Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 1163, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859523

ABSTRACT

Autotransporters (ATs) are a large family of bacterial secreted and outer membrane proteins that encompass a wide range of enzymatic activities frequently associated with pathogenic phenotypes. We present the structural and functional characterisation of a subtilase autotransporter, Ssp, from the opportunistic pathogen Serratia marcescens. Although the structures of subtilases have been well documented, this subtilisin-like protein is associated with a 248 residue ß-helix and itself includes three finger-like protrusions around its active site involved in substrate interactions. We further reveal that the activity of the subtilase AT is required for entry into epithelial cells as well as causing cellular toxicity. The Ssp structure not only provides details about the subtilase ATs, but also reveals a common framework and function to more distantly related ATs. As such these findings also represent a significant step forward toward understanding the molecular mechanisms underlying the functional divergence in the large AT superfamily.


Subject(s)
Antineoplastic Agents , Subtilisin , Type V Secretion Systems , Biological Transport
2.
Int J Mol Sci ; 24(6)2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36982580

ABSTRACT

Antigen 43 (Ag43) expression induces aggregation and biofilm formation that has consequences for bacterial colonisation and infection. Ag43 is secreted through the Type 5 subtype "a" secretion system (T5aSS) and is a prototypical member of the family of self-associating autotransporters (SAATs). As a T5aSS protein, Ag43 has a modular architecture comprised of (i) a signal peptide, (ii) a passenger domain that can be subdivided into three subdomains (SL, EJ, and BL), (iii) an autochaperone (AC) domain, and (iv) an outer membrane translocator. The cell-surface SL subdomain is directly involved in the "Velcro-handshake" mechanism resulting in bacterial autoaggregation. Ag43 is considered to have a ubiquitous distribution in E. coli genomes and many strains harbour multiple agn43 genes. However, recent phylogenetic analyses indicated the existence of four distinct Ag43 classes exhibiting different propensities for autoaggregation and interactions. Given the knowledge of the diversity and distribution of Ag43 in E. coli genomes is incomplete, we have performed a thorough in silico investigation across bacterial genomes. Our comprehensive analyses indicate that Ag43 passenger domains cluster in six phylogenetic classes associated with different SL subdomains. The diversity of Ag43 passenger domains is a result of the association of the SL subtypes with two different EJ-BL-AC modules. We reveal that agn43 is almost exclusively present among bacterial species of the Enterobacteriaceae family and essentially in the Escherichia genus (99.6%) but that it is not ubiquitous in E. coli. The gene is typically present as a single copy but up to five copies of agn43 with different combinations of classes can be observed. The presence of agn43 as well as its different classes appeared to differ between Escherichia phylogroups. Strikingly, agn43 is present in 90% of E. coli from E phylogroup. Our results shed light on Ag43 diversity and provide a rational framework for investigating its role in E. coli ecophysiology and physiopathology.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Escherichia coli/physiology , Escherichia coli Proteins/metabolism , Adhesins, Escherichia coli/metabolism , Bacterial Outer Membrane Proteins/genetics , Phylogeny , Prevalence
3.
Antioxidants (Basel) ; 12(2)2023 Feb 04.
Article in English | MEDLINE | ID: mdl-36829940

ABSTRACT

The formation of disulphide bonds is an essential step in the folding of many proteins that enter the secretory pathway; therefore, it is not surprising that eukaryotic and prokaryotic organisms have dedicated enzymatic systems to catalyse this process. In bacteria, one such enzyme is disulphide bond-forming protein A (DsbA), a thioredoxin-like thiol oxidase that catalyses the oxidative folding of proteins required for virulence and fitness. A large body of work on DsbA proteins, particularly Escherichia coli DsbA (EcDsbA), has demonstrated the key role that the Cys30-XX-Cys33 catalytic motif and its unique redox properties play in the thiol oxidase activity of this enzyme. Using mutational and functional analyses, here we identify that a set of charged residues, which form an acidic groove on the non-catalytic face of the enzyme, further modulate the activity of EcDsbA. Our high-resolution structures indicate that these residues form a water-mediated proton wire that can transfer protons from the bulk solvent to the active site. Our results support the view that proton shuffling may facilitate the stabilisation of the buried Cys33 thiolate formed during the redox reaction and promote the correct direction of the EcDsbA-substrate thiol-disulphide exchange. Comparison with other proteins of the same class and proteins of the thioredoxin-superfamily in general suggest that a proton relay system appears to be a conserved catalytic feature among this widespread superfamily of proteins. Furthermore, this study also indicates that the acidic groove of DsbA could be a promising allosteric site to develop novel DsbA inhibitors as antibacterial therapeutics.

4.
Front Immunol ; 13: 921272, 2022.
Article in English | MEDLINE | ID: mdl-35860281

ABSTRACT

Autotransporters are the core component of a molecular nano-machine that delivers cargo proteins across the outer membrane of Gram-negative bacteria. Part of the type V secretion system, this large family of proteins play a central role in controlling bacterial interactions with their environment by promoting adhesion to surfaces, biofilm formation, host colonization and invasion as well as cytotoxicity and immunomodulation. As such, autotransporters are key facilitators of fitness and pathogenesis and enable co-operation or competition with other bacteria. Recent years have witnessed a dramatic increase in the number of autotransporter sequences reported and a steady rise in functional studies, which further link these proteins to multiple virulence phenotypes. In this review we provide an overview of our current knowledge on classical autotransporter proteins, the archetype of this protein superfamily. We also carry out a phylogenetic analysis of their functional domains and present a new classification system for this exquisitely diverse group of bacterial proteins. The sixteen phylogenetic divisions identified establish sensible relationships between well characterized autotransporters and inform structural and functional predictions of uncharacterized proteins, which may guide future research aimed at addressing multiple unanswered aspects in this group of therapeutically important bacterial factors.


Subject(s)
Bacterial Proteins , Type V Secretion Systems , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gram-Negative Bacteria/metabolism , Phylogeny , Type V Secretion Systems/genetics , Type V Secretion Systems/metabolism , Virulence
5.
NPJ Biofilms Microbiomes ; 8(1): 20, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35396507

ABSTRACT

The formation of aggregates and biofilms enhances bacterial colonisation and infection progression by affording protection from antibiotics and host immune factors. Despite these advantages there is a trade-off, whereby bacterial dissemination is reduced. As such, biofilm development needs to be controlled to suit adaptation to different environments. Here we investigate members from one of largest groups of bacterial adhesins, the autotransporters, for their critical role in the assembly of bacterial aggregates and biofilms. We describe the structural and functional characterisation of autotransporter Ag43 variants from different Escherichia coli pathotypes. We show that specific interactions between amino acids on the contacting interfaces of adjacent Ag43 proteins drives a common mode of trans-association that leads to cell clumping. Furthermore, subtle variation of these interactions alters aggregation kinetics and the degree of compacting within cell clusters. Together, our structure-function investigation reveals an underlying molecular basis for variations in the density of bacterial communities.


Subject(s)
Adhesins, Escherichia coli , Escherichia coli Proteins , Adhesins, Escherichia coli/chemistry , Bacterial Adhesion , Biofilms , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
6.
Antioxid Redox Signal ; 35(1): 21-39, 2021 07.
Article in English | MEDLINE | ID: mdl-33607928

ABSTRACT

Aims: Thioredoxin (TRX)-fold proteins are ubiquitous in nature. This redox scaffold has evolved to enable a variety of functions, including redox regulation, protein folding, and oxidative stress defense. In bacteria, the TRX-like disulfide bond (Dsb) family mediates the oxidative folding of multiple proteins required for fitness and pathogenic potential. Conventionally, Dsb proteins have specific redox functions with monomeric and dimeric Dsbs exclusively catalyzing thiol oxidation and disulfide isomerization, respectively. This contrasts with the eukaryotic disulfide forming machinery where the modular TRX protein disulfide isomerase (PDI) mediates thiol oxidation and disulfide reshuffling. In this study, we identified and structurally and biochemically characterized a novel Dsb-like protein from Salmonella enterica termed bovine colonization factor protein H (BcfH) and defined its role in virulence. Results: In the conserved bovine colonization factor (bcf) fimbrial operon, the Dsb-like enzyme BcfH forms a trimeric structure, exceptionally uncommon among the large and evolutionary conserved TRX superfamily. This protein also displays very unusual catalytic redox centers, including an unwound α-helix holding the redox active site and a trans-proline instead of the conserved cis-proline active site loop. Remarkably, BcfH displays both thiol oxidase and disulfide isomerase activities contributing to Salmonella fimbrial biogenesis. Innovation and Conclusion: Typically, oligomerization of bacterial Dsb proteins modulates their redox function, with monomeric and dimeric Dsbs mediating thiol oxidation and disulfide isomerization, respectively. This study demonstrates a further structural and functional malleability in the TRX-fold protein family. BcfH trimeric architecture and unconventional catalytic sites permit multiple redox functions emulating in bacteria the eukaryotic PDI dual oxidoreductase activity. Antioxid. Redox Signal. 35, 21-39.


Subject(s)
Bacterial Proteins/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Protein Disulfide-Isomerases/metabolism , Salmonella enterica/pathogenicity , Bacterial Proteins/ultrastructure , Operon/genetics , Oxidation-Reduction , Oxidative Stress/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/ultrastructure , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/ultrastructure , Protein Folding , Protein Structure, Tertiary , Salmonella enterica/enzymology , Salmonella enterica/genetics , Salmonella enterica/metabolism , Thioredoxins/metabolism
7.
Angew Chem Int Ed Engl ; 59(25): 10129-10135, 2020 06 15.
Article in English | MEDLINE | ID: mdl-31826303

ABSTRACT

Environmental polarity is an important factor that drives biomolecular interactions to regulate cell function. Herein, a general method of using the fluorogenic probe NTPAN-MI is reported to quantify the subcellular polarity change in response to protein unfolding. NTPAN-MI fluorescence is selectively activated upon labeling unfolded proteins with exposed thiols, thereby reporting on the extent of proteostasis. NTPAN-MI also reveals the collapse of the host proteome caused by influenza A virus infection. The emission profile of NTPAN-MI contains information of the local polarity of the unfolded proteome, which can be resolved through spectral phasor analysis. Under stress conditions that disrupt different checkpoints of protein quality control, distinct patterns of dielectric constant distribution in the cytoplasm can be observed. However, in the nucleus, the unfolded proteome was found to experience a more hydrophilic environment across all the stress conditions, indicating the central role of nucleus in the stress response process.


Subject(s)
Proteome , Unfolded Protein Response/genetics , Cell Nucleus , Cytoplasm , Drug Design , Fluorescent Dyes/chemical synthesis , HEK293 Cells , Humans
8.
Sci Rep ; 9(1): 18019, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792243

ABSTRACT

The ClpP protease is found in all kingdoms of life, from bacteria to humans. In general, this protease forms a homo-oligomeric complex composed of 14 identical subunits, which associates with its cognate ATPase in a symmetrical manner. Here we show that, in contrast to this general architecture, the Clp protease from Mycobacterium smegmatis (Msm) forms an asymmetric hetero-oligomeric complex ClpP1P2, which only associates with its cognate ATPase through the ClpP2 ring. Our structural and functional characterisation of this complex demonstrates that asymmetric docking of the ATPase component is controlled by both the composition of the ClpP1 hydrophobic pocket (Hp) and the presence of a unique C-terminal extension in ClpP1 that guards this Hp. Our structural analysis of MsmClpP1 also revealed openings in the side-walls of the inactive tetradecamer, which may represent sites for product egress.


Subject(s)
Bacterial Proteins/ultrastructure , Endopeptidase Clp/ultrastructure , Mycobacterium smegmatis/metabolism , Protein Multimerization , Protein Subunits/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/ultrastructure , Bacterial Proteins/metabolism , Crystallography, X-Ray , Endopeptidase Clp/metabolism , Molecular Docking Simulation , Protein Structure, Quaternary , Proteolysis
9.
J Biol Chem ; 294(44): 15876-15888, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31444272

ABSTRACT

The human pathogen Salmonella enterica serovar Typhimurium (S Typhimurium) contains a complex disulfide bond (Dsb) catalytic machinery. This machinery encompasses multiple Dsb thiol-disulfide oxidoreductases that mediate oxidative protein folding and a less-characterized suppressor of copper sensitivity (scs) gene cluster, associated with increased tolerance to copper. To better understand the function of the Salmonella Scs system, here we characterized two of its key components, the membrane protein ScsB and the periplasmic protein ScsC. Our results revealed that these two proteins form a redox pair in which the electron transfer from the periplasmic domain of ScsB (n-ScsB) to ScsC is thermodynamically driven. We also demonstrate that the Scs reducing pathway remains separate from the Dsb oxidizing pathways and thereby avoids futile redox cycles. Additionally, we provide new insight into the molecular mechanism underlying Scs-mediated copper tolerance in Salmonella We show that both ScsB and ScsC can bind toxic copper(I) with femtomolar affinities and transfer it to the periplasmic copper metallochaperone CueP. Our results indicate that the Salmonella Scs machinery has evolved a dual mode of action, capable of transferring reducing power to the oxidizing periplasm and protecting against copper stress by cooperating with the cue regulon, a major copper resistance mechanism in Salmonella. Overall, these findings expand our understanding of the functional diversity of Dsb-like systems, ranging from those mediating oxidative folding of proteins required for infection to those contributing to defense mechanisms against oxidative stress and copper toxicity, critical traits for niche adaptation and survival.


Subject(s)
Bacterial Proteins/metabolism , Copper/metabolism , Drug Resistance, Bacterial , Metallochaperones/metabolism , NADH, NADPH Oxidoreductases/metabolism , Salmonella/metabolism , Adaptation, Physiological , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Copper/toxicity , Metallochaperones/chemistry , Metallochaperones/genetics , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/genetics , Oxidation-Reduction , Periplasm/metabolism , Protein Binding , Protein Folding , Regulon , Salmonella/drug effects , Salmonella/enzymology
10.
Nat Commun ; 10(1): 1967, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036849

ABSTRACT

Autotransporters are the largest family of outer membrane and secreted proteins in Gram-negative bacteria. Most autotransporters are localised to the bacterial surface where they promote colonisation of host epithelial surfaces. Here we present the crystal structure of UpaB, an autotransporter that is known to contribute to uropathogenic E. coli (UPEC) colonisation of the urinary tract. We provide evidence that UpaB can interact with glycosaminoglycans and host fibronectin. Unique modifications to its core ß-helical structure create a groove on one side of the protein for interaction with glycosaminoglycans, while the opposite face can bind fibronectin. Our findings reveal far greater diversity in the autotransporter ß-helix than previously thought, and suggest that this domain can interact with host macromolecules. The relevance of these interactions during infection remains unclear.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Glycosaminoglycans/metabolism , Uropathogenic Escherichia coli/metabolism , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Virulence Factors/chemistry , Virulence Factors/metabolism
11.
J Biol Chem ; 293(43): 16559-16571, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30181210

ABSTRACT

The worldwide incidence of neisserial infections, particularly gonococcal infections, is increasingly associated with antibiotic-resistant strains. In particular, extensively drug-resistant Neisseria gonorrhoeae strains that are resistant to third-generation cephalosporins are a major public health concern. There is a pressing clinical need to identify new targets for the development of antibiotics effective against Neisseria-specific processes. In this study, we report that the bacterial disulfide reductase DsbD is highly prevalent and conserved among Neisseria spp. and that this enzyme is essential for survival of N. gonorrhoeae DsbD is a membrane-bound protein that consists of two periplasmic domains, n-DsbD and c-DsbD, which flank the transmembrane domain t-DsbD. In this work, we show that the two functionally essential periplasmic domains of Neisseria DsbD catalyze electron transfer reactions through unidirectional interdomain interactions, from reduced c-DsbD to oxidized n-DsbD, and that this process is not dictated by their redox potentials. Structural characterization of the Neisseria n- and c-DsbD domains in both redox states provides evidence that steric hindrance reduces interactions between the two periplasmic domains when n-DsbD is reduced, thereby preventing a futile redox cycle. Finally, we propose a conserved mechanism of electron transfer for DsbD and define the residues involved in domain-domain recognition. Inhibitors of the interaction of the two DsbD domains have the potential to be developed as anti-neisserial agents.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Disulfides/metabolism , Neisseria gonorrhoeae/enzymology , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Conformation , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Disulfides/chemistry , Models, Molecular , Oxidation-Reduction , Protein Domains
12.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 1): 31-38, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29372905

ABSTRACT

The membrane protein DsbD is a reductase that acts as an electron hub, translocating reducing equivalents from cytoplasmic thioredoxin to a number of periplasmic substrates involved in oxidative protein folding, cytochrome c maturation and oxidative stress defence. DsbD is a multi-domain protein consisting of a transmembrane domain (t-DsbD) flanked by two periplasmic domains (n-DsbD and c-DsbD). Previous studies have shown that DsbD is required for the survival of the obligate human pathogen Neisseria meningitidis. To help understand the structural and functional aspects of N. meningitidis DsbD, the two periplasmic domains which are required for electron transfer are being studied. Here, the expression, purification and biophysical properties of n-NmDsbD and c-NmDsbD are described. The crystallization and crystallographic analysis of n-NmDsbD and c-NmDsbD are also described in both redox states, which differ only in the presence or absence of a disulfide bond but which crystallized in completely different conditions. Crystals of n-NmDsbDOx, n-NmDsbDRed, c-NmDsbDOx and c-NmDsbDRed diffracted to 2.3, 1.6, 2.3 and 1.7 Šresolution and belonged to space groups P213, P321, P41 and P1211, respectively.


Subject(s)
Bacterial Proteins/chemistry , Neisseria meningitidis/enzymology , Oxidoreductases/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Biophysical Phenomena , Crystallization , Crystallography, X-Ray , Electron Transport , Models, Molecular , Neisseria meningitidis/genetics , Oxidoreductases/genetics , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Scattering, Small Angle , X-Ray Diffraction
13.
Sci Rep ; 8(1): 358, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321677

ABSTRACT

Hendra virus (HeV) is a paramyxovirus that causes lethal disease in humans, for which no vaccine or antiviral agent is available. HeV V protein is central to pathogenesis through its ability to interact with cytoplasmic host proteins, playing key antiviral roles. Here we use immunoprecipitation, siRNA knockdown and confocal laser scanning microscopy to show that HeV V shuttles to and from the nucleus through specific host nuclear transporters. Spectroscopic and small angle X-ray scattering studies reveal HeV V undergoes a disorder-to-order transition upon binding to either importin α/ß1 or exportin-1/Ran-GTP, dependent on the V N-terminus. Importantly, we show that specific inhibitors of nuclear transport prevent interaction with host transporters, and reduce HeV infection. These findings emphasize the critical role of host-virus interactions in HeV infection, and potential use of compounds targeting nuclear transport, such as the FDA-approved agent ivermectin, as anti-HeV agents.


Subject(s)
Hendra Virus/physiology , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Host-Pathogen Interactions , Nucleocytoplasmic Transport Proteins/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Nucleus/metabolism , Drug Discovery , Gene Knockdown Techniques , Hendra Virus/drug effects , Henipavirus Infections/genetics , Humans , Karyopherins/chemistry , Karyopherins/genetics , Karyopherins/metabolism , Models, Molecular , Molecular Conformation , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/metabolism , Exportin 1 Protein
14.
Antioxid Redox Signal ; 29(7): 653-666, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29237285

ABSTRACT

AIMS: DsbA catalyzes disulfide bond formation in secreted and outer membrane proteins in bacteria. In pathogens, DsbA is a major facilitator of virulence constituting a target for antivirulence antimicrobial development. However, many pathogens encode multiple and diverse DsbA enzymes for virulence factor folding during infection. The aim of this study was to determine whether our recently identified inhibitors of Escherichia coli K-12 DsbA can inhibit the diverse DsbA enzymes found in two important human pathogens and attenuate their virulence. RESULTS: DsbA inhibitors from two chemical classes (phenylthiophene and phenoxyphenyl derivatives) inhibited the virulence of uropathogenic E. coli and Salmonella enterica serovar Typhimurium, encoding two and three diverse DsbA homologues, respectively. Inhibitors blocked the virulence of dsbA null mutants complemented with structurally diverse DsbL and SrgA, suggesting that they were not selective for prototypical DsbA. Structural characterization of DsbA-inhibitor complexes showed that compounds from each class bind in a similar region of the hydrophobic groove adjacent to the Cys30-Pro31-His32-Cys33 (CPHC) active site. Modeling of DsbL- and SrgA-inhibitor interactions showed that these accessory enzymes could accommodate the inhibitors in their different hydrophobic grooves, supporting our in vivo findings. Further, we identified highly conserved residues surrounding the active site for 20 diverse bacterial DsbA enzymes, which could be exploited in developing inhibitors with a broad spectrum of activity. Innovation and Conclusion: We have developed tools to analyze the specificity of DsbA inhibitors in bacterial pathogens encoding multiple DsbA enzymes. This work demonstrates that DsbA inhibitors can be developed to target diverse homologues found in bacteria. Antioxid. Redox Signal. 29, 653-666.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli K12/drug effects , Escherichia coli Proteins/antagonists & inhibitors , Protein Disulfide-Isomerases/antagonists & inhibitors , Thiophenes/pharmacology , Anti-Bacterial Agents/chemistry , Enzyme Inhibitors/chemistry , Escherichia coli K12/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Microbial Sensitivity Tests , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Thiophenes/chemistry
15.
Proteomics ; 17(23-24)2017 Dec.
Article in English | MEDLINE | ID: mdl-28665015

ABSTRACT

Most bacteria produce adhesion molecules to facilitate the interaction with host cells and establish successful infections. An important group of bacterial adhesins belong to the autotransporter (AT) superfamily, the largest group of secreted and outer membrane proteins in Gram-negative bacteria. AT adhesins possess diverse functions that facilitate bacterial colonisation, survival and persistence, and as such are often associated with increased bacterial fitness and pathogenic potential. In this review, we will describe AIDA-I type AT adhesins, which comprise the biggest and most diverse group in the AT family. We will focus on Escherichia coli proteins and define general aspects of their biogenesis, distribution, structural properties and key roles in infection.


Subject(s)
Escherichia coli Infections/metabolism , Escherichia coli/pathogenicity , Adhesins, Escherichia coli/metabolism , Bacterial Adhesion , Escherichia coli Infections/microbiology , Gene Expression Regulation, Bacterial
16.
Methods Mol Biol ; 1549: 209-220, 2017.
Article in English | MEDLINE | ID: mdl-27975294

ABSTRACT

The dramatic increase in the number of protein sequences and structures deposited in biological databases has led to the development of many bioinformatics tools and programs to manage, validate, compare, and interpret this large volume of data. In addition, powerful tools are being developed to use this sequence and structural data to facilitate protein classification and infer biological function of newly identified proteins. This chapter covers freely available bioinformatics resources on the World Wide Web that are commonly used for protein structure analysis.


Subject(s)
Computational Biology/methods , Models, Molecular , Protein Conformation , Proteins/chemistry , Software , Databases, Protein , Ligands , Protein Binding , Reproducibility of Results , Structure-Activity Relationship , User-Computer Interface , Web Browser
17.
Structure ; 24(8): 1282-1291, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27427481

ABSTRACT

Dihydrodipicolinate synthase (DHDPS) catalyzes the first committed step in the lysine biosynthesis pathway of bacteria. The pathway can be regulated by feedback inhibition of DHDPS through the allosteric binding of the end product, lysine. The current dogma states that DHDPS from Gram-negative bacteria are inhibited by lysine but orthologs from Gram-positive species are not. The 1.65-Å resolution structure of the Gram-negative Legionella pneumophila DHDPS and the 1.88-Å resolution structure of the Gram-positive Streptococcus pneumoniae DHDPS bound to lysine, together with comprehensive functional analyses, show that this dogma is incorrect. We subsequently employed our crystallographic data with bioinformatics, mutagenesis, enzyme kinetics, and microscale thermophoresis to reveal that lysine-mediated inhibition is not defined by Gram staining, but by the presence of a His or Glu at position 56 (Escherichia coli numbering). This study has unveiled the molecular determinants defining lysine-mediated allosteric inhibition of bacterial DHDPS.


Subject(s)
Escherichia coli/enzymology , Feedback, Physiological , Hydro-Lyases/chemistry , Legionella pneumophila/enzymology , Lysine/chemistry , Streptococcus pneumoniae/enzymology , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Gene Expression , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Kinetics , Legionella pneumophila/genetics , Lysine/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Streptococcus pneumoniae/genetics , Substrate Specificity
18.
Molecules ; 21(7)2016 Jul 16.
Article in English | MEDLINE | ID: mdl-27438817

ABSTRACT

Recent years have witnessed a dramatic increase in bacterial antimicrobial resistance and a decline in the development of novel antibiotics. New therapeutic strategies are urgently needed to combat the growing threat posed by multidrug resistant bacterial infections. The Dsb disulfide bond forming pathways are potential targets for the development of antimicrobial agents because they play a central role in bacterial pathogenesis. In particular, the DsbA/DsbB system catalyses disulfide bond formation in a wide array of virulence factors, which are essential for many pathogens to establish infections and cause disease. These redox enzymes are well placed as antimicrobial targets because they are taxonomically widespread, share low sequence identity with human proteins, and many years of basic research have provided a deep molecular understanding of these systems in bacteria. In this review, we discuss disulfide bond catalytic pathways in bacteria and their significance in pathogenesis. We also review the use of different approaches to develop inhibitors against Dsb proteins as potential anti-virulence agents, including fragment-based drug discovery, high-throughput screening and other structure-based drug discovery methods.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Virulence Factors/antagonists & inhibitors , Virulence/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Computer Simulation , Drug Discovery , Escherichia coli K12/drug effects , Escherichia coli K12/metabolism , High-Throughput Screening Assays , Humans , Models, Molecular , Oxidative Stress/drug effects , Quantitative Structure-Activity Relationship
19.
mBio ; 7(2): e02046, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26980835

ABSTRACT

UNLABELLED: Urinary tract infection (UTI) is a disease of extremely high incidence in both community and nosocomial settings. UTIs cause significant morbidity and mortality, with approximately 150 million cases globally per year. Uropathogenic Escherichia coli (UPEC) is the primary cause of UTI and is generally treated empirically. However, the rapidly increasing incidence of UTIs caused by multidrug-resistant UPEC strains has led to limited available treatment options and highlights the urgent need to develop alternative treatment and prevention strategies. In this study, we performed a comprehensive analysis to define the regulation, structure, function, and immunogenicity of recently identified UPEC vaccine candidate C1275 (here referred to as IrmA). We showed that the irmA gene is highly prevalent in UPEC, is cotranscribed with the biofilm-associated antigen 43 gene, and is regulated by the global oxidative stress response OxyR protein. Localization studies identified IrmA in the UPEC culture supernatant. We determined the structure of IrmA and showed that it adopts a unique domain-swapped dimer architecture. The dimeric structure of IrmA displays similarity to those of human cytokine receptors, including the interleukin-2 receptor (IL-2R), interleukin-4 receptor (IL-4R), and interleukin-10 receptor (IL-10R) binding domains, and we showed that purified IrmA can bind to their cognate cytokines. Finally, we showed that plasma from convalescent urosepsis patients contains high IrmA antibody titers, demonstrating the strong immunogenicity of IrmA. Taken together, our results indicate that IrmA may play an important role during UPEC infection. IMPORTANCE: Uropathogenic E. coli (UPEC) is the primary cause of urinary tract infection (UTI), a disease of major significance to human health. Globally, the incidence of UPEC-mediated UTI is strongly associated with increasing antibiotic resistance, making this extremely common infection a major public health concern. In this report, we describe the regulatory, structural, functional, and immunogenic properties of a candidate UPEC vaccine antigen, IrmA. We demonstrate that IrmA is a small UPEC protein that forms a unique domain-swapped dimer with structural mimicry to several human cytokine receptors. We also show that IrmA binds to IL-2, IL-4, and IL-10, is strongly immunogenic in urosepsis patients, and is coexpressed with factors associated with biofilm formation. Overall, this work suggests a potential novel contribution for IrmA in UPEC infection.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Molecular Mimicry/genetics , Receptors, Interleukin/chemistry , Receptors, Interleukin/genetics , Uropathogenic Escherichia coli/chemistry , Uropathogenic Escherichia coli/genetics , Antibodies, Bacterial/blood , Cytokines/metabolism , Escherichia coli Proteins/metabolism , Humans , Uropathogenic Escherichia coli/immunology
20.
Proc Natl Acad Sci U S A ; 111(1): 457-62, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24335802

ABSTRACT

Aggregation and biofilm formation are critical mechanisms for bacterial resistance to host immune factors and antibiotics. Autotransporter (AT) proteins, which represent the largest group of outer-membrane and secreted proteins in Gram-negative bacteria, contribute significantly to these phenotypes. Despite their abundance and role in bacterial pathogenesis, most AT proteins have not been structurally characterized, and there is a paucity of detailed information with regard to their mode of action. Here we report the structure-function relationships of Antigen 43 (Ag43a), a prototypic self-associating AT protein from uropathogenic Escherichia coli. The functional domain of Ag43a displays a twisted L-shaped ß-helical structure firmly stabilized by a 3D hydrogen-bonded scaffold. Notably, the distinctive Ag43a L shape facilitates self-association and cell aggregation. Combining all our data, we define a molecular "Velcro-like" mechanism of AT-mediated bacterial clumping, which can be tailored to fit different bacterial lifestyles such as the formation of biofilms.


Subject(s)
Adhesins, Bacterial/chemistry , Adhesins, Escherichia coli/chemistry , Biofilms , Uropathogenic Escherichia coli/metabolism , Antigens, Bacterial/chemistry , Biological Transport , Cloning, Molecular , Crystallography, X-Ray , Hydrogen Bonding , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Urinary Tract Infections/microbiology , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...