Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Antibiotics (Basel) ; 11(9)2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36139968

ABSTRACT

New inhibitors of the bacterial transferase MraY from Aquifex aeolicus (MraYAA), based on the aminoribosyl uridine central core of known natural MraY inhibitors, have been designed to generate interaction of their oxadiazole linker with the key amino acids (H324 or H325) of the enzyme active site, as observed for the highly potent inhibitors carbacaprazamycin, muraymycin D2 and tunicamycin. A panel of ten compounds was synthetized notably thanks to a robust microwave-activated one-step sequence for the synthesis of the oxadiazole ring that involved the O-acylation of an amidoxime and subsequent cyclization. The synthetized compounds, with various hydrophobic substituents on the oxadiazole ring, were tested against the MraYAA transferase activity. Although with poor antibacterial activity, nine out of the ten compounds revealed the inhibition of the MraYAA activity in the range of 0.8 µM to 27.5 µM.

2.
Molecules ; 27(6)2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35335131

ABSTRACT

New inhibitors of the bacterial tranferase MraY are described. Their structure is based on an aminoribosyl uridine scaffold, which is known to be important for the biological activity of natural MraY inhibitors. A decyl alkyl chain was introduced onto this scaffold through various linkers. The synthesized compounds were tested against the MraYAA transferase activity, and the most active compound with an original (S,S)-tartaric diamide linker inhibits MraY activity with an IC50 equal to 0.37 µM. Their antibacterial activity was also evaluated on a panel of Gram-positive and Gram-negative strains; however, the compounds showed no antibacterial activity. Docking and molecular dynamics studies revealed that this new linker established two stabilizing key interactions with N190 and H325, as observed for the highly potent inhibitors carbacaprazamycin, muraymycin D2 and tunicamycin.


Subject(s)
Diamide , Transferases , Bacterial Proteins/chemistry , Molecular Dynamics Simulation , Transferases/chemistry , Transferases (Other Substituted Phosphate Groups) , Uridine/chemistry , Uridine/pharmacology
3.
Org Biomol Chem ; 19(26): 5844-5866, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34115086

ABSTRACT

The straightforward synthesis of aminoribosyl uridines substituted by a 5'-methylene-urea is described. Their convergent synthesis involves the urea formation from various activated amides and an azidoribosyl uridine substituted at the 5' position by an aminomethyl group. This common intermediate resulted from the diastereoselective glycosylation of a phthalimido uridine derivative with a ribosyl fluoride as a ribosyl donor. The inhibition of the MraY transferase activity by the synthetized 11 urea-containing inhibitors was evaluated and 10 compounds revealed MraY inhibition with IC50 ranging from 1.9 µM to 16.7 µM. Their antibacterial activity was also evaluated on a panel of Gram-positive and Gram-negative bacteria. Four compounds exhibited a good activity against Gram-positive bacterial pathogens with MIC ranging from 8 to 32 µg mL-1, including methicillin resistant Staphylococcus aureus (MRSA) and Enterococcus faecium. Interestingly, one compound also revealed antibacterial activity against Pseudomonas aeruginosa with MIC equal to 64 µg mL-1. Docking experiments predicted two modes of positioning of the active compounds urea chain in different hydrophobic areas (HS2 and HS4) within the MraY active site from Aquifex aeolicus. However, molecular dynamics simulations showed that the urea chain adopts a binding mode similar to that observed in structural model and targets the hydrophobic area HS2.


Subject(s)
Anti-Bacterial Agents
4.
Chem Asian J ; 15(1): 51-55, 2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31686429

ABSTRACT

Monocyclic ß-lactams revive the research field on antibiotics, which are threatened by the emergence of resistant bacteria. A six-step synthetic route was developed, providing easy access to new 3-amino-1-carboxymethyl-4-phenyl-ß-lactams, of which the penicillin-binding protein (PBP) inhibitory potency was demonstrated biochemically.


Subject(s)
Amino Acids/pharmacology , Anti-Bacterial Agents/pharmacology , Dipeptides/pharmacology , Imines/pharmacology , Lactams/pharmacology , Penicillin-Binding Proteins/antagonists & inhibitors , Amino Acids/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dipeptides/chemical synthesis , Dipeptides/chemistry , Imines/chemistry , Lactams/chemical synthesis , Lactams/chemistry , Molecular Structure , Penicillin-Binding Proteins/metabolism
5.
Chemistry ; 25(70): 16128-16140, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31596974

ABSTRACT

Innovative monocyclic ß-lactam entities create opportunities in the battle against resistant bacteria because of their PBP acylation potential, intrinsically high ß-lactamase stability and compact scaffold. α-Benzylidene-substituted 3-amino-1-carboxymethyl-ß-lactams were recently shown to be potent PBP inhibitors and constitute eligible anchor points for synthetic elaboration of the chemical space around the central ß-lactam ring. The present study discloses a 12-step synthesis of ten α-arylmethylidenecarboxylates using a microwave-assisted Wittig olefination as the crucial reaction step. The library was designed aiming at enhanced ß-lactam electrophilicity and extended electron flow after enzymatic attack. Additionally, increased ß-lactamase stability and intermolecular target interaction were envisioned by tackling both the substitution pattern of the aromatic ring and the ß-lactam C4-position. The significance of α-unsaturation was validated and the R39/PBP3 inhibitory potency shown to be augmented the most through decoration of the aromatic ring with electron-withdrawing groups. Furthermore, ring cleavage by representative ß-lactamases was ruled out, providing new insights in the SAR landscape of monocyclic ß-lactams as eligible PBP or ß-lactamase inhibitors.

6.
Nanoscale ; 11(25): 12275-12284, 2019 Jul 07.
Article in English | MEDLINE | ID: mdl-31211302

ABSTRACT

Understanding the molecular mechanism by which the signal of the presence of an antibiotic is transduced from outside to inside the bacterial cell is of fundamental interest for the ß-lactam antibiotic resistance problem, but remains difficult to accomplish. No approach has ever addressed entire penicillin receptors in a membrane environment. Here we describe a method to investigate the purified Bacillus licheniformis BlaR1 receptor -a membrane-bound penicillin receptor involved in ß-lactam resistance- embedded into a lipid bilayer in absence or presence of penicillin. By selecting a mutated receptor blocked in its signal transduction pathway just after its activation by penicillin, we revealed the very first step of receptor signalling by unfolding the receptor from its C-terminal end by AFM-based single-molecule force spectroscopy. We showed that the presence of the antibiotic entails significant conformational changes within the receptor. Our approach opens an avenue to study signal-transduction pathways mediated by membrane-bound proteins in a membrane environment.


Subject(s)
Bacterial Proteins/chemistry , Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Penicillins/chemistry
7.
Chemistry ; 24(57): 15254-15266, 2018 Oct 12.
Article in English | MEDLINE | ID: mdl-29882610

ABSTRACT

As a complement to the renowned bicyclic ß-lactam antibiotics, monocyclic analogues provide a breath of fresh air in the battle against resistant bacteria. In that framework, the present study discloses the in silico design and unprecedented ten-step synthesis of eleven nocardicin-like enantiomerically pure 2-{3-[2-(2-aminothiazol-4-yl)-2-(methoxyimino)acetamido]-2-oxoazetidin-1-yl}acetic acids starting from serine as a readily accessible precursor. The capability of this novel class of monocyclic 3-amino-ß-lactams to inhibit penicillin-binding proteins (PBPs) of various (resistant) bacteria was assessed, revealing the potential of α-benzylidenecarboxylates as interesting leads in the pursuit of novel PBP inhibitors. No deactivation by representative enzymes belonging to the four ß-lactamase classes was observed, while weak inhibition of class C ß-lactamase P99 was demonstrated.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Enterococcus faecium/drug effects , Escherichia coli/drug effects , Penicillin-Binding Proteins/antagonists & inhibitors , beta-Lactams/chemistry , beta-Lactams/pharmacology , Amination , Anti-Bacterial Agents/chemical synthesis , Bacterial Infections/drug therapy , Computer Simulation , Computer-Aided Design , Drug Design , Drug Resistance, Bacterial/drug effects , Enterococcus faecium/metabolism , Escherichia coli/metabolism , Escherichia coli Infections/drug therapy , Gram-Positive Bacterial Infections/drug therapy , Humans , Molecular Docking Simulation , Penicillin-Binding Proteins/metabolism , beta-Lactams/chemical synthesis
8.
Microb Drug Resist ; 23(1): 44-50, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27991847

ABSTRACT

The aim of this study is to characterize the factors related to peptidoglycan metabolism in isogenic hVISA/VISA ST100 strains. Recently, we reported the increase in IS256 transposition in invasive hVISA ST100 clinical strains isolated from the same patient (D1 and D2) before and after vancomycin treatment and two laboratory VISA mutants (D23C9 and D2P11) selected from D2 in independent experiments. High performance liquid chromatography-mass spectrometry (HPLC-MS) analysis of peptidoglycan muropeptides showed increased proportion of monomeric muropeptides and a concomitant decrease in the proportion of tetrameric muropeptide in D2 and derived mutants when compared to the original strain D1. In addition, strain D2 and its derived mutants showed an increase in cell wall thickness with increased pbp2 gene expression. The VISA phenotype was not stable in D2P11 and showed a reduced autolysis profile. On the other hand, the mutant D23C9 differentiates from D2 and D2P11 in the autolysis profile, and pbp4 transcription profile. D2-derived mutants exhibited differences in the susceptibility to other antimicrobials. Our results highlight the possibility of selection of different VISA phenotypes from a single hVISA-ST100 genetic background.


Subject(s)
Anti-Bacterial Agents/pharmacology , Phenotype , Selection, Genetic , Staphylococcus aureus/genetics , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Wall/chemistry , Cell Wall/drug effects , Cell Wall/metabolism , DNA Transposable Elements/drug effects , Gene Expression , Humans , Microbial Sensitivity Tests , Mutation , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Peptidoglycan/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/isolation & purification
9.
BMC Microbiol ; 16(1): 239, 2016 Oct 12.
Article in English | MEDLINE | ID: mdl-27729019

ABSTRACT

BACKGROUND: Proteins from the LytR-CpsA-Psr family are found in almost all Gram-positive bacteria. Although LCP proteins have been studied in other pathogens, their functions in enterococci remain uncharacterized. The Psr protein from Enterococcus hirae, here renamed LcpA, previously associated with the regulation of the expression of the low-affinity PBP5 and ß-lactam resistance, has been characterized. RESULTS: LcpA protein of E. hirae ATCC 9790 has been produced and purified with and without its transmembrane helix. LcpA appears, through different methods, to be localized in the membrane, in agreement with in silico predictions. The interaction of LcpA with E. hirae cell wall indicates that LcpA binds enterococcal peptidoglycan, regardless of the presence of secondary cell wall polymers. Immunolocalization experiments showed that LcpA and PBP5 are localized at the division site of E. hirae. CONCLUSIONS: LcpA belongs to the LytR-CpsA-Psr family. Its topology, localization and binding to peptidoglycan support, together with previous observations on defective mutants, that LcpA plays a role related to the cell wall metabolism, probably acting as a phosphotransferase catalyzing the attachment of cell wall polymers to the peptidoglycan.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Enterococcus hirae/metabolism , Peptidoglycan/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Cell Membrane/metabolism , Cell Wall/metabolism , Cloning, Molecular , DNA, Bacterial , Enterococcus hirae/cytology , Enterococcus hirae/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Genes, Bacterial , Phosphotransferases/metabolism , Protein Interaction Maps , Recombinant Proteins , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , beta-Lactam Resistance
10.
J Med Chem ; 58(24): 9712-21, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26588190

ABSTRACT

Penicillin-binding proteins represent well-established, validated, and still very promising targets for the design and development of new antibacterial agents. The transglycosylase domain of penicillin-binding proteins is especially important, as it catalyzes polymerization of glycan chains, using the peptidoglycan precursor lipid II as a substrate. On the basis of the previous discovery of a noncovalent small-molecule inhibitor of transglycosylase activity, we systematically explored the structure-activity relationships of these tryptamine-based inhibitors. The main aim was to reduce the nonspecific cytotoxic properties of the initial hit compound and concurrently to retain the mode of its inhibition. A focused library of tryptamine-based compounds was synthesized, characterized, and evaluated biochemically. The results presented here show the successful reduction of the nonspecific cytotoxicity, and the retention of the inhibition of transglycosylase enzymatic activity, as well as the ability of these compounds to bind to lipid II and to have antibacterial actions.


Subject(s)
Anti-Bacterial Agents/chemistry , Escherichia coli Proteins/antagonists & inhibitors , Penicillin-Binding Proteins/antagonists & inhibitors , Peptidoglycan Glycosyltransferase/antagonists & inhibitors , Serine-Type D-Ala-D-Ala Carboxypeptidase/antagonists & inhibitors , Tryptamines/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Enterococcus faecium/drug effects , Escherichia coli/drug effects , HEK293 Cells , Humans , Methicillin Resistance , Microbial Sensitivity Tests , Protein Binding , Staphylococcus aureus/drug effects , Structure-Activity Relationship , Tryptamines/pharmacology , Tryptamines/toxicity , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
11.
PLoS One ; 10(4): e0125377, 2015.
Article in English | MEDLINE | ID: mdl-25927608

ABSTRACT

Release of conserved cytoplasmic proteins is widely spread among Gram-positive and Gram-negative bacteria. Because these proteins display additional functions when located at the bacterial surface, they have been qualified as moonlighting proteins. The GAPDH is a glycolytic enzyme which plays an important role in the virulence processes of pathogenic microorganisms like bacterial invasion and host immune system modulation. However, GAPDH, like other moonlighting proteins, cannot be secreted through active secretion systems since they do not contain an N-terminal predicted signal peptide. In this work, we investigated the mechanism of GAPDH export and surface retention in Streptococcus pneumoniae, a major human pathogen. We addressed the role of the major autolysin LytA in the delivery process of GAPDH to the cell surface. Pneumococcal lysis is abolished in the ΔlytA mutant strain or when 1% choline chloride is added in the culture media. We showed that these conditions induce a marked reduction in the amount of surface-associated GAPDH. These data suggest that the presence of GAPDH at the surface of pneumococcal cells depends on the LytA-mediated lysis of a fraction of the cell population. Moreover, we demonstrated that pneumococcal GAPDH binds to the bacterial cell wall independently of the presence of the teichoic acids component, supporting peptidoglycan as a ligand to surface GAPDH. Finally, we showed that peptidoglycan-associated GAPDH recruits C1q from human serum but does not activate the complement pathway.


Subject(s)
Bacterial Proteins/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Peptidoglycan/metabolism , Pneumococcal Infections/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/metabolism , Bacterial Proteins/genetics , Bacteriolysis/genetics , Cell Membrane/metabolism , Cell Wall/metabolism , Complement C1q/immunology , Complement C1q/metabolism , Humans , Pneumococcal Infections/immunology , Protein Binding/immunology , Protein Transport , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/immunology
12.
Org Biomol Chem ; 13(26): 7193-222, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26008868

ABSTRACT

The straightforward synthesis of 5'-methylene-[1,4]-triazole-substituted aminoribosyl uridines is described. Two families of compounds were synthesized from a unique epoxide which was regioselectively opened by acetylide ions (for compounds II) or azide ions (for compounds III). Sequential diastereoselective glycosylation with a ribosyl fluoride derivative, Cu(i)-catalyzed azide-alkyne cycloaddition (CuAAC) with various complementary azide and alkyne partners afforded the targeted compounds after final deprotection. The biological activity of the 16 resulting compounds together with that of 14 previously reported compounds I, lacking the 5' methylene group, was evaluated on the MraY transferase activity. Out of the 30 tested compounds, 18 compounds revealed MraY inhibition with IC50 ranging from 15 to 150 µM. A molecular modeling study was performed to rationalize the observed structure-activity relationships (SAR), which allowed us to correlate the activity of the most potent compounds with an interaction involving Leu191 of MraYAA. The antibacterial activity was also evaluated and seven compounds exhibited a good activity against Gram-positive bacterial pathogens with MIC ranging from 8 to 32 µg mL(-1), including the methicillin resistant Staphylococcus aureus (MRSA).


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Models, Molecular , Transferases/antagonists & inhibitors , Triazoles/chemistry , Uridine/chemistry , Uridine/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Bacillus subtilis/enzymology , Bacterial Proteins/chemistry , Catalytic Domain , Chemistry Techniques, Synthetic , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests , Transferases/chemistry , Transferases (Other Substituted Phosphate Groups) , Uridine/chemical synthesis
13.
J Immunol ; 193(11): 5699-708, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25344472

ABSTRACT

Human L-ficolin is a soluble protein of the innate immune system able to sense pathogens through its fibrinogen (FBG) recognition domains and to trigger activation of the lectin complement pathway through associated serine proteases. L-Ficolin has been previously shown to recognize pneumococcal clinical isolates, but its ligands and especially its molecular specificity remain to be identified. Using solid-phase binding assays, serum and recombinant L-ficolins were shown to interact with serotype 2 pneumococcal strain D39 and its unencapsulated R6 derivative. Incubation of both strains with serum triggered complement activation, as measured by C4b and C3b deposition, which was decreased by using ficolin-depleted serum. Recombinant L-ficolin and its FBG-like recognition domain bound to isolated pneumococcal cell wall extracts, whereas binding to cell walls depleted of teichoic acid (TA) was decreased. Both proteins were also shown to interact with two synthetic TA compounds, each comprising part structures of the complete lipoteichoic acid molecule with two PCho residues. Competition studies and direct interaction measurements by surface plasmon resonance identified PCho as a novel L-ficolin ligand. Structural analysis of complexes of the FBG domain of L-ficolin and PCho revealed that the phosphate moiety interacts with amino acids previously shown to define an acetyl binding site. Consequently, binding of L-ficolin to immobilized acetylated BSA was inhibited by PCho and synthetic TA. Binding of serum L-ficolin to immobilized synthetic TA and PCho-conjugated BSA triggered activation of the lectin complement pathway, thus further supporting the hypothesis of L-ficolin involvement in host antipneumococcal defense.


Subject(s)
Lectins/metabolism , Pneumococcal Infections/immunology , Streptococcus pneumoniae/metabolism , Teichoic Acids/metabolism , Acetylation , Cell Wall/metabolism , Complement Activation , Complement C3b/metabolism , Complement C4b/metabolism , Fibrinogen/genetics , Host-Pathogen Interactions , Humans , Immunity, Innate , Lectins/genetics , Phosphorylcholine/chemistry , Protein Binding , Protein Structure, Tertiary/genetics , Streptococcus pneumoniae/immunology , Surface Plasmon Resonance , Teichoic Acids/chemistry , Ficolins
14.
Antimicrob Agents Chemother ; 57(12): 6358-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24060866

ABSTRACT

The opportunistic human pathogen Enterococcus faecium overproduces the low-affinity PBP5. In clinical strains, mutations in PBP5 further reduce its acylation rate by ß-lactams. Previous studies have reported that ceftaroline had poor inhibitory activity against ß-lactam-resistant E. faecium strains. In this study, we show that ceftaroline exhibits killing activity against our laboratory-derived ampicillin-resistant E. faecium mutant that overproduces a wild-type PBP5 and that ceftaroline inactivates PBP5 much faster than benzylpenicillin and faster than ceftobiprole.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Enterococcus faecium/drug effects , Enterococcus faecium/genetics , Ampicillin Resistance/genetics , Bacterial Proteins/genetics , beta-Lactam Resistance/genetics , Ceftaroline
15.
PLoS One ; 7(5): e36400, 2012.
Article in English | MEDLINE | ID: mdl-22623956

ABSTRACT

In Bacillus licheniformis 749/I, BlaP ß-lactamase is induced by the presence of a ß-lactam antibiotic outside the cell. The first step in the induction mechanism is the detection of the antibiotic by the membrane-bound penicillin receptor BlaR1 that is composed of two functional domains: a carboxy-terminal domain exposed outside the cell, which acts as a penicillin sensor, and an amino-terminal domain anchored to the cytoplasmic membrane, which works as a transducer-transmitter. The acylation of BlaR1 sensor domain by the antibiotic generates an intramolecular signal that leads to the activation of the L3 cytoplasmic loop of the transmitter by a single-point cleavage. The exact mechanism of L3 activation and the nature of the secondary cytoplasmic signal launched by the activated transmitter remain unknown. However, these two events seem to be linked to the presence of a HEXXH zinc binding motif of neutral zinc metallopeptidases. By different experimental approaches, we demonstrated that the L3 loop binds zinc ion, belongs to Gluzincin metallopeptidase superfamily and is activated by self-proteolysis.


Subject(s)
Bacillus/enzymology , Metalloendopeptidases/metabolism , Signal Transduction/genetics , beta-Lactamases/metabolism , Amino Acid Sequence , Bacillus/genetics , Bacterial Proteins/genetics , Base Sequence , Blotting, Western , Metalloendopeptidases/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Proteolysis , Rosaniline Dyes , Sequence Alignment , Sequence Analysis, DNA , Zinc/metabolism
16.
Bioorg Med Chem ; 20(12): 3915-24, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22579615

ABSTRACT

In response to the widespread use of ß-lactam antibiotics bacteria have evolved drug resistance mechanisms that include the production of resistant Penicillin Binding Proteins (PBPs). Boronic acids are potent ß-lactamase inhibitors and have been shown to display some specificity for soluble transpeptidases and PBPs, but their potential as inhibitors of the latter enzymes is yet to be widely explored. Recently, a (2,6-dimethoxybenzamido)methylboronic acid was identified as being a potent inhibitor of Actinomadura sp. R39 transpeptidase (IC(50): 1.3 µM). In this work, we synthesized and studied the potential of a number of acylaminomethylboronic acids as inhibitors of PBPs from different classes. Several derivatives inhibited PBPs of classes A, B and C from penicillin sensitive strains. The (2-nitrobenzamido)methylboronic acid was identified as a good inhibitor of a class A PBP (PBP1b from Streptococcus pneumoniae, IC(50) = 26 µM), a class B PBP (PBP2xR6 from Streptococcus pneumoniae, IC(50) = 138 µM) and a class C PBP (R39 from Actinomadura sp., IC(50) = 0.6 µM). This work opens new avenues towards the development of molecules that inhibit PBPs, and eventually display bactericidal effects, on distinct bacterial species.


Subject(s)
Boronic Acids/chemical synthesis , Boronic Acids/pharmacology , Penicillin-Binding Proteins/antagonists & inhibitors , Penicillin-Binding Proteins/classification , Actinomycetales/chemistry , Boronic Acids/chemistry , Dose-Response Relationship, Drug , Molecular Structure , Streptococcus pneumoniae/chemistry , Structure-Activity Relationship
17.
PLoS Pathog ; 8(3): e1002571, 2012.
Article in English | MEDLINE | ID: mdl-22438804

ABSTRACT

To resist to ß-lactam antibiotics Eubacteria either constitutively synthesize a ß-lactamase or a low affinity penicillin-binding protein target, or induce its synthesis in response to the presence of antibiotic outside the cell. In Bacillus licheniformis and Staphylococcus aureus, a membrane-bound penicillin receptor (BlaR/MecR) detects the presence of ß-lactam and launches a cytoplasmic signal leading to the inactivation of BlaI/MecI repressor, and the synthesis of a ß-lactamase or a low affinity target. We identified a dipeptide, resulting from the peptidoglycan turnover and present in bacterial cytoplasm, which is able to directly bind to the BlaI/MecI repressor and to destabilize the BlaI/MecI-DNA complex. We propose a general model, in which the acylation of BlaR/MecR receptor and the cellular stress induced by the antibiotic, are both necessary to generate a cell wall-derived coactivator responsible for the expression of an inducible ß-lactam-resistance factor. The new model proposed confirms and emphasizes the role of peptidoglycan degradation fragments in bacterial cell regulation.


Subject(s)
Bacillus/genetics , Gene Expression Regulation, Bacterial/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Peptidoglycan/metabolism , beta-Lactam Resistance/genetics , Acylation , Bacillus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Wall/chemistry , Cell Wall/metabolism , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , Dipeptides/chemistry , Dipeptides/metabolism , Enzyme Induction/genetics , Gene Expression Regulation, Bacterial/drug effects , Humans , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/drug effects , Penicillins/metabolism , Penicillins/pharmacology , Peptidoglycan/chemistry , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , beta-Lactamases/biosynthesis , beta-Lactamases/genetics
18.
Acta Chim Slov ; 59(2): 280-388, 2012 Jun.
Article in English | MEDLINE | ID: mdl-24061241

ABSTRACT

Penicillin-binding proteins are a well established, validated and still a very promising target for the design and development of new antibacterial agents. Based on our previous discovery of several noncovalent small-molecule inhibitor hits for resistant PBPs we decided to additionally explore the chemical space around these compounds. In order to clarify their structure-activity relationships for PBP inhibition two new series of compounds were synthesized, characterized and evaluated biochemically: the derivatives of anthranilic acid and naphthalene-sulfonamide derivatives. The target compounds were tested for their inhibitory activities on three different transpeptidases: PBP2a from methicillin-resistant Staphylococcus aureus (MRSA) strains, PBP5fm from Enterococcus faecium strains, and PBP1b from Streptococcus pneumoniae strains. The most promising results for both of these series of compounds were obtained against the PBP2a enzyme with the IC50 values in the micromolar range. Although these results do not represent a significant breakthrough in the field of noncovalent PBP inhibitors, they do provide useful structure-activity relationship data, and thus a more solid basis for the design of potent and noncovalent inhibitors of resistant PBPs.

19.
ACS Chem Biol ; 6(9): 943-51, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21732689

ABSTRACT

ß-Lactam antibiotics have long been a treatment of choice for bacterial infections since they bind irreversibly to Penicillin-Binding Proteins (PBPs), enzymes that are vital for cell wall biosynthesis. Many pathogens express drug-insensitive PBPs rendering ß-lactams ineffective, revealing a need for new types of PBP inhibitors active against resistant strains. We have identified alkyl boronic acids that are active against pathogens including methicillin-resistant S. aureus (MRSA). The crystal structures of PBP1b complexed to 11 different alkyl boronates demonstrate that in vivo efficacy correlates with the mode of inhibitor side chain binding. Staphylococcal membrane analyses reveal that the most potent alkyl boronate targets PBP1, an autolysis system regulator, and PBP2a, a low ß-lactam affinity enzyme. This work demonstrates the potential of boronate-based PBP inhibitors for circumventing ß-lactam resistance and opens avenues for the development of novel antibiotics that target Gram-positive pathogens.


Subject(s)
Boronic Acids/pharmacology , Cell Wall/drug effects , Drug Design , Penicillin-Binding Proteins/antagonists & inhibitors , Penicillin-Binding Proteins/chemistry , Staphylococcus aureus/drug effects , beta-Lactam Resistance/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Boronic Acids/chemistry , Boronic Acids/metabolism , Cell Wall/metabolism , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Penicillin-Binding Proteins/metabolism , Staphylococcus aureus/cytology , Staphylococcus aureus/enzymology , Stereoisomerism , Structure-Activity Relationship , beta-Lactams/chemistry , beta-Lactams/pharmacology
20.
PLoS One ; 6(5): e19418, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21573060

ABSTRACT

BACKGROUND: Penicillin-binding proteins (PBPs) are well known and validated targets for antibacterial therapy. The most important clinically used inhibitors of PBPs ß-lactams inhibit transpeptidase activity of PBPs by forming a covalent penicilloyl-enzyme complex that blocks the normal transpeptidation reaction; this finally results in bacterial death. In some resistant bacteria the resistance is acquired by active-site distortion of PBPs, which lowers their acylation efficiency for ß-lactams. To address this problem we focused our attention to discovery of novel noncovalent inhibitors of PBPs. METHODOLOGY/PRINCIPAL FINDINGS: Our in-house bank of compounds was screened for inhibition of three PBPs from resistant bacteria: PBP2a from Methicillin-resistant Staphylococcus aureus (MRSA), PBP2x from Streptococcus pneumoniae strain 5204, and PBP5fm from Enterococcus faecium strain D63r. Initial hit inhibitor obtained by screening was then used as a starting point for computational similarity searching for structurally related compounds and several new noncovalent inhibitors were discovered. Two compounds had promising inhibitory activities of both PBP2a and PBP2x 5204, and good in-vitro antibacterial activities against a panel of Gram-positive bacterial strains. CONCLUSIONS: We found new noncovalent inhibitors of PBPs which represent important starting points for development of more potent inhibitors of PBPs that can target penicillin-resistant bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Penicillin-Binding Proteins/antagonists & inhibitors , Penicillins/pharmacology , Anti-Bacterial Agents/chemistry , Enterococcus faecium/drug effects , Enterococcus faecium/metabolism , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/metabolism , Microbial Sensitivity Tests , Molecular Structure , Peptide Synthases/antagonists & inhibitors , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...