Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 66(5): 672-683.e4, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575662

ABSTRACT

The AAA+ Lon protease is conserved from bacteria to humans, performs crucial roles in protein homeostasis, and is implicated in bacterial pathogenesis and human disease. We investigated how Lon selectively degrades specific substrates among a diverse array of potential targets. We report the discovery of HspQ as a new Lon substrate, unique specificity-enhancing factor, and potent allosteric activator. Lon recognizes HspQ via a C-terminal degron, whose precise presentation, in synergy with multipartite contacts with the native core of HspQ, is required for allosteric Lon activation. Productive HspQ-Lon engagement enhances degradation of multiple new and known Lon substrates. Our studies reveal the existence and simultaneous utilization of two distinct substrate recognition sites on Lon, an HspQ binding site and an HspQ-modulated allosteric site. Our investigations unveil an unprecedented regulatory use of an evolutionarily conserved heat shock protein and present a distinctive mechanism for how Lon protease achieves temporally enhanced substrate selectivity.


Subject(s)
Bacterial Proteins/metabolism , Heat-Shock Proteins/metabolism , Protease La/metabolism , Yersinia pestis/enzymology , Allosteric Regulation , Bacterial Proteins/genetics , Binding Sites , Heat-Shock Proteins/genetics , Kinetics , Protease La/genetics , Protein Binding , Protein Folding , Proteolysis , Substrate Specificity , Yersinia pestis/genetics
2.
J Biol Chem ; 298(10): 102454, 2022 10.
Article in English | MEDLINE | ID: mdl-36063993

ABSTRACT

Nonribosomal peptide synthetase heterocyclization (Cy) domains generate biologically important oxazoline/thiazoline groups found in natural products, including pharmaceuticals and virulence factors such as some siderophores. Cy domains catalyze consecutive condensation and cyclodehydration reactions, although the mechanism is unknown. To better understand Cy domain catalysis, here we report the crystal structure of the second Cy domain (Cy2) of yersiniabactin synthetase from the causative agent of the plague, Yersinia pestis. Our high-resolution structure of Cy2 adopts a conformation that enables exploration of interactions with the extended thiazoline-containing cyclodehydration intermediate and the acceptor carrier protein (CP) to which it is tethered. We also report complementary electrostatic interfaces between Cy2 and its donor CP that mediate donor binding. Finally, we explored domain flexibility through normal mode analysis and identified small-molecule fragment-binding sites that may inform future antibiotic design targeting Cy function. Our results suggest how CP binding may influence global Cy conformations, with consequences for active-site remodeling to facilitate the separate condensation and cyclodehydration steps as well as potential inhibitor development.


Subject(s)
Catalytic Domain , Peptide Synthases , Yersinia pestis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Siderophores/metabolism , Yersinia pestis/chemistry , Yersinia pestis/enzymology
3.
Immunity ; 39(6): 1108-20, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24332031

ABSTRACT

The virulence of Salmonella is linked to its invasive capacity and suppression of adaptive immunity. This does not explain, however, the rapid dissemination of the pathogen after it breaches the gut. In our study, S. Typhimurium suppressed degranulation of local mast cells (MCs), resulting in limited neutrophil recruitment and restricting outflow of vascular contents into infection sites, thus facilitating bacterial spread. MC suppression was mediated by secreted effector protein (SptP), which shares structural homology with Yersinia YopH. SptP functioned by dephosphorylating the vesicle fusion protein N-ethylmalemide-sensitive factor and by blocking phosphorylation of Syk. Without SptP, orally challenged S. Typhimurium failed to suppress MC degranulation and exhibited limited colonization of the mesenteric lymph nodes. Administration of SptP to sites of E. coli infection markedly enhanced its virulence. Thus, SptP-mediated inactivation of local MCs is a powerful mechanism utilized by S. Typhimurium to impede early innate immunity.


Subject(s)
Bacterial Proteins/metabolism , Immunity, Innate/immunology , Mast Cells/microbiology , Protein Tyrosine Phosphatases/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/enzymology , Animals , Bacterial Proteins/genetics , Cell Degranulation , Humans , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mutation , Neutrophils/immunology , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Yersinia pestis/enzymology
4.
Anal Chem ; 91(3): 2392-2400, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30580515

ABSTRACT

Molecular recognition between a receptor and ligand is a fundamental event in bioanalytical assays, which guarantees the sensitivity and specificity of an assay for the detection of the target of interest. An intensive understanding of the interaction mechanism could be useful for desirable hapten design, directed antibody evolution in vitro, and assay improvement. To illustrate the structural information on class-specific monoclonal antibodies (mAbs) and dihydropteroate synthase (DHPS) against sulfonamides (SAs) we have previously prepared, we initially measured the kinetic parameters of mAb 4C7, 4D11, and DHPS, which showed that the affinities of 4C7 and 4D11 were in the pM to µM range, while DHPS was uniformly in the µM range. Three-dimensional quantitative structure-activity relationship analysis for 4C7 and 4D11 then revealed that the contributions from the stereochemical structure and electron density of the SAs were comparable to binding with mAb. To acquire insights into the structural basis of mAbs and DHPS during the recognition process, the crystal structures of 4C7 and its complex with sulfathiazole were determined using X-ray crystallography. The results showed the SAs orientation and hydrogen bonding interactions mainly contributed to the diverse SAs-mAb affinities. However, for DHPS, a nucleophilic substitution reaction occurred during the recognition process with the SAs, which contributed to the surprisingly uniform affinity for all the SAs tested. This study verified the previous hypotheses on antibody production against SAs and enhanced our understanding of antibody-SAs interactions, which provided useful information toward the rational design of novel haptens and directed evolution to produce class-specific antibodies as DHPS.


Subject(s)
Anti-Bacterial Agents/metabolism , Antibodies, Monoclonal/metabolism , Dihydropteroate Synthase/metabolism , Sulfonamides/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Bacillus anthracis/enzymology , Binding Sites , Dihydropteroate Synthase/chemistry , Escherichia coli/enzymology , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Substrate Specificity , Sulfonamides/chemistry , Sulfonamides/immunology , Yersinia pestis/enzymology
5.
J Biol Chem ; 292(24): 10002-10013, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28455448

ABSTRACT

Nonribosomal peptide synthesis involves the interplay between covalent protein modifications, conformational fluctuations, catalysis, and transient protein-protein interactions. Delineating the mechanisms involved in orchestrating these various processes will deepen our understanding of domain-domain communication in nonribosomal peptide synthetases (NRPSs) and lay the groundwork for the rational reengineering of NRPSs by swapping domains handling different substrates to generate novel natural products. Although many structural and biochemical studies of NRPSs exist, few studies have focused on the energetics and dynamics governing the interactions in these systems. Here, we present detailed binding studies of an adenylation domain and its partner carrier protein in apo-, holo-, and substrate-loaded forms. Results from fluorescence anisotropy, isothermal titration calorimetry, and NMR titrations indicated that covalent modifications to a carrier protein modulate domain communication, suggesting that chemical modifications to carrier proteins during NRPS synthesis may impart directionality to sequential NRPS domain interactions. Comparison of the structure and dynamics of an apo-aryl carrier protein with those of its modified forms revealed structural fluctuations induced by post-translational modifications and mediated by modulations of protein dynamics. The results provide a comprehensive molecular description of a carrier protein throughout its life cycle and demonstrate how a network of dynamic residues can propagate the molecular impact of chemical modifications throughout a protein and influence its affinity toward partner domains.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Coenzyme A Ligases/metabolism , Models, Molecular , Peptide Synthases/metabolism , Protein Modification, Translational , Protein Processing, Post-Translational , Yersinia pestis/metabolism , Amino Acid Substitution , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Apoproteins/chemistry , Apoproteins/genetics , Apoproteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Calorimetry , Carbon Isotopes , Carrier Proteins/chemistry , Carrier Proteins/genetics , Coenzyme A Ligases/chemistry , Coenzyme A Ligases/genetics , Fluorescence Polarization , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Kinetics , Mutation , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Peptide Synthases/chemistry , Peptide Synthases/genetics , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Titrimetry , Yersinia pestis/enzymology
6.
Anal Biochem ; 542: 63-75, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29180070

ABSTRACT

The rise of antibacterial resistance among human pathogens represents a problem that could change the landscape of healthcare unless new antibiotics are developed. The methyl erythritol phosphate (MEP) pathway represents an attractive series of targets for novel antibiotic design, considering each enzyme of the pathway is both essential and has no human homologs. Here we describe a pilot scale high-throughput screening (HTS) campaign against the first and second committed steps in the pathway, catalyzed by DXP reductoisomerase (IspC) and MEP cytidylyltransferase (IspD), using compounds present in the commercially available LOPAC1280 library as well as in an in-house natural product extract library. Hit compounds were characterized to deduce their mechanism of inhibition; most function through aggregation. The HTS workflow outlined here is useful for quickly screening a chemical library, while effectively identifying false positive compounds associated with assay constraints and aggregation.


Subject(s)
Aldose-Ketose Isomerases/antagonists & inhibitors , Anti-Bacterial Agents/analysis , Enzyme Inhibitors/analysis , High-Throughput Screening Assays , Nucleotidyltransferases/antagonists & inhibitors , Aldose-Ketose Isomerases/metabolism , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Nucleotidyltransferases/metabolism , Recombinant Proteins/metabolism , Yersinia pestis/drug effects , Yersinia pestis/enzymology
7.
Bioorg Chem ; 76: 437-448, 2018 02.
Article in English | MEDLINE | ID: mdl-29275262

ABSTRACT

Development of new antimicrobial agents is a good solution to overcome drug-resistance problems. From this perspective, new quinoxaline derivatives bearing various bioactive heterocyclic moieties (thiadiazoles, oxadiazoles, pyrazoles and thiazoles) were designed and synthesized. The newly synthesized compounds were evaluated for their in vitro antibacterial activity against nine bacterial human pathogenic strains using the disc diffusion assay. In general, most of the synthesized compounds exhibited good antibacterial activities. The thiazolyl 11c displayed significant antibacterial activities against P. aeruginosa (MIC, 12.5 µg/mL vs levofloxacin 12.5 µg/mL). Molecular docking studies indicated that the synthesized compounds could occupy both p-amino benzoic acid (PABA) and pterin binding pockets of the dihydropteroate synthase (DHPS), suggesting that the target compounds could act by the inhibition of bacterial DHPS enzyme. The results provide important information for the future design of more potent antibacterial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dihydropteroate Synthase/antagonists & inhibitors , Drug Design , Quinoxalines/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Catalytic Domain , Dihydropteroate Synthase/chemistry , Dihydropteroate Synthase/metabolism , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Levofloxacin/pharmacology , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Protein Binding , Quinoxalines/chemical synthesis , Quinoxalines/chemistry , Quinoxalines/metabolism , Structure-Activity Relationship , Yersinia pestis/enzymology
8.
PLoS Pathog ; 11(10): e1005222, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26484539

ABSTRACT

Activation and/or recruitment of the host plasmin, a fibrinolytic enzyme also active on extracellular matrix components, is a common invasive strategy of bacterial pathogens. Yersinia pestis, the bubonic plague agent, expresses the multifunctional surface protease Pla, which activates plasmin and inactivates fibrinolysis inhibitors. Pla is encoded by the pPla plasmid. Following intradermal inoculation, Y. pestis has the capacity to multiply in and cause destruction of the lymph node (LN) draining the entry site. The closely related, pPla-negative, Y. pseudotuberculosis species lacks this capacity. We hypothesized that tissue damage and bacterial multiplication occurring in the LN during bubonic plague were linked and both driven by pPla. Using a set of pPla-positive and pPla-negative Y. pestis and Y. pseudotuberculosis strains in a mouse model of intradermal injection, we found that pPla is not required for bacterial translocation to the LN. We also observed that a pPla-cured Y. pestis caused the same extensive histological lesions as the wild type strain. Furthermore, the Y. pseudotuberculosis histological pattern, characterized by infectious foci limited by inflammatory cell infiltrates with normal tissue density and follicular organization, was unchanged after introduction of pPla. However, the presence of pPla enabled Y. pseudotuberculosis to increase its bacterial load up to that of Y. pestis. Similarly, lack of pPla strongly reduced Y. pestis titers in LNs of infected mice. This pPla-mediated enhancing effect on bacterial load was directly dependent on the proteolytic activity of Pla. Immunohistochemistry of Pla-negative Y. pestis-infected LNs revealed extensive bacterial lysis, unlike the numerous, apparently intact, microorganisms seen in wild type Y. pestis-infected preparations. Therefore, our study demonstrates that tissue destruction and bacterial survival/multiplication are dissociated in the bubo and that the primary action of Pla is to protect bacteria from destruction rather than to alter the tissue environment to favor Y. pestis propagation in the host.


Subject(s)
Bacterial Proteins/metabolism , Plague/microbiology , Plague/pathology , Plasminogen Activators/metabolism , Yersinia pestis/pathogenicity , Animals , Disease Models, Animal , Immunohistochemistry , Mice , Mutagenesis, Site-Directed , Plague/enzymology , Virulence/physiology , Virulence Factors/metabolism , Yersinia pestis/enzymology , Yersinia pseudotuberculosis/enzymology , Yersinia pseudotuberculosis/pathogenicity , Yersinia pseudotuberculosis Infections/enzymology , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis Infections/pathology
9.
Appl Environ Microbiol ; 83(17)2017 09 01.
Article in English | MEDLINE | ID: mdl-28646112

ABSTRACT

Clustered regularly interspaced short palindromic repeat (CRISPR)-Cas12a (Cpf1) has emerged as an effective genome editing tool in many organisms. Here, we developed and optimized a CRISPR-Cas12a-assisted recombineering system to facilitate genetic manipulation in bacteria. Using this system, point mutations, deletions, insertions, and gene replacements can be easily generated on the chromosome or native plasmids in Escherichia coli, Yersinia pestis, and Mycobacterium smegmatis Because CRISPR-Cas12a-assisted recombineering does not require introduction of an antibiotic resistance gene into the chromosome to select for recombinants, it is an efficient approach for generating markerless and scarless mutations in bacteria.IMPORTANCE The CRISPR-Cas9 system has been widely used to facilitate genome editing in many bacteria. CRISPR-Cas12a (Cpf1), a new type of CRISPR-Cas system, allows efficient genome editing in bacteria when combined with recombineering. Cas12a and Cas9 recognize different target sites, which allows for more precise selection of the cleavage target and introduction of the desired mutation. In addition, CRISPR-Cas12a-assisted recombineering can be used for genetic manipulation of plasmids and plasmid curing. Finally, Cas12a-assisted recombineering in the generation of point mutations, deletions, insertions, and replacements in bacteria has been systematically analyzed. Taken together, our findings will guide efficient Cas12a-mediated genome editing in bacteria.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Cas Systems , Endonucleases/metabolism , Escherichia coli/genetics , Mycobacterium smegmatis/genetics , Recombination, Genetic , Yersinia pestis/genetics , Bacterial Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/genetics , Escherichia coli/enzymology , Escherichia coli/metabolism , Genetic Engineering , Mutation , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/metabolism , Plasmids/genetics , Plasmids/metabolism , Yersinia pestis/enzymology , Yersinia pestis/metabolism
10.
Org Biomol Chem ; 15(26): 5593-5601, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28639657

ABSTRACT

Dihydropteroate synthase (DHPS) catalyzes the condensation of 6-hydroxymethyl-7,8-dihydropterin pyrophosphate (DHPPP) with p-aminobenzoic acid (pABA) and is a well validated target for anti-malarial and anti-bacterial drugs. However, in recent years its utility as a therapeutic target has diminished considerably due to multiple mutations. As such, considerable structural biology and medicinal chemistry effort has been expended to understand and overcome this issue. To date no detailed computational analysis of the protein mechanism has been made despite the detailed crystal structures and multiple mechanistic proposals being made. In this study the mechanistic proposals for DHPS have been systematically investigated using a hybrid QM/MM method. We aimed to compare the energetics associated with SN1 and SN2 processes, whether the SN1 process involves a carbocation or neutral DHP intermediate, uncover the identity of the general base in the catalytic mechanism, and understand the differences in substrate vs. inhibitor reactivity. Our results suggest a reaction that follows an SN1 process with the rate determining step being C-O bond breaking to give a carbocation intermediate. Comparative studies on the inhibitor STZ confirm the experimental observations that it is also a DHPS substrate.


Subject(s)
Dihydropteroate Synthase/antagonists & inhibitors , Dihydropteroate Synthase/metabolism , Enzyme Inhibitors/pharmacology , Sulfonamides/pharmacology , Biocatalysis , Dihydropteroate Synthase/chemistry , Enzyme Inhibitors/chemistry , Molecular Dynamics Simulation , Quantum Theory , Substrate Specificity , Sulfonamides/chemistry , Yersinia pestis/enzymology
11.
Proc Natl Acad Sci U S A ; 111(52): 18709-14, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25453069

ABSTRACT

The arthropod-borne transmission route of Yersinia pestis, the bacterial agent of plague, is a recent evolutionary adaptation. Yersinia pseudotuberculosis, the closely related food-and water-borne enteric species from which Y. pestis diverged less than 6,400 y ago, exhibits significant oral toxicity to the flea vectors of plague, whereas Y. pestis does not. In this study, we identify the Yersinia urease enzyme as the responsible oral toxin. All Y. pestis strains, including those phylogenetically closest to the Y. pseudotuberculosis progenitor, contain a mutated ureD allele that eliminated urease activity. Restoration of a functional ureD was sufficient to make Y. pestis orally toxic to fleas. Conversely, deletion of the urease operon in Y. pseudotuberculosis rendered it nontoxic. Enzymatic activity was required for toxicity. Because urease-related mortality eliminates 30-40% of infective flea vectors, ureD mutation early in the evolution of Y. pestis was likely subject to strong positive selection because it significantly increased transmission potential.


Subject(s)
Bacterial Proteins , Evolution, Molecular , Gene Silencing , Insect Vectors/microbiology , Urease , Xenopsylla/microbiology , Yersinia pestis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Mutation , Plague/enzymology , Plague/genetics , Plague/pathology , Plague/transmission , Urease/genetics , Urease/metabolism , Yersinia pestis/enzymology , Yersinia pestis/genetics , Yersinia pestis/pathogenicity , Yersinia pseudotuberculosis/enzymology , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/pathogenicity
12.
Biochemistry ; 55(21): 2992-3006, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27136302

ABSTRACT

The enoyl-ACP reductase (ENR) catalyzes the last reaction in the elongation cycle of the bacterial type II fatty acid biosynthesis (FAS-II) pathway. While the FabI ENR is a well-validated drug target in organisms such as Mycobacterium tuberculosis and Staphylococcus aureus, alternate ENR isoforms have been discovered in other pathogens, including the FabV enzyme that is the sole ENR in Yersinia pestis (ypFabV). Previously, we showed that the prototypical ENR inhibitor triclosan was a poor inhibitor of ypFabV and that inhibitors based on the 2-pyridone scaffold were more potent [Hirschbeck, M. (2012) Structure 20 (1), 89-100]. These studies were performed with the T276S FabV variant. In the work presented here, we describe a detailed examination of the mechanism and inhibition of wild-type ypFabV and the T276S variant. The T276S mutation significantly reduces the affinity of diphenyl ether inhibitors for ypFabV (20-fold → 100-fold). In addition, while T276S ypFabV generally displays an affinity for 2-pyridone inhibitors higher than that of the wild-type enzyme, the 4-pyridone scaffold yields compounds with similar affinity for both wild-type and T276S ypFabV. T276 is located at the N-terminus of the helical substrate-binding loop, and structural studies coupled with site-directed mutagenesis reveal that alterations in this residue modulate the size of the active site portal. Subsequently, we were able to probe the mechanism of time-dependent inhibition in this enzyme family by extending the inhibition studies to include P142W ypFabV, a mutation that results in a gain of slow-onset inhibition for the 4-pyridone PT156.


Subject(s)
Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Phenyl Ethers/chemistry , Pyridones/chemistry , Yersinia pestis/enzymology , Catalysis , Catalytic Domain , Crystallization , Crystallography, X-Ray , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutation/genetics , NAD/metabolism , Protein Binding , Protein Conformation
13.
Biochemistry ; 55(7): 1091-9, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26818694

ABSTRACT

The bacterial system for fatty acid biosynthesis (FAS) contains several enzymes whose sequence and structure are highly conserved across a vast array of pathogens. This, coupled with their low homology and difference in organization compared to the equivalent system in humans, makes the FAS pathway an excellent target for antimicrobial drug development. To this end, we have cloned, expressed, and purified the ß-hydroxyacyl-acyl carrier protein dehydratase (FabZ) from both Francisella tularensis (FtFabZ) and Yersinia pestis (YpFabZ). We also solved the crystal structures and performed an enzymatic characterization of both enzymes and several mutant forms of YpFabZ. Additionally, we have discovered two novel inhibitors of FabZ, mangostin and stictic acid, which show similar potencies against both YpFabZ and FtFabZ. Lastly, we selected several compounds from the literature that have been shown to be active against single homologues of FabZ and tested them against both YpFabZ and FtFabZ. These results have revealed clues as to which scaffolds are likely to lead to broad-spectrum antimicrobials targeted against FabZ as well as modifications to existing FabZ inhibitors that may improve potency.


Subject(s)
Bacterial Proteins/chemistry , Francisella tularensis/enzymology , Hydro-Lyases/chemistry , Models, Molecular , Yersinia pestis/enzymology , Amino Acid Substitution , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Dimerization , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacology , Histidine/chemistry , Hydro-Lyases/antagonists & inhibitors , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Molecular Weight , Oxepins/chemistry , Oxepins/pharmacology , Point Mutation , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Xanthones/chemistry , Xanthones/pharmacology
14.
BMC Microbiol ; 16(1): 176, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27492011

ABSTRACT

BACKGROUND: The ability of Yersinia pestis to form a biofilm is an important characteristic in flea transmission of this pathogen. Y. pestis laterally acquired two plasmids (pPCP1and pMT1) and the ability to form biofilms when it evolved from Yersinia pseudotuberculosis. Small regulatory RNAs (sRNAs) are thought to play a crucial role in the processes of biofilm formation and pathogenesis. RESULTS: A pPCP1-derived sRNA HmsA (also known as sR084) was found to contribute to the enhanced biofilm formation phenotype of Y. pestis. The concentration of c-di-GMP was significantly reduced upon deletion of the hmsA gene in Y. pestis. The abundance of mRNA transcripts determining exopolysaccharide production, crucial for biofilm formation, was measured by primer extension, RT-PCR and lacZ transcriptional fusion assays in the wild-type and hmsA mutant strains. HmsA positively regulated biofilm synthesis-associated genes (hmsHFRS, hmsT and hmsCDE), but had no regulatory effect on the biofilm degradation-associated gene hmsP. Interestingly, the recently identified biofilm activator sRNA, HmsB, was rapidly degraded in the hmsA deletion mutant. Two genes (rovM and rovA) functioning as biofilm regulators were also found to be regulated by HmsA, whose regulatory effects were consistent with the HmsA-mediated biofilm phenotype. CONCLUSION: HmsA potentially functions as an activator of biofilm formation in Y. pestis, implying that sRNAs encoded on the laterally acquired plasmids might be involved in the chromosome-based regulatory networks implicated in Y. pestis-specific physiological processes.


Subject(s)
Bacterial Proteins/genetics , Biofilms/growth & development , Yersinia pestis/physiology , Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/genetics , Cyclic GMP/metabolism , Phenotype , Plasmids/genetics , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription Factors/genetics , Yersinia pestis/enzymology , Yersinia pestis/genetics , beta-Galactosidase/metabolism
15.
Bioorg Med Chem Lett ; 26(16): 3950-4, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27423480

ABSTRACT

The sulfonamide class of antibiotics has been in continuous use for over 70years. They are thought to act by directly inhibiting dihydropteroate synthase (DHPS), and also acting as prodrugs that sequester pterin pools by forming dead end pterin-sulfonamide conjugates. In this study, eight pterin-sulfonamide conjugates were synthesized using a novel synthetic strategy and their biochemical and microbiological properties were investigated. The conjugates were shown to competitively inhibit DHPS, and inhibition was enhanced by the presence of pyrophosphate that is crucial to catalysis and is known to promote an ordering of the DHPS active site. The co-crystal structure of Yersinia pestis DHPS bound to one of the more potent conjugates revealed a mode of binding that is similar to that of the enzymatic product analog pteroic acid. The antimicrobial activities of the pterin-sulfonamide conjugates were measured against Escherichia coli in the presence and absence of folate precursors and dependent metabolites. These results show that the conjugates have appreciable antibacterial activity and act by an on target, anti-folate pathway mechanism rather than as simple dead end products.


Subject(s)
Anti-Bacterial Agents/chemistry , Dihydropteroate Synthase/antagonists & inhibitors , Pterins/chemistry , Sulfonamides/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dihydropteroate Synthase/metabolism , Escherichia coli/drug effects , Folic Acid/chemistry , Molecular Docking Simulation , Structure-Activity Relationship , Yersinia pestis/enzymology
16.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 4): 986-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25849407

ABSTRACT

Acyl-CoA thioesterases catalyse the hydrolysis of the thioester bonds present within a wide range of acyl-CoA substrates, releasing free CoASH and the corresponding fatty-acyl conjugate. The TesB-type thioesterases are members of the TE4 thioesterase family, one of 25 thioesterase enzyme families characterized to date, and contain two fused hotdog domains in both prokaryote and eukaryote homologues. Only two structures have been elucidated within this enzyme family, and much of the current understanding of the TesB thioesterases has been based on the Escherichia coli structure. Yersinia pestis, a highly virulent bacterium, encodes only one TesB-type thioesterase in its genome; here, the structural and functional characterization of this enzyme are reported, revealing unique elements both within the protomer and quaternary arrangements of the hotdog domains which have not been reported previously in any thioesterase family. The quaternary structure, confirmed using a range of structural and biophysical techniques including crystallography, small-angle X-ray scattering, analytical ultracentrifugation and size-exclusion chromatography, exhibits a unique octameric arrangement of hotdog domains. Interestingly, the same biological unit appears to be present in both TesB structures solved to date, and is likely to be a conserved and distinguishing feature of TesB-type thioesterases. Analysis of the Y. pestis TesB thioesterase activity revealed a strong preference for octanoyl-CoA and this is supported by structural analysis of the active site. Overall, the results provide novel insights into the structure of TesB thioesterases which are likely to be conserved and distinguishing features of the TE4 thioesterase family.


Subject(s)
Thiolester Hydrolases/chemistry , Yersinia pestis/enzymology , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Multimerization , Protein Structure, Tertiary , Substrate Specificity , Thiolester Hydrolases/metabolism , Yersinia pestis/chemistry , Yersinia pestis/metabolism
17.
Environ Microbiol ; 17(4): 947-59, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25586342

ABSTRACT

The second messenger molecule cyclic diguanylate is essential for Yersinia pestis biofilm formation that is important for blockage-dependent plague transmission from fleas to mammals. Two diguanylate cyclases (DGCs) HmsT and Y3730 (HmsD) are responsible for biofilm formation in vitro and biofilm-dependent blockage in the oriental rat flea Xenopsylla cheopis respectively. Here, we have identified a tripartite signalling system encoded by the y3729-y3731 operon that is responsible for regulation of biofilm formation in different environments. We present genetic evidence that a putative inner membrane-anchored protein with a large periplasmic domain Y3729 (HmsC) inhibits HmsD DGC activity in vitro while an outer membrane Pal-like putative lipoprotein Y3731 (HmsE) counteracts HmsC to activate HmsD in the gut of X. cheopis. We propose that HmsE is a critical element in the transduction of environmental signal(s) required for HmsD-dependent biofilm formation.


Subject(s)
Biofilms/growth & development , Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/genetics , Phosphorus-Oxygen Lyases/genetics , Xenopsylla/microbiology , Yersinia pestis/enzymology , Animals , Base Sequence , Cyclic GMP/biosynthesis , DNA, Bacterial/genetics , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/metabolism , Phosphorus-Oxygen Lyases/biosynthesis , Phosphorus-Oxygen Lyases/metabolism , Plague/microbiology , Plague/transmission , Rats , Sequence Analysis, DNA , Signal Transduction/genetics , Yersinia pestis/metabolism , Yersinia pestis/physiology
18.
Arch Microbiol ; 197(3): 371-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25417006

ABSTRACT

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


Subject(s)
Acetyltransferases/isolation & purification , Acetyltransferases/metabolism , Nucleotidyltransferases/isolation & purification , Nucleotidyltransferases/metabolism , Yersinia pestis/enzymology , Acetyltransferases/chemistry , Acetyltransferases/genetics , Amino Acid Sequence , Enzyme Activation/drug effects , Escherichia coli/genetics , Hydrogen-Ion Concentration , Kinetics , Magnesium/pharmacology , Mercaptoethanol/pharmacology , Molecular Sequence Data , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Oxidants/pharmacology , Oxidation-Reduction , Sequence Alignment , Yersinia pestis/genetics
19.
Proc Natl Acad Sci U S A ; 109(31): 12710-5, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22802624

ABSTRACT

The Gram-negative bacteria Yersinia pestis, causative agent of plague, is extremely virulent. One mechanism contributing to Y. pestis virulence is the presence of a type-three secretion system, which injects effector proteins, Yops, directly into immune cells of the infected host. One of these Yop proteins, YopJ, is proapoptotic and inhibits mammalian NF-κB and MAP-kinase signal transduction pathways. Although the molecular mechanism remained elusive for some time, recent work has shown that YopJ acts as a serine/threonine acetyl-transferase targeting MAP2 kinases. Using Drosophila as a model system, we find that YopJ inhibits one innate immune NF-κB signaling pathway (IMD) but not the other (Toll). In fact, we show YopJ mediated serine/threonine acetylation and inhibition of dTAK1, the critical MAP3 kinase in the IMD pathway. Acetylation of critical serine/threonine residues in the activation loop of Drosophila TAK1 blocks phosphorylation of the protein and subsequent kinase activation. In addition, studies in mammalian cells show similar modification and inhibition of hTAK1. These data present evidence that TAK1 is a target for YopJ-mediated inhibition.


Subject(s)
Bacterial Proteins/metabolism , Immunity, Innate , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Serine O-Acetyltransferase/metabolism , Yersinia pestis/enzymology , Acetylation , Animals , Bacterial Proteins/immunology , Drosophila melanogaster , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Plague/immunology , Plague/metabolism , Serine O-Acetyltransferase/immunology , Yersinia pestis/immunology , Yersinia pestis/pathogenicity
20.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 4): 1074-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24699651

ABSTRACT

Yersinia pestis, the causative agent of bubonic plague, is able to survive in both extracellular and intracellular environments within the human host, although its intracellular survival within macrophages is poorly understood. A novel Y. pestis three-gene rip (required for intracellular proliferation) operon, and in particular ripA, has been shown to be essential for survival and replication in interferon γ-induced macrophages. RipA was previously characterized as a putative butyryl-CoA transferase proposed to yield butyrate, a known anti-inflammatory shown to lower macrophage-produced NO levels. RipA belongs to the family I CoA transferases, which share structural homology, a conserved catalytic glutamate which forms a covalent CoA-thioester intermediate and a flexible loop adjacent to the active site known as the G(V/I)G loop. Here, functional and structural analyses of several RipA mutants are presented in an effort to dissect the CoA transferase mechanism of RipA. In particular, E61V, M31G and F60M RipA mutants show increased butyryl-CoA transferase activities when compared with wild-type RipA. Furthermore, the X-ray crystal structures of E61V, M31G and F60M RipA mutants, when compared with the wild-type RipA structure, reveal important conformational changes orchestrated by a conserved acyl-group binding-pocket phenylalanine, Phe85, and the G(V/I)G loop. Binary structures of M31G RipA and F60M RipA with two distinct CoA substrate conformations are also presented. Taken together, these data provide CoA transferase reaction snapshots of an open apo RipA, a closed glutamyl-anhydride intermediate and an open CoA-thioester intermediate. Furthermore, biochemical analyses support essential roles for both the catalytic glutamate and the flexible G(V/I)G loop along the reaction pathway, although further research is required to fully understand the function of the acyl-group binding pocket in substrate specificity.


Subject(s)
Coenzyme A-Transferases/chemistry , Yersinia pestis/enzymology , Acyl Coenzyme A/metabolism , Catalytic Domain , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/metabolism , Models, Molecular , Mutation , Protein Structure, Quaternary
SELECTION OF CITATIONS
SEARCH DETAIL