhibited by the d-Ala-d-Lac-substituted PG stem structure in VRE. Compositions of mutanolysin-digested isolated cell walls of VRE grown with and without vancomycin resistance induction were determined by LC-MS. Muropeptides with PG stems terminating in d-Ala-d-Lac were found only in VRE grown in the presence of vancomycin. Percentages of muropeptides with a pentapeptide stem terminating in d-Ala-d-Lac for VRE grown in the presence of vancomycin were 26% for the midexponential phase (OD660 = 0.6) and 57% for the stationary growth phase (OD660 = 1.0). These high percentages indicate that d-Ala-d-Lac-substituted lipid II was efficiently utilized for PG biosynthesis in VRE.


Subject(s)
Alanine/analogs & derivatives , Cell Wall/metabolism , Dipeptides/metabolism , Enterococcus faecalis/metabolism , Lactates/metabolism , Peptidoglycan/isolation & purification , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Alanine/chemistry , Alanine/metabolism , Anti-Bacterial Agents/pharmacology , Cell Wall/chemistry , Cell Wall/drug effects , Dipeptides/chemistry , Endopeptidases/pharmacology , Enterococcus faecalis/chemistry , Enterococcus faecalis/drug effects , Lactates/chemistry , Magnetic Resonance Spectroscopy/methods , Mass Spectrometry , Peptidoglycan/chemistry , Peptidoglycan/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism , Vancomycin/pharmacology , Vancomycin Resistance
12.
Biochemistry ; 56(37): 4992-5001, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28825809

ABSTRACT

Defensins make up a class of cysteine-rich antimicrobial peptides, expressed by virtually all eukaryotes as part of their innate immune response. Because of their unique mode of action and rapid killing of pathogenic microbes, defensins are considered promising alternatives to clinically applied antibiotics. Copsin is a defensin-like peptide, previously identified in the mushroom Coprinopsis cinerea. It exerts its activity against a range of Gram-positive bacteria by binding to the peptidoglycan precursor lipid II and prevention of proper cell wall formation. In this study, we present a new workflow for the generation, production, and activity-driven selection of copsin derivatives, based on their expression in Pichia pastoris. One hundred fifty-two single-amino acid mutants and combinations thereof allowed the identification of k-copsin, a peptide variant exhibiting significantly enhanced activity against Bacillus subtilis and Staphylococcus aureus. Furthermore, we performed in silico characterizations of membrane interactions of copsin and k-copsin, in the presence and absence of lipid II. The molecular dynamics data highlighted a high variability in lipid II binding, with a preference for the MurNAc moiety with 47 and 35% of the total contacts for copsin and k-copsin, respectively. Mutated amino acids were located in loop regions of k-copsin and shown to be crucial in the perturbation of the bacterial membrane. These structural studies provide a better understanding of how defensins can be developed toward antibacterial therapies less prone to resistance issues.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacillus subtilis/drug effects , Defensins/pharmacology , Drug Design , Fungal Proteins/pharmacology , Models, Molecular , Staphylococcus aureus/drug effects , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Agaricales/metabolism , Amino Acid Substitution , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Bacillus subtilis/growth & development , Binding Sites , Cell Wall/drug effects , Cell Wall/metabolism , Computational Biology , Defensins/chemistry , Defensins/metabolism , Expert Systems , Fermentation , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Point Mutation , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Staphylococcus aureus/growth & development , Structure-Activity Relationship , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
13.
Biochim Biophys Acta ; 1858(5): 947-57, 2016 May.
Article in English | MEDLINE | ID: mdl-26523408

ABSTRACT

Understanding the mode of action of antibiotics is becoming more and more important in the time that microorganisms start to develop resistance. One very well validated target of several classes of antibiotics is the peptidoglycan precursor lipid II. In this review different classes of lipid II targeting antibiotics will be discussed in detail, including the lantibiotics, human invertebrate defensins and the recently discovered teixobactin. By hitting bacteria where it hurts, at the level of lipid II, we expect to be able to develop efficient antibacterial agents in the future. This article is part of a Special Issue entitled: Antimicrobial peptides edited by Karl Lohner and Kai Hilpert.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacteriocins/pharmacology , Defensins/pharmacology , Depsipeptides/pharmacology , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/isolation & purification , Bacillus subtilis/chemistry , Bacillus subtilis/metabolism , Bacteriocins/chemistry , Bacteriocins/isolation & purification , Defensins/chemistry , Defensins/isolation & purification , Depsipeptides/chemistry , Depsipeptides/isolation & purification , Escherichia coli/drug effects , Escherichia coli/growth & development , Lactococcus lactis/chemistry , Lactococcus lactis/metabolism , Molecular Docking Simulation , Molecular Sequence Data , Peptidoglycan/biosynthesis , Peptidoglycan/chemistry , Staphylococcus/chemistry , Staphylococcus/metabolism , Streptomyces/chemistry , Streptomyces/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/antagonists & inhibitors , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
14.
Biochim Biophys Acta ; 1858(5): 958-70, 2016 May.
Article in English | MEDLINE | ID: mdl-26498397

ABSTRACT

Antimicrobial peptides and proteins (AMPs) are widespread in the living kingdom. They are key effectors of defense reactions and mediators of competitions between organisms. They are often cationic and amphiphilic, which favors their interactions with the anionic membranes of microorganisms. Several AMP families do not directly alter membrane integrity but rather target conserved components of the bacterial membranes in a process that provides them with potent and specific antimicrobial activities. Thus, lipopolysaccharides (LPS), lipoteichoic acids (LTA) and the peptidoglycan precursor Lipid II are targeted by a broad series of AMPs. Studying the functional diversity of immune effectors tells us about the essential residues involved in AMP mechanism of action. Marine invertebrates have been found to produce a remarkable diversity of AMPs. Molluscan defensins and crustacean anti-LPS factors (ALF) are diverse in terms of amino acid sequence and show contrasted phenotypes in terms of antimicrobial activity. Their activity is directed essentially against Gram-positive or Gram-negative bacteria due to their specific interactions with Lipid II or Lipid A, respectively. Through those interesting examples, we discuss here how sequence diversity generated throughout evolution informs us on residues required for essential molecular interaction at the bacterial membranes and subsequent antibacterial activity. Through the analysis of molecular variants having lost antibacterial activity or shaped novel functions, we also discuss the molecular bases of functional divergence in AMPs. This article is part of a Special Issue entitled: Antimicrobial peptides edited by Karl Lohner and Kai Hilpert.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Defensins/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Teichoic Acids/antagonists & inhibitors , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/isolation & purification , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Crustacea/chemistry , Crustacea/physiology , Defensins/chemistry , Defensins/isolation & purification , Drug Resistance, Multiple, Bacterial/drug effects , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Gram-Positive Bacteria/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Molecular Sequence Data , Mollusca/chemistry , Mollusca/physiology , Sequence Alignment , Structure-Activity Relationship , Teichoic Acids/chemistry , Teichoic Acids/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/antagonists & inhibitors , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
15.
Biochemistry ; 55(23): 3303-14, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27158738

ABSTRACT

Lipid II is critical for peptidoglycan synthesis, which is the main component of the bacterial cell wall. Lipid II is a relatively conserved and important part of the cell wall biosynthesis pathway and is targeted by antibiotics such as the lantibiotics, which achieve their function by disrupting the biosynthesis of the cell wall. Given the urgent need for development of novel antibiotics to counter the growing threat of bacterial infection resistance, it is imperative that a thorough molecular-level characterization of the molecules targeted by antibiotics be achieved. To this end, we present a molecular dynamics simulation study of the conformational dynamics of Lipid II within a detailed model of the Staphylococcus aureus cell membrane. We show that Lipid II is able to adopt a range of conformations, even within the packed lipidic environment of the membrane. Our simulations also reveal dimerization of Lipid II mediated by cations. In the presence of the defensin peptide plectasin, the conformational lability of Lipid II allows it to form loose complexes with the protein, via a number of different binding modes.


Subject(s)
Cell Membrane/metabolism , Molecular Dynamics Simulation , Peptides/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Binding Sites , Models, Molecular , Peptides/chemistry , Protein Conformation , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
16.
J Biol Chem ; 290(5): 2630-43, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25480792

ABSTRACT

Inducible expression of chromosomal AmpC ß-lactamase is a major cause of ß-lactam antibiotic resistance in the Gram-negative bacteria Pseudomonas aeruginosa and Enterobacteriaceae. AmpC expression is induced by the LysR-type transcriptional regulator (LTTR) AmpR, which activates ampC expression in response to changes in peptidoglycan (PG) metabolite levels that occur during exposure to ß-lactams. Under normal conditions, AmpR represses ampC transcription by binding the PG precursor UDP-N-acetylmuramic acid (MurNAc)-pentapeptide. When exposed to ß-lactams, however, PG catabolites (1,6-anhydroMurNAc-peptides) accumulate in the cytosol, which have been proposed to competitively displace UDP-MurNAc-pentapeptide from AmpR and convert it into an activator of ampC transcription. Here we describe the molecular interactions between AmpR (from Citrobacter freundii), its DNA operator, and repressor UDP-MurNAc-pentapeptide. Non-denaturing mass spectrometry revealed AmpR to be a homotetramer that is stabilized by DNA containing the T-N11-A LTTR binding motif and revealed that it can bind four repressor molecules in an apparently stepwise manner. A crystal structure of the AmpR effector-binding domain bound to UDP-MurNAc-pentapeptide revealed that the terminal D-Ala-D-Ala motif of the repressor forms the primary contacts with the protein. This observation suggests that 1,6-anhydroMurNAc-pentapeptide may convert AmpR into an activator of ampC transcription more effectively than 1,6-anhydroMurNAc-tripeptide (which lacks the D-Ala-D-Ala motif). Finally, small angle x-ray scattering demonstrates that the AmpR·DNA complex adopts a flat conformation similar to the LTTR protein AphB and undergoes only a slight conformational change when binding UDP-MurNAc-pentapeptide. Modeling the AmpR·DNA tetramer bound to UDP-MurNAc-pentapeptide predicts that the UDP-MurNAc moiety of the repressor participates in modulating AmpR function.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Dipeptides/chemistry , Dipeptides/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , beta-Lactamases/metabolism , Peptidoglycan/metabolism , Protein Binding , Scattering, Small Angle , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
17.
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
18.
Chemistry ; 22(36): 12606-16, 2016 Aug 26.
Article in English | MEDLINE | ID: mdl-27388768

ABSTRACT

Research into antibacterial agents has recently gathered pace in light of the disturbing crisis of antimicrobial resistance. The development of modern tools offers the opportunity of reviving the fallen era of antibacterial discovery through uncovering novel lead compounds that target vital bacterial cell components, such as lipid II. This paper provides a summary of the role of lipid II as well as an overview and insight into the structural features of macrocyclic peptides that inhibit this bacterial cell wall component. The recent discovery of teixobactin, a new class of lipid II inhibitor has generated substantial research interests. As such, the significant progress that has been achieved towards its development as a promising antibacterial agent is discussed.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cell Wall/drug effects , Depsipeptides/pharmacology , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Anti-Bacterial Agents/chemistry , Depsipeptides/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/antagonists & inhibitors , Uridine Diphosphate N-Acetylmuramic Acid/chemistry
19.
Angew Chem Int Ed Engl ; 55(29): 8401-4, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27225706

ABSTRACT

ß-Lactams represent one of the most important classes of antibiotics discovered to date. These agents block Lipid II processing and cell wall biosynthesis through inactivation of penicillin-binding proteins (PBPs). PBPs enzymatically load cell wall building blocks from Lipid II carrier molecules onto the growing cell wall scaffold during growth and division. Lipid II, a bottleneck in cell wall biosynthesis, is the target of some of the most potent antibiotics in clinical use. Despite the immense therapeutic value of this biosynthetic pathway, the PBP-Lipid II association has not been established in live cells. To determine this key interaction, we designed an unnatural d-amino acid dipeptide that is metabolically incorporated into Lipid II molecules. By hijacking the peptidoglycan biosynthetic machinery, photoaffinity probes were installed in combination with click partners within Lipid II, thereby allowing, for the first time, demonstration of PBP interactions in vivo with Lipid II.


Subject(s)
Penicillin-Binding Proteins/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Bacillus subtilis/cytology , Bacillus subtilis/metabolism , Cell Wall/metabolism , Molecular Structure , Penicillin-Binding Proteins/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
20.
Angew Chem Int Ed Engl ; 55(43): 13553-13557, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27667506

ABSTRACT

RNA functionalization is challenging due to the instability of RNA and the limited range of available enzymatic reactions. We developed a strategy based on solid phase synthesis and post-functionalization to introduce an electrophilic site at the 3' end of tRNA analogues. The squarate diester used as an electrophile enabled sequential amidation and provided asymmetric squaramides with high selectivity. The squaramate-RNAs specifically reacted with the lysine of UDP-MurNAc-pentapeptide, a peptidoglycan precursor used by the aminoacyl-transferase FemXWv for synthesis of the bacterial cell wall. The peptidyl-RNA obtained with squaramate-RNA and unprotected UDP-MurNAc-pentapeptide efficiently inhibited FemXWv . The squaramate unit also promoted specific cross-linking of RNA to the catalytic Lys of FemXWv but not to related transferases recognizing different aminoacyl-tRNAs. Thus, squaramate-RNAs provide specificity for cross-linking with defined groups in complex biomolecules due to its unique reactivity.


Subject(s)
Aminoacyltransferases/metabolism , Cross-Linking Reagents/metabolism , Peptides/metabolism , RNA, Transfer/metabolism , RNA/biosynthesis , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Aminoacyltransferases/chemistry , Cross-Linking Reagents/chemistry , Models, Molecular , Molecular Conformation , Peptides/chemistry , RNA/chemistry , RNA, Transfer/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL