Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.731
Filter
2.
Korean J Intern Med ; 39(4): 659-667, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38986495

ABSTRACT

BACKGROUND/AIMS: Sensitization to staphylococcal superantigens (SAgs) could contribute to asthma severity. However, its relevance with eosinophilic phenotype has not yet been clarified. This study aimed to investigate associations between serum specific IgE levels to SAg and eosinophilic airway inflammation in adult asthmatics. METHODS: The serum specific IgE levels to 3 SAgs, including staphylococcal enterotoxin A (SEA) and B (SEB), and toxic shock syndrome toxin-1 (TSST-1) were measured by ImmunoCAP in 230 adult asthmatic patients and 50 healthy controls (HCs). Clinical characteristics and laboratory parameters, including serum total/free IgE, and 2 eosinophil-activation markers, eosinophil cationic protein (ECP), and eosinophil-derived neurotoxin (EDN), were analyzed according to blood eosinophil counts (BEC; 150 cells/µL) and serum specific IgE levels to 3 SAgs (0.35 kU/L). RESULTS: Asthmatic patients showed higher serum specific IgE levels to 3 SAgs than HCs (p < 0.05 for all). The serum total/clinfree IgE levels were significantly higher in asthmatics with positive IgE responses to 3 SAgs than those without (p < 0.05 for all). There were no significant differences in clinical parameters including age, asthma severity, comorbidities, or smoking according to IgE responses to 3 SAgs. Patients with positive IgE responses to SEB (not to SEA/TSST-1) had higher serum specific IgE levels to house dust mites and ECP/EDN as well as higher BEC with positive correlations between serum SEB-specific IgE levels and BEC/ECP/EDN (p < 0.05 for all). CONCLUSION: These findings suggest that serum SEB-specific IgE levels could contribute to eosinophil activation as well as IgE production in adult asthma.


Subject(s)
Asthma , Enterotoxins , Eosinophils , Immunoglobulin E , Phenotype , Superantigens , Humans , Enterotoxins/immunology , Immunoglobulin E/blood , Male , Asthma/immunology , Asthma/blood , Asthma/diagnosis , Female , Middle Aged , Adult , Eosinophils/immunology , Case-Control Studies , Superantigens/immunology , Superantigens/blood , Biomarkers/blood , Aged , Eosinophilia/immunology , Eosinophilia/blood , Eosinophilia/diagnosis , Eosinophil Cationic Protein/blood , Bacterial Toxins/immunology , Bacterial Toxins/blood , Eosinophil-Derived Neurotoxin/blood
3.
mSphere ; 9(7): e0050524, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38990001

ABSTRACT

During surveillance of Staphylococcus aureus in lesions from patients with atopic dermatitis (AD), we isolated Staphylococcus argenteus, a species registered in 2011 as a new member of the genus Staphylococcus and previously considered a lineage of S. aureus. Genome sequence comparisons between S. argenteus isolates and representative S. aureus clinical isolates from various origins revealed that the S. argenteus genome from AD patients closely resembles that of S. aureus causing skin infections. We previously reported that 17%-22% of S. aureus isolated from skin infections produce staphylococcal enterotoxin Y (SEY), which predominantly induces T-cell proliferation via the T-cell receptor (TCR) Vα pathway. Complete genome sequencing of S. argenteus isolates revealed a gene encoding a protein similar to superantigen SEY, designated as SargEY, on its chromosome. Population structure analysis of S. argenteus revealed that these isolates are ST2250 lineage, which was the only lineage positive for the SEY-like gene among S. argenteus. Recombinant SargEY demonstrated immunological cross-reactivity with anti-SEY serum. SargEY could induce proliferation of human CD4+ and CD8+ T cells, as well as production of TNF-α and IFN-γ. SargEY showed emetic activity in a marmoset monkey model. SargEY and SET (a phylogenetically close but uncharacterized SE) revealed their dependency on TCR Vα in inducing human T-cell proliferation. Additionally, TCR sequencing revealed other previously undescribed Vα repertoires induced by SEH. SargEY and SEY may play roles in exacerbating the respective toxin-producing strains in AD. IMPORTANCE: Staphylococcus aureus is frequently isolated from active lesions of atopic dermatitis (AD) patients. We reported that 17%-22% of S. aureus isolated from AD patients produced a novel superantigen staphylococcal enterotoxin Y (SEY). Unlike many S. aureus superantigens that activate T cells via T-cell receptor (TCR) Vß, SEY activates T cells via TCR Vα and stimulates cytokine secretion. Staphylococcus argenteus was isolated from AD patients during the surveillance for S. aureus. Phylogenetic comparison of the genome indicated that the isolate was very similar to S. aureus causing skin infections. The isolate encoded a SEY-like protein, designated SargEY, which, like SEY, activated T cells via the TCR Vα. ST2250 is the only lineage positive for SargEY gene. ST2250 S. argenteus harboring a superantigen SargEY gene may be a novel staphylococcal clone that infects human skin and is involved in the exacerbation of AD.


Subject(s)
Dermatitis, Atopic , Enterotoxins , Genome, Bacterial , Staphylococcus , Superantigens , Humans , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/immunology , Superantigens/genetics , Superantigens/immunology , Staphylococcus/genetics , Staphylococcus/immunology , Staphylococcus/classification , Enterotoxins/genetics , Enterotoxins/immunology , Animals , Phylogeny , Genomics , Whole Genome Sequencing , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology
4.
Hum Vaccin Immunother ; 20(1): 2360338, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38857905

ABSTRACT

Staphylococcal Enterotoxin B (SEB), produced by Staphylococcus aureus (S. aureus), is a powerful superantigen that induces severe immune disruption and toxic shock syndrome (TSS) upon binding to MHC-II and TCR. Despite its significant impact on the pathogenesis of S. aureus, there are currently no specific therapeutic interventions available to counteract the mechanism of action exerted by this toxin. In this study, we have identified a human monoclonal antibody, named Hm0487, that specifically targets SEB by single-cell sequencing using PBMCs isolated from volunteers enrolled in a phase I clinical trial of the five-antigen S. aureus vaccine. X-ray crystallography studies revealed that Hm0487 exhibits high affinity for a linear B cell epitope in SEB (SEB138-147), which is located distantly from the site involved in the formation of the MHC-SEB-TCR ternary complex. Furthermore, in vitro studies demonstrated that Hm0487 significantly impacts the interaction of SEB with both receptors and the binding to immune cells, probably due to an allosteric effect on SEB rather than competing with receptors for binding sites. Moreover, both in vitro and in vivo studies validated that Hm0487 displayed efficient neutralizing efficacy in models of lethal shock and sepsis induced by either SEB or bacterial challenge. Our findings unveil an alternative mechanism for neutralizing the pathogenesis of SEB by Hm0487, and this antibody provides a novel strategy for mitigating both SEB-induced toxicity and S. aureus infection.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Enterotoxins , Enterotoxins/immunology , Enterotoxins/antagonists & inhibitors , Humans , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Animals , Crystallography, X-Ray , Staphylococcus aureus/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Epitopes, B-Lymphocyte/immunology , Mice , Shock, Septic/immunology , Shock, Septic/prevention & control , Female , Leukocytes, Mononuclear/immunology , Staphylococcal Vaccines/immunology , Antibodies, Bacterial/immunology , Superantigens/immunology
5.
Biosens Bioelectron ; 261: 116458, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38852321

ABSTRACT

Herein, a colorimetric-fluorescent hybrid bifunctional nanobead with Janus structure (J-cf-HBN) was synthesized via one-pot microemulsification. Oleylamine-coated AuNPs and aggregation-induced emission luminogens (AIEgens) were suggested as building blocks to obtain high-performance colorimetric-fluorescent signals. The as-prepared J-cf-HBNs were used as a signal amplification probe to construct an immunochromatographic assay (J-cf-HBNs-ICA) platform for the ultrasensitive detection of staphylococcal enterotoxin B (SEB) in milk samples. Owing to the rational spatial distribution of AuNPs and AIEgens, the J-cf-HBNs present a highly retained photoluminescence and enhanced colorimetric signals. Combined with a pair of highly affinitive anti-SEB antibodies, the J-cf-HBN-ICA platform enabled the fast naked-eye visualization and fluorescent quantitative detection of SEB in various milk matrices. Given the advantages of the dual-mode high-performance J-cf-HBNs, the proposed strip achieved a high sensitivity for SEB qualitative determination with a visual limit of detection (LOD) of 1.56 ng mL-1 and exhibited ultrasensitivity for SEB quantitative detection with a LOD of 0.09 ng mL-1, which is 139-fold lower than that of ELISA using same antibodies. In conclusion, this work provides new insights into the construction of multimode immunochromatographic methods for food safety detection in the field.


Subject(s)
Biosensing Techniques , Colorimetry , Enterotoxins , Gold , Limit of Detection , Metal Nanoparticles , Milk , Milk/chemistry , Enterotoxins/analysis , Enterotoxins/immunology , Enterotoxins/isolation & purification , Animals , Biosensing Techniques/methods , Colorimetry/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Fluorescent Dyes/chemistry , Food Contamination/analysis , Chromatography, Affinity/methods , Immunoassay/methods
6.
Commun Biol ; 7(1): 733, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886509

ABSTRACT

Claudins are a 27-member family of ~25 kDa membrane proteins that integrate into tight junctions to form molecular barriers at the paracellular spaces between endothelial and epithelial cells. As the backbone of tight junction structure and function, claudins are attractive targets for modulating tissue permeability to deliver drugs or treat disease. However, structures of claudins are limited due to their small sizes and physicochemical properties-these traits also make therapy development a challenge. Here we report the development of a synthetic antibody fragment (sFab) that binds human claudin-4 and the determination of a high-resolution structure of it bound to claudin-4/enterotoxin complexes using cryogenic electron microscopy. Structural and biophysical results reveal this sFabs mechanism of select binding to human claudin-4 over other homologous claudins and establish the ability of sFabs to bind hard-to-target claudins to probe tight junction structure and function. The findings provide a framework for tight junction modulation by sFabs for tissue-selective therapies.


Subject(s)
Claudin-4 , Claudin-4/metabolism , Humans , Tight Junctions/metabolism , Cryoelectron Microscopy , Enterotoxins/metabolism , Enterotoxins/chemistry , Enterotoxins/immunology , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/metabolism , Protein Binding , Models, Molecular
7.
Int Immunopharmacol ; 132: 111943, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38581989

ABSTRACT

BACKGROUND: Staphylococcal enterotoxin C2 (SEC2) is used as an immunotherapeutic drug in China. However, SEC2 are limited due to its immunosuppressive and toxic effects. A SEC2 2M-118 (H118A/T20L/G22E) mutant generated by site-directed mutagenesis was studied to elucidate the underlying antitumor mechanism. METHODS: The effects of 2M-118 on mouse fibrosarcoma (Meth-A) cells and cytokine responses were tested in vitro using a transwell assay and ELISA, respectively. 2M-118 effect on immune function in tumor-bearing mice was tested. Cytokine levels and antitumor responses were measured using ELISA and flow cytometry, respectively. TUNEL staining and immunohistochemistry were employed to detect the tumor apoptosis and CD4+ and CD8+ tumor infiltrating lymphocytes (TILs) in tumor tissue. RESULTS: 2M-118 demonstrated the growth inhibition on tumor cells, increase of cytokines production (IL-2, IFN-γ, and TNF-α) and splenocyte proliferation in vitro. 2M-118 effectively inhibited tumor development and increased lymphocytes and cytokines in a tumor-bearing mouse model. Additionally, 2M-118 regulated the tumormicroenvironment by reducing the number of myeloid-derived suppressor cells (MDSCs), increasing the number of TILs, and inducing tumorcell apoptosis. CONCLUSION: 2M-118 promotes immune function and enhances antitumor response. This indicates that 2M-118 could potentially be developed as a novel anti-tumor drug with-highefficiencyandlowtoxicity.


Subject(s)
Cytokines , Enterotoxins , Animals , Enterotoxins/immunology , Cell Line, Tumor , Mice , Cytokines/metabolism , Mice, Inbred BALB C , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Fibrosarcoma/drug therapy , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Apoptosis/drug effects , Immunity, Cellular/drug effects , Female , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Mutation , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects
9.
Front Immunol ; 13: 827048, 2022.
Article in English | MEDLINE | ID: mdl-35237272

ABSTRACT

Previous studies on immune responses following COVID-19 vaccination in patients with common variable immunodeficiency (CVID) were inconclusive with respect to the ability of the patients to produce vaccine-specific IgG antibodies, while patients with milder forms of primary antibody deficiency such as immunoglobulin isotype deficiency or selective antibody deficiency have not been studied at all. In this study we examined antigen-specific activation of CXCR5-positive and CXCR5-negative CD4+ memory cells and also isotype-specific and functional antibody responses in patients with CVID as compared to other milder forms of primary antibody deficiency and healthy controls six weeks after the second dose of BNT162b2 vaccine against SARS-CoV-2. Expression of the activation markers CD25 and CD134 was examined by multi-color flow cytometry on CD4+ T cell subsets stimulated with SARS-CoV-2 spike peptides, while in parallel IgG and IgA antibodies and surrogate virus neutralization antibodies against SARS-CoV-2 spike protein were measured by ELISA. The results show that in CVID and patients with other milder forms of antibody deficiency normal IgG responses (titers of spike protein-specific IgG three times the detection limit or more) were associated with intact vaccine-specific activation of CXCR5-negative CD4+ memory T cells, despite defective activation of circulating T follicular helper cells. In contrast, CVID IgG nonresponders showed defective vaccine-specific and superantigen-induced activation of both CD4+T cell subsets. In conclusion, impaired TCR-mediated activation of CXCR5-negative CD4+ memory T cells following stimulation with vaccine antigen or superantigen identifies patients with primary antibody deficiency and impaired IgG responses after BNT162b2 vaccination.


Subject(s)
BNT162 Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , COVID-19/immunology , Primary Immunodeficiency Diseases/immunology , SARS-CoV-2/immunology , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/prevention & control , Common Variable Immunodeficiency/immunology , Enterotoxins/immunology , Female , Humans , Immunoglobulin G/immunology , Lymphocyte Activation , Male , Memory T Cells/immunology , Middle Aged , Receptors, Antigen, T-Cell/immunology , Receptors, CXCR5/immunology , T Follicular Helper Cells/immunology , Vaccination
10.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35165181

ABSTRACT

Staphylococcus aureus is a foremost bacterial pathogen responsible for a vast array of human diseases. Staphylococcal superantigens (SAgs) constitute a family of exotoxins from S. aureus that bind directly to major histocompatibility complex (MHC) class II and T cell receptors to drive extensive T cell activation and cytokine release. Although these toxins have been implicated in serious disease, including toxic shock syndrome, the specific pathological mechanisms remain unclear. Herein, we aimed to elucidate how SAgs contribute to pathogenesis during bloodstream infections and utilized transgenic mice encoding human MHC class II to render mice susceptible to SAg activity. We demonstrate that SAgs contribute to S. aureus bacteremia by massively increasing bacterial burden in the liver, and this was mediated by CD4+ T cells that produced interferon gamma (IFN-γ) to high levels in a SAg-dependent manner. Bacterial burdens were reduced by blocking IFN-γ, phenocopying SAg-deletion mutant strains, and inhibiting a proinflammatory response. Infection kinetics and flow cytometry analyses suggested that this was a macrophage-driven mechanism, which was confirmed through macrophage-depletion experiments. Experiments in human cells demonstrated that excessive IFN-γ allowed S. aureus to replicate efficiently within macrophages. This indicates that SAgs promote bacterial survival by manipulating the immune response to inhibit effective clearing of S. aureus Altogether, this work implicates SAg toxins as critical therapeutic targets for preventing persistent or severe S. aureus disease.


Subject(s)
Interferon-gamma/immunology , Staphylococcal Infections/immunology , Superantigens/immunology , Animals , Bacteremia , Enterotoxins/immunology , Exotoxins/immunology , Histocompatibility Antigens Class II/immunology , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Staphylococcus aureus/pathogenicity , T-Lymphocytes/immunology , Virulence Factors/immunology
11.
J Vet Sci ; 23(1): e7, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34841745

ABSTRACT

BACKGROUND: Enterotoxigenic Escherichia coli (ETEC) infection is a primary cause of livestock diarrhea. Therefore, effective vaccines are needed to reduce the incidence of ETEC infection. OBJECTIVES: Our study aimed to develop a multivalent ETEC vaccine targeting major virulence factors of ETEC, including enterotoxins and fimbriae. METHODS: SLS (STa-LTB-STb) recombinant enterotoxin and fimbriae proteins (F4, F5, F6, F18, and F41) were prepared to develop a multivalent vaccine. A total of 65 mice were immunized subcutaneously by vaccines and phosphate-buffered saline (PBS). The levels of specific immunoglobulin G (IgG) and pro-inflammatory cytokines were determined at 0, 7, 14 and 21 days post-vaccination (dpv). A challenge test with a lethal dose of ETEC was performed, and the survival rate of the mice in each group was recorded. Feces and intestine washes were collected to measure the concentrations of secretory immunoglobulin A (sIgA). RESULTS: Anti-SLS and anti-fimbriae-specific IgG in serums of antigen-vaccinated mice were significantly higher than those of the control group. Immunization with the SLS enterotoxin and multivalent vaccine increased interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) concentrations. Compared to diarrheal symptoms and 100% death of mice in the control group, mice inoculated with the multivalent vaccine showed an 80% survival rate without any symptom of diarrhea, while SLS and fimbriae vaccinated groups showed 60 and 70% survival rates, respectively. CONCLUSIONS: Both SLS and fimbriae proteins can serve as vaccine antigens, and the combination of these two antigens can elicit stronger immune responses. The results suggest that the multivalent vaccine can be successfully used for preventing ETEC in important livestock.


Subject(s)
Diarrhea , Escherichia coli Infections , Escherichia coli Vaccines/immunology , Animals , Antibodies, Bacterial/blood , Diarrhea/prevention & control , Diarrhea/veterinary , Disease Models, Animal , Enterotoxigenic Escherichia coli , Enterotoxins/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/veterinary , Escherichia coli Proteins/immunology , Fimbriae Proteins/immunology , Immunoglobulin A/analysis , Immunoglobulin G/blood , Mice , Vaccines, Combined/immunology
12.
Front Immunol ; 12: 732938, 2021.
Article in English | MEDLINE | ID: mdl-34887850

ABSTRACT

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.


Subject(s)
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.
Front Immunol ; 12: 723689, 2021.
Article in English | MEDLINE | ID: mdl-34489975

ABSTRACT

The inflammatory activity of staphylococcal enterotoxin B (SEB) relies on its capacity to trigger polyclonal T-cell activation by binding both T-cell receptor (TCR) and costimulatory receptor CD28 on T cells and MHC class II and B7 molecules on antigen presenting cells (APC). Previous studies highlighted that SEB may bind TCR and CD28 molecules independently of MHC class II, yet the relative contribution of these interactions to the pro-inflammatory function of SEB remained unclear. Here, we show that binding to MHC class II is dispensable for the inflammatory activity of SEB, whereas binding to TCR, CD28 and B7 molecules is pivotal, in both human primary T cells and Jurkat T cell lines. In particular, our finding is that binding of SEB to B7 molecules suffices to trigger both TCR- and CD28-mediated inflammatory signalling. We also provide evidence that, by strengthening the interaction between CD28 and B7, SEB favours the recruitment of the TCR into the immunological synapse, thus inducing lethal inflammatory signalling.


Subject(s)
CD28 Antigens/immunology , Enterotoxins/immunology , Histocompatibility Antigens Class II/metabolism , Receptors, Antigen, T-Cell/metabolism , Antigen-Presenting Cells/immunology , Cell Communication , Cells, Cultured , Humans , Lymphocyte Activation , Signal Transduction , T-Lymphocytes/immunology
14.
mSphere ; 6(4): e0060821, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34319127

ABSTRACT

Atopic dermatitis (AD) is a condition affecting 30 million persons in the United States. AD patients are heavily infected with Staphylococcus aureus on the skin. A particularly severe form of AD is eczema herpeticum (ADEH), where the patients' AD is complicated by S. aureus and herpes simplex virus (HSV) infection. This study examined the S. aureus strains from 15 ADEH patients, provided blinded, and showed a high association of ADEH with strains that produce toxic shock syndrome toxin-1 (TSST-1; 73%) compared to 10% production by typical AD isolates from patients without EH and those from another unrelated condition, cystic fibrosis. The ADEH isolates produced the superantigens associated with TSS (TSST-1 and staphylococcal enterotoxins A, B, and C). This association may in part explain the potential severity of ADEH. We also examined the effect of TSST-1 and HSV-1 on human epithelial cells and keratinocytes. TSST-1 used CD40 as its receptor on epithelial cells, and HSV-1 either directly or indirectly interacted with CD40. The consequence of these interactions was chemokine production, which is capable of causing harmful inflammation, with epidermal/keratinocyte barrier disruption. Human epithelial cells treated first with TSST-1 and then HSV-1 resulted in enhanced chemokine production. Finally, we showed that TSST-1 modestly increased HSV-1 replication but did not increase viral plaque size. Our data suggest that ADEH is associated with production of the major TSS-associated superantigens, together with HSV reactivation. The superantigens plus HSV may damage the skin barrier by causing harmful inflammation, thereby leading to increased symptoms. IMPORTANCE Atopic dermatitis (eczema, AD) with concurrent herpes simplex virus infection (eczema herpeticum, ADEH) is a severe form of AD. We show that ADEH patients are colonized with Staphylococcus aureus that primarily produces the superantigen toxic shock syndrome toxin-1 (TSST-1); however, significantly but to a lesser extent the superantigens staphylococcal enterotoxins A, B, and C are also represented in ADEH. Our studies showed that TSST-1 uses the immune costimulatory molecule CD40 as its epithelial cell receptor. Herpes simplex virus (HSV) also interacted directly or indirectly with CD40 on epithelial cells. Treatment of epithelial cells with TSST-1 and then HSV-1 resulted in enhanced chemokine production. We propose that this combination of exposures (TSST-1 and then HSV) leads to opening of epithelial and skin barriers to facilitate potentially serious ADEH.


Subject(s)
Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Enterotoxins/genetics , Enterotoxins/metabolism , Herpesvirus 1, Human/metabolism , Kaposi Varicelliform Eruption/microbiology , Staphylococcus aureus/pathogenicity , Superantigens/genetics , Superantigens/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/pharmacology , CD40 Antigens/immunology , Chemokines/immunology , Enterotoxins/immunology , Enterotoxins/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/virology , HaCaT Cells , Herpesvirus 1, Human/immunology , Humans , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/virology , Staphylococcus aureus/metabolism , Superantigens/immunology , Superantigens/pharmacology
15.
Infect Immun ; 89(10): e0027421, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34097471

ABSTRACT

The pathology associated with Clostridioides difficile disease is caused in large part by TcdB, an intracellular bacterial toxin that inactivates small GTPases. Despite C. difficile causing enteric disease, antitoxin IgG is a clear correlate of protection against infection-associated pathology. Immunization with TcdB-based immunogens or passive transfer of monoclonal antibodies specific for the TcdB carboxy-terminal domain (CTD) confers protection following C. difficile infection. Whether the mechanism by which circulating IgG is delivered to the gut depends on specific receptor-mediated transport or is solely reflective of infection-induced damage to the gut remains unclear. Here, we tested the hypothesis that neonatal Fc receptor (FcRn) is required for the delivery of systemic TcdB-specific IgG to the gut and protection against C. difficile-associated pathology. FcRn-expressing mice and FcRn-deficient littermates were immunized subcutaneously with Alhydrogel adjuvant-adsorbed CTD before challenge with live C. difficile spores. FcRn was required for the delivery of systemic TcdB-specific IgG to the gut and for vaccine-induced protection against C. difficile-associated disease. The lack of FcRn expression had minimal effects on the composition of the gut microbiome and did not affect susceptibility to C. difficile infection in nonimmunized mice. In further experiments, intraperitoneal injection of immune sera in FcRn-deficient mice led to the transport of protective IgG to the gut independently of infection, confirming a reported method of bypassing the FcRn. Our results reveal an FcRn-dependent mechanism by which systemic immunization-induced IgG protects the gut during enteric C. difficile infection. These findings may be beneficial for the targeting of C. difficile-specific IgG to the gut.


Subject(s)
Clostridioides difficile/immunology , Clostridium Infections/immunology , Digestive System/immunology , Digestive System/microbiology , Disease Susceptibility/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Receptors, Fc/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antitoxins/immunology , Bacterial Toxins/immunology , Clostridium Infections/microbiology , Disease Susceptibility/microbiology , Enterotoxins/immunology , Female , Immunity/immunology , Immunization/methods , Male , Mice , Mice, Inbred C57BL , Vaccination/methods
16.
Front Immunol ; 12: 649235, 2021.
Article in English | MEDLINE | ID: mdl-34017331

ABSTRACT

Staphylococcus aureus (S. aureus) is an important zoonotic food-borne pathogen causing severe invasive infections, such as sepsis, pneumonia, food poisoning, toxic shock syndrome and autoimmune diseases. Staphylococcal enterotoxin O (SEO) is a new type of enterotoxins of S. aureus with superantigenic and emetic activity. However, it is still unclear about SEO-induced host inflammatory response. Therefore, the mechanism of SEO-induced interleukin-1ß (IL-1ß) secretion in mouse neutrophils was investigated in this study. Our results showed that recombinant SEO had superantigenic activity with high level of gamma interferon (IFN-γ) production in mouse spleen cells and induced inflammatory cytokines expression including IL-1α, IL-1ß, IL-6 and TNF-α in neutrophils under the action of ATP. In addition, SEO-induced IL-1ß secretion was dependent on activation of Toll like receptor 4 (TLR4), nuclear factor kappa B (NF-κB) and c-jun N-terminal kinase (JNK) signaling pathways. However, SEO-induced IL-1ß secretion was abolished in the neutrophils of NLRP3-/- mice compared with those of wild type mice, indicating that activation of NLRP3 inflammasome mediated IL-1ß secretion during neutrophils stimulation with SEO under the action of ATP. Moreover, this process of SEO+ATP-induced IL-1ß secretion was dependent on potassium (K+) efflux. Taken together, our study suggests that activation of TLR4/JNK/NLRP3 inflammasome signaling pathway mediate maturation and secretion of IL-1ß and provides a new insight on S. aureus virulence factor-induced host immune response.


Subject(s)
Adenosine Triphosphate/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/immunology , Staphylococcal Infections/immunology , Animals , Disease Models, Animal , Enterotoxins/immunology , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neutrophils/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Virulence Factors/immunology
17.
Clin Exp Immunol ; 205(3): 306-315, 2021 09.
Article in English | MEDLINE | ID: mdl-33999404

ABSTRACT

Typical murine models of allergic inflammation are induced by the combination of ovalbumin and aluminum hydroxide. However, accumulating evidence indicates that, in models of asthma and atopic dermatitis, allergic inflammation can be generated in the absence of aluminum hydroxide. Moreover, co-administration of Staphylococcus aureus enterotoxin B with ovalbumin can enhance inflammation. The objective of this study was to establish a rapid and mast cell-dependent murine model of allergic inflammation by inducing allergic peritonitis using ovalbumin and S. aureus enterotoxin B. Allergic peritonitis was induced in C57BL/6 mice by subcutaneous sensitization and intraperitoneal challenge with ovalbumin and S. aureus enterotoxin B. Disease characteristics were assessed by flow cytometry, enzyme-linked immunosorbent assay (ELISA), trypan blue exclusion and colorimetric assays. The time-course of the allergic peritonitis revealed a peak of peritoneal inflammation 48 h after challenge, as assessed by total cells and eosinophil counts. The decrease of cell numbers started 96 h post-challenge, with complete clearance within 168 h. Moreover, significantly higher levels of tryptase and increased vascular permeability were found 30 min following challenge. Allergic inflammation induction by ovalbumin and S. aureus enterotoxin B was impaired in mast cell-deficient mice and partially restored by mice reconstitution with bone marrow-derived mast cells, indicating the mast cell role in this model. We present a novel model of allergic peritonitis that is mast cell-dependent, simple and robust. Moreover, the use of S. aureus enterotoxin B better resembles human allergic inflammation, which is known to be characterized by the colonization of S. aureus.


Subject(s)
Asthma/immunology , Dermatitis, Atopic/immunology , Mast Cells/immunology , Peritonitis/immunology , Animals , Disease Models, Animal , Enterotoxins/immunology , Female , Immunization/methods , Immunoglobulin E/blood , Inflammation/immunology , Inflammation/pathology , Male , Mast Cells/transplantation , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Peritonitis/pathology , Staphylococcus aureus/metabolism
18.
Biochem Biophys Res Commun ; 558: 168-174, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33932776

ABSTRACT

Staphylococcal enterotoxins are one of the most important causative agents of food poisoning. These molecules function as both gastrointestinal toxins and superantigens (SAgs) which can simultaneously bind MHC-II and T cell receptor leading to a non-specific polyclonal T cell activation and massive proinflammatory cytokine release. Common symptoms include vomiting and diarrhea; however, in more severe cases, systemic dissemination may result in toxic shock syndrome and can be lethal in a few hours. Only small amounts of these heat-stable toxins are needed to cause the disease. Therefore, it is highly important to detect quickly low concentrations of SAgs in biological samples. In this work, we report a surface plasmon resonance (SPR)-based capture immunoassay for the detection of the SAg SEG. We analyzed the use of different amplification strategies. The SPR-based double-antibody sandwich approach could detect picomolar levels of SEG. The use of antibody-coated silica nanoparticles (AbSiNPs) as an alternative enhancing reagent also detected SEG in the picomolar range. Although AbSiNPs did not improve the limit of detection, for the same amount of SAg tested, AbSiNPs gave a higher response level than free antibodies. This work highlights the suitability of silica nanoparticles for signal amplification in SPR-based biosensors. Overall, SPR biosensors offer the capability for continuous real-time monitoring and high sensitivity that can be befitting for the detection of enterotoxins in food industries, laboratories and regulatory agencies.


Subject(s)
Enterotoxins/analysis , Immunoassay/methods , Superantigens/analysis , Surface Plasmon Resonance/methods , Amino Acid Sequence , Animals , Antibodies, Bacterial , Biosensing Techniques/methods , Coated Materials, Biocompatible , Enterotoxins/genetics , Enterotoxins/immunology , Food Microbiology , Humans , Limit of Detection , Nanoparticles , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Silicon Dioxide , Staphylococcal Food Poisoning/diagnosis , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Superantigens/genetics , Superantigens/immunology
19.
Front Immunol ; 12: 654872, 2021.
Article in English | MEDLINE | ID: mdl-34054818

ABSTRACT

Enhancement of mucosal immune responses in children and infants using novel adjuvants such as double mutant heat labile toxin (dmLT) is an important goal in the enteric vaccine field. dmLT has been shown to enhance mucosal IgA responses to the oral inactivated enterotoxigenic Escherichia coli (ETEC) vaccine ETVAX. dmLT can enhance IL-17A production from adult T cells, which may increase the production and secretion of mucosal IgA antibodies. However, the adjuvant mechanism remains to be fully elucidated and might differ between infants and adults due to age-related differences in the development of the immune system. The main objective of this study was to determine how dmLT influences antigen presenting cells and T cells from infants compared to adults, and the role of IL-1ß for mediating the adjuvant activity. Peripheral blood mononuclear cells (PBMCs) from Bangladeshi infants (6-11 months) and adults (18-40 years) were stimulated with the mitogen phytohaemagglutinin (PHA), the superantigen Staphylococcal enterotoxin B (SEB), ETVAX whole cell component (WCC) or E. coli lipopolysaccharide (LPS) ± dmLT, and cytokine production was measured using ELISA and electrochemiluminescence assays. The adjuvant dmLT significantly enhanced SEB- and PHA-induced IL-17A, but not IFN-γ responses, in PBMCs from both infants and adults. Blocking experiments using an IL-1 receptor antagonist demonstrated the importance of IL-1 signaling for the adjuvant effect. dmLT, ETVAX WCC and LPS induced dose-dependent IL-1ß responses of comparable magnitudes in infant and adult cells. Depletion experiments suggested that IL-1ß was mainly produced by monocytes. dmLT enhanced IL-1ß responses to low doses of WCC and LPS, and the adjuvant effect appeared over a wider dose-range of WCC in infants. dmLT and WCC also induced IL-6, IL-23 and IL-12p70 production in both age groups and dmLT tended to particularly enhance IL-23 responses to WCC. Our results show that dmLT can induce IL-1ß as well as other cytokines, which in turn may enhance IL-17A and potentially modulate other immunological responses in both infants and adults. Thus, dmLT may have an important function in promoting immune responses to the ETVAX vaccine, as well as other whole cell- or LPS-based vaccines in infants in low- and middle-income countries.


Subject(s)
Adjuvants, Immunologic , Antigens, Bacterial/immunology , Cytokines/biosynthesis , Lipopolysaccharides/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccines, Inactivated/immunology , Adult , Age Factors , Bacterial Toxins/immunology , Enterotoxins/immunology , Escherichia coli Proteins/immunology , Humans , Infant , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation , Monocytes/immunology , Monocytes/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL