Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
Nat Commun ; 14(1): 5151, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620344

ABSTRACT

Peptidoglycan (PG) is an essential structural component of the bacterial cell wall that is synthetized during cell division and elongation. PG forms an extracellular polymer crucial for cellular viability, the synthesis of which is the target of many antibiotics. PG assembly requires a glycosyltransferase (GT) to generate a glycan polymer using a Lipid II substrate, which is then crosslinked to the existing PG via a transpeptidase (TP) reaction. A Shape, Elongation, Division and Sporulation (SEDS) GT enzyme and a Class B Penicillin Binding Protein (PBP) form the core of the multi-protein complex required for PG assembly. Here we used single particle cryo-electron microscopy to determine the structure of a cell elongation-specific E. coli RodA-PBP2 complex. We combine this information with biochemical, genetic, spectroscopic, and computational analyses to identify the Lipid II binding sites and propose a mechanism for Lipid II polymerization. Our data suggest a hypothesis for the movement of the glycan strand from the Lipid II polymerization site of RodA towards the TP site of PBP2, functionally linking these two central enzymatic activities required for cell wall peptidoglycan biosynthesis.


Subject(s)
Escherichia coli , Peptidyl Transferases , Cryoelectron Microscopy , Escherichia coli/genetics , Peptidoglycan , Molecular Biology , Anti-Bacterial Agents , Glycosyltransferases
2.
Proc Natl Acad Sci U S A ; 120(24): e2302580120, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37276423

ABSTRACT

AmiA and AmiB are peptidoglycan-hydrolyzing enzymes from Escherichia coli that are required to break the peptidoglycan layer during bacterial cell division and maintain integrity of the cell envelope. In vivo, the activity of AmiA and AmiB is tightly controlled through their interactions with the membrane-bound FtsEX-EnvC complex. Activation of AmiA and AmiB requires access to a groove in the amidase-activating LytM domain of EnvC which is gated by ATP-driven conformational changes in FtsEX-EnvC complex. Here, we present a high-resolution structure of the isolated AmiA protein, confirming that it is autoinhibited in the same manner as AmiB and AmiC, and a complex of the AmiB enzymatic domain bound to the activating EnvC LytM domain. In isolation, the active site of AmiA is blocked by an autoinhibitory helix that binds directly to the catalytic zinc and fills the volume expected to accommodate peptidoglycan binding. In the complex, binding of the EnvC LytM domain induces a conformational change that displaces the amidase autoinhibitory helix and reorganizes the active site for activity. Our structures, together with complementary mutagenesis work, defines the conformational changes required to activate AmiA and/or AmiB through their interaction with their cognate activator EnvC.


Subject(s)
Escherichia coli Proteins , Escherichia coli Proteins/metabolism , Peptidoglycan/metabolism , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Escherichia coli/metabolism , Amidohydrolases/metabolism , Bacterial Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 120(8): e2215237120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36787358

ABSTRACT

Acinetobacter baumannii is a gram-negative bacterial pathogen that causes challenging nosocomial infections. ß-lactam targeting of penicillin-binding protein (PBP)-mediated cell wall peptidoglycan (PG) formation is a well-established antimicrobial strategy. Exposure to carbapenems or zinc (Zn)-deprived growth conditions leads to a rod-to-sphere morphological transition in A. baumannii, an effect resembling that caused by deficiency in the RodA-PBP2 PG synthesis complex required for cell wall elongation. While it is recognized that carbapenems preferentially acylate PBP2 in A. baumannii and therefore block the transpeptidase function of the RodA-PBP2 system, the molecular details underpinning cell wall elongation inhibition upon Zn starvation remain undefined. Here, we report the X-ray crystal structure of A. baumannii PBP2, revealing an unexpected Zn coordination site in the transpeptidase domain required for protein stability. Mutations in the Zn-binding site of PBP2 cause a loss of bacterial rod shape and increase susceptibility to ß-lactams, therefore providing a direct rationale for cell wall shape maintenance and Zn homeostasis in A. baumannii. Furthermore, the Zn-coordinating residues are conserved in various ß- and γ-proteobacterial PBP2 orthologs, consistent with a widespread Zn-binding requirement for function that has been previously unknown. Due to the emergence of resistance to virtually all marketed antibiotic classes, alternative or complementary antimicrobial strategies need to be explored. These findings offer a perspective for dual inhibition of Zn-dependent PG synthases and metallo-ß-lactamases by metal chelating agents, considered the most sought-after adjuvants to restore ß-lactam potency against gram-negative bacteria.


Subject(s)
Acinetobacter baumannii , Peptidyl Transferases , Acinetobacter baumannii/metabolism , Peptidyl Transferases/metabolism , Zinc/metabolism , Cell Shape , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Penicillin-Binding Proteins/metabolism , beta-Lactams/pharmacology , Carbapenems/pharmacology , Chelating Agents/pharmacology , Binding Sites , Bacterial Proteins/metabolism
4.
Front Cell Infect Microbiol ; 12: 863712, 2022.
Article in English | MEDLINE | ID: mdl-35967845

ABSTRACT

Escherichia coli is one of the most common Gram-negative pathogens and is responsible for infection leading to neonatal meningitis and sepsis. The FtsZ protein is a bacterial tubulin homolog required for cell division in most species, including E. coli. Several agents that block cell division have been shown to mislocalise FtsZ, including the bacteriophage λ-encoded Kil peptide, resulting in defective cell division and a filamentous phenotype, making FtsZ an attractive target for antimicrobials. In this study, we have used an in vitro meningitis model system for studying the effect of bacteriophages on FtsZ using fluorescent E. coli EV36/FtsZ-mCherry and K12/FtsZ-mNeon strains. We show localisation of FtsZ to the bacterial cell midbody as a single ring during normal growth conditions, and mislocalisation of FtsZ producing filamentous multi-ringed bacterial cells upon addition of the known inhibitor Kil peptide. We also show that when bacteriophages K1F-GFP and T7-mCherry were applied to their respective host strains, these phages can inhibit FtsZ and block bacterial cell division leading to a filamentous multi-ringed phenotype, potentially delaying lysis and increasing progeny number. This occurs in the exponential growth phase, as actively dividing hosts are needed. We present that ZapA protein is needed for phage inhibition by showing a phenotype recovery with a ZapA mutant strain, and we show that FtsI protein is also mislocalised upon phage infection. Finally, we show that the T7 peptide gp0.4 is responsible for the inhibition of FtsZ in K12 strains by observing a phenotype recovery with a T7Δ0.4 mutant.


Subject(s)
Bacterial Proteins , Bacteriophages , Carrier Proteins , Cytoskeletal Proteins , Escherichia coli Proteins , Escherichia coli , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriophages/genetics , Bacteriophages/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptides/genetics , Peptides/metabolism
5.
Plant Physiol ; 190(1): 165-179, 2022 08 29.
Article in English | MEDLINE | ID: mdl-35471580

ABSTRACT

Accumulating evidence suggests that peptidoglycan, consistent with a bacterial cell wall, is synthesized around the chloroplasts of many photosynthetic eukaryotes, from glaucophyte algae to early-diverging land plants including pteridophyte ferns, but the biosynthetic pathway has not been demonstrated. Here, we employed mass spectrometry and enzymology in a two-fold approach to characterize the synthesis of peptidoglycan in chloroplasts of the moss Physcomitrium (Physcomitrella) patens. To drive the accumulation of peptidoglycan pathway intermediates, P. patens was cultured with the antibiotics fosfomycin, D-cycloserine, and carbenicillin, which inhibit key peptidoglycan pathway proteins in bacteria. Mass spectrometry of the trichloroacetic acid-extracted moss metabolome revealed elevated levels of five of the predicted intermediates from uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) through the uridine diphosphate N-acetylmuramic acid (UDP-MurNAc)-D,L-diaminopimelate (DAP)-pentapeptide. Most Gram-negative bacteria, including cyanobacteria, incorporate meso-diaminopimelic acid (D,L-DAP) into the third residue of the stem peptide of peptidoglycan, as opposed to L-lysine, typical of most Gram-positive bacteria. To establish the specificity of D,L-DAP incorporation into the P. patens precursors, we analyzed the recombinant protein UDP-N-acetylmuramoyl-L-alanyl-D-glutamate-2,6-diaminopimelate ligase (MurE) from both P. patens and the cyanobacterium Anabaena sp. (Nostoc sp. strain PCC 7120). Both ligases incorporated D,L-DAP in almost complete preference to L-Lys, consistent with the mass spectrophotometric data, with catalytic efficiencies similar to previously documented Gram-negative bacterial MurE ligases. We discuss how these data accord with the conservation of active site residues common to DL-DAP-incorporating bacterial MurE ligases and of the probability of a horizontal gene transfer event within the plant peptidoglycan pathway.


Subject(s)
Cell Wall , Peptidoglycan , Bacteria/metabolism , Cell Wall/metabolism , Chloroplasts/metabolism , Gram-Negative Bacteria/metabolism , Ligases/metabolism , Lysine/metabolism , Peptidoglycan/chemistry , Peptidoglycan/genetics , Peptidoglycan/metabolism , Uridine Diphosphate/metabolism
6.
Protein Pept Lett ; 29(5): 448-459, 2022.
Article in English | MEDLINE | ID: mdl-35382715

ABSTRACT

BACKGROUND: Peptidoglycan (PG) is a key structural component of the bacterial cell wall and interruption of its biosynthesis is a validated target for antimicrobials. Of the enzymes involved in PG biosynthesis, D-alanyl,D-alanine ligase B (DdlB) is responsible for the condensation of two alanines, forming D-Ala-D-Ala, which is required for subsequent extracellular transpeptidase crosslinking of the mature peptidoglycan polymer. OBJECTIVE: We aimed at the biophysical characterization of recombinant Escherichia coli DdlB (EcDdlB), considering parameters of melting temperature (Tm), calorimetry and Van't Hoff enthalpy changes of denaturation ( ΔHUcal and ΔHUvH ), as well as characterization of elements of secondary structure at three different pHs. METHODS: DdlB was overexpressed in E. coli BL21 and purified by affinity chromatography. Thermal stability and structural characteristics of the purified enzyme were analyzed by circular dichroism (CD), differential scanning calorimetry and fluorescence spectroscopy. RESULTS: The stability of EcDdlB increased with proximity to its pI of 5.0, reaching the maximum at pH 5.4 with Tm and ΔHUvH U of 52.68 ºC and 484 kJ.mol-1, respectively. Deconvolutions of the CD spectra at 20 ºC showed a majority percentage of α-helix at pH 5.4 and 9.4, whereas for pH 7.4, an equal contribution of ß-structures and α-helices was calculated. Thermal denaturation process of EcDdlB proved to be irreversible with an increase in ß-structures that can contribute to the formation of protein aggregates. CONCLUSION: Such results will be useful for energy minimization of structural models aimed at virtual screening simulations, providing useful information in the search for drugs that inhibit peptidoglycan synthesis.


Subject(s)
Escherichia coli , Peptidoglycan , Alanine , Calorimetry, Differential Scanning , Circular Dichroism , Escherichia coli/genetics , Ligases , Protein Denaturation , Protein Structure, Secondary , Thermodynamics
7.
Nature ; 604(7905): 371-376, 2022 04.
Article in English | MEDLINE | ID: mdl-35388216

ABSTRACT

The outer membrane of Gram-negative bacteria has an external leaflet that is largely composed of lipopolysaccharide, which provides a selective permeation barrier, particularly against antimicrobials1. The final and crucial step in the biosynthesis of lipopolysaccharide is the addition of a species-dependent O-antigen to the lipid A core oligosaccharide, which is catalysed by the O-antigen ligase WaaL2. Here we present structures of WaaL from Cupriavidus metallidurans, both in the apo state and in complex with its lipid carrier undecaprenyl pyrophosphate, determined by single-particle cryo-electron microscopy. The structures reveal that WaaL comprises 12 transmembrane helices and a predominantly α-helical periplasmic region, which we show contains many of the conserved residues that are required for catalysis. We observe a conserved fold within the GT-C family of glycosyltransferases and hypothesize that they have a common mechanism for shuttling the undecaprenyl-based carrier to and from the active site. The structures, combined with genetic, biochemical, bioinformatics and molecular dynamics simulation experiments, offer molecular details on how the ligands come in apposition, and allows us to propose a mechanistic model for catalysis. Together, our work provides a structural basis for lipopolysaccharide maturation in a member of the GT-C superfamily of glycosyltransferases.


Subject(s)
Ligases , Lipopolysaccharides , O Antigens , Bacterial Proteins/chemistry , Carbon-Oxygen Ligases/chemistry , Carbon-Oxygen Ligases/genetics , Cryoelectron Microscopy , Glycosyltransferases , Gram-Negative Bacteria , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism
8.
Int J Mol Sci ; 22(23)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34884635

ABSTRACT

Bacteria must maintain the ability to modify and repair the peptidoglycan layer without jeopardising its essential functions in cell shape, cellular integrity and intermolecular interactions. A range of new experimental techniques is bringing an advanced understanding of how bacteria regulate and achieve peptidoglycan synthesis, particularly in respect of the central role played by complexes of Sporulation, Elongation or Division (SEDs) and class B penicillin-binding proteins required for cell division, growth and shape. In this review we highlight relationships implicated by a bioinformatic approach between the outer membrane, cytoskeletal components, periplasmic control proteins, and cell elongation/division proteins to provide further perspective on the interactions of these cell division, growth and shape complexes. We detail the network of protein interactions that assist in the formation of peptidoglycan and highlight the increasingly dynamic and connected set of protein machinery and macrostructures that assist in creating the cell envelope layers in Gram-negative bacteria.


Subject(s)
Cell Membrane/metabolism , Gram-Negative Bacteria/metabolism , Penicillin-Binding Proteins/metabolism , Peptidoglycan/metabolism , Periplasmic Proteins/metabolism , Bacterial Proteins/metabolism , Cell Wall/metabolism
9.
Front Microbiol ; 12: 737396, 2021.
Article in English | MEDLINE | ID: mdl-34737730

ABSTRACT

Cell division in Streptococcus pneumoniae (pneumococcus) is performed and regulated by a protein complex consisting of at least 14 different protein elements; known as the divisome. Recent findings have advanced our understanding of the molecular events surrounding this process and have provided new understanding of the mechanisms that occur during the division of pneumococcus. This review will provide an overview of the key protein complexes and how they are involved in cell division. We will discuss the interaction of proteins in the divisome complex that underpin the control mechanisms for cell division and cell wall synthesis and remodelling that are required in S. pneumoniae, including the involvement of virulence factors and capsular polysaccharides.

10.
J Med Chem ; 64(23): 17326-17345, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34845906

ABSTRACT

Herein, we report the design and synthesis of inhibitors of Mycobacterium tuberculosis (Mtb) phospho-MurNAc-pentapeptide translocase I (MurX), the first membrane-associated step of peptidoglycan synthesis, leveraging the privileged structure of the sansanmycin family of uridylpeptide natural products. A number of analogues bearing hydrophobic amide modifications to the pseudo-peptidic end of the natural product scaffold were generated that exhibited nanomolar inhibitory activity against Mtb MurX and potent activity against Mtb in vitro. We show that a lead analogue bearing an appended neopentylamide moiety possesses rapid antimycobacterial effects with a profile similar to the frontline tuberculosis drug isoniazid. This molecule was also capable of inhibiting Mtb growth in macrophages where mycobacteria reside in vivo and reduced mycobacterial burden in an in vivo zebrafish model of tuberculosis.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Oligopeptides/pharmacology , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Uridine/analogs & derivatives , Animals , Antitubercular Agents/pharmacology , Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Hydrophobic and Hydrophilic Interactions , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Oligopeptides/chemistry , Transferases (Other Substituted Phosphate Groups)/chemistry , Uridine/chemistry , Uridine/pharmacology , Zebrafish
11.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article in English | MEDLINE | ID: mdl-33785594

ABSTRACT

Survival in the human host requires bacteria to respond to unfavorable conditions. In the important Gram-positive pathogen Streptococcus pneumoniae, cell wall biosynthesis proteins MurM and MurN are tRNA-dependent amino acyl transferases which lead to the production of branched muropeptides. We demonstrate that wild-type cells experience optimal growth under mildly acidic stressed conditions, but ΔmurMN strain displays growth arrest and extensive lysis. Furthermore, these stress conditions compromise the efficiency with which alanyl-tRNAAla synthetase can avoid noncognate mischarging of tRNAAla with serine, which is toxic to cells. The observed growth defects are rescued by inhibition of the stringent response pathway or by overexpression of the editing domain of alanyl-tRNAAla synthetase that enables detoxification of tRNA misacylation. Furthermore, MurM can incorporate seryl groups from mischarged Seryl-tRNAAlaUGC into cell wall precursors with exquisite specificity. We conclude that MurM contributes to the fidelity of translation control and modulates the stress response by decreasing the pool of mischarged tRNAs. Finally, we show that enhanced lysis of ΔmurMN pneumococci is caused by LytA, and the murMN operon influences macrophage phagocytosis in a LytA-dependent manner. Thus, MurMN attenuates stress responses with consequences for host-pathogen interactions. Our data suggest a causal link between misaminoacylated tRNA accumulation and activation of the stringent response. In order to prevent potential corruption of translation, consumption of seryl-tRNAAla by MurM may represent a first line of defense. When this mechanism is overwhelmed or absent (ΔmurMN), the stringent response shuts down translation to avoid toxic generation of mistranslated/misfolded proteins.


Subject(s)
Bacterial Proteins/metabolism , Cell Division , Cell Wall/metabolism , Peptide Synthases/metabolism , RNA, Transfer/metabolism , Streptococcus pneumoniae/metabolism , Animals , Bacterial Proteins/genetics , Cell Line , Macrophages/immunology , Macrophages/microbiology , Mice , Operon , Peptide Synthases/genetics , Phagocytosis , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity
12.
Structure ; 29(7): 731-742.e6, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33740396

ABSTRACT

Branched Lipid II, required for the formation of indirectly crosslinked peptidoglycan, is generated by MurM, a protein essential for high-level penicillin resistance in the human pathogen Streptococcus pneumoniae. We have solved the X-ray crystal structure of Staphylococcus aureus FemX, an isofunctional homolog, and have used this as a template to generate a MurM homology model. Using this model, we perform molecular docking and molecular dynamics to examine the interaction of MurM with the phospholipid bilayer and the membrane-embedded Lipid II substrate. Our model suggests that MurM is associated with the major membrane phospholipid cardiolipin, and experimental evidence confirms that the activity of MurM is enhanced by this phospholipid and inhibited by its direct precursor phosphatidylglycerol. The spatial association of pneumococcal membrane phospholipids and their impact on MurM activity may therefore be critical to the final architecture of peptidoglycan and the expression of clinically relevant penicillin resistance in this pathogen.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cardiolipins/metabolism , Penicillin Resistance , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Streptococcus pneumoniae/growth & development , Binding Sites , Cell Membrane/metabolism , Crystallography, X-Ray , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Phosphatidylglycerols/metabolism , Protein Conformation , Sequence Homology, Amino Acid , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
13.
Anim Reprod Sci ; 225: 106672, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33421820

ABSTRACT

Physiological stress responses have been associated with lesser pregnancy rates in beef cattle. Alternative cattle restraint methods have been developed to reduce this negative effect. The objective was to determine if there was a difference in pregnancy rate as a result of fixed-time artificial insemination (FTAI) in Bos taurus females when using a breeding box compared with a squeeze chute for restraining cows when conducting FTAI. There were FTAI treatment regimens imposed on cows and heifers at two separate locations (Virginia Tech University, VT; and R.A. Brown Ranch, RAB) with the FTAI being conducted while animals were restrained in a squeeze chute (n = 169) or a breeding box (n = 162). There was imposing of a 7-day Co-Sync + CIDR pre-breeding treatment regimen. Females were randomly assigned to treatments just prior to breeding. Immediately following breeding, blood was collected using coccygeal venipuncture procedures for assessment of plasma cortisol concentration. Chute and exit scores (1-5) were determined at the time of FTAI. Pregnancy rate, cortisol, chute score, and exit score were analyzed. There was no difference in pregnancy rate between treatments (57 ± 26% and 69 ± 26 %, respectively P = 0.59).There were differences in chute (1.52 ± 0.05 and 1.27 ± 0.05, respectively P < 0.05) and exit (1.26 ± 0.04 and 1.08 ± 0.04, respectively P < 0.05) scores between the two treatment groups. Results from the present study are not conclusive in ascertaining effects of the two types of restraint during FTAI on reproductive efficiency.


Subject(s)
Cattle/physiology , Insemination, Artificial/veterinary , Pregnancy Rate , Restraint, Physical/veterinary , Stress, Physiological , Animals , Cattle/blood , Estrus Synchronization/methods , Female , Hydrocortisone/blood , Pregnancy
14.
Elife ; 92020 12 14.
Article in English | MEDLINE | ID: mdl-33315009

ABSTRACT

The Gram-negative outer-membrane envelops the bacterium and functions as a permeability barrier against antibiotics, detergents, and environmental stresses. Some virulence factors serve to maintain the integrity of the outer membrane, including DolP (formerly YraP) a protein of unresolved structure and function. Here, we reveal DolP is a lipoprotein functionally conserved amongst Gram-negative bacteria and that loss of DolP increases membrane fluidity. We present the NMR solution structure for Escherichia coli DolP, which is composed of two BON domains that form an interconnected opposing pair. The C-terminal BON domain binds anionic phospholipids through an extensive membrane:protein interface. This interaction is essential for DolP function and is required for sub-cellular localisation of the protein to the cell division site, providing evidence of subcellular localisation of these phospholipids within the outer membrane. The structure of DolP provides a new target for developing therapies that disrupt the integrity of the bacterial cell envelope.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Protein Transport/physiology , Anti-Bacterial Agents/metabolism , Cell Wall/metabolism , Escherichia coli/metabolism , Gram-Negative Bacteria/metabolism , Lipoproteins/metabolism , Virulence Factors/metabolism
15.
Proc Natl Acad Sci U S A ; 117(45): 28355-28365, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33097670

ABSTRACT

FtsEX is a bacterial ABC transporter that regulates the activity of periplasmic peptidoglycan amidases via its interaction with the murein hydrolase activator, EnvC. In Escherichia coli, FtsEX is required to separate daughter cells after cell division and for viability in low-osmolarity media. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for amidase activation, but the process itself is poorly understood. Here we present the 2.1 Å structure of the FtsX periplasmic domain in complex with its periplasmic partner, EnvC. The EnvC-FtsX periplasmic domain complex has a 1-to-2 stoichiometry with two distinct FtsX-binding sites located within an antiparallel coiled coil domain of EnvC. Residues involved in amidase activation map to a previously identified groove in the EnvC LytM domain that is here found to be occluded by a "restraining arm" suggesting a self-inhibition mechanism. Mutational analysis, combined with bacterial two-hybrid screens and in vivo functional assays, verifies the FtsEX residues required for EnvC binding and experimentally test a proposed mechanism for amidase activation. We also define a predicted link between FtsEX and integrity of the outer membrane. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for resistance to membrane-attacking antibiotics and detergents to which E. coli would usually be considered intrinsically resistant. These structural and functional data provide compelling mechanistic insight into FtsEX-mediated regulation of EnvC and its downstream control of periplasmic peptidoglycan amidases.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/chemistry , Cell Cycle Proteins/chemistry , Cell Division/physiology , Endopeptidases/chemistry , Periplasm/metabolism , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Crystallography, X-Ray , DNA Mutational Analysis , Endopeptidases/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins , Models, Molecular , Mutation , N-Acetylmuramoyl-L-alanine Amidase/genetics , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Periplasm/chemistry , Protein Binding , Protein Conformation , Protein Domains , Protein Interaction Domains and Motifs
16.
Sci Rep ; 10(1): 5727, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32235931

ABSTRACT

The glycopeptide antibiotic vancomycin has been widely used to treat infections of Gram-positive bacteria including Clostridium difficile and methicillin-resistant Staphylococcus aureus. However, since its introduction, high level vancomycin resistance has emerged. The genes responsible require the action of the two-component regulatory system VanSR to induce expression of resistance genes. The mechanism of detection of vancomycin by this two-component system has yet to be elucidated. Diverging evidence in the literature supports activation models in which the VanS protein binds either vancomycin, or Lipid II, to induce resistance. Here we investigated the interaction between vancomycin and VanS from Streptomyces coelicolor (VanSSC), a model Actinomycete. We demonstrate a direct interaction between vancomycin and purified VanSSC, and traced these interactions to the extracellular region of the protein, which we reveal adopts a predominantly α-helical conformation. The VanSSC-binding epitope within vancomycin was mapped to the N-terminus of the peptide chain, distinct from the binding site for Lipid II. In targeting a separate site on vancomycin, the effective VanS ligand concentration includes both free and lipid-bound molecules, facilitating VanS activation. This is the first molecular description of the VanS binding site within vancomycin, and could direct engineering of future therapeutics.


Subject(s)
Bacterial Proteins/metabolism , Streptomyces coelicolor/metabolism , Transcription Factors/metabolism , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Bacterial Proteins/genetics , Binding Sites , Gene Expression Regulation, Bacterial , Streptomyces coelicolor/drug effects , Streptomyces coelicolor/genetics , Transcription Factors/genetics
17.
J Am Chem Soc ; 142(11): 5034-5048, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32048840

ABSTRACT

Penicillin binding proteins (PBPs) catalyzing transpeptidation reactions that stabilize the peptidoglycan component of the bacterial cell wall are the targets of ß-lactams, the most clinically successful antibiotics to date. However, PBP-transpeptidation enzymology has evaded detailed analysis, because of the historical unavailability of kinetically competent assays with physiologically relevant substrates and the previously unappreciated contribution of protein cofactors to PBP activity. By re-engineering peptidoglycan synthesis, we have constructed a continuous spectrophotometric assay for transpeptidation of native or near native peptidoglycan precursors and fragments by Escherichia coli PBP1B, allowing us to (a) identify recognition elements of transpeptidase substrates, (b) reveal a novel mechanism of stereochemical editing within peptidoglycan transpeptidation, (c) assess the impact of peptidoglycan substrates on ß-lactam targeting of transpeptidation, and (d) demonstrate that both substrates have to be bound before transpeptidation occurs. The results allow characterization of high molecular weight PBPs as enzymes and not merely the targets of ß-lactam acylation.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Penicillin-Binding Proteins/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan/chemistry , Polyisoprenyl Phosphate Monosaccharides/chemistry , Polyisoprenyl Phosphate Oligosaccharides/chemistry , Serine-Type D-Ala-D-Ala Carboxypeptidase/chemistry , Bacterial Outer Membrane Proteins/chemistry , Biocatalysis , Enzyme Assays/methods , Kinetics , Stereoisomerism , Substrate Specificity
19.
J Perioper Pract ; 30(6): 176-182, 2020 06.
Article in English | MEDLINE | ID: mdl-31524069

ABSTRACT

Royal Navy Operating Department Practitioners are employed in a number of different roles, during peacetime, humanitarian aid operations and periods of war. In recent times, Royal Navy Operating Department Practitioners have deployed on active operations in addition to working in NHS hospitals at home in the United Kingdom. This article will explore the different avenues and experiences of Operating Department Practitioners who are currently serving in the Royal Navy. The reader will then also gain an insight into the different echelons of care provided by the Defence Medical Services to the United Kingdom Armed Forces and Allied Nations. The article will then consider the unique experiences available to Royal Navy Operating Department Practitioners in this multi-faceted role which offers the opportunity to explore work patterns in different environments.


Subject(s)
Hospitals, Military/standards , Military Medicine/standards , Operating Rooms/standards , Perioperative Care/standards , Practice Guidelines as Topic , Ships/standards , Adult , Female , Humans , Male , Middle Aged , United Kingdom
20.
Sci Rep ; 9(1): 18712, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31822696

ABSTRACT

The E. coli membrane protein ZipA, binds to the tubulin homologue FtsZ, in the early stage of cell division. We isolated ZipA in a Styrene Maleic Acid lipid particle (SMALP) preserving its position and integrity with native E. coli membrane lipids. Direct binding of ZipA to FtsZ is demonstrated, including FtsZ fibre bundles decorated with ZipA. Using Cryo-Electron Microscopy, small-angle X-ray and neutron scattering, we determine the encapsulated-ZipA structure in isolation, and in complex with FtsZ to a resolution of 1.6 nm. Three regions can be identified from the structure which correspond to, SMALP encapsulated membrane and ZipA transmembrane helix, a separate short compact tether, and ZipA globular head which binds FtsZ. The complex extends 12 nm from the membrane in a compact structure, supported by mesoscale modelling techniques, measuring the movement and stiffness of the regions within ZipA provides molecular scale analysis and visualisation of the early divisome.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Division/physiology , Cytoskeletal Proteins/metabolism , Escherichia coli Proteins/metabolism , Bacterial Proteins/physiology , Carrier Proteins/physiology , Carrier Proteins/ultrastructure , Cell Cycle Proteins/physiology , Cell Cycle Proteins/ultrastructure , Cryoelectron Microscopy/methods , Cytoskeletal Proteins/physiology , Escherichia coli/metabolism , Escherichia coli Proteins/physiology , Escherichia coli Proteins/ultrastructure , Membrane Proteins/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...