Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 293(47): 18123-18137, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30275012

RESUMEN

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.


Asunto(s)
Clostridioides difficile/metabolismo , Glicoproteínas de Membrana/metabolismo , Polisacáridos/metabolismo , Esporas Bacterianas/crecimiento & desarrollo , Clostridioides difficile/genética , Clostridioides difficile/crecimiento & desarrollo , Glicosilación , Espectrometría de Masas , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Peso Molecular , Polisacáridos/química , Conformación Proteica , Esporas Bacterianas/genética , Esporas Bacterianas/metabolismo
2.
Mol Microbiol ; 100(1): 204-28, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26690930

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Clostridioides difficile/fisiología , Regulación Bacteriana de la Expresión Génica , Esporas Bacterianas , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Eliminación de Secuencia
3.
J Biol Chem ; 290(40): 24453-69, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26283789

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Clostridioides difficile/metabolismo , GMP Cíclico/análogos & derivados , Regulación Bacteriana de la Expresión Génica , Metaloproteasas/metabolismo , Péptido Hidrolasas/metabolismo , Adhesinas Bacterianas/metabolismo , Secuencias de Aminoácidos , Aminoaciltransferasas/metabolismo , Membrana Celular/metabolismo , GMP Cíclico/química , Cisteína Endopeptidasas/metabolismo , Perfilación de la Expresión Génica , Microscopía Fluorescente , Mutación , Oligonucleótidos/metabolismo , Peptidoglicano/química , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Espectrometría de Masas en Tándem , Factores de Virulencia/metabolismo
4.
Mol Microbiol ; 98(2): 329-42, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26179020

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteriófagos/crecimiento & desarrollo , Pared Celular/química , Clostridioides difficile/metabolismo , Clostridioides difficile/virología , Animales , Proteínas Bacterianas/genética , Bacteriófagos/patogenicidad , Bacteriófagos/fisiología , Pared Celular/metabolismo , Clostridioides difficile/química , Clostridioides difficile/genética , ADN Viral/genética , Humanos , Análisis de Secuencia de ADN
5.
Mol Microbiol ; 96(3): 596-608, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25649385

RESUMEN

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.


Asunto(s)
Secuencias de Aminoácidos , Pared Celular/metabolismo , Clostridioides difficile/metabolismo , Proteínas de la Membrana/metabolismo , Polisacáridos Bacterianos/metabolismo , Técnicas de Silenciamiento del Gen , Unión Proteica , Secuencias Repetitivas de Aminoácido
6.
BMC Genomics ; 16: 392, 2015 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-25981746

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas/genética , Clostridium sordellii/genética , Clostridium sordellii/patogenicidad , Genoma Bacteriano/genética , Plásmidos/metabolismo , Factores de Virulencia/genética , Mapeo Cromosómico , Clostridium sordellii/clasificación , Transferencia de Gen Horizontal , Sitios Genéticos/genética , Neuraminidasa/genética , Filogenia , Plásmidos/genética , Análisis de Secuencia de ADN , Fosfolipasas de Tipo C/genética
7.
Mol Microbiol ; 92(5): 1025-38, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24720767

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Clostridioides difficile/patogenicidad , Enterocolitis Seudomembranosa/metabolismo , Esporas Bacterianas/patogenicidad , Animales , Clostridioides difficile/metabolismo , Cricetinae , Regulación Bacteriana de la Expresión Génica , Ratones , Esporas Bacterianas/metabolismo
8.
BMC Microbiol ; 15: 280, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26679502

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Clostridioides difficile/genética , Enterotoxinas/genética , Mutagénesis Insercional/genética , Factores de Transcripción/genética , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Supervivencia Celular/efectos de los fármacos , Pared Celular/genética , Chlorocebus aethiops , Clostridioides difficile/metabolismo , Clostridioides difficile/fisiología , Enterotoxinas/metabolismo , Enterotoxinas/toxicidad , Regulación Bacteriana de la Expresión Génica , Células HT29 , Humanos , Mutagénesis Insercional/inmunología , Transcripción Genética , Regulación hacia Arriba , Células Vero
9.
Nat Genet ; 38(7): 779-86, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16804543

RESUMEN

We determined the complete genome sequence of Clostridium difficile strain 630, a virulent and multidrug-resistant strain. Our analysis indicates that a large proportion (11%) of the genome consists of mobile genetic elements, mainly in the form of conjugative transposons. These mobile elements are putatively responsible for the acquisition by C. difficile of an extensive array of genes involved in antimicrobial resistance, virulence, host interaction and the production of surface structures. The metabolic capabilities encoded in the genome show multiple adaptations for survival and growth within the gut environment. The extreme genome variability was confirmed by whole-genome microarray analysis; it may reflect the organism's niche in the gut and should provide information on the evolution of virulence in this organism.


Asunto(s)
Clostridioides difficile/genética , Clostridioides difficile/patogenicidad , Adaptación Fisiológica , Proteínas Bacterianas/genética , Secuencia de Bases , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/fisiología , Conjugación Genética , Elementos Transponibles de ADN/genética , ADN Bacteriano/genética , Farmacorresistencia Bacteriana Múltiple/genética , Enterocolitis Seudomembranosa/etiología , Enterocolitis Seudomembranosa/microbiología , Tracto Gastrointestinal/microbiología , Genoma Bacteriano , Humanos , Datos de Secuencia Molecular , Mosaicismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Esporas Bacterianas/fisiología , Virulencia/genética
10.
Cell Microbiol ; 15(10): 1674-87, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23517059

RESUMEN

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.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Clostridioides difficile/fisiología , Colágeno/metabolismo , Interacciones Huésped-Patógeno , Animales , Fibroblastos/metabolismo , Fibroblastos/microbiología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Cinética , Lactococcus lactis/genética , Lactococcus lactis/fisiología , Ratones , Microscopía Confocal , Unión Proteica
11.
J Biol Chem ; 287(2): 1538-44, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22128177

RESUMEN

Clostridium difficile infection is a leading cause of antibiotic-associated diarrhea, placing considerable economic pressure on healthcare systems and resulting in significant morbidity and mortality. The pathogen produces a proteinaceous array on its cell surface known as the S-layer, consisting primarily of the major S-layer protein SlpA and a family of SlpA homologs. CwpV is the largest member of this family and is expressed in a phase-variable manner. The protein is post-translationally processed into two fragments that form a noncovalent, heterodimeric complex. To date, no specific proteases capable of cleaving CwpV have been identified. Using site-directed mutagenesis we show that CwpV undergoes intramolecular autoproteolysis, most likely facilitated by a N-O acyl shift, with Thr-413 acting as the source of a nucleophile driving this rearrangement. We demonstrate that neighboring residues are also important for correct processing of CwpV. Based on protein structural predictions and analogy to the glycosylasparaginase family of proteins, it appears likely that these residues play key roles in determining the correct protein fold and interact directly with Thr-413 to promote nucleophilic attack. Furthermore, using a cell-free protein synthesis assay we show that CwpV maturation requires neither cofactors nor auxiliary enzymes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Clostridioides difficile/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteolisis , Pared Celular/genética , Clostridioides difficile/genética , Glicoproteínas de Membrana/genética
12.
PLoS Pathog ; 7(4): e1002024, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21533071

RESUMEN

Clostridium difficile is the main cause of antibiotic-associated diarrhea, leading to significant morbidity and mortality and putting considerable economic pressure on healthcare systems. Current knowledge of the molecular basis of pathogenesis is limited primarily to the activities and regulation of two major toxins. In contrast, little is known of mechanisms used in colonization of the enteric system. C. difficile expresses a proteinaceous array on its cell surface known as the S-layer, consisting primarily of the major S-layer protein SlpA and a family of SlpA homologues, the cell wall protein (CWP) family. CwpV is the largest member of this family and is expressed in a phase variable manner. Here we show CwpV promotes C. difficile aggregation, mediated by the C-terminal repetitive domain. This domain varies markedly between strains; five distinct repeat types were identified and were shown to be antigenically distinct. Other aspects of CwpV are, however, conserved. All CwpV types are expressed in a phase variable manner. Using targeted gene knock-out, we show that a single site-specific recombinase RecV is required for CwpV phase variation. CwpV is post-translationally cleaved at a conserved site leading to formation of a complex of cleavage products. The highly conserved N-terminus anchors the CwpV complex to the cell surface. Therefore CwpV function, regulation and processing are highly conserved across C. difficile strains, whilst the functional domain exists in at least five antigenically distinct forms. This hints at a complex evolutionary history for CwpV.


Asunto(s)
Pared Celular/metabolismo , Clostridioides difficile/metabolismo , Evolución Molecular , Glicoproteínas de Membrana/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Pared Celular/genética , Pared Celular/inmunología , Clostridioides difficile/genética , Clostridioides difficile/inmunología , Técnicas de Silenciamiento del Gen , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Estructura Terciaria de Proteína , Recombinasas/genética , Recombinasas/inmunología , Recombinasas/metabolismo
13.
J Biol Chem ; 286(31): 27483-93, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21659510

RESUMEN

Protein translocation across the cytoplasmic membrane is an essential process in all bacteria. The Sec system, comprising at its core an ATPase, SecA, and a membrane channel, SecYEG, is responsible for the majority of this protein transport. Recently, a second parallel Sec system has been described in a number of gram-positive species. This accessory Sec system is characterized by the presence of a second copy of the energizing ATPase, SecA2; where it has been studied, SecA2 is responsible for the translocation of a subset of Sec substrates. In common with many pathogenic gram-positive species, Clostridium difficile possesses two copies of SecA. Here, we describe the first characterization of the C. difficile accessory Sec system and the identification of its major substrates. Using inducible antisense RNA expression and dominant-negative alleles of secA1 and secA2, we demonstrate that export of the S-layer proteins (SLPs) and an additional cell wall protein (CwpV) is dependent on SecA2. Accumulation of the cytoplasmic precursor of the SLPs SlpA and other cell wall proteins was observed in cells expressing dominant-negative secA1 or secA2 alleles, concomitant with a decrease in the levels of mature SLPs in the cell wall. Furthermore, expression of either dominant-negative allele or antisense RNA knockdown of SecA1 or SecA2 dramatically impaired growth, indicating that both Sec systems are essential in C. difficile.


Asunto(s)
Proteínas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Clostridioides difficile/genética , Mutación , Plásmidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Infect Immun ; 80(8): 2704-11, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22615253

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Infecciones por Clostridium/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Factores de Transcripción/metabolismo , Animales , Proteínas Bacterianas/genética , Clostridioides difficile/genética , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/transmisión , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Mutación , Organismos Libres de Patógenos Específicos , Factores de Transcripción/genética , Virulencia
15.
Bioorg Med Chem ; 20(2): 614-21, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21752656

RESUMEN

Clostridium difficile, a leading cause of hospital-acquired bacterial infection, is coated in a dense surface layer (S-layer) that is thought to provide both physicochemical protection and a scaffold for host-pathogen interactions. The key structural components of the S-layer are two proteins derived from a polypeptide precursor, SlpA, via proteolytic cleavage by the protease Cwp84. Here, we report the design, synthesis and in vivo characterization of a panel of protease inhibitors and activity-based probes (ABPs) designed to target S-layer processing in live C. difficile cells. Inhibitors based on substrate-mimetic peptides bearing a C-terminal Michael acceptor warhead were found to be promising candidates for further development.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Clostridioides difficile/metabolismo , Cisteína Endopeptidasas/química , Inhibidores de Proteasas/química , Proteínas Bacterianas/metabolismo , Clostridioides difficile/enzimología , Cisteína Endopeptidasas/metabolismo , Interacciones Huésped-Patógeno , Péptidos/síntesis química , Péptidos/química , Inhibidores de Proteasas/síntesis química , Relación Estructura-Actividad
16.
Nat Commun ; 13(1): 970, 2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35217634

RESUMEN

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.


Asunto(s)
Clostridioides difficile , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Clostridioides difficile/genética
17.
J Bacteriol ; 193(13): 3276-85, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21531808

RESUMEN

Clostridium difficile expresses a number of cell wall proteins, including the abundant high-molecular-weight and low-molecular-weight S-layer proteins (SLPs). These proteins are generated by posttranslational cleavage of the precursor SlpA by the cysteine protease Cwp84. We compared the phenotypes of C. difficile strains containing insertional mutations in either cwp84 or its paralog cwp13 and complemented with plasmids expressing wild-type or mutant forms of their genes. We show that the presence of uncleaved SlpA in the cell wall of the cwp84 mutant results in aberrant retention of other cell wall proteins at the cell surface, as demonstrated by secretion of the proteins Cwp66 and Cwp2 into the growth medium. These phenotypes are restored by complementation with a plasmid expressing wild-type Cwp84 enzyme but not with one encoding a Cys116Ala substitution in the active site. The cwp13 mutant cleaved the SlpA precursor normally and had a wild-type-like colony phenotype. Both Cwp84 and Cwp13 are produced as proenzymes which are processed by cleavage to produce mature enzymes. In the case of Cwp84, this cleavage does not appear to be autocatalytic, whereas in Cwp13 autocatalysis was demonstrated as a Cys109Ala mutant did not undergo processing. Cwp13 appears to have a role in processing of Cwp84 but is not essential for Cwp84 activity. Cwp13 cleaves SlpA in the HMW SLP domain, which we suggest may reflect a role in cleavage and degradation of misfolded proteins at the cell surface.


Asunto(s)
Pared Celular/metabolismo , Clostridioides difficile/enzimología , Clostridioides difficile/metabolismo , Cisteína Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Clostridioides difficile/genética , Cisteína Endopeptidasas/genética , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Mutagénesis Insercional , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Procesamiento Proteico-Postraduccional
18.
Infect Immun ; 79(6): 2295-302, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21482682

RESUMEN

Clostridium difficile is a leading cause of nosocomial infection in the developed world. Two toxins, A and B, produced by most strains of C. difficile are implicated as virulence factors, yet only recently has the requirement of these for infection been investigated by genetic manipulation. Current vaccine strategies are focused mostly on parenteral delivery of toxoids. In this work, we have used bacterial spores (Bacillus subtilis) as a delivery vehicle to evaluate the carboxy-terminal repeat domains of toxins A and B as protective antigens. Our findings are important and show that oral immunization of the repeat domain of toxin A is sufficient to confer protection in a hamster model of infection designed to closely mimic the human course of infection. Importantly, neutralizing antibodies to the toxin A repeat domain were shown to be cross-reactive with the analogous domain of toxin B and, being of high avidity, provided protection against challenge with a C. difficile strain producing toxins A and B (A(+)B(+)). Thus, although many strains produce both toxins, antibodies to only toxin A can mediate protection. Animals vaccinated with recombinant spores were fully able to survive reinfection, a property that is particularly important for a disease with which patients are prone to relapse. We show that mucosal immunization, not parenteral delivery, is required to generate secretory IgA and that production of these neutralizing polymeric antibodies correlates with protection. This work demonstrates that an effective vaccine against C. difficile can be designed around two attributes, mucosal delivery and the repeat domain of toxin A.


Asunto(s)
Bacillus subtilis/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/prevención & control , Enterotoxinas/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Cricetinae , Protección Cruzada/inmunología , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/microbiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Mesocricetus , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Esporas Bacterianas/inmunología , Vacunas Sintéticas/inmunología
19.
Sci Rep ; 11(1): 3244, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547340

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas , Clostridioides difficile/fisiología , GMP Cíclico/análogos & derivados , ADN Bacteriano/metabolismo , Biopelículas/crecimiento & desarrollo , Clostridioides difficile/ultraestructura , Infecciones por Clostridium/microbiología , GMP Cíclico/metabolismo , Humanos
20.
Mol Microbiol ; 71(5): 1308-22, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19183279

RESUMEN

Clostridium difficile expresses a surface layer (S-layer) which coats the surface of the bacterium and acts as an adhesin facilitating interaction of the bacterium with host enteric cells. The S-layer contains a high-molecular-weight S-layer protein (HMW SLP) and its low-molecular-weight partner protein (LMW SLP). We show that these proteins form a tightly associated non-covalent complex, the H/L complex, and we identify the regions of both proteins responsible for complex formation. The 2.4 A X-ray crystal structure of a truncated derivative of the LMW SLP reveals two domains. Domain 1 has a two-layer sandwich architecture while domain 2, predicted to orientate towards the external environment, contains a novel fold. Small-angle X-ray scattering analysis of the H/L complex shows an elongated molecule, with the two SLPs arranged 'end-to-end' interacting with each other through a small contact area. Alignment of LMW SLPs, which exhibit high sequence diversity, reveals a core of conserved residues that could reflect functional conservation, while allowing for immune evasion through sequence variation. These structures are the first described for the S-layer of a bacterial pathogen, and provide insights into the assembly and biogenesis of the S-layer.


Asunto(s)
Proteínas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Glicoproteínas de Membrana/metabolismo , Secuencia de Aminoácidos , Pared Celular/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA