Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 228
Filter
1.
Angew Chem Int Ed Engl ; 63(21): e202317552, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38497459

ABSTRACT

Celiac disease (CeD) is an autoimmune disorder triggered by gluten proteins, affecting approximately 1 % of the global population. The 33-mer deamidated gliadin peptide (DGP) is a metabolically modified wheat-gluten superantigen for CeD. Here, we demonstrate that the 33-mer DGP spontaneously assembles into oligomers with a diameter of approximately 24 nm. The 33-mer DGP oligomers present two main secondary structural motifs-a major polyproline II helix and a minor ß-sheet structure. Importantly, in the presence of 33-mer DGP oligomers, there is a statistically significant increase in the permeability in the gut epithelial cell model Caco-2, accompanied by the redistribution of zonula occludens-1, a master tight junction protein. These findings provide novel molecular and supramolecular insights into the impact of 33-mer DGP in CeD and highlight the relevance of gliadin peptide oligomerization.


Subject(s)
Celiac Disease , Enterocytes , Gliadin , Humans , Celiac Disease/metabolism , Celiac Disease/pathology , Caco-2 Cells , Gliadin/chemistry , Gliadin/metabolism , Enterocytes/metabolism , Superantigens/chemistry , Superantigens/metabolism , Permeability
2.
Biochemistry ; 61(7): 616-624, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35285627

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen producing many immune evasion molecules targeting various components of the host immune defense, including the Staphylococcal superantigen-like protein (SSL 1-14) family. Despite sharing similar structures with the powerful superantigens (SAgs), which cause massive T cell activation, SSLs interfere with a wide range of innate immune defenses. SSLs are divided into two subgroups, SSLs that contain a conserved carbohydrate Sialyl Lewis X [Neu5Acα2-3Galß1-4(Fucα1-3) GlcNAcß, SLeX] binding site and SSLs that lack the SLeX binding site. SSL2-6 and SSL11 possess the SLeX binding site. Our previous studies showed that SSL11 arrests cell motility by inducing cell adhesion in differentiated HL60 (dHL60) cells, while SSL7 did not bind dHL60 cells. SSL7-based chimeras were engineered by exchanging the SSL7 sequence with the corresponding SSL11 sequence and assaying for a gain of SSL11 function, namely, the induction of cell spreading and motility arrest. In addition to the SLeX-binding site, we observed that three beta-strands ß6, ß7, and ß9 and the N-terminal residues, Y16 and Y17, transitioned SSL7 to gain SSL11 activities. These studies define the structure-function properties of SSL11 that may allow SSL11 to inhibit S. aureus clearance by the host innate immune system, allowing S. aureus to maintain a carrier state in humans, an understudied aspect of S. aureus pathogenesis.


Subject(s)
Staphylococcal Infections , Superantigens , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Humans , Neutrophils , Protein Binding , Staphylococcus aureus/metabolism , Superantigens/chemistry , Superantigens/metabolism
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34548394

ABSTRACT

Microorganisms have coevolved diverse mechanisms to impair host defenses. A major one, superantigens, can result in devastating effects on the immune system. While all known superantigens induce vast immune cell proliferation and come from opportunistic pathogens, recently, proteins with similar broad specificity to antibody variable (V) domain families were identified in a commensal microbiota. These proteins, identified in the human commensal Ruminococcus gnavus, are called immunoglobulin-binding protein (Ibp) A and B and have been shown to activate B cells in vitro expressing either human VH3 or murine VH5/6/7. Here, we provide molecular and functional studies revealing the basis of this Ibp/immunoglobulin (Ig) interaction. The crystal structure and biochemical assays of a truncated IbpA construct in complex with mouse VH5 antigen-binding fragment (Fab) shows a binding of Ig heavy chain framework residues to the Ibp Domain D and the C-terminal heavy chain binding domain (HCBD). We used targeted mutagenesis of contact residues and affinity measurements and performed studies of the Fab-IbpA complex to determine the stoichiometry between Ibp and VH domains, suggesting Ibp may serve to cluster full-length IgA antibodies in vivo. Furthermore, in vitro stimulation experiments indicate that binding of the Ibp HCBD alone is sufficient to activate responsive murine B cell receptors. The presence of these proteins in a commensal microbe suggest that binding a broad repertoire of immunoglobulins, particularly in the gut/microbiome environment, may provide an important function in the maintenance of host/microbiome homeostasis contrasting with the pathogenic role of structurally homologous superantigens expressed by pathogens.


Subject(s)
Antibodies, Monoclonal/metabolism , B-Lymphocytes/metabolism , Clostridiales/metabolism , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Variable Region/metabolism , Receptors, Antigen, B-Cell/metabolism , Superantigens/metabolism , Animals , Antibodies, Monoclonal/chemistry , B-Lymphocytes/immunology , Binding Sites , Clostridiales/growth & development , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Mice , Mice, Inbred C57BL , Receptors, Antigen, B-Cell/chemistry , Superantigens/chemistry
4.
Int J Mol Sci ; 21(21)2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33113941

ABSTRACT

The human genome comprises 8% sequences of retroviral origin, so-called human endogenous retroviruses (HERVs). Most of these proviral sequences are defective, but some possess open reading frames. They can lead to the formation of viral transcripts, when activated by intrinsic and extrinsic factors. HERVs are thought to play a pathological role in inflammatory diseases and cancer. Since the consequences of activated proviral sequences in the human body are largely unexplored, selected envelope proteins of human endogenous retroviruses associated with inflammatory diseases, namely HERV-K18, HERV-K113, and HERV-Fc1, were investigated in the present study. A formation of glycosylated envelope proteins was demonstrated in different mammalian cell lines. Nevertheless, protein maturation seemed to be incomplete as no transport to the plasma membrane was observed. Instead, the proteins remained in the ER where they induced the expression of genes involved in unfolded protein response, such as HSPA5 and sXBP1. Furthermore, low expression levels of native envelope proteins were increased by codon optimization. Cell-free expression systems showed that both the transcriptional and translational level is affected. By generating different codon-optimized variants of HERV-K113 envelope, the influence of single rare t-RNA pools in certain cell lines was demonstrated. The mRNA secondary structure also appears to play an important role in the translation of the tested viral envelope proteins. In summary, the formation of certain HERV proteins is basically possible. However, their complete maturation and thus full biologic activity seems to depend on additional factors that might be disease-specific and await elucidation in the future.


Subject(s)
Endogenous Retroviruses/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Superantigens/genetics , Superantigens/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , A549 Cells , Animals , COS Cells , Cell Line , Cell-Free System , Chlorocebus aethiops , Endogenous Retroviruses/chemistry , Endogenous Retroviruses/genetics , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation , Glycosylation , HEK293 Cells , Humans , Membrane Proteins/chemistry , Molecular Conformation , Nucleic Acid Conformation , Open Reading Frames , Protein Biosynthesis , RNA, Messenger/chemistry , Superantigens/chemistry , Transcription, Genetic , Viral Envelope Proteins/chemistry
5.
Proc Natl Acad Sci U S A ; 117(41): 25254-25262, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32989130

ABSTRACT

Multisystem Inflammatory Syndrome in Children (MIS-C) associated with COVID-19 is a newly recognized condition in children with recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These children and adult patients with severe hyperinflammation present with a constellation of symptoms that strongly resemble toxic shock syndrome, an escalation of the cytotoxic adaptive immune response triggered upon the binding of pathogenic superantigens to T cell receptors (TCRs) and/or major histocompatibility complex class II (MHCII) molecules. Here, using structure-based computational models, we demonstrate that the SARS-CoV-2 spike (S) glycoprotein exhibits a high-affinity motif for binding TCRs, and may form a ternary complex with MHCII. The binding epitope on S harbors a sequence motif unique to SARS-CoV-2 (not present in other SARS-related coronaviruses), which is highly similar in both sequence and structure to the bacterial superantigen staphylococcal enterotoxin B. This interaction between the virus and human T cells could be strengthened by a rare mutation (D839Y/N/E) from a European strain of SARS-CoV-2. Furthermore, the interfacial region includes selected residues from an intercellular adhesion molecule (ICAM)-like motif shared between the SARS viruses from the 2003 and 2019 pandemics. A neurotoxin-like sequence motif on the receptor-binding domain also exhibits a high tendency to bind TCRs. Analysis of the TCR repertoire in adult COVID-19 patients demonstrates that those with severe hyperinflammatory disease exhibit TCR skewing consistent with superantigen activation. These data suggest that SARS-CoV-2 S may act as a superantigen to trigger the development of MIS-C as well as cytokine storm in adult COVID-19 patients, with important implications for the development of therapeutic approaches.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Receptors, Antigen, T-Cell/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Superantigens/metabolism , Systemic Inflammatory Response Syndrome/immunology , Amino Acid Motifs , Betacoronavirus/chemistry , Betacoronavirus/genetics , Betacoronavirus/metabolism , COVID-19 , Coronavirus Infections/genetics , Coronavirus Infections/pathology , Enterotoxins/chemistry , Epitopes, T-Lymphocyte , Humans , Intercellular Adhesion Molecule-1/chemistry , Models, Molecular , Mutation , Neurotoxins/chemistry , Pandemics , Pneumonia, Viral/genetics , Pneumonia, Viral/pathology , Protein Binding , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Superantigens/chemistry , Superantigens/genetics , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/pathology
6.
Toxins (Basel) ; 12(9)2020 09 10.
Article in English | MEDLINE | ID: mdl-32927913

ABSTRACT

Staphylococcal enterotoxins are the most common cause of foodborne intoxications (staphylococcal food poisoning) and cause a wide range of diseases. With at least six variants staphylococcal enterotoxin C (SEC) stands out as particularly diverse amongst the 25 known staphylococcal enterotoxins. Some variants present unique and even host-specific features. Here, we review the role of SEC in human and animal health with a particular focus on its role as a causative agent for foodborne intoxications. We highlight structural features unique to SEC and its variants, particularly, the emetic and superantigen activity, as well as the roles of SEC in mastitis and in dairy products. Information about the genetic organization as well as regulatory mechanisms including the accessory gene regulator and food-related stressors are provided.


Subject(s)
Antigens, Bacterial/metabolism , Enterotoxins/metabolism , Food Microbiology , Staphylococcal Food Poisoning/microbiology , Staphylococcus/metabolism , Superantigens/metabolism , Animal Feed/microbiology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Enterotoxins/chemistry , Enterotoxins/genetics , Genetic Variation , Host Specificity , Humans , Protein Conformation , Staphylococcus/genetics , Staphylococcus/pathogenicity , Structure-Activity Relationship , Superantigens/chemistry , Superantigens/genetics , Virulence
7.
Front Immunol ; 11: 1279, 2020.
Article in English | MEDLINE | ID: mdl-32695105

ABSTRACT

Chagas disease is an endemic chronic parasitosis in Latin America affecting more than 7 million people. Around 100 million people are currently at risk of acquiring the infection; however, no effective vaccine has been developed yet. Trypanosoma cruzi is the etiological agent of this parasitosis and as an intracellular protozoan it can reside within different tissues, mainly muscle cells, evading host immunity and allowing progression towards the chronic stage of the disease. Considering this intracellular parasitism triggers strong cellular immunity that, besides being necessary to limit infection, is not sufficient to eradicate the parasite from tissues, a differential immune response is required and new strategies for vaccines against Chagas disease need to be explored. In this work, we designed, cloned and expressed a chimeric molecule, named NCz-SEGN24A, comprising a parasite antigen, the N-terminal domain of the major cysteine protease of T. cruzi, cruzipain (Nt-Cz), and a non-toxic form of the staphylococcal superantigen (SAg) G, SEG, with the residue Asn24 mutated to Ala (N24A). The mutant SAg SEGN24A, retains its ability to trigger classical activation of macrophages without inducing T cell apoptosis. To evaluate, as a proof of concept, the immunogenicity and efficacy of the chimeric immunogen vs. its individual antigens, C3H mice were immunized intramuscularly with NCz-SEGN24A co-adjuvanted with CpG-ODN, or the recombinant proteins Nt-Cz plus SEGN24A with the same adjuvant. Vaccinated mice significantly produced Nt-Cz-specific IgG titers after immunization and developed higher IgG2a than IgG1 titers. Specific cell-mediated immunity was assessed by in-vivo DTH and significant responses were obtained. To assess protection, mice were challenged with trypomastigotes of T. cruzi. Both schemes reduced the parasite load throughout the acute phase, but only mice immunized with NCz-SEGN24A showed significant differences against control; moreover, these mice maintained 100% survival. These results encourage testing mutated superantigens fused to specific antigens as immune modulators against pathogens.


Subject(s)
Antigens, Bacterial/immunology , Chagas Disease/prevention & control , Cross Protection/immunology , Cysteine Endopeptidases/immunology , Protozoan Proteins/immunology , Superantigens/immunology , Trypanosoma cruzi/immunology , Animals , Antibodies, Neutralizing , Antibodies, Protozoan/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Protozoan/immunology , Chagas Disease/immunology , Chagas Disease/parasitology , Cysteine Endopeptidases/genetics , Disease Models, Animal , Immunity, Cellular , Immunity, Humoral , Immunization , Mice , Parasite Load , Protein Conformation , Protein Domains/immunology , Protozoan Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Superantigens/chemistry , Superantigens/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Int J Mol Sci ; 20(23)2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31775346

ABSTRACT

Staphylococcus aureus strains produce a unique family of immunostimulatory exotoxins termed as bacterial superantigens (SAgs), which cross-link major histocompatibility complex class II (MHC II) molecule and T-cell receptor (TCR) to stimulate large numbers of T cells at extremely low concentrations. SAgs are associated with food poisoning and toxic shock syndrome. To date, 26 genetically distinct staphylococcal SAgs have been reported. This study reports the first X-ray structure of newly characterized staphylococcal enterotoxin N (SEN). SEN possesses the classical two domain architecture that includes an N-terminal oligonucleotide-binding fold and a C-terminal ß-grasp domain. Amino acid and structure alignments revealed that several critical amino acids that are proposed to be responsible for MHC II and TCR molecule engagements are variable in SEN, suggesting that SEN may adopt a different binding mode to its cellular receptors. This work helps better understand the mechanisms of action of SAgs.


Subject(s)
Enterotoxins/chemistry , Enterotoxins/metabolism , Histocompatibility Antigens Class II/metabolism , Protein Conformation , Receptors, Antigen, T-Cell/metabolism , Staphylococcus aureus/metabolism , Superantigens/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Histocompatibility Antigens Class II/chemistry , Humans , Models, Molecular , Receptors, Antigen, T-Cell/chemistry , Sequence Homology , Superantigens/chemistry
9.
Protein Eng Des Sel ; 32(8): 359-366, 2019 12 31.
Article in English | MEDLINE | ID: mdl-31641749

ABSTRACT

Staphylococcus aureus interacts with the human immune system through the production of secreted factors. Key among these is protein A, a B-cell superantigen capable of interacting with both antibody Fc and VH regions. Here, we review structural and molecular features of this important example of naturally occurring bacterial superantigens, as well as engineered variants and their application in biotechnology.


Subject(s)
Immunoglobulin Fc Fragments/immunology , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology , Superantigens/immunology , Amino Acid Sequence , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Protein Binding , Protein Engineering/methods , Protein Folding , Sequence Homology, Amino Acid , Staphylococcal Protein A/chemistry , Staphylococcal Protein A/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Superantigens/chemistry , Superantigens/genetics
10.
J Allergy Clin Immunol ; 144(2): 514-523.e5, 2019 08.
Article in English | MEDLINE | ID: mdl-30995457

ABSTRACT

BACKGROUND: Variable heavy chain (VH) family frameworks (FWRs) have been reported to affect antibody receptor and superantigen binding; however, such effects in IgE remain largely unknown. Given that VH family biases have been previously reported in IgE of certain allergies, there is a need to investigate this phenomenon for biotechnological and therapeutic purposes. OBJECTIVE: We sought to investigate the effects of VH families on IgE interaction with FcεRIα, anti-IgE omalizumab, antigen, and superantigen protein A (spA) by using the pertuzumab and trastuzumab IgE models. METHODS: Pertuzumab VH1-VH7 family variants of IgE with the same complementarity-determining regions were investigated with regard to their binding interactions to FcεRIα, Her2, omalizumab, and spA. Notable FcεRIα-IgE observations were cross-checked against appropriate trastuzumab IgE VH variants. Computational structural modeling and simulations were also performed for insight into the mechanism of interactions with various VH FWRs. RESULTS: The pertuzumab VH5 IgE variant, but not the trastuzumab VH5 IgE, was found to interact with FcεRIα significantly longer than the respective VH family variants within each model antibody. No significant differences in interaction were found between IgE and omalizumab for the pertuzumab VH variants. Although trastuzumab VH3 interacted with spA, none of our pertuzumab VH variants, including VH3, associated with spA. CONCLUSION: We found unexpected varying allosteric communications caused by the VH family FWRs to the FcεRIα-, Her2-, and spA-binding regions of pertuzumab IgE, with implications for use of IgE/anti-IgE therapeutics to treat allergy and IgE therapeutics in allergo-oncology.


Subject(s)
Antigens, Bacterial/chemistry , Immunoglobulin E/chemistry , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/immunology , Receptors, IgE/chemistry , Superantigens/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antigens, Bacterial/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/therapy , Immunoglobulin E/immunology , Immunoglobulin E/therapeutic use , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/therapeutic use , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/therapeutic use , Immunotherapy , Omalizumab/chemistry , Omalizumab/immunology , Receptors, IgE/immunology , Superantigens/immunology , Trastuzumab/chemistry , Trastuzumab/immunology
11.
Sci Rep ; 9(1): 4211, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862940

ABSTRACT

Methicillin resistant Staphylococcus aureus (MRSA) is a major human pathogen, which causes superficial to lethal clinical infections. Neutrophils are the most abundant leukocytes in the blood and are the first defense mechanism against S. aureus infections. Here we show Staphylococcal Superantigen-Like protein 11 (SSL11) from MRSA USA300_FPR3757 mediated differentiated human neutrophil-like cells (dHL60) motility arrest by inducing cell adhesion and "locking" cells in adhesion stage, without inducing oxidative burst. Pre-incubation of SSL11 with the glycan Sialyl Lewis X blocked SSL11 function and de-glycosylation of dHL60 cells by PNGase F abolished SSL11 binding, suggesting that SSL11 functions via interacting with glycans. This is the first description of a bacterial toxin inhibiting neutrophil motility by inducing adhesion and "locking" cells in an adhesion stage. Therefore, this study might provide a new target against S. aureus infections.


Subject(s)
Bacterial Proteins/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Methicillin-Resistant Staphylococcus aureus/chemistry , Neutrophils/metabolism , Superantigens/pharmacology , Bacterial Proteins/chemistry , HL-60 Cells , Humans , Neutrophils/pathology , Superantigens/chemistry
12.
Sci Rep ; 9(1): 3279, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824769

ABSTRACT

Superantigens (SAgs) play a major role in the pathogenesis of Staphylococcus aureus and are associated with several diseases, including food poisoning, bacterial arthritis, and toxic shock syndrome. Monoclonal antibodies to these SAgs, primarily TSST-1, SEB and SEA have been shown to provide protection in animal studies and to reduce clinical severity in bacteremic patients. Here we quantify the pre-existing antibodies against SAgs in many human plasma and IVIG samples and demonstrate that in a major portion of the population these antibody titers are suboptimal and IVIG therapy only incrementally elevates the anti-SAg titers. Our in vitro neutralization studies show that a combination of antibodies against SEA, SEB,and TSST-1 can provide broad neutralization of staphylococcal SAgs. We report a single fusion protein (TBA225) consisting of the toxoid versions of TSST-1, SEB and SEA and demonstrate its immunogenicity and protective efficacy in a mouse model of toxic shock. Antibodies raised against this fusion vaccine provide broad neutralization of purified SAgs and culture supernatants of multiple clinically relevant S. aureus strains. Our data strongly supports the use of this fusion protein as a component of an anti-virulence based multivalent toxoid vaccine against S. aureus disease.


Subject(s)
Enterotoxins/toxicity , Recombinant Fusion Proteins/pharmacology , Staphylococcal Toxoid/pharmacology , Staphylococcus aureus , Superantigens/toxicity , Animals , Enterotoxins/chemistry , Enterotoxins/genetics , Enterotoxins/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Staphylococcal Toxoid/chemistry , Staphylococcal Toxoid/genetics , Staphylococcal Toxoid/immunology , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Superantigens/chemistry , Superantigens/genetics , Superantigens/immunology
13.
Proc Natl Acad Sci U S A ; 115(37): E8707-E8716, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150373

ABSTRACT

Antibodies classically bind antigens via their complementarity-determining regions, but an alternative mode of interaction involving V-domain framework regions has been observed for some B cell "superantigens." We report the crystal structure of an antibody employing both modes of interaction simultaneously and binding two antigen molecules. This human antibody from an allergic individual binds to the grass pollen allergen Phl p 7. Not only are two allergen molecules bound to each antibody fragment (Fab) but also each allergen molecule is bound by two Fabs: One epitope is recognized classically, the other in a superantigen-like manner. A single allergen molecule thus cross-links two identical Fabs, contrary to the one-antibody-one-epitope dogma, which dictates that a dimeric allergen at least is required for this to occur. Allergens trigger immediate hypersensitivity reactions by cross-linking receptor-bound IgE molecules on effector cells. We found that monomeric Phl p 7 induced degranulation of basophils sensitized solely with this monoclonal antibody expressed as an IgE, demonstrating that the dual specificity has functional consequences. The monomeric state of Phl p 7 and two structurally related allergens was confirmed by size-exclusion chromatography and multiangle laser light scattering, and the results were supported by degranulation studies with the related allergens, a second patient-derived allergen-specific antibody lacking the nonclassical binding site, and mutagenesis of the nonclassically recognized allergen epitope. The antibody dual reactivity and cross-linking mechanism not only have implications for understanding allergenicity and allergen potency but, importantly, also have broader relevance to antigen recognition by membrane Ig and cross-linking of the B cell receptor.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Plant/immunology , Calcium-Binding Proteins/immunology , Epitopes/immunology , Superantigens/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibody Specificity/immunology , Antigens, Plant/chemistry , Antigens, Plant/metabolism , Basophils/immunology , Basophils/physiology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cell Degranulation/immunology , Cross Reactions/immunology , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/metabolism , Humans , Immunoglobulin E/chemistry , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Superantigens/chemistry , Superantigens/metabolism
14.
Toxins (Basel) ; 10(4)2018 03 27.
Article in English | MEDLINE | ID: mdl-29584685

ABSTRACT

Staphylococcal Enterotoxins (SEs) are superantigens (SAg) originally produced by S. aureus, but their presence in coagulase negative staphylococci (CNS) has long been suspected. This study aims to better characterize a novel C-like enterotoxin expressed by clinical S. epidermidis strains, called SECepi. We isolated and characterized SECepi for its molecular and functional properties. The toxin was structurally modeled according to its significant similarity with S. aureus SEC3. Most of SEC amino acid residues important for the formation of the trimolecular Major Histocompatibility Complex II MHCII-SEC-T Cell Receptor TCR complex are conserved in SECepi. The functional properties of SECepi were estimated after cloning, expression in E. coli, and purification. The recombinant SECepi toxin exhibits biological characteristics of a SAg including stimulation of human T-cell mitogenicity, inducing and releasing high cytokines levels: IL-2, -4, -6, -8, -10, IFN-γ, TNF-α and GM-CSF at a dose as low as 3.7 pM. Compared to SECaureus, the production of pro-sepsis cytokine IL-6 is significantly higher with SECepi-activated lymphocytes. Furthermore, SECepi is stable to heat, pepsin or trypsin hydrolysis. The SECepi superantigen produced by CNS is functionally very close to that of S. aureus, possibly inducing a systemic inflammatory response at least comparable to that of SECaureus, and may account for S. epidermidis pathogenicity.


Subject(s)
Enterotoxins , Staphylococcus epidermidis/physiology , Superantigens , Cell Proliferation/drug effects , Cytokines/metabolism , Enterotoxins/chemistry , Enterotoxins/metabolism , Enterotoxins/pharmacology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Superantigens/chemistry , Superantigens/metabolism , Superantigens/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology
15.
Biotechnol Lett ; 39(9): 1413-1423, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28653102

ABSTRACT

OBJECTIVES: To identify and characterize staphylococcus exotoxin-like (SET) protein Set11 from Staphylococcus aureus Mu50 strain and its possible targets proteins from human blood/serum. RESULTS: Set11 is a member of the staphylococcal superantigen-like (SSL) proteins (also called Staphylococcus exotoxin-like (SET) proteins) family that is found in staphylococcal strain Mu50. Its structure and function, however, remain unknown. We performed bioinformatics analysis of Set11: it had 90% sequence identity to SSL7 in NCTC 8325 strain, indicating Set11 is a SSL7 ortholog. SSL7 in ATCC 12598 strain binds complement C5 to inhibit complement system. To investigate if Set11 binds C5, we made the homology model of Set11 and the Set11-C5 complex model based on SSL7 and SSL7-C5 structures, respectively. Structural analysis and sequence alignment reveal that the residues in SSL7 involved in C5 binding are conserved in Set11, indicating C5 as the potential target for Set11. To identify new targets of Set11, we cloned, expressed and purified Set11 and performed CNBr-pull down combined mass spectrum assays using human blood and serum. CONCLUSIONS: We identified Set11 as the ortholog of SSL7 and determined C5, fibronectin 1 isoform 3 preproprotein, albumin, alpha-1-microglobulin precursor and complement C3 processor as the potential target proteins for Set11, indicating new functions of Set11/SSL7.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Complement C5/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Superantigens/genetics , Superantigens/metabolism , Albumins/metabolism , Alpha-Globulins/metabolism , Bacterial Proteins/chemistry , Cloning, Molecular , Complement C3/metabolism , Computational Biology , Cytokines/metabolism , Fibronectins , Gene Expression , Humans , Mass Spectrometry , Models, Molecular , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Serum/chemistry , Substrate Specificity , Superantigens/chemistry
16.
J Parasitol ; 103(5): 579-583, 2017 10.
Article in English | MEDLINE | ID: mdl-28581897

ABSTRACT

B-cell superantigens (BC-SAgs) are immunoevasins that have evolved in response to innate catalytic IgM antibodies; germ-line encoded immunoglobulins present in the preimmune repertoire independent of prior antigen exposure. Catalysis is the result of a 2-step process that involves first the formation of a non-covalent bond between the BC-SAg and the immunoglobulin followed by covalent bond formation at the catalytic site resulting in target hydrolysis. Tc24 is a recently described Trypanosoma cruzi BC-SAg hypothesized to play a role in evading the humoral response early in the infection period. We previously demonstrated that exposure to Tc24 following immunization or infection resulted in the depletion of the catalytic IgM response, leaving a gap in the catalytic IgM repertoire. The present report compares the BC-SAg properties of wild-type Tc24 (Tc24-WT) to that of 2 recombinant Tc24 isoforms: Tc24-C2 (Cys to Ser mutations in the 2 most-proximal Cys residues) and Tc24-C4 (Cys to Ser mutations in all 4 Cys residues present). BC-SAg activity was assessed by immunizing mice with the respective isoforms and examining the ability of IgM purified from the respective groups to hydrolyze the 3 Tc24 isoforms. In addition, the ability of IgM purified from naive mice to hydrolyze the Tc24 isoforms was also assessed. Immunization with Tc24-WT, Tc24-C2, or Tc24-C4 resulted in loss of IgM-mediated hydrolysis of Tc24-WT. However, the ability of IgM purified from naive mice (previously shown to hydrolyze Tc24-WT) was less effective in hydrolyzing the 2 Tc24 isoforms. These data demonstrate that although the BC-SAg site in the mutants remained intact, their reduced susceptibility to IgM-mediated hydrolysis suggested that structural changes resulting from the Cys to Ser mutations altered accessibility to the catalytic site in the 2 isoforms.


Subject(s)
Cysteine/genetics , Immunoglobulin M/immunology , Superantigens/genetics , Trypanosoma cruzi/genetics , Trypanosoma cruzi/immunology , Animals , Antibodies, Protozoan/immunology , B-Lymphocytes/immunology , Calcium-Binding Proteins/immunology , Chagas Disease/immunology , Cysteine/chemistry , Hydrolysis , Immunity, Innate/immunology , Immunization , Mice , Mutation , Protein Isoforms/immunology , Protozoan Vaccines , Sequence Alignment , Serine/chemistry , Serine/genetics , Superantigens/chemistry , Superantigens/immunology
17.
Infect Disord Drug Targets ; 17(2): 130-142, 2017.
Article in English | MEDLINE | ID: mdl-28034364

ABSTRACT

Toxins are one among the numerous virulence factors produced by the bacteria. These are powerful poisonous substances enabling the bacteria to encounter the defense mechanism of human body. The pathogenic system of Staphylococcus aureus is evolved with various exotoxins that cause detrimental effects on human immune system. Four toxins namely enterotoxin A, exfoliative toxin A, TSST-1 and γ-hemolysin were downloaded from Uniprot database and were analyzed to understand the nature of the toxins and for drug target validation. The results inferred that the toxins were found to interact with many protein partners and no homologous sequences for human proteome were found, and based on similarity search in Drugbank, the targets were identified as novel drug targets.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Staphylococcus aureus/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Computer Simulation , Enterotoxins/chemistry , Enterotoxins/metabolism , Exfoliatins/chemistry , Exfoliatins/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Humans , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Superantigens/chemistry , Superantigens/metabolism , Virulence Factors/chemistry , Virulence Factors/metabolism
18.
Proc Natl Acad Sci U S A ; 113(42): E6437-E6446, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27708164

ABSTRACT

Full T-cell activation requires interaction between the costimulatory receptors B7-2 and CD28. By binding CD28, bacterial superantigens elicit harmful inflammatory cytokine overexpression through an unknown mechanism. We show that, by engaging not only CD28 but also its coligand B7-2 directly, superantigens potently enhance the avidity between B7-2 and CD28, inducing thereby T-cell hyperactivation. Using the same 12-aa ß-strand-hinge-α-helix domain, superantigens engage both B7-2 and CD28 at their homodimer interfaces, areas remote from where these coreceptors interact, implying that inflammatory signaling can be controlled through the receptor homodimer interfaces. Short B7-2 dimer interface mimetic peptides bind diverse superantigens, prevent superantigen binding to cell-surface B7-2 or CD28, attenuate inflammatory cytokine overexpression, and protect mice from lethal superantigen challenge. Thus, superantigens induce a cytokine storm not only by mediating the interaction between MHC-II molecule and T-cell receptor but also, critically, by promoting B7-2/CD28 coreceptor engagement, forcing the principal costimulatory axis to signal excessively. Our results reveal a role for B7-2 as obligatory receptor for superantigens. B7-2 homodimer interface mimotopes prevent superantigen lethality by blocking the superantigen-host costimulatory receptor interaction.


Subject(s)
B7-2 Antigen/metabolism , CD28 Antigens/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Superantigens/immunology , Amino Acid Sequence , Animals , B7-2 Antigen/chemistry , B7-2 Antigen/genetics , Cell Line, Tumor , Cytokines/genetics , Enterotoxins/chemistry , Enterotoxins/immunology , Female , Humans , Mice , Models, Molecular , Molecular Mimicry , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Protein Binding/immunology , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Fusion Proteins , Signal Transduction , Superantigens/chemistry , Superantigens/metabolism
19.
Sci Rep ; 6: 25796, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27180909

ABSTRACT

Superantigens are toxins produced by Staphylococcus aureus, called staphylococcal enterotoxins (abbreviated SEA to SEU). They can cross-link the T cell receptor (TCR) and major histocompatibility complex class II, triggering a massive T cell activation and hence disease. Due to high stability and toxicity, superantigens are potential agents of bioterrorism. Hence, antagonists may not only be useful in the treatment of disease but also serve as countermeasures to biological warfare. Of particular interest are inhibitors against SEA and SEB. SEA is the main cause of food poisoning, while SEB is a common toxin manufactured as a biological weapon. Here, we present the crystal structures of SEA in complex with TCR and SEE in complex with the same TCR, complemented with computational alanine-scanning mutagenesis of SEA, SEB, SEC3, SEE, and SEH. We have identified two common areas that contribute to the general TCR binding for these superantigens. This paves the way for design of single antagonists directed towards multiple toxins.


Subject(s)
Enterotoxins/chemistry , Enterotoxins/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Amino Acid Motifs , Amino Acid Sequence , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Mutagenesis/genetics , Protein Structure, Secondary , Superantigens/chemistry , Superantigens/metabolism
20.
Methods Mol Biol ; 1396: 1-17, 2016.
Article in English | MEDLINE | ID: mdl-26676033

ABSTRACT

Superantigens secreted by Staphylococcus aureus and Streptococcus pyogenes interact with the T-cell receptor and major histocompatibility class II molecules on antigen-presenting cells to elicit a massive cytokine release and activation of T cells in higher numbers than that seen with ordinary antigens. Because of this unique ability, superantigens have been implicated as etiological agents for many different types of diseases, including toxic shock syndrome, infective endocarditis, pneumonia, and inflammatory skin diseases. This review covers the main animal models that have been developed in order to identify the roles of superantigens in human disease.


Subject(s)
Disease Models, Animal , Staphylococcus aureus/immunology , Streptococcus pyogenes/immunology , Superantigens/immunology , Animals , Dermatitis/etiology , Endocarditis/etiology , Humans , Pneumonia/etiology , Shock, Septic/etiology , Superantigens/chemistry , Superantigens/metabolism , Vomiting/etiology
SELECTION OF CITATIONS
SEARCH DETAIL