Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.734
Filter
1.
Immunohorizons ; 8(9): 606-621, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39240270

ABSTRACT

The human body harbors a substantial population of bacteria, which may outnumber host cells. Thus, there are multiple interactions between both cell types. Given the common presence of Staphylococcus aureus in the human body and the role of Th17 cells in controlling this pathogen on mucous membranes, we sought to investigate the effect of α-hemolysin, which is produced by this bacterium, on differentiating Th17 cells. RNA sequencing analysis revealed that α-hemolysin influences the expression of signature genes for Th17 cells as well as genes involved in epigenetic regulation. We observed alterations in various histone marks and genome methylation levels via whole-genome bisulfite sequencing. Our findings underscore how bacterial proteins can significantly influence the transcriptome, epigenome, and phenotype of human Th17 cells, highlighting the intricate and complex nature of the interaction between immune cells and the microbiota.


Subject(s)
Bacterial Toxins , Epigenesis, Genetic , Hemolysin Proteins , Staphylococcus aureus , Th17 Cells , Staphylococcus aureus/immunology , Staphylococcus aureus/genetics , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Hemolysin Proteins/metabolism , Humans , Th17 Cells/immunology , Th17 Cells/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , DNA Methylation , Cell Differentiation , Transcriptome
2.
Front Cell Infect Microbiol ; 14: 1403219, 2024.
Article in English | MEDLINE | ID: mdl-39253327

ABSTRACT

Introduction: Despite years of efforts to develop new antibiotics for eradicating multidrug-resistant (MDR) and multi-virulent Methicillin-Resistant Staphylococcus aureus (MRSA) and Vancomycin-Resistant Staphylococcus aureus (VRSA) infections, treatment failures and poor prognoses in most cases have been common. Therefore, there is an urgent need for new therapeutic approaches targeting virulence arrays. Our aim is to discover new anti-virulence therapies targeting MRSA and VRSA virulence arrays. Methodology: We employed phenotypic, molecular docking, and genetic studies to screen for anti-virulence activities among selected promising compounds: Coumarin, Simvastatin, and Ibuprofen. Results: We found that nearly all detected MRSA and VRSA strains exhibited MDR and multi-virulent profiles. The molecular docking results aligned with the phenotypic and genetic assessments of virulence production. Biofilm and hemolysin productions were inhibited, and all virulence genes were downregulated upon treatment with sub-minimum inhibitory concentration (sub-MIC) of these promising compounds. Ibuprofen was the most active compound, exhibiting the highest inhibition and downregulation of virulence gene products. Moreover, in vivo and histopathological studies confirmed these results. Interestingly, we observed a significant decrease in wound area and improvements in re-epithelialization and tissue organization in the Ibuprofen and antimicrobial treated group compared with the group treated with antimicrobial alone. These findings support the idea that a combination of Ibuprofen and antimicrobial drugs may offer a promising new therapy for MRSA and VRSA infections. Conclusion: We hope that our findings can be implemented in clinical practice to assist physicians in making the most suitable treatment decisions.


Subject(s)
Anti-Bacterial Agents , Biofilms , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Molecular Docking Simulation , Staphylococcal Infections , Vancomycin-Resistant Staphylococcus aureus , Virulence Factors , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Methicillin-Resistant Staphylococcus aureus/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Biofilms/drug effects , Virulence Factors/genetics , Vancomycin-Resistant Staphylococcus aureus/drug effects , Animals , Virulence/drug effects , Ibuprofen/pharmacology , Ibuprofen/therapeutic use , Humans , Coumarins/pharmacology , Coumarins/therapeutic use , Mice , Disease Models, Animal , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics , Drug Resistance, Multiple, Bacterial
3.
Pestic Biochem Physiol ; 204: 106068, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277415

ABSTRACT

The insecticidal crystalline (Cry) and vegetative insecticidal (Vip) proteins derived from Bacillus thuringiensis (Bt) are used globally to manage insect pests, including the cotton bollworm, Helicoverpa armigera, one of the world's most damaging agricultural pests. Cry proteins bind to the ATP-binding cassette transporter C2 (ABCC2) receptor on the membrane surface of larval midgut cells, resulting in Cry toxin pores, and ultimately leading to cell swelling and/or lysis. Insect aquaporin (AQP) proteins within the membranes of larval midgut cells are proposed to allow the rapid influx of water into enterocytes following the osmotic imbalance triggered by the formation of Cry toxin pores. Here, we examined the involvement of H. armigera AQPs in Cry1Ac-induced osmotic cell swelling. We identified and characterized eight H. armigera AQPs and demonstrated that five are functional water channel proteins. Three of these (HaDrip1, HaPrip, and HaEglp1) were found to be expressed in the larval midgut. Xenopus laevis oocytes co-expressing the known Cry1Ac receptor HaABCC2 and each of the three HaAQPs displayed abnormal morphology and were lysed following exposure to Cry1Ac, suggesting a rapid influx of water was induced after Cry1Ac pore formation. In contrast, oocytes producing either HaABCC2 or HaAQP alone failed to swell or lyse after treatment with Cry1Ac, implying that both Cry1Ac pore formation and HaAQP function are needed for osmotic cell swelling. However, CRISPR/Cas9-mediated knockout of any one of the three HaAQP genes failed to cause significant changes in susceptibility to the Bt toxins Cry1Ac, Cry2Ab, or Vip3Aa. Our findings suggest that the multiple HaAQPs produced in larval midgut cells compensate for each other in allowing for the rapid influx of water in H. armigera midgut cells following Cry toxin pore formation, and that mutations affecting a single HaAQP are unlikely to confer resistance to Bt proteins.


Subject(s)
Aquaporins , Bacillus thuringiensis Toxins , Bacterial Proteins , Endotoxins , Hemolysin Proteins , Larva , Moths , Animals , Bacillus thuringiensis Toxins/toxicity , Hemolysin Proteins/toxicity , Hemolysin Proteins/pharmacology , Hemolysin Proteins/metabolism , Endotoxins/toxicity , Endotoxins/pharmacology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Moths/drug effects , Moths/metabolism , Moths/genetics , Larva/drug effects , Larva/metabolism , Aquaporins/metabolism , Aquaporins/genetics , Insect Proteins/metabolism , Insect Proteins/genetics , Bacillus thuringiensis/metabolism , Bacillus thuringiensis/genetics , Xenopus laevis , Oocytes/metabolism , Oocytes/drug effects , Insecticides/toxicity , Insecticides/pharmacology , Osmosis , Helicoverpa armigera
4.
Virulence ; 15(1): 2399798, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39229975

ABSTRACT

Staphylococcus aureus is the most common cause of skin and soft tissue infections (SSTIs) with Methicillin-Resistant S. aureus (MRSA) strains being a major contributor in both community and hospital settings. S. aureus relies on metabolic diversity and a large repertoire of virulence factors to cause disease. This includes α-hemolysin (Hla), an integral player in tissue damage found in various models, including SSTIs. Previously, we identified a role for the Spx adapter protein, YjbH, in the regulation of several virulence factors and as an inhibitor of pathogenesis in a sepsis model. In this study, we found that YjbH is critical for tissue damage during SSTI, and its absence leads to decreased proinflammatory chemokines and cytokines in the skin. We identified no contribution of YjbI, encoded on the same transcript as YjbH. Using a combination of reporters and quantitative hemolysis assays, we demonstrated that YjbH impacts Hla expression and activity both in vitro and in vivo. Additionally, expression of Hla from a non-native promoter reversed the tissue damage phenotype of the ΔyjbIH mutant. Lastly, we identified reduced Agr activity as the likely cause for reduced Hla production in the ΔyjbH mutant. This work continues to define the importance of YjbH in the pathogenesis of S. aureus infection as well as identify a new pathway important for Hla production.


Subject(s)
Bacterial Proteins , Bacterial Toxins , Gene Expression Regulation, Bacterial , Hemolysin Proteins , Staphylococcus aureus , Trans-Activators , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/immunology , Staphylococcus aureus/genetics , Mice , Animals , Trans-Activators/genetics , Trans-Activators/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/pathology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/immunology , Skin/microbiology , Skin/pathology , Skin/immunology , Virulence Factors/genetics , Humans , Soft Tissue Infections/microbiology , Soft Tissue Infections/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cytokines/metabolism , Cytokines/immunology , Cytokines/genetics
5.
Biochem J ; 481(19): 1349-1377, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39268843

ABSTRACT

Cholesterol-dependent cytolysins (CDCs) are the distinct class of ß-barrel pore-forming toxins (ß-PFTs) that attack eukaryotic cell membranes, and form large, oligomeric, transmembrane ß-barrel pores. Listeriolysin O (LLO) is a prominent member in the CDC family. As documented for the other CDCs, membrane cholesterol is essential for the pore-forming functionality of LLO. However, it remains obscure how exactly cholesterol facilitates its pore formation. Here, we show that cholesterol promotes both membrane-binding and oligomerization of LLO. We demonstrate cholesterol not only facilitates membrane-binding, it also enhances the saturation threshold of LLO-membrane association, and alteration of the cholesterol-recognition motif in the LLO mutant (LLOT515G-L516G) compromises its pore-forming efficacy. Interestingly, such defect of LLOT515G-L516G could be rescued in the presence of higher membrane cholesterol levels, suggesting cholesterol can augment the pore-forming efficacy of LLO even in the absence of a direct toxin-cholesterol interaction. Furthermore, we find the membrane-binding and pore-forming abilities of LLOT515G-L516G, but not those of LLO, correlate with the cholesterol-dependent rigidity/ordering of the membrane lipid bilayer. Our data further suggest that the line tension derived from the lipid phase heterogeneity of the cholesterol-containing membranes could play a pivotal role in LLO function, particularly in the absence of cholesterol binding. Therefore, in addition to its receptor-like role, we conclude cholesterol can further facilitate the pore-forming, membrane-damaging functionality of LLO by asserting the optimal physicochemical environment in membranes. To the best of our knowledge, this aspect of the cholesterol-mediated regulation of the CDC mode of action has not been appreciated thus far.


Subject(s)
Bacterial Toxins , Cholesterol , Heat-Shock Proteins , Hemolysin Proteins , Cholesterol/metabolism , Hemolysin Proteins/metabolism , Hemolysin Proteins/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Cell Membrane/metabolism , Humans , Protein Binding , Membrane Lipids/metabolism , Membrane Lipids/chemistry
6.
Arch Microbiol ; 206(10): 401, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261350

ABSTRACT

Staphylococcus aureus is a notorious pathogen predominantly involved in skin and soft tissue infections, exhibiting a distinct innate sex bias. This study explores the influence of testosterone on the virulence of S. aureus and elucidates its underlying mechanisms. Utilizing a skin abscess model in intact and castrated male mice, we assessed the effects of testosterone on S. aureus pathogenicity. Compared to controls, castrated mice showed significantly reduced abscess sizes and decreased bacterial loads, highlighting the role of testosterone in modulating the severity of S. aureus infections. In vitro experiments revealed that testosterone enhances the hemolytic activity, cytotoxicity, and oxidative stress resistance of S. aureus. Real-time quantitative PCR analysis showed a significant upregulation of the genes encoding α-hemolysin (hla) and phenol-soluble modulin (psmα). Importantly, testosterone treatment significantly enhanced the expression of the accessory gene regulator (Agr) quorum-sensing system components (agrC, agrA, agrB, agrD), while the SaeRS system (saeR, saeS, and sbi) exhibited only slight changes. Gene knockout experiments revealed that deletion of agrC, rather than saeRS and agrBD, abolishes the testosterone-induced enhancement of hemolysis and gene expression, underscoring the key role of AgrC. Molecular docking simulations indicated a direct interaction between testosterone and AgrC protein, with a strong binding affinity at the active site residue SER201. This study provides new insights into the mechanistic basis of how testosterone enhances the pathogenicity of S. aureus, potentially contributing to increased male susceptibility to S. aureus infections and offering a targeted approach for therapeutic interventions.


Subject(s)
Bacterial Proteins , Staphylococcal Infections , Staphylococcus aureus , Testosterone , Male , Testosterone/pharmacology , Testosterone/metabolism , Animals , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Mice , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence , Staphylococcal Infections/microbiology , Trans-Activators/genetics , Trans-Activators/metabolism , Gene Expression Regulation, Bacterial , Quorum Sensing , Molecular Docking Simulation , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Abscess/microbiology , Hemolysis , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics
7.
Virulence ; 15(1): 2388219, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39192628

ABSTRACT

Clostridium perfringens type A causes gas gangrene, which involves muscle infection. Both alpha toxin (PLC), encoded by the plc gene, and perfringolysin O (PFO), encoded by the pfoA gene, are important when type A strains cause gas gangrene in a mouse model. This study used the differentiated C2C12 muscle cell line to test the hypothesis that one or both of those toxins contributes to gas gangrene pathogenesis by releasing growth nutrients from muscle cells. RT-qPCR analyses showed that the presence of differentiated C2C12 cells induces C. perfringens type A strain ATCC3624 to upregulate plc and pfoA expression, as well as increase expression of several regulatory genes, including virS/R, agrB/D, and eutV/W. The VirS/R two component regulatory system (TCRS) and its coupled Agr-like quorum sensing system, along with the EutV/W TCRS (which regulates expression of genes involved in ethanolamine [EA] utilization), were shown to mediate the C2C12 cell-induced increase in plc and pfoA expression. EA was demonstrated to increase toxin gene expression. ATCC3624 growth increased in the presence of differentiated C2C12 muscle cells and this effect was shown to involve both PFO and PLC. Those membrane-active toxins were each cytotoxic for differentiated C2C12 cells, suggesting they support ATCC3624 growth by releasing nutrients from differentiated C2C12 cells. These findings support a model where, during gas gangrene, increased production of PFO and PLC in the presence of muscle cells causes more damage to those host cells, which release nutrients like EA that are then used to support C. perfringens growth in muscle.


Subject(s)
Bacterial Toxins , Clostridium perfringens , Gas Gangrene , Type C Phospholipases , Clostridium perfringens/genetics , Clostridium perfringens/growth & development , Clostridium perfringens/metabolism , Clostridium perfringens/physiology , Mice , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Line , Gas Gangrene/microbiology , Type C Phospholipases/genetics , Type C Phospholipases/metabolism , Cell Differentiation , Muscle Cells/microbiology , Muscle Cells/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Quorum Sensing
8.
J Agric Food Chem ; 72(33): 18708-18719, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39106049

ABSTRACT

The extensive use of Bacillus thuringiensis (Bt) in pest management has driven the evolution of pest resistance to Bt toxins, particularly Cry1Ac. Effective management of Bt resistance necessitates a good understanding of which pest proteins interact with Bt toxins. In this study, we screened a Helicoverpa armigera larval midgut cDNA library and captured 208 potential Cry1Ac-interacting proteins. Among these, we further examined the interaction between Cry1Ac and a previously unknown Cry1Ac-interacting protein, HaDALP (H. armigera death-associated LIM-only protein), as well as its role in toxicology. The results revealed that HaDALP specifically binds to both the Cry1Ac protoxin and activated toxin, significantly enhancing cell and larval tolerance to Cry1Ac. Additionally, HaDALP was overexpressed in a Cry1Ac-resistant H. armigera strain. These findings reveal a greater number of Cry1Ac-interacting proteins than previously known and demonstrate, for the first time, that HaDALP reduces Cry1Ac toxicity by sequestering both the protoxin and activated toxin.


Subject(s)
Bacillus thuringiensis Toxins , Bacterial Proteins , Endotoxins , Hemolysin Proteins , Insect Proteins , Insecticides , Larva , Moths , Animals , Bacillus thuringiensis Toxins/metabolism , Bacillus thuringiensis Toxins/toxicity , Bacillus thuringiensis Toxins/chemistry , Endotoxins/metabolism , Endotoxins/genetics , Endotoxins/toxicity , Hemolysin Proteins/metabolism , Hemolysin Proteins/pharmacology , Hemolysin Proteins/toxicity , Hemolysin Proteins/genetics , Moths/metabolism , Moths/drug effects , Moths/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/toxicity , Insect Proteins/metabolism , Insect Proteins/genetics , Larva/metabolism , Larva/drug effects , Larva/growth & development , Larva/genetics , Insecticides/toxicity , Insecticides/pharmacology , Insecticides/chemistry , Bacillus thuringiensis/chemistry , Bacillus thuringiensis/metabolism , Bacillus thuringiensis/genetics , Insecticide Resistance/genetics , Pest Control, Biological , Helicoverpa armigera
9.
BMC Plant Biol ; 24(1): 781, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39148017

ABSTRACT

BACKGROUND: Sudden temperature drops, resulting from extreme weather events, often occur during the boll-setting period of cotton in Xinjiang, China, causing decreased expression of Bacillus thuringiensis (Bt) insecticidal proteins in cotton bolls. The precise threshold temperatures and durations that lead to significant changes in Cry1Ac endotoxin levels under low temperatures remain unclear. To address this, we investigated the effects of different temperatures and stress durations on Cry1Ac endotoxin levels in cotton bolls. In 2020-2021, two Bt transgenic cotton varieties, conventional Sikang1 and hybrid Sikang3, were selected as experimental materials. Various low temperatures (ranging from 16 to 20 °C) with different durations (12 h, 24 h and 48 h) were applied during the peak boll-setting period. RESULTS: As the temperature decreased, the Cry1Ac endotoxin content in the boll shell, fiber, and seed exhibited a declining trend. Moreover, the threshold temperature which caused a significant reduction in Cry1Ac endotoxin content increased with the prolonged duration of low-temperature stress. Among the components of cotton bolls, seeds were most affected by low-temperature stress, with the threshold temperature for a significant reduction in Cry1Ac endotoxin content ranging from 17 °C to 19 °C. Correlation analysis indicated that low temperatures led to a decrease in protein synthesis capacity and an increase in degradation ability, resulting in reduced Cry1Ac endotoxin content. Pathway analysis revealed that both free amino acid and peptidase had significant negative effects on Cry1Ac endotoxin content. CONCLUSION: In summary, when the daily average temperature was ≤ 19 °C, implementing cultural practices to reduce free amino acid content and peptidase activity could serve as effective cold defense strategies for Bt cotton production.


Subject(s)
Bacillus thuringiensis Toxins , Bacterial Proteins , Cold Temperature , Endotoxins , Gossypium , Hemolysin Proteins , Nitrogen , Seeds , Gossypium/genetics , Gossypium/metabolism , Hemolysin Proteins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Seeds/metabolism , Nitrogen/metabolism , Plants, Genetically Modified , Bacillus thuringiensis
10.
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
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.
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
13.
Microb Pathog ; 194: 106815, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032674

ABSTRACT

Cancer is one of the main causes of death in the world. Resistance to anticancer treatments in patients with advanced solid tumors leads to new treatments. Therefore, more alternative anticancer methods have been found over time with greater specificity against tumor cells and with less or no adverse effects on normal cells. Bacterial spores of obligate anaerobes exclusively germinate in the hypoxic/necrotic areas and not in the well oxygenated areas of the body. This unique phenomenon has been exploited in using bacterial spores as a remedy for cancer. Bacterial toxins also play a significant role in either directly killing tumor cells or altering the cellular processes of the tumor cells which ultimately leads to the inhibition and regression of the solid tumor. In the microbial environment, pathogens such as Staphylococcus aureus, Bacillus cereus, or Streptococcus pyogenes produce hemolysin. This protein is used as an anti-cancer protein. To identify the production of hemolysin by bacteria, which can destroy cancer cells more effectively, different bacterial strains were first cultured in blood agar culture medium. The Strains that completely lysed red blood cells, creating transparent zones, were selected for further investigation. Then, to find out which strains have more ability to lyse red blood cells, the qualitative method of halo diameter measurement was used. Also, using quantitative methods, hemolysin strength in microtubes was determined compared to control samples. The results of the hemolysis in the microtube and the qualitative test results showed similar results. In the next step, the cell viability test was performed with the partially purified proteins. Then, bioinformatics studies such as secondary structure investigation, physicochemical properties, pseudo amino acid composition, and molecular docking were performed. The results of molecular docking showed that the hemolysin protein has the highest affinity for the cholesterol of the cytoplasmic membrane, respectively, of Bacillus subtilis, Bacillus cereus, and Staphylococcus aureus bacteria which play a significant role in either directly killing tumor cells or altering the cellular processes of the tumor cells which ultimately leads to the inhibition and regression of the solid tumor.


Subject(s)
Antineoplastic Agents , Computational Biology , Hemolysin Proteins , Staphylococcus aureus , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics , Humans , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Staphylococcus aureus/genetics , Antineoplastic Agents/pharmacology , Bacillus cereus/metabolism , Bacillus cereus/drug effects , Bacillus cereus/genetics , Hemolysis/drug effects , Erythrocytes/drug effects , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacteria/metabolism , Bacteria/drug effects , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/genetics , Molecular Docking Simulation , Neoplasms/drug therapy
14.
Nat Microbiol ; 9(9): 2448-2461, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38965331

ABSTRACT

Interactions between microbiota and enteric pathogens can promote colonization resistance or enhance pathogenesis. The pathobiont Enterococcus faecalis increases enterohaemorrhagic E. coli (EHEC) virulence by upregulating Type 3 Secretion System (T3SS) expression, effector translocation, and attaching and effacing (AE) lesion formation on enterocytes, but the mechanisms underlying this remain unknown. Using co-infection of organoids, metabolomics, supplementation experiments and bacterial genetics, here we show that co-culture of EHEC with E. faecalis increases the xanthine-hypoxanthine pathway activity and adenine biosynthesis. Adenine or E. faecalis promoted T3SS gene expression, while transcriptomics showed upregulation of adeP expression, which encodes an adenine importer. Mechanistically, adenine relieved High hemolysin activity (Hha)-dependent repression of T3SS gene expression in EHEC and promoted AE lesion formation in an AdeP-dependent manner. Microbiota-derived purines, such as adenine, support multiple beneficial host responses; however, our data show that this metabolite also increases EHEC virulence, highlighting the complexity of pathogen-microbiota-host interactions in the gut.


Subject(s)
Adenine , Enterococcus faecalis , Enterohemorrhagic Escherichia coli , Gene Expression Regulation, Bacterial , Type III Secretion Systems , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/metabolism , Virulence , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Enterococcus faecalis/pathogenicity , Adenine/metabolism , Adenine/pharmacology , Animals , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Mice , Escherichia coli Infections/microbiology , Humans , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics , Host-Pathogen Interactions , Coculture Techniques , Enterocytes/microbiology , Enterocytes/metabolism , Xanthine/metabolism , Hypoxanthine/metabolism , Virulence Factors/metabolism , Virulence Factors/genetics , Gastrointestinal Microbiome
15.
Microb Pathog ; 194: 106801, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39025378

ABSTRACT

Listeria monocytogenes (L. monocytogenes) is a prevalent foodborne pathogen with a remarkable capacity to form biofilms on utensil surfaces. The Listeriolysin O (LLO) exhibits hemolytic activity, which is responsible for causing human infections. In this study, we investigated the inhibitory effect and mechanism of oregano essential oil (OEO) on L. monocytogenes, evaluated the effects on its biofilm removal and hemolytic activity. The minimum inhibitory concentration (MIC) of OEO against L. monocytogenes was 0.03 % (v/v). L. monocytogenes was treated with OEO at 3/2 MIC for 30 min the bacteria was decreased below the detection limit (10 CFU/mL) in PBS and TSB (the initial bacterial load was about 6.5 log CFU/mL). The level of L. monocytogenes in minced pork co-cultured with OEO (15 MIC) about 2.5 log CFU/g lower than that in the untreated group. The inhibitory mechanisms of OEO against planktonic L. monocytogenes encompassed perturbation of cellular morphology, elevation in reactive oxygen species levels, augmentation of lipid oxidation extent, hyperpolarization of membrane potential, and reduction in intracellular ATP concentration. In addition, OEO reduced biofilm coverage on the surface of glass slides by 62.03 % compared with the untreated group. Meanwhile, OEO (1/8 MIC) treatment reduced the hemolytic activity of L. monocytogenes to 24.6 % compared with the positive control. Molecular docking suggested carvacrol and thymol might reduce the hemolytic activity of L. monocytogenes. The results of this study demonstrate that OEO exhibits inhibitory effects against L. monocytogenes, biofilms and LLO, which had potential as natural antimicrobial for the inhibition of L. monocytogenes.


Subject(s)
Anti-Bacterial Agents , Bacterial Toxins , Biofilms , Hemolysin Proteins , Listeria monocytogenes , Microbial Sensitivity Tests , Oils, Volatile , Origanum , Reactive Oxygen Species , Listeria monocytogenes/drug effects , Biofilms/drug effects , Biofilms/growth & development , Oils, Volatile/pharmacology , Oils, Volatile/chemistry , Origanum/chemistry , Hemolysin Proteins/metabolism , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/pharmacology , Bacterial Toxins/metabolism , Bacterial Toxins/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Anti-Bacterial Agents/pharmacology , Animals , Heat-Shock Proteins/metabolism , Hemolysis/drug effects , Swine , Adenosine Triphosphate/metabolism , Membrane Potentials/drug effects , Molecular Docking Simulation , Cymenes
16.
Microbiol Spectr ; 12(8): e0061224, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38980021

ABSTRACT

Streptococcus suis type 2 (SS2) is a zoonotic pathogen capable of eliciting meningitis, presenting significant challenges to both the swine industry and public health. Suilysin (Sly), one of SS2 most potent virulence determinants, releases a surfeit of inflammatory agents following red blood cell lysis. Notably, while current research on Sly role in SS2-induced meningitis predominantly centers on its interaction with the blood-brain barrier (BBB), the repercussions of Sly hemolytic products on BBB function have largely been sidestepped. In this vein, our study delves into the ramifications of Sly-induced hemolysis on BBB integrity. We discern that Sly hemolytic derivatives exacerbate the permeability of Sly-induced in vitro BBB models. Within these Sly hemolytic products, Interleukin-33 (IL-33) disrupts the expression and distribution of Claudin-5 in brain microvascular endothelial cells, facilitating the release of Interleukin-6 (IL-6) and Interleukin-8 (IL-8), thereby amplifying BBB permeability. Preliminary mechanistic insights suggest that IL-33-driven expression of IL-6 and IL-8 is orchestrated by the p38-mitogen-activated protein kinase signaling, whereas matrix metalloproteinase 9 mediates IL-33-induced suppression of Claudin-5. To validate these in vitro findings, an SS2-infected mouse model was established, and upon intravenous administration of growth stimulation expressed gene 2 (ST2) antibodies, in vivo results further underscored the pivotal role of the IL-33/ST2 axis during SS2 cerebral invasion. In summation, this study pioneerly illuminates the involvement of Sly hemolytic products in SS2-mediated BBB compromise and spotlights the instrumental role and primary mechanism of IL-33 therein. These insights enrich our comprehension of SS2 meningitis pathogenesis, laying pivotal groundwork for therapeutic advancements against SS2-induced meningitis.IMPORTANCEThe treatment of meningitis caused by Streptococcus suis type 2 (SS2) has always been a clinical challenge. Elucidating the molecular mechanisms by which SS2 breaches the blood-brain barrier (BBB) is crucial for the development of meningitis therapeutics. Suilysin (Sly) is one of the most important virulence factors of SS2, which can quickly lyse red blood cells and release large amounts of damage-associated molecular patterns, such as hemoglobin, IL-33, cyclophilin A, and so on. However, the impact of these hemolytic products on the function of BBB is unknown and ignored. This study is the first to investigate the effect of Sly hemolytic products on BBB function. The data are crucial for the study of the pathogenesis of SS2 meningitis and can provide an important reference for the development of meningitis therapeutics.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Hemolysin Proteins , Hemolysis , Interleukin-33 , Streptococcus suis , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Animals , Mice , Interleukin-33/metabolism , Humans , Hemolysin Proteins/metabolism , Streptococcus suis/pathogenicity , Endothelial Cells/microbiology , Endothelial Cells/metabolism , Streptococcal Infections/microbiology , Interleukin-6/metabolism , Interleukin-6/genetics , Interleukin-8/metabolism , Swine , Matrix Metalloproteinase 9/metabolism
17.
ACS Infect Dis ; 10(8): 2886-2898, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39079033

ABSTRACT

Vibrio cholerae (V. cholerae), the etiological agent of cholera, employs various virulence factors to adapt and thrive within both aquatic and human host environments. Among these factors, the type VI secretion system (T6SS) stands out as one of the crucial determinants of its pathogenicity. Valine glycine repeat protein G1 (VgrG1) and hemolysin coregulated protein (HCP) are considered major effector molecules of T6SS. Previous studies have highlighted that VgrG1 interacts with HCP proteins. Additionally, it has been shown that VgrG1 possesses an actin cross-linking domain (ACD) with actin-binding activity. Interestingly, it was reported that purified HCP protein treatment increased the stress fibers within cells. Therefore, we hypothesize that HCP may interact with host cell actin, potentially playing a role in the cytoskeletal rearrangement during V. cholerae infection. To test this hypothesis, we characterized HCP from the V. cholerae O139 serotype and demonstrated its interaction with actin monomers. In silico analysis and experimental validation revealed the presence of an actin-binding site within HCP. Furthermore, overexpression of HCP resulted in its colocalization with actin stress fibers in host cells. Our findings establish HCP as an effector molecule for potent host cell actin cytoskeleton remodeling during V. cholerae infection, providing new insights into bacterial pathogenicity mechanisms. Understanding the interplay between bacterial effectors and host cell components is crucial for developing targeted therapeutic interventions against cholera and related infectious diseases.


Subject(s)
Actin Cytoskeleton , Bacterial Proteins , Vibrio cholerae , Vibrio cholerae/pathogenicity , Vibrio cholerae/metabolism , Vibrio cholerae/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Humans , Actin Cytoskeleton/metabolism , Host-Pathogen Interactions , Protein Binding , Virulence Factors/metabolism , Virulence Factors/genetics , Actins/metabolism , Cholera/microbiology , Hemolysin Proteins/metabolism
18.
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
19.
Mol Microbiol ; 122(2): 255-270, 2024 08.
Article in English | MEDLINE | ID: mdl-39030901

ABSTRACT

The flagellar MS-ring, uniquely constituted by FliF, is essential for flagellar biogenesis and functionality in several bacteria. The aim of this study was to dissect the role of FliF in the Gram-positive and peritrichously flagellated Bacillus cereus. We demonstrate that fliF forms an operon with the upstream gene fliE. In silico analysis of B. cereus ATCC 14579 FliF identifies functional domains and amino acid residues that are essential for protein functioning. The analysis of a ΔfliF mutant of B. cereus, constructed in this study using an in frame markerless gene replacement method, reveals that the mutant is unexpectedly able to assemble flagella, although in reduced amounts compared to the parental strain. Nevertheless, motility is completely abolished by fliF deletion. FliF deprivation causes the production of submerged biofilms and affects the ability of B. cereus to adhere to gastrointestinal mucins. We additionally show that the fliF deletion does not compromise the secretion of the three components of hemolysin BL, a toxin secreted through the flagellar type III secretion system. Overall, our findings highlight the important role of B. cereus FliF in flagella-related functions, being the protein required for complete flagellation, motility, mucin adhesion, and pellicle biofilms.


Subject(s)
Bacillus cereus , Bacterial Proteins , Biofilms , Flagella , Operon , Bacillus cereus/metabolism , Bacillus cereus/genetics , Flagella/metabolism , Flagella/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Biofilms/growth & development , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics , Bacterial Adhesion , Gene Expression Regulation, Bacterial , Gene Deletion , Membrane Proteins
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL