Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
1.
Nature ; 632(8023): 39-49, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085542

ABSTRACT

In this Review, we explore natural product antibiotics that do more than simply inhibit an active site of an essential enzyme. We review these compounds to provide inspiration for the design of much-needed new antibacterial agents, and examine the complex mechanisms that have evolved to effectively target bacteria, including covalent binders, inhibitors of resistance, compounds that utilize self-promoted entry, those that evade resistance, prodrugs, target corrupters, inhibitors of 'undruggable' targets, compounds that form supramolecular complexes, and selective membrane-acting agents. These are exemplified by ß-lactams that bind covalently to inhibit transpeptidases and ß-lactamases, siderophore chimeras that hijack import mechanisms to smuggle antibiotics into the cell, compounds that are activated by bacterial enzymes to produce reactive molecules, and antibiotics such as aminoglycosides that corrupt, rather than merely inhibit, their targets. Some of these mechanisms are highly sophisticated, such as the preformed ß-strands of darobactins that target the undruggable ß-barrel chaperone BamA, or teixobactin, which binds to a precursor of peptidoglycan and then forms a supramolecular structure that damages the membrane, impeding the emergence of resistance. Many of the compounds exhibit more than one notable feature, such as resistance evasion and target corruption. Understanding the surprising complexity of the best antimicrobial compounds provides a roadmap for developing novel compounds to address the antimicrobial resistance crisis by mining for new natural products and inspiring us to design similarly sophisticated antibiotics.


Subject(s)
Anti-Bacterial Agents , Bacteria , Biological Products , Animals , Humans , Aminoglycosides/pharmacology , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacteria/drug effects , Bacteria/enzymology , Bacteria/metabolism , beta Lactam Antibiotics/chemistry , beta Lactam Antibiotics/pharmacology , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/metabolism , Drug Design , Drug Resistance, Bacterial/drug effects , Peptidyl Transferases/antagonists & inhibitors , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/metabolism , Siderophores/metabolism , Siderophores/chemistry , Siderophores/pharmacology
2.
Nat Struct Mol Biol ; 29(2): 152-161, 2022 02.
Article in English | MEDLINE | ID: mdl-35165455

ABSTRACT

Ribosome-targeting antibiotics serve as powerful antimicrobials and as tools for studying the ribosome, the catalytic peptidyl transferase center (PTC) of which is targeted by many drugs. The classic PTC-acting antibiotic chloramphenicol (CHL) and the newest clinically significant linezolid (LZD) were considered indiscriminate inhibitors of protein synthesis that cause ribosome stalling at every codon of every gene being translated. However, recent discoveries have shown that CHL and LZD preferentially arrest translation when the ribosome needs to polymerize particular amino acid sequences. The molecular mechanisms that underlie the context-specific action of ribosome inhibitors are unknown. Here we present high-resolution structures of ribosomal complexes, with or without CHL, carrying specific nascent peptides that support or negate the drug action. Our data suggest that the penultimate residue of the nascent peptide directly modulates antibiotic affinity to the ribosome by either establishing specific interactions with the drug or by obstructing its proper placement in the binding site.


Subject(s)
Chloramphenicol/chemistry , Chloramphenicol/pharmacology , Peptidyl Transferases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Models, Molecular , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Static Electricity , Thermus thermophilus/drug effects , Thermus thermophilus/metabolism
3.
Cell Chem Biol ; 28(9): 1321-1332.e5, 2021 09 16.
Article in English | MEDLINE | ID: mdl-33826941

ABSTRACT

Effective treatment of tuberculosis is frequently hindered by the emerging antimicrobial resistance of Mycobacterium tuberculosis. The present study evaluates monocyclic ß-lactam compounds targeting the mycobacterial cell wall remodeling. Novel N-thio-ß-lactams were designed, synthesized, and characterized on the L,D-transpeptidase-2, a validated target in M. tuberculosis. The candidates were evaluated in biochemical assays identifying five compounds presenting target-specific kinetic constants equal or superior to meropenem, a carbapenem currently considered for tuberculosis therapy. Mass spectrometry in line with the crystal structures of five target-ligand complexes revealed that the N-thio-ß-lactams act via an unconventional mode of adduct formation, transferring the thio-residues from the lactam ring to the active-site cysteine of LdtMt2. The resulting stable adducts lead to a long-term inactivation of the target protein. Finally, the candidates were evaluated in vitro against a drug-susceptible and multidrug-resistant clinical isolates of M. tuberculosis, confirming the antimycobacterial effect of these novel compounds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Mycobacterium tuberculosis/drug effects , Peptidyl Transferases/antagonists & inhibitors , beta-Lactams/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/metabolism , beta-Lactams/chemical synthesis , beta-Lactams/chemistry
4.
Chem Biol Drug Des ; 96(2): 836-849, 2020 08.
Article in English | MEDLINE | ID: mdl-32271987

ABSTRACT

The issue of antibiotic resistance is becoming progressively serious these days, and the feasible solution to address it is to develop and discover novel antibiotics. The diterpene natural pleuromutilin is a prominent candidate for its special mode of action by inhibiting protein synthesis. In this study, a series of novel pleuromutilin derivatives with chalcone moiety was designed and synthesized, and their antibacterial activities were assessed in vitro. As suggested from the results, most of compounds exhibited potent activities against two methicillin-resistant Staphylococcus aureus (MRSA) ATCC 33591 and 43300. The further modification of the chalcone structure, aza-cyclic derivatives were afforded and then assessed, and potent activities against the tested strains were reported. The preliminary docking studies were conducted to explore the interactions between target molecules and binding site.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Chalcones/chemical synthesis , Diterpenes/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Peptidyl Transferases/antagonists & inhibitors , Polycyclic Compounds/chemical synthesis , Anti-Bacterial Agents/pharmacology , Binding Sites , Chalcones/pharmacology , Diterpenes/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Molecular Docking Simulation , Polycyclic Compounds/pharmacology , Protein Binding , Structure-Activity Relationship , Pleuromutilins
5.
Molecules ; 25(6)2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32245177

ABSTRACT

Pyrazolo[1,5-a]pyrimidines 5a-c, 9a-c and 13a-i were synthesized for evaluation of their in vitro antimicrobial properties against some microorganisms and their immunomodulatory activity. The biological activities of pyrazolo[1,5-a]pyrimidines showed that the pyrazolo[1,5-a]pyrimidines (5c, 9a, 9c, 13a, 13c, 13d, 13e and 13h) displayed promising antimicrobial and immunomodulatory activities. Studying the in silico predicted physicochemical, pharmacokinetic, ADMET and drug-likeness properties for the pyrazolo[1,5-a]pyrimidines 5a-c, 9a-c and 13a-i confirmed that most of the compounds (i) were within the range set by Lipinski's rule of five, (ii) show higher gastrointestinal absorption and inhibition of some CYP isoforms, and (iii) have a carcinogenicity test that was predicted as negative and hERG test that presented medium risk. Moreover, the molecular docking study demonstrated that the compounds 5c, 9a, 9c, 13a, 13c, 13d, 13e and 13h are potent inhibitors of 14-alpha demethylase, transpeptidase and alkaline phosphatase enzymes. This study could be valuable in the discovery of a new series of drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Fungi/drug effects , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , 14-alpha Demethylase Inhibitors/pharmacology , Alkaline Phosphatase/antagonists & inhibitors , Aspergillus/drug effects , Caco-2 Cells , Candida albicans/drug effects , Carcinogenicity Tests/adverse effects , Computer Simulation , Drug Design , Enterococcus faecalis/drug effects , Escherichia coli/drug effects , Fusarium/drug effects , Humans , Molecular Docking Simulation , Molecular Structure , Peptidyl Transferases/antagonists & inhibitors , Pseudomonas aeruginosa/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrazoles/toxicity , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/toxicity , Salmonella typhi/drug effects , Staphylococcus aureus/drug effects , Structure-Activity Relationship
6.
Eur J Med Chem ; 194: 112262, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32248005

ABSTRACT

The enzymes involved in bacterial cell wall synthesis are established antibiotic targets, and continue to be a central focus for antibiotic development. Bacterial penicillin-binding proteins (and, in some bacteria, l,d-transpeptidases) form essential peptide cross-links in the cell wall. Although the ß-lactam class of antibiotics target these enzymes, bacterial resistance threatens their clinical use, and there is an urgent unmet need for new antibiotics. However, the search for new antibiotics targeting the bacterial cell wall is hindered by a number of obstacles associated with screening the enzymes involved in peptidoglycan synthesis. This review describes recent approaches for measuring the activity and inhibition of penicillin-binding proteins and l,d-transpeptidases, highlighting strategies that are poised to serve as valuable tools for high-throughput screening of transpeptidase inhibitors, supporting the development of new antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cell Wall/drug effects , Drug Discovery , Peptidyl Transferases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacteria/metabolism , Cell Wall/metabolism , Microbial Sensitivity Tests , Molecular Structure , Penicillin-Binding Proteins/antagonists & inhibitors , Penicillin-Binding Proteins/metabolism , Peptidyl Transferases/metabolism
7.
Biochem Biophys Res Commun ; 523(1): 6-9, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31822344

ABSTRACT

Tuberculosis has attracted increased attention worldwide due to its high morality and its resistance to treatment with traditional antibacterial drugs. The l,d-transpeptidase LdtMt2 confers resistance to traditional ß-lactams and is considered a target for anti-Tuberculosis treatment. Carbapenems are proposed to inhibit Mycobacterium tuberculosis by repressing the activity of LdtMt2. The interaction mechanisms between LdtMt2 and carbapenems have been revealed by LdtMt2-carbapenem adduct structures along with various biochemical assays. Interestingly, the lack of the 1-ß-methyl group in imipenem may be related to its high binding ability to LdtMt2. However, there is limited evidence on the interaction mode of LdtMt2 and panipenem, another carbapenem lacking the 1-ß-methyl group. Herein, we identified the biochemical features of panipenem binding to LdtMt2. We further suggest that the presence of the 1-ß-methyl group in carbapenems is indeed related to the ligand affinity of LdtMt2 and that the presence of the Y308 and Y318 residues in LdtMt2 stabilized the conformation of the LdtMt2-carbepenem adduct. Our research provides a structural basis for the development of novel carbapenems against L,D-transpeptidases.


Subject(s)
Enzyme Inhibitors/pharmacology , Peptidyl Transferases/antagonists & inhibitors , Thienamycins/pharmacology , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/chemistry , Peptidyl Transferases/metabolism , Thienamycins/chemistry
8.
Chembiochem ; 21(3): 368-372, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31322798

ABSTRACT

Mycobacterium tuberculosis l,d-transpeptidases (Ldts), which are involved in cell-wall biosynthesis, have emerged as promising targets for the treatment of tuberculosis. However, an efficient method for testing inhibition of these enzymes is not currently available. We present a fluorescence-based assay for LdtMt2 , which is suitable for high-throughput screening. Two fluorogenic probes were identified that release a fluorophore upon reaction with LdtMt2 , thus making it possible to assess the availability of the catalytic site in the presence of inhibitors. The assay was applied to a panel of ß-lactam antibiotics and related inhibitors; the results validate observations that the (carba)penem subclass of ß-lactams are more potent Ldt inhibitors than other ß-lactam classes, though unexpected variations in potency were observed. The method will enable systematic structure-activity relationship studies on Ldts, thereby facilitating the identification of new antibiotics active against M. tuberculosis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacology , Mycobacterium tuberculosis/drug effects , Peptidyl Transferases/antagonists & inhibitors , beta-Lactams/pharmacology , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/chemistry , Fluorescence , Fluorescent Dyes/chemistry , High-Throughput Screening Assays , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/metabolism , Peptidyl Transferases/metabolism , beta-Lactams/chemistry
9.
J Mol Model ; 25(11): 328, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31656981

ABSTRACT

Virtual screening is a useful in silico approach to identify potential leads against various targets. It is known that carbapenems (doripenem and faropenem) do not show any reasonable inhibitory activities against L,D-transpeptidase 5 (LdtMt5) and also an adduct of meropenem exhibited slow acylation. Since these drugs are active against L,D-transpeptidase 2 (LdtMt2), understanding the differences between these two enzymes is essential. In this study, a ligand-based virtual screening of 12,766 compounds followed by molecular dynamics (MD) simulations was applied to identify potential leads against LdtMt5. To further validate the obtained virtual screening ranking for LdtMt5, we screened the same libraries of compounds against LdtMt2 which had more experimetal and calculated binding energies reported. The observed consistency between the binding affinities of LdtMt2 validates the obtained virtual screening binding scores for LdtMt5. We subjected 37 compounds with docking scores ranging from - 7.2 to - 9.9 kcal mol-1 obtained from virtual screening for further MD analysis. A set of compounds (n = 12) from four antibiotic classes with ≤ - 30 kcal mol-1 molecular mechanics/generalized born surface area (MM-GBSA) binding free energies (ΔGbind) was characterized. A final set of that, all ß-lactams (n = 4), was considered. The outcome of this study provides insight into the design of potential novel leads for LdtMt5. Graphical abstract.


Subject(s)
Antitubercular Agents/pharmacology , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Anti-Bacterial Agents/pharmacology , Ligands , Meropenem/pharmacology , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Peptidyl Transferases/antagonists & inhibitors , Protein Binding/drug effects
10.
Chem Commun (Camb) ; 55(69): 10214-10217, 2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31380528

ABSTRACT

The l,d-transpeptidases (Ldts) are promising antibiotic targets for treating tuberculosis. We report screening of cysteine-reactive inhibitors against LdtMt2 from Mycobacterium tuberculosis. Structural studies on LdtMt2 with potent inhibitor ebselen reveal opening of the benzisoselenazolone ring by a nucleophilic cysteine, forming a complex involving extensive hydrophobic interactions with a substrate-binding loop.


Subject(s)
Azoles/chemistry , Azoles/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Organoselenium Compounds/chemistry , Organoselenium Compounds/pharmacology , Peptidyl Transferases/antagonists & inhibitors , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Benzene Derivatives/chemistry , Benzene Derivatives/pharmacology , Cysteine/metabolism , Humans , Isoindoles , Molecular Docking Simulation , Mycobacterium tuberculosis/drug effects , Peptidyl Transferases/metabolism , Tuberculosis/drug therapy , Tuberculosis/microbiology
11.
ACS Infect Dis ; 5(7): 1169-1176, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31056908

ABSTRACT

In most bacteria, ß-lactam antibiotics inhibit the last cross-linking step of peptidoglycan synthesis by acylation of the active-site Ser of d,d-transpeptidases belonging to the penicillin-binding protein (PBP) family. In mycobacteria, cross-linking is mainly ensured by l,d-transpeptidases (LDTs), which are promising targets for the development of ß-lactam-based therapies for multidrug-resistant tuberculosis. For this purpose, fluorescence spectroscopy is used to investigate the efficacy of LDT inactivation by ß-lactams but the basis for fluorescence quenching during enzyme acylation remains unknown. In contrast to what has been reported for PBPs, we show here using a model l,d-transpeptidase (Ldtfm) that fluorescence quenching of Trp residues does not depend upon direct hydrophobic interaction between Trp residues and ß-lactams. Rather, Trp fluorescence was quenched by the drug covalently bound to the active-site Cys residue of Ldtfm. Fluorescence quenching was not quantitatively determined by the size of the drug and was not specific of the thioester link connecting the ß-lactam carbonyl to the catalytic Cys as quenching was also observed for acylation of the active-site Ser of ß-lactamase BlaC from M. tuberculosis. Fluorescence quenching was extensive for reaction intermediates containing an amine anion and for acylenzymes containing an imine stabilized by mesomeric effect, but not for acylenzymes containing a protonated ß-lactam nitrogen. Together, these results indicate that the extent of fluorescence quenching is determined by the status of the ß-lactam nitrogen. Thus, fluorescence kinetics can provide information not only on the efficacy of enzyme inactivation but also on the structure of the covalent adducts responsible for enzyme inactivation.


Subject(s)
Peptidyl Transferases/chemistry , Tryptophan/chemistry , beta-Lactams/pharmacology , Acylation , Catalytic Domain , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/antagonists & inhibitors , Peptidyl Transferases/metabolism , Serine/chemistry , Spectrometry, Fluorescence , beta-Lactamases/metabolism , beta-Lactams/chemistry
12.
Sci Rep ; 9(1): 5634, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948752

ABSTRACT

Oxazolidinones are synthetic antibiotics used for treatment of infections caused by Gram-positive bacteria. They target the bacterial protein synthesis machinery by binding to the peptidyl transferase centre (PTC) of the ribosome and interfering with the peptidyl transferase reaction. Cadazolid is the first member of quinoxolidinone antibiotics, which are characterized by combining the pharmacophores of oxazolidinones and fluoroquinolones, and it is evaluated for treatment of Clostridium difficile gastrointestinal infections that frequently occur in hospitalized patients. In vitro protein synthesis inhibition by cadazolid was shown in Escherichia coli and Staphylococcus aureus, including an isolate resistant against linezolid, the prototypical oxazolidinone antibiotic. To better understand the mechanism of inhibition, we determined a 3.0 Å cryo-electron microscopy structure of cadazolid bound to the E. coli ribosome in complex with mRNA and initiator tRNA. Here we show that cadazolid binds with its oxazolidinone moiety in a binding pocket in close vicinity of the PTC as observed previously for linezolid, and that it extends its unique fluoroquinolone moiety towards the A-site of the PTC. In this position, the drug inhibits protein synthesis by interfering with the binding of tRNA to the A-site, suggesting that its chemical features also can enable the inhibition of linezolid-resistant strains.


Subject(s)
Oxazolidinones/metabolism , Oxazolidinones/pharmacology , Protein Synthesis Inhibitors/pharmacology , Acetamides/pharmacology , Anti-Bacterial Agents/pharmacology , Clostridium Infections/drug therapy , Cryoelectron Microscopy/methods , Escherichia coli/metabolism , Fluoroquinolones/pharmacology , Humans , Microbial Sensitivity Tests , Peptidyl Transferases/antagonists & inhibitors , RNA, Transfer, Met/metabolism , Ribosomes/metabolism , Staphylococcus aureus/metabolism
13.
ACS Infect Dis ; 5(6): 863-872, 2019 06 14.
Article in English | MEDLINE | ID: mdl-30848883

ABSTRACT

The high-molecular mass penicillin-binding proteins (PBPs) are the essential targets of the ß-lactam classes of antibacterial drugs. In the Gram-negative pathogen Escherichia coli, these include PBP1a, PBP1b, PBP2, and PBP3. Techniques that enable facile measurement of the potency of inhibition of these targets are valuable for understanding structure-activity relationships in programs aimed at discovering new antibiotics to combat drug-resistant infections. Continuous fluorescence anisotropy-based assays for inhibition of soluble constructs of PBP1a, PBP2, and PBP3 from the serious Gram-negative bacterial pathogens Pseudomonas aeruginosa and Acinetobacter baumannii and PBP3 from E. coli using the fluorescent phenoxypenicillin analogue BOCILLIN FL have been described previously, but this technique was not useful for PBP2 from E. coli due to a lack of change in fluorescence anisotropy or intensity upon reaction. Here, we report that a fluorescent analogue of ampicillin, 5-carboxytetramethylrhodamine-ampicillin (5-TAMRA-ampicillin), was useful as the indicator in a continuous fluorescence anisotropy-based kinetic assay for inhibition of a soluble construct of PBP2 from E. coli. The assay enables measurement of the bimolecular rate constant for inhibition kinact /Ki. This measurement was made for representative drugs from four classes of ß-lactams and for the diazabicyclooctenone ETX2514. 5-TAMRA-ampicillin was also useful in a fluorescence anisotropy-based assay for P. aeruginosa PBP2 and in fluorescence intensity-based assays with PBP1a and PBP3 from P. aeruginosa and A. baumannii and PBP3 from E. coli.


Subject(s)
Ampicillin/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/drug effects , Penicillin-Binding Proteins/antagonists & inhibitors , Peptidyl Transferases/antagonists & inhibitors , Rhodamines/pharmacology , Acinetobacter baumannii/enzymology , Ampicillin/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Escherichia coli/enzymology , Fluorescence Polarization , Kinetics , Microbial Sensitivity Tests , Pseudomonas aeruginosa/enzymology , beta-Lactams/pharmacology
14.
RNA ; 25(5): 600-606, 2019 05.
Article in English | MEDLINE | ID: mdl-30733327

ABSTRACT

The 70S ribosome is a major target for antibacterial drugs. Two of the classical antibiotics, chloramphenicol (CHL) and erythromycin (ERY), competitively bind to adjacent but separate sites on the bacterial ribosome: the catalytic peptidyl transferase center (PTC) and the nascent polypeptide exit tunnel (NPET), respectively. The previously reported competitive binding of CHL and ERY might be due either to a direct collision of the two drugs on the ribosome or due to a drug-induced allosteric effect. Because of the resolution limitations, the available structures of these antibiotics in complex with bacterial ribosomes do not allow us to discriminate between these two possible mechanisms. In this work, we have obtained two crystal structures of CHL and ERY in complex with the Thermus thermophilus 70S ribosome at a higher resolution (2.65 and 2.89 Å, respectively) allowing unambiguous placement of the drugs in the electron density maps. Our structures provide evidence of the direct collision of CHL and ERY on the ribosome, which rationalizes the observed competition between the two drugs.


Subject(s)
Anti-Bacterial Agents/chemistry , Chloramphenicol/chemistry , Erythromycin/chemistry , Ribosome Subunits/drug effects , Thermus thermophilus/drug effects , Anti-Bacterial Agents/pharmacology , Binding Sites , Binding, Competitive , Chloramphenicol/pharmacology , Crystallography, X-Ray , Erythromycin/pharmacology , Escherichia coli/chemistry , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Models, Molecular , Peptidyl Transferases/antagonists & inhibitors , Peptidyl Transferases/chemistry , Peptidyl Transferases/genetics , Peptidyl Transferases/metabolism , Protein Binding , Protein Biosynthesis , Protein Conformation , Ribosome Subunits/genetics , Ribosome Subunits/metabolism , Ribosome Subunits/ultrastructure , Thermus thermophilus/chemistry , Thermus thermophilus/genetics , Thermus thermophilus/metabolism
15.
J Mol Graph Model ; 87: 204-210, 2019 03.
Article in English | MEDLINE | ID: mdl-30554066

ABSTRACT

Tuberculosis (TB) is one of the world's deadliest diseases resulting from infection by the bacterium, Mycobacterium tuberculosis (M.tb). The L,D-transpeptidase enzymes catalyze the synthesis of 3 → 3 transpeptide linkages which are predominant in the peptidoglycan of the M.tb cell wall. Carbapenems is class of ß-lactams that inactivate L,D-transpeptidases by acylation, although differences in antibiotic side chains modulate drug binding and acylation rates. Herein, we used a two-layered our Own N-layer integrated Molecular Mechanics ONIOM method to investigate the catalytic mechanism of L,D-transpeptidase 5 (LdtMt5) by ß-lactam derivatives. LdtMt5 complexes with six ß-lactams, ZINC03788344 (1), ZINC02462884 (2), ZINC03791246 (3), ZINC03808351 (4), ZINC03784242 (5) and ZINC02475683 (6) were simulated. The QM region (high-level) comprises the ß-lactam, one water molecule and the Cys360 catalytic residue, while the rest of the LdtMt5 residues were treated with AMBER force field. The activation energies (ΔG#) were calculated with B3LYP, M06-2X and ωB97X density functionals with 6-311++G(2d, 2p) basis set. The ΔG# for the acylation of LdtMt5 by the selected ß-lactams were obtained as 13.67, 20.90, 22.88, 24.29, 27.86 and 28.26 kcal mol-1respectively. Several of the compounds showed an improved ΔG# when compared to the previously calculated energies for imipenem and meropenem for the acylation step for LdtMt5. This model provides further validation of the catalytic inhibition mechanism of LDTs with atomistic detail.


Subject(s)
Peptidyl Transferases/chemistry , Quantitative Structure-Activity Relationship , beta-Lactams/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Peptidyl Transferases/antagonists & inhibitors , beta-Lactams/pharmacology
16.
Drug Dev Res ; 80(1): 11-18, 2019 02.
Article in English | MEDLINE | ID: mdl-30312987

ABSTRACT

Tuberculosis (TB) is a disease that has afflicted mankind for thousands of years, but in the last seven decades, much progress has been made in anti-TB therapy. Early drugs, such as para-aminosalicylic acid, streptomycin, isoniazid, and rifamycins were very effective in combatting the disease, giving rise to the hope that TB would be eradicated from the face of the earth by 2010. Despite that optimism, TB continues to kill more than a million people annually worldwide. A major reason for our inability to contain TB is the emergence drug resistance in Mycobacterium tuberculosis. This commentary is based on our recent publication on the structure of l,d-transpeptidase enzyme, relevant to drug resistance. As a background, we briefly outline the history and development of anti-TB therapy. Based on the crystal structure, we suggest a potential direction for designing more potent drugs against TB.


Subject(s)
Antitubercular Agents/administration & dosage , Drug Delivery Systems/methods , Drug Resistance, Bacterial/drug effects , Mycobacterium tuberculosis/drug effects , Peptidyl Transferases/antagonists & inhibitors , Tuberculosis/drug therapy , Animals , Cell Wall/drug effects , Cell Wall/metabolism , Drug Delivery Systems/trends , Drug Resistance, Bacterial/physiology , Humans , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Tuberculosis/enzymology
17.
Tuberculosis (Edinb) ; 113: 222-230, 2018 12.
Article in English | MEDLINE | ID: mdl-30514506

ABSTRACT

Mycobacterium tuberculosis is the causative agent of Tuberculosis. Formation of 3 → 3 crosslinks in the peptidoglycan layer of M. tuberculosis is catalyzed by l,d-transpeptidases. These enzymes can confer resistance against classical ß-lactams that inhibit enzymes that generate 4 → 3 peptidoglycan crosslinks. The focus of this study is to investigate the catalytic role of water molecules in the acylation mechanism of the ß-lactam ring within two models; 4- and 6-membered ring systems using two-layered our Own N-layer integrated Molecular Mechanics ONIOM (B3LYP/6-311++G(2d,2p): AMBER) model. The obtained thermochemical parameters revealed that the 6-membered ring model best describes the inhibition mechanism of acylation which indicates the role of water in the preference of 6-membered ring reaction pathway. This finding is in accordance with experimental data for the rate-limiting step of cysteine protease with the same class of inhibitor and binding affinity for both inhibitors. As expected, the ΔG# results also reveal that the 6-membered ring reaction pathway is the most favourable. The electrostatic potential (ESP) and the natural bond orbital analysis (NBO) showed stronger interactions in 6-membered ring transition state (TS-6) mechanism involving water in the active site of the enzyme. This study could be helpful in the development of novel antibiotics against l,d-transpeptidase.


Subject(s)
Bacterial Proteins/metabolism , Models, Molecular , Mycobacterium tuberculosis/enzymology , Peptidoglycan/metabolism , Peptidyl Transferases/metabolism , Water/metabolism , Acylation , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Binding Sites , Catalysis , Catalytic Domain , Imipenem/chemistry , Imipenem/metabolism , Imipenem/pharmacology , Kinetics , Meropenem/chemistry , Meropenem/metabolism , Meropenem/pharmacology , Molecular Structure , Mycobacterium tuberculosis/drug effects , Peptidyl Transferases/antagonists & inhibitors , Peptidyl Transferases/chemistry , Protein Binding
18.
J Comput Aided Mol Des ; 32(6): 687-701, 2018 06.
Article in English | MEDLINE | ID: mdl-29845435

ABSTRACT

Tuberculosis remains a dreadful disease that has claimed many human lives worldwide and elimination of the causative agent Mycobacterium tuberculosis also remains elusive. Multidrug-resistant TB is rapidly increasing worldwide; therefore, there is an urgent need for improving the current antibiotics and novel drug targets to successfully curb the TB burden. L,D-Transpeptidase 2 is an essential protein in Mtb that is responsible for virulence and growth during the chronic stage of the disease. Both D,D- and L,D-transpeptidases are inhibited concurrently to eradicate the bacterium. It was recently discovered that classic penicillins only inhibit D,D-transpeptidases, while L,D-transpeptidases are blocked by carbapenems. This has contributed to drug resistance and persistence of tuberculosis. Herein, a hybrid two-layered ONIOM (B3LYP/6-31G+(d): AMBER) model was used to extensively investigate the binding interactions of LdtMt2 complexed with four carbapenems (biapenem, imipenem, meropenem, and tebipenem) to ascertain molecular insight of the drug-enzyme complexation event. In the studied complexes, the carbapenems together with catalytic triad active site residues of LdtMt2 (His187, Ser188 and Cys205) were treated at with QM [B3LYP/6-31+G(d)], while the remaining part of the complexes were treated at MM level (AMBER force field). The resulting Gibbs free energy (ΔG), enthalpy (ΔH) and entropy (ΔS) for all complexes showed that the carbapenems exhibit reasonable binding interactions towards LdtMt2. Increasing the number of amino acid residues that form hydrogen bond interactions in the QM layer showed significant impact in binding interaction energy differences and the stabilities of the carbapenems inside the active pocket of LdtMt2. The theoretical binding free energies obtained in this study reflect the same trend of the experimental  observations. The electrostatic, hydrogen bonding and Van der Waals interactions between the carbapenems and LdtMt2 were also assessed. To further examine the nature of intermolecular interactions for carbapenem-LdtMt2 complexes, AIM and NBO analysis were performed for the QM region (carbapenems and the active residues of LdtMt2) of the complexes. These analyses revealed that the hydrogen bond interactions and charge transfer from the bonding to anti-bonding orbitals between catalytic residues of the enzyme and selected ligands enhances the binding and stability of carbapenem-LdtMt2 complexes. The two-layered ONIOM (B3LYP/6-31+G(d): Amber) model was used to evaluate the efficacy of FDA approved carbapenems antibiotics towards LdtMt2.


Subject(s)
Anti-Bacterial Agents/chemistry , Antitubercular Agents/chemistry , Bacterial Proteins/chemistry , Carbapenems/chemistry , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/chemistry , Catalytic Domain , Hydrogen Bonding , Peptidyl Transferases/antagonists & inhibitors , Protein Binding , Protein Conformation , Quantum Theory , Stereoisomerism , Thermodynamics
19.
J Antibiot (Tokyo) ; 71(8): 757-761, 2018 08.
Article in English | MEDLINE | ID: mdl-29700424

ABSTRACT

A new metabolite, cyclic dipeptide, cis-(3S,8aS)-3-(3,4-dihydroxybenzyl)hexahydropyrrolo[1,2-a]pyrazine-1,4-dione, named JS-3 was isolated from Streptomyces sp. 8812 fermentation broth. Its chemical structure was established by means of spectroscopic analysis. A wide-range-screening study, which included inhibition assay of DD-carboxypeptidase/transpeptidase activity, determination of antibacterial, antifungal, and antiproliferative activities as well as free-radical scavenging was performed. To authors knowledge, this is the first isolation of such compound from natural sources and the first one from bacteria, Streptomyces.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Carboxypeptidases/antagonists & inhibitors , Diketopiperazines/pharmacology , Dipeptides/pharmacology , Peptidyl Transferases/antagonists & inhibitors , Streptomyces/metabolism , Anti-Bacterial Agents/isolation & purification , Antifungal Agents/isolation & purification , Bacteria/drug effects , Diketopiperazines/isolation & purification , Diketopiperazines/metabolism , Dipeptides/isolation & purification , Dipeptides/metabolism , Fermentation , Fungi/drug effects
20.
AAPS J ; 20(2): 44, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29524047

ABSTRACT

The final step of peptidoglycan (PG) synthesis in all bacteria is the formation of cross-linkage between PG-stems. The cross-linking between amino acids in different PG chains gives the peptidoglycan cell wall a 3-dimensional structure and adds strength and rigidity to it. There are two distinct types of cross-linkages in bacterial cell walls. D,D-transpeptidase (D,D-TPs) generate the classical 4➔3 cross-linkages and the L,D-transpeptidase (L,D-TPs) generate the 3➔3 non-classical peptide cross-linkages. The present study is aimed at understanding the nature of drug resistance associated with L,D-TP and gaining insights for designing novel antibiotics against multi-drug resistant bacteria. Penicillin and cephalosporin classes of ß-lactams cannot inhibit L,D-TP function; however, carbapenems inactivate its function. We analyzed the structure of L,D-TP of Mycobacterium tuberculosis in the apo form and in complex with meropenem and imipenem. The periplasmic region of L,D-TP folds into three domains. The catalytic residues are situated in the C-terminal domain. The acylation reaction occurs between carbapenem antibiotics and the catalytic Cys-354 forming a covalent complex. This adduct formation mimics the acylation of L,D-TP with the donor PG-stem. A novel aspect of this study is that in the crystal structures of the apo and the carbapenem complexes, the N-terminal domain has a muropeptide unit non-covalently bound to it. Another interesting observation is that the calcium complex crystallized as a dimer through head and tail interactions between the monomers.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Resistance, Multiple, Bacterial , Mycobacterium tuberculosis/physiology , Peptidyl Transferases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Calcium/metabolism , Catalytic Domain/drug effects , Crystallography, X-Ray , Imipenem/chemistry , Imipenem/pharmacology , Meropenem/chemistry , Meropenem/pharmacology , Mycobacterium tuberculosis/drug effects , Peptidoglycan/biosynthesis , Peptidyl Transferases/chemistry , Peptidyl Transferases/isolation & purification , Peptidyl Transferases/metabolism , Protein Binding , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL