Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 765
Filter
1.
Nature ; 593(7857): 125-129, 2021 05.
Article in English | MEDLINE | ID: mdl-33854236

ABSTRACT

Antibiotics that target Gram-negative bacteria in new ways are needed to resolve the antimicrobial resistance crisis1-3. Gram-negative bacteria are protected by an additional outer membrane, rendering proteins on the cell surface attractive drug targets4,5. The natural compound darobactin targets the bacterial insertase BamA6-the central unit of the essential BAM complex, which facilitates the folding and insertion of outer membrane proteins7-13. BamA lacks a typical catalytic centre, and it is not obvious how a small molecule such as darobactin might inhibit its function. Here we resolve the mode of action of darobactin at the atomic level using a combination of cryo-electron microscopy, X-ray crystallography, native mass spectrometry, in vivo experiments and molecular dynamics simulations. Two cyclizations pre-organize the darobactin peptide in a rigid ß-strand conformation. This creates a mimic of the recognition signal of native substrates with a superior ability to bind to the lateral gate of BamA. Upon binding, darobactin replaces a lipid molecule from the lateral gate to use the membrane environment as an extended binding pocket. Because the interaction between darobactin and BamA is largely mediated by backbone contacts, it is particularly robust against potential resistance mutations. Our results identify the lateral gate as a functional hotspot in BamA and will allow the rational design of antibiotics that target this bacterial Achilles heel.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/drug effects , Escherichia coli/enzymology , Phenylpropionates/chemistry , Phenylpropionates/pharmacology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , Drug Design , Escherichia coli/cytology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Mass Spectrometry , Molecular Dynamics Simulation , Protein Structure, Secondary
2.
Nature ; 589(7843): 597-602, 2021 01.
Article in English | MEDLINE | ID: mdl-33361818

ABSTRACT

Isoprenoids are vital for all organisms, in which they maintain membrane stability and support core functions such as respiration1. IspH, an enzyme in the methyl erythritol phosphate pathway of isoprenoid synthesis, is essential for Gram-negative bacteria, mycobacteria and apicomplexans2,3. Its substrate, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP), is not produced in metazoans, and in humans and other primates it activates cytotoxic Vγ9Vδ2 T cells at extremely low concentrations4-6. Here we describe a class of IspH inhibitors and refine their potency to nanomolar levels through structure-guided analogue design. After modification of these compounds into prodrugs for delivery into bacteria, we show that they kill clinical isolates of several multidrug-resistant bacteria-including those from the genera Acinetobacter, Pseudomonas, Klebsiella, Enterobacter, Vibrio, Shigella, Salmonella, Yersinia, Mycobacterium and Bacillus-yet are relatively non-toxic to mammalian cells. Proteomic analysis reveals that bacteria treated with these prodrugs resemble those after conditional IspH knockdown. Notably, these prodrugs also induce the expansion and activation of human Vγ9Vδ2 T cells in a humanized mouse model of bacterial infection. The prodrugs we describe here synergize the direct killing of bacteria with a simultaneous rapid immune response by cytotoxic γδ T cells, which may limit the increase of antibiotic-resistant bacterial populations.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/immunology , Lymphocyte Activation/drug effects , Microbial Viability/drug effects , Oxidoreductases/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/drug effects , Animals , Drug Resistance, Microbial , Drug Resistance, Multiple , Enzyme Inhibitors/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Half-Life , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Sensitivity Tests , Molecular Docking Simulation , Oxidoreductases/deficiency , Oxidoreductases/genetics , Oxidoreductases/metabolism , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Substrate Specificity , Swine/blood , T-Lymphocytes, Cytotoxic/immunology
3.
Genes Dev ; 33(11-12): 718-732, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30975721

ABSTRACT

The stationary phase promoter specificity subunit σS (RpoS) is delivered to the ClpXP machinery for degradation dependent on the adaptor RssB. This adaptor-specific degradation of σS provides a major point for regulation and transcriptional reprogramming during the general stress response. RssB is an atypical response regulator and the only known ClpXP adaptor that is inhibited by multiple but dissimilar antiadaptors (IraD, IraP, and IraM). These are induced by distinct stress signals and bind to RssB in poorly understood manners to achieve stress-specific inhibition of σS turnover. Here we present the first crystal structure of RssB bound to an antiadaptor, the DNA damage-inducible IraD. The structure reveals that RssB adopts a compact closed architecture with extensive interactions between its N-terminal and C-terminal domains. The structural data, together with mechanistic studies, suggest that RssB plasticity, conferred by an interdomain glutamate-rich flexible linker, is critical for regulation of σS degradation. Structural modulation of interdomain linkers may thus constitute a general strategy for tuning response regulators.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins/chemistry , Escherichia coli Proteins/chemistry , Sigma Factor/chemistry , Sigma Factor/metabolism , Transcription Factors/chemistry , Bacterial Proteins/chemistry , Crystallography, X-Ray , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Models, Molecular , Protein Conformation , Protein Conformation, alpha-Helical , Protein Domains , Protein Stability , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
4.
Nat Chem Biol ; 20(7): 867-876, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38238495

