Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
1.
Microb Biotechnol ; 17(6): e14478, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38850267

ABSTRACT

Clostridioides difficile (CD) infections are defined by toxins A (TcdA) and B (TcdB) along with the binary toxin (CDT). The emergence of the 'hypervirulent' (Hv) strain PR 027, along with PR 176 and 181, two decades ago, reshaped CD infection epidemiology in Europe. This study assessed MALDI-TOF mass spectrometry (MALDI-TOF MS) combined with machine learning (ML) and Deep Learning (DL) to identify toxigenic strains (producing TcdA, TcdB with or without CDT) and Hv strains. In total, 201 CD strains were analysed, comprising 151 toxigenic (24 ToxA+B+CDT+, 22 ToxA+B+CDT+ Hv+ and 105 ToxA+B+CDT-) and 50 non-toxigenic (ToxA-B-) strains. The DL-based classifier exhibited a 0.95 negative predictive value for excluding ToxA-B- strains, showcasing accuracy in identifying this strain category. Sensitivity in correctly identifying ToxA+B+CDT- strains ranged from 0.68 to 0.91. Additionally, all classifiers consistently demonstrated high specificity (>0.96) in detecting ToxA+B+CDT+ strains. The classifiers' performances for Hv strain detection were linked to high specificity (≥0.96). This study highlights MALDI-TOF MS enhanced by ML techniques as a rapid and cost-effective tool for identifying CD strain virulence factors. Our results brought a proof-of-concept concerning the ability of MALDI-TOF MS coupled with ML techniques to detect virulence factor and potentially improve the outbreak's management.


Subject(s)
Clostridioides difficile , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Virulence Factors , Clostridioides difficile/genetics , Clostridioides difficile/classification , Clostridioides difficile/chemistry , Clostridioides difficile/pathogenicity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Virulence Factors/genetics , Virulence Factors/analysis , Humans , Clostridium Infections/microbiology , Clostridium Infections/diagnosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Machine Learning , Deep Learning , Sensitivity and Specificity , Enterotoxins/analysis , Enterotoxins/genetics
2.
J Bacteriol ; 204(2): e0041121, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34807726

ABSTRACT

The Gram-positive bacterium Clostridioides difficile is a primary cause of hospital-acquired diarrhea, threatening both immunocompromised and healthy individuals. An important aspect of defining mechanisms that drive C. difficile persistence and virulence relies on developing a more complete understanding of sporulation. C. difficile sporulation is the single determinant of transmission and complicates treatment and prevention due to the chemical and physical resilience of spores. By extension, the identification of druggable targets that significantly attenuate sporulation would have a significant impact on thwarting C. difficile infection. By use of a new CRISPR-Cas9 nickase genome editing methodology, stop codons were inserted early in the coding sequence for clpP1 and clpP2 to generate C. difficile mutants that no longer produced the corresponding isoforms of caseinolytic protease P (ClpP). The data show that genetic ablation of ClpP isoforms leads to altered sporulation phenotypes with the clpP1/clpP2 double mutant exhibiting asporogenic behavior. A small screen of known ClpP inhibitors in a fluorescence-based biochemical assay identified bortezomib as an inhibitor of C. difficile ClpP that produces dose-dependent inhibition of purified ClpP. Incubation of C. difficile cultures in the presence of bortezomib reveals antisporulation effects approaching that observed in the clpP1/clpP2 double mutant. This work identifies ClpP as a key contributor to C. difficile sporulation and provides compelling support for the pursuit of small-molecule ClpP inhibitors as C. difficile antisporulating agents. IMPORTANCE Due to diverse roles of ClpP and the reliance of pathogens upon this system for infection, it has emerged as a target for antimicrobial development. Biology regulated by ClpP is organism dependent and has not been defined in Clostridioides difficile. This work identifies ClpP as a key contributor to C. difficile sporulation and provides compelling support for the pursuit of small-molecule ClpP inhibitors as antisporulating agents. The identification of new approaches and/or drug targets that reduce C. difficile sporulation would be transformative and are expected to find high utility in prophylaxis, transmission attenuation, and relapse prevention. Discovery of the ClpP system as a major driver to sporulation also provides a new avenue of inquiry for advancing the understanding of sporulation.


Subject(s)
Bacterial Proteins/genetics , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial , Spores, Bacterial/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bortezomib/pharmacology , Clostridioides difficile/chemistry , Clostridioides difficile/drug effects , Clostridium Infections/microbiology , Gene Editing/methods , Humans , Mutation , Phenotype , Protein Isoforms/genetics , Spores, Bacterial/metabolism , Virulence
3.
Nat Commun ; 12(1): 6285, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725358

ABSTRACT

Clostridium difficile causes life-threatening diarrhea and is the leading cause of healthcare-associated bacterial infections in the United States. TcdA and TcdB bacterial toxins are primary determinants of disease pathogenesis and are attractive therapeutic targets. TcdA and TcdB contain domains that use UDP-glucose to glucosylate and inactivate host Rho GTPases, resulting in cytoskeletal changes causing cell rounding and loss of intestinal integrity. Transition state analysis revealed glucocationic character for the TcdA and TcdB transition states. We identified transition state analogue inhibitors and characterized them by kinetic, thermodynamic and structural analysis. Iminosugars, isofagomine and noeuromycin mimic the transition state and inhibit both TcdA and TcdB by forming ternary complexes with Tcd and UDP, a product of the TcdA- and TcdB-catalyzed reactions. Both iminosugars prevent TcdA- and TcdB-induced cytotoxicity in cultured mammalian cells by preventing glucosylation of Rho GTPases. Iminosugar transition state analogues of the Tcd toxins show potential as therapeutics for C. difficile pathology.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Clostridioides difficile/drug effects , Clostridioides difficile/enzymology , Clostridium Infections/microbiology , Enterotoxins/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Enterotoxins/chemistry , Enterotoxins/metabolism , Humans , Kinetics
4.
Acta Crystallogr F Struct Biol Commun ; 77(Pt 8): 269-274, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34341193

ABSTRACT

In many prokaryotes, the first step of threonine metabolism is catalysed by the enzyme threonine dehydrogenase (TDH), which uses NAD+ to oxidize its substrate to 2-amino-3-ketobutyrate. The absence of a functional TDH gene in humans suggests that inhibitors of this enzyme may have therapeutic potential against pathogens which are reliant on this enzyme. Here, TDH from Clostridium difficile has been cloned and overexpressed, and the X-ray structure of the apoenzyme form has been determined at 2.6 Šresolution.


Subject(s)
Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Cross Infection , X-Ray Diffraction/methods , Amino Acid Sequence , Crystallography, X-Ray/methods , Humans , Protein Structure, Secondary , Protein Structure, Tertiary
5.
Anaerobe ; 70: 102379, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33940167

ABSTRACT

BACKGROUND: Sporulation is a complex cell differentiation programme shared by many members of the Firmicutes, the end result of which is a highly resistant, metabolically inert spore that can survive harsh environmental insults. Clostridioides difficile spores are essential for transmission of disease and are also required for recurrent infection. However, the molecular basis of sporulation is poorly understood, despite parallels with the well-studied Bacillus subtilis system. The spore envelope consists of multiple protective layers, one of which is a specialised layer of peptidoglycan, called the cortex, that is essential for the resistant properties of the spore. We set out to identify the enzymes required for synthesis of cortex peptidoglycan in C. difficile. METHODS: Bioinformatic analysis of the C. difficile genome to identify putative homologues of Bacillus subtilis spoVD was combined with directed mutagenesis and microscopy to identify and characterise cortex-specific PBP activity. RESULTS: Deletion of CDR20291_2544 (SpoVDCd) abrogated spore formation and this phenotype was completely restored by complementation in cis. Analysis of SpoVDCd revealed a three domain structure, consisting of dimerization, transpeptidase and PASTA domains, very similar to B. subtilis SpoVD. Complementation with SpoVDCd domain mutants demonstrated that the PASTA domain was dispensable for formation of morphologically normal spores. SpoVDCd was also seen to localise to the developing spore by super-resolution confocal microscopy. CONCLUSIONS: We have identified and characterised a cortex specific PBP in C. difficile. This is the first characterisation of a cortex-specific PBP in C. difficile and begins the process of unravelling cortex biogenesis in this important pathogen.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Penicillin-Binding Proteins/metabolism , Spores, Bacterial/metabolism , Bacterial Proteins/genetics , Cell Wall/chemistry , Cell Wall/genetics , Cell Wall/metabolism , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Hot Temperature , Penicillin-Binding Proteins/genetics , Spores, Bacterial/genetics , Spores, Bacterial/growth & development
6.
Mol Microbiol ; 115(4): 684-698, 2021 04.
Article in English | MEDLINE | ID: mdl-33140473

ABSTRACT

Autolysin is a lytic enzyme that hydrolyzes peptidoglycans of the bacterial cell wall, with a catalytic domain and cell wall-binding (CWB) domains, to be involved in different physiological functions that require bacterial cell wall remodeling. We identified a novel autolysin, Acd24020, from Clostridioides (Clostridium) difficile (C. difficile), with an endopeptidase catalytic domain belonging to the NlpC/P60 family and three bacterial Src-homology 3 domains as CWB domains. The catalytic domain of Acd24020 (Acd24020-CD) exhibited C. difficile-specific lytic activity equivalent to Acd24020, indicating that Acd24020-CD has full-function as a lytic enzyme by itself. To elucidate the specific peptidoglycan-recognition and catalytic reaction mechanisms of Acd24020-CD, biochemical characterization, X-ray structure determination, a modeling study of the enzyme/substrate complex, and mutagenesis analysis were performed. Acd24020-CD has an hourglass-shaped substrate-binding groove across the molecule, which is responsible for recognizing the direct 3-4 cross-linking structure unique to C. difficile peptidoglycan. Based on the X-ray structure and modeling study, we propose a dynamic Cys/His catalyzing mechanism, in which the catalytic Cys299 and His354 residues dynamically change their conformations to complement each step of the enzyme catalytic reaction.


Subject(s)
Clostridioides difficile/chemistry , Clostridioides difficile/physiology , N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/physiology , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/physiology , Catalytic Domain , Cell Wall/metabolism , Clostridioides difficile/enzymology , Crystallography, X-Ray , Hydrolysis , Models, Molecular , Mutagenesis , N-Acetylmuramoyl-L-alanine Amidase/isolation & purification , Peptidoglycan/metabolism , Protein Conformation , Protein Domains
7.
mSphere ; 5(6)2020 11 18.
Article in English | MEDLINE | ID: mdl-33208520

ABSTRACT

Clostridioides difficile is an obligately anaerobic, spore-forming, Gram-positive pathogenic bacterium that is considered the leading cause of nosocomial diarrhea worldwide. Recent studies have attempted to understand the biology of the outermost layer of C. difficile spores, the exosporium, which is believed to contribute to early interactions with the host. The fundamental role of the cysteine-rich proteins CdeC and CdeM has been described. However, the molecular details behind the mechanism of exosporium assembly are missing. The underlying mechanisms that govern exosporium assembly in C. difficile remain poorly studied, in part due to difficulties in obtaining pure soluble recombinant proteins of the C. difficile exosporium. In this work, we observed that CdeC was able to form organized inclusion bodies (IBs) in Escherichia coli filled with lamella-like structures separated by an interspace of 5 to 15 nm; however, CdeC expression in an E. coli strain with a more oxidative environment led to the loss of the lamella-like organization of CdeC IBs. Additionally, dithiothreitol (DTT) treatment of CdeC inclusion bodies released monomeric soluble forms of CdeC. Deletions in different portions of CdeC did not affect CdeC's ability to aggregate and form oligomers stable under denaturation conditions but affected CdeC's self-assembly properties. Overall, these observations have important implications in further studies elucidating the role of CdeC in the exosporium assembly of C. difficile spores.IMPORTANCE The endospore of Clostridioides difficile is the vehicle for transmission and persistence of the pathogen, and, specifically, the exosporium is the first contact between the host and the spore. The underlying mechanisms that govern exosporium assembly in C. difficile remain understudied, in part due to difficulties in obtaining pure soluble recombinant proteins of the C. difficile exosporium. Understanding the exosporium assembly's molecular bases may be essential to developing new therapies against C. difficile infection.


Subject(s)
Bacterial Proteins/metabolism , Clostridioides difficile/pathogenicity , Inclusion Bodies/metabolism , Spores, Bacterial/metabolism , Bacterial Proteins/genetics , Cell Wall/chemistry , Cell Wall/metabolism , Clostridioides difficile/chemistry , Clostridioides difficile/metabolism , Cysteine/chemistry , Cysteine/metabolism , Escherichia coli/metabolism , Spores, Bacterial/chemistry
8.
J Bacteriol ; 202(22)2020 10 22.
Article in English | MEDLINE | ID: mdl-32868401

ABSTRACT

Clostridioides difficile is an anaerobic Gram-positive bacterium that can produce the large clostridial toxins toxin A and toxin B, encoded within the pathogenicity locus (PaLoc). The PaLoc also encodes the sigma factor TcdR, which positively regulates toxin gene expression, and TcdC, which is a putative negative regulator of toxin expression. TcdC is proposed to be an anti-sigma factor; however, several studies failed to show an association between the tcdC genotype and toxin production. Consequently, the TcdC function is not yet fully understood. Previous studies have characterized TcdC as a membrane-associated protein with the ability to bind G-quadruplex structures. The binding to the DNA secondary structures is mediated through the oligonucleotide/oligosaccharide binding fold (OB-fold) domain present at the C terminus of the protein. This domain was previously also proposed to be responsible for the inhibitory effect on toxin gene expression, implicating a cytoplasmic localization of the OB-fold. In this study, we aimed to obtain topological information on the C terminus of TcdC and demonstrate that the C terminus of TcdC is located extracellularly. In addition, we show that the membrane association of TcdC is dependent on a membrane-proximal cysteine residue and that mutating this residue results in the release of TcdC from the bacterial cell. The extracellular location of TcdC is not compatible with the direct binding of the OB-fold domain to intracellular nucleic acid or protein targets and suggests a mechanism of action that is different from that of the characterized anti-sigma factors.IMPORTANCE The transcription of C. difficile toxins TcdA and TcdB is directed by the sigma factor TcdR. TcdC has been proposed to be an anti-sigma factor. The activity of TcdC has been mapped to its C terminus, and the N terminus serves as the membrane anchor. Acting as an anti-sigma factor requires a cytoplasmic localization of the C terminus of TcdC. Using cysteine accessibility analysis and a HiBiT-based system, we show that the TcdC C terminus is located extracellularly, which is incompatible with its role as anti-sigma factor. Furthermore, mutating a cysteine residue at position 51 resulted in the release of TcdC from the bacteria. The codon-optimized version of the HiBiT (HiBiTopt) extracellular detection system is a valuable tool for topology determination of membrane proteins, increasing the range of systems available to tackle important aspects of C. difficile development.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Clostridioides difficile/chemistry , Enterotoxins/chemistry , Repressor Proteins/chemistry , Bacterial Proteins/genetics , Cell Membrane/chemistry , Clostridioides difficile/genetics , Gene Expression Regulation, Bacterial , Repressor Proteins/genetics , Sigma Factor/genetics
9.
Lett Appl Microbiol ; 71(6): 596-604, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32920822

ABSTRACT

Clostridium difficile can cause antibiotic-associated diarrhoea or pseudo-membranous colitis in humans and animals. Currently, the various methods such as microbiological culture, cytotoxic assay, ELISA and polymerase chain reaction have been used to detect Clostridium difficile infection (CDI). These conventional methods, however, require long detection time and professional staff. The paper is to describe a simple strategy which employs immunomagnetic separation and aptamer-mediated colorimetric assay for the detection of toxin B of C. difficile (TcdB) in the stool samples. HRP-labelled aptamer against TcdB selected by SELEX was firstly captured on the surface of magnetic beads (MB) by DNA hybridization with a complementary strand. In the presence of TcdB, aptamer specifically recognized and bound TcdB, disturbing the DNA hybridization and causing the release of HRP-aptamer from MB. This reduced the catalytic capacity of HRP and consequently the absorption intensity. As there was a relationship between the decrease in the absorption intensity and target concentration, a quantitative analysis of TcdB can be accomplished by the measurement of the absorption intensity. Under the optimal conditions, the assay system is able to detect TcdB at a concentration down to 5 ng ml-1 . Moreover the method had specificity of 97% and sensitivity of 66% and the system remained excellent stability within 4 weeks. The proposed method is a valuable screening procedure for CDI and can be extended readily to detection of other clinically important pathogens.


Subject(s)
Bacterial Proteins/analysis , Bacterial Toxins/analysis , Clostridioides difficile/metabolism , Colorimetry/methods , Immunomagnetic Separation/methods , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Biological Assay , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Clostridium Infections/microbiology , Diarrhea/microbiology , Feces/microbiology , Humans , Immunomagnetic Separation/instrumentation , Polymerase Chain Reaction
10.
Int J Mol Sci ; 21(17)2020 Aug 26.
Article in English | MEDLINE | ID: mdl-32858965

ABSTRACT

SecA protein is a major component of the general bacterial secretory system. It is an ATPase that couples nucleotide hydrolysis to protein translocation. In some Gram-positive pathogens, a second paralogue, SecA2, exports a different set of substrates, usually virulence factors. To identify SecA2 features different from SecA(1)s, we determined the crystal structure of SecA2 from Clostridioides difficile, an important nosocomial pathogen, in apo and ATP-γ-S-bound form. The structure reveals a closed monomer lacking the C-terminal tail (CTT) with an otherwise similar multidomain organization to its SecA(1) homologues and conserved binding of ATP-γ-S. The average in vitro ATPase activity rate of C. difficile SecA2 was 2.6 ± 0.1 µmolPi/min/µmol. Template-based modeling combined with evolutionary conservation analysis supports a model where C. difficile SecA2 in open conformation binds the target protein, ensures its movement through the SecY channel, and enables dimerization through PPXD/HWD cross-interaction of monomers during the process. Both approaches exposed regions with differences between SecA(1) and SecA2 homologues, which are in agreement with the unique adaptation of SecA2 proteins for a specific type of substrate, a role that can be addressed in further studies.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Clostridioides difficile/enzymology , Adenosine Triphosphatases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Conserved Sequence , Crystallography, X-Ray , Evolution, Molecular , Models, Molecular , Protein Conformation
11.
Braz J Microbiol ; 51(3): 1139-1143, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32367261

ABSTRACT

Clostridioides (Clostridium) difficile is responsible for most cases of nosocomial diarrhea and, despite the high prevalence of the disease worldwide, the best laboratory diagnostic approach to diagnose C. difficile infection (CDI) is a subject of ongoing debate. Although the use of multiple tests is recommended, the cost of these algorithms commonly exceeds the affordability in some countries. Thus, to improve CDI diagnosis in a university hospital in Brazil, this study analyzed two immunochromatographic tests and one enzyme immunoassay (ELISA) to evaluate the detection of glutamate dehydrogenase (GDH) and A/B toxins of C. difficile. Stool samples of 89 adult patients presenting nosocomial diarrhea during hospitalization were included. The toxigenic culture was used as the reference method. GDH detection by both commercial tests showed high sensitivity (100%) and specificity (92.1%). On the other hand, toxin-based methods showed a sensitivity between 19.2 and 57.7%. In conclusion, the results suggest that rapid tests for GDH detection are not only suitable for CDI diagnosis as screening tests but also as a single method.


Subject(s)
Bacterial Proteins/analysis , Bacterial Toxins/analysis , Clostridioides difficile/enzymology , Clostridium Infections/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Glutamate Dehydrogenase/analysis , Immunoassay/methods , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Brazil , Clostridioides , Clostridioides difficile/chemistry , Clostridioides difficile/metabolism , Clostridium Infections/microbiology , Diagnostic Tests, Routine/methods , Glutamate Dehydrogenase/metabolism , Hospitals, University , Humans
12.
Biochem J ; 477(8): 1459-1478, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32242623

ABSTRACT

Clostridioides difficile is a spore-forming bacterial pathogen that is the leading cause of hospital-acquired gastroenteritis. C. difficile infections begin when its spore form germinates in the gut upon sensing bile acids. These germinants induce a proteolytic signaling cascade controlled by three members of the subtilisin-like serine protease family, CspA, CspB, and CspC. Notably, even though CspC and CspA are both pseudoproteases, they are nevertheless required to sense germinants and activate the protease, CspB. Thus, CspC and CspA are part of a growing list of pseudoenzymes that play important roles in regulating cellular processes. However, despite their importance, the structural properties of pseudoenzymes that allow them to function as regulators remain poorly understood. Our recently solved crystal structure of CspC revealed that its pseudoactive site residues align closely with the catalytic triad of CspB, suggesting that it might be possible to 'resurrect' the ancestral protease activity of the CspC and CspA pseudoproteases. Here, we demonstrate that restoring the catalytic triad to these pseudoproteases fails to resurrect their protease activity. We further show that the pseudoactive site substitutions differentially affect the stability and function of the CspC and CspA pseudoproteases: the substitutions destabilized CspC and impaired spore germination without affecting CspA stability or function. Thus, our results surprisingly reveal that the presence of a catalytic triad does not necessarily predict protease activity. Since homologs of C. difficile CspA occasionally carry an intact catalytic triad, our results indicate that bioinformatic predictions of enzyme activity may underestimate pseudoenzymes in rare cases.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Clostridioides difficile/enzymology , Spores, Bacterial/growth & development , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Catalysis , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Clostridioides difficile/growth & development , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Developmental , Spores, Bacterial/enzymology , Spores, Bacterial/genetics
13.
ACS Chem Biol ; 15(4): 1050-1058, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32191024

ABSTRACT

Clostridiodes (Clostridium) difficile is an anaerobic Gram-positive, spore-forming nosocomial, gastrointestinal pathogen causing C. difficile-associated disease with symptoms ranging from mild cases of antibiotic-associated diarrhea to fatal pseudomembranous colitis. We developed murine monoclonal antibodies (mAbs) specific for a conserved cell surface antigen, lipoteichoic acid (LTA)of C. difficile. The mAbs were characterized in terms of their thermal stability, solubility, and their binding to LTA by surface plasmon resonance and competitive ELISA. Synthetic LTA molecules were prepared in order to better define the minimum epitope required to mimic the natural antigen, and three repeat units of the polymer were required for optimal recognition. One of the murine mAbs was chimerized with human constant region domains and was found to recognize the target antigen identically to the mouse version. These mAbs may be useful as therapeutics (standalone, in conjunction with known antitoxin approaches, or as delivery vehicles for antibody drug conjugates targeting the bacterium), as diagnostic agents, and in infection control applications.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Clostridioides difficile/immunology , Lipopolysaccharides/immunology , Teichoic Acids/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Clostridioides difficile/chemistry , Humans , Mice , Protein Stability
14.
J Clin Microbiol ; 58(2)2020 01 28.
Article in English | MEDLINE | ID: mdl-31776192

ABSTRACT

The Singulex Clarity C. diff toxins A/B (Clarity) assay is an automated, ultrasensitive immunoassay for the detection of Clostridioides difficile toxins in stool. In this study, the performance of the Clarity assay was compared to that of a multistep algorithm using an enzyme immunoassay (EIA) for detection of glutamate dehydrogenase (GDH) and toxins A and B arbitrated by a semiquantitative cell cytotoxicity neutralization assay (CCNA). The performance of the assay was evaluated using 211 residual deidentified stool samples tested with a GDH-and-toxin EIA (C. Diff Quik Chek Complete; Techlab), with GDH-and-toxin discordant samples tested with CCNA. The stool samples were stored at -80°C before being tested with the Clarity assay. For samples discordant between Clarity and the standard-of-care algorithm, the samples were tested with PCR (Xpert C. difficile; Cepheid), and chart review was performed. The testing algorithm resulted in 34 GDH+/toxin+, 53 GDH-/toxin-, and 124 GDH+/toxin- samples, of which 39 were CCNA+ and 85 were CCNA- Clarity had 96.2% negative agreement with GDH-/toxin- samples, 100% positive agreement with GDH+/toxin+ samples, and 95.3% agreement with GDH+/toxin-/CCNA- samples. The Clarity result was invalid for one sample. Clarity agreed with 61.5% of GDH+/toxin-/CCNA+ samples, 90.0% of GDH+/toxin-/CCNA+ (high-positive) samples, and 31.6% of GDH+/toxin-/CCNA+ (low-positive) samples. The Singulex Clarity C. diff toxins A/B assay demonstrated high agreement with a testing algorithm utilizing a GDH-and-toxin EIA and CCNA. This novel automated assay may offer an accurate, stand-alone solution for C. difficile infection (CDI) diagnostics, and further prospective clinical studies are merited.


Subject(s)
Bacterial Proteins/analysis , Bacterial Toxins/analysis , Clostridioides difficile/chemistry , Clostridioides difficile/enzymology , Enterotoxins/analysis , Glutamate Dehydrogenase/analysis , Immunoenzyme Techniques/standards , Adult , Algorithms , Automation, Laboratory , Clostridium Infections/diagnosis , Feces/chemistry , Female , Humans , Male , Sensitivity and Specificity
15.
J Clin Microbiol ; 57(11)2019 11.
Article in English | MEDLINE | ID: mdl-31434726

ABSTRACT

Clostridioides difficile infection (CDI) is one of the most common health care-associated infections, resulting in significant morbidity, mortality, and economic burden. Diagnosis of CDI relies on the assessment of clinical presentation and laboratory tests. We evaluated the clinical performance of ultrasensitive single-molecule counting technology for detection of C. difficile toxins A and B. Stool specimens from 298 patients with suspected CDI were tested with the nucleic acid amplification test (NAAT; BD MAX Cdiff assay or Xpert C. difficile assay) and Singulex Clarity C. diff toxins A/B assay. Specimens with discordant results were tested with the cell cytotoxicity neutralization assay (CCNA), and the results were correlated with disease severity and outcome. There were 64 NAAT-positive and 234 NAAT-negative samples. Of the 32 NAAT+/Clarity- and 4 NAAT-/Clarity+ samples, there were 26 CCNA- and 4 CCNA- samples, respectively. CDI relapse was more common in NAAT+/toxin+ patients than in NAAT+/toxin- and NAAT-/toxin- patients. The clinical specificity of Clarity and NAAT was 97.4% and 89.0%, respectively, and overdiagnosis was more than three times more common in NAAT+/toxin- than in NAAT+/toxin+ patients. The Clarity assay was superior to NAATs for the diagnosis of CDI, by reducing overdiagnosis and thereby increasing clinical specificity, and the presence of toxins was associated with negative patient outcomes.


Subject(s)
Clostridium Infections/diagnosis , Enterotoxins/isolation & purification , Immunoassay/methods , Single Molecule Imaging/methods , Adult , Aged , Bacteriological Techniques/methods , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Clostridium Infections/microbiology , Feces/chemistry , Feces/microbiology , Female , Humans , Male , Medical Overuse , Middle Aged , Nucleic Acid Amplification Techniques , Sensitivity and Specificity
16.
J Med Chem ; 62(17): 7739-7750, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31429553

ABSTRACT

Frizzled 7 (FZD7) receptors have been shown to play a central role in intestinal stem cell regeneration and, more recently, in Clostridium difficile pathogenesis. Yet, targeting FZD7 receptors with small ligands has not been explored as an approach to block C. difficile pathogenesis. Here, we report the discovery of high affinity peptides that selectively bind to FZD7 receptors. We describe an integrated approach for lead optimization, utilizing structure-based rational design and directed evolution, to enhance the peptide binding affinity while still maintaining FZD7 receptor selectivity. This work yielded new peptide leads with picomolar binding constants to FZD7 as measured by biophysical methods. The new peptides block the interaction between C. difficile toxin B (TcdB) and FZD receptors and perturb C. difficile pathogenesis in epithelial cells. As such, our findings provide a proof of concept that targeting FZD receptors could be a viable pharmacological approach to protect epithelial cells from TcdB pathogenicity.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Clostridioides difficile/chemistry , Epithelial Cells/drug effects , Frizzled Receptors/antagonists & inhibitors , Peptides/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Epithelial Cells/metabolism , Frizzled Receptors/chemistry , Frizzled Receptors/metabolism , Humans , Models, Molecular , Molecular Structure , Peptides/chemistry , Structure-Activity Relationship
17.
J Med Microbiol ; 68(10): 1445-1454, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31429817

ABSTRACT

Purpose. Clostridium difficile spores are extremely resilient to high temperatures. Sublethal temperatures are associated with the 'reactivation' of dormant spores, and are utilized to maximize C. difficile spore recovery. Spore eradication is of vital importance to the food industry. The current study seeks to elucidate the transient and persisting effects of heating C. difficile spores at various temperatures.Methods. Spores of five C. difficile strains of different ribotypes (001, 015, 020, 027 and 078) were heated at 50, 60 and 70-80 °C for 60 min in phosphate-buffered saline (PBS) and enumerated at 0, 15, 30, 45 and 60 min. GInaFiT was used to model the kinetics of spore inactivation. In subsequent experiments, spores were transferred to enriched brain heart infusion (BHI) broths after 10 min of 80 °C heat treatment in PBS; samples were enumerated at 90 min and 24 h.Results. The spores of all strains demonstrated log-linear inactivation with tailing when heated for 60 min at 80 °C [(x̄=7.54±0.04 log10 vs 4.72±0.09 log10 colony-forming units (c.f.u.) ml- 1; P<0.001]. At 70 °C, all strains except 078 exhibited substantial decline in recovery over 60 min. Interestingly, 50 °C heat treatment had an inhibitory effect on 078 spore recovery at 0 vs 60 min (7.61±0.06 log10 c.f.u. ml- 1 vs 6.13±0.05 log10 c.f.u. ml- 1; P<0.001). Heating at 70/80 °C inhibited the initial germination and outgrowth of both newly produced and aged spores in enriched broths. This inhibition appeared to be transient; after 24 h vegetative counts were higher in heat-treated vs non-heat-treated spores (x̄=7.65±0.04 log10 c.f.u. ml- 1 vs 6.79±0.06 log10 c.f.u. ml- 1; P<0.001).Conclusions. The 078 spores were more resistant to the inhibitory effects of higher temperatures. Heat initially inhibits spore germination, but the subsequent outgrowth of vegetative populations accelerates after the initial inhibitory period.


Subject(s)
Clostridioides difficile/growth & development , Spores, Bacterial/chemistry , Clostridioides difficile/chemistry , Clostridioides difficile/classification , Clostridioides difficile/physiology , Hot Temperature , Humans , Kinetics , Microbial Viability , Ribotyping , Spores, Bacterial/growth & development , Spores, Bacterial/physiology
18.
Nat Struct Mol Biol ; 26(8): 712-719, 2019 08.
Article in English | MEDLINE | ID: mdl-31308519

ABSTRACT

Clostridium difficile is an opportunistic pathogen that establishes in the colon when the gut microbiota are disrupted by antibiotics or disease. C. difficile infection (CDI) is largely caused by two virulence factors, TcdA and TcdB. Here, we report a 3.87-Å-resolution crystal structure of TcdB holotoxin that captures a unique conformation of TcdB at endosomal pH. Complementary biophysical studies suggest that the C-terminal combined repetitive oligopeptides (CROPs) domain of TcdB is dynamic and can sample open and closed conformations that may facilitate modulation of TcdB activity in response to environmental and cellular cues during intoxication. Furthermore, we report three crystal structures of TcdB-antibody complexes that reveal how antibodies could specifically inhibit the activities of individual TcdB domains. Our studies provide novel insight into the structure and function of TcdB holotoxin and identify intrinsic vulnerabilities that could be exploited to develop new therapeutics and vaccines for the treatment of CDI.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Clostridioides difficile/chemistry , Amino Acid Sequence , Antibodies, Neutralizing/immunology , Antigen-Antibody Complex/chemistry , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Conserved Sequence , Crystallography, X-Ray , Endosomes/chemistry , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Liposomes , Membrane Potentials , Models, Molecular , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid
19.
Nat Microbiol ; 4(10): 1760-1769, 2019 10.
Article in English | MEDLINE | ID: mdl-31160825

ABSTRACT

Clostridium difficile toxin A (TcdA) is a major exotoxin contributing to disruption of the colonic epithelium during C. difficile infection. TcdA contains a carbohydrate-binding combined repetitive oligopeptides (CROPs) domain that mediates its attachment to cell surfaces, but recent data suggest the existence of CROPs-independent receptors. Here, we carried out genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9)-mediated screens using a truncated TcdA lacking the CROPs, and identified sulfated glycosaminoglycans (sGAGs) and low-density lipoprotein receptor (LDLR) as host factors contributing to binding and entry of TcdA. TcdA recognizes the sulfation group in sGAGs. Blocking sulfation and glycosaminoglycan synthesis reduces TcdA binding and entry into cells. Binding of TcdA to the colonic epithelium can be reduced by surfen, a small molecule that masks sGAGs, by GM-1111, a sulfated heparan sulfate analogue, and by sulfated cyclodextrin, a sulfated small molecule. Cells lacking LDLR also show reduced sensitivity to TcdA, although binding between LDLR and TcdA are not detected, suggesting that LDLR may facilitate endocytosis of TcdA. Finally, GM-1111 reduces TcdA-induced fluid accumulation and tissue damage in the colon in a mouse model in which TcdA is injected into the caecum. These data demonstrate in vivo and pathological relevance of TcdA-sGAGs interactions, and reveal a potential therapeutic approach of protecting colonic tissues by blocking these interactions.


Subject(s)
Bacterial Toxins/metabolism , Clostridioides difficile/chemistry , Enterotoxins/metabolism , Glycosaminoglycans/metabolism , Receptors, LDL/metabolism , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Cell Membrane/metabolism , Colon/drug effects , Colon/metabolism , Endocytosis , Enterotoxins/chemistry , Enterotoxins/genetics , Enterotoxins/toxicity , Glycosaminoglycans/deficiency , HeLa Cells , Heparitin Sulfate/analogs & derivatives , Heparitin Sulfate/pharmacology , Humans , Intestinal Mucosa/metabolism , Mice , Mutation , Oligopeptides/genetics , Protein Binding , Receptors, LDL/deficiency
20.
ACS Chem Biol ; 14(7): 1528-1535, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31184849

ABSTRACT

Clostridioides difficile infection (CDI) is a leading cause of significant morbidity, mortality, and healthcare-related costs in the United States. After standard therapy, recurrence rates remain high, and multiple recurrences are not uncommon. Causes include treatments employing broad-spectrum agents that disrupt the normal host microbiota, as well as treatment-resistant spore formation by C. difficile. Thus, novel druggable anti-C. difficile targets that promote narrow-spectrum eradication and inhibition of sporulation are desired. As a critical rate-limiting step within the FAS-II bacterial fatty acid synthesis pathway, which supplies precursory component phospholipids found in bacterial cytoplasmic and spore-mediated membranes, enoyl-acyl carrier protein (ACP) reductase II (FabK) represents such a target. FabK is essential in C. difficile (CdFabK) and is structurally and mechanistically distinct from other isozymes found in gut microbiota species, making CdFabK an attractive narrow-spectrum target. We report here the kinetic evaluation of CdFabK, the biochemical activity of a series of phenylimidazole analogues, and microbiological data suggesting these compounds' selective antibacterial activity against C. difficile versus several other prominent gut organisms. The compounds display promising, selective, low micromolar CdFabK inhibitory activity without significantly affecting the growth of other gut organisms, and the series prototype (1b) is shown to be competitive for the CdFabK cofactor and uncompetitive for the substrate. A series analogue (1g) shows maintained inhibitory activity while also possessing increased solubility. These findings represent the basis for future drug discovery efforts by characterizing the CdFabK enzyme while demonstrating its druggability and potential role as a narrow-spectrum antidifficile target.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/enzymology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Clostridioides difficile/chemistry , Clostridioides difficile/metabolism , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...