Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nucleic Acids Res ; 50(20): 11415-11425, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36350642

ABSTRACT

Xenobiotic nucleic acids (XNAs) offer tremendous potential for synthetic biology, biotechnology, and molecular medicine but their ability to mimic nucleic acids still needs to be explored. Here, to study the ability of XNA oligonucleotides to mimic tRNA, we synthesized three L-Ala-tXNAs analogs. These molecules were used in a non-ribosomal peptide synthesis involving a bacterial Fem transferase. We compared the ability of this enzyme to use amino-acyl tXNAs containing 1',5'-anhydrohexitol (HNA), 2'-fluoro ribose (2'F-RNA) and 2'-fluoro arabinose. L-Ala-tXNA containing HNA or 2'F-RNA were substrates of the Fem enzyme. The synthesis of peptidyl-XNA and the resolution of their structures in complex with the enzyme show the impact of the XNA on protein binding. For the first time we describe functional tXNA in an in vitro assay. These results invite to test tXNA also as substitute for tRNA in translation.


Subject(s)
Amino Acids , RNA, Transfer, Ala , Nucleic Acids/chemistry , Oligonucleotides/chemistry , Peptides , RNA, Transfer, Ala/chemistry
2.
Nucleic Acids Res ; 49(2): 684-699, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33367813

ABSTRACT

The sequence of tRNAs is submitted to evolutionary constraints imposed by their multiple interactions with aminoacyl-tRNA synthetases, translation elongation factor Tu in complex with GTP (EF-Tu•GTP), and the ribosome, each being essential for accurate and effective decoding of messenger RNAs. In Staphylococcus aureus, an additional constraint is imposed by the participation of tRNAGly isoacceptors in the addition of a pentaglycine side chain to cell-wall peptidoglycan precursors by transferases FmhB, FemA and FemB. Three tRNAGly isoacceptors poorly interacting with EF-Tu•GTP and the ribosome were previously identified. Here, we show that these 'non-proteogenic' tRNAs are preferentially recognized by FmhB based on kinetic analyses and on synthesis of stable aminoacyl-tRNA analogues acting as inhibitors. Synthesis of chimeric tRNAs and of helices mimicking the tRNA acceptor arms revealed that this discrimination involves identity determinants exclusively present in the D and T stems and loops of non-proteogenic tRNAs, which belong to an evolutionary lineage only present in the staphylococci. EF-Tu•GTP competitively inhibited FmhB by sequestration of 'proteogenic' aminoacyl-tRNAs in vitro. Together, these results indicate that competition for the Gly-tRNAGly pool is restricted by both limited recognition of non-proteogenic tRNAs by EF-Tu•GTP and limited recognition of proteogenic tRNAs by FmhB.


Subject(s)
Peptidoglycan/biosynthesis , RNA, Bacterial/metabolism , RNA, Transfer, Gly/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/metabolism , Base Sequence , Binding, Competitive , Cell Wall/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Nucleic Acid Conformation , Peptide Elongation Factor Tu/metabolism , Protein Binding
3.
PLoS Pathog ; 15(1): e1007537, 2019 01.
Article in English | MEDLINE | ID: mdl-30668586

ABSTRACT

c-di-AMP is an important second messenger molecule that plays a pivotal role in regulating fundamental cellular processes, including osmotic and cell wall homeostasis in many Gram-positive organisms. In the opportunistic human pathogen Staphylococcus aureus, c-di-AMP is produced by the membrane-anchored DacA enzyme. Inactivation of this enzyme leads to a growth arrest under standard laboratory growth conditions and a re-sensitization of methicillin-resistant S. aureus (MRSA) strains to ß-lactam antibiotics. The gene coding for DacA is part of the conserved three-gene dacA/ybbR/glmM operon that also encodes the proposed DacA regulator YbbR and the essential phosphoglucosamine mutase GlmM, which is required for the production of glucosamine-1-phosphate, an early intermediate of peptidoglycan synthesis. These three proteins are thought to form a complex in vivo and, in this manner, help to fine-tune the cellular c-di-AMP levels. To further characterize this important regulatory complex, we conducted a comprehensive structural and functional analysis of the S. aureus DacA and GlmM enzymes by determining the structures of the S. aureus GlmM enzyme and the catalytic domain of DacA. Both proteins were found to be dimers in solution as well as in the crystal structures. Further site-directed mutagenesis, structural and enzymatic studies showed that multiple DacA dimers need to interact for enzymatic activity. We also show that DacA and GlmM form a stable complex in vitro and that S. aureus GlmM, but not Escherichia coli or Pseudomonas aeruginosa GlmM, acts as a strong inhibitor of DacA function without the requirement of any additional cellular factor. Based on Small Angle X-ray Scattering (SAXS) data, a model of the complex revealed that GlmM likely inhibits DacA by masking the active site of the cyclase and preventing higher oligomer formation. Together these results provide an important mechanistic insight into how c-di-AMP production can be regulated in the cell.


Subject(s)
Adenylyl Cyclase Inhibitors/metabolism , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/ultrastructure , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Wall/metabolism , Dinucleoside Phosphates/antagonists & inhibitors , Dinucleoside Phosphates/metabolism , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Operon/genetics , Phosphoglucomutase/metabolism , Phosphorus-Oxygen Lyases/metabolism , Protein Domains , Scattering, Small Angle , Second Messenger Systems/genetics , Staphylococcal Infections/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/physiology , X-Ray Diffraction/methods
4.
J Bacteriol ; 202(23)2020 11 04.
Article in English | MEDLINE | ID: mdl-32958631

ABSTRACT

Colicin M is an enzymatic bacteriocin produced by some Escherichia coli strains which provokes cell lysis of competitor strains by hydrolysis of the cell wall peptidoglycan undecaprenyl-PP-MurNAc(-pentapeptide)-GlcNAc (lipid II) precursor. The overexpression of a gene, cbrA (formerly yidS), was shown to protect E. coli cells from the deleterious effects of this colicin, but the underlying resistance mechanism was not established. We report here that a major structural modification of the undecaprenyl-phosphate carrier lipid and of its derivatives occurred in membranes of CbrA-overexpressing cells, which explains the acquisition of resistance toward this bacteriocin. Indeed, a main fraction of these lipids, including the lipid II peptidoglycan precursor, now displayed a saturated isoprene unit at the α-position, i.e., the unit closest to the colicin M cleavage site. Only unsaturated forms of these lipids were normally detectable in wild-type cells. In vitro and in vivo assays showed that colicin M did not hydrolyze α-saturated lipid II, clearly identifying this substrate modification as the resistance mechanism. These saturated forms of undecaprenyl-phosphate and lipid II remained substrates of the different enzymes participating in peptidoglycan biosynthesis and carrier lipid recycling, allowing this colicin M-resistance mechanism to occur without affecting this essential pathway.IMPORTANCE Overexpression of the chromosomal cbrA gene allows E. coli to resist colicin M (ColM), a bacteriocin specifically hydrolyzing the undecaprenyl-PP-MurNAc(-pentapeptide)-GlcNAc (lipid II) peptidoglycan precursor of targeted cells. This resistance results from a CbrA-dependent modification of the precursor structure, i.e., reduction of the α-isoprenyl bond of C55-carrier lipid moiety that is proximal to ColM cleavage site. This modification, observed here for the first time in eubacteria, annihilates the ColM activity without affecting peptidoglycan biogenesis. These data, which further increase our knowledge of the substrate specificity of this colicin, highlight the capability of E. coli to generate reduced forms of C55-carrier lipid and its derivatives. Whether the function of this modification is only relevant with respect to ColM resistance is now questioned.


Subject(s)
Anti-Bacterial Agents/pharmacology , Colicins/pharmacology , Drug Resistance, Bacterial , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , Flavoproteins/metabolism , Peptidoglycan/metabolism , Polyisoprenyl Phosphates/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Flavoproteins/genetics , Peptidoglycan/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
5.
Microbiology (Reading) ; 166(6): 567-578, 2020 06.
Article in English | MEDLINE | ID: mdl-32375990

ABSTRACT

Clostridium difficile 630 possesses a cryptic but functional gene cluster vanGCd homologous to the vanG operon of Enterococcus faecalis. Expression of vanGCd in the presence of subinhibitory concentrations of vancomycin is accompanied by peptidoglycan amidation on the meso-DAP residue. In this paper, we report the presence of two potential asparagine synthetase genes named asnB and asnB2 in the C. difficile genome whose products were potentially involved in this peptidoglycan structure modification. We found that asnB expression was only induced when C. difficile was grown in the presence of vancomycin, yet independently from the vanGCd resistance and regulation operons. In addition, peptidoglycan precursors were not amidated when asnB was inactivated. No change in vancomycin MIC was observed in the asnB mutant strain. In contrast, overexpression of asnB resulted in the amidation of most of the C. difficile peptidoglycan precursors and in a weak increase of vancomycin susceptibility. AsnB activity was confirmed in E. coli. In contrast, the expression of the second asparagine synthetase, AsnB2, was not induced in the presence of vancomycin. In summary, our results demonstrate that AsnB is responsible for peptidoglycan amidation of C. difficile in the presence of vancomycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aspartate-Ammonia Ligase/metabolism , Bacterial Proteins/metabolism , Clostridioides difficile/drug effects , Clostridioides difficile/enzymology , Peptidoglycan/metabolism , Vancomycin/pharmacology , Aspartate-Ammonia Ligase/genetics , Bacterial Proteins/genetics , Clostridioides difficile/genetics , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial , Genome, Bacterial , Multigene Family , Operon
6.
J Bacteriol ; 201(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30988031

ABSTRACT

Certain Pseudomonas aeruginosa strains produce a homolog of colicin M, namely, PaeM, that specifically inhibits peptidoglycan biosynthesis of susceptible P. aeruginosa strains by hydrolyzing the lipid II intermediate precursor. Two variants of this pyocin were identified whose sequences mainly differed in the N-terminal protein moiety, i.e., the region involved in the binding to the FiuA outer membrane receptor and translocation into the periplasm. The antibacterial activity of these two variants, PaeM1 and PaeM2, was tested against various P. aeruginosa strains comprising reference strains PAO1 and PA14, PaeM-producing strains, and 60 clinical isolates. Seven of these strains, including PAO1, were susceptible to only one variant (2 to PaeM1 and 5 to PaeM2), and 11 were affected by both. The remaining strains, including PA14 and four PaeM1 producers, were resistant to both variants. The differences in the antibacterial spectra of the two PaeM homologs prompted us to investigate the molecular determinants allowing their internalization into P. aeruginosa cells, taking the PAO1 strain that is susceptible to PaeM2 but resistant to PaeM1 as the indicator strain. Heterologous expression of fiuA gene orthologs from different strains into PAO1, site-directed mutagenesis experiments, and construction of PaeM chimeric proteins provided evidence that the cell susceptibility and discrimination differences between the PaeM variants resulted from a polymorphism of both the pyocin and the outer membrane receptor FiuA. Moreover, we found that a third component, TonB1, a protein involved in iron transport in P. aeruginosa, working together with FiuA and the ExbB/ExbD complex, was directly implicated in this discrimination.IMPORTANCE Bacterial antibiotic resistance constitutes a threat to human health, imposing the need for identification of new targets and development of new strategies to fight multiresistant pathogens. Bacteriocins and other weapons that bacteria have themselves developed to kill competitors are therefore of great interest and a valuable source of inspiration for us. Attention was paid here to two variants of a colicin M homolog (PaeM) produced by certain strains of P. aeruginosa that inhibit the growth of their congeners by blocking cell wall peptidoglycan synthesis. Molecular determinants allowing recognition of these pyocins by the outer membrane receptor FiuA were identified, and a receptor polymorphism affecting the susceptibility of P. aeruginosa clinical strains was highlighted, providing new insights into the potential use of these pyocins as an alternative to antibiotics.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Drug Resistance, Bacterial , Polymorphism, Genetic , Pseudomonas aeruginosa/genetics , Pyocins/pharmacology , Anti-Bacterial Agents/pharmacology , Cell Wall/chemistry , Mutagenesis, Site-Directed , Peptidoglycan/chemistry , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Receptors, Cell Surface
7.
J Enzyme Inhib Med Chem ; 34(1): 1010-1017, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31072165

ABSTRACT

The Mur ligases form a series of consecutive enzymes that participate in the intracellular steps of bacterial peptidoglycan biosynthesis. They therefore represent interesting targets for antibacterial drug discovery. MurC, D, E and F are all ATP-dependent ligases. Accordingly, with the aim being to find multiple inhibitors of these enzymes, we screened a collection of ATP-competitive kinase inhibitors, on Escherichia coli MurC, D and F, and identified five promising scaffolds that inhibited at least two of these ligases. Compounds 1, 2, 4 and 5 are multiple inhibitors of the whole MurC to MurF cascade that act in the micromolar range (IC50, 32-368 µM). NMR-assisted binding studies and steady-state kinetics studies performed on aza-stilbene derivative 1 showed, surprisingly, that it acts as a competitive inhibitor of MurD activity towards D-glutamic acid, and additionally, that its binding to the D-glutamic acid binding site is independent of the enzyme closure promoted by ATP.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Ligases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Kinetics , Ligases/metabolism , Molecular Structure , Structure-Activity Relationship
8.
Chemistry ; 24(56): 14911-14915, 2018 Oct 09.
Article in English | MEDLINE | ID: mdl-30020544

ABSTRACT

Conjugation of RNA with multiple partners to obtain mimics of complex biomolecules is limited by the identification of orthogonal reactions. Here, lipid-carbohydrate-peptidyl-RNA conjugates were obtained by post-functionalization reactions, solid-phase synthesis, and enzymatic steps, to generate molecules mimicking the substrates of FmhB, an essential peptidoglycan synthesis enzyme of Staphylococcus aureus. Mimics of Gly-tRNAGly and lipid intermediate II (undecaprenyl-diphospho-disaccharide-pentapeptide) were combined in a single "bi-substrate" inhibitor (IC50 =56 nm). The synthetic route was exploited to generate substrates and inhibitors containing d-lactate residue (d-Lac) instead of d-Ala at the C-terminus of the pentapeptide stem, a modification responsible for vancomycin resistance in the enterococci. The substitution impaired recognition of peptidoglycan precursors by FmhB. The associated fitness cost may account for limited dissemination of vancomycin resistance genes in S. aureus.


Subject(s)
Carbohydrates/chemistry , Cell Wall/enzymology , Enzyme Inhibitors/chemistry , Lipids/chemistry , RNA/chemistry , Solid-Phase Synthesis Techniques/methods , Staphylococcus aureus/enzymology , Bacterial Proteins/antagonists & inhibitors , Carbohydrates/chemical synthesis , Carbohydrates/pharmacology , Cell Wall/drug effects , Cell Wall/metabolism , Drug Discovery , Drug Resistance, Bacterial , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Lipids/chemical synthesis , Lipids/pharmacology , Peptidoglycan/metabolism , RNA/chemical synthesis , RNA/pharmacology , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Substrate Specificity
9.
Chemistry ; 24(22): 5743-5747, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29389045

ABSTRACT

The bacterial cell wall peptidoglycan contains unusual l- and d-amino acids assembled as branched peptides. Insight into the biosynthesis of the polymer has been hampered by limited access to substrates and to suitable polymerization assays. Here we report the full synthesis of the peptide stem of peptidoglycan precursors from two pathogenic bacteria, Enterococcus faecium and Mycobacterium tuberculosis, and the development of a sensitive post-derivatization assay for their cross-linking by l,d-transpeptidases. Access to series of stem peptides showed that amidation of free carboxyl groups is essential for optimal enzyme activity, in particular the amidation of diaminopimelate (DAP) residues for the cross-linking activity of the l,d-transpeptidase LdtMt2 from M. tuberculosis. Accordingly, construction of a conditional mutant established the essential role of AsnB indicating that this DAP amidotransferase is an attractive target for the development of anti-mycobacterial drugs.


Subject(s)
Enterococcus faecium/enzymology , Mycobacterium tuberculosis/enzymology , Peptidoglycan/biosynthesis , Peptidyl Transferases/metabolism , Transaminases/metabolism , Cell Wall/metabolism , Enterococcus faecium/chemistry , Enterococcus faecium/genetics , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Peptidyl Transferases/drug effects , beta-Lactams/chemistry
10.
Bioorg Med Chem Lett ; 27(15): 3529-3533, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28579123

ABSTRACT

MurA is an intracellular bacterial enzyme that is essential for peptidoglycan biosynthesis, and is therefore an important target for antibacterial drug discovery. We report the synthesis, in silico studies and extensive structure-activity relationships of a series of quinazolinone-based inhibitors of MurA from Escherichia coli. 3-Benzyloxyphenylquinazolinones showed promising inhibitory potencies against MurA, in the low micromolar range, with an IC50 of 8µM for the most potent derivative (58). Furthermore, furan-substituted quinazolinones (38, 46) showed promising antibacterial activities, with MICs from 1µg/mL to 8µg/mL, concomitant with their MurA inhibitory potencies. These data represent an important step towards the development of novel antimicrobial agents to combat increasing bacterial resistance.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/enzymology , Quinazolinones/chemistry , Quinazolinones/pharmacology , Alkyl and Aryl Transferases/metabolism , Anti-Bacterial Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Quinazolinones/chemical synthesis , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 27(4): 944-949, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28077258

ABSTRACT

We report on the successful application of ProBiS-CHARMMing web server in the discovery of new inhibitors of MurA, an enzyme that catalyzes the first committed cytoplasmic step of bacterial peptidoglycan synthesis. The available crystal structures of Escherichia coli MurA in the Protein Data Bank have binding sites whose small volume does not permit the docking of drug-like molecules. To prepare the binding site for docking, the ProBiS-CHARMMing web server was used to simulate the induced-fit effect upon ligand binding to MurA, resulting in a larger, more holo-like binding site. The docking of a filtered ZINC compound library to this enlarged binding site was then performed and resulted in three compounds with promising inhibitory potencies against MurA. Compound 1 displayed significant inhibitory potency with IC50 value of 1µM. All three compounds have novel chemical structures, which could be used for further optimization of small-molecule MurA inhibitors.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/metabolism , Carbohydrate Sequence , Drug Discovery , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Peptidoglycan/metabolism
12.
J Biol Chem ; 290(21): 13079-94, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25847251

ABSTRACT

A gene named ltsA was earlier identified in Rhodococcus and Corynebacterium species while screening for mutations leading to increased cell susceptibility to lysozyme. The encoded protein belonged to a huge family of glutamine amidotransferases whose members catalyze amide nitrogen transfer from glutamine to various specific acceptor substrates. We here describe detailed physiological and biochemical investigations demonstrating the specific role of LtsA protein from Corynebacterium glutamicum (LtsACg) in the modification by amidation of cell wall peptidoglycan diaminopimelic acid (DAP) residues. A morphologically altered but viable ΔltsA mutant was generated, which displays a high susceptibility to lysozyme and ß-lactam antibiotics. Analysis of its peptidoglycan structure revealed a total loss of DAP amidation, a modification that was found in 80% of DAP residues in the wild-type polymer. The cell peptidoglycan content and cross-linking were otherwise not modified in the mutant. Heterologous expression of LtsACg in Escherichia coli yielded a massive and toxic incorporation of amidated DAP into the peptidoglycan that ultimately led to cell lysis. In vitro assays confirmed the amidotransferase activity of LtsACg and showed that this enzyme used the peptidoglycan lipid intermediates I and II but not, or only marginally, the UDP-MurNAc pentapeptide nucleotide precursor as acceptor substrates. As is generally the case for glutamine amidotransferases, either glutamine or NH4(+) could serve as the donor substrate for LtsACg. The enzyme did not amidate tripeptide- and tetrapeptide-truncated versions of lipid I, indicating a strict specificity for a pentapeptide chain length.


Subject(s)
Amides/chemistry , Bacterial Proteins/metabolism , Corynebacterium/metabolism , Diaminopimelic Acid/chemistry , Muramidase/metabolism , Peptidoglycan/metabolism , Transaminases/metabolism , Amides/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Blotting, Western , Cell Wall/metabolism , Cells, Cultured , Corynebacterium/genetics , Corynebacterium/growth & development , Diaminopimelic Acid/metabolism , Immunoenzyme Techniques , Microscopy, Electron, Transmission , Molecular Sequence Data , Mutation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transaminases/genetics
13.
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
14.
Arch Microbiol ; 197(3): 371-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25417006

ABSTRACT

Antibiotic resistance has emerged as a real threat to mankind, rendering many compounds ineffective in the fight against bacterial infection, including for significant diseases such as plague caused by Yersinia pestis. Essential genes have been identified as promising targets for inhibiting with new classes of compounds. Previously, the gene encoding the bifunctional UDP-N-acetylglucosamine pyrophosphorylase/glucosamine-1-phosphate N-acetyltransferase enzyme GlmU was confirmed as an essential gene in Yersinia. As a step towards exploiting this target for antimicrobial screening, we undertook a biochemical characterisation of the Yersinia GlmU. Effects of pH and magnesium concentration on the acetyltransferase and uridyltransferase activities were analysed, and kinetic parameters were determined. The acetyltransferase activity, which is strongly increased in the presence of reducing agent, was shown to be susceptible to oxidation and thiol-specific reagents.


Subject(s)
Acetyltransferases/isolation & purification , Acetyltransferases/metabolism , Nucleotidyltransferases/isolation & purification , Nucleotidyltransferases/metabolism , Yersinia pestis/enzymology , Acetyltransferases/chemistry , Acetyltransferases/genetics , Amino Acid Sequence , Enzyme Activation/drug effects , Escherichia coli/genetics , Hydrogen-Ion Concentration , Kinetics , Magnesium/pharmacology , Mercaptoethanol/pharmacology , Molecular Sequence Data , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Oxidants/pharmacology , Oxidation-Reduction , Sequence Alignment , Yersinia pestis/genetics
15.
Mol Microbiol ; 89(4): 612-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23782343

ABSTRACT

vanGCd, a cryptic gene cluster highly homologous to the vanG gene cluster of Enterococcus faecalis is largely spread in Clostridium difficile. Since emergence of vancomycin resistance would have dramatic clinical consequences, we have evaluated the capacity of the vanGCd cluster to confer resistance. We showed that expression of vanGCd is inducible by vancomycin and that VanGCd , VanXYCd and VanTCd are functional, exhibiting D-Ala : D-Ser ligase, D,D-dipeptidase and D-Ser racemase activities respectively. In other bacteria, these enzymes are sufficient to promote vancomycin resistance. Trans-complementation of C. difficile with the vanC resistance operon of Enterococcus gallinarum faintly impacted the MIC of vancomycin, but did not promote vancomycin resistance in C. difficile. Sublethal concentration of vancomycin led to production of UDP-MurNAc-pentapeptide[D-Ser], suggesting that the vanGCd gene cluster is able to modify the peptidoglycan precursors. Our results indicated amidation of UDP-MurNAc-tetrapeptide, UDP-MurNAc-pentapeptide[D-Ala] and UDP-MurNAc-pentapeptide[D-Ser]. This modification is passed on the mature peptidoglycan where a muropeptide Tetra-Tetra is amidated on the meso-diaminopimelic acid. Taken together, our results suggest that the vanGCd gene cluster is functional and is prevented from promoting vancomycin resistance in C. difficile.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Genes, Bacterial , Multigene Family , Vancomycin Resistance , Vancomycin/pharmacology , Bacteria , Bacterial Proteins/genetics , Drug Resistance, Microbial/genetics , Enterococcus , Enterococcus faecalis , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Genetic Complementation Test , Microbial Sensitivity Tests , Peptide Synthases/genetics
16.
J Biol Chem ; 287(44): 37395-405, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22977250

ABSTRACT

Colicin M (ColM) is the only enzymatic colicin reported to date that inhibits cell wall peptidoglycan biosynthesis. It catalyzes the specific degradation of the lipid intermediates involved in this pathway, thereby provoking lysis of susceptible Escherichia coli cells. A gene encoding a homologue of ColM was detected within the exoU-containing genomic island A carried by certain pathogenic Pseudomonas aeruginosa strains. This bacteriocin (pyocin) that we have named PaeM was crystallized, and its structure with and without an Mg(2+) ion bound was solved. In parallel, site-directed mutagenesis of conserved PaeM residues from the C-terminal domain was performed, confirming their essentiality for the protein activity both in vitro (lipid II-degrading activity) and in vivo (cytotoxicity against a susceptible P. aeruginosa strain). Although PaeM is structurally similar to ColM, the conformation of their active sites differs radically; in PaeM, residues essential for enzymatic activity and cytotoxicity converge toward a same pocket, whereas in ColM they are spread along a particularly elongated active site. We have also isolated a minimal domain corresponding to the C-terminal half of the PaeM protein and exhibiting a 70-fold higher enzymatic activity as compared with the full-length protein. This isolated domain of the PaeM bacteriocin was further shown to kill E. coli cells when addressed to the periplasm of these bacteria.


Subject(s)
Bacteriocins/chemistry , Colicins/chemistry , Phosphoric Diester Hydrolases/chemistry , Pseudomonas aeruginosa/metabolism , Amino Acid Sequence , Amino Acid Substitution , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteriocins/metabolism , Bacteriocins/pharmacology , Catalytic Domain , Colicins/metabolism , Colicins/pharmacology , Conserved Sequence , Crystallography, X-Ray , Escherichia coli/drug effects , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/pharmacology , Protein Structure, Secondary , Structural Homology, Protein , Substrate Specificity
17.
Chemistry ; 19(4): 1357-63, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-23197408

ABSTRACT

Peptidyl-RNA conjugates have various applications in studying the ribosome and enzymes participating in tRNA-dependent pathways such as Fem transferases in peptidoglycan synthesis. Herein a convergent synthesis of peptidyl-RNAs based on Huisgen-Sharpless cycloaddition for the final ligation step is developed. Azides and alkynes are introduced into tRNA and UDP-MurNAc-pentapeptide, respectively. Synthesis of 2'-azido RNA helix starts from 2'-azido-2'-deoxyadenosine that is coupled to deoxycytidine by phosphoramidite chemistry. The resulting dinucleotide is deprotected and ligated to a 22-nt RNA helix mimicking the acceptor arm of Ala-tRNA(Ala) by T4 RNA ligase. For alkyne UDP-MurNAc-pentapeptide, meso-cystine is enzymatically incorporated into the peptidoglycan precursor and reduced, and L-Cys is converted to dehydroalanine with O-(mesitylenesulfonyl)hydroxylamine. Reaction of but-3-yne-1-thiol with dehydroalanine affords the alkyne-containing UDP-MurNAc-pentapeptide. The Cu(I)-catalyzed azide alkyne cycloaddition reaction in the presence of tris[(1-hydroxypropyl-1H-1,2,3-triazol-4-yl)methyl]amine provided the peptidyl-RNA conjugate, which was tested as an inhibitor of non-ribosomal FemX(Wv) aminoacyl transferase. The bi-substrate analogue was found to inhibit FemX(Wv) with an IC(50) of (89±9) pM, as both moieties of the peptidyl-RNA conjugate contribute to high-affinity binding.


Subject(s)
Aminoacyltransferases/metabolism , Oligopeptides/chemistry , RNA/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Aminoacyltransferases/antagonists & inhibitors , Catalysis , Copper/chemistry , Cycloaddition Reaction , Kinetics , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Protein Binding , RNA/chemical synthesis , RNA/metabolism , RNA Ligase (ATP)/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
18.
Biochem Soc Trans ; 40(6): 1522-7, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23176510

ABSTRACT

Colicins are proteins produced by some strains of Escherichia coli to kill competitors belonging to the same species. Among them, ColM (colicin M) is the only one that blocks the biosynthesis of peptidoglycan, a specific bacterial cell-wall polymer essential for cell integrity. ColM acts in the periplasm by hydrolysing the phosphoester bond of the peptidoglycan lipid intermediate (lipid II). ColM cytotoxicity is dependent on FkpA of the targeted cell, a chaperone with peptidylprolyl cis-trans isomerase activity. Dissection of ColM was used to delineate the catalytic domain and to identify the active-site residues. The in vitro activity of the isolated catalytic domain towards lipid II was 50-fold higher than that of the full-length bacteriocin. Moreover, this domain was bactericidal in the absence of FkpA under conditions that bypass the import mechanism (FhuA-TonB machinery). Thus ColM undergoes a maturation process driven by FkpA that is not required for the activity of the isolated catalytic domain. Genes encoding proteins with similarity to the catalytic domain of ColM were identified in pathogenic strains of Pseudomonas and other genera. ColM acts on several structures of lipid II representative of the diversity of peptidoglycan chemotypes. All together, these data open the way to the potential use of ColM-related bacteriocins as broad spectrum antibacterial agents.


Subject(s)
Anti-Bacterial Agents/metabolism , Colicins/metabolism , Escherichia coli/enzymology , Peptidoglycan/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antibiosis , Bacteriocins/chemistry , Bacteriocins/metabolism , Bacteriocins/pharmacology , Colicins/chemistry , Colicins/pharmacology , Humans , Models, Molecular , Protein Conformation , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
19.
Arch Microbiol ; 194(6): 505-12, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22231476

ABSTRACT

Chlamydiaceae are obligate intracellular bacteria that do not synthesise detectable peptidoglycan although they possess an almost complete arsenal of genes encoding peptidoglycan biosynthetic activities. In this paper, the murF gene from Chlamydia trachomatis was shown to be capable of complementing a conditional Escherichia coli mutant impaired in UDP-MurNAc-tripeptide:D-Ala-D-Ala ligase activity. Recombinant MurF from C. trachomatis was overproduced and purified from E. coli. It exhibited ATP-dependent UDP-MurNAc-X-γ-D-Glu-meso-A(2)pm:D-Ala-D-Ala ligase activity in vitro. No significant difference of kinetic parameters was seen when X was L-Ala, L-Ser or Gly. The L-Lys-containing UDP-MurNAc-tripeptide was a poorer substrate as compared to the meso-A(2)pm-containing one. Based on the respective substrate specificities of the chlamydial MurC, MurE, MurF and Ddl enzymes, a sequence L-Ala/L-Ser/Gly-γ-D-Glu-meso-A(2)pm-D-Ala-D-Ala is expected for the chlamydial pentapeptide stem, with Gly at position 1 being less likely.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/enzymology , Escherichia coli/metabolism , Ligases/metabolism , Bacterial Proteins/genetics , Chlamydia trachomatis/genetics , Chlamydia trachomatis/metabolism , Dipeptides/metabolism , Escherichia coli/genetics , Genetic Complementation Test , Ligases/genetics , Peptidoglycan/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Transformation, Bacterial
20.
Mol Microbiol ; 75(4): 874-85, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20025663

ABSTRACT

Bypass of the penicillin-binding proteins by an L,D-transpeptidase (Ldt(fm)) confers cross-resistance to beta-lactam and glycopeptide antibiotics in mutants of Enterococcus faecium selected in vitro. Ldt(fm) is produced by the parental strain D344S although it insignificantly contributes to peptidoglycan cross-linking as pentapeptide stems cannot be used as acyl donors by this enzyme. Here we show that production of the tetrapeptide substrate of Ldt(fm) is controlled by a two-component regulatory system (DdcRS) and a metallo-D,D-carboxypeptidase (DdcY). The locus was silent in D344S and its activation was due to amino acid substitutions in DdcS or DdcR that led to production of DdcY and hydrolysis of the C-terminal D-Ala residue of the cytoplasmic peptidoglycan precursor UDP-MurNAc-pentapeptide. The T(161)A and T(161)M substitutions affected a position of DdcS known to be essential for the phosphatase activity of related sensor kinases. Complete elimination of UDP-MurNAc-pentapeptide, which was required specifically for resistance to glycopeptides, involved substitutions in DdcY that increased the catalytic efficiency of the enzyme (E(127)K) and affected its interaction with the cell envelope (I(14)N). The ddc locus displays striking similarities with portions of the van vancomycin resistance gene clusters, suggesting possible routes of emergence of cross-resistance to glycopeptides and beta-lactams in natural conditions.


Subject(s)
Enterococcus faecium/metabolism , Peptidoglycan/metabolism , Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Base Sequence , Cell Wall/metabolism , Drug Resistance, Bacterial , Enterococcus faecium/drug effects , Enterococcus faecium/enzymology , Glycopeptides/pharmacology , Molecular Sequence Data , Peptidoglycan/biosynthesis , Peptidoglycan/chemistry , Proteins/genetics , Substrate Specificity , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism , beta-Lactams/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL