Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 67: 116819, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35635930

ABSTRACT

A series of salicylanilide compounds was previously identified as antibacterial agents that inhibit the peptidoglycan formation. To find the exact binding mode, we synthesized a benzophenone-containing salicylanilide compound (1) and used it as a photoaffinity probe to label Acinetobacter baumannii penicillin-binding protein (PBP1b). After incubation and photo-irradiation, the labeled protein was subjected to trypsin digestion, dialysis enrichment, LC-ESI-MS/MS analysis, and Mascot search to reveal an octadecapeptide sequence 364RQLRTEYQESDLTNQGLR381 that was labeled at E372. Our molecular docking experiments suggest a hydrophobic pocket surrounded by R367 and E372 is the binding site of salicylanilide 1. The pocket lies in between the transglycosylase and transpeptidase domains, thus binding of salicylanilide 1 can block the propagation pathway to disrupt the growth of peptidoglycan chain.


Subject(s)
Peptidoglycan Glycosyltransferase , Benzophenones/pharmacology , Escherichia coli/metabolism , Molecular Docking Simulation , Peptidoglycan , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/metabolism , Photoaffinity Labels , Salicylanilides , Tandem Mass Spectrometry
2.
PLoS Genet ; 17(4): e1009366, 2021 04.
Article in English | MEDLINE | ID: mdl-33857142

ABSTRACT

SEDS family peptidoglycan (PG) glycosyltransferases, RodA and FtsW, require their cognate transpeptidases PBP2 and FtsI (class B penicillin binding proteins) to synthesize PG along the cell cylinder and at the septum, respectively. The activities of these SEDS-bPBPs complexes are tightly regulated to ensure proper cell elongation and division. In Escherichia coli FtsN switches FtsA and FtsQLB to the active forms that synergize to stimulate FtsWI, but the exact mechanism is not well understood. Previously, we isolated an activation mutation in ftsW (M269I) that allows cell division with reduced FtsN function. To try to understand the basis for activation we isolated additional substitutions at this position and found that only the original substitution produced an active mutant whereas drastic changes resulted in an inactive mutant. In another approach we isolated suppressors of an inactive FtsL mutant and obtained FtsWE289G and FtsIK211I and found they bypassed FtsN. Epistatic analysis of these mutations and others confirmed that the FtsN-triggered activation signal goes from FtsQLB to FtsI to FtsW. Mapping these mutations, as well as others affecting the activity of FtsWI, on the RodA-PBP2 structure revealed they are located at the interaction interface between the extracellular loop 4 (ECL4) of FtsW and the pedestal domain of FtsI (PBP3). This supports a model in which the interaction between the ECL4 of SEDS proteins and the pedestal domain of their cognate bPBPs plays a critical role in the activation mechanism.


Subject(s)
Bacterial Proteins/ultrastructure , Escherichia coli Proteins/ultrastructure , Membrane Proteins/ultrastructure , Multiprotein Complexes/ultrastructure , Penicillin-Binding Proteins/ultrastructure , Peptidoglycan Glycosyltransferase/ultrastructure , Protein Conformation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/genetics , Peptidoglycan/chemistry , Peptidoglycan/genetics , Peptidoglycan/ultrastructure , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/genetics , Peptidyl Transferases/chemistry , Peptidyl Transferases/genetics , Peptidyl Transferases/ultrastructure
3.
Nat Microbiol ; 5(6): 813-820, 2020 06.
Article in English | MEDLINE | ID: mdl-32152588

ABSTRACT

The shape, elongation, division and sporulation (SEDS) proteins are a highly conserved family of transmembrane glycosyltransferases that work in concert with class B penicillin-binding proteins (bPBPs) to build the bacterial peptidoglycan cell wall1-6. How these proteins coordinate polymerization of new glycan strands with their crosslinking to the existing peptidoglycan meshwork is unclear. Here, we report the crystal structure of the prototypical SEDS protein RodA from Thermus thermophilus in complex with its cognate bPBP at 3.3 Å resolution. The structure reveals a 1:1 stoichiometric complex with two extensive interaction interfaces between the proteins: one in the membrane plane and the other at the extracytoplasmic surface. When in complex with a bPBP, RodA shows an approximately 10 Å shift of transmembrane helix 7 that exposes a large membrane-accessible cavity. Negative-stain electron microscopy reveals that the complex can adopt a variety of different conformations. These data define the bPBP pedestal domain as the key allosteric activator of RodA both in vitro and in vivo, explaining how a SEDS-bPBP complex can coordinate its dual enzymatic activities of peptidoglycan polymerization and crosslinking to build the cell wall.


Subject(s)
Models, Molecular , Multiprotein Complexes/chemistry , Penicillin-Binding Proteins/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Protein Multimerization , Binding Sites , Cell Wall/metabolism , Molecular Structure , Multiprotein Complexes/metabolism , Penicillin-Binding Proteins/metabolism , Peptidoglycan/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Protein Binding , Protein Conformation , Structure-Activity Relationship
4.
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
5.
J Mol Biol ; 431(18): 3501-3519, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31301409

ABSTRACT

Even with the emergence of antibiotic resistance, penicillin and the wider family of ß-lactams have remained the single most important family of antibiotics. The periplasmic/extra-cytoplasmic targets of penicillin are a family of enzymes with a highly conserved catalytic activity involved in the final stage of bacterial cell wall (peptidoglycan) biosynthesis. Named after their ability to bind penicillin, rather than their catalytic activity, these key targets are called penicillin-binding proteins (PBPs). Resistance is predominantly mediated by reducing the target drug concentration via ß-lactamases; however, naturally transformable bacteria have also acquired target-mediated resistance by inter-species recombination. Here we focus on structural based interpretations of amino acid alterations associated with the emergence of resistance within clinical isolates and include new PBP3 structures along with new, and improved, PBP-ß-lactam co-structures.


Subject(s)
Penicillin-Binding Proteins/chemistry , Serine-Type D-Ala-D-Ala Carboxypeptidase/chemistry , beta-Lactam Resistance/physiology , beta-Lactams/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Haemophilus influenzae/enzymology , Models, Molecular , Mutation , Neisseria gonorrhoeae/enzymology , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/genetics , Peptidoglycan Glycosyltransferase/metabolism , Protein Conformation , Protein Domains , Pseudomonas aeruginosa/enzymology , Sequence Alignment , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , beta-Lactamases/chemistry , beta-Lactamases/metabolism , beta-Lactams/pharmacology
6.
Mol Microbiol ; 110(3): 335-356, 2018 11.
Article in English | MEDLINE | ID: mdl-30044025

ABSTRACT

Bacteria surround their cytoplasmic membrane with an essential, stress-bearing peptidoglycan (PG) layer consisting of glycan chains linked by short peptides into a mesh-like structure. Growing and dividing cells expand their PG layer using inner-membrane anchored PG synthases, including Penicillin-binding proteins (PBPs), which participate in dynamic protein complexes to facilitate cell wall growth. In Escherichia coli, and presumably other Gram-negative bacteria, growth of the mainly single layered PG is regulated by outer membrane-anchored lipoproteins. The lipoprotein LpoB is required to activate PBP1B, which is a major, bi-functional PG synthase with glycan chain polymerising (glycosyltransferase) and peptide cross-linking (transpeptidase) activities. In this work we show how the binding of LpoB to the regulatory UB2H domain of PBP1B activates both activities. Binding induces structural changes in the UB2H domain, which transduce to the two catalytic domains by distinct allosteric pathways. We also show how an additional regulator protein, CpoB, is able to selectively modulate the TPase activation by LpoB without interfering with GTase activation.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/metabolism , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/metabolism , Peptidoglycan/biosynthesis , Serine-Type D-Ala-D-Ala Carboxypeptidase/chemistry , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Allosteric Regulation , Membrane Proteins/metabolism , Protein Binding , Protein Conformation
7.
mBio ; 9(3)2018 06 12.
Article in English | MEDLINE | ID: mdl-29895635

ABSTRACT

Clostridium difficile is the major etiologic agent of antibiotic-associated intestinal disease. Pathogenesis of C. difficile is mainly attributed to the production and secretion of toxins A and B. Unlike most clostridial toxins, toxins A and B have no signal peptide, and they are therefore secreted by unusual mechanisms involving the holin-like TcdE protein and/or autolysis. In this study, we characterized the cell surface protein Cwp19, a newly identified peptidoglycan-degrading enzyme containing a novel catalytic domain. We purified a recombinant His6-tagged Cwp19 protein and showed that it has lytic transglycosylase activity. Moreover, we observed that Cwp19 is involved in cell autolysis and that a C. difficilecwp19 mutant exhibited delayed autolysis in stationary phase compared to the wild type when bacteria were grown in brain heart infusion (BHI) medium. Wild-type cell autolysis is correlated to strong alterations of cell wall thickness and integrity and to release of cytoplasmic material. Furthermore, we demonstrated that toxins were released into the extracellular medium as a result of Cwp19-induced autolysis when cells were grown in BHI medium. In contrast, Cwp19 did not induce autolysis or toxin release when cells were grown in tryptone-yeast extract (TY) medium. These data provide evidence for the first time that TcdE and bacteriolysis are coexisting mechanisms for toxin release, with their relative contributions in vitro depending on growth conditions. Thus, Cwp19 is an important surface protein involved in autolysis of vegetative cells of C. difficile that mediates the release of the toxins from the cell cytosol in response to specific environment conditions.IMPORTANCEClostridium difficile-associated disease is mainly known as a health care-associated infection. It represents the most problematic hospital-acquired infection in North America and Europe and exerts significant economic pressure on health care systems. Virulent strains of C. difficile generally produce two toxins that have been identified as the major virulence factors. The mechanism for release of these toxins from bacterial cells is not yet fully understood but is thought to be partly mediated by bacteriolysis. Here we identify a novel peptidoglycan hydrolase in C. difficile, Cwp19, exhibiting lytic transglycosylase activity. We show that Cwp19 contributes to C. difficile cell autolysis in the stationary phase and, consequently, to toxin release, most probably as a response to environmental conditions such as nutritional signals. These data highlight that Cwp19 constitutes a promising target for the development of new preventive and curative strategies.


Subject(s)
Bacterial Proteins/metabolism , Bacteriolysis , Clostridioides difficile/enzymology , Clostridioides difficile/growth & development , Peptidoglycan Glycosyltransferase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Cell Wall/genetics , Cell Wall/metabolism , Clostridioides difficile/genetics , Clostridioides difficile/physiology , Clostridium Infections/microbiology , Gene Expression Regulation, Bacterial , Humans , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/genetics
8.
Biochemistry ; 56(48): 6317-6320, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29131935

ABSTRACT

Formation of catenanes by proteins is rare, with few known examples. We report herein the X-ray structure of a catenane dimer of lytic transglycosylase SltB1 of Pseudomonas aeruginosa. The enzyme is soluble and exists in the periplasmic space, where it modifies the bacterial cell wall. The catenane dimer exhibits the protein monomers in a noncovalent chain-link arrangement, whereby a stretch of 51 amino acids (to become a loop and three helices) from one monomer threads through the central opening of the structure of the partner monomer. The protein folds after threading in a manner that leaves two helices (α1 and α2) as stoppers to impart stability to the dimer structure. The symmetric embrace by the two SltB1 molecules occludes both active sites entirely, an arrangement that is sustained by six electrostatic interactions between the two monomers. In light of the observation of these structural motifs in all members of Family 3 lytic transglycosylases, catenanes might be present for those enzymes, as well. The dimeric catenane might represent a regulated form of SltB1.


Subject(s)
Crystallography, X-Ray , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Models, Molecular , Protein Conformation , Protein Folding
9.
Biochem Biophys Res Commun ; 484(4): 850-856, 2017 03 18.
Article in English | MEDLINE | ID: mdl-28163025

ABSTRACT

Glycogen branching enzyme (GBE) catalyzes the formation of α-1,6-branching points during glycogenesis by cleaving α-1,4 bonds and making new α-1,6 bonds. Most GBEs belong to the glycoside hydrolase 13 family (GH13), but new GBEs in the GH57 family have been isolated from Archaea. Here, we determined the crystal structure of a GH57 GBE from the hyperthermophilic archaeon Pyrococcus horikoshii (PhGBE) at a resolution of 2.3 Å. PhGBE exhibits both α-1,6-branching activity and endo-α-1,4 hydrolytic activity. PhGBE has a central (ß/α)7-barrel domain that contains an embedded helix domain and an α-helix-rich C-terminal domain. The active-site cleft is located at the interface of the central and C-terminal domains. Amino acid substitution at Trp22, which is separate from the catalytic nucleophilic residue, abolished both enzymatic activities, indicating that Trp22 might be responsible for substrate recognition. We also observed that shortening of the flexible loop near the catalytic residue changed branched chain lengths of the reaction products with increased hydrolytic activity. Taken together, our findings propose a molecular mechanism for how GH57 GBEs exhibit the two activities and where the substrate binds the enzyme.


Subject(s)
1,4-alpha-Glucan Branching Enzyme/chemistry , 1,4-alpha-Glucan Branching Enzyme/ultrastructure , Glycogen/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/ultrastructure , Pyrococcus horikoshii/enzymology , Binding Sites , Enzyme Activation , Protein Binding , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
10.
Adv Exp Med Biol ; 925: 41-56, 2017.
Article in English | MEDLINE | ID: mdl-27722959

ABSTRACT

The flagellum is an important macromolecular machine for many pathogenic bacteria. It is a hetero-oligomeric structure comprised of three major sub-structures: basal body, hook and thin helical filament. An important step during flagellum assembly is the localized and controlled degradation of the peptidoglycan sacculus to allow for the insertion of the rod as well as to facilitate anchoring for proper motor function. The peptidoglycan lysis events require specialized lytic enzymes, ß-N-acetylglucosaminidases and lytic transglycosylases, which differ in flagellated proteobacteria. Due to their autolytic activity, these enzymes need to be controlled in order to prevent cellular lysis. This review summarizes are current understanding of the peptidoglycan lysis events required for flagellum assembly and motility with a main focus on Gram-negative bacteria.


Subject(s)
Acetylglucosaminidase/genetics , Bacterial Proteins/genetics , Flagella/genetics , Gene Expression Regulation, Bacterial , Peptidoglycan Glycosyltransferase/genetics , Acetylglucosaminidase/chemistry , Acetylglucosaminidase/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacteriolysis/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/ultrastructure , Flagella/enzymology , Flagella/ultrastructure , Helicobacter pylori/enzymology , Helicobacter pylori/genetics , Helicobacter pylori/ultrastructure , Multigene Family , Peptidoglycan/metabolism , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/metabolism , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/ultrastructure , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Salmonella typhimurium/ultrastructure , Sequence Alignment
11.
Infect Immun ; 85(2)2017 02.
Article in English | MEDLINE | ID: mdl-27895129

ABSTRACT

The pathogenicity of the Gram-negative plant-pathogenic bacterium Xanthomonas campestris pv. vesicatoria depends on a type III secretion (T3S) system, which spans both bacterial membranes and translocates effector proteins into plant cells. The assembly of the T3S system presumably involves the predicted lytic transglycosylase (LT) HpaH, which is encoded adjacent to the T3S gene cluster. Bacterial LTs degrade peptidoglycan and often promote the formation of membrane-spanning macromolecular protein complexes. In the present study, we show that HpaH localizes to the bacterial periplasm and binds to peptidoglycan as well as to components of the T3S system, including the predicted periplasmic inner rod proteins HrpB1 and HrpB2 as well as the pilus protein HrpE. In vivo translocation assays revealed that HpaH promotes the translocation of various effector proteins and of early substrates of the T3S system, suggesting a general contribution of HpaH to type III-dependent protein export. Mutant studies and the analysis of reporter fusions showed that the N-terminal region of HpaH contributes to protein function and is proteolytically cleaved. The N-terminally truncated HpaH cleavage product is secreted into the extracellular milieu by a yet-unknown transport pathway, which is independent of the T3S system.


Subject(s)
Peptidoglycan Glycosyltransferase/metabolism , Type III Secretion Systems , Xanthomonas campestris/physiology , Xanthomonas vesicatoria/physiology , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Codon, Initiator , Gene Expression , Gene Expression Regulation, Bacterial , Models, Molecular , Molecular Conformation , Peptidoglycan/chemistry , Peptidoglycan/metabolism , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/genetics , Plants/microbiology , Protein Binding , Protein Biosynthesis , Protein Interaction Domains and Motifs , Protein Transport , Proteolysis , Recombinant Fusion Proteins/metabolism , Sequence Deletion
12.
J Biol Chem ; 292(3): 979-993, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27899450

ABSTRACT

In Escherichia coli, the peptidoglycan cell wall is synthesized by bifunctional penicillin-binding proteins such as PBP1b that have both transpeptidase and transglycosylase activities. The PBP1b transpeptidase domain is a major target of ß-lactams, and therefore it is important to attain a detailed understanding of its inhibition. The peptidoglycan glycosyltransferase domain of PBP1b is also considered an excellent antibiotic target yet is not exploited by any clinically approved antibacterials. Herein, we adapt a pyrophosphate sensor assay to monitor PBP1b-catalyzed glycosyltransfer and present an improved crystallographic model for inhibition of the PBP1b glycosyltransferase domain by the potent substrate analog moenomycin. We elucidate the structure of a previously disordered region in the glycosyltransferase active site and discuss its implications with regards to peptidoglycan polymerization. Furthermore, we solve the crystal structures of E. coli PBP1b bound to multiple different ß-lactams in the transpeptidase active site and complement these data with gel-based competition assays to provide a detailed structural understanding of its inhibition. Taken together, these biochemical and structural data allow us to propose new insights into inhibition of both enzymatic domains in PBP1b.


Subject(s)
Escherichia coli K12/chemistry , Escherichia coli Proteins/chemistry , Penicillin-Binding Proteins/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Serine-Type D-Ala-D-Ala Carboxypeptidase/chemistry , beta-Lactams/chemistry , Crystallography, X-Ray , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Penicillin-Binding Proteins/genetics , Peptidoglycan Glycosyltransferase/genetics , Protein Domains , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics
13.
Chembiochem ; 17(23): 2250-2256, 2016 12 02.
Article in English | MEDLINE | ID: mdl-27709766

ABSTRACT

Surface plasmon resonance (SPR) is one of the most powerful label-free methods to determine the kinetic parameters of molecular interactions in real time and in a highly sensitive way. Penicillin-binding proteins (PBPs) are peptidoglycan synthesis enzymes present in most bacteria. Established protocols to analyze interactions of PBPs by SPR involve immobilization to an ampicillin-coated chip surface (a ß-lactam antibiotic mimicking its substrate), thereby forming a covalent complex with the PBPs transpeptidase (TP) active site. However, PBP interactions measured with a substrate-bound TP domain potentially affect interactions near the TPase active site. Furthermore, in vivo PBPs are anchored in the inner membrane by an N-terminal transmembrane helix, and hence immobilization at the C-terminal TPase domain gives an orientation contrary to the in vivo situation. We designed a new procedure: immobilization of PBP by copper-free click chemistry at an azide incorporated in the N terminus. In a proof-of-principle study, we immobilized Escherichia coli PBP1B on an SPR chip surface and used this for the analysis of the well-characterized interaction of PBP1B with LpoB. The site-specific incorporation of the azide affords control over protein orientation, thereby resulting in a homogeneous immobilization on the chip surface. This method can be used to study topology-dependent interactions of any (membrane) protein.


Subject(s)
Escherichia coli Proteins/chemistry , Immobilized Proteins/chemistry , Penicillin-Binding Proteins/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Serine-Type D-Ala-D-Ala Carboxypeptidase/chemistry , Surface Plasmon Resonance , Azides/chemistry , Azides/metabolism , Cyclooctanes/chemistry , Cyclooctanes/metabolism , Escherichia coli Proteins/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Immobilized Proteins/metabolism , Models, Molecular , Molecular Structure , Penicillin-Binding Proteins/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Surface Properties
14.
Drug Resist Updat ; 28: 91-104, 2016 09.
Article in English | MEDLINE | ID: mdl-27620957

ABSTRACT

Antimicrobial resistance is one of the most serious health threats. Cell-wall remodeling processes are tightly regulated to warrant bacterial survival and in some cases are directly linked to antibiotic resistance. Remodeling produces cell-wall fragments that are recycled but can also act as messengers for bacterial communication, as effector molecules in immune response and as signaling molecules triggering antibiotic resistance. This review is intended to provide state-of-the-art information about the molecular mechanisms governing this process and gather structural information of the different macromolecular machineries involved in peptidoglycan recycling in Gram-negative bacteria. The growing body of literature on the 3D structures of the corresponding macromolecules reveals an extraordinary complexity. Considering the increasing incidence and widespread emergence of Gram-negative multidrug-resistant pathogens in clinics, structural information on the main actors of the recycling process paves the way for designing novel antibiotics disrupting cellular communication in the recycling-resistance pathway.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Wall/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Gene Expression Regulation, Bacterial , Gram-Negative Bacteria/drug effects , Peptidoglycan/metabolism , Biological Transport , Cell Wall/chemistry , Cell Wall/metabolism , Gram-Negative Bacteria/enzymology , Gram-Negative Bacteria/genetics , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Hexosaminidases/genetics , Hexosaminidases/metabolism , Humans , Models, Molecular , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/classification , Peptidoglycan Glycosyltransferase/genetics , Peptidoglycan Glycosyltransferase/metabolism , Protein Domains , Protein Structure, Secondary , beta-Lactamases/genetics , beta-Lactamases/metabolism
15.
Nature ; 537(7622): 634-638, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27525505

ABSTRACT

Elongation of rod-shaped bacteria is mediated by a dynamic peptidoglycan-synthetizing machinery called the Rod complex. Here we report that, in Bacillus subtilis, this complex is functional in the absence of all known peptidoglycan polymerases. Cells lacking these enzymes survive by inducing an envelope stress response that increases the expression of RodA, a widely conserved core component of the Rod complex. RodA is a member of the SEDS (shape, elongation, division and sporulation) family of proteins, which have essential but ill-defined roles in cell wall biogenesis during growth, division and sporulation. Our genetic and biochemical analyses indicate that SEDS proteins constitute a family of peptidoglycan polymerases. Thus, B. subtilis and probably most bacteria use two distinct classes of polymerase to synthesize their exoskeleton. Our findings indicate that SEDS family proteins are core cell wall synthases of the cell elongation and division machinery, and represent attractive targets for antibiotic development.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/metabolism , Cell Wall/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Peptidoglycan/biosynthesis , Polymerization , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/cytology , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Division , Cell Wall/chemistry , Drug Design , Drug Resistance, Bacterial/drug effects , Mutation , Oligosaccharides/pharmacology , Penicillin-Binding Proteins/classification , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/genetics , Phenotype
16.
Methods Mol Biol ; 1440: 171-84, 2016.
Article in English | MEDLINE | ID: mdl-27311672

ABSTRACT

Bacterial cell wall peptidoglycan is synthesized from its precursor lipid II by two enzymatic reactions. First, glycosyltransferases polymerize the glycan strands and second, DD-transpeptidases form cross-links between peptides of neighboring strands. Most bacteria possess bifunctional peptidoglycan synthesis enzymes capable of catalyzing both reactions. Here, we describe a continuous fluorescence glycosyltransferase assay using Dansyl-labeled lipid II as substrate. Progression of the reaction is monitored by the reduction in fluorescence over time. The assay is suitable to investigate the effect of protein interaction partners on the glycan strand synthesis activity of peptidoglycan polymerases.


Subject(s)
Escherichia coli/enzymology , Fluorescent Dyes/chemistry , Peptidoglycan Glycosyltransferase/isolation & purification , Cell Wall/metabolism , Escherichia coli/metabolism , Fluorometry , Peptidoglycan/biosynthesis , Peptidoglycan Glycosyltransferase/chemistry
17.
J Biol Chem ; 291(29): 14915-26, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27226615

ABSTRACT

Bacterial spores are the most resistant form of life known on Earth and represent a serious problem for (i) bioterrorism attack, (ii) horizontal transmission of microbial pathogens in the community, and (iii) persistence in patients and in a nosocomial environment. Stage II sporulation protein D (SpoIID) is a lytic transglycosylase (LT) essential for sporulation. The LT superfamily is a potential drug target because it is active in essential bacterial processes involving the peptidoglycan, which is unique to bacteria. However, the absence of structural information for the sporulation-specific LT enzymes has hindered mechanistic understanding of SpoIID. Here, we report the first crystal structures with and without ligands of the SpoIID family from two community relevant spore-forming pathogens, Bacillus anthracis and Clostridium difficile. The structures allow us to visualize the overall architecture, characterize the substrate recognition model, identify critical residues, and provide the structural basis for catalysis by this new family of enzymes.


Subject(s)
Bacterial Proteins/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Amino Acid Sequence , Bacillus anthracis/enzymology , Bacillus anthracis/genetics , Bacillus anthracis/pathogenicity , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Catalytic Domain/genetics , Clostridioides difficile/enzymology , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Conserved Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Peptidoglycan Glycosyltransferase/genetics , Phylogeny , Protein Conformation , Sequence Homology, Amino Acid , Spores, Bacterial/enzymology , Structural Homology, Protein
18.
J Bacteriol ; 198(13): 1847-56, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27114466

ABSTRACT

UNLABELLED: SltF was identified previously as an autolysin required for the assembly of flagella in the alphaproteobacteria, but the nature of its peptidoglycan lytic activity remained unknown. Sequence alignment analyses suggest that it could function as either a muramidase, lytic transglycosylase, or ß-N-acetylglucosaminidase. Recombinant SltF from Rhodobacter sphaeroides was purified to apparent homogeneity, and it was demonstrated to function as a lytic transglycosylase based on enzymatic assays involving mass spectrometric analyses. Circular dichroism (CD) analysis determined that it is composed of 83.4% α-structure and 1.48% ß-structure and thus is similar to family 1A lytic transglycosylases. However, alignment of apparent SltF homologs identified in the genome database defined a new subfamily of the family 1 lytic transglycosylases. SltF was demonstrated to be endo-acting, cleaving within chains of peptidoglycan, with optimal activity at pH 7.0. Its activity is modulated by two flagellar rod proteins, FlgB and FlgF: FlgB both stabilizes and stimulates SltF activity, while FlgF inhibits it. Invariant Glu57 was confirmed as the sole catalytic acid/base residue of SltF. IMPORTANCE: The bacterial flagellum is comprised of a basal body, hook, and helical filament, which are connected by a rod structure. With a diameter of approximately 4 nm, the rod is larger than the estimated pore size within the peptidoglycan sacculus, and hence its insertion requires the localized and controlled lysis of this essential cell wall component. In many beta- and gammaproteobacteria, this lysis is catalyzed by the ß-N-acetylglucosaminidase domain of FlgJ. However, FlgJ of the alphaproteobacteria lacks this activity and instead it recruits a separate enzyme, SltF, for this purpose. In this study, we demonstrate that SltF functions as a newly identified class of lytic transglycosylases and that its autolytic activity is uniquely modulated by two rod proteins, FlgB and FlgF.


Subject(s)
Bacterial Proteins/metabolism , Flagella/metabolism , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Rhodobacter sphaeroides/enzymology , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Flagella/chemistry , Flagella/genetics , Molecular Sequence Data , N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/genetics , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/genetics , Rhodobacter sphaeroides/genetics , Rhodobacter sphaeroides/metabolism , Sequence Alignment
19.
Philos Trans R Soc Lond B Biol Sci ; 370(1679)2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26370943

ABSTRACT

Peptidoglycan (PG) is an essential component in the cell wall of nearly all bacteria, forming a continuous, mesh-like structure, called the sacculus, around the cytoplasmic membrane to protect the cell from bursting by its turgor. Although PG synthases, the penicillin-binding proteins (PBPs), have been studied for 70 years, useful in vitro assays for measuring their activities were established only recently, and these provided the first insights into the regulation of these enzymes. Here, we review the current knowledge on the glycosyltransferase and transpeptidase activities of PG synthases. We provide new data showing that the bifunctional PBP1A and PBP1B from Escherichia coli are active upon reconstitution into the membrane environment of proteoliposomes, and that these enzymes also exhibit DD-carboxypeptidase activity in certain conditions. Both novel features are relevant for their functioning within the cell. We also review recent data on the impact of protein-protein interactions and other factors on the activities of PBPs. As an example, we demonstrate a synergistic effect of multiple protein-protein interactions on the glycosyltransferase activity of PBP1B, by its cognate lipoprotein activator LpoB and the essential cell division protein FtsN.


Subject(s)
Escherichia coli Proteins/metabolism , Penicillin-Binding Proteins/metabolism , Peptidoglycan/biosynthesis , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/classification , Kinetics , Models, Molecular , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/classification , Peptidoglycan/chemistry , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/metabolism , Substrate Specificity
20.
Biochem Pharmacol ; 93(2): 141-50, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25462814

ABSTRACT

The glycosyltransferases of family 51 (GT51) catalyze the polymerization of lipid II to form linear glycan chains, which, after cross linking by the transpeptidases, form the net-like peptidoglycan macromolecule. The essential function of the GT makes it an attractive antimicrobial target; therefore a better understanding of its function and its mechanism of interaction with substrates could help in the design and the development of new antibiotics. In this work, we have used a surface plasmon resonance Biacore(®) biosensor, based on an amine derivative of moenomycin A immobilized on a sensor chip surface, to investigate the mechanism of binding of substrate analogous inhibitors to the GT. Addition of increasing concentrations of moenomycin A to the Staphylococcus aureus MtgA led to reduced binding of the protein to the sensor chip as expected. Remarkably, in the presence of low concentrations of the most active disaccharide inhibitors, binding of MtgA to immobilized moenomycin A was found to increase; in contrast competition with moenomycin A occurred only at high concentrations. This finding suggests that at low concentrations, the lipid II analogs bind to the acceptor site and induce a cooperative binding of moenomycin A to the donor site. Our results constitute the first indication of the existence of a positive cooperativity between the acceptor and the donor sites of peptidoglycan GTs. In addition, our study indicates that a modification of two residues (L119N and F120S) within the hydrophobic region of MtgA can yield monodisperse forms of the protein with apparently no change in its secondary structure content, but this is at the expense of the enzyme function.


Subject(s)
Microbial Interactions/physiology , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/metabolism , Staphylococcus aureus/metabolism , Bambermycins/metabolism , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Surface Plasmon Resonance/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...