Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Curr Opin Chem Biol ; 74: 102287, 2023 06.
Article in English | MEDLINE | ID: mdl-36948086

ABSTRACT

How has metabolomics helped our understanding of infectious diseases? With the threat of antimicrobial resistance to human health around the world, metabolomics has emerged as a powerful tool to comprehensively characterize metabolic pathways to identify new drug targets. However, its output is constrained to known metabolites and their metabolic pathways. Recent advances in instrumentation, methodologies, and computational mass spectrometry have accelerated the use of metabolomics to understand pathogen-host metabolic interactions. This short review discusses a selection of recent publications using metabolomics in infectious/bacterial diseases. These studies unravel the links between metabolic adaptations to environments and host metabolic responses. Moreover, they highlight the importance of enzyme function and metabolite characterization in identifying new drug targets and biomarkers, as well as precision medicine in monitoring therapeutics and diagnosing diseases.


Subject(s)
Bacteria , Metabolomics , Humans , Metabolomics/methods , Mass Spectrometry/methods , Biomarkers , Metabolic Networks and Pathways
2.
Curr Opin Microbiol ; 69: 102191, 2022 10.
Article in English | MEDLINE | ID: mdl-35970040

ABSTRACT

Tuberculosis (TB) persists as a major global health issue and a leading cause of death by a single infectious agent. The global burden of TB is further exacerbated by the continuing emergence and dissemination of strains of Mycobacterium tuberculosis resistant to multiple antibiotics. The need for novel drugs that can be used to shorten the course for current TB drug regimens as well as combat the persistent threat of antibiotic resistance has never been greater. There have been significant advances in the discovery of de novo TB treatments, with the first TB-specific drugs in 45 years approved for use. However, there are still issues that restrict the pipeline of new antitubercular chemotherapies. The rate of failure of TB drug candidates in clinical trials remains high, while the validation of new TB drug targets and subsequent identification of novel inhibitors remains modest.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Drug Delivery Systems , Humans , Mycobacterium tuberculosis/genetics , Tuberculosis/drug therapy , Tuberculosis/microbiology
3.
Methods Enzymol ; 665: 29-47, 2022.
Article in English | MEDLINE | ID: mdl-35379439

ABSTRACT

Most of the chemical diversity present in the natural world derives from the incredible ability of enzymes to act on and control metabolism. Yet, thousands of enzymes have no defined function. The capacity to probe, investigate and assign previously unknown enzyme function with speed and confidence is therefore highly sought-after. Metabolomics is becoming a dominant player in the field of functional genomics and, when coupled with genetic tools and protein biochemistry techniques, has enabled unbiased, de novo annotation of orphan enzymes both in vitro and ex vivo. In this chapter, we describe two distinct experimental and analytical metabolomic methodologies used to reveal enzyme function. Activity-based metabolomic profiling (ABMP) is an in vitro technique that enables tracking of enzyme-induced changes in a complex metabolite extract. Global metabolomic profiling permits the comparison of extracted cellular metabolome of groups of samples (e.g., wild-type versus mutant bacteria). The methods we describe present the advantage of generating cell extracts containing a broad range of metabolites in their native states, which can then be used to identify substrates for orphan enzymes. This chapter aims to provide a guide for the use of these metabolomic techniques by scientists interested in identifying bona fide physiological substrates of orphan enzymes and the metabolic pathways they belong to.


Subject(s)
Metabolome , Metabolomics , Bacteria , Metabolic Networks and Pathways , Metabolomics/methods
4.
Eur J Med Chem ; 230: 114105, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35065413

ABSTRACT

There is a pressing need for new drugs against tuberculosis (TB) to combat the growing resistance to current antituberculars. Herein a novel strategy is described for hit generation against promising TB targets involving X-ray crystallographic screening in combination with phenotypic screening. This combined approach (XP Screen) affords both a validation of target engagement as well as determination of in cellulo activity. The utility of this method is illustrated by way of an XP Screen against CYP121A1, a cytochrome P450 enzyme from Mycobacterium tuberculosis (Mtb) championed as a validated drug discovery target. A focused screening set was synthesized and tested by such means, with several members of the set showing promising activity against Mtb strain H37Rv. One compound was observed as an X-ray hit against CYP121A1 and showed improved activity against Mtb strain H37Rv under multiple assay conditions (pan-assay activity). Data obtained during X-ray crystallographic screening were utilized in a structure-based campaign to design a limited number of analogues (less than twenty), many of which also showed pan-assay activity against Mtb strain H37Rv. These included the benzo[b][1,4]oxazine derivative (MIC90 6.25 µM), a novel hit compound suitable as a starting point for a more involved hit to lead candidate medicinal chemistry campaign.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Antitubercular Agents/pharmacology , Drug Design , Humans , Tuberculosis/drug therapy , X-Rays
5.
Nat Chem Biol ; 16(6): 686-694, 2020 06.
Article in English | MEDLINE | ID: mdl-32203411

ABSTRACT

The broad-spectrum antibiotic D-cycloserine (DCS) is a key component of regimens used to treat multi- and extensively drug-resistant tuberculosis. DCS, a structural analog of D-alanine, binds to and inactivates two essential enzymes involved in peptidoglycan biosynthesis, alanine racemase (Alr) and D-Ala:D-Ala ligase. Inactivation of Alr is thought to proceed via a mechanism-based irreversible route, forming an adduct with the pyridoxal 5'-phosphate cofactor, leading to bacterial death. Inconsistent with this hypothesis, Mycobacterium tuberculosis Alr activity can be detected after exposure to clinically relevant DCS concentrations. To address this paradox, we investigated the chemical mechanism of Alr inhibition by DCS. Inhibition of M. tuberculosis Alr and other Alrs is reversible, mechanistically revealed by a previously unidentified DCS-adduct hydrolysis. Dissociation and subsequent rearrangement to a stable substituted oxime explains Alr reactivation in the cellular milieu. This knowledge provides a novel route for discovery of improved Alr inhibitors against M. tuberculosis and other bacteria.


Subject(s)
Alanine Racemase/metabolism , Antibiotics, Antitubercular/chemistry , Cycloserine/chemistry , Recombinant Proteins/metabolism , Alanine/chemistry , Alanine/metabolism , Alanine Racemase/genetics , Amino Acid Sequence , Antibiotics, Antitubercular/metabolism , Bacterial Proteins/metabolism , Binding Sites , Cycloserine/metabolism , Escherichia coli , Isoxazoles/chemistry , Ligases/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Oximes/chemistry , Protein Binding , Protein Conformation , Recombinant Proteins/genetics
6.
Nat Commun ; 10(1): 4177, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519879

ABSTRACT

Drug resistant infections represent one of the most challenging medical problems of our time. D-cycloserine is an antibiotic used for six decades without significant appearance and dissemination of antibiotic resistant strains, making it an ideal model compound to understand what drives resistance evasion. We therefore investigated why Mycobacterium tuberculosis fails to become resistant to D-cycloserine. To address this question, we employed a combination of bacterial genetics, genomics, biochemistry and fitness analysis in vitro, in macrophages and in mice. Altogether, our results suggest that the ultra-low rate of emergence of D-cycloserine resistance mutations is the dominant biological factor delaying the appearance of clinical resistance to this antibiotic. Furthermore, we also identified potential compensatory mechanisms able to minimize the severe fitness costs of primary D-cycloserine resistance conferring mutations.


Subject(s)
Cycloserine/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Animals , Antibiotics, Antitubercular/pharmacology , Blotting, Western , Drug Resistance, Bacterial/genetics , Genotype , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Monocytes/drug effects , Monocytes/metabolism , Mutation/genetics , Mycobacterium tuberculosis/genetics
7.
Mol Microbiol ; 112(4): 1284-1307, 2019 10.
Article in English | MEDLINE | ID: mdl-31389636

ABSTRACT

Bacterial nutrition is an essential aspect of host-pathogen interaction. For the intracellular pathogen Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis in humans, fatty acids derived from lipid droplets are considered the major carbon source. However, many other soluble nutrients are available inside host cells and may be used as alternative carbon sources. Lactate and pyruvate are abundant in human cells and fluids, particularly during inflammation. In this work, we study Mtb metabolism of lactate and pyruvate combining classic microbial physiology with a 'multi-omics' approach consisting of transposon-directed insertion site sequencing (TraDIS), RNA-seq transcriptomics, proteomics and stable isotopic labelling coupled with mass spectrometry-based metabolomics. We discovered that Mtb is well adapted to use both lactate and pyruvate and that their metabolism requires gluconeogenesis, valine metabolism, the Krebs cycle, the GABA shunt, the glyoxylate shunt and the methylcitrate cycle. The last two pathways are traditionally associated with fatty acid metabolism and, unexpectedly, we found that in Mtb the methylcitrate cycle operates in reverse, to allow optimal metabolism of lactate and pyruvate. Our findings reveal a novel function for the methylcitrate cycle as a direct route for the biosynthesis of propionyl-CoA, the essential precursor for the biosynthesis of the odd-chain fatty acids.


Subject(s)
Lactic Acid/metabolism , Mycobacterium tuberculosis/metabolism , Pyruvic Acid/metabolism , Acyl Coenzyme A/metabolism , Bacterial Proteins/metabolism , Citrate (si)-Synthase/metabolism , Citrates/metabolism , Fatty Acids/metabolism , Gene Expression Regulation, Bacterial/genetics , Glyoxylates , Tuberculosis/microbiology
8.
ChemistryOpen ; 8(7): 995-1011, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31367508

ABSTRACT

The emergence of untreatable drug-resistant strains of Mycobacterium tuberculosis is a major public health problem worldwide, and the identification of new efficient treatments is urgently needed. Mycobacterium tuberculosis cytochrome P450 CYP121A1 is a promising drug target for the treatment of tuberculosis owing to its essential role in mycobacterial growth. Using a rational approach, which includes molecular modelling studies, three series of azole pyrazole derivatives were designed through two synthetic pathways. The synthesized compounds were biologically evaluated for their inhibitory activity towards M. tuberculosis and their protein binding affinity (K D). Series 3 biarylpyrazole imidazole derivatives were the most effective with the isobutyl (10 f) and tert-butyl (10 g) compounds displaying optimal activity (MIC 1.562 µg/mL, K D 0.22 µM (10 f) and 4.81 µM (10 g)). The spectroscopic data showed that all the synthesised compounds produced a type II red shift of the heme Soret band indicating either direct binding to heme iron or (where less extensive Soret shifts are observed) putative indirect binding via an interstitial water molecule. Evaluation of biological and physicochemical properties identified the following as requirements for activity: LogP >4, H-bond acceptors/H-bond donors 4/0, number of rotatable bonds 5-6, molecular volume >340 Å3, topological polar surface area <40 Å2.

9.
Bioorg Med Chem ; 27(8): 1546-1561, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30837169

ABSTRACT

The rise in multidrug resistant (MDR) cases of tuberculosis (TB) has led to the need for the development of TB drugs with different mechanisms of action. The genome sequence of Mycobacterium tuberculosis (Mtb) revealed twenty different genes coding for cytochrome P450s. CYP121A1 catalyzes a CC crosslinking reaction of dicyclotyrosine (cYY) producing mycocyclosin and current research suggests that either mycocyclosin is essential or the overproduction of cYY is toxic to Mtb. A series of 1,4-dibenzyl-2-imidazol-1-yl-methylpiperazine derivatives were designed and synthesised as cYY mimics. The derivatives substituted in the 4-position of the phenyl rings with halides or alkyl group showed promising antimycobacterial activity (MIC 6.25 µg/mL), with the more lipophilic branched alkyl derivatives displaying optimal binding affinity with CYP121A1 (iPr KD = 1.6 µM; tBu KD = 1.2 µM). Computational studies revealed two possible binding modes within the CYP121A1 active site both of which would effectively block cYY from binding.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Dipeptides/chemistry , Dipeptides/pharmacology , Mycobacterium tuberculosis/enzymology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Antitubercular Agents/chemical synthesis , Cytochrome P-450 Enzyme Inhibitors/chemical synthesis , Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/chemistry , Dipeptides/chemical synthesis , Drug Design , Humans , Molecular Docking Simulation , Mycobacterium tuberculosis/drug effects , Peptides, Cyclic/chemical synthesis , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology , Tuberculosis/drug therapy
10.
Mol Microbiol ; 111(4): 951-964, 2019 04.
Article in English | MEDLINE | ID: mdl-30600561

ABSTRACT

Biosynthesis of the nucleotide sugar precursor dTDP-L-rhamnose is critical for the viability and virulence of many human pathogenic bacteria, including Streptococcus pyogenes (Group A Streptococcus; GAS), Streptococcus mutans and Mycobacterium tuberculosis. Streptococcal pathogens require dTDP-L-rhamnose for the production of structurally similar rhamnose polysaccharides in their cell wall. Via heterologous expression in S. mutans, we confirmed that GAS RmlB and RmlC are critical for dTDP-L-rhamnose biosynthesis through their action as dTDP-glucose-4,6-dehydratase and dTDP-4-keto-6-deoxyglucose-3,5-epimerase enzymes respectively. Complementation with GAS RmlB and RmlC containing specific point mutations corroborated the conservation of previous identified catalytic residues. Bio-layer interferometry was used to identify and confirm inhibitory lead compounds that bind to GAS dTDP-rhamnose biosynthesis enzymes RmlB, RmlC and GacA. One of the identified compounds, Ri03, inhibited growth of GAS, other rhamnose-dependent streptococcal pathogens as well as M. tuberculosis with an IC50 of 120-410 µM. Importantly, we confirmed that Ri03 inhibited dTDP-L-rhamnose formation in a concentration-dependent manner through a biochemical assay with recombinant rhamnose biosynthesis enzymes. We therefore conclude that inhibitors of dTDP-L-rhamnose biosynthesis, such as Ri03, affect streptococcal and mycobacterial viability and can serve as lead compounds for the development of a new class of antibiotics that targets dTDP-rhamnose biosynthesis in pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Hydro-Lyases/metabolism , Nucleoside Diphosphate Sugars/biosynthesis , Racemases and Epimerases/metabolism , Streptococcus/enzymology , Thymine Nucleotides/biosynthesis , Anti-Bacterial Agents/pharmacology , Biosynthetic Pathways , Hydro-Lyases/genetics , Inhibitory Concentration 50 , Racemases and Epimerases/genetics , Streptococcus/drug effects
11.
Microbiology (Reading) ; 164(11): 1457-1470, 2018 11.
Article in English | MEDLINE | ID: mdl-29957170

ABSTRACT

The initial adaptive transcriptional response to nitrogen (N) starvation in Escherichia coli involves large-scale alterations to the transcriptome mediated by the transcriptional activator, NtrC. One of these NtrC-activated genes is yeaG, which encodes a conserved bacterial kinase. Although it is known that YeaG is required for optimal survival under sustained N starvation, the molecular basis by which YeaG benefits N starved E. coli remains elusive. By combining transcriptomics with targeted metabolomics analyses, we demonstrate that the methionine biosynthesis pathway becomes transcriptionally dysregulated in ΔyeaG bacteria experiencing sustained N starvation. It appears the ability of MetJ, the master transcriptional repressor of methionine biosynthesis genes, to effectively repress transcription of genes under its control is compromised in ΔyeaG bacteria under sustained N starvation, resulting in transcriptional derepression of MetJ-regulated genes. Although the aberrant biosynthesis does not appear to be a contributing factor for the compromised viability of ΔyeaG bacteria experiencing sustained N starvation, this study identifies YeaG as a novel regulatory factor in E. coli affecting the transcription of methionine biosynthesis genes under sustained N starvation.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/genetics , Methionine/biosynthesis , Nitrogen/metabolism , Protein Serine-Threonine Kinases/genetics , Transcription, Genetic/genetics , Apoproteins/genetics , Escherichia coli/genetics , Gene Deletion , PII Nitrogen Regulatory Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics
12.
Biochemistry ; 57(24): 3387-3401, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29684272

ABSTRACT

The biosynthetic pathway of peptidoglycan is essential for Mycobacterium tuberculosis. We report here the acetyltransferase substrate specificity and catalytic mechanism of the bifunctional N-acetyltransferase/uridylyltransferase from M. tuberculosis (GlmU). This enzyme is responsible for the final two steps of the synthesis of UDP- N-acetylglucosamine, which is an essential precursor of peptidoglycan, from glucosamine 1-phosphate, acetyl-coenzyme A, and uridine 5'-triphosphate. GlmU utilizes ternary complex formation to transfer an acetyl from acetyl-coenzyme A to glucosamine 1-phosphate to form N-acetylglucosamine 1-phosphate. Steady-state kinetic studies and equilibrium binding experiments indicate that GlmU follows a steady-state ordered kinetic mechanism, with acetyl-coenzyme A binding first, which triggers a conformational change in GlmU, followed by glucosamine 1-phosphate binding. Coenzyme A is the last product to dissociate. Chemistry is partially rate-limiting as indicated by pH-rate studies and solvent kinetic isotope effects. A novel crystal structure of a mimic of the Michaelis complex, with glucose 1-phosphate and acetyl-coenzyme A, helps us to propose the residues involved in deprotonation of glucosamine 1-phosphate and the loop movement that likely generates the active site required for glucosamine 1-phosphate to bind. Together, these results pave the way for the rational discovery of improved inhibitors against M. tuberculosis GlmU, some of which might become candidates for antibiotic discovery programs.


Subject(s)
Bacterial Proteins/metabolism , Biocatalysis , Multienzyme Complexes/metabolism , Uridine Diphosphate N-Acetylglucosamine/biosynthesis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Kinetics , Magnesium Chloride/chemistry , Magnesium Chloride/pharmacology , Molecular Structure , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Mycobacterium tuberculosis/enzymology , Substrate Specificity , Uridine Diphosphate N-Acetylglucosamine/chemistry
13.
Nat Commun ; 8(1): 1939, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29208891

ABSTRACT

D-cycloserine is an antibiotic which targets sequential bacterial cell wall peptidoglycan biosynthesis enzymes: alanine racemase and D-alanine:D-alanine ligase. By a combination of structural, chemical and mechanistic studies here we show that the inhibition of D-alanine:D-alanine ligase by the antibiotic D-cycloserine proceeds via a distinct phosphorylated form of the drug. This mechanistic insight reveals a bimodal mechanism of action for a single antibiotic on different enzyme targets and has significance for the design of future inhibitor molecules based on this chemical structure.


Subject(s)
Antibiotics, Antitubercular/pharmacology , Cycloserine/pharmacology , Peptide Synthases/antagonists & inhibitors , Alanine Racemase , Antibiotics, Antitubercular/metabolism , Cycloserine/metabolism , Escherichia coli , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/drug effects , Peptide Synthases/drug effects , Phosphorylation
14.
J Med Chem ; 60(24): 10257-10267, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29185746

ABSTRACT

Three series of biarylpyrazole imidazole and triazoles are described, which vary in the linker between the biaryl pyrazole and imidazole/triazole group. The imidazole and triazole series with the short -CH2- linker displayed promising antimycobacterial activity, with the imidazole-CH2- series (7) showing low MIC values (6.25-25 µg/mL), which was also influenced by lipophilicity. Extending the linker to -C(O)NH(CH2)2- resulted in a loss of antimycobacterial activity. The binding affinity of the compounds with CYP121A1 was determined by UV-visible optical titrations with KD values of 2.63, 35.6, and 290 µM, respectively, for the tightest binding compounds 7e, 8b, and 13d from their respective series. Both binding affinity assays and docking studies of the CYP121A1 inhibitors suggest type II indirect binding through interstitial water molecules, with key binding residues Thr77, Val78, Val82, Val83, Met86, Ser237, Gln385, and Arg386, comparable with the binding interactions observed with fluconazole and the natural substrate dicyclotyrosine.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/chemistry , Mycobacterium tuberculosis/drug effects , Small Molecule Libraries/chemistry , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Cytochrome P-450 Enzyme Inhibitors/chemical synthesis , Cytochrome P-450 Enzyme System/metabolism , Drug Evaluation, Preclinical/methods , Microbial Sensitivity Tests , Molecular Docking Simulation , Pyrazoles/chemistry , Small Molecule Libraries/pharmacology , Spectrophotometry, Ultraviolet
15.
J Med Chem ; 60(20): 8647-8660, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28968083

ABSTRACT

Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is the infectious disease responsible for the highest number of deaths worldwide. Herein, 22 new N-oxide-containing compounds were synthesized followed by in vitro and in vivo evaluation of their antitubercular potential against Mtb. Compound 8 was found to be the most promising compound, with MIC90 values of 1.10 and 6.62 µM against active and nonreplicating Mtb, respectively. Additionally, we carried out in vivo experiments to confirm the safety and efficacy of compound 8; the compound was found to be orally bioavailable and highly effective, leading to a reduction of Mtb to undetectable levels in a mouse model of infection. Microarray-based initial studies on the mechanism of action suggest that compound 8 blocks translation. Altogether, these results indicate that benzofuroxan derivative 8 is a promising lead compound for the development of a novel chemical class of antitubercular drugs.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacokinetics , Biological Availability , Caco-2 Cells , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/pharmacokinetics , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Oxides/chemistry , Spectrum Analysis/methods
16.
Nat Commun ; 8(1): 203, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28781362

ABSTRACT

ATP-phosphoribosyltransferase (ATP-PRT) is a hexameric enzyme in conformational equilibrium between an open and seemingly active state and a closed and presumably inhibited form. The structure-function relationship of allosteric regulation in this system is still not fully understood. Here, we develop a screening strategy for modulators of ATP-PRT and identify 3-(2-thienyl)-L-alanine (TIH) as an allosteric activator of this enzyme. Kinetic analysis reveals co-occupancy of the allosteric sites by TIH and L-histidine. Crystallographic and native ion-mobility mass spectrometry data show that the TIH-bound activated form of the enzyme closely resembles the inhibited L-histidine-bound closed conformation, revealing the uncoupling between ATP-PRT open and closed conformations and its functional state. These findings suggest that dynamic processes are responsible for ATP-PRT allosteric regulation and that similar mechanisms might also be found in other enzymes bearing a ferredoxin-like allosteric domain.Active and inactive state ATP-phosphoribosyltransferases (ATP-PRTs) are believed to have different conformations. Here the authors show that in both states, ATP-PRT has a similar structural arrangement, suggesting that dynamic alterations are involved in ATP-PRT regulation by allosteric modulators.


Subject(s)
ATP Phosphoribosyltransferase/chemistry , ATP Phosphoribosyltransferase/genetics , ATP Phosphoribosyltransferase/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation , Allosteric Site , Histidine/chemistry , Histidine/metabolism , Kinetics , Models, Molecular
17.
Biochem J ; 474(16): 2897-2899, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798160

ABSTRACT

Regulation of metabolic pathways by signal transduction and transcriptional cascades can alter cellular levels of metabolites. Metabolites themselves can also have regulatory activity as shown in a new study published in the Biochemical Journal Tsuchiya et al. describe a novel antibody and mass spectrometry-based method for identifying proteins that are reversibly modified with Coenzyme A (CoA). Analysis of the 'CoAlated proteome' under conditions of oxidative and metabolic stress revealed a bias towards the modification of metabolic enzymes by CoA. Furthermore, CoAlation was shown to alter the activity of target proteins. These results suggest that CoAlation is a widespread post-translational modification that may have important roles in the metabolic response to stress.


Subject(s)
Coenzyme A/metabolism , Liver/metabolism , Myocardium/metabolism , Protein Processing, Post-Translational , Animals , Antibodies/chemistry , Cysteine/metabolism , HEK293 Cells , Humans , Male , Metabolic Networks and Pathways , Organ Culture Techniques , Oxidation-Reduction , Pyrophosphatases/chemistry , Rats , Rats, Sprague-Dawley , Signal Transduction , Stress, Physiological , Nudix Hydrolases
18.
Antimicrob Agents Chemother ; 60(10): 6091-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27480853

ABSTRACT

The increasing global prevalence of drug resistance among many leading human pathogens necessitates both the development of antibiotics with novel mechanisms of action and a better understanding of the physiological activities of preexisting clinically effective drugs. Inhibition of peptidoglycan (PG) biosynthesis and cross-linking has traditionally enjoyed immense success as an antibiotic target in multiple bacterial pathogens, except in Mycobacterium tuberculosis, where it has so far been underexploited. d-Cycloserine, a clinically approved antituberculosis therapeutic, inhibits enzymes within the d-alanine subbranch of the PG-biosynthetic pathway and has been a focus in our laboratory for understanding peptidoglycan biosynthesis inhibition and for drug development in studies of M. tuberculosis During our studies on alternative inhibitors of the d-alanine pathway, we discovered that the canonical alanine racemase (Alr) inhibitor ß-chloro-d-alanine (BCDA) is a very poor inhibitor of recombinant M. tuberculosis Alr, despite having potent antituberculosis activity. Through a combination of enzymology, microbiology, metabolomics, and proteomics, we show here that BCDA does not inhibit the d-alanine pathway in intact cells, consistent with its poor in vitro activity, and that it is instead a mechanism-based inactivator of glutamate racemase (MurI), an upstream enzyme in the same early stage of PG biosynthesis. This is the first report to our knowledge of inhibition of MurI in M. tuberculosis and thus provides a valuable tool for studying this essential and enigmatic enzyme and a starting point for future MurI-targeted antibacterial development.


Subject(s)
Amino Acid Isomerases/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/chemistry , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , beta-Alanine/analogs & derivatives , Amino Acid Isomerases/antagonists & inhibitors , Amino Acid Isomerases/genetics , Amino Acid Isomerases/metabolism , Amino Acid Sequence , Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cloning, Molecular , Enzyme Inhibitors/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Microbial Sensitivity Tests , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Peptidoglycan/biosynthesis , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity , beta-Alanine/chemistry , beta-Alanine/pharmacology
19.
ACS Infect Dis ; 2(5): 352-360, 2016 May 13.
Article in English | MEDLINE | ID: mdl-27231718

ABSTRACT

The mechanisms that lead to phenotypic antibacterial tolerance in bacteria remain poorly understood. We investigate whether changes in NaCl concentration toward physiologically higher values affect antibacterial efficacy against Mycobacterium tuberculosis (Mtb), the causal agent of human tuberculosis. Indeed, multiclass phenotypic antibacterial tolerance is observed during Mtb growth in physiologic saline. This includes changes in sensitivity to ethionamide, ethambutol, d-cycloserine, several aminoglycosides, and quinolones. By employing organism-wide metabolomic and lipidomic approaches combined with phenotypic tests, we identified a time-dependent biphasic adaptive response after exposure of Mtb to physiological levels of NaCl. A first rapid, extensive, and reversible phase was associated with changes in core and amino acid metabolism. In a second phase, Mtb responded with a substantial remodelling of plasma membrane and outer lipid membrane composition. We demonstrate that phenotypic tolerance at physiological concentrations of NaCl is the result of changes in plasma and outer membrane lipid remodeling and not changes in core metabolism. Altogether, these results indicate that physiologic saline-induced antibacterial tolerance is kinetically coupled to cell envelope changes and demonstrate that metabolic changes and growth arrest are not the cause of phenotypic tolerance observed in Mtb exposed to physiologic concentrations of NaCl. Importantly, this work uncovers a role for bacterial cell envelope remodeling in antibacterial tolerance, alongside well-documented allterations in respiration, metabolism, and growth rate.

20.
J Med Chem ; 59(7): 3272-302, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27002486

ABSTRACT

The essential enzyme CYP121 is a target for drug development against antibiotic resistant strains of Mycobacterium tuberculosis. A triazol-1-yl phenol fragment 1 was identified to bind to CYP121 using a cascade of biophysical assays. Synthetic merging and optimization of 1 produced a 100-fold improvement in binding affinity, yielding lead compound 2 (KD = 15 µM). Deconstruction of 2 into its component retrofragments allowed the group efficiency of structural motifs to be assessed, the identification of more LE scaffolds for optimization and highlighted binding affinity hotspots. Structure-guided addition of a metal-binding pharmacophore onto LE retrofragment scaffolds produced low nanomolar (KD = 15 nM) CYP121 ligands. Elaboration of these compounds to target binding hotspots in the distal active site afforded compounds with excellent selectivity against human drug-metabolizing P450s. Analysis of the factors governing ligand potency and selectivity using X-ray crystallography, UV-vis spectroscopy, and native mass spectrometry provides insight for subsequent drug development.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cytochrome P-450 Enzyme System/chemistry , Drug Design , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Humans , Ligands , Mycobacterium tuberculosis/enzymology , Protein Binding , Protein Structure, Tertiary , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...