ABSTRACT

The bacterial ribosome is an essential drug target as many clinically important antibiotics bind and inhibit its functional centers. The catalytic peptidyl transferase center (PTC) is targeted by the broadest array of inhibitors belonging to several chemical classes. One of the most abundant and clinically prevalent resistance mechanisms to PTC-acting drugs in Gram-positive bacteria is C8-methylation of the universally conserved A2503 nucleobase by Cfr methylase in 23S ribosomal RNA. Despite its clinical importance, a sufficient understanding of the molecular mechanisms underlying Cfr-mediated resistance is currently lacking. Here, we report a set of high-resolution structures of the Cfr-modified 70S ribosome containing aminoacyl- and peptidyl-transfer RNAs. These structures reveal an allosteric rearrangement of nucleotide A2062 upon Cfr-mediated methylation of A2503 that likely contributes to the reduced potency of some PTC inhibitors. Additionally, we provide the structural bases behind two distinct mechanisms of engaging the Cfr-methylated ribosome by the antibiotics iboxamycin and tylosin.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Drug Resistance, Bacterial/drug effects , Ribosomes/metabolism , Ribosomes/drug effects , Ribosomes/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , RNA, Ribosomal, 23S/chemistry , RNA, Ribosomal, 23S/metabolism , Methyltransferases/metabolism , Methyltransferases/chemistry , Methyltransferases/antagonists & inhibitors , Methylation , Models, Molecular , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli/genetics
5.
Nature ; 576(7787): 459-464, 2019 12.
Article in English | MEDLINE | ID: mdl-31747680

ABSTRACT

The current need for novel antibiotics is especially acute for drug-resistant Gram-negative pathogens1,2. These microorganisms have a highly restrictive permeability barrier, which limits the penetration of most compounds3,4. As a result, the last class of antibiotics that acted against Gram-negative bacteria was developed in the 1960s2. We reason that useful compounds can be found in bacteria that share similar requirements for antibiotics with humans, and focus on Photorhabdus symbionts of entomopathogenic nematode microbiomes. Here we report a new antibiotic that we name darobactin, which was obtained using a screen of Photorhabdus isolates. Darobactin is coded by a silent operon with little production under laboratory conditions, and is ribosomally synthesized. Darobactin has an unusual structure with two fused rings that form post-translationally. The compound is active against important Gram-negative pathogens both in vitro and in animal models of infection. Mutants that are resistant to darobactin map to BamA, an essential chaperone and translocator that folds outer membrane proteins. Our study suggests that bacterial symbionts of animals contain antibiotics that are particularly suitable for development into therapeutics.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/pathogenicity , Phenylpropionates/isolation & purification , Phenylpropionates/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Line , Disease Models, Animal , Drug Discovery , Drug Resistance, Microbial/drug effects , Drug Resistance, Microbial/genetics , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Gastrointestinal Microbiome/drug effects , Gram-Negative Bacteria/genetics , Humans , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Mutation , Nematoda/microbiology , Operon/genetics , Photorhabdus/chemistry , Photorhabdus/genetics , Photorhabdus/isolation & purification , Substrate Specificity , Symbiosis
6.
Proc Natl Acad Sci U S A ; 119(11): e2119417119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35263219

ABSTRACT

Colistin is considered the last-line antimicrobial for the treatment of multidrug-resistant gram-negative bacterial infections. The emergence and spread of superbugs carrying the mobile colistin resistance gene (mcr) have become the most serious and urgent threat to healthcare. Here, we discover that silver (Ag+), including silver nanoparticles, could restore colistin efficacy against mcr-positive bacteria. We show that Ag+ inhibits the activity of the MCR-1 enzyme via substitution of Zn2+ in the active site. Unexpectedly, a tetra-silver center was found in the active-site pocket of MCR-1 as revealed by the X-ray structure of the Ag-bound MCR-1, resulting in the prevention of substrate binding. Moreover, Ag+effectively slows down the development of higher-level resistance and reduces mutation frequency. Importantly, the combined use of Ag+ at a low concentration with colistin could relieve dermonecrotic lesions and reduce the bacterial load of mice infected with mcr-1­carrying pathogens. This study depicts a mechanism of Ag+ inhibition of MCR enzymes and demonstrates the potentials of Ag+ as broad-spectrum inhibitors for the treatment of mcr-positive bacterial infection in combination with colistin.


Subject(s)
Anti-Bacterial Agents , Colistin , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins , Escherichia coli , Silver , Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Microbial Sensitivity Tests , Plasmids/genetics , Silver/pharmacology
7.
J Infect Dis ; 230(1): e149-e158, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052707

ABSTRACT

BACKGROUND: Cytochrome bd complexes are respiratory oxidases found exclusively in prokaryotes that are important during infection for numerous bacterial pathogens. METHODS: In silico docking was employed to screen approved drugs for their ability to bind to the quinol site of Escherichia coli cytochrome bd-I. Respiratory inhibition was assessed with oxygen electrodes using membranes isolated from E. coli and methicillin-resistant Staphylococcus aureus strains expressing single respiratory oxidases (ie, cytochromes bd, bo', or aa3). Growth/viability assays were used to measure bacteriostatic and bactericidal effects. RESULTS: The steroid drugs ethinylestradiol and quinestrol inhibited E. coli bd-I activity with median inhibitory concentration (IC50) values of 47 ± 28.9 µg/mL (158 ± 97.2 µM) and 0.2 ± 0.04 µg/mL (0.5 ± 0.1 µM), respectively. Quinestrol inhibited growth of an E. coli "bd-I only" strain with an IC50 of 0.06 ± 0.02 µg/mL (0.2 ± 0.07 µM). Growth of an S. aureus "bd only" strain was inhibited by quinestrol with an IC50 of 2.2 ± 0.43 µg/mL (6.0 ± 1.2 µM). Quinestrol exhibited potent bactericidal effects against S. aureus but not E. coli. CONCLUSIONS: Quinestrol inhibits cytochrome bd in E. coli and S. aureus membranes and inhibits the growth of both species, yet is only bactericidal toward S. aureus.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Methicillin-Resistant Staphylococcus aureus , Methicillin-Resistant Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Escherichia coli/enzymology , Anti-Bacterial Agents/pharmacology , Molecular Docking Simulation , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Microbial Sensitivity Tests , Steroids/pharmacology , Steroids/chemistry , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , Cytochrome b Group , Cytochromes/antagonists & inhibitors , Cytochromes/metabolism
8.
J Bacteriol ; 206(7): e0011224, 2024 07 25.
Article in English | MEDLINE | ID: mdl-38856220

ABSTRACT

Urinary tract infections (UTIs) are a major global health problem and are caused predominantly by uropathogenic Escherichia coli (UPEC). UTIs are a leading cause of prescription antimicrobial use. Incessant increase in antimicrobial resistance in UPEC and other uropathogens poses a serious threat to the current treatment practices. Copper is an effector of nutritional immunity that impedes the growth of pathogens during infection. We hypothesized that copper would augment the toxicity of select small molecules against bacterial pathogens. We conducted a small molecule screening campaign with a library of 51,098 molecules to detect hits that inhibit a UPEC ΔtolC mutant in a copper-dependent manner. A molecule, denoted as E. coli inhibitor or ECIN, was identified as a copper-responsive inhibitor of wild-type UPEC strains. Our gene expression and metal content analysis results demonstrate that ECIN works in concert with copper to exacerbate Cu toxicity in UPEC. ECIN has a broad spectrum of activity against pathogens of medical and veterinary significance including Acinetobacter baumannii, Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus. Subinhibitory levels of ECIN eliminate UPEC biofilm formation. Transcriptome analysis of UPEC treated with ECIN reveals induction of multiple stress response systems. Furthermore, we demonstrate that L-cysteine rescues the growth of UPEC exposed to ECIN. In summary, we report the identification and characterization of a novel copper-responsive small molecule inhibitor of UPEC.IMPORTANCEUrinary tract infection (UTI) is a ubiquitous infectious condition affecting millions of people annually. Uropathogenic Escherichia coli (UPEC) is the predominant etiological agent of UTI. However, UTIs are becoming increasingly difficult to resolve with antimicrobials due to increased antimicrobial resistance in UPEC and other uropathogens. Here, we report the identification and characterization of a novel copper-responsive small molecule inhibitor of UPEC. In addition to E. coli, this small molecule also inhibits pathogens of medical and veterinary significance including Acinetobacter baumannii, Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus.


Subject(s)
Anti-Bacterial Agents , Copper , Microbial Sensitivity Tests , Urinary Tract Infections , Uropathogenic Escherichia coli , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/genetics , Copper/pharmacology , Anti-Bacterial Agents/pharmacology , Urinary Tract Infections/microbiology , Urinary Tract Infections/drug therapy , Small Molecule Libraries/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Gene Expression Regulation, Bacterial/drug effects , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/drug therapy
9.
J Am Chem Soc ; 146(30): 20845-20856, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39041457

ABSTRACT

We recently reported on small-molecule inhibitors of the GroES/GroEL chaperone system as potential antibiotics against Escherichia coli and the ESKAPE pathogens but were unable to establish GroES/GroEL as the cellular target, leading to cell death. In this study, using two of our most potent bis-sulfonamido-2-phenylbenzoxazoles (PBZs), we established the binding site of the PBZ molecules using cryo-EM and found that GroEL was the cellular target responsible for the mode of action. Cryo-EM revealed that PBZ1587 binds at the GroEL ring-ring interface (RRI). A cellular reporter assay confirmed that PBZ1587 engaged GroEL in cells, but cellular rescue experiments showed potential off-target effects. This prompted us to explore a closely related analogue, PBZ1038, which is also bound to the RRI. Biochemical characterization showed potent inhibition of Gram-negative chaperonins but much lower potency of chaperonin from a Gram-positive organism, Enterococcus faecium. A cellular reporter assay showed that PBZ1038 also engaged GroEL in cells and that the cytotoxic phenotype could be rescued by a chromosomal copy of E. faecium GroEL/GroES or by expressing a recalcitrant RRI mutant. These data argue that PBZ1038's antimicrobial action is exerted through inhibition of GroES/GroEL, validating this chaperone system as an antibiotic target.


Subject(s)
Anti-Bacterial Agents , Chaperonin 10 , Escherichia coli , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Chaperonin 10/metabolism , Chaperonin 10/antagonists & inhibitors , Chaperonin 10/chemistry , Escherichia coli/drug effects , Chaperonin 60/metabolism , Chaperonin 60/antagonists & inhibitors , Chaperonin 60/chemistry , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Benzoxazoles/chemical synthesis , Microbial Sensitivity Tests , Molecular Structure , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry
10.
J Chem Inf Model ; 64(15): 5964-5976, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39011748

ABSTRACT

AcrB, a key component in bacterial efflux processes, exhibits distinct binding pockets that influence inhibitor interactions. In addition to the well-known distal binding pocket within the periplasmic domain, a noteworthy pocket amidst the transmembrane (TM) helices serves as an alternate binding site for inhibitors. The bacterial efflux mechanism involves a pivotal functional rotation of the TM protein, inducing conformational changes in each protomer and propelling drugs toward the outer membrane domain. Surprisingly, inhibitors binding to the TM domain display a preference for L protomers over T protomers. Metadynamics simulations elucidate that Lys940 in the TM domain of AcrB can adopt two conformations in L protomers, whereas the energy barrier for such transitions is higher in T protomers. This phenomenon results in stable inhibitor binding in l protomers. Upon a detailed analysis of unbinding pathways using random accelerated molecular dynamics and umbrella sampling, we have identified three distinct routes for ligand exit from the allosteric site, specifically involving regions within the TM domains─TM4, TM5, and TM10. To explore allosteric crosstalk, we focused on the following key residues: Val452 from the TM domain and Ala831 from the porter domain. Surprisingly, our findings reveal that inhibitor binding disrupts this communication. The shortest path connecting Val452 and Ala831 increases upon inhibitor binding, suggesting sabotage of the natural interdomain communication dynamics. This result highlights the intricate interplay between inhibitor binding and allosteric signaling within our studied system.


Subject(s)
Escherichia coli Proteins , Molecular Dynamics Simulation , Multidrug Resistance-Associated Proteins , Allosteric Regulation , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/metabolism , Binding Sites , Ligands , Allosteric Site , Protein Conformation , Protein Binding , Protein Multimerization
11.
Mol Cell ; 61(3): 329-340, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26805573

ABSTRACT

Intramembrane proteases signal by releasing proteins from the membrane, but despite their importance, their enzymatic mechanisms remain obscure. We probed rhomboid proteases with reversible, mechanism-based inhibitors that allow precise kinetic analysis and faithfully mimic the transition state structurally. Unexpectedly, inhibition by peptide aldehydes is non-competitive, revealing that in the Michaelis complex, substrate does not contact the catalytic center. Structural analysis in a membrane revealed that all extracellular loops of rhomboid make stabilizing interactions with substrate, but mainly through backbone interactions, explaining rhomboid's broad sequence selectivity. At the catalytic site, the tetrahedral intermediate lies covalently attached to the catalytic serine alone, with the oxyanion stabilized by unusual tripartite interactions with the side chains of H150, N154, and the backbone of S201. We also visualized unexpected substrate-enzyme interactions at the non-essential P2/P3 residues. These "extra" interactions foster potent rhomboid inhibition in living cells, thereby opening avenues for rational design of selective rhomboid inhibitors.


Subject(s)
Aldehydes/pharmacology , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , DNA-Binding Proteins/antagonists & inhibitors , Drug Design , Escherichia coli Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Peptides/pharmacology , Protease Inhibitors/pharmacology , Aldehydes/chemistry , Aldehydes/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Catalysis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/metabolism , Enzyme Stability , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Kinetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity
12.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125622

ABSTRACT

Bacteria are known to be constantly adapting to become resistant to antibiotics. Currently, efficient antibacterial compounds are still available; however, it is only a matter of time until these compounds also become inefficient. Ribonucleases are the enzymes responsible for the maturation and degradation of RNA molecules, and many of them are essential for microbial survival. Members of the PNPase and RNase II families of exoribonucleases have been implicated in virulence in many pathogens and, as such, are valid targets for the development of new antibacterials. In this paper, we describe the use of virtual high-throughput screening (vHTS) to identify chemical compounds predicted to bind to the active sites within the known structures of RNase II and PNPase from Escherichia coli. The subsequent in vitro screening identified compounds that inhibited the activity of these exoribonucleases, with some also affecting cell viability, thereby providing proof of principle for utilizing the known structures of these enzymes in the pursuit of new antibacterials.


Subject(s)
Anti-Bacterial Agents , Enzyme Inhibitors , Escherichia coli , Exoribonucleases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Exoribonucleases/antagonists & inhibitors , Exoribonucleases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Escherichia coli/drug effects , Escherichia coli/enzymology , Catalytic Domain , High-Throughput Screening Assays/methods , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Bacteria/drug effects , Bacteria/enzymology
13.
Nat Methods ; 17(1): 86-92, 2020 01.
Article in English | MEDLINE | ID: mdl-31740817

ABSTRACT

Our ability to connect genotypic variation to biologically important phenotypes has been seriously limited by the gap between live-cell microscopy and library-scale genomic engineering. Here, we show how in situ genotyping of a library of strains after time-lapse imaging in a microfluidic device overcomes this problem. We determine how 235 different CRISPR interference knockdowns impact the coordination of the replication and division cycles of Escherichia coli by monitoring the location of replication forks throughout on average >500 cell cycles per knockdown. Subsequent in situ genotyping allows us to map each phenotype distribution to a specific genetic perturbation to determine which genes are important for cell cycle control. The single-cell time-resolved assay allows us to determine the distribution of single-cell growth rates, cell division sizes and replication initiation volumes. The technology presented in this study enables genome-scale screens of most live-cell microscopy assays.


Subject(s)
CRISPR-Cas Systems , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Image Processing, Computer-Assisted/methods , Metabolic Engineering/methods , Microfluidic Analytical Techniques/methods , Microscopy/methods , Cell Cycle , DNA Replication , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Library , Genotype , Phenotype
14.
Proc Natl Acad Sci U S A ; 117(43): 26608-26615, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33046652

ABSTRACT

Stochastic pulsatile dynamics have been observed in an increasing number of biological circuits with known mechanism involving feedback control and bistability. Surprisingly, recent single-cell experiments in Escherichia coli flagellar synthesis showed that flagellar genes are activated in stochastic pulses without the means of feedback. However, the mechanism for pulse generation in these feedbackless circuits has remained unclear. Here, by developing a system-level stochastic model constrained by a large set of single-cell E. coli flagellar synthesis data from different strains and mutants, we identify the general underlying design principles for generating stochastic transcriptional pulses without feedback. Our study shows that an inhibitor (YdiV) of the transcription factor (FlhDC) creates a monotonic ultrasensitive switch that serves as a digital filter to eliminate small-amplitude FlhDC fluctuations. Furthermore, we find that the high-frequency (fast) fluctuations of FlhDC are filtered out by integration over a timescale longer than the timescale of the input fluctuations. Together, our results reveal a filter-and-integrate design for generating stochastic pulses without feedback. This filter-and-integrate mechanism enables a general strategy for cells to avoid premature activation of the expensive downstream gene expression by filtering input fluctuations in both intensity and time so that the system only responds to input signals that are both strong and persistent.


Subject(s)
Gene Expression Regulation, Bacterial , Models, Biological , Stochastic Processes , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/physiology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Time Factors , Trans-Activators/metabolism
15.
Proc Natl Acad Sci U S A ; 117(10): 5260-5268, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32094196

ABSTRACT

A critical problem in the fight against bacterial infection is the rising rates of resistance and the lack of new antibiotics. The discovery of new targets or new antibacterial mechanisms is a potential solution but is becoming more difficult. Here we report an antibacterial mechanism that safeguards intestine cells from enteropathogenic Escherichia coli (EPEC) by shutting down an infection-responsive signal of the host intestine cell. A key step in EPEC infection of intestinal cells involves Tir-induced actin reorganization. Nck mediates this event by binding with Tir through its SH2 domain (Nck-SH2) and with WIP through its second SH3 domain (Nck-SH3.2). Here we report the design of a synthetic peptide that reacts precisely with a unique cysteine of the Nck-SH3.2 domain, blocks the binding site of the Nck protein, and prevents EPEC infection of Caco-2 cells. Oral update of this nontoxic peptide before EPEC administration safeguards mice from EPEC infection and diarrhea. This study demonstrates domain-specific blockage of an SH3 domain of a multidomain adaptor protein inside cells and the inhibition of Tir-induced rearrangement of the host actin cytoskeleton as a previously unknown antibacterial mechanism.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antimicrobial Cationic Peptides/pharmacology , Enteropathogenic Escherichia coli/drug effects , Escherichia coli Infections/prevention & control , Escherichia coli Proteins/antagonists & inhibitors , Host-Pathogen Interactions/drug effects , Oncogene Proteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Actins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antimicrobial Cationic Peptides/therapeutic use , Caco-2 Cells , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Protein Binding , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Signal Transduction , src Homology Domains
16.
J Biol Chem ; 296: 100653, 2021.
Article in English | MEDLINE | ID: mdl-33845047

ABSTRACT

The transcription terminator Rho regulates many physiological processes in bacteria, such as antibiotic sensitivity, DNA repair, RNA remodeling, and so forth, and hence, is a potential antimicrobial target, which is unexplored. The bacteriophage P4 capsid protein, Psu, moonlights as a natural Rho antagonist. Here, we report the design of novel peptides based on the C-terminal region of Psu using phenotypic screening methods. The resultant 38-mer peptides, in addition to containing mutagenized Psu sequences, also contained plasmid sequences, fused to their C termini. Expression of these peptides inhibited the growth of Escherichia coli and specifically inhibited Rho-dependent termination in vivo. Peptides 16 and 33 exhibited the best Rho-inhibitory properties in vivo. Direct high-affinity binding of these two peptides to Rho also inhibited the latter's RNA-dependent ATPase and transcription termination functions in vitro. These two peptides remained functional even if eight to ten amino acids were deleted from their C termini. In silico modeling and genetic and biochemical evidence revealed that these two peptides bind to the primary RNA-binding site of the Rho hexamer near its subunit interfaces. In addition, the gene expression profiles of these peptides and Psu overlapped significantly. These peptides also inhibited the growth of Mycobacteria and inhibited the activities of Rho proteins from Mycobacterium tuberculosis, Xanthomonas, Vibrio cholerae, and Salmonella enterica. Our results showed that these novel anti-Rho peptides mimic the Rho-inhibition function of the ∼42-kDa dimeric bacteriophage P4 capsid protein, Psu. We conclude that these peptides and their C-terminal deletion derivatives could provide a basis on which to design novel antimicrobial peptides.


Subject(s)
Capsid Proteins/pharmacology , Drug Design , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/metabolism , Mycobacterium tuberculosis/drug effects , Peptide Fragments/pharmacology , Terminator Regions, Genetic , Xanthomonas/drug effects , Amino Acid Sequence , Escherichia coli/genetics , Escherichia coli/growth & development , Mycobacterium tuberculosis/growth & development , Peptide Library , Plasmids , Protein Binding , Sequence Homology , Xanthomonas/growth & development
17.
EMBO J ; 37(3): 413-426, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29237698

ABSTRACT

To maintain genome integrity, organisms employ DNA damage response, the underlying principles of which are conserved from bacteria to humans. The bacterial small RNA OxyS of Escherichia coli is induced upon oxidative stress and has been implicated in protecting cells from DNA damage; however, the mechanism by which OxyS confers genome stability remained unknown. Here, we revealed an OxyS-induced molecular checkpoint relay, leading to temporary cell cycle arrest to allow damage repair. By repressing the expression of the essential transcription termination factor nusG, OxyS enables read-through transcription into a cryptic prophage encoding kilR The KilR protein interferes with the function of the major cell division protein FtsZ, thus imposing growth arrest. This transient growth inhibition facilitates DNA damage repair, enabling cellular recovery, thereby increasing viability following stress. The OxyS-mediated growth arrest represents a novel tier of defense, introducing a new regulatory concept into bacterial stress response.


Subject(s)
Cell Cycle Checkpoints/genetics , DNA Damage/genetics , DNA Repair/genetics , Escherichia coli Proteins/genetics , Escherichia coli/growth & development , Escherichia coli/genetics , Repressor Proteins/genetics , Bacterial Proteins/genetics , Cell Division/genetics , Cytoskeletal Proteins/genetics , Escherichia coli Proteins/antagonists & inhibitors , Genomic Instability/genetics , Oxidative Stress/physiology , Peptide Elongation Factors/antagonists & inhibitors , Peptide Elongation Factors/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription, Genetic/genetics
18.
Nature ; 533(7604): 557-560, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27088606

ABSTRACT

Antibiotic-resistant bacterial infection is a serious threat to public health. Peptidoglycan biosynthesis is a well-established target for antibiotic development. MraY (phospho-MurNAc-pentapeptide translocase) catalyses the first and an essential membrane step of peptidoglycan biosynthesis. It is considered a very promising target for the development of new antibiotics, as many naturally occurring nucleoside inhibitors with antibacterial activity target this enzyme. However, antibiotics targeting MraY have not been developed for clinical use, mainly owing to a lack of structural insight into inhibition of this enzyme. Here we present the crystal structure of MraY from Aquifex aeolicus (MraYAA) in complex with its naturally occurring inhibitor, muraymycin D2 (MD2). We show that after binding MD2, MraYAA undergoes remarkably large conformational rearrangements near the active site, which lead to the formation of a nucleoside-binding pocket and a peptide-binding site. MD2 binds the nucleoside-binding pocket like a two-pronged plug inserting into a socket. Further interactions it makes in the adjacent peptide-binding site anchor MD2 to and enhance its affinity for MraYAA. Surprisingly, MD2 does not interact with three acidic residues or the Mg(2+) cofactor required for catalysis, suggesting that MD2 binds to MraYAA in a manner that overlaps with, but is distinct from, its natural substrate, UDP-MurNAc-pentapeptide. We have determined the principles of MD2 binding to MraYAA, including how it avoids the need for pyrophosphate and sugar moieties, which are essential features for substrate binding. The conformational plasticity of MraY could be the reason that it is the target of many structurally distinct inhibitors. These findings can inform the design of new inhibitors targeting MraY as well as its paralogues, WecA and TarO.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Cell Wall/metabolism , Monosaccharides/biosynthesis , Nucleosides/pharmacology , Oligopeptides/biosynthesis , Peptides/pharmacology , Transferases/antagonists & inhibitors , Transferases/chemistry , Anti-Bacterial Agents/chemistry , Bacteria/enzymology , Bacterial Proteins/metabolism , Catalytic Domain/drug effects , Cell Wall/chemistry , Cell Wall/drug effects , Conserved Sequence , Crystallography, X-Ray , Drug Design , Escherichia coli Proteins/antagonists & inhibitors , Magnesium/metabolism , Models, Molecular , Nucleosides/chemistry , Peptides/chemistry , Peptidoglycan/biosynthesis , Protein Binding , Protein Conformation/drug effects , Structure-Activity Relationship , Transferases/metabolism , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
19.
Nature ; 534(7606): 277-280, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27279228

ABSTRACT

In order to survive, bacteria continually sense, and respond to, environmental fluctuations. Stringent control represents a key bacterial stress response to nutrient starvation that leads to rapid and comprehensive reprogramming of metabolic and transcriptional patterns. In general, transcription of genes for growth and proliferation is downregulated, while those important for survival and virulence are upregulated. Amino acid starvation is sensed by depletion of the aminoacylated tRNA pools, and this results in accumulation of ribosomes stalled with non-aminoacylated (uncharged) tRNA in the ribosomal A site. RelA is recruited to stalled ribosomes and activated to synthesize a hyperphosphorylated guanosine analogue, (p)ppGpp, which acts as a pleiotropic secondary messenger. However, structural information about how RelA recognizes stalled ribosomes and discriminates against aminoacylated tRNAs is missing. Here we present the cryo-electron microscopy structure of RelA bound to the bacterial ribosome stalled with uncharged tRNA. The structure reveals that RelA utilizes a distinct binding site compared to the translational factors, with a multi-domain architecture that wraps around a highly distorted A-site tRNA. The TGS (ThrRS, GTPase and SpoT) domain of RelA binds the CCA tail to orient the free 3' hydroxyl group of the terminal adenosine towards a ß-strand, such that an aminoacylated tRNA at this position would be sterically precluded. The structure supports a model in which association of RelA with the ribosome suppresses auto-inhibition to activate synthesis of (p)ppGpp and initiate the stringent response. Since stringent control is responsible for the survival of pathogenic bacteria under stress conditions, and contributes to chronic infections and antibiotic tolerance, RelA represents a good target for the development of novel antibacterial therapeutics.


Subject(s)
Amino Acids/deficiency , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , GTP Pyrophosphokinase/metabolism , RNA, Transfer/metabolism , Ribosomes/metabolism , Stress, Physiological , Adenosine/metabolism , Binding Sites , Cryoelectron Microscopy , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Escherichia coli/ultrastructure , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/ultrastructure , GTP Pyrophosphokinase/antagonists & inhibitors , GTP Pyrophosphokinase/genetics , GTP Pyrophosphokinase/ultrastructure , Gene Expression Regulation, Bacterial , Guanosine Tetraphosphate/chemistry , Guanosine Tetraphosphate/metabolism , Metabolic Networks and Pathways , Models, Molecular , Phosphorylation , Protein Biosynthesis , Protein Conformation , Protein Structure, Tertiary , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/ultrastructure , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/chemistry , Ribosomes/ultrastructure , Second Messenger Systems
20.
Nucleic Acids Res ; 48(7): 3776-3788, 2020 04 17.
Article in English | MEDLINE | ID: mdl-31960065

ABSTRACT

All enzymes face a challenge of discriminating cognate substrates from similar cellular compounds. Finding a correct substrate is especially difficult for the Escherichia coli Nudix hydrolase RppH, which triggers 5'-end-dependent RNA degradation by removing orthophosphate from the 5'-diphosphorylated transcripts. Here we show that RppH binds and slowly hydrolyzes NTPs, NDPs and (p)ppGpp, which each resemble the 5'-end of RNA. A series of X-ray crystal structures of RppH-nucleotide complexes, trapped in conformations either compatible or incompatible with hydrolysis, explain the low reaction rates of mononucleotides and suggest two distinct mechanisms for their hydrolysis. While RppH adopts the same catalytic arrangement with 5'-diphosphorylated nucleotides as with RNA, the enzyme hydrolyzes 5'-triphosphorylated nucleotides by extending the active site with an additional Mg2+ cation, which coordinates another reactive nucleophile. Although the average intracellular pH minimizes the hydrolysis of nucleotides by slowing their reaction with RppH, they nevertheless compete with RNA for binding and differentially inhibit the reactivity of RppH with triphosphorylated and diphosphorylated RNAs. Thus, E. coli RppH integrates various signals, such as competing non-cognate substrates and a stimulatory protein factor DapF, to achieve the differential degradation of transcripts involved in cellular processes important for the adaptation of bacteria to different growth conditions.


Subject(s)
Acid Anhydride Hydrolases/chemistry , Acid Anhydride Hydrolases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , RNA/metabolism , Acid Anhydride Hydrolases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Amino Acid Isomerases/metabolism , Catalytic Domain , Escherichia coli Proteins/antagonists & inhibitors , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Hydrogen-Ion Concentration , Magnesium/chemistry , Models, Molecular , Nucleotides/chemistry , Nucleotides/metabolism , RNA/chemistry , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL