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










Publication year range
1.
Mol Microbiol ; 112(5): 1453-1470, 2019 11.
Article in English | MEDLINE | ID: mdl-31483905

ABSTRACT

D-Ala-D-Ala ligase, encoded by ddl genes, is responsible for the synthesis of a dipeptide, D-Ala-D-Ala, an essential precursor of bacterial peptidoglycan. In Clostridioides difficile, the single ddl gene is located upstream of the ddlR gene, which encodes a putative transcriptional regulator. Using mutational and transcriptional analysis and DNA-binding assays, DdlR was found to be a direct activator of the ddl ddlR operon. DdlR is a member of the MocR/GabR-type proteins that have aminotransferase-like, pyridoxal 5'-phosphate-binding domains. A DdlR mutation that prevented covalent binding of pyridoxal 5'-phosphate abolished the ability of DdlR to activate transcription. Addition of D-Ala-D-Ala to the medium inactivated DdlR, reducing dipeptide biosynthesis. In contrast, D-Ala-D-Ala limitation caused a dramatic increase in expression from the ddl promoter. Though uncommon for transcription regulators, C. difficile DdlR is essential, as the ddlR null mutant cells could not grow even in complex laboratory media in the absence of D-Ala-D-Ala. A dyad symmetry sequence, which is located immediately upstream of the -35 region of the ddl promoter, serves as an important element of the DdlR-binding site. This sequence is conserved upstream of putative DdlR targets in other bacteria of classes Clostridia and Bacilli, indicating a similar mode of regulation of these genes.


Subject(s)
Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial/genetics , Peptide Synthases/genetics , Peptidoglycan/biosynthesis , Clostridioides difficile/genetics , DNA-Binding Proteins/genetics , Peptide Synthases/metabolism , Promoter Regions, Genetic/genetics , Transcriptional Activation/genetics
2.
Mol Microbiol ; 111(6): 1671-1688, 2019 06.
Article in English | MEDLINE | ID: mdl-30882947

ABSTRACT

For the human pathogen Clostridioides (also known as Clostridium) difficile, the ability to adapt to nutrient availability is critical for its proliferation and production of toxins during infection. Synthesis of the toxins is regulated by the availability of certain carbon sources, fermentation products and amino acids (e.g. proline, cysteine, isoleucine, leucine and valine). The effect of proline is attributable at least in part to its role as an inducer and substrate of D-proline reductase (PR), a Stickland reaction that regenerates NAD+ from NADH. Many Clostridium spp. use Stickland metabolism (co-fermentation of pairs of amino acids) to generate ATP and NAD+ . Synthesis of PR is activated by PrdR, a proline-responsive regulatory protein. Here we report that PrdR, in the presence of proline, represses other NAD+ -generating pathways, such as the glycine reductase and succinate-acetyl CoA utilization pathways leading to butyrate production, but does so indirectly by affecting the activity of Rex, a global redox-sensing regulator that responds to the NAD+ /NADH ratio. Our results indicate that PR activity is the favored mechanism for NAD+ regeneration and that both Rex and PrdR influence toxin production. Using the hamster model of C. difficile infection, we revealed the importance of PrdR-regulated Stickland metabolism in the virulence of C. difficile.


Subject(s)
Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial , Gene Products, rex/genetics , NAD/metabolism , Proline/metabolism , Amino Acid Oxidoreductases/metabolism , Animals , Clostridioides difficile/pathogenicity , Female , Gene Products, rex/antagonists & inhibitors , Mesocricetus , Multienzyme Complexes , Oxidation-Reduction , Regeneration , Virulence
3.
PLoS One ; 14(1): e0206896, 2019.
Article in English | MEDLINE | ID: mdl-30699117

ABSTRACT

Toxin synthesis and endospore formation are two of the most critical factors that determine the outcome of infection by Clostridioides difficile. The two major toxins, TcdA and TcdB, are the principal factors causing damage to the host. Spores are the infectious form of C. difficile, permit survival of the bacterium during antibiotic treatment and are the predominant cell form that leads to recurrent infection. Toxin production and sporulation have their own specific mechanisms of regulation, but they share negative regulation by the global regulatory protein CodY. Determining the extent of such regulation and its detailed mechanism is important for understanding the linkage between two apparently independent biological phenomena and raises the possibility of creating new ways of limiting infection. The work described here shows that a codY null mutant of a hypervirulent (ribotype 027) strain is even more virulent than its parent in a mouse model of infection and that the mutant expresses most sporulation genes prematurely during exponential growth phase. Moreover, examining the expression patterns of mutants producing CodY proteins with different levels of residual activity revealed that expression of the toxin genes is dependent on total CodY inactivation, whereas most sporulation genes are turned on when CodY activity is only partially diminished. These results suggest that, in wild-type cells undergoing nutrient limitation, sporulation genes can be turned on before the toxin genes.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Ribotyping , Spores, Bacterial/physiology , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins/toxicity , Biosynthetic Pathways/drug effects , Biosynthetic Pathways/genetics , Clostridioides difficile/genetics , Diarrhea/microbiology , Ethanolamine/metabolism , Gene Expression Regulation, Bacterial/drug effects , Genes, Bacterial , Mice, Inbred C57BL , Multigene Family , Operon/genetics , Point Mutation/genetics , Protein Domains , Spores, Bacterial/genetics , Transcription, Genetic/drug effects , Virulence/genetics
4.
Infect Immun ; 86(11)2018 11.
Article in English | MEDLINE | ID: mdl-30150259

ABSTRACT

The symptoms of Clostridium difficile infection (CDI) are attributed largely to two C. difficile toxins, TcdA and TcdB. Significant efforts have been devoted to developing vaccines targeting both toxins through parenteral immunization routes. However, C. difficile is an enteric pathogen, and mucosal/oral immunization would be particularly useful to protect the host against CDI, considering that the gut is the main site of disease onset and progression. Moreover, vaccines directed only against toxins do not target the cells and spores that transmit the disease. Previously, we constructed a chimeric vaccine candidate, mTcd138, comprised of the glucosyltransferase and cysteine proteinase domains of TcdB and the receptor binding domain of TcdA. In this study, to develop an oral vaccine that can target both C. difficile toxins and colonization/adhesion factors, we expressed mTcd138 in a nontoxigenic C. difficile (NTCD) strain, resulting in strain NTCD_mTcd138. Oral immunization with spores of NTCD_mTcd138 provided mice full protection against infection with a hypervirulent C. difficile strain, UK6 (ribotype 027). The protective strength and efficacy of NTCD_mTcd138 were further evaluated in the acute CDI hamster model. Oral immunization with spores of NTCD_mTcd138 also provided hamsters significant protection against infection with 2 × 104 UK6 spores, a dose 200-fold higher than the lethal dose of UK6 in hamsters. These results imply that the genetically modified, nontoxigenic C. difficile strain expressing mTcd138 may represent a novel mucosal vaccine candidate against CDI.


Subject(s)
Bacterial Proteins/immunology , Bacterial Toxins/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Clostridioides difficile/immunology , Clostridium Infections/prevention & control , Enterotoxins/immunology , Administration, Oral , Animals , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Bacterial Vaccines/genetics , Clostridioides difficile/genetics , Clostridium Infections/immunology , Cricetinae , Disease Models, Animal , Enterotoxins/genetics , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
5.
Sci Rep ; 7(1): 14672, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116155

ABSTRACT

Clostridium difficile is a significant concern as a nosocomial pathogen, and genetic tools are important when analyzing the physiology of such organisms so that the underlying physiology/pathogenesis of the organisms can be studied. Here, we used TargeTron to investigate the role of selenoproteins in C. difficile Stickland metabolism and found that a TargeTron insertion into selD, encoding the selenophosphate synthetase that is essential for the specific incorporation of selenium into selenoproteins, results in a significant growth defect and a global loss of selenium incorporation. However, because of potential polar effects of the TargeTron insertion, we developed a CRISPR-Cas9 mutagenesis system for C. difficile. This system rapidly and efficiently introduces site-specific mutations into the C. difficile genome (20-50% mutation frequency). The selD CRISPR deletion mutant had a growth defect in protein-rich medium and mimicked the phenotype of a generated TargeTron selD mutation. Our findings suggest that Stickland metabolism could be a target for future antibiotic therapies and that the CRISPR-Cas9 system can introduce rapid and efficient modifications into the C. difficile genome.


Subject(s)
Clostridioides difficile/metabolism , Gene Editing/methods , Selenoproteins/metabolism , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Clostridioides difficile/genetics , Electrophoresis, Polyacrylamide Gel , Genes, Bacterial/genetics , Phosphotransferases/genetics , Phosphotransferases/metabolism , Selenium/metabolism , Selenoproteins/genetics
6.
Infect Immun ; 84(8): 2389-405, 2016 08.
Article in English | MEDLINE | ID: mdl-27297391

ABSTRACT

The pathogenicity of Clostridium difficile is linked to its ability to produce two toxins: TcdA and TcdB. The level of toxin synthesis is influenced by environmental signals, such as phosphotransferase system (PTS) sugars, biotin, and amino acids, especially cysteine. To understand the molecular mechanisms of cysteine-dependent repression of toxin production, we reconstructed the sulfur metabolism pathways of C. difficile strain 630 in silico and validated some of them by testing C. difficile growth in the presence of various sulfur sources. High levels of sulfide and pyruvate were produced in the presence of 10 mM cysteine, indicating that cysteine is actively catabolized by cysteine desulfhydrases. Using a transcriptomic approach, we analyzed cysteine-dependent control of gene expression and showed that cysteine modulates the expression of genes involved in cysteine metabolism, amino acid biosynthesis, fermentation, energy metabolism, iron acquisition, and the stress response. Additionally, a sigma factor (SigL) and global regulators (CcpA, CodY, and Fur) were tested to elucidate their roles in the cysteine-dependent regulation of toxin production. Among these regulators, only sigL inactivation resulted in the derepression of toxin gene expression in the presence of cysteine. Interestingly, the sigL mutant produced less pyruvate and H2S than the wild-type strain. Unlike cysteine, the addition of 10 mM pyruvate to the medium for a short time during the growth of the wild-type and sigL mutant strains reduced expression of the toxin genes, indicating that cysteine-dependent repression of toxin production is mainly due to the accumulation of cysteine by-products during growth. Finally, we showed that the effect of pyruvate on toxin gene expression is mediated at least in part by the two-component system CD2602-CD2601.


Subject(s)
Clostridioides difficile/physiology , Cysteine/metabolism , Enterocolitis, Pseudomembranous/microbiology , Amino Acids/metabolism , Animals , Bacterial Toxins/biosynthesis , Bacterial Toxins/genetics , Cell Line , Chlorocebus aethiops , Energy Metabolism/genetics , Gene Expression Regulation, Bacterial , Homocysteine/metabolism , Hydrogen Sulfide/metabolism , Intracellular Space/metabolism , Metabolic Networks and Pathways , Pyruvic Acid/metabolism , Vero Cells
7.
J Bacteriol ; 198(15): 2113-30, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27246573

ABSTRACT

UNLABELLED: Clostridium difficile must form a spore to survive outside the gastrointestinal tract. The factors that trigger sporulation in C. difficile remain poorly understood. Previous studies have suggested that a link exists between nutritional status and sporulation initiation in C. difficile In this study, we investigated the impact of the global nutritional regulator CodY on sporulation in C. difficile strains from the historical 012 ribotype and the current epidemic 027 ribotype. Sporulation frequencies were increased in both backgrounds, demonstrating that CodY represses sporulation in C. difficile The 027 codY mutant exhibited a greater increase in spore formation than the 012 codY mutant. To determine the role of CodY in the observed sporulation phenotypes, we examined several factors that are known to influence sporulation in C. difficile Using transcriptional reporter fusions and quantitative reverse transcription-PCR (qRT-PCR) analysis, we found that two loci associated with the initiation of sporulation, opp and sinR, are regulated by CodY. The data demonstrate that CodY is a repressor of sporulation in C. difficile and that the impact of CodY on sporulation and expression of specific genes is significantly influenced by the strain background. These results suggest that the variability of CodY-dependent regulation is an important contributor to virulence and sporulation in current epidemic isolates. This report provides further evidence that nutritional state, virulence, and sporulation are linked in C. difficile IMPORTANCE: This study sought to examine the relationship between nutrition and sporulation in C. difficile by examining the global nutritional regulator CodY. CodY is a known virulence and nutritional regulator of C. difficile, but its role in sporulation was unknown. Here, we demonstrate that CodY is a negative regulator of sporulation in two different ribotypes of C. difficile We also demonstrate that CodY regulates known effectors of sporulation, Opp and SinR. These results support the idea that nutrient limitation is a trigger for sporulation in C. difficile and that the response to nutrient limitation is coordinated by CodY. Additionally, we demonstrate that CodY has an altered role in sporulation regulation for some strains.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/physiology , Gene Expression Regulation, Bacterial/physiology , Repressor Proteins/metabolism , Spores, Bacterial/physiology , Bacterial Proteins/genetics , Mutation , Repressor Proteins/genetics
8.
Microb Cell ; 3(11): 573-575, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-28357327

ABSTRACT

Regulation of biological functions requires factors (proteins, peptides or chemicals) able to sense and translate environmental conditions or any circumstances in order to modulate the transcription of a gene, the stability of a transcript or the activity of a protein. Quorum sensing is a regulation mechanism connecting cell density to the physiological state of a single cell. In bacteria, quorum sensing coordinates virulence, cell fate and commitment to sporulation and other adaptation properties. The critical role of such regulatory systems was demonstrated in pathogenicity and adaptation of bacteria from the Bacillus cereus group (i.e. B. cereus and Bacillus thuringiensis). Furthermore, using insects as a model of infection, it was shown that sequential activation of several quorum sensing systems allowed bacteria to switch from a virulence state to a necrotrophic lifestyle, allowing their survival in the host cadaver, and ultimately to the commitment into sporulation. The chronological development of these physiological states is directed by quorum sensors forming the RNPP family. Among them, NprR combines two distinct functions connecting sporulation to necrotrophism in B. thuringiensis. In the absence of its cognate signaling peptide (NprX), NprR negatively controls sporulation by acting as a phosphatase. In the presence of NprX, it acts as a transcription factor regulating a set of genes involved in the survival of the bacteria in the insect cadaver.

9.
Antimicrob Agents Chemother ; 59(7): 4199-205, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941230

ABSTRACT

The increasing incidence and severity of infection by Clostridium difficile have stimulated attempts to develop new antimicrobial therapies. We report here the relative abilities of two antibiotics (metronidazole and vancomycin) in current use for treating C. difficile infection and of a third antimicrobial, surotomycin, to kill C. difficile cells at various stages of development and to inhibit the production of the toxin proteins that are the major virulence factors. The results indicate that none of the drugs affects the viability of spores at 8× MIC or 80× MIC and that all of the drugs kill exponential-phase cells when provided at 8× MIC. In contrast, none of the drugs killed stationary-phase cells or inhibited toxin production when provided at 8× MIC and neither vancomycin nor metronidazole killed stationary-phase cells when provided at 80× MIC. Surotomycin, on the other hand, did kill stationary-phase cells when provided at 80× MIC but did so without inducing lysis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/biosynthesis , Clostridioides difficile/drug effects , Clostridioides difficile/metabolism , Enterotoxins/biosynthesis , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Bacterial Toxins/genetics , Cell Wall/drug effects , Clostridioides difficile/genetics , Enterotoxins/genetics , Gene Expression Regulation, Bacterial/drug effects , Metronidazole/pharmacology , Microbial Sensitivity Tests , Mutation/genetics , Spores, Bacterial/drug effects , Vancomycin/pharmacology , Virulence Factors/metabolism
10.
Res Microbiol ; 166(4): 375-83, 2015 May.
Article in English | MEDLINE | ID: mdl-25445566

ABSTRACT

Synthesis of the major toxin proteins of the diarrheal pathogen, Clostridium difficile, is dependent on the activity of TcdR, an initiation (sigma) factor of RNA polymerase. The synthesis of TcdR and the activation of toxin gene expression are responsive to multiple components in the bacterium's nutritional environment, such as the presence of certain sugars, amino acids, and fatty acids. This review summarizes current knowledge about the mechanisms responsible for repression of toxin synthesis when glucose or branched-chain amino acids or proline are in excess and the pathways that lead to synthesis of butyrate, an activator of toxin synthesis. The regulatory proteins implicated in these mechanisms also play key roles in modulating bacterial metabolic pathways, suggesting that C. difficile pathogenesis is intimately connected to the bacterium's metabolic state.


Subject(s)
Bacterial Toxins/biosynthesis , Clostridioides difficile/growth & development , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial , Metabolic Networks and Pathways , Amino Acids/metabolism , Butyrates/metabolism , Clostridioides difficile/genetics , Fatty Acids/metabolism , Gene Regulatory Networks , Glucose/metabolism , Sigma Factor/metabolism , Virulence
11.
Infect Immun ; 82(10): 4080-91, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25024365

ABSTRACT

Clostridium difficile-associated disease (CDAD) constitutes a large majority of nosocomial diarrhea cases in industrialized nations and is mediated by the effects of two secreted toxins, toxin A (TcdA) and toxin B (TcdB). Patients who develop strong antitoxin antibody responses can clear C. difficile infection and remain disease free. Key toxin-neutralizing epitopes have been found within the carboxy-terminal receptor binding domains (RBDs) of TcdA and TcdB, which has generated interest in developing the RBD as a viable vaccine target. While numerous platforms have been studied, very little data describes the potential of DNA vaccination against CDAD. Therefore, we created highly optimized plasmids encoding the RBDs from TcdA and TcdB in which any putative N-linked glycosylation sites were altered. Mice and nonhuman primates were immunized intramuscularly, followed by in vivo electroporation, and in these animal models, vaccination induced significant levels of both anti-RBD antibodies (blood and stool) and RBD-specific antibody-secreting cells. Further characterization revealed that sera from immunized mice and nonhuman primates could detect RBD protein from transfected cells, as well as neutralize purified toxins in an in vitro cytotoxicity assay. Mice that were immunized with plasmids or given nonhuman-primate sera were protected from a lethal challenge with purified TcdA and/or TcdB. Moreover, immunized mice were significantly protected when challenged with C. difficile spores from homologous (VPI 10463) and heterologous, epidemic (UK1) strains. These data demonstrate the robust immunogenicity and efficacy of a TcdA/B RBD-based DNA vaccine in preclinical models of acute toxin-associated and intragastric, spore-induced colonic disease.


Subject(s)
Antibodies, Bacterial/blood , Antitoxins/blood , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Enterotoxins/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Cross Protection , Electrophoresis , Enterotoxins/genetics , Female , Injections, Intramuscular , Macaca mulatta , Mice , Mice, Inbred C57BL , Neutralization Tests , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Survival Analysis , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
12.
Int J Mol Sci ; 15(3): 4415-30, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24625665

ABSTRACT

Clostridium difficile is a nosocomial pathogen that causes a serious toxin-mediated enteric disease in humans. Reducing C. difficile toxin production could significantly minimize its pathogenicity and improve disease outcomes in humans. This study investigated the efficacy of two, food-grade, plant-derived compounds, namely trans-cinnamaldehyde (TC) and carvacrol (CR) in reducing C. difficile toxin production and cytotoxicity in vitro. Three hypervirulent C. difficile isolates were grown with or without the sub-inhibitory concentrations of TC or CR, and the culture supernatant and the bacterial pellet were collected for total toxin quantitation, Vero cell cytotoxicity assay and RT-qPCR analysis of toxin-encoding genes. The effect of CR and TC on a codY mutant and wild type C. difficile was also investigated. Carvacrol and TC substantially reduced C. difficile toxin production and cytotoxicity on Vero cells. The plant compounds also significantly down-regulated toxin production genes. Carvacrol and TC did not inhibit toxin production in the codY mutant of C. difficile, suggesting a potential codY-mediated anti-toxigenic mechanism of the plant compounds. The antitoxigenic concentrations of CR and TC did not inhibit the growth of beneficial gut bacteria. Our results suggest that CR and TC could potentially be used to control C. difficile, and warrant future studies in vivo.


Subject(s)
Acrolein/analogs & derivatives , Bacterial Toxins/biosynthesis , Clostridioides difficile/drug effects , Monoterpenes/pharmacology , Acrolein/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Cymenes , Gene Expression Regulation, Bacterial/drug effects , Microbial Sensitivity Tests , Microbial Viability/drug effects , Microbial Viability/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
J Bacteriol ; 195(3): 629-36, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23204460

ABSTRACT

Clostridium perfringens is an anaerobic Gram-positive pathogen that causes many human and animal diseases, including food poisoning and gas gangrene. C. perfringens lacks flagella but possesses type IV pili (TFP). We have previously shown that C. perfringens can glide across an agar surface in long filaments composed of individual bacteria attached end to end and that two TFP-associated proteins, PilT and PilC, are needed for this. To discover additional gene products that play a role in gliding, we developed a plasmid-based mariner transposon mutagenesis system that works effectively in C. perfringens. More than 10,000 clones were screened for mutants that lacked the ability to move away from the edge of a colony. Twenty-four mutants (0.24%) were identified that fit the criteria. The genes containing insertions that affected gliding motility fell into nine different categories. One gene, CPE0278, which encodes a homolog of the SagA cell wall-dependent endopeptidase, acquired distinct transposon insertions in two independent mutants. sagA mutants were unable to form filaments due to a complete lack of end-to-end connections essential for gliding motility. Complementation of the sagA mutants with a wild-type copy of the gene restored gliding motility. We constructed an in-frame deletion mutation in the sagA gene and found that this mutant had a phenotype similar to those of the transposon mutants. We hypothesize that the sagA mutant strains are unable to form the molecular complexes which are needed to keep the cells in an end-to-end orientation, leading to separation of daughter cells and the inability to carry out gliding motility.


Subject(s)
Clostridium perfringens/physiology , DNA Transposable Elements/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Movement/physiology , Transposases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chromosome Mapping , Chromosomes, Bacterial/genetics , Clostridium perfringens/classification , Clostridium perfringens/genetics , Clostridium perfringens/ultrastructure , DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic/physiology , Genetic Complementation Test , Mutagenesis , Mutation , Plasmids/genetics , RNA, Bacterial/genetics , RNA, Ribosomal/genetics , Transposases/genetics
14.
J Antimicrob Chemother ; 68(3): 515-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23208832

ABSTRACT

OBJECTIVES: Fidaxomicin, which was recently approved for the treatment of Clostridium difficile-associated diarrhoea, demonstrates narrow-spectrum bactericidal activity via inhibition of RNA polymerase. In this study we evaluated its inhibitory activity versus C. difficile toxin gene expression and toxin production by quantifying toxin mRNA and protein. METHODS: The effects of fidaxomicin, its major metabolite (OP-1118), vancomycin and metronidazole on toxin A and toxin B production were determined by assaying culture supernatants of two C. difficile isolates (ATCC 43255, a high-level toxin-producing strain, and UK-14, a NAP1/027/BI epidemic strain) using a commercial ELISA. The effects of the drugs on toxin gene expression were assessed in stationary-phase cells of C. difficile strain UK-1 (NAP1/027/BI type epidemic strain) and in the closely related non-epidemic strain CD196 by quantitative RT-PCR. RESULTS: Subinhibitory levels (1/4× MIC) of fidaxomicin or OP-1118 (but not vancomycin or metronidazole) strongly suppressed toxin production in C. difficile (≥ 60%) through at least 1 week of culture. Additionally, transcripts from the pathogenicity loci (tcdR, tcdA and tcdB) were nearly completely inhibited by both fidaxomicin (2× MIC) and OP-1118 (2.5× MIC), but not vancomycin (2.5× MIC). CONCLUSIONS: Both fidaxomicin and OP-1118 are able to inhibit toxin production in vitro, which may explain prior post-treatment observations of less frequent detectable toxin in fidaxomicin-treated patients (27 subjects) than those treated with vancomycin (8 patients).


Subject(s)
Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Clostridioides difficile/drug effects , Enterotoxins/antagonists & inhibitors , Bacterial Proteins/biosynthesis , Bacterial Toxins/biosynthesis , Culture Media/chemistry , Enterotoxins/biosynthesis , Enzyme-Linked Immunosorbent Assay , Fidaxomicin , Gene Expression/drug effects , Gene Expression Profiling , Humans , Metronidazole/metabolism , Vancomycin/metabolism
15.
J Bacteriol ; 195(4): 844-54, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23222730

ABSTRACT

Clostridium difficile, a proteolytic Gram-positive anaerobe, has emerged as a significant nosocomial pathogen. Stickland fermentation reactions are thought to be important for growth of C. difficile and appear to influence toxin production. In Stickland reactions, pairs of amino acids donate and accept electrons, generating ATP and reducing power in the process. Reduction of the electron acceptors proline and glycine requires the d-proline reductase (PR) and the glycine reductase (GR) enzyme complexes, respectively. Addition of proline in the medium increases the level of PR protein but decreases the level of GR. We report the identification of PrdR, a protein that activates transcription of the PR-encoding genes in the presence of proline and negatively regulates the GR-encoding genes. The results suggest that PrdR is a central metabolism regulator that controls preferential utilization of proline and glycine to produce energy via the Stickland reactions.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial/physiology , Proline/metabolism , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Bacterial Proteins/genetics , Clostridioides difficile/genetics , Escherichia coli , Fermentation , Gene Expression Regulation, Enzymologic/physiology , Glycine/metabolism , Molecular Structure , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Operon , Plasmids/genetics , Proline/chemistry
16.
Clin Infect Dis ; 55 Suppl 2: S162-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22752866

ABSTRACT

Fidaxomicin (FDX) is a novel antimicrobial agent with narrow-spectrum and potent bactericidal activity against Clostridium difficile. In recent clinical trials, FDX was superior to vancomycin in preventing recurrences of C. difficile infection. A possible mechanism of reducing recurrence may be through an inhibitory effect on sporulation. The effect of FDX and its major metabolite, OP-1118, on C. difficile growth and sporulation kinetics was compared with that of vancomycin, metronidazole, and rifaximin. Drugs at subminimum inhibitory concentrations (sub-MICs) were added to cells at an early stationary phase of growth; this was followed by collection of cells at various intervals for quantitation of total viable cell and heat-resistant spore counts on taurocholate-containing media. The effect of the drugs at 2-2.5× MIC on the expression of sporulation genes in C. difficile was also compared using quantitative reverse-transcriptase polymerase chain reaction. Both FDX and OP-1118 (1/4× MIC) inhibited sporulation when added to early-stationary-phase cells in C. difficile strains, including the epidemic NAP1/BI/027 strain. In contrast, vancomycin, metronidazole, and rifaximin (at similar sub-MICs) did not inhibit sporulation. The number of spores following treatment with comparator drugs increased to the same level as the no-drug control treatment. Expression of mother cell-specific (spoIIID) and forespore-specific (spoIIR) sporulation genes also was inhibited by FDX and OP-1118 but not significantly by vancomycin. Both FDX and OP-1118 (unlike vancomycin, rifaximin, and metronidazole) effectively inhibited sporulation by C. difficile. The inhibitory effect of FDX on C. difficile sporulation may contribute to its superior performance in sustaining clinical response and reducing recurrences and may also be beneficial in decreasing shedding and transmission of this pathogen.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Spores, Bacterial/drug effects , Bacterial Load , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Clostridioides difficile/isolation & purification , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Fidaxomicin , Gene Expression Regulation, Bacterial , Genes, Bacterial , Metronidazole/pharmacology , Microbial Sensitivity Tests , Rifamycins/pharmacology , Rifaximin , Spores, Bacterial/genetics , Spores, Bacterial/growth & development , Vancomycin/pharmacology
17.
Curr Protoc Microbiol ; Chapter 9: Unit 9A.2, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21400677

ABSTRACT

Clostridium difficile is a Gram-positive, spore forming, anaerobic, intestinal bacterium and is the most common cause of antibiotic-associated colitis. For many years this organism was considered genetically intractable, but in the past 10 years, multiple methods have been developed or adapted for genetic manipulation of C. difficile. This unit describes the molecular techniques used for genetic modification of this organism, including methods for gene disruption, complementation, plasmid introduction and integration, and cross-species conjugations.


Subject(s)
Clostridioides difficile/genetics , Genetic Techniques
18.
Proc Natl Acad Sci U S A ; 104(47): 18490-5, 2007 Nov 20.
Article in English | MEDLINE | ID: mdl-17998541

ABSTRACT

Gram-positive bacteria use a wealth of extracellular signaling peptides, so-called autoinducers, to regulate gene expression according to population densities. These "quorum sensing" systems control vital processes such as virulence, sporulation, and gene transfer. Using x-ray analysis, we determined the structure of PlcR, the major virulence regulator of the Bacillus cereus group, and obtained mechanistic insights into the effects of autoinducer binding. Our structural and phylogenetic analysis further suggests that all of those quorum sensors that bind directly to their autoinducer peptide derive from a common ancestor and form a single family (the RNPP family, for Rap/NprR/PlcR/PrgX) with conserved features. As a consequence, fundamentally different processes in different bacterial genera appear regulated by essentially the same autoinducer recognition mechanism. Our results shed light on virulence control by PlcR and elucidate origin and evolution of multicellular behavior in bacteria.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Evolution, Molecular , Gram-Positive Bacteria/metabolism , Gram-Positive Bacteria/pathogenicity , Quorum Sensing , Trans-Activators/chemistry , Trans-Activators/metabolism , Bacterial Proteins/classification , Bacterial Proteins/genetics , Crystallography, X-Ray , Gram-Positive Bacteria/chemistry , Gram-Positive Bacteria/genetics , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Structural Homology, Protein , Trans-Activators/classification , Trans-Activators/genetics , Virulence
19.
Proteomics ; 7(10): 1719-28, 2007 May.
Article in English | MEDLINE | ID: mdl-17486558

ABSTRACT

Using 2-DE, transcriptional gene fusions and cell cytotoxicity assays, we followed changes in the Bacillus cereus strain ATCC14579 secretome, gene expression and culture supernatant cytotoxicity from the end of the vegetative phase up to 5 h after entry into the stationary phase. The concentration of each of the 22 proteins in the culture supernatant was determined at various times. In addition, the stability of the proteins was studied. Fifteen of these proteins, including 14 members of the virulence regulon PlcR, were known or predicted to be secreted. All of the secreted proteins reached a maximum concentration during early stationary phase, but there were significant differences in the kinetics of their concentrations. The time courses of protein concentrations were in agreement with gene expression data, except for cytotoxin CytK, which was unstable, and for the metalloprotease InhA1. Supernatant cytoxicity also peaked in early stationary phase, and the kinetics of cytotoxicity paralleled the time course of concentration of the PlcR-controlled toxin, CytK. Our concomitant study of the time course of protein concentrations, gene expression and supernatant cytotoxicity reveals that the pathogenic potential of B. cereus peaks during the transition state. It also suggests that there is diversity in the regulation of gene expression within the PlcR regulon.


Subject(s)
Bacillus cereus/chemistry , Bacillus cereus/physiology , Bacterial Proteins/analysis , Bacillus cereus/metabolism , Bacterial Proteins/genetics , Electrophoresis, Gel, Two-Dimensional , Exocytosis , Gene Expression , Molecular Sequence Data , Time Factors
20.
Appl Environ Microbiol ; 71(12): 8903-10, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16332888

ABSTRACT

Bacillus thuringiensis and Bacillus cereus are closely related. B. thuringiensis is well known for its entomopathogenic properties, principally due to the synthesis of plasmid-encoded crystal toxins. B. cereus appears to be an emerging opportunistic human pathogen. B. thuringiensis and B. cereus produce many putative virulence factors which are positively controlled by the pleiotropic transcriptional regulator PlcR. The inactivation of plcR decreases but does not abolish virulence, indicating that additional factors like flagella may contribute to pathogenicity. Therefore, we further analyzed a mutant (B. thuringiensis 407 Cry(-) DeltaflhA) previously described as being defective in flagellar apparatus assembly and in motility as well as in the production of hemolysin BL and phospholipases. A large picture of secreted proteins was obtained by two-dimensional electrophoresis analysis, which revealed that flagellar proteins are not secreted and that production of several virulence-associated factors is reduced in the flhA mutant. Moreover, we quantified the effect of FlhA on plcA and hblC gene transcription. The results show that the flhA mutation results in a significant reduction of plcA and hblC transcription. These results indicate that the transcription of several PlcR-regulated virulence factors is coordinated with the flagellar apparatus. Consistently, the flhA mutant also shows a strong decrease in cytotoxicity towards HeLa cells and in virulence against Galleria mellonella larvae following oral and intrahemocoelic inoculation. The decrease in virulence may be due to both a lack of flagella and a lower production of secreted factors. Hence, FlhA appears to be an essential virulence factor with a pleiotropic role.


Subject(s)
Bacillus thuringiensis/genetics , Bacillus thuringiensis/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Insecta/microbiology , Membrane Proteins/physiology , Trans-Activators/genetics , Transcription, Genetic , Animals , Bacillus cereus/genetics , Bacillus cereus/pathogenicity , Bacillus thuringiensis/growth & development , Bacterial Proteins/biosynthesis , Cell Survival , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Escherichia coli/genetics , HeLa Cells , Humans , Kinetics , Trans-Activators/biosynthesis , Virulence , beta-Galactosidase/metabolism , beta-Glucosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...