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










Publication year range
1.
J Colloid Interface Sci ; 652(Pt A): 718-726, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37611471

ABSTRACT

Highly persistent, drug-resistant and transmissible healthcare pathogens such as Clostridioides difficile (C. difficile) and Candida auris (C. auris) are responsible for causing antibiotic-associated fatal diarrhea and invasive candidiasis, respectively. In this study, we demonstrate that these potentially lethal gastrointestinal microbes can be rapidly inactivated on the solid surface of a self-disinfecting anionic block polymer that inherently generates a water surface layer that is highly acidic (pH < 1) upon hydration. Due to thermodynamic incompatibility between its chemical sequences, the polymer spontaneously self-organizes into a nanostructure that enables proton migration from the interior of a film to the surface via contiguous nanoscale hydrophilic channels, as discerned here by scanning electron and atomic force microscopies, as well as X-ray photoelectron spectroscopy. Here, we report that two strains of C. difficile in the vegetative state and two species of Candida, Candida albicans (C. albicans) and C. auris, are, in most cases, inactivated to the limit of minimum detection. Corresponding electron and optical microscopy images reveal that, upon exposure to the hydrated polymer, the outer microbial membranes display evidence of damage and intracellular material is expelled. Combined with our previous studies of rapid bacterial and viral inactivation, these antimicrobial results are highly encouraging and, if translatable to clinical conditions in the form of self-standing polymer films or coatings, are expected to benefit the welfare of patients in healthcare facilities by continuously preventing the spread of such potentially dangerous microbes.


Subject(s)
Candidiasis , Clostridioides difficile , Humans , Candida , Candida albicans , Antifungal Agents
2.
Curr Opin Microbiol ; 65: 95-100, 2022 02.
Article in English | MEDLINE | ID: mdl-34781095

ABSTRACT

Clostridioides difficile produces toxins TcdA and TcdB during infection. Since the severity of the illness is directly correlated with the level of toxins produced, researchers have long been interested in the regulation mechanisms of toxin production. The advent of new genetics and mutagenesis technologies in C. difficile has allowed a slew of new investigations in the last decade, which considerably improved our understanding of this crucial regulatory network. The current body of work shows that the toxin regulatory network overlaps with the regulatory networks of sporulation, motility, and key metabolic pathways. This implies that toxin production is a complicated process initiated by bacteria in response to numerous host factors during infection. We summarize the existing knowledge about the toxin gene regulatory network here.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Clostridioides difficile/genetics , Enterotoxins/genetics , Enterotoxins/metabolism
3.
Mol Microbiol ; 116(5): 1347-1360, 2021 11.
Article in English | MEDLINE | ID: mdl-34606654

ABSTRACT

Clostridioides difficile is the causative agent of antibiotic-associated diarrhea and is the leading cause of nosocomial infection in developed countries. An increasing number of C. difficile infections are attributed to epidemic strains that produce more toxins and spores. C. difficile spores are the major factor for the transmission and persistence of the organism. Previous studies have identified global regulators that influence sporulation in C. difficile. This study discovers that PdcB, a phosphodiesterase, enhances sporulation in C. difficile strain UK1. Through genetic and biochemical assays, we show that phase-variable expression of pdcB results in hypo- and hyper-sporulation phenotypes. In the "ON" orientation, the identified promotor is in the right orientation to drive the expression of pdcB. Production of the PdcB phosphodiesterase reduces the intracellular cyclic-di-GMP (c-di-GMP) concentration, resulting in a hyper-sporulation phenotype. Loss of PdcB due to the pdcB promoter being in the OFF orientation or mutation of pdcB results in increased c-di-GMP levels and a hypo-sporulation phenotype. Additionally, we demonstrate that CodY binds to the upstream region of pdcB. DNA inversion reorients the CodY binding site so that in the OFF orientation, CodY binds a site that is upstream of the pdcB promoter and can further repress gene expression.


Subject(s)
Clostridioides difficile/physiology , Cyclic GMP/analogs & derivatives , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Spores, Bacterial/metabolism , Transcription Factors/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Clostridium Infections/microbiology , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial , Mutation , Promoter Regions, Genetic
4.
Microbiology (Reading) ; 167(8)2021 08.
Article in English | MEDLINE | ID: mdl-34410904

ABSTRACT

Cellobiose metabolism is linked to the virulence properties in numerous bacterial pathogens. Here, we characterized a putative cellobiose PTS operon of Clostridiodes difficile to investigate the role of cellobiose metabolism in C. difficile pathogenesis. Our gene knockout experiments demonstrated that the putative cellobiose operon enables uptake of cellobiose into C. difficile and allows growth when cellobiose is provided as the sole carbon source in minimal medium. Additionally, using reporter gene fusion assays and DNA pulldown experiments, we show that its transcription is regulated by CelR, a novel transcriptional repressor protein, which directly binds to the upstream region of the cellobiose operon to control its expression. We have also identified cellobiose metabolism to play a significant role in C. difficile physiology as observed by the reduction of sporulation efficiency when cellobiose uptake was compromised in the mutant strain. In corroboration to in vitro study findings, our in vivo hamster challenge experiment showed a significant reduction of pathogenicity by the cellobiose mutant strain in both the primary and the recurrent infection model - substantiating the role of cellobiose metabolism in C. difficile pathogenesis.


Subject(s)
Cellobiose , Clostridioides difficile , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Clostridioides , Clostridioides difficile/genetics , Gene Expression Regulation, Bacterial , Operon
5.
mSphere ; 5(6)2020 11 04.
Article in English | MEDLINE | ID: mdl-33148827

ABSTRACT

Clostridioides difficile is the leading cause of nosocomial infection and is the causative agent of antibiotic-associated diarrhea. The severity of the disease is directly associated with toxin production, and spores are responsible for the transmission and persistence of the organism. Previously, we characterized sin locus regulators SinR and SinR' (we renamed it SinI), where SinR is the regulator of toxin production and sporulation. The SinI regulator acts as its antagonist. In Bacillus subtilis, Spo0A, the master regulator of sporulation, controls SinR by regulating the expression of its antagonist, sinI However, the role of Spo0A in the expression of sinR and sinI in C. difficile had not yet been reported. In this study, we tested spo0A mutants in three different C. difficile strains, R20291, UK1, and JIR8094, to understand the role of Spo0A in sin locus expression. Western blot analysis revealed that spo0A mutants had increased SinR levels. Quantitative reverse transcription-PCR (qRT-PCR) analysis of its expression further supported these data. By carrying out genetic and biochemical assays, we show that Spo0A can bind to the upstream region of this locus to regulates its expression. This study provides vital information that Spo0A regulates the sin locus, which controls critical pathogenic traits such as sporulation, toxin production, and motility in C. difficileIMPORTANCEClostridioides difficile is the leading cause of antibiotic-associated diarrheal disease in the United States. During infection, C. difficile spores germinate, and the vegetative bacterial cells produce toxins that damage host tissue. In C. difficile, the sin locus is known to regulate both sporulation and toxin production. In this study, we show that Spo0A, the master regulator of sporulation, controls sin locus expression. Results from our study suggest that Spo0A directly regulates the expression of this locus by binding to its upstream DNA region. This observation adds new detail to the gene regulatory network that connects sporulation and toxin production in this pathogen.


Subject(s)
Bacterial Proteins/genetics , Clostridioides difficile/genetics , Gene Expression Regulation, Bacterial/genetics , Genetic Loci , Spores, Bacterial/physiology , Suppression, Genetic , Bacterial Proteins/metabolism , Clostridioides difficile/pathogenicity , Clostridioides difficile/physiology , Mutation , Promoter Regions, Genetic , Protein Binding , Spores, Bacterial/genetics
6.
Protein Expr Purif ; 174: 105662, 2020 10.
Article in English | MEDLINE | ID: mdl-32387144

ABSTRACT

The tobacco etch virus (TEV) protease has become a popular choice for cleaving fusion proteins because of its high stringency in sequence recognition. Procedures for isolating recombinant protein from the cytoplasm of E. coli require rupturing of the cell wall via enzymatic treatment combined with sonication or French press. Here we present an expedited method for producing laboratory-grade TEV protease in E. coli using a freeze-thaw method, followed by purification with immobilized metal affinity chromatography. Protease is obtained by expression from the pDZ2087 plasmid in BL21 (DE3) cells. Proteolysis resulting from this product, cleaves a maltose-binding protein fusion to completion at a fusion-to-protease molar ratio of 50:1.


Subject(s)
Endopeptidases , Escherichia coli , Gene Expression , Endopeptidases/biosynthesis , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/isolation & purification , Escherichia coli/enzymology , Escherichia coli/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
7.
Anaerobe ; 59: 1-7, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31077800

ABSTRACT

Clostridioides difficile is a Gram-positive, anaerobic bacterium. It is known that C. difficile is one of the major causes of antibiotic associated diarrhea. The enhanced antibiotic resistance observed in C. difficile is the result of highly resistant spores produced by the bacterium. In Bacillus subtilis, the sin operon is involved in sporulation inhibition. Two proteins coded within this operon, SinR and SinI, have an antagonistic relationship; SinR acts as an inhibitor to sporulation whereas SinI represses the activity of SinR, thus allowing the bacterium to sporulate. In a previous study, we examined the sin locus in C. difficile and named the two genes associated with this operon sinR and sinR', analogous to sinR and sinI in B. subtilis, respectively. We have shown that SinR and SinR' have pleiotropic roles in pathogenesis pathways and interact antagonistically with each other. Unlike B. subtilis SinI, SinR' in C. difficile carries two domains: the HTH domain and the Multimerization Domain (MD). In this study, we first performed a GST Pull-down experiment to determine the domain within SinR' that interacts with SinR. Second, the effect of these two domains on three phenotypes; sporulation, motility, and toxin production was examined. The findings of this study confirmed the prediction that the Multimerization Domain (MD) of SinR' is responsible for the interaction between SinR and SinR'. It was also discovered that SinR' regulates sporulation, toxin production and motility primarily by inhibiting SinR activity through the Multimerization Domain (MD).


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/biosynthesis , Clostridioides difficile/growth & development , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial , Locomotion , Spores, Bacterial/growth & development , Protein Binding , Protein Interaction Mapping
8.
PLoS Pathog ; 14(3): e1006940, 2018 03.
Article in English | MEDLINE | ID: mdl-29529083

ABSTRACT

Clostridium difficile is the primary cause of nosocomial diarrhea and pseudomembranous colitis. It produces dormant spores, which serve as an infectious vehicle responsible for transmission of the disease and persistence of the organism in the environment. In Bacillus subtilis, the sin locus coding SinR (113 aa) and SinI (57 aa) is responsible for sporulation inhibition. In B. subtilis, SinR mainly acts as a repressor of its target genes to control sporulation, biofilm formation, and autolysis. SinI is an inhibitor of SinR, so their interaction determines whether SinR can inhibit its target gene expression. The C. difficile genome carries two sinR homologs in the operon that we named sinR and sinR', coding for SinR (112 aa) and SinR' (105 aa), respectively. In this study, we constructed and characterized sin locus mutants in two different C. difficile strains R20291 and JIR8094, to decipher the locus's role in C. difficile physiology. Transcriptome analysis of the sinRR' mutants revealed their pleiotropic roles in controlling several pathways including sporulation, toxin production, and motility in C. difficile. Through various genetic and biochemical experiments, we have shown that SinR can regulate transcription of key regulators in these pathways, which includes sigD, spo0A, and codY. We have found that SinR' acts as an antagonist to SinR by blocking its repressor activity. Using a hamster model, we have also demonstrated that the sin locus is needed for successful C. difficile infection. This study reveals the sin locus as a central link that connects the gene regulatory networks of sporulation, toxin production, and motility; three key pathways that are important for C. difficile pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Movement/physiology , Clostridioides difficile/metabolism , Clostridium Infections/microbiology , Operon , Spores, Bacterial/physiology , Amino Acid Sequence , Animals , Bacillus subtilis/genetics , Bacillus subtilis/growth & development , Bacillus subtilis/metabolism , Bacterial Proteins/genetics , Cecum/metabolism , Cecum/microbiology , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Clostridium Infections/genetics , Clostridium Infections/metabolism , Gene Expression Regulation, Bacterial , Mesocricetus , Mice , Rabbits , Regulon , Sequence Homology
9.
mSphere ; 2(1)2017.
Article in English | MEDLINE | ID: mdl-28217744

ABSTRACT

Clostridium difficile is an important nosocomial pathogen and the leading cause of hospital-acquired diarrhea. Antibiotic use is the primary risk factor for the development of C. difficile-associated disease because it disrupts normally protective gut flora and enables C. difficile to colonize the colon. C. difficile damages host tissue by secreting toxins and disseminates by forming spores. The toxin-encoding genes, tcdA and tcdB, are part of a pathogenicity locus, which also includes the tcdR gene that codes for TcdR, an alternate sigma factor that initiates transcription of tcdA and tcdB genes. We created a tcdR mutant in epidemic-type C. difficile strain R20291 in an attempt to identify the global role of tcdR. A site-directed mutation in tcdR affected both toxin production and sporulation in C. difficile R20291. Spores of the tcdR mutant were more heat sensitive than the wild type (WT). Nearly 3-fold more taurocholate was needed to germinate spores from the tcdR mutant than to germinate the spores prepared from the WT strain. Transmission electron microscopic analysis of the spores also revealed a weakly assembled exosporium on the tcdR mutant spores. Accordingly, comparative transcriptome analysis showed many differentially expressed sporulation genes in the tcdR mutant compared to the WT strain. These data suggest that regulatory networks of toxin production and sporulation in C. difficile strain R20291 are linked with each other. IMPORTANCEC. difficile infects thousands of hospitalized patients every year, causing significant morbidity and mortality. C. difficile spores play a pivotal role in the transmission of the pathogen in the hospital environment. During infection, the spores germinate, and the vegetative bacterial cells produce toxins that damage host tissue. Thus, sporulation and toxin production are two important traits of C. difficile. In this study, we showed that a mutation in tcdR, the toxin gene regulator, affects both toxin production and sporulation in epidemic-type C. difficile strain R20291.

10.
PLoS One ; 11(10): e0165579, 2016.
Article in English | MEDLINE | ID: mdl-27768767

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0160107.].

11.
PLoS One ; 11(7): e0160107, 2016.
Article in English | MEDLINE | ID: mdl-27467167

ABSTRACT

Clostridium difficile is the principal cause of antibiotic-associated diarrhea. Major metabolic requirements for colonization and expansion of C. difficile after microbiota disturbance have not been fully determined. In this study, we show that glutamate utilization is important for C. difficile to establish itself in the animal gut. When the gluD gene, which codes for glutamate dehydrogenase (GDH), was disrupted, the mutant C. difficile was unable to colonize and cause disease in a hamster model. Further, from the complementation experiment it appears that extracellular GDH may be playing a role in promoting C. difficile colonization and disease progression. Quantification of free amino acids in the hamster gut during C. difficile infection showed that glutamate is among preferred amino acids utilized by C. difficile during its expansion. This study provides evidence of the importance of glutamate metabolism for C. difficile pathogenesis.


Subject(s)
Clostridioides difficile/enzymology , Glutamate Dehydrogenase/metabolism , Animals , Clostridioides difficile/growth & development , Cricetinae
12.
Appl Microbiol Biotechnol ; 100(1): 279-88, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26433970

ABSTRACT

D-lactic acid is used as a monomer in the production of poly-D-lactic acid (PDLA), which is used to form heat-resistant stereocomplex poly-lactic acid. To produce cost-effective D-lactic acid by using all sugars derived from biomass efficiently, xylose-assimilating genes encoding xylose isomerase and xylulokinase were cloned into an L-lactate-deficient strain, Lactobacillus plantarum. The resulting recombinant strain, namely L. plantarum NCIMB 8826 ∆ldhL1-pLEM-xylAB, was able to produce D-lactic acid (at optical purity >99 %) from xylose at a yield of 0.53 g g(-1). Simultaneous utilization of glucose and xylose to produce D-lactic acid was also achieved by this strain, and 47.2 g L(-1) of D-lactic acid was produced from 37.5 g L(-1) glucose and 19.7 g L(-1) xylose. Corn stover and soybean meal extract (SBME) were evaluated as cost-effective medium components for D-lactic acid production. Optimization of medium composition using response surface methodology resulted in 30 % reduction in enzyme loading and 70 % reduction in peptone concentration. In addition, we successfully demonstrated D-lactic acid fermentation from corn stover and SBME in a fed-batch fermentation, which yielded 61.4 g L(-1) D-lactic acid with an overall yield of 0.77 g g(-1). All these approaches are geared to attaining high D-lactic acid production from biomass sugars to produce low-cost, highly thermostable biodegradable plastics.


Subject(s)
Lactic Acid/metabolism , Lactobacillus plantarum/genetics , Lactobacillus plantarum/metabolism , Metabolic Engineering/methods , Xylose/metabolism , Aldose-Ketose Isomerases/genetics , Aldose-Ketose Isomerases/metabolism , Batch Cell Culture Techniques , Biomass , Biotransformation , Culture Media/chemistry , Fermentation , Glucose/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Glycine max/metabolism , Zea mays/metabolism
13.
J Bacteriol ; 197(15): 2600-9, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26013487

ABSTRACT

UNLABELLED: Clostridium difficile is a major nosocomial pathogen and the principal causative agent of antibiotic-associated diarrhea. The toxigenic C. difficile strains that cause disease secrete virulence factors, toxin A and toxin B, that cause colonic injury and inflammation. C. difficile toxins have no export signature and are secreted by an unusual mechanism that involves TcdE, a holin-like protein. We isolated a TcdE mutant of the epidemic R20291 strain with impaired toxin secretion, which was restored by complementation with functional TcdE. In the TcdE open reading frame (ORF), we identified three possible translation start sites; each translated isoform may play a specific role in TcdE-controlled toxin release. We created plasmid constructs that express only one of the three TcdE isoforms and complemented the TcdE mutant with these isoforms. Western blot analysis of the complemented strains demonstrated that TcdE is translated efficiently from the start codon at the 25th and 27th positions in the predicted ORF, producing proteins with 142 amino acids (TcdE142) and 140 amino acids (TcdE140), respectively. TcdE166 was not detected when expressed from its own ribosomal binding site (RBS). The effects of all three TcdE isoforms on C. difficile cell viability and toxin release were determined. Among the three isoforms, overexpression of TcdE166 and TcdE142 had a profound effect on cell viability compared to the TcdE140 isoform. Similarly, TcdE166 and TcdE142 facilitated toxin release more efficiently than did TcdE140. The importance of these variations among TcdE isoforms and their role in toxin release are discussed. IMPORTANCE: C. difficile is a nosocomial pathogen that has become the most prevalent cause of antibiotic-associated diarrhea in North America and in several countries in Europe. Most strains of C. difficile produce two high-molecular-weight toxins that are regarded as the primary virulence factors. The mechanism by which these large toxins are secreted from bacterial cells is not yet clear but involves TcdE, a holin-like protein. In this work, we show that TcdE could be translated from three different start codons, resulting in the production of three TcdE isoforms. Furthermore, we investigated the role of these isoforms in toxin release and cell lysis in C. difficile. An understanding of TcdE-dependent toxin secretion may be helpful for the development of strategies for preventing and treating C. difficile infections.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Base Sequence , Gene Expression Regulation, Bacterial/physiology , Humans , Molecular Sequence Data , Mutation , Protein Isoforms
14.
Microbiology (Reading) ; 160(Pt 1): 47-55, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24145018

ABSTRACT

Clostridium difficile produces an NAD-specific glutamate dehydrogenase (GDH), which converts l-glutamate into α-ketoglutarate through an irreversible reaction. The enzyme GDH is detected in the stool samples of patients with C. difficile-associated disease and serves as one of the diagnostic tools to detect C. difficile infection (CDI). We demonstrate here that supernatant fluids of C. difficile cultures contain GDH. To understand the role of GDH in the physiology of C. difficile, an isogenic insertional mutant of gluD was created in strain JIR8094. The mutant failed to produce and secrete GDH as shown by Western blot analysis. Various phenotypic assays were performed to understand the importance of GDH in C. difficile physiology. In TY (tryptose yeast extract) medium, the gluD mutant grew slower than the parent strain. Complementation of the gluD mutant with the functional gluD gene reversed the growth defect in TY medium. The presence of extracellular GDH may have a functional role in the pathogenesis of CDI. In support of this assumption we found higher sensitivity to H2O2 in the gluD mutant as compared to the parent strain. Complementation of the gluD mutant with the functional gluD gene reversed the H2O2 sensitivity.


Subject(s)
Clostridioides difficile/drug effects , Clostridioides difficile/enzymology , Drug Resistance, Bacterial , Glutamate Dehydrogenase/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Culture Media/chemistry , Gene Knockout Techniques , Genetic Complementation Test , Glutamate Dehydrogenase/genetics , Mutagenesis, Insertional
15.
J Bacteriol ; 195(18): 4246-54, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23873908

ABSTRACT

Toxigenic Clostridium sordellii causes uncommon but highly lethal infections in humans and animals. Recently, an increased incidence of C. sordellii infections has been reported in women undergoing obstetric interventions. Pathogenic strains of C. sordellii produce numerous virulence factors, including sordellilysin, phospholipase, neuraminidase, and two large clostridial glucosylating toxins, TcsL and TcsH. Recent studies have demonstrated that TcsL toxin is an essential virulence factor for the pathogenicity of C. sordellii. In this study, we identified and characterized TcsR as the toxin gene (tcsL) regulator in C. sordellii. High-throughput sequencing of two C. sordellii strains revealed that tcsR lies within a genomic region that encodes TcsL, TcsH, and TcsE, a putative holin. By using ClosTron technology, we inactivated the tcsR gene in strain ATCC 9714. Toxin production and tcsL transcription were decreased in the tcsR mutant strain. However, the complemented tcsR mutant produced large amounts of toxins, similar to the parental strain. Expression of the Clostridium difficile toxin gene regulator tcdR also restored toxin production to the C. sordellii tcsR mutant, showing that these sigma factors are functionally interchangeable.


Subject(s)
Bacterial Proteins , Bacterial Toxins/metabolism , Gene Expression Regulation, Bacterial , Sigma Factor , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Clostridium Infections/microbiology , Clostridium sordellii/genetics , Clostridium sordellii/metabolism , Clostridium sordellii/pathogenicity , Female , Genes, Regulator , Genome, Bacterial , High-Throughput Nucleotide Sequencing , Humans , Molecular Sequence Data , Sequence Analysis, DNA , Sigma Factor/chemistry , Sigma Factor/genetics , Sigma Factor/metabolism , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism
16.
PLoS Pathog ; 8(6): e1002727, 2012.
Article in English | MEDLINE | ID: mdl-22685398

ABSTRACT

The pathogenesis of Clostridium difficile, the major cause of antibiotic-associated diarrhea, is mainly associated with the production and activities of two major toxins. In many bacteria, toxins are released into the extracellular environment via the general secretion pathways. C. difficile toxins A and B have no export signature and their secretion is not explainable by cell lysis, suggesting that they might be secreted by an unusual mechanism. The TcdE protein encoded within the C. difficile pathogenicity locus (PaLoc) has predicted structural features similar to those of bacteriophage holin proteins. During many types of phage infection, host lysis is driven by an endolysin that crosses the cytoplasmic membrane through a pore formed by holin oligomerization. We demonstrated that TcdE has a holin-like activity by functionally complementing a λ phage deprived of its holin. Similar to λ holin, TcdE expressed in Escherichia coli and C. difficile formed oligomers in the cytoplamic membrane. A C. difficile tcdE mutant strain grew at the same rate as the wild-type strain, but accumulated a dramatically reduced amount of toxin proteins in the medium. However, the complemented tcdE mutant released the toxins efficiently. There was no difference in the abundance of tcdA and tcdB transcripts or of several cytoplasmic proteins in the mutant and the wild-type strains. In addition, TcdE did not overtly affect membrane integrity of C. difficile in the presence of TcdA/TcdB. Thus, TcdE acts as a holin-like protein to facilitate the release of C. difficile toxins to the extracellular environment, but, unlike the phage holins, does not cause the non-specific release of cytosolic contents. TcdE appears to be the first example of a bacterial protein that releases toxins into the environment by a phage-like system.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Enterotoxins/metabolism , Gene Expression Regulation, Bacterial/physiology , Amino Acid Sequence , Animals , Bacterial Toxins , Base Sequence , Blotting, Western , Chlorocebus aethiops , Clostridioides difficile/genetics , Clostridium Infections/genetics , Clostridium Infections/metabolism , Flow Cytometry , Gene Knockdown Techniques , Molecular Sequence Data , Vero Cells
17.
PLoS Pathog ; 7(10): e1002317, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22022270

ABSTRACT

Nosocomial infections are increasingly being recognised as a major patient safety issue. The modern hospital environment and associated health care practices have provided a niche for the rapid evolution of microbial pathogens that are well adapted to surviving and proliferating in this setting, after which they can infect susceptible patients. This is clearly the case for bacterial pathogens such as Methicillin Resistant Staphylococcus aureus (MRSA) and Vancomycin Resistant Enterococcus (VRE) species, both of which have acquired resistance to antimicrobial agents as well as enhanced survival and virulence properties that present serious therapeutic dilemmas for treating physicians. It has recently become apparent that the spore-forming bacterium Clostridium difficile also falls within this category. Since 2000, there has been a striking increase in C. difficile nosocomial infections worldwide, predominantly due to the emergence of epidemic or hypervirulent isolates that appear to possess extended antibiotic resistance and virulence properties. Various hypotheses have been proposed for the emergence of these strains, and for their persistence and increased virulence, but supportive experimental data are lacking. Here we describe a genetic approach using isogenic strains to identify a factor linked to the development of hypervirulence in C. difficile. This study provides evidence that a naturally occurring mutation in a negative regulator of toxin production, the anti-sigma factor TcdC, is an important factor in the development of hypervirulence in epidemic C. difficile isolates, presumably because the mutation leads to significantly increased toxin production, a contentious hypothesis until now. These results have important implications for C. difficile pathogenesis and virulence since they suggest that strains carrying a similar mutation have the inherent potential to develop a hypervirulent phenotype.


Subject(s)
Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/biosynthesis , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/microbiology , Enterotoxins/biosynthesis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Bacterial Toxins/genetics , Chlorocebus aethiops , Cloning, Molecular , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Cricetinae , Cross Infection/microbiology , Enterotoxins/genetics , Mesocricetus , Mutation , Plasmids , Repressor Proteins/biosynthesis , Vero Cells , Virulence Factors/metabolism
18.
J Virol ; 83(23): 12037-45, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776116

ABSTRACT

Clostridium difficile has been identified as the most important single identifiable cause of nosocomial antibiotic-associated diarrhea and colitis. Virulent strains of C. difficile produce two large protein toxins, toxin A and toxin B, which are involved in pathogenesis. In this study, we examined the effect of lysogeny by PhiCD119 on C. difficile toxin production. Transcriptional analysis demonstrated a decrease in the expression of pathogenicity locus (PaLoc) genes tcdA, tcdB, tcdR, tcdE, and tcdC in PhiCD119 lysogens. During this study we found that repR, a putative repressor gene of PhiCD119, was expressed in C. difficile lysogens and that its product, RepR, could downregulate tcdA::gusA and tcdR::gusA reporter fusions in Escherichia coli. We cloned and purified a recombinant RepR containing a C-terminal six-His tag and documented its binding to the upstream regions of tcdR in C. difficile PaLoc and in repR upstream region in PhiCD119 by gel shift assays. DNA footprinting experiments revealed similarities between the RepR binding sites in tcdR and repR upstream regions. These findings suggest that presence of a CD119-like temperate phage can influence toxin gene regulation in this nosocomially important pathogen.


Subject(s)
Bacterial Toxins/biosynthesis , Bacteriophages/genetics , Clostridioides difficile/physiology , Clostridioides difficile/virology , Gene Expression Regulation, Bacterial , Prophages/genetics , Artificial Gene Fusion , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , DNA Footprinting , DNA, Bacterial/metabolism , Electrophoretic Mobility Shift Assay , Genes, Reporter , Glucuronidase/genetics , Glucuronidase/metabolism , Humans , Lysogeny , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , Repressor Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/isolation & purification , Viral Proteins/metabolism
19.
J Bacteriol ; 188(10): 3716-20, 2006 May.
Article in English | MEDLINE | ID: mdl-16672625

ABSTRACT

Clostridium difficile produces two toxins, A and B, which act together to cause pseudomembraneous colitis. The genes encoding these toxins, tcdA and tcdB, are part of the pathogenicity locus, which also includes tcdC, a putative negative regulator of the toxin genes. In this study, we demonstrate that TcdC is a membrane-associated protein in C. difficile.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Membrane Proteins/metabolism , Repressor Proteins/metabolism , Adenosine Triphosphatases/metabolism , Antibodies , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Toxins/genetics , Base Sequence , Clostridioides difficile/genetics , DNA Primers , Kinetics , Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/isolation & purification
20.
J Bacteriol ; 188(7): 2568-77, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16547044

ABSTRACT

In this study, we have isolated a temperate phage (PhiCD119) from a pathogenic Clostridium difficile strain and sequenced and annotated its genome. This virus has an icosahedral capsid and a contractile tail covered by a sheath and contains a double-stranded DNA genome. It belongs to the Myoviridae family of the tailed phages and the order Caudovirales. The genome was circularly permuted, with no physical ends detected by sequencing or restriction enzyme digestion analysis, and lacked a cos site. The DNA sequence of this phage consists of 53,325 bp, which carries 79 putative open reading frames (ORFs). A function could be assigned to 23 putative gene products, based upon bioinformatic analyses. The PhiCD119 genome is organized in a modular format, which includes modules for lysogeny, DNA replication, DNA packaging, structural proteins, and host cell lysis. The PhiCD119 attachment site attP lies in a noncoding region close to the putative integrase (int) gene. We have identified the phage integration site on the C. difficile chromosome (attB) located in a noncoding region just upstream of gene gltP, which encodes a carrier protein for glutamate and aspartate. This genetic analysis represents the first complete DNA sequence and annotation of a C. difficile phage.


Subject(s)
Bacteriophages/classification , Bacteriophages/genetics , Clostridioides difficile/virology , Genome, Viral , DNA Replication/genetics , DNA, Viral , Genes, Viral/genetics , Lysogeny , Open Reading Frames/genetics , Recombination, Genetic/genetics , Virus Assembly/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...