Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.369
Filter
1.
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
2.
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
3.
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
4.
Pestic Biochem Physiol ; 204: 106096, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277420

ABSTRACT

Transgenic crops producing insecticidal proteins from Bacillus thuringiensis (Bt) have revolutionized pest control. However, the evolution of resistance by target pests poses a significant threat to the long-term success of Bt crops. Understanding the genetics and mechanisms underlying Bt resistance is crucial for developing resistance detection methods and management tactics. The T92C mutation in a tetraspanin gene (HaTSPAN1), resulting in the L31S substitution, is associated with dominant resistance to Cry1Ac in a major pest, Helicoverpa armigera. Previous studies using CRISPR/Cas9 technique have demonstrated that knockin of the HaTSPAN1 T92C mutation confers a 125-fold resistance to Cry1Ac in the susceptible SCD strain of H. armigera. In this study, we employed the piggyBac transposon system to create two transgenic H. armigera strains based on SCD: one expressing the wild-type HaTSPAN1 gene (SCD-TSPANwt) and another expressing the T92C mutant form of HaTSPAN1 (SCD-TSPANmt). The SCD-TSPANmt strain exhibited an 82-fold resistance to Cry1Ac compared to the recipient SCD strain, while the SCD-TSPANwt strain remained susceptible. The Cry1Ac resistance followed an autosomal dominant inheritance mode and was genetically linked with the transgene locus in the SCD-TSPANmt strain of H. armigera. Our results further confirm the causal association between the T92C mutation of HaTSPAN1 and dominant resistance to Cry1Ac in H. armigera. Additionally, they suggest that the piggyBac-mediated transformation system we used in H. armigera is promising for functional investigations of candidate Bt resistance genes from other lepidopteran pests.


Subject(s)
Bacillus thuringiensis Toxins , Bacterial Proteins , Endotoxins , Hemolysin Proteins , Insecticide Resistance , Moths , Animals , Endotoxins/genetics , Endotoxins/pharmacology , Bacillus thuringiensis Toxins/pharmacology , Hemolysin Proteins/genetics , Hemolysin Proteins/pharmacology , Hemolysin Proteins/toxicity , Moths/drug effects , Moths/genetics , Insecticide Resistance/genetics , Bacterial Proteins/genetics , Alleles , Plants, Genetically Modified/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Bacillus thuringiensis/genetics , Insecticides/pharmacology , Insecticides/toxicity , Helicoverpa armigera
5.
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
6.
Curr Genet ; 70(1): 13, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39101952

ABSTRACT

Bacillus thuringiensis is the most widely used biopesticide, targets a diversity of insect pests belonging to several orders. However, information regarding the B. thuringiensis strains and toxins targeting Zeugodacus cucurbitae is very limited. Therefore, in the present study, we isolated and identified five indigenous B. thuringiensisstrains toxic to larvae of Z. cucurbitae. However, of five strains NBAIR BtPl displayed the highest mortality (LC50 = 37.3 µg/mL) than reference strain B. thuringiensis var. israelensis (4Q1) (LC50 = 45.41 µg/mL). Therefore, the NBAIR BtPl was considered for whole genome sequencing to identify the cry genes present in it. Whole genome sequencing of our strain revealed genome size of 6.87 Mb with 34.95% GC content. Homology search through the BLAST algorithm revealed that NBAIR BtPl is 99.8% similar to B. thuringiensis serovar tolworthi, and gene prediction through Prokka revealed 7406 genes, 7168 proteins, 5 rRNAs, and 66 tRNAs. BtToxin_Digger analysis of NBAIR BtPl genome revealed four cry gene families: cry1, cry2, cry8Aa1, and cry70Aa1. When tested for the presence of these four cry genes in other indigenous strains, results showed that cry70Aa1 was absent. Thus, the study provided a basis for predicting cry70Aa1 be the possible reason for toxicity. In this study apart from novel genes, we also identified other virulent genes encoding zwittermicin, chitinase, fengycin, and bacillibactin. Thus, the current study aids in predicting potential toxin-encoding genes responsible for toxicity to Z. cucurbitae and thus paves the way for the development of B. thuringiensis-based formulations and transgenic crops for management of dipteran pests.


Subject(s)
Bacillus thuringiensis , Bacterial Proteins , Genome, Bacterial , Whole Genome Sequencing , Bacillus thuringiensis/genetics , Animals , Bacterial Proteins/genetics , Bacillus thuringiensis Toxins/genetics , Endotoxins/genetics , Pest Control, Biological , Tephritidae/genetics , Tephritidae/microbiology , Hemolysin Proteins/genetics , Larva/genetics , Phylogeny
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.
Appl Microbiol Biotechnol ; 108(1): 432, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39102054

ABSTRACT

Infections caused by Staphylococcus aureus pose a significant global public problem. Therefore, new antibiotics and therapeutic strategies are needed to combat this pathogen. This investigation delves into the effects of iclaprim, a newly discovered inhibitor of folic acid synthesis, on S. aureus virulence. The phenotypic and genotypic effects of iclaprim were thoroughly examined in relation to virulence factors, biofilm formation, and dispersal, as well as partial virulence-encoding genes associated with exoproteins, adherence, and regulation in S. aureus MW2, N315, and ATCC 25923. Then, the in vivo effectiveness of iclaprim on S. aureus pathogenicity was explored by a Galleria mellonella larvae infection model. The use of iclaprim at sub-inhibitory concentrations (sub-MICs) resulted in a reduction of α-hemolysin (Hla) production and a differential effect on the activity of coagulase in S. aureus strains. The results of biofilm formation and eradication assay showed that iclaprim was highly effective in depolymerizing the mature biofilm of S. aureus strains at concentrations of 1 MIC or greater, however, inhibited the biofilm-forming ability of only strains N315 and ATCC 25923 at sub-MICs. Interestingly, treatment of strains with sub-MICs of iclaprim resulted in significant stimulation or suppression of most virulence-encoding genes expression. Iclaprim did not affect the production of δ-hemolysin or staphylococcal protein A (SpA), nor did it impact the total activity of proteases, nucleases, and lipases. In vivo testing showed that sub-MICs of iclaprim significantly improves infected larvae survival. The present study offered valuable insights towards a better understating of the influence of iclaprim on different strains of S. aureus. The findings suggest that iclaprim may have potential as an anti-virulence and antibiofilm agent, thus potentially mitigating the pathogenicity of S. aureus and improving clinical outcomes associated with infections caused by this pathogen. KEY POINTS: • Iclaprim effectively inhibits α-hemolysin production and biofilm formation in a strain-dependent manner and was an excellent depolymerizing agent of mature biofilm • Iclaprim affected the mRNA expression of virulence-encoding genes associated with exoproteins, adherence, and regulation • In vivo study in G. mellonella larvae challenged with S. aureus exhibited that iclaprim improves larvae survival.


Subject(s)
Anti-Bacterial Agents , Biofilms , Larva , Microbial Sensitivity Tests , Staphylococcal Infections , Staphylococcus aureus , Virulence Factors , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/genetics , Biofilms/drug effects , Animals , Virulence Factors/genetics , Anti-Bacterial Agents/pharmacology , Virulence/drug effects , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Larva/microbiology , Moths/microbiology , Hemolysin Proteins/genetics , Folic Acid/pharmacology , Folic Acid/biosynthesis , Folic Acid Antagonists/pharmacology , Coagulase/metabolism , Disease Models, Animal , Pyrimidines
10.
Int J Mol Sci ; 25(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39126052

ABSTRACT

Exopolysaccharides (EPSs) are carbohydrate polymers that are synthesized and secreted into the extracellular during the growth of microorganisms. Bacillus thuringiensis (Bt) is a type of entomopathogenic bacterium, that produces various insecticidal proteins and EPSs. In our previous study, the EPSs produced by Bt strains were first found to enhance the toxicity of insecticidal crystal proteins against Plutella xylostella. However, the response of the intestinal bacterial communities of P. xylostella under the action of EPSs is still unelucidated. In this study, 16S rRNA amplicon sequencing was used to characterize the intestinal bacterial communities in P. xylostella treated with EPSs alone, Cry1Ac protoxin alone, and both the Cry1Ac protoxin and EPSs. Compared with the control group, alpha diversity indices, the Chao1 and ACE indices were significantly altered after treatment with EPSs alone, and no significant difference was observed between the groups treated with Cry1Ac protoxin alone and Cry1Ac protoxin + EPSs. However, compared with the gut bacterial community feeding on Cry1Ac protoxin alone, the relative abundance of 31 genera was significantly changed in the group treated with Cry1Ac protoxin and EPSs. The intestinal bacteria, through the oral of Cry1Ac protoxin and EPSs, significantly enhanced the toxicity of the Cry1Ac protoxin towards the axenic P. xylostella. In addition, the relative abundance of the 16S rRNA gene in the chloroplasts of Brassica campestris decreased after adding EPSs. Taken together, these results show the vital contribution of the gut microbiota to the Bt strain-killing activity, providing new insights into the mechanism of the synergistic insecticidal activity of Bt proteins and EPSs.


Subject(s)
Bacillus thuringiensis Toxins , Bacterial Proteins , Endotoxins , Gastrointestinal Microbiome , Hemolysin Proteins , Moths , Animals , Gastrointestinal Microbiome/drug effects , Endotoxins/genetics , Hemolysin Proteins/genetics , Hemolysin Proteins/pharmacology , Moths/drug effects , Moths/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , RNA, Ribosomal, 16S/genetics , Bacillus thuringiensis/genetics , Insecticides/pharmacology
11.
J Agric Food Chem ; 72(36): 19689-19698, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39189874

ABSTRACT

Synergistic factors can enhance the toxicity of Bt toxins and delay the development of Bt resistance. Previous research has demonstrated that a Helicoverpa armigera cadherin fragment (HaCad-TBR) increased the toxicity of Cry1Ac in Plutella xylostella larvae but did not have a synergistic effect on Cry1B, Cry1C, and Cry1F toxins. In this study, a fusion protein (HaCad-TBR-2D3 VL) derived from HaCad-TBR and a Bt Cry1-specific antibody peptide was expressed in Escherichia coli. The HaCad-TBR-2D3 VL enhanced Cry1Ac toxicity more efficiently in insects and Sf9 cells than HaCad-TBR and also significantly increased the toxicity of Cry1B, Cry1C, and Cry1F toxins in insects. Further investigation indicated that the improved stability in insect midguts and higher binding capacity with Bt toxins contributed to the enhanced synergism of HaCad-TBR-2D3 VL over HaCad-TBR. This study suggested that Bt antibody fragments can potentially broaden the synergistic range of Bt receptor fragments, providing a theoretical foundation for developing broad-spectrum synergists for other biopesticides.


Subject(s)
Bacillus thuringiensis Toxins , Bacterial Proteins , Cadherins , Endotoxins , Hemolysin Proteins , Insect Proteins , Larva , Moths , Recombinant Fusion Proteins , Animals , Cadherins/genetics , Cadherins/metabolism , Cadherins/immunology , Cadherins/chemistry , Hemolysin Proteins/chemistry , Hemolysin Proteins/pharmacology , Hemolysin Proteins/immunology , Hemolysin Proteins/genetics , Endotoxins/immunology , Endotoxins/chemistry , Endotoxins/pharmacology , Endotoxins/metabolism , Endotoxins/genetics , Bacillus thuringiensis Toxins/chemistry , Bacillus thuringiensis Toxins/pharmacology , Moths/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Insect Proteins/immunology , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Proteins/metabolism , Larva/growth & development , Peptides/chemistry , Peptides/immunology , Peptides/pharmacology , Antibodies/immunology , Antibodies/chemistry , Bacillus thuringiensis/chemistry , Bacillus thuringiensis/genetics , Bacillus thuringiensis/metabolism , Insecticides/chemistry , Insecticides/pharmacology , Pest Control, Biological
12.
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
13.
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
14.
Biomolecules ; 14(7)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39062509

ABSTRACT

Cry toxins, produced by the bacterium Bacillus thuringiensis, are of significant agronomic value worldwide due to their potent and highly specific activity against various insect orders. However, some of these pore-forming toxins display specific activity against a range of human cancer cells whilst possessing no known insecticidal activity; Cry41Aa is one such toxin. Cry41Aa has similarities to its insecticidal counterparts in both its 3-domain toxic core structure and pore-forming abilities, but how it has evolved to target human cells is a mystery. This work shows that some insecticidal Cry toxins can enhance the toxicity of Cry41Aa against hepatocellular carcinoma cells, despite possessing no intrinsic toxicity themselves. This interesting crossover is not limited to human cancer cells, as Cry41Aa was found to inhibit some Aedes-active Cry toxins in mosquito larval assays. Here, we present findings that suggest that Cry41Aa shares a receptor with several insecticidal toxins, indicating a stronger evolutionary relationship than their divergent activities might suggest.


Subject(s)
Bacillus thuringiensis Toxins , Bacillus thuringiensis , Bacterial Proteins , Endotoxins , Hemolysin Proteins , Humans , Endotoxins/chemistry , Endotoxins/genetics , Endotoxins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacillus thuringiensis/genetics , Bacillus thuringiensis/metabolism , Bacillus thuringiensis Toxins/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Hemolysin Proteins/pharmacology , Animals , Insecticides/chemistry , Insecticides/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Aedes/drug effects , Aedes/genetics , Cell Line, Tumor
15.
Sci Rep ; 14(1): 15216, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956138

ABSTRACT

Here, we present the whole genome sequence of Bt S2160-1, a potential alternative to the mosquitocidal model strain, Bti. One chromosome genome and four mega-plasmids were contained in Bt S2160-1, and 13 predicted genes encoding predicted insecticidal crystal proteins were identified clustered on one plasmid pS2160-1p2 containing two pathogenic islands (PAIs) designed as PAI-1 (Cry54Ba, Cry30Ea4, Cry69Aa-like, Cry50Ba2-like, Cry4Ca1-like, Cry30Ga2, Cry71Aa-like, Cry72Aa-like, Cry70Aa-like, Cyt1Da2-like and Vpb4C1-like) and PAI-2 (Cyt1Aa-like, and Tpp80Aa1-like). The clusters appear to represent mosquitocidal toxin islands similar to pathogenicity islands. Transcription/translation of 10 of the 13 predicted genes was confirmed by whole-proteome analysis using LTQ-Orbitrap LC-MS/MS. In summary, the present study identified the existence of a mosquitocidal toxin island in Bacillus thuringiensis, and provides important genomic information for understanding the insecticidal mechanism of B. thuringiensis.


Subject(s)
Bacillus thuringiensis , Bacterial Proteins , Insecticides , Proteomics , Bacillus thuringiensis/genetics , Bacillus thuringiensis/metabolism , Proteomics/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Insecticides/pharmacology , Whole Genome Sequencing/methods , Genome, Bacterial , Endotoxins/genetics , Bacillus thuringiensis Toxins , Genomic Islands , Proteome , Plasmids/genetics , Tandem Mass Spectrometry , Animals , Hemolysin Proteins/genetics
16.
Gene ; 927: 148753, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38972556

ABSTRACT

BACKGROUND: Transgenic insect-resistant rice offers an environmentally friendly approach to mitigate yield losses caused by lepidopteran pests, such as stem borers. Bt (Bacillus thuringiensis) genes encode insecticidal proteins and are widely used to confer insect resistance to genetically modified crops. This study investigated the integration, inheritance, and expression characteristics of codon-optimised synthetic Bt genes, cry1C* and cry2A*, in transgenic early japonica rice lines. METHODS: The early japonica rice cultivar, Songgeng 9 (Oryza sativa), was transformed with cry1C* or cry2A*, which are driven by the ubi promoter via Agrobacterium tumefaciens-mediated transformation. Molecular analyses, including quantitative PCR (qPCR), enzyme-linked immunosorbent assay (ELISA), and Southern blot analysis were performed to confirm transgene integration, inheritance, transcriptional levels, and protein expression patterns across different tissues and developmental stages. RESULTS: Stable transgenic early japonica lines exhibiting single-copy transgene integration were established. Transcriptional analysis revealed variations in Bt gene expression among lines, tissues, and growth stages, with higher expression levels observed in leaves than in other organs. Notably, cry2A* exhibited consistently higher mRNA and protein levels than cry1C* across all examined tissues and developmental time points. Bt protein accumulation followed the trend of leaves > stem sheaths > young panicles > brown rice, with peak expression during the filling stage in the vegetative tissues. CONCLUSIONS: Synthetic cry2A* displayed markedly elevated transcription and translation compared to cry1C* in the transgenic early japonica rice lines examined. Distinct spatiotemporal patterns of Bt gene expression were elucidated, providing insights into the potential insect resistance conferred by these genes in rice. These findings will contribute to the development of insect-resistant japonica rice varieties and facilitate the rational deployment of Bt crops.


Subject(s)
Bacterial Proteins , Endotoxins , Oryza , Plants, Genetically Modified , Oryza/genetics , Oryza/parasitology , Plants, Genetically Modified/genetics , Animals , Endotoxins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Hemolysin Proteins/genetics , Bacillus thuringiensis Toxins/genetics , Bacillus thuringiensis/genetics , Moths/genetics , Gene Expression Regulation, Plant , Pest Control, Biological/methods
17.
Funct Integr Genomics ; 24(4): 129, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39039331

ABSTRACT

Genetically modified (GM) crops, expressing Bacillus thuringiensis (Bt) insecticidal toxins, have substantially transformed agriculture. Despite rapid adoption, their environmental and economic benefits face scrutiny due to unsustainable agricultural practices and the emergence of resistant pests like Spodoptera frugiperda, known as the fall armyworm (FAW). FAW's adaptation to Bt technology in corn and cotton compromises the long-term efficacy of Bt crops. To advance the understanding of the genetic foundations of resistance mechanisms, we conducted an exploratory comparative transcriptomic analysis of two divergent FAW populations. One population exhibited practical resistance to the Bt insecticidal proteins Cry1A.105 and Cry2Ab2, expressed in the genetically engineered MON-89Ø34 - 3 maize, while the other population remained susceptible to these proteins. Differential expression analysis supported that Cry1A.105 and Cry2Ab2 significantly affect the FAW physiology. A total of 247 and 254 differentially expressed genes were identified in the Cry-resistant and susceptible populations, respectively. By integrating our findings with established literature and databases, we underscored 53 gene targets potentially involved in FAW's resistance to Cry1A.105 and Cry2Ab2. In particular, we considered and discussed the potential roles of the differentially expressed genes encoding ABC transporters, G protein-coupled receptors, the P450 enzymatic system, and other Bt-related detoxification genes. Based on these findings, we emphasize the importance of exploratory transcriptomic analyses to uncover potential gene targets involved with Bt insecticidal proteins resistance, and to support the advantages of GM crops in the face of emerging challenges.


Subject(s)
Bacillus thuringiensis Toxins , Bacterial Proteins , Endotoxins , Hemolysin Proteins , Insecticide Resistance , Spodoptera , Transcriptome , Spodoptera/drug effects , Spodoptera/genetics , Animals , Endotoxins/genetics , Endotoxins/pharmacology , Hemolysin Proteins/genetics , Hemolysin Proteins/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Insecticide Resistance/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/parasitology , Zea mays/genetics , Zea mays/parasitology , Gene Expression Profiling
18.
ACS Appl Mater Interfaces ; 16(30): 40100-40110, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39038810

ABSTRACT

Biological and solid-state nanopores are at the core of transformative techniques and nanodevices, democratizing the examination of matter and biochemical reactions at the single-molecule level, with low cost, portability, and simplicity in operation. One of the crucial hurdles in such endeavors is the fast analyte translocation, which limits characterization, and a rich number of strategies have been explored over the years to overcome this. Here, by site-directed mutagenesis on the α-hemolysin protein nanopore (α-HL), sought to replace selected amino acids with glycine, electrostatic binding sites were induced on the nanopore's vestibule and constriction region and achieved in the most favorable case a 20-fold increase in the translocation time of short single-stranded DNA (ssDNA) at neutral pH, with respect to the wild-type (WT) nanopore. We demonstrated an efficient tool of controlling the ssDNA translocation time, via the interplay between the nanopore-ssDNA surface electrostatic interactions and electroosmotic flow, all mediated by the pH-dependent ionization of amino acids lining the nanopore's translocation pathway. Our data also reveal the nonmonotonic, pH-induced alteration of ssDNA average translocation time. Unlike mildly acidic conditions (pH ∼ 4.7), at a pH ∼ 2.8 maintained symmetrically or asymmetrically across the WT α-HL, we evidenced the manifestation of a dominant electroosmotic flow, determining the speeding up of the ssDNA translocation across the nanopore by counteracting the ssDNA-nanopore attractive electrostatic interactions. We envision potential applications of the presented approach by enabling easy-to-use, real-time detection of short ssDNA sequences, without the need for complex biochemical modifications to the nanopore to mitigate the fast translocation of such sequences.


Subject(s)
DNA, Single-Stranded , Electroosmosis , Hemolysin Proteins , Mutagenesis, Site-Directed , Nanopores , Hydrogen-Ion Concentration , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Static Electricity
19.
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
20.
Braz J Microbiol ; 55(3): 2071-2083, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38904690

ABSTRACT

Co-infection of Lactococcus garvieae and Aeromonas hydrophila, has been confirmed from diseased Nile Tilapia (Oreochromis niloticus), Chithralada strain cultured in a freshwater rearing pond of Alappuzha district of Kerala, India. The aetiological agents behind the disease outbreak were bacteriologically proven and confirmed by 16SrRNA sequencing and phylogenetic analysis. PCR detection of the virulent genes, showed existence of adhesin and hemolysin in L. garvieae and aerolysin in A. hydrophila strain obtained. To fulfil Koch's postulates, challenge experiments were conducted and median lethal dose (LD50) of L. garvieae and A. hydrophila was calculated as 1 × 105.91 CFU per mL and 1 × 105.2 CFU per mL respectively. Histopathologically, eyes, spleen, and kidney were the predominantly infected organs by L. garvieae and A. hydrophila. Out of the 13 antibiotics tested to check antibiotic susceptibility, L. garvieae showed resistance to almost 7 antibiotics tested, with a resistance to Ciprofloxacin while A. hydrophila was found resistant to Streptomycin and Erythromycin. Understanding the complex interaction between Gram-positive and Gram-negative bacteria in the disease process and pathogenesis in fish host will contribute to efficient treatment strategies. As a preliminary investigation into this complex interaction, the present study is aimed at phenotypic and genotypic characterization, pathogenicity evaluation, and antibiotic susceptibility of the co-infecting pathogens in a diseased sample of freshwater-farmed Nile tilapia.


Subject(s)
Aeromonas hydrophila , Anti-Bacterial Agents , Cichlids , Coinfection , Fish Diseases , Gram-Negative Bacterial Infections , Lactococcus , Phylogeny , Animals , Aeromonas hydrophila/genetics , Aeromonas hydrophila/isolation & purification , Aeromonas hydrophila/pathogenicity , Aeromonas hydrophila/classification , Aeromonas hydrophila/drug effects , Cichlids/microbiology , India , Fish Diseases/microbiology , Lactococcus/genetics , Lactococcus/isolation & purification , Lactococcus/classification , Lactococcus/pathogenicity , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/veterinary , Coinfection/microbiology , Coinfection/veterinary , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/veterinary , RNA, Ribosomal, 16S/genetics , Aquaculture , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL