Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 296: 100041, 2021.
Article in English | MEDLINE | ID: mdl-33162395

ABSTRACT

O-acetyl serine sulfhydrylase (OASS), referred to as cysteine synthase (CS), synthesizes cysteine from O-acetyl serine (OAS) and sulfur in bacteria and plants. The inherent challenge for CS is to overcome 4 to 6 log-folds stronger affinity for its natural inhibitor, serine acetyltransferase (SAT), as compared with its affinity for substrate, OAS. Our recent study showed that CS employs a novel competitive-allosteric mechanism to selectively recruit its substrate in the presence of natural inhibitor. In this study, we trace the molecular features that control selective substrate recruitment. To generalize our findings, we used CS from three different bacteria (Haemophilus, Salmonella, and Mycobacterium) as our model systems and analyzed structural and substrate-binding features of wild-type CS and its ∼13 mutants. Results show that CS uses a noncatalytic residue, M120, located 20 Šaway from the reaction center, to discriminate in favor of substrate. M120A and background mutants display significantly reduced substrate binding, catalytic efficiency, and inhibitor binding. Results shows that M120 favors the substrate binding by selectively enhancing the affinity for the substrate and disengaging the inhibitor by 20 to 286 and 5- to 3-folds, respectively. Together, M120 confers a net discriminative force in favor of substrate by 100- to 858-folds.


Subject(s)
Cysteine Synthase/metabolism , Allosteric Regulation , Amino Acid Sequence , Amino Acid Substitution , Catalysis , Circular Dichroism , Crystallography, X-Ray , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Methionine/chemistry , Protein Conformation , Substrate Specificity
2.
Eur J Med Chem ; 192: 112157, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32145643

ABSTRACT

The l-cysteine is crucial for growth, survival, defense against oxidative stress, and pathogenesis of Entamoeba histolytica. The de novo biosynthesis of l-cysteine in E. histolytica, has a two-step pathway, where O-acetylserine sulfhydrylase (OASS) catalyses the last step by converting OAS to l-cysteine. This pathway is absent in humans and hence represents a promising target for novel therapeutics. E. histolytica expresses three isoforms of OASS and knockdown studies showed the importance of these enzymes for the survival of the pathogen. Here, we report the crystal structure of OASS isoform 3 from E. histolytica to 1.54 Å resolution. The active site geometries and kinetics of EhOASS3 and EhOASS1 structures were found to be very similar. Small-molecule libraries were screened against EhOASS3 and compounds were shortlisted based on the docking scores. F3226-1387 showed best inhibition with IC50 of 38 µM against EhOASS3 and was able to inhibit the growth of the organism to 72%.


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Entamoeba histolytica/cytology , Entamoeba histolytica/enzymology , Enzyme Inhibitors/pharmacology , Crystallography, X-Ray , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Entamoeba histolytica/growth & development , Enzyme Inhibitors/chemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship
3.
ACS Infect Dis ; 6(3): 467-478, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31887254

ABSTRACT

Increasing rates of drug-resistant Gram-negative (GN) infections, combined with a lack of new GN-effective antibiotic classes, are driving the need for the discovery of new agents. Bacterial metabolism represents an underutilized mechanism of action in current antimicrobial therapies. Therefore, we sought to identify novel antimetabolites that disrupt key metabolic pathways and explore the specific impacts of these agents on bacterial metabolism. This study describes the successful application of this approach to discover a new series of chemical probes, N-(phenyl)thioacetamide-linked 1,2,3-triazoles (TAT), that target cysteine synthase A (CysK), an enzyme unique to bacteria that is positioned at a key juncture between several fundamental pathways. The TAT class was identified using a high-throughput screen against Escherichia coli designed to identify modulators of pathways related to folate biosynthesis. TAT analog synthesis demonstrated a clear structure-activity relationship, and activity was confirmed against GN antifolate-resistant clinical isolates. Spontaneous TAT resistance mutations were tracked to CysK, and mode of action studies led to the identification of a false product formation mechanism between the CysK substrate O-acetyl-l-serine and the TATs. Global transcriptional responses to TAT treatment revealed that these antimetabolites impose substantial disruption of key metabolic networks beyond cysteine biosynthesis. This study highlights the potential of antimetabolite drug discovery as a promising approach to the discovery of novel GN antibiotics and the pharmacological promise of TAT CysK probes.


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Cysteine/biosynthesis , Escherichia coli/drug effects , Thioacetamide/pharmacology , Triazoles/pharmacology , Anti-Bacterial Agents/pharmacology , Antimetabolites/pharmacology , Drug Discovery , Escherichia coli/enzymology , High-Throughput Screening Assays , Metabolic Networks and Pathways/drug effects , Thioacetamide/chemistry , Triazoles/chemistry
4.
J Biomol Struct Dyn ; 37(2): 481-492, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29415627

ABSTRACT

OASS is a specific enzyme that helps Leishmania parasite to survive the oxidative stress condition in human macrophages. SAT C-terminal peptides in several organisms, including Leishmania, were reported to inhibit or reduce the activity of OASS. Small peptide and small molecules mimicking the SAT C-terminal residues are designed and tested for the inhibition of OASS in different organisms. Hence, in this study, all the possible tetra-peptide combinations were designed and screened based on the docking ability with Leishmania donovani OASS (Ld-OASS). The top ranked peptides were further validated for the stability using 50 ns molecular dynamic simulation. In order to identify the better binding capability of the peptides, the top peptides complexed with Ld-OASS were also subjected to molecular dynamic simulation. The docking and simulation results favored the peptide EWSI to possess greater advantage than previously reported peptide (DWSI) in binding with Ld-OASS active site. Also, screening of non-peptide inhibitor of Asinex Biodesign library based on the shape similarity of EWSI and DWSI was performed. The top similar molecules of each peptides were docked on to Ld-OASS active site and subsequently simulated for 20 ns. The results suggested that the ligand that shares high shape similarity with EWSI possess better binding capability than the ligand that shares high shape similarity with DWSI. This study revealed that the tetra-peptide EWSI had marginal advantage over DWSI in binding with Ld-OASS, thereby providing basis for defining a pharmacophoric scaffold for the design of peptidomimetic inhibitors as well as non-peptide inhibitors of Ld-OASS. Communicated by Ramaswamy H. Sarma.


Subject(s)
Cysteine Synthase/chemistry , Drug Discovery , Enzyme Inhibitors/chemistry , Leishmania donovani/enzymology , Models, Molecular , Peptides/chemistry , Quantitative Structure-Activity Relationship , Cysteine Synthase/antagonists & inhibitors , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Hydrogen Bonding , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptides/pharmacology
5.
J Enzyme Inhib Med Chem ; 34(1): 31-43, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30362368

ABSTRACT

The lack of efficacy of current antibacterials to treat multidrug resistant bacteria poses a life-threatening alarm. In order to develop enhancers of the antibacterial activity, we carried out a medicinal chemistry campaign aiming to develop inhibitors of enzymes that synthesise cysteine and belong to the reductive sulphur assimilation pathway, absent in mammals. Previous studies have provided a novel series of inhibitors for O-acetylsulfhydrylase - a key enzyme involved in cysteine biosynthesis. Despite displaying nanomolar affinity, the most active representative of the series was not able to interfere with bacterial growth, likely due to poor permeability. Therefore, we rationally modified the structure of the hit compound with the aim of promoting their passage through the outer cell membrane porins. The new series was evaluated on the recombinant enzyme from Salmonella enterica serovar Typhimurium, with several compounds able to keep nanomolar binding affinity despite the extent of chemical manipulation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carboxylic Acids/pharmacology , Cyclopropanes/pharmacology , Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Carboxylic Acids/chemical synthesis , Carboxylic Acids/chemistry , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Cysteine Synthase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/drug effects , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Microbial Sensitivity Tests , Molecular Structure , Salmonella typhimurium/enzymology , Structure-Activity Relationship
6.
J Enzyme Inhib Med Chem ; 33(1): 1444-1452, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30221554

ABSTRACT

Several bacteria rely on the reductive sulphur assimilation pathway, absent in mammals, to synthesise cysteine. Reduction of virulence and decrease in antibiotic resistance have already been associated with mutations on the genes that codify cysteine biosynthetic enzymes. Therefore, inhibition of cysteine biosynthesis has emerged as a promising strategy to find new potential agents for the treatment of bacterial infection. Following our previous efforts to explore OASS inhibition and to expand and diversify our library, a scaffold hopping approach was carried out, with the aim of identifying a novel fragment for further development. This novel chemical tool, endowed with favourable pharmacological characteristics, was successfully developed, and a preliminary Structure-Activity Relationship investigation was carried out.


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Drug Design , Small Molecule Libraries/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/enzymology , Bacteria/genetics , Binding Sites , Biological Assay , Computer Simulation , DNA, Recombinant/chemistry , DNA, Recombinant/genetics , Ligands , Models, Molecular , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
7.
J Enzyme Inhib Med Chem ; 33(1): 1343-1351, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30251899

ABSTRACT

O-acetylserine sulfhydrylase (OASS) is the pyridoxal 5'-phosphate dependent enzyme that catalyses the formation of L-cysteine in bacteria and plants. Its inactivation is pursued as a strategy for the identification of novel antibiotics that, targeting dispensable proteins, holds a great promise for circumventing resistance development. In the present study, we have investigated the reactivity of Salmonella enterica serovar Typhimurium OASS-A and OASS-B isozymes with fluoroalanine derivatives. Monofluoroalanine reacts with OASS-A and OASS-B forming either a stable or a metastable α-aminoacrylate Schiff's base, respectively, as proved by spectral changes. This finding indicates that monofluoroalanine is a substrate analogue, as previously found for other beta-halogenalanine derivatives. Trifluoroalanine caused different and time-dependent absorbance and fluorescence spectral changes for the two isozymes and is associated with irreversible inhibition. The time course of enzyme inactivation was found to be characterised by a biphasic behaviour. Partially distinct inactivation mechanisms for OASS-A and OASS-B are proposed.


Subject(s)
Alanine/analogs & derivatives , Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Alanine/chemical synthesis , Alanine/chemistry , Alanine/pharmacology , Cysteine Synthase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Salmonella enterica/enzymology , Structure-Activity Relationship
8.
Plant Cell Physiol ; 59(5): 1072-1083, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29490083

ABSTRACT

In Arabidopsis thaliana, cyanide is produced concomitantly with ethylene biosynthesis and is mainly detoxified by the ß-cyanoalanine synthase CAS-C1. In roots, CAS-C1 activity is essential to maintain a low level of cyanide for proper root hair development. Root hair elongation relies on polarized cell expansion at the growing tip, and we have observed that CAS-C1 locates in mitochondria and accumulates in root hair tips during root hair elongation, as shown by observing the fluorescence in plants transformed with the translational construct ProC1:CASC1-GFP, containing the complete CAS-C1 gene fused to green fluorescent protein (GFP). Mutants in the SUPERCENTIPEDE (SCN1) gene, that regulate the NADPH oxidase gene ROOT HAIR DEFECTIVE 2 (RHD2)/AtrbohC, are affected at the very early steps of the development of root hair that do not elongate and do not show a preferential localization of the GFP accumulation in the tips of the root hair primordia. Root hairs of mutants in CAS-C1 or RHD2/AtrbohC, whose protein product catalyzes the generation of ROS and the Ca2+ gradient, start to grow out correctly, but they do not elongate. Genetic crosses between the cas-c1 mutant and scn1 or rhd2 mutants were performed, and the detailed phenotypic and molecular characterization of the double mutants demonstrates that scn1 mutation is epistatic to cas-c1 and cas-c1 is epistatic to rhd2 mutation, indicating that CAS-C1 acts in early steps of the root hair development process. Moreover, our results show that the role of CAS-C1 in root hair elongation is independent of H2O2 production and of a direct NADPH oxidase inhibition by cyanide.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Arabidopsis/growth & development , Cyanides/toxicity , Cysteine Synthase/metabolism , Lyases/metabolism , NADPH Oxidases/metabolism , Plant Roots/enzymology , Plant Roots/growth & development , Adenosine Triphosphate/metabolism , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/genetics , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/genetics , Enzyme Activation/drug effects , Epistasis, Genetic/drug effects , Gene Expression Regulation, Plant/drug effects , Hydrogen Peroxide/metabolism , Hydroxocobalamin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Mutation/genetics , NADPH Oxidases/antagonists & inhibitors , Phenotype , Plant Roots/drug effects , Superoxides/metabolism
9.
J Chem Inf Model ; 58(3): 710-723, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29481752

ABSTRACT

Saturation transfer difference (STD) is an NMR technique conventionally applied in drug discovery to identify ligand moieties relevant for binding to protein cavities. This is important to direct medicinal chemistry efforts in small-molecule optimization processes. However, STD does not provide any structural details about the ligand-target complex under investigation. Herein, we report the application of a new integrated approach, which combines enhanced sampling methods with STD experiments, for the characterization of ligand-target complexes that are instrumental for drug design purposes. As an example, we have studied the interaction between StOASS-A, a potential antibacterial target, and an inhibitor previously reported. This approach allowed us to consider the ligand-target complex from a dynamic point of view, revealing the presence of an accessory subpocket which can be exploited to design novel StOASS-A inhibitors. As a proof of concept, a small library of derivatives was designed and evaluated in vitro, displaying the expected activity.


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/metabolism , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Salmonella typhimurium/enzymology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Cysteine Synthase/chemistry , Drug Design , Ligands , Magnetic Resonance Spectroscopy/methods , Molecular Docking Simulation , Molecular Dynamics Simulation , Salmonella typhimurium/drug effects , Thermodynamics
10.
Bioorg Med Chem Lett ; 27(19): 4582-4587, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28882483

ABSTRACT

CysK1 and CysK2 are two members of the cysteine/S-sulfocysteine synthase family in Mycobacterium tuberculosis, responsible for the de novo biosynthesis of l-cysteine, which is subsequently used as a building block for mycothiol. This metabolite is the first line defense of this pathogen against reactive oxygen and nitrogen species released by host macrophages after phagocytosis. In a previous medicinal chemistry campaign we had developed urea-based inhibitors of the cysteine synthase CysM with bactericidal activity against dormant M. tuberculosis. In this study we extended these efforts by examination of the in vitro activities of a library consisting of 71 urea compounds against CysK1 and CysK2. Binding was established by fluorescence spectroscopy and inhibition by enzyme assays. Several of the compounds inhibited these two cysteine synthases, with the most potent inhibitor displaying an IC50 value of 2.5µM for CysK1 and 6.6µM for CysK2, respectively. Four of the identified molecules targeting CysK1 and CysK2 were also among the top ten inhibitors of CysM, suggesting that potent compounds could be developed with activity against all three enzymes.


Subject(s)
Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Urea/pharmacology , Cysteine Synthase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
11.
Biochemistry ; 56(37): 5011-5025, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28805060

ABSTRACT

By classical competitive antagonism, a substrate and competitive inhibitor must bind mutually exclusively to the active site. The competitive inhibition of O-acetyl serine sulfhydrylase (OASS) by the C-terminus of serine acetyltransferase (SAT) presents a paradox, because the C-terminus of SAT binds to the active site of OASS with an affinity that is 4-6 log-fold (104-106) greater than that of the substrate. Therefore, we employed multiple approaches to understand how the substrate gains access to the OASS active site under physiological conditions. Single-molecule and ensemble approaches showed that the active site-bound high-affinity competitive inhibitor is actively dissociated by the substrate, which is not consistent with classical views of competitive antagonism. We employed fast-flow kinetic approaches to demonstrate that substrate-mediated dissociation of full length SAT-OASS (cysteine regulatory complex) follows a noncanonical "facilitated dissociation" mechanism. To understand the mechanism by which the substrate induces inhibitor dissociation, we resolved the crystal structures of enzyme·inhibitor·substrate ternary complexes. Crystal structures reveal a competitive allosteric binding mechanism in which the substrate intrudes into the inhibitor-bound active site and disengages the inhibitor before occupying the site vacated by the inhibitor. In summary, here we reveal a new type of competitive allosteric binding mechanism by which one of the competitive antagonists facilitates the dissociation of the other. Together, our results indicate that "competitive allostery" is the general feature of noncanonical "facilitated/accelerated dissociation" mechanisms. Further understanding of the mechanistic framework of "competitive allosteric" mechanism may allow us to design a new family of "competitive allosteric drugs/small molecules" that will have improved selectivity and specificity as compared to their competitive and allosteric counterparts.


Subject(s)
Alanine/analogs & derivatives , Bacterial Proteins/antagonists & inhibitors , Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Haemophilus influenzae/enzymology , Models, Molecular , Salmonella enterica/metabolism , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Alanine/pharmacology , Allosteric Regulation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Binding, Competitive , Catalytic Domain , Crystallography, X-Ray , Cysteine Synthase/chemistry , Cysteine Synthase/genetics , Cysteine Synthase/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Haemophilus influenzae/metabolism , Kinetics , Ligands , Molecular Conformation , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Oligopeptides/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Salmonella enterica/enzymology , Serine/chemistry , Serine/metabolism , Serine O-Acetyltransferase/chemistry , Serine O-Acetyltransferase/genetics , Serine O-Acetyltransferase/metabolism , Serine O-Acetyltransferase/pharmacology
12.
J Enzyme Inhib Med Chem ; 31(sup4): 78-87, 2016.
Article in English | MEDLINE | ID: mdl-27578398

ABSTRACT

Cysteine is a building block for many biomolecules that are crucial for living organisms. O-Acetylserine sulfhydrylase (OASS), present in bacteria and plants but absent in mammals, catalyzes the last step of cysteine biosynthesis. This enzyme has been deeply investigated because, beside the biosynthesis of cysteine, it exerts a series of "moonlighting" activities in bacteria. We have previously reported a series of molecules capable of inhibiting Salmonella typhimurium (S. typhymurium) OASS isoforms at nanomolar concentrations, using a combination of computational and spectroscopic approaches. The cyclopropane-1,2-dicarboxylic acids presented herein provide further insights into the binding mode of small molecules to OASS enzymes. Saturation transfer difference NMR (STD-NMR) was used to characterize the molecule/enzyme interactions for both OASS-A and B. Most of the compounds induce a several fold increase in fluorescence emission of the pyridoxal 5'-phosphate (PLP) coenzyme upon binding to either OASS-A or OASS-B, making these compounds excellent tools for the development of competition-binding experiments.


Subject(s)
Cyclopropanes/pharmacology , Cysteine Synthase/antagonists & inhibitors , Dicarboxylic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Fluorometry , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Cysteine Synthase/chemistry , Cysteine Synthase/metabolism , Dicarboxylic Acids/chemical synthesis , Dicarboxylic Acids/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
13.
J Med Chem ; 59(14): 6848-59, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27379713

ABSTRACT

Cysteine is an important amino acid in the redox defense of Mycobacterium tuberculosis, primarily as a building block of mycothiol. Genetic studies have implicated de novo cysteine biosynthesis in pathogen survival in infected macrophages, in particular for persistent M. tuberculosis. Here, we report on the identification and characterization of potent inhibitors of CysM, a critical enzyme in cysteine biosynthesis during dormancy. A screening campaign of 17 312 compounds identified ligands that bind to the active site with micromolar affinity. These were characterized in terms of their inhibitory potencies and structure-activity relationships through hit expansion guided by three-dimensional structures of enzyme-inhibitor complexes. The top compound binds to CysM with 300 nM affinity and displays selectivity over the mycobacterial homologues CysK1 and CysK2. Notably, two inhibitors show significant potency in a nutrient-starvation model of dormancy of Mycobacterium tuberculosis, with little or no cytotoxicity toward mammalian cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cell Line , Cysteine Synthase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/enzymology , Structure-Activity Relationship
14.
FEBS Lett ; 590(7): 943-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27072053

ABSTRACT

O-acetylserine sulfhydrylase (OASS), the enzyme catalysing the last step of cysteine biosynthesis in bacteria, is involved in antibiotic resistance and biofilm formation. Since mammals lack OASS, it is a potential target for antimicrobials. However, a limited number of inhibitors has been developed and crystallized in complex with OASS. STD-NMR was applied to study the interaction of the inhibitory pentapeptide MNYDI with the CysK and CysM OASS isozymes from Salmonella Typhimurium. Results are in excellent agreement with docking and SAR studies and confirm the important role played by the C-terminal Ile5 and the arylic moiety at P3 in dictating affinity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Models, Molecular , Oligopeptides/pharmacology , Salmonella typhimurium/enzymology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cysteine Synthase/chemistry , Cysteine Synthase/genetics , Cysteine Synthase/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Epitope Mapping , Haemophilus influenzae/enzymology , Hydrogen Bonding , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Docking Simulation , Nuclear Magnetic Resonance, Biomolecular , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Library , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structural Homology, Protein
15.
J Med Chem ; 59(6): 2567-78, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26894308

ABSTRACT

Cysteine is a building block for several biomolecules that are crucial for living organisms. The last step of cysteine biosynthesis is catalyzed by O-acetylserine sulfydrylase (OASS), a highly conserved pyridoxal 5'-phosphate (PLP)-dependent enzyme, present in different isoforms in bacteria, plants, and nematodes, but absent in mammals. Beside the biosynthesis of cysteine, OASS exerts a series of "moonlighting" activities in bacteria, such as transcriptional regulation, contact-dependent growth inhibition, swarming motility, and induction of antibiotic resistance. Therefore, the discovery of molecules capable of inhibiting OASS would be a valuable tool to unravel how this protein affects the physiology of unicellular organisms. As a continuation of our efforts toward the synthesis of OASS inhibitors, in this work we have used a combination of computational and spectroscopic approaches to rationally design, synthesize, and test a series of substituted 2-phenylcyclopropane carboxylic acids that bind to the two S. typhymurium OASS isoforms at nanomolar concentrations.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Carboxylic Acids/chemical synthesis , Carboxylic Acids/pharmacology , Cyclopropanes/chemical synthesis , Cyclopropanes/pharmacology , Cysteine Synthase/antagonists & inhibitors , Salmonella typhimurium/drug effects , Salmonella typhimurium/enzymology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Isoenzymes/antagonists & inhibitors , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Models, Molecular , Protein Binding , Pyridoxal Phosphate/chemistry , Salmonella typhimurium/growth & development , Structure-Activity Relationship
16.
Curr Top Med Chem ; 16(9): 948-59, 2016.
Article in English | MEDLINE | ID: mdl-26303427

ABSTRACT

The cysteine biosynthetic pathway is of fundamental importance for the growth, survival, and pathogenicity of the many pathogens. This pathway is present in many species but is absent in mammals. The ability of pathogens to counteract the oxidative defences of a host is critical for the survival of these pathogens during their long latent phases, especially in anaerobic pathogens such as Entamoeba histolytica, Leishmania donovani, Trichomonas vaginalis, and Salmonella typhimurium. All of these organisms rely on the de novo cysteine biosynthetic pathway to assimilate sulphur and maintain a ready supply of cysteine. The de novo cysteine biosynthetic pathway, on account of its being important for the survival of pathogens and at the same time being absent in mammals, is an important drug target for diseases such as amoebiasis, trichomoniasis & tuberculosis. Cysteine biosynthesis is catalysed by two enzymes: serine acetyl transferase (SAT) followed by O-acetylserine sulfhydrylase (OASS). OASS is well studied, and with the availability of crystal structures of this enzyme in different conformations, it is a suitable template for structure-based inhibitor development. Moreover, OASS is highly conserved, both structurally and sequence-wise, among the above-mentioned organisms. There have been several reports of inhibitor screening and development against this enzyme from different organisms such as Salmonella typhimurium, Mycobacterium tuberculosis and Entamoeba histolytica. All of these inhibitors have been reported to display micromolar to nanomolar binding affinities for the open conformation of the enzyme. In this review, we highlight the structural similarities of this enzyme in different organisms and the attempts for inhibitor development so far. We also propose that the intermediate state of the enzyme may be the ideal target for the design of effective highaffinity inhibitors.


Subject(s)
Biosynthetic Pathways/drug effects , Cysteine Synthase/antagonists & inhibitors , Cysteine/biosynthesis , Drug Design , Enzyme Inhibitors/pharmacology , Cysteine Synthase/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
17.
Curr Med Chem ; 22(2): 187-213, 2015.
Article in English | MEDLINE | ID: mdl-25388010

ABSTRACT

The rising emergence of antibiotic resistance urges the search for new strategies to defeat microorganisms that lead to persistent infections of the host. Tolerant to antibiotics, slowly replicating bacteria often cause latent and persistent infections that are the most challenging for pharmacological treatment. Persistence inside the host requires an extensive re-programming of the pathogen metabolic functions, due to the extremely hostile environment they face. Therefore, targeting key metabolic functions could result in better antibiotic treatments, shortened latency periods, and increased susceptibility to traditional antibiotics. Bacteria, differently from mammals, assimilate inorganic sulfur into cysteine, the precursor of a number of key metabolites including reducing agents, cofactors and membrane components. Inhibition of cysteine biosynthesis was proven to interfere heavily with the ability of pathogens to fight oxidative stress, to infect the host and to establish long-term infections. This review has the purpose of i) briefly summarizing the key structural and functional properties of transporters and enzymes involved in sulfur assimilation, ii) presenting biological evidence that supports the exploitation of this pathway for the identification of potential targets and, iii) highlighting intense efforts and advancements in the search of promising candidates for the development of novel compounds that enhance antibiotics therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/metabolism , Sulfur/metabolism , Amino Acid Sequence , Animals , Cysteine/biosynthesis , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/metabolism , Drug Resistance, Microbial , Humans , Models, Molecular , Molecular Sequence Data
18.
Biochim Biophys Acta ; 1854(9): 1175-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25484279

ABSTRACT

The alarming increase of drug resistance in Mycobacterium tuberculosis strains poses a severe threat to human health. Chemotherapy is particularly challenging because M. tuberculosis can persist in the lungs of infected individuals; estimates of the WHO indicate that about 1/3 of the world population is infected with latent tuberculosis providing a large reservoir for relapse and subsequent spread of the disease. Persistent M. tuberculosis shows considerable tolerance towards conventional antibiotics making treatment particularly difficult. In this phase the bacilli are exposed to oxygen and nitrogen radicals generated as part of the host response and redox-defense mechanisms are thus vital for the survival of the pathogen. Sulfur metabolism and de novo cysteine biosynthesis have been shown to be important for the redox homeostasis in persistent M. tuberculosis and these pathways could provide promising targets for novel antibiotics for the treatment of the latent form of the disease. Recent research has provided evidence for three de novo metabolic routes of cysteine biosynthesis in M. tuberculosis, each with a specific PLP dependent cysteine synthase with distinct substrate specificities. In this review we summarize our present understanding of these pathways, with a focus on the advances on functional and mechanistic characterization of mycobacterial PLP dependent cysteine synthases, their role in the various pathways to cysteine, and first attempts to develop specific inhibitors of mycobacterial cysteine biosynthesis. This article is part of a Special Issue entitled: Cofactor-dependent proteins: evolution, chemical diversity and bio-applications.


Subject(s)
Cysteine Synthase/chemistry , Mycobacterium tuberculosis/enzymology , Pyridoxal Phosphate/physiology , Anti-Bacterial Agents/pharmacology , Cysteine/biosynthesis , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/metabolism , Humans , Mycobacterium tuberculosis/drug effects
19.
PLoS One ; 8(10): e77558, 2013.
Article in English | MEDLINE | ID: mdl-24167577

ABSTRACT

The last step of cysteine biosynthesis in bacteria and plants is catalyzed by O-acetylserine sulfhydrylase. In bacteria, two isozymes, O-acetylserine sulfhydrylase-A and O-acetylserine sulfhydrylase-B, have been identified that share similar binding sites, although the respective specific functions are still debated. O-acetylserine sulfhydrylase plays a key role in the adaptation of bacteria to the host environment, in the defense mechanisms to oxidative stress and in antibiotic resistance. Because mammals synthesize cysteine from methionine and lack O-acetylserine sulfhydrylase, the enzyme is a potential target for antimicrobials. With this aim, we first identified potential inhibitors of the two isozymes via a ligand- and structure-based in silico screening of a subset of the ZINC library using FLAP. The binding affinities of the most promising candidates were measured in vitro on purified O-acetylserine sulfhydrylase-A and O-acetylserine sulfhydrylase-B from Salmonella typhimurium by a direct method that exploits the change in the cofactor fluorescence. Two molecules were identified with dissociation constants of 3.7 and 33 µM for O-acetylserine sulfhydrylase-A and O-acetylserine sulfhydrylase-B, respectively. Because GRID analysis of the two isoenzymes indicates the presence of a few common pharmacophoric features, cross binding titrations were carried out. It was found that the best binder for O-acetylserine sulfhydrylase-B exhibits a dissociation constant of 29 µM for O-acetylserine sulfhydrylase-A, thus displaying a limited selectivity, whereas the best binder for O-acetylserine sulfhydrylase-A exhibits a dissociation constant of 50 µM for O-acetylserine sulfhydrylase-B and is thus 8-fold selective towards the former isozyme. Therefore, isoform-specific and isoform-independent ligands allow to either selectively target the isozyme that predominantly supports bacteria during infection and long-term survival or to completely block bacterial cysteine biosynthesis.


Subject(s)
Anti-Bacterial Agents , Bacteria , Bacterial Proteins/antagonists & inhibitors , Cysteine Synthase/antagonists & inhibitors , Enzyme Inhibitors , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/enzymology , Bacteria/growth & development , Bacterial Proteins/metabolism , Cysteine/biosynthesis , Cysteine Synthase/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism
20.
Biomed Res Int ; 2013: 390920, 2013.
Article in English | MEDLINE | ID: mdl-24073401

ABSTRACT

In our presented research, we made an attempt to predict the 3D model for cysteine synthase (A2GMG5_TRIVA) using homology-modeling approaches. To investigate deeper into the predicted structure, we further performed a molecular dynamics simulation for 10 ns and calculated several supporting analysis for structural properties such as RMSF, radius of gyration, and the total energy calculation to support the predicted structured model of cysteine synthase. The present findings led us to conclude that the proposed model is stereochemically stable. The overall PROCHECK G factor for the homology-modeled structure was -0.04. On the basis of the virtual screening for cysteine synthase against the NCI subset II molecule, we present the molecule 1-N, 4-N-bis [3-(1H-benzimidazol-2-yl) phenyl] benzene-1,4-dicarboxamide (ZINC01690699) having the minimum energy score (-13.0 Kcal/Mol) and a log P value of 6 as a potential inhibitory molecule used to inhibit the growth of T. vaginalis infection.


Subject(s)
Antitrichomonal Agents/pharmacology , Antitrichomonal Agents/therapeutic use , Cysteine Synthase/antagonists & inhibitors , Cysteine Synthase/chemistry , Molecular Dynamics Simulation , Trichomonas Infections/drug therapy , Trichomonas/enzymology , Catalytic Domain , Cysteine Synthase/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions/drug effects , Ligands , Reproducibility of Results , Software , Substrate Specificity/drug effects , Thermodynamics , Trichomonas/drug effects , User-Computer Interface
SELECTION OF CITATIONS
SEARCH DETAIL
...