Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.540
Filter
1.
PLoS One ; 19(8): e0307512, 2024.
Article in English | MEDLINE | ID: mdl-39093838

ABSTRACT

The multifunctional autoprocessing repeat-in-toxin (MARTX) toxin is the primary virulence factor of Vibrio vulnificus displaying cytotoxic and hemolytic properties. The cysteine protease domain (CPD) is responsible for activating the MARTX toxin by cleaving the toxin precursor and releasing the mature toxin fragments. To investigate the structural determinants for inositol hexakisphosphate (InsP6)-mediated activation of the CPD, we determined the crystal structures of unprocessed and ß-flap truncated MARTX CPDs of Vibrio vulnificus strain MO6-24/O in complex with InsP6 at 1.3 and 2.2Å resolution, respectively. The CPD displays a conserved domain with a central seven-stranded ß-sheet flanked by three α-helices. The scissile bond Leu3587-Ala3588 is bound in the catalytic site of the InsP6-loaded form of the Cys3727Ala mutant. InsP6 interacts with the conserved basic cleft and the ß-flap inducing the active conformation of catalytic residues. The ß-flap of the post-CPD is flexible in the InsP6-unbound state. The structure of the CPD Δß-flap showed an inactive conformation of the catalytic residues due to the absence of interaction between the active site and the ß-flap. This study confirms the InsP6-mediated activation of the MARTX CPDs in which InsP6-binding induces conformational changes of the catalytic residues and the ß-flap that holds the N terminus of the CPD in the active site, facilitating hydrolysis of the scissile bond.


Subject(s)
Phytic Acid , Vibrio vulnificus , Vibrio vulnificus/enzymology , Vibrio vulnificus/genetics , Vibrio vulnificus/metabolism , Phytic Acid/metabolism , Catalytic Domain , Cysteine Proteases/metabolism , Cysteine Proteases/chemistry , Cysteine Proteases/genetics , Crystallography, X-Ray , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Protein Domains , Models, Molecular , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Amino Acid Sequence
2.
Arch Microbiol ; 206(9): 381, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39153128

ABSTRACT

The bacterial type II toxin-antitoxin (TA) system is a rich genetic element that participates in various physiological processes. Aeromonas veronii is the main bacterial pathogen threatening the freshwater aquaculture industry. However, the distribution of type II TA system in A. veronii was seldom documented and its roles in the life activities of A. veronii were still unexplored. In this study, a novel type II TA system AvtA-AvtT was predicted in a fish pathogen Aeromonas veronii biovar sobria with multi-drug resistance using TADB 2.0. Through an Escherichia coli host killing and rescue assay, we demonstrated that AvtA and AvtT worked as a genuine TA system, and the predicted toxin AvtT actually functioned as an antitoxin while the predicted antitoxin AvtA actually functioned as a toxin. The binding ability of AvtA with AvtT proteins were confirmed by dot blotting analysis and co-immunoprecipitation assay. Furthermore, we found that the toxin and antitoxin labelled with fluorescent proteins were co-localized. In addition, it was found that the transcription of AvtAT bicistronic operon was repressed by the AvtAT protein complex. Deletion of avtA gene and avtT gene had no obvious effect on the drug susceptibility. This study provides first characterization of type II TA system AvtA-AvtT in aquatic pathogen A. veronii.


Subject(s)
Aeromonas veronii , Bacterial Proteins , Toxin-Antitoxin Systems , Aeromonas veronii/genetics , Aeromonas veronii/metabolism , Toxin-Antitoxin Systems/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Operon , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/drug effects , Antitoxins/genetics , Antitoxins/metabolism , Gene Expression Regulation, Bacterial
3.
Gut Microbes ; 16(1): 2390133, 2024.
Article in English | MEDLINE | ID: mdl-39132815

ABSTRACT

Vancomycin (VAN) treatment in Clostridioides difficile infection (CDI) suffers from a relatively high rate of recurrence, with a variety of reasons behind this, including biofilm-induced recurrent infections. C. difficile can form monophyletic or symbiotic biofilms with other microbes in the gut, and these biofilms protect C. difficile from being killed by antibiotics. In this study, we analyzed the ecological relationship between Bacteroides thetaiotaomicron and C. difficile and their formation of symbiotic biofilm in the VAN environment. The production of symbiotic biofilm formed by C. difficile and B. thetaiotaomicron was higher than that of C. difficile and B. thetaiotaomicron alone in the VAN environment. In symbiotic biofilms, C. difficile was characterized by increased production of the toxin protein TcdA and TcdB, up-regulation of the expression levels of the virulence genes tcdA and tcdB, enhanced bacterial cell swimming motility and c-di-GMP content, and increased adhesion to Caco-2 cells. The scanning electron microscope (SEM) combined with confocal laser scanning microscopy (CLSM) results indicated that the symbiotic biofilm was elevated in thickness, dense, and had an increased amount of mixed bacteria, while the fluorescence in situ hybridization (FISH) probe and plate colony counting results further indicated that the symbiotic biofilm had a significant increase in the amount of C. difficile cells, and was able to better tolerate the killing of the simulated intestinal fluid. Taken together, C. difficile and B. thetaiotaomicron become collaborative in the VAN environment, and targeted deletion or attenuation of host gut B. thetaiotaomicron content may improve the actual efficacy of VAN in CDI treatment.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Bacteroides thetaiotaomicron , Biofilms , Clostridioides difficile , Symbiosis , Vancomycin , Biofilms/drug effects , Biofilms/growth & development , Clostridioides difficile/drug effects , Clostridioides difficile/physiology , Clostridioides difficile/genetics , Humans , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Caco-2 Cells , Bacteroides thetaiotaomicron/drug effects , Bacteroides thetaiotaomicron/metabolism , Bacteroides thetaiotaomicron/physiology , Bacteroides thetaiotaomicron/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Enterotoxins/metabolism , Enterotoxins/genetics , Bacterial Adhesion/drug effects
4.
Proc Natl Acad Sci U S A ; 121(33): e2406775121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39116134

ABSTRACT

Biofilm-protected pathogenic Staphylococcus aureus causes chronic infections that are difficult to treat. An essential building block of these biofilms are functional amyloid fibrils that assemble from phenol-soluble modulins (PSMs). PSMα1 cross-seeds other PSMs into cross-ß amyloid folds and is therefore a key element in initiating biofilm formation. However, the paucity of high-resolution structures hinders efforts to prevent amyloid assembly and biofilm formation. Here, we present a 3.5 Å resolution density map of the major PSMα1 fibril form revealing a left-handed cross-ß fibril composed of two C2-symmetric U-shaped protofilaments whose subunits are unusually tilted out-of-plane. Monomeric α-helical PSMα1 is extremely cytotoxic to cells, despite the moderate toxicity of the cross-ß fibril. We suggest mechanistic insights into the PSM functional amyloid formation and conformation transformation on the path from monomer-to-fibril formation. Details of PSMα1 assembly and fibril polymorphism suggest how S. aureus utilizes functional amyloids to form biofilms and establish a framework for developing therapeutics against infection and antimicrobial resistance.


Subject(s)
Amyloid , Biofilms , Staphylococcus aureus , Staphylococcus aureus/metabolism , Staphylococcus aureus/physiology , Biofilms/growth & development , Amyloid/metabolism , Amyloid/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/chemistry , Protein Conformation , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Models, Molecular
5.
ACS Appl Mater Interfaces ; 16(29): 37390-37400, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39007843

ABSTRACT

Staphylococcus aureus (S. aureus) is an opportunistic infectious pathogen, which causes a high mortality rate during bloodstream infections. The early detection of virulent strains in patients' blood samples is of medical interest for rapid diagnosis. The main virulent factors identified in patient isolates include leukocidins that bind to specific membrane receptors and lyse immune cells and erythrocytes. Duffy antigen receptor for chemokines (DARC) on the surface of specific cells is a main target of leukocidins such as gamma-hemolysin AB (HlgAB) and leukocidin ED (LukED). Among them, HlgAB is a conserved and critical leukocidin that binds to DARC and forms pores on the cell membranes, leading to cell lysis. Current methods are based on ELISA or bacterial culture, which takes hours to days. For detecting HlgAB with faster response and higher sensitivity, we developed a biosensor that combines single-walled carbon nanotube field effect transistors (swCNT-FETs) with immobilized DARC receptors as biosensing elements. DARC was purified from a bacterial expression system and successfully reconstituted into nanodiscs that preserve binding capability for HlgAB. Dynamic light scattering (DLS) and scanning electron microscopy (SEM) showed an increase of the DARC-containing nanodisc size in the presence of HlgAB, indicating the formation of HlgAB prepore or pore complexes. We demonstrate that this sensor can specifically detect the leukocidins HlgA and HlgAB in a quantitative manner within the dynamic range of 1 fM to 100 pM with an LOD of 0.122 fM and an LOQ of 0.441 fM. The sensor was challenged with human serum spiked with HlgAB as simulated clinical samples. After dilution for decreasing nonspecific binding, it selectively detected the toxin with a similar detection range and apparent dissociation constant as in the buffer. This biosensor was demonstrated with remarkable sensitivity to detect HlgAB rapidly and has the potential as a tool for fundamental research and clinical applications, although this sensor cannot differentiate between HlgAB and LukED as both have the same receptor.


Subject(s)
Biosensing Techniques , Duffy Blood-Group System , Leukocidins , Staphylococcus aureus , Biosensing Techniques/methods , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/metabolism , Leukocidins/chemistry , Leukocidins/metabolism , Humans , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/chemistry , Nanotubes, Carbon/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism
6.
Vet Microbiol ; 295: 110168, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964035

ABSTRACT

Glaesserella parasuis is an important porcine pathogen that commonly colonizes the upper respiratory tract of pigs and is prone to causing Glässer's disease under complex conditions. As yet, the disease has led to serious economic losses to the swine industry worldwide. Studies so far have found that several virulence factors are associated with the pathogenicity of G. parasuis, but the pathogenic mechanism is still not fully understood. Cytolethal distending toxin (CDT), a potential virulence factor in G. parasuis, is involved in cytotoxicity, serum resistance, adherence to and invasion of host cells in vitro. Here, to further investigate the pathogenic role of CDT during G. parasuis infection in vitro and in vivo, a double cdt1 and cdt2 deletion mutant (Δcdt1Δcdt2) without selectable marker was first generated in G. parasuis JS0135 strain by continuous natural transformations and replica plating. Morphological observation and lactate dehydrogenase assay showed that the Δcdt1Δcdt2 mutant was defective in cytotoxicity. Additionally, the Δcdt1Δcdt2 mutant was more susceptible to phagocytosis caused by 3D4/2 macrophages compared to the wild-type JS0135 strain. Moreover, by focusing on clinical signs, necropsy, bacterial recovery and pathological observation, we found that the deletion of cdt1 and cdt2 genes led to a significant attenuation of virulence in G. parasuis. Taken together, these findings suggest that as an important virulence factor, CDT can significantly affect the pathogenicity of G. parasuis.


Subject(s)
Bacterial Toxins , Haemophilus parasuis , Phagocytosis , Swine Diseases , Animals , Swine , Haemophilus parasuis/pathogenicity , Haemophilus parasuis/genetics , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Bacterial Toxins/metabolism , Swine Diseases/microbiology , Virulence , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Haemophilus Infections/immunology , Virulence Factors/genetics , Macrophages/microbiology , Cell Line
7.
Cell Host Microbe ; 32(7): 1039-1041, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38991498

ABSTRACT

Bacteria have evolved anti-viral defenses, but the mechanisms of sensing and stopping infection are still under investigation. In this issue of Cell Host & Microbe, Mets, Kurata, Ernits et al. describe how direct sensing of a phage protein by a bacterial toxin-antitoxin-associated chaperone unleashes toxin activity to prevent infection.


Subject(s)
Bacteriophages , Molecular Chaperones , Molecular Chaperones/metabolism , Bacteriophages/physiology , Toxin-Antitoxin Systems , Bacterial Toxins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Bacteria/virology , Bacteria/metabolism , Bacteria/genetics
8.
Subcell Biochem ; 104: 245-267, 2024.
Article in English | MEDLINE | ID: mdl-38963490

ABSTRACT

Bacteria encode a wide range of survival and immunity systems, including CRISPR-Cas, restriction-modification systems, and toxin-antitoxin systems involved in defence against bacteriophages, as well as survival during challenging growth conditions or exposure to antibiotics. Toxin-antitoxin (TA) systems are small two- or three-gene cassettes consisting of a metabolic regulator (the "toxin") and its associated antidote (the "antitoxin"), which also often functions as a transcriptional regulator. TA systems are widespread in the genomes of pathogens but are also present in commensal bacterial species and on plasmids. For mobile elements such as plasmids, TA systems play a role in maintenance, and increasing evidence now points to roles of chromosomal toxin-antitoxin systems in anti-phage defence. Moreover, the widespread occurrence of toxin-antitoxin systems in the genomes of pathogens has been suggested to relate to survival during host infection as well as in persistence during antibiotic treatment. Upon repeated exposure to antibiotics, TA systems have been shown to acquire point mutations as well as more dramatic rearrangements such as in-frame deletions with potential relevance for bacterial survival and pathogenesis. In this review, we present an overview of the known functional and structural consequences of mutations and rearrangements arising in bacterial toxin-antitoxin systems and discuss their relevance for survival and persistence of pathogenic species.


Subject(s)
Bacteria , Toxin-Antitoxin Systems , Toxin-Antitoxin Systems/genetics , Bacteria/genetics , Bacteria/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
9.
Microbiol Res ; 286: 127812, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38954992

ABSTRACT

Clostridioides difficile infection (CDI) poses a significant health threat due to high recurrence rates. Antimicrobial agents are commonly used to manage CDI-related diarrhoea; however, by aggravating intestinal dysbiosis, antibiotics enable C. difficile spores germination and production of toxins, the main virulence factors. Therefore, the binding of exotoxins using adsorbents represents an attractive alternative medication for the prevention and treatment of relapses. In this study, we provided evidence that the natural insoluble polysaccharides, named ABR119, extracted by plant cell cultures, effectively trap C. difficile toxins. In our experiments, ABR119 exhibited no cytotoxicity in vitro and was safely administered in vivo. In the animal model of C. difficile-associated colitis, ABR119 (50 mg/kg body weight) significantly reduced the colonic myeloperoxidase activity and severity of inflammation, preventing body weight loss. These effects were not evident when we treated animals with wheat bran polysaccharides. We did not detect bacterial killing effects of ABR119 against C. difficile nor against bacterial species of the normal gut microbiota. Moreover, ABR119 did not interfere in vitro with the antimicrobial activities of most clinically used antibiotics. In summary, ABR119 holds promise for treating and preventing C. difficile colitis by trapping the bacterial toxins, warranting further studies to assess the ABR119 potential in human infections caused by C. difficile.


Subject(s)
Anti-Bacterial Agents , Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Colitis , Disease Models, Animal , Polysaccharides , Polysaccharides/pharmacology , Polysaccharides/metabolism , Clostridioides difficile/drug effects , Clostridioides difficile/pathogenicity , Animals , Colitis/microbiology , Colitis/drug therapy , Colitis/prevention & control , Colitis/chemically induced , Clostridium Infections/prevention & control , Clostridium Infections/microbiology , Clostridium Infections/drug therapy , Bacterial Toxins/metabolism , Anti-Bacterial Agents/pharmacology , Humans , Plant Cells , Mice , Colon/microbiology , Colon/drug effects , Gastrointestinal Microbiome/drug effects
10.
Cells ; 13(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38994956

ABSTRACT

Clostridioides difficile (C. difficile) is responsible for a spectrum of nosocomial/antibiotic-associated gastrointestinal diseases that are increasing in global incidence and mortality rates. The C. difficile pathogenesis is due to toxin A and B (TcdA/TcdB), both causing cytopathic and cytotoxic effects and inflammation. Recently, we demonstrated that TcdB induces cytopathic and cytotoxic (apoptosis and necrosis) effects in enteric glial cells (EGCs) in a dose/time-dependent manner and described the underlying signaling. Despite the role played by lipids in host processes activated by pathogens, to counter infection and/or induce cell death, to date no studies have investigated lipid changes induced by TcdB/TcdA. Here, we evaluated the modification of lipid composition in our in vitro model of TcdB infection. Apoptosis, cell cycle, cell viability, and lipidomic profiles were evaluated in EGCs treated for 24 h with two concentrations of TcdB (0.1 ng/mL; 10 ng/mL). In EGCs treated with the highest concentration of TcdB, not only an increased content of total lipids was observed, but also lipidome changes, allowing the separation of TcdB-treated cells and controls into different clusters. The statistical analyses also allowed us to ascertain which lipid classes and lipid molecular species determine the clusterization. Changes in lipid species containing inositol as polar head and plasmalogen phosphatidylethanolamine emerged as key indicators of altered lipid metabolism in TcdB-treated EGCs. These results not only provide a picture of the phospholipid profile changes but also give information regarding the lipid metabolism pathways altered by TcdB, and this might represent an important step for developing strategies against C. difficile infection.


Subject(s)
Bacterial Proteins , Bacterial Toxins , Neuroglia , Phospholipids , Neuroglia/metabolism , Neuroglia/drug effects , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Bacterial Toxins/pharmacology , Phospholipids/metabolism , Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Lipidomics , Humans
11.
Cells ; 13(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38994991

ABSTRACT

Clostridium perfringens (C. perfringens), a Gram-positive bacterium, produces a variety of toxins and extracellular enzymes that can lead to disease in both humans and animals. Common symptoms include abdominal swelling, diarrhea, and intestinal inflammation. Severe cases can result in complications like intestinal hemorrhage, edema, and even death. The primary toxins contributing to morbidity in C. perfringens-infected intestines are CPA, CPB, CPB2, CPE, and PFO. Amongst these, CPB, CPB2, and CPE are implicated in apoptosis development, while CPA is associated with cell death, increased intracellular ROS levels, and the release of the inflammatory factor IL-18. However, the exact mechanism by which PFO toxins exert their effects in the infected gut is still unidentified. This study demonstrates that a C. perfringens PFO toxin infection disrupts the intestinal epithelial barrier function through in vitro and in vivo models. This study emphasizes the notable influence of PFO toxins on intestinal barrier integrity in the context of C. perfringens infections. It reveals that PFO toxins increase ROS production by causing mitochondrial damage, triggering pyroptosis in IPEC-J2 cells, and consequently resulting in compromised intestinal barrier function. These results offer a scientific foundation for developing preventive and therapeutic approaches against C. perfringens infections.


Subject(s)
Bacterial Toxins , Clostridium perfringens , Epithelial Cells , Hemolysin Proteins , Intestinal Mucosa , Pyroptosis , Reactive Oxygen Species , Clostridium perfringens/pathogenicity , Bacterial Toxins/toxicity , Bacterial Toxins/metabolism , Pyroptosis/drug effects , Animals , Hemolysin Proteins/metabolism , Hemolysin Proteins/toxicity , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Reactive Oxygen Species/metabolism , Cell Line , Mice , Humans , Mitochondria/metabolism , Mitochondria/drug effects
12.
Harmful Algae ; 137: 102655, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39003021

ABSTRACT

Microseira wollei, a globally distributed freshwater bloom-forming benthic cyanobacterium, is known for its production of cyanotoxins and taste and odor (T&O). While CYN (Cylindrospermopsin)-producing populations of M. wollei are confined to Australia, PST (Paralytic shellfish toxins)-producing populations have been exclusively documented in North America. In this study, four benthic cyanobacterial strains, isolated from West Lake in China, were identified as M. wollei based on morphological and phylogenetic analyses. Detection of sxtA gene and UPLC-MS/MS analysis conclusively confirmed the PST-producing capability of M. wollei CHAB5998. In the phylogenetic tree of 16S rDNA, M. wollei strains formed a monophyletic group with two subclades. Notably, non-PST-producing Chinese strains clustered with Australian strains in Clade II, while all other strains, including PST-producing ones, clustered in Clade I. Additionally, CHAB5998 contains ten PST variants, of which STX, NEO, GTX2, GTX3, GTX5 and C1 were identified for the first time in M. wollei. Sequence analysis of PST biosynthetic gene cluster (sxt) genes indicated potential base variations, gene rearrangements, insertions, and deletions in the strain CHAB5998. Also, sxt gene has a longer evolutionary history in M. wollei than that in cyanobacteria from Nostocales. Multiple recombination breakpoints detected in sxt genes and the inconsistency in the topology of the phylogenetic trees between sxt and 16S rDNA suggested that multiple horizontal gene transfers (HGT) have occurred. Overall, the present study marks the first documented occurrence of PST-producing M. wollei outside of North America and identifies it as the first toxic freshwater benthic cyanobacterium in China. This revelation implies that benthic cyanobacteria may pose a higher environmental risk in China than previously acknowledged.


Subject(s)
Bacterial Toxins , Cyanobacteria , Phylogeny , Cyanobacteria/metabolism , Cyanobacteria/genetics , Cyanobacteria/classification , China , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Cyanobacteria Toxins , RNA, Ribosomal, 16S/genetics , Marine Toxins/metabolism
13.
Commun Biol ; 7(1): 839, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987278

ABSTRACT

Clostridioides difficile causes a wide range of intestinal diseases through the action of two main cytotoxins, TcdA and TcdB. Ingested spores germinate in the intestine establishing a population of cells that produce toxins and spores. The pathogenicity locus, PaLoc, comprises several genes, including those coding for TcdA/B, for the holin-like TcdE protein, and for TcdR, an auto-regulatory RNA polymerase sigma factor essential for tcdA/B and tcdE expression. Here we show that tcdR, tcdA, tcdB and tcdE are expressed in a fraction of the sporulating cells, in either the whole sporangium or in the forespore. The whole sporangium pattern is due to protracted expression initiated in vegetative cells by σD, which primes the TcdR auto-regulatory loop. In contrast, the forespore-specific regulatory proteins σG and SpoVT control TcdR production and tcdA/tcdB and tcdE expression in this cell. We detected TcdA at the spore surface, and we show that wild type and ΔtcdA or ΔtcdB spores but not ΔtcdR or ΔtcdA/ΔtcdB spores are cytopathic against HT29 and Vero cells, indicating that spores may serve as toxin-delivery vehicles. Since the addition of TcdA and TcdB enhance binding of spores to epithelial cells, this effect may occur independently of toxin production by vegetative cells.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Spores, Bacterial , Spores, Bacterial/metabolism , Spores, Bacterial/genetics , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Animals , Chlorocebus aethiops , Vero Cells , Enterotoxins/metabolism , Enterotoxins/genetics
14.
PLoS One ; 19(7): e0306998, 2024.
Article in English | MEDLINE | ID: mdl-38985791

ABSTRACT

Infectious and foodborne diseases pose significant global threats, with devastating consequences in low- and middle-income countries. Ozone, derived from atmospheric oxygen, exerts antimicrobial effects against various microorganisms, and degrades fungal toxins, which were initially recognized in the healthcare and food industries. However, highly concentrated ozone gas can be detrimental to human health. In addition, ozonated water is unstable and has a short half-life. Therefore, ultrafine-bubble technology is expected to overcome these issues. Ultrafine bubbles, which are nanoscale entitles that exist in water for considerable durations, have previously demonstrated bactericidal effects against various bacterial species, including antibiotic-resistant strains. This present study investigated the effects of ozone ultrafine bubble water (OUFBW) on various bacterial toxins. This study revealed that OUFBW treatment abolished the toxicity of pneumolysin, a pneumococcal pore-forming toxin, and leukotoxin, a toxin that causes leukocyte injury. Silver staining confirmed the degradation of pneumolysin, leukotoxin, and staphylococcal enterotoxin A, which are potent gastrointestinal toxins, following OUFB treatment. In addition, OUFBW treatment significantly inhibited NF-κB activation by Pam3CSK4, a synthetic triacylated lipopeptide that activates Toll-like receptor 2. Additionally, OUFBW exerted bactericidal activity against Staphylococcus aureus, including an antibiotic-resistant strain, without displaying significant toxicity toward human neutrophils or erythrocytes. These results suggest that OUFBW not only sterilizes bacteria but also degrades bacterial toxins.


Subject(s)
Bacterial Toxins , Ozone , Ozone/chemistry , Ozone/pharmacology , Humans , Bacterial Toxins/metabolism , Water/chemistry , NF-kappa B/metabolism , Bacterial Proteins/metabolism
15.
World J Microbiol Biotechnol ; 40(9): 265, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990361

ABSTRACT

The increasing prevalence of infections related to methicillin-resistant Staphylococcus aureus (MRSA) necessitates the exploration of innovative therapeutic strategies that diverge from conventional antibiotic treatments. This is imperative to effectively combat resistance and manage these infections. The adoption of antivirulence strategies has emerged as a particularly promising avenue. This approach applies a heightened selective pressure on pathogens, thereby diminishing the likelihood of bacteria evolving resistance to antibiotics. In our pursuit of novel therapeutics for treating MRSA infections, we have focused on agents that inhibit the virulence of S. aureus without impeding its growth, aiming to minimize the development of drug resistance. α-Hemolysin, a critical virulence factor encoded by the hla gene, is a cytotoxin that forms pores in host cell membranes and plays a pivotal role in the progression of disease during bacterial infections. Herein, we identified that norwogonin could effectively inhibit Hla production via targeting agrAC, a crucial protein in quorum sensing, resulting in dose-dependent inhibition of hemolytic activity without suppressing S. aureus growth. In vitro assays illustrated that norwogonin decreased the thermal stability of agrAC, providing evidence of interaction between norwogonin and agrAC. Meanwhile, norwogonin alleviated Hla-mediated A549 cell damage and reduced lactate dehydrogenase release. In vivo studies suggested that norwogonin treatment blocked the establishment of a mouse model of pneumonia caused by S. aureus USA300. Notably, norwogonin enhanced the antibacterial potency of oxacillin. In conclusion, norwogonin is a promising candidate for treating S. aureus infections, offering a novel alternative to traditional antibiotics by targeting virulence factors and enhancing the efficacy of existing treatments.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Hemolysin Proteins , Methicillin-Resistant Staphylococcus aureus , Virulence Factors , Animals , Female , Humans , Mice , A549 Cells , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Toxins/metabolism , Disease Models, Animal , Hemolysin Proteins/metabolism , Hemolysis/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice, Inbred BALB C , Quorum Sensing/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Virulence/drug effects , Virulence Factors/metabolism
16.
Infect Immun ; 92(8): e0030024, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39057915

ABSTRACT

The cGAS/STING sensor system drives innate immune responses to intracellular microbial double-stranded DNA (dsDNA) and bacterial cyclic nucleotide second messengers (e.g., c-di-AMP). STING-dependent cell-intrinsic responses can increase resistance to microbial infection and speed pathogen clearance. Correspondingly, STING activation and signaling are known to be targeted for suppression by effectors from several bacterial pathogens. Whether STING responses are also positively regulated through sensing of specific bacterial effectors is less clear. We find that STING activation through dsDNA, by its canonical ligand 2'-3' cGAMP, or the small molecule DMXAA is potentiated following intracellular delivery of the AB5 toxin family member pertussis toxin from Bordetella pertussis or the B subunit of cholera toxin from Vibrio cholerae. Entry of pertussis toxin or cholera toxin B into mouse macrophages triggers markers of endoplasmic reticulum (ER) stress and enhances ligand-dependent STING responses at the level of STING receptor activation in a manner that is independent of toxin enzymatic activity. Our results provide an example in which STING responses integrate information about the presence of relevant ER-transiting bacterial toxins into the innate inflammatory response and may help to explain the in vivo adjuvant effects of catalytically inactive toxins.


Subject(s)
Bacterial Toxins , Endoplasmic Reticulum Stress , Immunity, Innate , Membrane Proteins , Animals , Mice , Endoplasmic Reticulum Stress/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Signal Transduction , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Humans
17.
World J Microbiol Biotechnol ; 40(9): 286, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39083107

ABSTRACT

Staphylococcus aureus is a gram-positive bacteria, and its virulence factors can cause many kinds of infections, such as pneumonia, sepsis, enteritis and osteomyelitis. Traditional antibiotics can not only kill bacteria, but also easily lead to bacterial resistance. Jingfang Mixture (JFM) has the effects of inducing sweating and relieving the exterior, dispelling wind and eliminating dampness, and is commonly used in clinic to prevent and treat epidemic diseases and infectious diseases. The main purpose of this study is to explore the inhibitory effect of JFM on alpha-hemolysin (Hla) of S. aureus and to alleviate the damage caused by Hla. We found that JFM could inhibit the hemolytic activity, transcription level and neutralizing activity of Hla in a dose-dependent manner at the concentrations of 125, 250 and 500 µg/mL, without affecting the growth of bacteria. In addition, JFM reduced the damage of Hla to A549 cells and the release of lactate dehydrogenase (LDH). We also observed that in the S. aureus - induced pneumonia mouse model, JFM could significantly prolong the life of mice, reduce the bacterial load in the lungs, significantly improve the pathological state of the lungs and alleviate the damage caused by inflammatory factors, and the pathogenicity of gene deletion strain DU 1090 of S. aureus to pneumonia mice was also significantly reduced. In conclusion, this study proved that JFM is a potential drug against S. aureus infection, and this study provided a preliminary study for better guidance of clinical drug use.


Subject(s)
Anti-Bacterial Agents , Hemolysin Proteins , Staphylococcal Infections , Staphylococcus aureus , Animals , Female , Humans , Mice , A549 Cells , Anti-Bacterial Agents/pharmacology , Bacterial Load/drug effects , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Hemolysin Proteins/metabolism , Hemolysis/drug effects , Lung/microbiology , Lung/drug effects , Mice, Inbred BALB C , Pneumonia, Staphylococcal/drug therapy , Pneumonia, Staphylococcal/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Virulence Factors/genetics
18.
BMC Microbiol ; 24(1): 266, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39026145

ABSTRACT

BACKGROUND: Legionella pneumophila is a Gram-negative intracellular bacillus and is the causative agent of a severe form of pneumonia called Legionnaires' disease which accounts for 2-9% of cases of community acquired pneumonia. It produces an extremely large protein belonging to the RTX (Repeats in ToXin) family, called RtxA, and we previously reported that RtxA is transported by a dedicated type 1 secretion system (T1SS) to the cell surface. RTX proteins have been shown to participate in the virulence or biofilm formation of various bacteria, the most studied models being the pore forming hemolysin A (HlyA) of Escherichia coli and the biofilm associated protein LapA of P. fluorescens. LapA localization depends on the enzymatic release by LapD/LapG complex activity. This study aimed to elucidate the dual localization (cell surface associated or released state) of L. pneumophila RTX protein (RtxA) and whether this released versus sequestered state of RtxA plays a role in L. pneumophila virulence. RESULTS: The hereby work reveals that, in vitro, LapG periplasmic protease cleaves RtxA N-terminus in the middle of a di-alanine motif (position 108-109). Consistently, a strain lacking LapG protease maintains RtxA on the cell surface, whereas a strain lacking the c-di-GMP receptor LapD does not exhibit cell surface RtxA because of its continuous cleavage and release, as in the LapA-D-G model of Pseudomonas fluorescens. Interestingly, our data point out a key role of RtxA in enhancing the infection process of amoeba cells, regardless of its location (embedded or released); therefore, this may be the result of a secondary role of this surface protein. CONCLUSIONS: This is the first experimental identification of the cleavage site within the RTX protein family. The primary role of RtxA in Legionella is still questionable as in many other bacterial species, hence it sounds reasonable to propose a major function in biofilm formation, promoting cell aggregation when RtxA is embedded in the outer membrane and facilitating biofilm dispersion in case of RtxA release. The role of RtxA in enhancing the infection process may be a result of its action on host cells (i.e., PDI interaction or pore-formation), and independently of its status (embedded or released).


Subject(s)
Bacterial Proteins , Legionella pneumophila , Legionella pneumophila/pathogenicity , Legionella pneumophila/metabolism , Legionella pneumophila/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Virulence , Bacterial Toxins/metabolism , Biofilms/growth & development , Legionnaires' Disease/microbiology , Type I Secretion Systems/metabolism , Type I Secretion Systems/genetics , Cell Membrane/metabolism
19.
Sci Total Environ ; 948: 174745, 2024 Oct 20.
Article in English | MEDLINE | ID: mdl-39032754

ABSTRACT

Harmful cyanobacterial blooms will be more intense and frequent in the future, contaminating surface waters with cyanotoxins and posing a threat to communities heavily reliant on surface water usage for crop irrigation. Constructed wetlands (CWs) are proposed to ensure safe crop irrigation, but more research is needed before implementation. The present study operated 28 mesocosms in continuous mode mimicking horizontal sub-surface flow CWs. Mesocosms were fed with synthetic lake water and spiked periodically with two cyanotoxins, microcystin-LR (MC-LR) and cylindrospermopsin (CYN), at environmentally relevant cyanotoxins concentrations (10 µg L-1). The influence of various design factors, including plant species, porous media, and seasonality, was explored. The mesocosms achieved maximum MC-LR and CYN mass removal rates of 95 % and 98 %, respectively. CYN removal is reported for the first time in CWs mimicking horizontal sub-surface flow CWs. Planted mesocosms consistently outperformed unplanted mesocosms, with Phragmites australis exhibiting superior cyanotoxin mass removal compared to Juncus effusus. Considering evapotranspiration, J. effusus yielded the least cyanotoxin-concentrated effluent due to the lower water losses in comparison with P. australis. Using the P-kC* model, different scaling-up scenarios for future piloting were calculated and discussed. Additionally, bacterial community structure was analyzed through correlation matrices and differential taxa analyses, offering valuable insights into their removal of cyanotoxins. Nevertheless, attempts to validate microcystin-LR biotransformation via the known mlrA gene degradation pathway were unfruitful, indicating alternative enzymatic degradation pathways occurring in such complex CW systems. Further investigation into the precise molecular mechanisms of removal and the identification of transformation products is needed for the comprehensive understanding of cyanotoxin mitigation in CW. This study points towards the feasibility of horizontal sub-surface flow CWs to be employed to control cyanotoxins in irrigation or recreational waters.


Subject(s)
Alkaloids , Bacterial Toxins , Biodegradation, Environmental , Cyanobacteria Toxins , Marine Toxins , Microcystins , Uracil , Wetlands , Microcystins/metabolism , Marine Toxins/metabolism , Alkaloids/metabolism , Uracil/analogs & derivatives , Uracil/metabolism , Bacterial Toxins/metabolism , Water Pollutants, Chemical/metabolism , Water Pollutants, Chemical/analysis , Cyanobacteria/metabolism
20.
Infect Immun ; 92(8): e0013324, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38953668

ABSTRACT

Staphylococcus aureus α-hemolysin (Hla) is a pore-forming toxin critical for the pathogenesis of skin and soft tissue infections, which causes the pathognomonic lesion of cutaneous necrosis (dermonecrosis) in mouse models. To determine the mechanism by which dermonecrosis develops during S. aureus skin infection, mice were given control serum, Hla-neutralizing antiserum, or an inhibitor of Hla receptor [A-disintegrin and metalloprotease 10 (ADAM10) inhibitor] followed by subcutaneous infection by S. aureus, and the lesions were evaluated using immunohistochemistry and immunofluorescence. Hla induced apoptosis in the vascular endothelium at 6 hours post-infection (hpi), followed by apoptosis in keratinocytes at 24 hpi. The loss of vascular endothelial (VE)-cadherin expression preceded the loss of epithelial-cadherin expression. Hla also induced hypoxia in the keratinocytes at 24 hpi following vascular injury. Treatment with Hla-neutralizing antibody or ADAM10 inhibitor attenuated early cleavage of VE-cadherin, cutaneous hypoxia, and dermonecrosis. These findings suggest that Hla-mediated vascular injury with cutaneous hypoxia underlies the pathogenesis of S. aureus-induced dermonecrosis.


Subject(s)
ADAM10 Protein , Bacterial Toxins , Cadherins , Hemolysin Proteins , Keratinocytes , Necrosis , Staphylococcus aureus , Animals , Hemolysin Proteins/metabolism , Hemolysin Proteins/toxicity , Mice , Bacterial Toxins/toxicity , Bacterial Toxins/metabolism , Staphylococcus aureus/pathogenicity , Keratinocytes/microbiology , Keratinocytes/metabolism , ADAM10 Protein/metabolism , Cadherins/metabolism , Apoptosis , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Antigens, CD/metabolism , Membrane Proteins/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Staphylococcal Skin Infections/immunology , Skin/pathology , Skin/microbiology , Female , Endothelium, Vascular/pathology , Endothelium, Vascular/microbiology , Endothelium, Vascular/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL