Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 771
1.
Int J Biol Macromol ; 257(Pt 1): 128666, 2024 Feb.
Article En | MEDLINE | ID: mdl-38070805

Staphylococcus enterotoxin B (SEB) interacts with MHC-II molecules to overactivate immune cells and thereby to produce excessive pro-inflammatory cytokines. Disrupting the interactions between SEB and MHC-II helps eliminate the lethal threat posed by SEB. In this study, a de novo computational approach was used to design protein binders targeting SEB. The MHC-II binding domain of SEB was selected as the target, and the possible promising binding mode was broadly explored. The obtained original binder was folded into triple-helix bundles and contained 56 amino acids with molecular weight 5.9 kDa. The interface of SEB and the binder was highly hydrophobic. ProteinMPNN optimization further enlarged the hydrophobic region of the binder and improved the stability of the binder-SEB complex. In vitro study demonstrated that the optimized binder significantly inhibited the inflammatory response induced by SEB. Overall, our research demonstrated the applicability of this approach in de novo designing protein binders against SEB, and thereby providing potential therapeutics for SEB induced diseases.


Enterotoxins , Histocompatibility Antigens Class II , Enterotoxins/chemistry , Cytokines/metabolism
2.
Int J Biol Macromol ; 253(Pt 2): 126822, 2023 Dec 31.
Article En | MEDLINE | ID: mdl-37703983

Staphylococcus enterotoxin B (SEB) is one of the primary virulence factors of Staphylococcus aureus but there is still a lack of targeted drugs. SEB activates immune cells via interacting with MHC-II on antigen-presenting cells, leading to the production of large amounts of pro-inflammatory cytokines. Blocking the interaction between SEB and MHC-II can avert the overactivation of immune cells. Nanobodies are the smallest functional antibodies that can bind stably to antigens. In this study, an ideal approach to obtain specific nanobodies without immunizing camelids was introduced. We constructed a library containing up to 5 × 108 nanobodies, and then screened those targeting SEB by using yeast surface display (YSD) technique and fluorescence-activated cell sorting (FACS). A total of 8 nanobodies with divergent complementarity-determining regions (CDRs) sequences were identified and one candidate Nb8 with high affinity to SEB was isolated. In vitro study demonstrated that Nb8 significantly inhibited SEB-induced inflammatory response. Molecular docking simulation indicated that the unique CDR3 sequence contributed to the binding of Nb8 to the MHC-II binding domain of SEB and accordingly cut off the connection between SEB and MHC-II. Our efforts contributed to the development of specific nanobodies for eliminating the threats of SEB.


Saccharomyces cerevisiae , Single-Domain Antibodies , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Molecular Docking Simulation , Enterotoxins/chemistry
3.
Int J Biol Macromol ; 237: 124172, 2023 May 15.
Article En | MEDLINE | ID: mdl-36966860

Despite the great potential of protein drugs as intracellular therapeutic agents, the unmet challenge in breaking through the cell membrane barrier and delivering them to intracellular targets remains. Therefore, developing safe and effective delivery vehicles is critical for fundamental biomedical research and clinical applications. In this study, we designed an octopus-like self-releasing intracellular protein transporter, the LEB5, based on the heat-labile enterotoxin. This carrier comprises five identical units, each of which has three main components: a linker, a self-releasing enzyme sensitivity loop, and the LTB transport domain. The LEB5 comprises five purified monomers that self-assemble to create a pentamer with ganglioside GM1 binding capacity. The fluorescent protein EGFP was used as a reporter system to identify the LEB5 features. The high-purity fusion protein ELEB monomer was produced from modified bacteria carrying pET24a(+)-eleb recombinant plasmids. EGFP protein could effectively detach from LEB5 by low dosage trypsin, according to electrophoresis analysis. The transmission electron microscopy results indicate that both LEB5 and ELEB5 pentamers exhibit a relatively regularly spherical shape, and the differential scanning calorimetry measurements further suggest that these proteins possess excellent thermal stability. Fluorescence microscopy revealed that LEB5 translocated EGFP into different cell types. Flow cytometry showed cellular differences in the transport capacity of LEB5. According to the confocal microscopy, fluorescence analysis and western blotting data, EGFP was transferred to the endoplasmic reticulum by the LEB5 carrier, detached from LEB5 by cleavage of the enzyme-sensitive loop, and released into the cytoplasm. Within the dosage range of LEB5 10-80 µg/mL, cell counting kit-8 assay revealed no significant changes in cell viability. These results demonstrated that LEB5 is a safe and effective intracellular self-releasing delivery vehicle capable of transporting and releasing protein medicines into cells.


Bacterial Toxins , Escherichia coli Proteins , Octopodiformes , Animals , Octopodiformes/metabolism , Bacterial Toxins/chemistry , Enterotoxins/chemistry
4.
Molecules ; 28(3)2023 Jan 23.
Article En | MEDLINE | ID: mdl-36770798

Heat-stable enterotoxin (STa) produced by Enterotoxigenic E. coli is responsible for causing acute diarrhea in infants in developing countries. However, the chemical synthesis of STa peptides with the native conformation and the correct intra-molecular disulfide bonds is a major hurdle for vaccine development. To address this issue, we herein report on the design and preparation of STa analogs and a convenient chemical method for obtaining STa molecules with the correct conformation. To develop an STa vaccine, we focused on a structure in a type II ß-turn in the STa molecule and introduced a D-Lys residue as a conjugation site for carrier proteins. In addition, the -Glu-Leu- sequence in the STa molecule was replaced with a -Asp-Val- sequence to decrease the toxic activity of the peptide to make it more amenable for use in vaccinations. To solve several issues associated with the synthesis of STa, such as the formation of non-native disulfide isomers, the native disulfide pairings were regioselectively formed in a stepwise manner. A native form or topological isomer of the designed STa peptide, which possesses a right-handed or a left-handed spiral structure, respectively, were synthesized in high synthetic yields. The conformation of the synthetic STa peptide was also confirmed by CD and NMR spectroscopy. To further utilize the designed STa peptide, it was labeled with fluorescein for fluorescent detection, since recent studies have also focused on the use of STa for detecting cancer cells, such as Caco-2 and T84. The labeled STa peptide was able to specifically and efficiently detect 293T cells expressing the recombinant STa receptor (GC-C) protein and Caco-2 cells. The findings reported here provide an outline of the molecular basis for using STa for vaccine development and in the detection of cancer cells.


Bacterial Toxins , Enterotoxigenic Escherichia coli , Escherichia coli Proteins , Neoplasms , Humans , Enterotoxins/genetics , Enterotoxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/chemistry , Hot Temperature , Caco-2 Cells , Enterotoxigenic Escherichia coli/genetics , Enterotoxigenic Escherichia coli/metabolism , Peptides/metabolism , Vaccine Development , Disulfides , Guanylate Cyclase/metabolism
5.
Int J Mol Sci ; 23(17)2022 Sep 01.
Article En | MEDLINE | ID: mdl-36077344

C. novyi type A produces the alpha-toxin (TcnA) that belongs to the large clostridial glucosylating toxins (LCGTs) and is able to modify small GTPases by N-acetylglucosamination on conserved threonine residues. In contrast, other LCGTs including Clostridioides difficile toxin A and toxin B (TcdA; TcdB) modify small GTPases by mono-o-glucosylation. Both modifications inactivate the GTPases and cause strong effects on GTPase-dependent signal transduction pathways and the consequent reorganization of the actin cytoskeleton leading to cell rounding and finally cell death. However, the effect of TcnA on target cells is largely unexplored. Therefore, we performed a comprehensive screening approach of TcnA treated HEp-2 cells and analyzed their proteome and their phosphoproteome using LC-MS-based methods. With this data-dependent acquisition (DDA) approach, 5086 proteins and 9427 phosphosites could be identified and quantified. Of these, 35 proteins were found to be significantly altered after toxin treatment, and 1832 phosphosites were responsive to TcnA treatment. By analyzing the TcnA-induced proteomic effects of HEp-2 cells, 23 common signaling pathways were identified to be altered, including Actin Cytoskeleton Signaling, Epithelial Adherens Junction Signaling, and Signaling by Rho Family GTPases. All these pathways are also regulated after application of TcdA or TcdB of C. difficile. After TcnA treatment the regulation on phosphorylation level was much stronger compared to the proteome level, in terms of both strength of regulation and the number of regulated phosphosites. Interestingly, various signaling pathways such as Signaling by Rho Family GTPases or Integrin Signaling were activated on proteome level while being inhibited on phosphorylation level or vice versa as observed for the Role of BRCA1 in DNA Damage Response. ZIP kinase, as well as Calmodulin-dependent protein kinases IV & II, were observed as activated while Aurora-A kinase and CDK kinases tended to be inhibited in cells treated with TcnA based on their substrate regulation pattern.


Bacterial Toxins , Clostridioides difficile , Monomeric GTP-Binding Proteins , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Enterotoxins/chemistry , Glycosylation , Monomeric GTP-Binding Proteins/metabolism , Proteome/metabolism , Proteomics/methods , Type C Phospholipases/metabolism , rho GTP-Binding Proteins/metabolism
6.
Int J Mol Sci ; 23(14)2022 Jul 06.
Article En | MEDLINE | ID: mdl-35886856

Some prevention strategies, including vaccines and antibiotic alternatives, have been developed to reduce enterotoxigenic Escherichia coli proliferation in animal production. In this study, a wild-type strain of BE311 with a virulent heat-stable enterotoxin gene identical to E. coli K99 was isolated for its high potential for gene expression ability. The whole genome of E. coli BE311 was sequenced for gene analyses and editing. Subsequently, the fluorescent gene mCherry was successfully knocked into the genome of E. coli BE311 by CRISPR/Cas9. The E. coli BE311−mCherry strain was precisely quantified through the fluorescence intensity and red colony counting. The inflammatory factors in different intestinal tissues all increased significantly after an E. coli BE311−mCherry challenge in Sprague−Dawley rats (p < 0.05). The heat-stable enterotoxin gene of E. coli BE311 was knocked out, and an attenuated vaccine host E. coli BE311-STKO was constructed. Flow cytometry showed apoptotic cell numbers were lower following a challenge of IPEC-J2 cells with E. coli BE311-STKO than with E. coli BE311. Therefore, the E. coli BE311−mCherry and E. coli BE311-STKO strains that were successfully constructed based on the gene knock-in and knock-out technology could be used as ideal candidates in ETEC challenge models and for the development of attenuated vaccines.


Enterotoxigenic Escherichia coli , Escherichia coli Infections , Escherichia coli Proteins , Animals , CRISPR-Cas Systems/genetics , Enterotoxigenic Escherichia coli/genetics , Enterotoxins/chemistry , Enterotoxins/genetics , Escherichia coli Infections/genetics , Escherichia coli Proteins/metabolism , Gene Editing/methods , Optical Imaging/methods , Rats , Rats, Sprague-Dawley , Whole Genome Sequencing
7.
Int J Mol Sci ; 23(9)2022 Apr 19.
Article En | MEDLINE | ID: mdl-35562899

Rising incidences and mortalities have drawn attention to Clostridioides difficile infections (CDIs) in recent years. The main virulence factors of this bacterium are the exotoxins TcdA and TcdB, which glucosylate Rho-GTPases and thereby inhibit Rho/actin-mediated processes in cells. This results in cell rounding, gut barrier disruption and characteristic clinical symptoms. So far, treatment of CDIs is limited and mainly restricted to some antibiotics, often leading to a vicious circle of antibiotic-induced disease recurrence. Here, we demonstrate the protective effect of the human antimicrobial peptide α-defensin-6 against TcdA, TcdB and the combination of both toxins in vitro and in vivo and unravel the underlying molecular mechanism. The defensin prevented toxin-mediated glucosylation of Rho-GTPases in cells and protected human cells, model epithelial barriers as well as zebrafish embryos from toxic effects. In vitro analyses revealed direct binding to TcdB in an SPR approach and the rapid formation of TcdB/α-defensin-6 complexes, as analyzed with fluorescent TcdB by time-lapse microscopy. In conclusion, the results imply that α-defensin-6 rapidly sequesters the toxin into complexes, which prevents its cytotoxic activity. These findings extend the understanding of how human peptides neutralize bacterial protein toxins and might be a starting point for the development of novel therapeutic options against CDIs.


Bacterial Toxins , Clostridioides difficile , Clostridium Infections , alpha-Defensins , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridium Infections/microbiology , Enterotoxins/chemistry , Humans , Zebrafish/metabolism , alpha-Defensins/pharmacology , rho GTP-Binding Proteins/metabolism
8.
Glycobiology ; 32(5): 391-403, 2022 04 21.
Article En | MEDLINE | ID: mdl-34972864

The heat-labile enterotoxins of Escherichia coli and cholera toxin of Vibrio cholerae are related in structure and function. Each of these oligomeric toxins is comprised of one A polypeptide and five B polypeptides. The B-subunits bind to gangliosides, which are followed by uptake into the intoxicated cell and activation of the host's adenylate cyclase by the A-subunits. There are two antigenically distinct groups of these toxins. Group I includes cholera toxin and type I heat-labile enterotoxin of E. coli; group II contains the type II heat-labile enterotoxins of E. coli. Three variants of type II toxins, designated LT-IIa, LT-IIb and LT-IIc have been described. Earlier studies revealed the crystalline structure of LT-IIb. Herein the carbohydrate binding specificity of LT-IIc B-subunits was investigated by glycosphingolipid binding studies on thin-layer chromatograms and in microtiter wells. Binding studies using a large variety of glycosphingolipids showed that LT-IIc binds with high affinity to gangliosides with a terminal Neu5Acα3Gal or Neu5Gcα3Gal, e.g. the gangliosides GM3, GD1a and Neu5Acα3-/Neu5Gcα3--neolactotetraosylceramide and Neu5Acα3-/Neu5Gcα3-neolactohexaosylceramide. The crystal structure of LT-IIc B-subunits alone and with bound LSTd/sialyl-lacto-N-neotetraose d pentasaccharide uncovered the molecular basis of the ganglioside recognition. These studies revealed common and unique functional structures of the type II family of heat-labile enterotoxins.


Bacterial Toxins , Escherichia coli Proteins , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cholera Toxin/metabolism , Enterotoxins/chemistry , Enterotoxins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , G(M1) Ganglioside/metabolism , Gangliosides/metabolism , Hot Temperature
9.
Sci Rep ; 12(1): 34, 2022 01 07.
Article En | MEDLINE | ID: mdl-34997016

Cholera toxin (CT) and Escherichia coli heat-labile enterotoxin (LT) are structurally similar AB5-type protein toxins. They move from the cell surface to the endoplasmic reticulum where the A1 catalytic subunit is separated from its holotoxin by protein disulfide isomerase (PDI), thus allowing the dissociated A1 subunit to enter the cytosol for a toxic effect. Despite similar mechanisms of toxicity, CT is more potent than LT. The difference has been attributed to a more stable domain assembly for CT as compared to LT, but this explanation has not been directly tested and is arguable as toxin disassembly is an indispensable step in the cellular action of these toxins. We show here that PDI disassembles CT more efficiently than LT, which provides a possible explanation for the greater potency of the former toxin. Furthermore, direct examination of CT and LT domain assemblies found no difference in toxin stability. Using novel analytic geometry approaches, we provide a detailed characterization of the positioning of the A subunit with respect to the B pentamer and demonstrate significant differences in the interdomain architecture of CT and LT. Protein docking analysis further suggests that these global structural differences result in distinct modes of PDI-toxin interactions. Our results highlight previously overlooked structural differences between CT and LT that provide a new model for the PDI-assisted disassembly and differential potency of these toxins.


Cholera Toxin/chemistry , Cholera Toxin/metabolism , Enterotoxins/chemistry , Enterotoxins/metabolism , Glycosides/chemistry , Glycosides/metabolism , Protein Disulfide-Isomerases/metabolism , Triterpenes/chemistry , Triterpenes/metabolism , Catalytic Domain , Cholera Toxin/toxicity , Enterotoxins/toxicity , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Hot Temperature , Molecular Docking Simulation , Protein Disulfide-Isomerases/chemistry , Protein Stability
10.
EMBO Rep ; 23(1): e53597, 2022 01 05.
Article En | MEDLINE | ID: mdl-34817920

Clostridioides difficile infections have emerged as the leading cause of healthcare-associated infectious diarrhea. Disease symptoms are mainly caused by the virulence factors, TcdA and TcdB, which are large homologous multidomain proteins. Here, we report a 2.8 Å resolution cryo-EM structure of native TcdA, unveiling its conformation at neutral pH. The structure uncovers the dynamic movement of the CROPs domain which is induced in response to environmental acidification. Furthermore, the structure reveals detailed information about the interaction area between the CROPs domain and the tip of the delivery and receptor-binding domain, which likely serves to shield the C-terminal part of the hydrophobic pore-forming region from solvent exposure. Similarly, extensive interactions between the globular subdomain and the N-terminal part of the pore-forming region suggest that the globular subdomain shields the upper part of the pore-forming region from exposure to the surrounding solvent. Hence, the TcdA structure provides insights into the mechanism of preventing premature unfolding of the pore-forming region at neutral pH, as well as the pH-induced inter-domain dynamics.


Bacterial Toxins , Clostridioides difficile , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides , Enterotoxins/chemistry , Enterotoxins/metabolism
11.
ACS Chem Biol ; 17(1): 118-128, 2022 01 21.
Article En | MEDLINE | ID: mdl-34965093

Clostridium difficile infection is mediated by two major exotoxins: toxins A (TcdA) and B (TcdB). Inhibiting the biocatalytic activities of these toxins with targeted peptide-based drugs can reduce the risk of C. difficile infection. In this work, we used a computational strategy that integrates a peptide binding design (PepBD) algorithm and explicit-solvent atomistic molecular dynamics simulation to determine promising toxin A-targeting peptides that can recognize and bind to the catalytic site of the TcdA glucosyltransferase domain (GTD). Our simulation results revealed that two out of three in silico discovered peptides, viz. the neutralizing peptides A (NPA) and B (NPB), exhibit lower binding free energies when bound to the TcdA GTD than the phage-display discovered peptide, viz. the reference peptide (RP). These peptides may serve as potential inhibitors against C. difficile infection. The efficacy of the peptides RP, NPA, and NPB to neutralize the cytopathic effects of TcdA was tested in vitro in human jejunum cells. Both phage-display peptide RP and in silico peptide NPA were found to exhibit strong toxin-neutralizing properties, thereby preventing the TcdA toxicity. However, the in silico peptide NPB demonstrates a relatively low efficacy against TcdA.


Bacterial Toxins/antagonists & inhibitors , Computer Simulation , Enterotoxins/antagonists & inhibitors , Peptides/pharmacology , Bacterial Toxins/chemistry , Drug Design , Enterotoxins/chemistry , Models, Molecular , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Domains , Reproducibility of Results
12.
Front Immunol ; 12: 732938, 2021.
Article En | MEDLINE | ID: mdl-34887850

Staphylococcal enterotoxin B (SEB) simultaneously crosslinks MHC class II antigen and TCR, promoting proliferation of T cells and releasing a large number of toxic cytokines. In this report, we computationally examined the possibility of using a single-chain biparatopic bispecific antibody to target SEB and prevent TCR binding. The design was inspired by the observation that mixing two anti-SEB antibodies 14G8 and 6D3 can block SEB-TCR activation, and we used 14G8-6D3-SEB tertiary crystal structure as a template. Twelve simulation systems were constructed to systematically examine the effects of the designed bispecific scFV MB102a, including isolated SEB, MB102a with different linkers, MB102a-SEB complex, MB102a-SEB-TCRß complex, MB102a-SEB-TCR-MHC II complex, and MB102a-SEB-MHC II. Our all atom molecular dynamics simulations (total 18,900 ns) confirmed that the designed single-chain bispecific antibody may allosterically prevent SEB-TCRß chain binding and inhibit SEB-TCR-MHC II formation. Subsequent analysis indicated that the binding of scFV to SEB correlates with SEB-TCR binding site motion and weakens SEB-TCR interactions.


Antibodies, Bacterial/immunology , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Enterotoxins/immunology , Animals , Antibodies, Bacterial/chemistry , Antibodies, Bacterial/genetics , Antibodies, Bispecific/genetics , Binding Sites, Antibody , Computer Simulation , Crystallography, X-Ray , Drug Design , Enterotoxins/chemistry , Humans , Mice , Molecular Dynamics Simulation , Protein Conformation , Protein Engineering , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
13.
Int J Mol Sci ; 22(22)2021 Nov 13.
Article En | MEDLINE | ID: mdl-34830170

Claudin (CLDN) proteins are commonly expressed in cancers and targeted in novel therapeutic approaches. The C-terminal of Clostridium perfringens enterotoxin (C-CPE) efficiently binds several claudins. In this study, recombinant C-CPE conjugated to gold nanoparticles (AuNPs) has been used for prostate adenocarcinoma (PAC) and transitional cell carcinoma (TCC) cell killing in vitro using gold-nanoparticle-mediated laser perforation (GNOME-LP). A PAC and TCC cell lines, as well as red fluorescence variants, allowing deep tissue imaging, were used. CLDN-3, -4, and -7 expression was confirmed by qPCR and immunofluorescences. The binding of C-CPE-AuNPs complexes on the cell surface was examined by scanning electron microscopy (SEM). Further, transcriptome analysis was carried out to evaluate the effect of C-CPE binder on the biological response of treated cells. Directed C-CPE-AuNP binding verified the capability to target CLDN receptors. Transcriptome analysis showed that C-CPE binding may activate immune and inflammatory responses but does not directly affect cell survival. Cancer cells ablation was demonstrated using a combination of GNOME-LP and C-CPE-AuNPs treatment reducing tumor cell viability to less than 10% depending on cell line. The fluorescent cell lines and the verified proof of concept in vitro provide the basis for perspective xenograft studies in an animal model.


Adenocarcinoma , Carcinoma, Transitional Cell , Dog Diseases , Enterotoxins , Gold , Laser Therapy , Metal Nanoparticles , Prostatic Neoplasms , Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Adenocarcinoma/veterinary , Animals , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/therapy , Carcinoma, Transitional Cell/veterinary , Cell Line, Tumor , Clostridium perfringens/chemistry , Dog Diseases/metabolism , Dog Diseases/therapy , Dogs , Enterotoxins/chemistry , Enterotoxins/pharmacology , Gold/chemistry , Gold/pharmacology , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Prostatic Neoplasms/veterinary
14.
Nat Commun ; 12(1): 6285, 2021 11 01.
Article En | MEDLINE | ID: mdl-34725358

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


Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Clostridioides difficile/drug effects , Clostridioides difficile/enzymology , Clostridium Infections/microbiology , Enterotoxins/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Clostridioides difficile/chemistry , Clostridioides difficile/genetics , Enterotoxins/chemistry , Enterotoxins/metabolism , Humans , Kinetics
15.
PLoS One ; 16(10): e0257824, 2021.
Article En | MEDLINE | ID: mdl-34618824

Infectious gastrointestinal diseases are frequently caused by toxins secreted by pathogens which may impair physiological functions of the intestines, for instance by cholera toxin or by heat-labile enterotoxin. To obtain a functional model of the human intestinal epithelium for studying toxin-induced disease mechanisms, differentiated enterocyte-like Caco-2 cells were co-cultured with goblet cell-like HT29-MTX cells. These co-cultures formed a functional epithelial barrier, as characterized by a high electrical resistance and the presence of physiological intestinal properties such as glucose transport and chloride secretion which could be demonstrated electrophysiologically and by measuring protein expression. When the tissues were exposed to cholera toxin or heat-labile enterotoxin in the Ussing chamber, cholera toxin incubation resulted in an increase in short-circuit currents, indicating an increase in apical chloride secretion. This is in line with typical cholera toxin-induced secretory diarrhea in humans, while heat-labile enterotoxin only showed an increase in short-circuit-current in Caco-2 cells. This study characterizes for the first time the simultaneous measurement of physiological properties on a functional and structural level combined with the epithelial responses to bacterial toxins. In conclusion, using this model, physiological responses of the intestine to bacterial toxins can be investigated and characterized. Therefore, this model can serve as an alternative to the use of laboratory animals for characterizing pathophysiological mechanisms of enterotoxins at the intestinal level.


Bacterial Toxins/metabolism , Cholera Toxin/metabolism , Communicable Diseases/microbiology , Gastrointestinal Diseases/microbiology , Bacterial Toxins/chemistry , Caco-2 Cells , Chlorides/metabolism , Cholera Toxin/chemistry , Coculture Techniques , Communicable Diseases/pathology , Enterotoxins/chemistry , Enterotoxins/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gastrointestinal Diseases/pathology , Glucose/metabolism , HT29 Cells , Humans , Intestinal Mucosa/drug effects
16.
Toxins (Basel) ; 13(9)2021 08 29.
Article En | MEDLINE | ID: mdl-34564613

Staphylococcal enterotoxin A (SEA), which is a superantigen toxin protein, binds to cytokine receptor gp130. Gp130 activates intracellular signaling pathways, including the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway. The effects of SEA on the JAK/STAT signaling pathway in mouse spleen cells were examined. After treatment with SEA, mRNA expression levels of interferon gamma (IFN-γ) and suppressor of cytokine-signaling 1 (SOCS1) increased. SEA-induced IFN-γ and SOCS1 expression were decreased by treatment with (-)-epigallocatechin gallate (EGCG). The phosphorylated STAT3, Tyr705, increased significantly in a SEA concentration-dependent manner in mouse spleen cells. Although (-)-3″-Me-EGCG did not inhibit SEA-induced phosphorylated STAT3, EGCG and (-)-4″-Me-EGCG significantly inhibited SEA-induced phosphorylated STAT3. It was thought that the hydroxyl group at position 3 of the galloyl group in the EGCG was responsible for binding to SEA and suppressing SEA-induced phosphorylation of STAT3. Through protein thermal shift assay in vitro, the binding of the gp130 receptor to SEA and the phosphorylation of STAT3 were inhibited by the interaction between EGCG and SEA. As far as we know, this is the first report to document that EGCG inhibits the binding of the gp130 receptor to SEA and the associated phosphorylation of STAT3.


Catechin/analogs & derivatives , Catechin/metabolism , Enterotoxins/chemistry , Enterotoxins/metabolism , Enterotoxins/toxicity , Janus Kinases/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured/drug effects , Disease Models, Animal , Gene Expression Regulation, Bacterial , Humans , Mice , Signal Transduction/drug effects , Spleen/drug effects , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics
17.
Nihon Saikingaku Zasshi ; 76(2): 139-148, 2021.
Article Ja | MEDLINE | ID: mdl-34039783

Staphylococcus aureus food poisoning was shown by Dack et al. in 1930 to be caused by staphylococcal enterotoxin (SE) produced by S. aureus, rather than by the bacterial infection. However, the emetic mechanism of SE has remained unclear. In this study, we analyzed the emetic activity of SE in several emetic animal models and tried to elucidate the mechanism of emesis. We established a small primate, common marmoset, as a novel emetic model for SE. We also analyzed the immunofluorescence analysis of the gastrointestinal tract of the common marmoset and found that SE binds to submucosal mast cells in the gastrointestinal tract and SE induces degranulation of the mast cells. Furthermore, we showed that SE induces histamine releases, which is inhibited by mast cell stabilizer. In addition, treatment of common marmosets with either mast cell stabilizer or histamine H1 receptor antagonists suppressed the emetic response induced by SE. These results indicate that orally administered SE binds to submucosal mast cells in the gastrointestinal tract and causes degranulation, resulting in the release of histamine, which in turn causes emesis.


Enterotoxins/adverse effects , Histamine Release/drug effects , Vomiting/chemically induced , Vomiting/drug therapy , Amino Acid Sequence , Animals , Callithrix , Cell Degranulation/drug effects , Disease Models, Animal , Enterotoxins/chemistry , Enterotoxins/metabolism , Histamine Antagonists/pharmacology , Histamine Antagonists/therapeutic use , Intestinal Mucosa/cytology , Mast Cells/metabolism , Mast Cells/physiology , Protein Conformation , Shrews , Vomiting/metabolism
18.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article En | MEDLINE | ID: mdl-33876770

The bacterium Clostridium perfringens causes severe, sometimes lethal gastrointestinal disorders in humans, including enteritis and enterotoxemia. Type F strains produce an enterotoxin (CpE) that causes the third most common foodborne illness in the United States. CpE induces gut breakdown by disrupting barriers at cell-cell contacts called tight junctions (TJs), which are formed and maintained by claudins. Targeted binding of CpE to specific claudins, encoded by its C-terminal domain (cCpE), loosens TJ barriers to trigger molecular leaks between cells. Cytotoxicity results from claudin-bound CpE complexes forming pores in cell membranes. In mammalian tissues, ∼24 claudins govern TJ barriers-but the basis for CpE's selective targeting of claudins in the gut was undetermined. We report the structure of human claudin-4 in complex with cCpE, which reveals that enterotoxin targets a motif conserved in receptive claudins and how the motif imparts high-affinity CpE binding to these but not other subtypes. The structural basis of CpE targeting is supported by binding affinities, kinetics, and half-lives of claudin-enterotoxin complexes and by the cytotoxic effects of CpE on claudin-expressing cells. By correlating the binding residence times of claudin-CpE complexes we determined to claudin expression patterns in the gut, we uncover that the primary CpE receptors differ in mice and humans due to sequence changes in the target motif. These findings provide the molecular and structural element CpE employs for subtype-specific targeting of claudins during pathogenicity of C. perfringens in the gut and a framework for new strategies to treat CpE-based illnesses in domesticated mammals and humans.


Claudin-4/chemistry , Enterotoxins/chemistry , Tight Junctions/drug effects , Animals , Binding Sites , Claudin-4/metabolism , Clostridium perfringens , Enterotoxins/toxicity , Humans , Molecular Docking Simulation , Protein Binding , Sf9 Cells , Spodoptera , Tight Junctions/metabolism
19.
J Zhejiang Univ Sci B ; 22(4): 305-309, 2021 Apr 15.
Article En | MEDLINE | ID: mdl-33835764

Staphylococcal enterotoxin A (SEA) synthesized by Staphylococcus aureus is a foodborne and heat-stable toxin, which is a great threat to human health (Pexaraet al., 2010). Highly sensitive antibodies are a key factor in the immunological detection of SEA, which is one of the most effective ways to detect SEA because of its accuracy, agility, and efficiency (Nouri et al., 2018). In this study, we constructed a tetravalent anti-SEA antibody gene by linking the tetramerization domain of human p53 to the C-terminus of the anti-SEA single-chain variable fragment (scFv), which was then transformed into Escherichia coli BL21 (DE3) for the production of a SEA-specific tetravalent antibody. Successful expression of the tetravalent antibody was determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blot. An indirect non-competitive enzyme-linked immunosorbent assay (ELISA) revealed that the tetravalent antibody exhibited SEA-specific binding activity. A sandwich ELISA demonstrated that compared to the scFv monomer, the tetravalent antibody was more sensitive in detecting SEA. Molecular docking analysis revealed that the SEA interacted with the scFv mainly on the opposite side of the residue linked to p53. Thus, this study indicated that genetically engineered tetramerization is a potential way to improve the sensitivity of SEA-specific scFv.


Enterotoxins/analysis , Single-Chain Antibodies/immunology , Enterotoxins/chemistry , Enterotoxins/immunology , Enzyme-Linked Immunosorbent Assay , Genetic Engineering , Molecular Docking Simulation , Protein Multimerization , Single-Chain Antibodies/chemistry
20.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article En | MEDLINE | ID: mdl-33805767

Novel therapeutics are needed to treat pathologies associated with the Clostridioides difficile binary toxin (CDT), particularly when C. difficile infection (CDI) occurs in the elderly or in hospitalized patients having illnesses, in addition to CDI, such as cancer. While therapies are available to block toxicities associated with the large clostridial toxins (TcdA and TcdB) in this nosocomial disease, nothing is available yet to treat toxicities arising from strains of CDI having the binary toxin. Like other binary toxins, the active CDTa catalytic subunit of CDT is delivered into host cells together with an oligomeric assembly of CDTb subunits via host cell receptor-mediated endocytosis. Once CDT arrives in the host cell's cytoplasm, CDTa catalyzes the ADP-ribosylation of G-actin leading to degradation of the cytoskeleton and rapid cell death. Although a detailed molecular mechanism for CDT entry and host cell toxicity is not yet fully established, structural and functional resemblances to other binary toxins are described. Additionally, unique conformational assemblies of individual CDT components are highlighted herein to refine our mechanistic understanding of this deadly toxin as is needed to develop effective new therapeutic strategies for treating some of the most hypervirulent and lethal strains of CDT-containing strains of CDI.


Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Clostridioides difficile/pathogenicity , Cross Infection/drug therapy , Enterocolitis, Pseudomembranous/drug therapy , Enterotoxins/antagonists & inhibitors , ADP-Ribosylation/drug effects , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/deficiency , Actins/genetics , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Binding Sites , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Cross Infection/metabolism , Cross Infection/microbiology , Cross Infection/pathology , Endocytosis/drug effects , Enterocolitis, Pseudomembranous/metabolism , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Enterotoxins/chemistry , Enterotoxins/genetics , Enterotoxins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , Protein Structure, Secondary
...