Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 970, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35217634

ABSTRACT

Many bacteria and archaea possess a two-dimensional protein array, or S-layer, that covers the cell surface and plays crucial roles in cell physiology. Here, we report the crystal structure of SlpA, the main S-layer protein of the bacterial pathogen Clostridioides difficile, and use electron microscopy to study S-layer organisation and assembly. The SlpA crystal lattice mimics S-layer assembly in the cell, through tiling of triangular prisms above the cell wall, interlocked by distinct ridges facing the environment. Strikingly, the array is very compact, with pores of only ~10 Å in diameter, compared to other S-layers (30-100 Å). The surface-exposed flexible ridges are partially dispensable for overall structure and assembly, although a mutant lacking this region becomes susceptible to lysozyme, an important molecule in host defence. Thus, our work gives insights into S-layer organisation and provides a basis for development of C. difficile-specific therapeutics.


Subject(s)
Clostridioides difficile , Bacterial Proteins/metabolism , Cell Wall/metabolism , Clostridioides difficile/genetics
2.
Sci Rep ; 11(1): 3244, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547340

ABSTRACT

Clostridioides difficile is the leading cause of nosocomial antibiotic-associated diarrhoea worldwide, yet there is little insight into intestinal tract colonisation and relapse. In many bacterial species, the secondary messenger cyclic-di-GMP mediates switching between planktonic phase, sessile growth and biofilm formation. We demonstrate that c-di-GMP promotes early biofilm formation in C. difficile and that four cell surface proteins contribute to biofilm formation, including two c-di-GMP regulated; CD2831 and CD3246, and two c-di-GMP-independent; CD3392 and CD0183. We demonstrate that C. difficile biofilms are composed of extracellular DNA (eDNA), cell surface and intracellular proteins, which form a protective matrix around C. difficile vegetative cells and spores, as shown by a protective effect against the antibiotic vancomycin. We demonstrate a positive correlation between biofilm biomass, sporulation frequency and eDNA abundance in all five C. difficile lineages. Strains 630 (RT012), CD305 (RT023) and M120 (RT078) contain significantly more eDNA in their biofilm matrix than strains R20291 (RT027) and M68 (RT017). DNase has a profound effect on biofilm integrity, resulting in complete disassembly of the biofilm matrix, inhibition of biofilm formation and reduced spore germination. The addition of exogenous DNase could be exploited in treatment of C. difficile infection and relapse, to improve antibiotic efficacy.


Subject(s)
Bacterial Proteins/metabolism , Biofilms , Clostridioides difficile/physiology , Cyclic GMP/analogs & derivatives , DNA, Bacterial/metabolism , Biofilms/growth & development , Clostridioides difficile/ultrastructure , Clostridium Infections/microbiology , Cyclic GMP/metabolism , Humans
3.
J Biol Chem ; 293(47): 18123-18137, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30275012

ABSTRACT

Clostridium difficile is a bacterial pathogen that causes major health challenges worldwide. It has a well-characterized surface (S)-layer, a para-crystalline proteinaceous layer surrounding the cell wall. In many bacterial and archaeal species, the S-layer is glycosylated, but no such modifications have been demonstrated in C. difficile. Here, we show that a C. difficile strain of S-layer cassette type 11, Ox247, has a complex glycan attached via an O-linkage to Thr-38 of the S-layer low-molecular-weight subunit. Using MS and NMR, we fully characterized this glycan. We present evidence that it is composed of three domains: (i) a core peptide-linked tetrasaccharide with the sequence -4-α-Rha-3-α-Rha-3-α-Rha-3-ß-Gal-peptide; (ii) a repeating pentasaccharide with the sequence -4-ß-Rha-4-α-Glc-3-ß-Rha-4-(α-Rib-3-)ß-Rha-; and (iii) a nonreducing end-terminal 2,3 cyclophosphoryl-rhamnose attached to a ribose-branched sub-terminal rhamnose residue. The Ox247 genome contains a 24-kb locus containing genes for synthesis and protein attachment of this glycan. Mutations in genes within this locus altered or completely abrogated formation of this glycan, and their phenotypes suggested that this S-layer modification may affect sporulation, cell length, and biofilm formation of C. difficile In summary, our findings indicate that the S-layer protein of SLCT-11 strains displays a complex glycan and suggest that this glycan is required for C. difficile sporulation and control of cell shape, a discovery with implications for the development of antimicrobials targeting the S-layer.


Subject(s)
Clostridioides difficile/metabolism , Membrane Glycoproteins/metabolism , Polysaccharides/metabolism , Spores, Bacterial/growth & development , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Glycosylation , Mass Spectrometry , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Molecular Weight , Polysaccharides/chemistry , Protein Conformation , Spores, Bacterial/genetics , Spores, Bacterial/metabolism
4.
mBio ; 9(1)2018 01 16.
Article in English | MEDLINE | ID: mdl-29339424

ABSTRACT

A major virulence factor in Clostridium sordellii-mediated infection is the toxin TcsL, which is encoded within a region of the genome called the pathogenicity locus (PaLoc). C. sordellii isolates carry the PaLoc on the pCS1 family of plasmids, of which there are four characterized members. Here, we determined the potential mobility of pCS1 plasmids and characterized a fifth unique pCS1 member. Using a derivative of the pCS1-1 plasmid from strain ATCC 9714 which had been marked with the ermB erythromycin resistance gene, conjugative transfer into a recipient C. sordellii isolate, R28058, was demonstrated. Bioinformatic analysis of pCS1-1 identified a novel conjugation gene cluster defined as the C. sordellii transfer (cst) locus. Interruption of genes within the cst locus resulted in loss of pCS1-1 transfer, which was restored upon complementation in trans These studies provided clear evidence that genes within the cst locus are essential for the conjugative transfer of pCS1-1. The cst locus is present on all pCS1 subtypes, and homologous loci were identified on toxin-encoding plasmids from Clostridium perfringens and Clostridium botulinum and also carried within genomes of Clostridium difficile isolates, indicating that it is a widespread clostridial conjugation locus. The results of this study have broad implications for the dissemination of toxin genes and, potentially, antibiotic resistance genes among members of a diverse range of clostridial pathogens, providing these microorganisms with a survival advantage within the infected host.IMPORTANCEC. sordellii is a bacterial pathogen that causes severe infections in humans and animals, with high mortality rates. While the pathogenesis of C. sordellii infections is not well understood, it is known that the toxin TcsL is an important virulence factor. Here, we have shown the ability of a plasmid carrying the tcsL gene to undergo conjugative transfer between distantly related strains of C. sordellii, which has far-reaching implications for the ability of C. sordellii to acquire the capacity to cause disease. Plasmids that carry tcsL encode a previously uncharacterized conjugation locus, and individual genes within this locus were shown to be required for conjugative transfer. Furthermore, homologues on toxin plasmids from other clostridial species were identified, indicating that this region represents a novel clostridial conjugation locus. The results of this study have broad implications for the dissemination of virulence genes among members of a diverse range of clostridial pathogens.


Subject(s)
Clostridium sordellii/genetics , Conjugation, Genetic , Gene Transfer, Horizontal , Genetic Loci , Plasmids , Computational Biology , Genes, Bacterial , Multigene Family
5.
Sci Rep ; 7(1): 9204, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28835650

ABSTRACT

Clostridium difficile is a gastrointestinal pathogen but how the bacterium colonises this niche is still little understood. Sortase enzymes covalently attach specific bacterial proteins to the peptidoglycan cell wall and are often involved in colonisation by pathogens. Here we show C. difficile proteins CD2537 and CD3392 are functional substrates of sortase SrtB. Through manipulation of the C-terminal regions of these proteins we show the SPKTG motif is essential for covalent attachment to the cell wall. Two additional putative substrates, CD0183 which contains an SPSTG motif, and CD2768 which contains an SPQTG motif, are not cleaved or anchored to the cell wall by sortase. Finally, using an in vivo asymmetric cleavage assay, we show that despite containing a conserved SPKTG motif, in the absence of SrtB these proteins are localised to disparate cellular compartments.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Cysteine Endopeptidases/metabolism , Amino Acid Motifs , Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Cell Wall/metabolism , Cysteine Endopeptidases/chemistry , Protein Binding , Protein Transport , Proteolysis , Substrate Specificity
6.
Sci Rep ; 6: 23463, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26996606

ABSTRACT

The uses of fluorescent reporters derived from green fluorescent protein have proved invaluable for the visualisation of biological processes in bacteria grown under aerobic conditions. However, their requirement for oxygen has limited their application in obligate anaerobes such as Clostridium difficile. Fluorescent proteins derived from Light, Oxygen or Voltage sensing (LOV) domains have been shown to bridge this limitation, but their utility as translational fusions to monitor protein expression and localisation in a strict anaerobic bacterium has not been reported. Here we demonstrate the utility of phiLOV in three species of Clostridium and its application as a marker of real-time protein translation and dynamics through genetic fusion with the cell division protein, FtsZ. Time lapse microscopy of dividing cells suggests that Z ring assembly arises through the extension of the FtsZ arc starting from one point on the circumference. Furthermore, through incorporation of phiLOV into the flagella subunit, FliC, we show the potential of bacterial LOV-based fusion proteins to be successfully exported to the extracellular environment.


Subject(s)
Biosensing Techniques/methods , Clostridioides difficile/genetics , Molecular Imaging/methods , Optical Imaging/methods , Protein Biosynthesis , Bacterial Proteins/biosynthesis , Clostridioides difficile/metabolism , Cytoskeletal Proteins/biosynthesis , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Protein Transport
7.
Mol Microbiol ; 100(1): 204-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26690930

ABSTRACT

Engulfment of the forespore by the mother cell is a universal feature of endosporulation. In Bacillus subtilis, the forespore protein SpoIIQ and the mother cell protein SpoIIIAH form a channel, essential for endosporulation, through which the developing spore is nurtured. The two proteins also form a backup system for engulfment. Unlike in B. subtilis, SpoIIQ of Clostridium difficile has intact LytM zinc-binding motifs. We show that spoIIQ or spoIIIAH deletion mutants of C. difficile result in anomalous engulfment, and that disruption of the SpoIIQ LytM domain via a single amino acid substitution (H120S) impairs engulfment differently. SpoIIQ and SpoIIQ(H120S) interact with SpoIIIAH throughout engulfment. SpoIIQ, but not SpoIIQ(H120S) , binds Zn(2+) , and metal absence alters the SpoIIQ-SpoIIIAH complex in vitro. Possibly, SpoIIQ(H120S) supports normal engulfment in some cells but not a second function of the complex, required following engulfment completion. We show that cells of the spoIIQ or spoIIIAH mutants that complete engulfment are impaired in post-engulfment, forespore and mother cell-specific gene expression, suggesting a channel-like function. Both engulfment and a channel-like function may be ancestral functions of SpoIIQ-SpoIIIAH while the requirement for engulfment was alleviated through the emergence of redundant mechanisms in B. subtilis and related organisms.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/physiology , Gene Expression Regulation, Bacterial , Spores, Bacterial , Amino Acid Sequence , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Protein Binding , Protein Interaction Domains and Motifs , Sequence Deletion
8.
BMC Microbiol ; 15: 280, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26679502

ABSTRACT

BACKGROUND: The symptoms of Clostridium difficile infection are mediated primarily by two toxins, TcdA and TcdB, the expression of which is governed by a multitude of factors including nutrient availability, growth phase and cell stress. Several global regulators have been implicated in the regulation of toxin expression, such as CcpA and CodY. RESULTS: During attempts to insertionally inactivate a putative secondary cell wall polysaccharide synthesis gene, we obtained several mutants containing off-target insertions. One mutant displayed an unusual branched colony morphology and was investigated further. Marker recovery revealed an insertion in mfd, a gene encoding a transcription-coupled repair factor. The mfd mutant exhibited pleiotropic effects, in particular increased expression of both toxin A and B (TcdA and TcdB) compared to the parental strain. Western blotting and cellular cytotoxicity assays revealed increased expression across all time points over a 24 h period, with inactivation of mfd resulting in at least a 10 fold increase in cell cytotoxicity. qRT-PCR demonstrated the upregulation of both toxins occurred on a transcriptional level. All effects of the mfd mutation were complemented by a plasmid-encoded copy of mfd, showing the effects are not due to polar effects of the intron insertion or to second site mutations. CONCLUSIONS: This study adds Mfd to the repertoire of factors involved in regulation of toxin expression in Clostridium difficile. Mfd is known to remove RNA polymerase molecules from transcriptional sites where it has stalled due to repressor action, preventing transcriptional read through. The consistently high levels of toxin in the C. difficile mfd mutant indicate this process is inefficient leading to transcriptional de-repression.


Subject(s)
Bacterial Proteins/genetics , Bacterial Toxins/genetics , Clostridioides difficile/genetics , Enterotoxins/genetics , Mutagenesis, Insertional/genetics , Transcription Factors/genetics , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Cell Survival/drug effects , Cell Wall/genetics , Chlorocebus aethiops , Clostridioides difficile/metabolism , Clostridioides difficile/physiology , Enterotoxins/metabolism , Enterotoxins/toxicity , Gene Expression Regulation, Bacterial , HT29 Cells , Humans , Mutagenesis, Insertional/immunology , Transcription, Genetic , Up-Regulation , Vero Cells
9.
Chem Biol ; 22(11): 1562-1573, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26584780

ABSTRACT

Bacterial lipoproteins are surface exposed, anchored to the membrane by S-diacylglyceryl modification of the N-terminal cysteine thiol. They play important roles in many essential cellular processes and in bacterial pathogenesis. For example, Clostridium difficile is a Gram-positive anaerobe that causes severe gastrointestinal disease; however, its lipoproteome remains poorly characterized. Here we describe the application of metabolic tagging with alkyne-tagged lipid analogs, in combination with quantitative proteomics, to profile protein lipidation across diverse C. difficile strains and on inactivation of specific components of the lipoprotein biogenesis pathway. These studies provide the first comprehensive map of the C. difficile lipoproteome, demonstrate the existence of two active lipoprotein signal peptidases, and provide insights into lipoprotein function, implicating the lipoproteome in transmission of this pathogen.


Subject(s)
Clostridioides difficile/physiology , Lipoproteins/metabolism , Proteome/analysis , Proteomics , Alkynes/chemistry , Biocatalysis , Chromatography, High Pressure Liquid , Lipoproteins/chemistry , Myristic Acid/chemistry , Spores, Bacterial/metabolism , Tandem Mass Spectrometry
10.
J Biol Chem ; 290(40): 24453-69, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26283789

ABSTRACT

In Gram-positive pathogens, surface proteins may be covalently anchored to the bacterial peptidoglycan by sortase, a cysteine transpeptidase enzyme. In contrast to other Gram-positive bacteria, only one single sortase enzyme, SrtB, is conserved between strains of Clostridium difficile. Sortase-mediated peptidase activity has been reported in vitro, and seven potential substrates have been identified. Here, we demonstrate the functionality of sortase in C. difficile. We identify two sortase-anchored proteins, the putative adhesins CD2831 and CD3246, and determine the cell wall anchor structure of CD2831. The C-terminal PPKTG sorting motif of CD2831 is cleaved between the threonine and glycine residues, and the carboxyl group of threonine is amide-linked to the side chain amino group of diaminopimelic acid within the peptidoglycan peptide stem. We show that CD2831 protein levels are elevated in the presence of high intracellular cyclic diGMP (c-diGMP) concentrations, in agreement with the control of CD2831 expression by a c-diGMP-dependent type II riboswitch. Low c-diGMP levels induce the release of CD2831 and presumably CD3246 from the surface of cells. This regulation is mediated by proteolytic cleavage of CD2831 and CD3246 by the zinc metalloprotease ZmpI, whose expression is controlled by a type I c-diGMP riboswitch. These data reveal a novel regulatory mechanism for expression of two sortase substrates by the secondary messenger c-diGMP, on which surface anchoring is dependent.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/metabolism , Clostridioides difficile/metabolism , Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial , Metalloproteases/metabolism , Peptide Hydrolases/metabolism , Adhesins, Bacterial/metabolism , Amino Acid Motifs , Aminoacyltransferases/metabolism , Cell Membrane/metabolism , Cyclic GMP/chemistry , Cysteine Endopeptidases/metabolism , Gene Expression Profiling , Microscopy, Fluorescence , Mutation , Oligonucleotides/metabolism , Peptidoglycan/chemistry , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Tandem Mass Spectrometry , Virulence Factors/metabolism
11.
Mol Microbiol ; 98(2): 329-42, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26179020

ABSTRACT

Bacteriophages are present in virtually all ecosystems, and bacteria have developed multiple antiphage strategies to counter their attacks. Clostridium difficile is an important pathogen causing severe intestinal infections in humans and animals. Here we show that the conserved cell-surface protein CwpV provides antiphage protection in C. difficile. This protein, for which the expression is phase-variable, is classified into five types, each differing in their repeat-containing C-terminal domain. When expressed constitutively from a plasmid or the chromosome of locked 'ON' cells of C. difficile R20291, CwpV conferred antiphage protection. Differences in the level of phage protection were observed depending on the phage morphological group, siphophages being the most sensitive with efficiency of plaquing (EOP) values of < 5 × 10(-7) for phages ϕCD38-2, ϕCD111 and ϕCD146. Protection against the myophages ϕMMP01 and ϕCD52 was weaker, with EOP values between 9.0 × 10(-3) and 1.1 × 10(-1). The C-terminal domain of CwpV carries the antiphage activity and its deletion, or part of it, significantly reduced the antiphage protection. CwpV does not affect phage adsorption, but phage DNA replication is prevented, suggesting a mechanism reminiscent of superinfection exclusion systems normally encoded on prophages. CwpV thus represents a novel ubiquitous host-encoded and phase-variable antiphage system in C. difficile.


Subject(s)
Bacterial Proteins/metabolism , Bacteriophages/growth & development , Cell Wall/chemistry , Clostridioides difficile/metabolism , Clostridioides difficile/virology , Animals , Bacterial Proteins/genetics , Bacteriophages/pathogenicity , Bacteriophages/physiology , Cell Wall/metabolism , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , DNA, Viral/genetics , Humans , Sequence Analysis, DNA
12.
BMC Genomics ; 16: 392, 2015 May 16.
Article in English | MEDLINE | ID: mdl-25981746

ABSTRACT

BACKGROUND: Clostridium sordellii can cause severe infections in animals and humans, the latter associated with trauma, toxic shock and often-fatal gynaecological infections. Strains can produce two large clostridial cytotoxins (LCCs), TcsL and TcsH, related to those produced by Clostridium difficile, Clostridium novyi and Clostridium perfringens, but the genetic basis of toxin production remains uncharacterised. RESULTS: Phylogenetic analysis of the genome sequences of 44 strains isolated from human and animal infections in the UK, US and Australia placed the species into four clades. Although all strains originated from animal or clinical disease, only 5 strains contained LCC genes: 4 strains contain tcsL alone and one strain contains tcsL and tcsH. Four toxin-positive strains were found within one clade. Where present, tcsL and tcsH were localised in a pathogenicity locus, similar to but distinct from that present in C. difficile. In contrast to C. difficile, where the LCCs are chromosomally localised, the C. sordellii tcsL and tcsH genes are localised on plasmids. Our data suggest gain and loss of entire toxigenic plasmids in addition to horizontal transfer of the pathogenicity locus. A high quality, annotated sequence of ATCC9714 reveals many putative virulence factors including neuraminidase, phospholipase C and the cholesterol-dependent cytolysin sordellilysin that are highly conserved between all strains studied. CONCLUSIONS: Genome analysis of C. sordellii reveals that the LCCs, the major virulence factors, are localised on plasmids. Many strains do not contain the LCC genes; it is probable that in several of these cases the plasmid has been lost upon laboratory subculture. Our data are consistent with LCCs being the primary virulence factors in the majority of infections, but LCC-negative strains may precipitate certain categories of infection. A high quality genome sequence reveals putative virulence factors whose role in virulence can be investigated.


Subject(s)
Bacterial Toxins/genetics , Clostridium sordellii/genetics , Clostridium sordellii/pathogenicity , Genome, Bacterial/genetics , Plasmids/metabolism , Virulence Factors/genetics , Chromosome Mapping , Clostridium sordellii/classification , Gene Transfer, Horizontal , Genetic Loci/genetics , Neuraminidase/genetics , Phylogeny , Plasmids/genetics , Sequence Analysis, DNA , Type C Phospholipases/genetics
13.
Mol Microbiol ; 96(3): 596-608, 2015 May.
Article in English | MEDLINE | ID: mdl-25649385

ABSTRACT

Gram-positive surface proteins can be covalently or non-covalently anchored to the cell wall and can impart important properties on the bacterium in respect of cell envelope organisation and interaction with the environment. We describe here a mechanism of protein anchoring involving tandem CWB2 motifs found in a large number of cell wall proteins in the Firmicutes. In the Clostridium difficile cell wall protein family, we show the three tandem repeats of the CWB2 motif are essential for correct anchoring to the cell wall. CWB2 repeats are non-identical and cannot substitute for each other, as shown by the secretion into the culture supernatant of proteins containing variations in the patterns of repeats. A conserved Ile Leu Leu sequence within the CWB2 repeats is essential for correct anchoring, although a preceding proline residue is dispensable. We propose a likely genetic locus encoding synthesis of the anionic polymer PSII and, using RNA knock-down of key genes, reveal subtle effects on cell wall composition. We show that the anionic polymer PSII binds two cell wall proteins, SlpA and Cwp2, and these interactions require the CWB2 repeats, defining a new mechanism of protein anchoring in Gram-positive bacteria.


Subject(s)
Amino Acid Motifs , Cell Wall/metabolism , Clostridioides difficile/metabolism , Membrane Proteins/metabolism , Polysaccharides, Bacterial/metabolism , Gene Knockdown Techniques , Protein Binding , Repetitive Sequences, Amino Acid
14.
mBio ; 6(2): e02383, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25714712

ABSTRACT

UNLABELLED: Clostridium difficile is the most common cause of antibiotic-associated intestinal infections and a significant cause of morbidity and mortality. Infection with C. difficile requires disruption of the intestinal microbiota, most commonly by antibiotic usage. Therapeutic intervention largely relies on a small number of broad-spectrum antibiotics, which further exacerbate intestinal dysbiosis and leave the patient acutely sensitive to reinfection. Development of novel targeted therapeutic interventions will require a detailed knowledge of essential cellular processes, which represent attractive targets, and species-specific processes, such as bacterial sporulation. Our knowledge of the genetic basis of C. difficile infection has been hampered by a lack of genetic tools, although recent developments have made some headway in addressing this limitation. Here we describe the development of a method for rapidly generating large numbers of transposon mutants in clinically important strains of C. difficile. We validated our transposon mutagenesis approach in a model strain of C. difficile and then generated a comprehensive transposon library in the highly virulent epidemic strain R20291 (027/BI/NAP1) containing more than 70,000 unique mutants. Using transposon-directed insertion site sequencing (TraDIS), we have identified a core set of 404 essential genes, required for growth in vitro. We then applied this technique to the process of sporulation, an absolute requirement for C. difficile transmission and pathogenesis, identifying 798 genes that are likely to impact spore production. The data generated in this study will form a valuable resource for the community and inform future research on this important human pathogen. IMPORTANCE: Clostridium difficile is a common cause of potentially fatal intestinal infections in hospital patients, particularly those who have been treated with antibiotics. Our knowledge of this bacterium has been hampered by a lack of tools for dissecting the organism. We have developed a method to study the function of every gene in the bacterium simultaneously. Using this tool, we have identified a set of 404 genes that are required for growth of the bacteria in the laboratory. C. difficile also produces a highly resistant spore that can survive in the environment for a long time and is a requirement for transmission of the bacteria between patients. We have applied our genetic tool to identify all of the genes required for production of a spore. All of these genes represent attractive targets for new drugs to treat infection.


Subject(s)
Clostridioides difficile/growth & development , Genes, Bacterial , Genes, Essential , Genetic Testing/methods , High-Throughput Screening Assays , Mutagenesis, Insertional/methods , Spores, Bacterial/growth & development , Clostridioides difficile/genetics , DNA Transposable Elements , Humans , Spores, Bacterial/genetics
15.
Mol Microbiol ; 92(5): 1025-38, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24720767

ABSTRACT

The BclA protein is a major component of the outermost layer of spores of a number of bacterial species and Clostridium difficile carries three bclA genes. Using insertional mutagenesis each gene was characterized and spores devoid of these proteins had surface aberrations, reduced hydrophobicity and germinated faster than wild-type spores. Therefore the BclA proteins were likely major components of the spore surface and when absent impaired the protective shield effect of this outermost layer. Analysis of infection and colonization in mice and hamsters revealed that the 50% infectious dose (ID50 ) of spores was significantly higher (2-logs) in the bclA1(-) mutant compared to the isogenic wild-type control, but that levels of toxins (A and B) were indistinguishable from animals dosed with wild-type spores. bclA1(-) spores germinated faster than wild-type spores yet mice were less susceptible to infection suggesting that BclA1 must play a key role in the initial (i.e. pre-spore germination) stages of infection. We also show that the ID50 was higher in mice infected with R20291, a 'hypervirulent' 027 strain, that carries a truncated BclA1 protein.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/metabolism , Spores, Bacterial/pathogenicity , Animals , Clostridioides difficile/metabolism , Cricetinae , Gene Expression Regulation, Bacterial , Mice , Spores, Bacterial/metabolism
16.
Nat Rev Microbiol ; 12(3): 211-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24509785

ABSTRACT

The outer surface of many archaea and bacteria is coated with a proteinaceous surface layer (known as an S-layer), which is formed by the self-assembly of monomeric proteins into a regularly spaced, two-dimensional array. Bacteria possess dedicated pathways for the secretion and anchoring of the S-layer to the cell wall, and some Gram-positive species have large S-layer-associated gene families. S-layers have important roles in growth and survival, and their many functions include the maintenance of cell integrity, enzyme display and, in pathogens and commensals, interaction with the host and its immune system. In this Review, we discuss our current knowledge of S-layer and related proteins, including their structures, mechanisms of secretion and anchoring and their diverse functions.


Subject(s)
Bacteria/metabolism , Membrane Glycoproteins/metabolism , Animals , Archaea/genetics , Archaea/metabolism , Bacteria/genetics , Cell Wall/metabolism , Genetic Variation , Humans , Membrane Glycoproteins/genetics , Models, Biological , Multigene Family/physiology , Protein Structure, Tertiary
17.
PLoS One ; 8(5): e64011, 2013.
Article in English | MEDLINE | ID: mdl-23691138

ABSTRACT

Clostridium difficile is the leading cause of hospital acquired diarrhoea in industrialised countries. Under conditions that are not favourable for growth, the pathogen produces metabolically dormant endospores via asymmetric cell division. These are extremely resistant to both chemical and physical stress and provide the mechanism by which C. difficile can evade the potentially fatal consequences of exposure to heat, oxygen, alcohol, and certain disinfectants. Spores are the primary infective agent and must germinate to allow for vegetative cell growth and toxin production. While spore germination in Bacillus is well understood, little is known about C. difficile germination and outgrowth. Here we use genome-wide transcriptional analysis to elucidate the temporal gene expression patterns in C. difficile 630 endospore germination. We have optimized methods for large scale production and purification of spores. The germination characteristics of purified spores have been characterized and RNA extraction protocols have been optimized. Gene expression was highly dynamic during germination and outgrowth, and was found to involve a large number of genes. Using this genome-wide, microarray approach we have identified 511 genes that are significantly up- or down-regulated during C. difficile germination (p≤0.01). A number of functional groups of genes appeared to be co-regulated. These included transport, protein synthesis and secretion, motility and chemotaxis as well as cell wall biogenesis. These data give insight into how C. difficile re-establishes its metabolism, re-builds the basic structures of the vegetative cell and resumes growth.


Subject(s)
Clostridioides difficile/genetics , Gene Expression Profiling , Spores, Bacterial , Transcription, Genetic , Clostridioides difficile/physiology , Oligonucleotide Array Sequence Analysis , RNA, Bacterial/genetics
18.
Cell Microbiol ; 15(10): 1674-87, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23517059

ABSTRACT

Clostridium difficile is the leading cause of antibiotic-associated diarrhoea and pseudomembranous colitis. While the role of toxins in pathogenesis has been extensively described, the contribution of surface determinants to intestinal colonization is still poorly understood. We focused our study on a novel member of the MSCRAMM family, named CbpA (Collagen binding protein A), for its adhesive properties towards collagen. We demonstrate that CbpA, which carries an LPXTG-like cell wall anchoring domain, is expressed on the bacterial surface of C. difficile and that the recombinant protein binds at high affinity to collagens I and V (apparent Kd in the order of 10(-9 ) M). These findings were validated by confocal microscopy studies showing the colocalization of the protein with type I and V collagen fibres produced by human fibroblasts and mouse intestinal tissues. However, the collagen binding activity of the wild-type C. difficile 630 strain was indistinguishable to the cbpA knock-out strain. To overcome this apparent clostridial adherence redundancy, we engineered a Lactococcus lactis strain for the heterologous expression of CbpA. When exposed on the surface of L. lactis, CbpA significantly enhances the ability of the bacterium to interact with collagen and to adhere to ECM-producing cells. The binding activity of L. lactis-CbpA strain was prevented by an antiserum raised against CbpA, demonstrating the specificity of the interaction. These results suggest that CbpA is a newsurface-exposed adhesin contributing to the C. difficile interaction with the host.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Clostridioides difficile/physiology , Collagen/metabolism , Host-Pathogen Interactions , Animals , Fibroblasts/metabolism , Fibroblasts/microbiology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Kinetics , Lactococcus lactis/genetics , Lactococcus lactis/physiology , Mice , Microscopy, Confocal , Protein Binding
19.
Infect Immun ; 80(8): 2704-11, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22615253

ABSTRACT

Clostridium difficile is a major cause of chronic antibiotic-associated diarrhea and a significant health care-associated pathogen that forms highly resistant and infectious spores. Spo0A is a highly conserved transcriptional regulator that plays a key role in initiating sporulation in Bacillus and Clostridium species. Here, we use a murine model to study the role of the C. difficile spo0A gene during infection and transmission. We demonstrate that C. difficile spo0A mutant derivatives can cause intestinal disease but are unable to persist within and effectively transmit between mice. Thus, the C. difficile Spo0A protein plays a key role in persistent infection, including recurrence and host-to-host transmission in mice.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Clostridium Infections/microbiology , Gene Expression Regulation, Bacterial/physiology , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Clostridium Infections/transmission , Mice , Mice, Inbred C57BL , Mutagenesis , Mutation , Specific Pathogen-Free Organisms , Transcription Factors/genetics , Virulence
20.
Vaccine ; 30(20): 3047-52, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22414558

ABSTRACT

The current commercially available vaccine used to prevent tetanus disease following infection with the anaerobic bacterium Clostridium tetani is safe and effective. However, tetanus remains a major source of mortality in developing countries. In 2008, neonatal tetanus was estimated to have caused >59,000 deaths, accounting for 1% of worldwide infant mortality, primarily in poorer nations. The cost of multiple vaccine doses administered by injection necessary to achieve protective levels of anti-tetanus toxoid antibodies is the primary reason for low vaccine coverage. Herein, we show that a novel vaccine strategy using a cytomegalovirus (CMV)-based vaccine platform induces protective levels of anti-tetanus antibodies that are durable (lasting >13 months) in mice following only a single dose. This study demonstrates the ability of a 'single-dose' CMV-based vaccine strategy to induce durable protection, and supports the potential for a tetanus vaccine based on CMV to impact the incidence of tetanus in developing countries.


Subject(s)
Antibodies, Bacterial/blood , Antitoxins/blood , Cytomegalovirus/genetics , Genetic Vectors , Peptide Fragments/immunology , Tetanus Toxin/immunology , Tetanus Toxoid/immunology , Tetanus/prevention & control , Animals , Disease Models, Animal , Mice , Peptide Fragments/genetics , Tetanus Toxin/genetics , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...