Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(3): e1012072, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38452154

ABSTRACT

Streptococcus pyogenes is a human-specific pathogen that commonly colonizes the upper respiratory tract and skin, causing a wide variety of diseases ranging from pharyngitis to necrotizing fasciitis and toxic shock syndrome. S. pyogenes has a repertoire of secreted virulence factors that promote infection and evasion of the host immune system including the cytolysins streptolysin O (SLO) and streptolysin S (SLS). S. pyogenes does not naturally infect the upper respiratory tract of mice although mice transgenic for MHC class II human leukocyte antigens (HLA) become highly susceptible. Here we used HLA-transgenic mice to assess the role of both SLO and SLS during both nasopharyngeal and skin infection. Using S. pyogenes MGAS8232 as a model strain, we found that an SLS-deficient strain exhibited a 100-fold reduction in bacterial recovery from the nasopharynx and a 10-fold reduction in bacterial burden in the skin, whereas an SLO-deficient strain did not exhibit any infection defects in these models. Furthermore, depletion of neutrophils significantly restored the bacterial burden of the SLS-deficient bacteria in skin, but not in the nasopharynx. In mice nasally infected with the wildtype S. pyogenes, there was a marked change in localization of the tight junction protein ZO-1 at the site of infection, demonstrating damage to the nasal epithelia that was absent in mice infected with the SLS-deficient strain. Overall, we conclude that SLS is required for the establishment of nasopharyngeal infection and skin infection in HLA-transgenic mice by S. pyogenes MGAS8232 and provide evidence that SLS contributes to nasopharyngeal infection through the localized destruction of nasal epithelia.


Subject(s)
Streptococcal Infections , Streptococcus pyogenes , Humans , Mice , Animals , Streptococcus pyogenes/metabolism , Streptolysins/genetics , Streptolysins/metabolism , Mice, Transgenic , Streptococcal Infections/metabolism , Bacterial Proteins/metabolism , Nasopharynx
2.
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
3.
Nat Commun ; 11(1): 5018, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33024089

ABSTRACT

The re-emergence of scarlet fever poses a new global public health threat. The capacity of North-East Asian serotype M12 (emm12) Streptococcus pyogenes (group A Streptococcus, GAS) to cause scarlet fever has been linked epidemiologically to the presence of novel prophages, including prophage ΦHKU.vir encoding the secreted superantigens SSA and SpeC and the DNase Spd1. Here, we report the molecular characterization of ΦHKU.vir-encoded exotoxins. We demonstrate that streptolysin O (SLO)-induced glutathione efflux from host cellular stores is a previously unappreciated GAS virulence mechanism that promotes SSA release and activity, representing the first description of a thiol-activated bacterial superantigen. Spd1 is required for resistance to neutrophil killing. Investigating single, double and triple isogenic knockout mutants of the ΦHKU.vir-encoded exotoxins, we find that SpeC and Spd1 act synergistically to facilitate nasopharyngeal colonization in a mouse model. These results offer insight into the pathogenesis of scarlet fever-causing GAS mediated by prophage ΦHKU.vir exotoxins.


Subject(s)
Exotoxins/metabolism , Prophages/genetics , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/virology , Animals , Bacterial Proteins/pharmacology , Cell Line , Erythrocytes/drug effects , Exotoxins/genetics , Female , Glutathione/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Pharynx/cytology , Scarlet Fever/epidemiology , Scarlet Fever/microbiology , Streptococcus pyogenes/genetics , Streptolysins/pharmacology , Superantigens/genetics , Superantigens/metabolism
5.
PLoS One ; 14(8): e0219777, 2019.
Article in English | MEDLINE | ID: mdl-31398210

ABSTRACT

BACKGROUND: Injection drug use-associated endocarditis (IDUaIE) incidence in Ontario has recently been associated with hydromorphone prescribing rates. Staphylococcus aureus causes the majority of cases of IDUaIE in Ontario and across North America. Hydromorphone controlled-release (Hydromorphone-CR) requires a complex technique for injection and therefore provides multiple opportunities for contamination. Hydromorphone-CR contains several excipients, which could enhance staphylococcal survival and increase risk of contaminating the injectate. METHODS: Used injection drug preparation equipment (cookers/filters) was collected from persons who inject drugs (PWID), rinsed with water, and plated on Mannitol salt agar. Bacterial isolates from bacteremic PWID were used to assess the survival of S. aureus and Streptococcus pyogenes on cookers/filters with Hydromorphone-CR, hydromorphone immediate-release (Hydromorphone-IR) or oxycodone controlled-release (Oxycodone-CR). The solutions spiked with S. aureus were heated and the remaining viable bacteria enumerated. RESULTS: S. aureus was detected in 12/87 (14%, 95%CI 8-23%) cookers/filters samples used for injection of Hydromorphone-CR. Hydromorphone-CR was the only opioid associated with greater survival of methicillin-sensitive S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA) on cookers/filters when compared to sterile water vehicle control. There was a ~2 log reduction in the number of S. aureus that survived when cookers/filters were heated. CONCLUSION: 14% of all cookers/filters used in the preparation of Hydromorphone-CR were contaminated with S. aureus. Hydromorphone-CR prolongs the survival of MRSA and MSSA in cookers/filters. Heating cookers/filters may be a harm-reduction strategy.


Subject(s)
Bacteremia/microbiology , Drug Compounding/instrumentation , Endocarditis, Bacterial/microbiology , Equipment Contamination , Hydromorphone/administration & dosage , Microbial Viability , Staphylococcus aureus/physiology , Administration, Oral , Delayed-Action Preparations , Injections , Risk
6.
Infect Genet Evol ; 61: 160-175, 2018 07.
Article in English | MEDLINE | ID: mdl-29530660

ABSTRACT

Streptococcus pyogenes is a human-specific and globally prominent bacterial pathogen that despite causing numerous human infections, this bacterium is normally found in an asymptomatic carrier state. This review provides an overview of both bacterial and human factors that likely play an important role in nasopharyngeal colonization and pharyngitis, as well as the development of acute rheumatic fever and rheumatic heart disease. Here we highlight a recently described role for bacterial superantigens in promoting acute nasopharyngeal infection, and discuss how these immune system activating toxins could be crucial to initiate the autoimmune process in rheumatic heart disease.


Subject(s)
Pharyngitis/microbiology , Rheumatic Fever/microbiology , Streptococcus pyogenes , Superantigens , Carrier State/microbiology , Humans , Nasopharynx/microbiology , Streptococcal Infections/microbiology
7.
Proc Natl Acad Sci U S A ; 114(38): 10226-10231, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28794279

ABSTRACT

The globally prominent pathogen Streptococcus pyogenes secretes potent immunomodulatory proteins known as superantigens (SAgs), which engage lateral surfaces of major histocompatibility class II molecules and T-cell receptor (TCR) ß-chain variable domains (Vßs). These interactions result in the activation of numerous Vß-specific T cells, which is the defining activity of a SAg. Although streptococcal SAgs are known virulence factors in scarlet fever and toxic shock syndrome, mechanisms by how SAgs contribute to the life cycle of S. pyogenes remain poorly understood. Herein, we demonstrate that passive immunization against the Vß8-targeting SAg streptococcal pyrogenic exotoxin A (SpeA), or active immunization with either wild-type or a nonfunctional SpeA mutant, protects mice from nasopharyngeal infection; however, only passive immunization, or vaccination with inactive SpeA, resulted in high-titer SpeA-specific antibodies in vivo. Mice vaccinated with wild-type SpeA rendered Vß8+ T cells poorly responsive, which prevented infection. This phenotype was reproduced with staphylococcal enterotoxin B, a heterologous SAg that also targets Vß8+ T cells, and rendered mice resistant to infection. Furthermore, antibody-mediated depletion of T cells prevented nasopharyngeal infection by S. pyogenes, but not by Streptococcus pneumoniae, a bacterium that does not produce SAgs. Remarkably, these observations suggest that S. pyogenes uses SAgs to manipulate Vß-specific T cells to establish nasopharyngeal infection.

8.
J Bacteriol ; 198(19): 2732-42, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27457715

ABSTRACT

UNLABELLED: Toxic shock syndrome toxin 1 (TSST-1) is a Staphylococcus aureus superantigen that has been implicated in both menstrual and nonmenstrual toxic shock syndrome (TSS). Despite the important role of TSST-1 in severe human disease, a comprehensive understanding of staphylococcal regulatory factors that control TSST-1 expression remains incomplete. The S. aureus exotoxin expression (Sae) operon contains a well-characterized two-component system that regulates a number of important exotoxins in S. aureus, although regulation of TSST-1 by the Sae system has not been investigated. We generated a defined deletion mutant of the Sae histidine kinase sensor (saeS) in the prototypic menstrual TSS strain S. aureus MN8. Mutation of saeS resulted in a complete loss of TSST-1 expression. Using both luciferase reporter experiments and quantitative real-time PCR, we demonstrate that the Sae system is an important transcriptional activator of TSST-1 expression. Recombinant SaeR was able to bind directly to the tst promoter to a region containing two SaeR consensus binding sites. Although the stand-alone SarA transcriptional regulator has been shown to be both a positive and a negative regulator of TSST-1, deletion of sarA in S. aureus MN8 resulted in a dramatic overexpression of TSST-1. As expected, mutation of agr also reduced TSST-1 expression, but this phenotype appeared to be independent of Sae. A double mutation of saeS and sarA resulted in the loss of TSST-1 expression. This work indicates that the Sae system is a dominant and direct transcriptional activator that is required for expression of TSST-1. IMPORTANCE: The TSST-1 superantigen is an exotoxin, produced by some strains of S. aureus, that has a clear role in both menstrual and nonmenstrual TSS. Although the well-characterized agr quorum sensing system is a known positive regulator of TSST-1, the molecular mechanisms that directly control TSST-1 expression are only partially understood. Our studies demonstrate that the Sae two-component regulatory system is a positive transcriptional regulator that binds directly to the TSST-1 promoter, and furthermore, our data suggest that Sae is required for expression of TSST-1. This work highlights how major regulatory circuits can converge to fine-tune exotoxin expression and suggests that the Sae regulatory system may be an important target for antivirulence strategies.


Subject(s)
Bacterial Toxins/metabolism , Enterotoxins/metabolism , Gene Expression Regulation, Bacterial/physiology , Protein Kinases/metabolism , Staphylococcus aureus/metabolism , Superantigens/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Enterotoxins/genetics , Promoter Regions, Genetic , Protein Kinases/genetics , Staphylococcus aureus/genetics , Superantigens/genetics
9.
Methods Mol Biol ; 1396: 95-107, 2016.
Article in English | MEDLINE | ID: mdl-26676040

ABSTRACT

Streptococcus pyogenes is a globally prominent human-specific pathogen that is responsible for an enormous burden of infectious disease. Despite intensive experimental efforts to understand the molecular correlates that contribute to invasive infections, there has been less focus on S. pyogenes carriage and local infection of the nasopharynx. This chapter describes an acute nasopharyngeal infection model in mice that is utilized in our laboratory to study the role of superantigen toxins in the biology of S. pyogenes. We also describe a method to detect superantigen-specific T cell activation in vivo.


Subject(s)
Nasopharyngitis/immunology , Nasopharyngitis/microbiology , Streptococcus pyogenes/immunology , Superantigens/immunology , Animals , Bacterial Load , Disease Models, Animal , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
10.
Toxins (Basel) ; 7(5): 1821-36, 2015 May 22.
Article in English | MEDLINE | ID: mdl-26008236

ABSTRACT

Superantigens (SAgs) are potent microbial toxins that function to activate large numbers of T cells in a T cell receptor (TCR) Vß-specific manner, resulting in excessive immune system activation. Staphylococcus aureus possesses a large repertoire of distinct SAgs, and in the context of host-pathogen interactions, staphylococcal SAg research has focused primarily on the role of these toxins in severe and invasive diseases. However, the contribution of SAgs to colonization by S. aureus remains unclear. We developed a two-week nasal colonization model using SAg-sensitive transgenic mice expressing HLA-DR4, and evaluated the role of SAgs using two well-studied stains of S. aureus. S. aureus Newman produces relatively low levels of staphylococcal enterotoxin A (SEA), and although we did not detect significant TCR-Vß specific changes during wild-type S. aureus Newman colonization, S. aureus Newman Δsea established transiently higher bacterial loads in the nose. S. aureus COL produces relatively high levels of staphylococcal enterotoxin B (SEB), and colonization with wild-type S. aureus COL resulted in clear Vß8-specific T cell skewing responses. S. aureus COL Δseb established consistently higher bacterial loads in the nose. These data suggest that staphylococcal SAgs may be involved in regulating bacterial densities during nasal colonization.


Subject(s)
Antigens, Bacterial/immunology , Enterotoxins/immunology , Nose/microbiology , Staphylococcus aureus , Superantigens/immunology , Animals , Bacterial Load , Female , HLA-DR4 Antigen , Liver/microbiology , Lung/microbiology , Lymph Nodes/immunology , Male , Mice, Transgenic , T-Lymphocytes/immunology
11.
PLoS Pathog ; 10(5): e1004155, 2014 May.
Article in English | MEDLINE | ID: mdl-24875883

ABSTRACT

Establishing the genetic determinants of niche adaptation by microbial pathogens to specific hosts is important for the management and control of infectious disease. Streptococcus pyogenes is a globally prominent human-specific bacterial pathogen that secretes superantigens (SAgs) as 'trademark' virulence factors. SAgs function to force the activation of T lymphocytes through direct binding to lateral surfaces of T cell receptors and class II major histocompatibility complex (MHC-II) molecules. S. pyogenes invariably encodes multiple SAgs, often within putative mobile genetic elements, and although SAgs are documented virulence factors for diseases such as scarlet fever and the streptococcal toxic shock syndrome (STSS), how these exotoxins contribute to the fitness and evolution of S. pyogenes is unknown. Here we show that acute infection in the nasopharynx is dependent upon both bacterial SAgs and host MHC-II molecules. S. pyogenes was rapidly cleared from the nasal cavity of wild-type C57BL/6 (B6) mice, whereas infection was enhanced up to ∼10,000-fold in B6 mice that express human MHC-II. This phenotype required the SpeA superantigen, and vaccination with an MHC -II binding mutant toxoid of SpeA dramatically inhibited infection. Our findings indicate that streptococcal SAgs are critical for the establishment of nasopharyngeal infection, thus providing an explanation as to why S. pyogenes produces these potent toxins. This work also highlights that SAg redundancy exists to avoid host anti-SAg humoral immune responses and to potentially overcome host MHC-II polymorphisms.


Subject(s)
Bacterial Proteins/metabolism , Exotoxins/metabolism , Histocompatibility Antigens Class II/immunology , Membrane Proteins/metabolism , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Superantigens/immunology , Acute Disease , Animals , Bacterial Proteins/immunology , Exotoxins/immunology , Humans , Membrane Proteins/immunology , Mice, Inbred C57BL , Nasopharynx/immunology , Receptors, Antigen, T-Cell/immunology , Streptococcal Infections/prevention & control , Streptococcus pyogenes/genetics , Superantigens/genetics , T-Lymphocytes/immunology
12.
BMC Infect Dis ; 13: 381, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23957431

ABSTRACT

BACKGROUND: Fournier's gangrene is a rare necrotizing soft tissue infection of the scrotum and penis. We report, to our knowledge, the first case of Fournier's gangrene caused by Streptococcus dysgalactiae subsp. equisimilis (SDSE), a strain of pyogenic ß-hemolytic streptococci that is increasingly being recognized as an important human pathogen. CASE PRESENTATION: We describe a healthy 59 year-old Caucasian male who presented to the emergency department with Fournier's gangrene of the penis and scrotum, with extension to the anterior abdominal wall. He underwent urgent surgical debridement of his scrotum, penis, and anterior abdomen. Swabs from the scrotum grew Gram-positive cocci, which were initially identified as Streptococcus anginosus group by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS). However, polymerase chain reaction (PCR) amplification and sequencing of the 16S rRNA gene identified the isolate as Streptococcus dysgalatiae subspecies equisimilis (SDSE). The incidences of invasive S. anginosus group and SDSE infections at the London Health Sciences Centre, a tertiary-care institution in southwestern Ontario, were determined between August 1, 2011 and August 31, 2012, revealing a slightly lower rate of SDSE (3.2 cases per 100,000 population) than other studies. CONCLUSIONS: This case highlights a unique disease manifestation of the emerging human pathogen Streptococcus dysgalatiae subspecies equisimilis that has not been previously reported. This case also underscores the limitations of MALDI-TOF MS in differentiating between closely-related streptococcal species which may have different pathogenic profiles.


Subject(s)
Fournier Gangrene/microbiology , Penile Diseases/microbiology , Streptococcal Infections/epidemiology , Tertiary Care Centers , Cohort Studies , Fournier Gangrene/diagnosis , Fournier Gangrene/epidemiology , Humans , Incidence , Male , Middle Aged , Penile Diseases/diagnosis , Penile Diseases/epidemiology , Retrospective Studies , Streptococcal Infections/diagnosis , Streptococcus/isolation & purification
13.
J Biol Chem ; 286(6): 4871-81, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21127057

ABSTRACT

Superantigens (SAgs) are microbial toxins defined by their ability to activate T lymphocytes in a T cell receptor (TCR) ß-chain variable domain (Vß)-specific manner. Although existing structural information indicates that diverse bacterial SAgs all uniformly engage the Vß second complementarity determining region (CDR2ß) loop, the molecular rules that dictate SAg-mediated T cell activation and Vß specificity are not fully understood. Herein we report the crystal structure of human Vß2.1 (hVß2.1) in complex with the toxic shock syndrome toxin-1 (TSST-1) SAg, and mutagenesis of hVß2.1 indicates that the non-canonical length of CDR2ß is a critical determinant for recognition by TSST-1 as well as the distantly related SAg streptococcal pyrogenic exotoxin C. Frame work (FR) region 3 is uniquely critical for TSST-1 function explaining the fine Vß-specificity exhibited by this SAg. Furthermore, domain swapping experiments with SAgs, which use distinct domains to engage both CDR2ß and FR3/4ß revealed that the CDR2ß contacts dictate T lymphocyte Vß-specificity. These findings demonstrate that the TCR CDR2ß loop is the critical determinant for functional recognition and Vß-specificity by diverse bacterial SAgs.


Subject(s)
Bacterial Toxins/chemistry , Complementarity Determining Regions/chemistry , Enterotoxins/chemistry , Lymphocyte Activation , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Superantigens/chemistry , T-Lymphocytes/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Crystallography, X-Ray , Enterotoxins/genetics , Enterotoxins/immunology , Exotoxins/chemistry , Exotoxins/genetics , Exotoxins/immunology , Humans , Mutagenesis , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Superantigens/genetics , Superantigens/immunology , T-Lymphocytes/immunology
14.
Nat Med ; 15(6): 641-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19465927

ABSTRACT

Staphylococcal superantigens are pyrogenic exotoxins that cause massive T cell activation leading to toxic shock syndrome and death. Despite the strong adaptive immune response induced by these toxins, infections by superantigen-producing staphylococci are very common clinical events. We hypothesized that this may be partly a result of staphylococcal strains having developed strategies that downregulate the T cell response to these toxins. Here we show that the human interleukin-2 response to staphylococcal superantigens is inhibited by the simultaneous presence of bacteria. Such a downregulatory effect is the result of peptidoglycan-embedded molecules binding to Toll-like receptor 2 and inducing interleukin-10 production and apoptosis of antigen-presenting cells. We corroborated these findings in vivo by showing substantial prevention of mortality after simultaneous administration of staphylococcal enterotoxin B with either heat-killed staphylococci or Staphylococcus aureus peptidoglycan in mouse models of superantigen-induced toxic shock syndrome.


Subject(s)
Cell Wall/immunology , Lymphocyte Activation/immunology , Shock, Septic/prevention & control , Staphylococcus aureus/immunology , Superantigens/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 2/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Bacterial/immunology , Apoptosis , Down-Regulation/immunology , Humans , Interleukin-2/immunology , Ligands , Mice , NF-kappa B/metabolism , Shock, Septic/immunology
15.
J Immunol ; 181(5): 3384-92, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18714010

ABSTRACT

Superantigens (SAgs) are microbial toxins that bind to both TCR beta-chain variable domains (Vbetas) and MHC class II molecules, resulting in the activation of T cells in a Vbeta-specific manner. It is now well established that different isoforms of MHC II molecules can play a significant role in the immune response to bacterial SAgs. In this work, using directed mutational studies in conjunction with functional analyses, we provide a complete functional map of the low-affinity MHC II alpha-chain binding interface of the SAg streptococcal pyrogenic exotoxin C (SpeC) and identify a functional epitope in the beta-barrel domain that is required for the activation of T cells. Using cell lines that exclusively express individual MHC II isoforms, our studies provide a molecular basis for the selectivity of SpeC-MHC II recognition, and provide one mechanism by how SAgs are capable of distinguishing between different MHC II alleles.


Subject(s)
Bacterial Proteins/immunology , Epitopes/immunology , Exotoxins/immunology , Histocompatibility Antigens Class II/metabolism , Protein Interaction Domains and Motifs/immunology , Bacterial Proteins/metabolism , Cell Line , Epitope Mapping , Exotoxins/metabolism , Humans , Jurkat Cells , Lymphocyte Activation , T-Lymphocytes/immunology
16.
J Mol Biol ; 371(1): 210-21, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17560605

ABSTRACT

Superantigens (SAGs) interact with host immune receptors to induce a massive release of inflammatory cytokines that can lead to toxic shock syndrome and death. Bacterial SAGs can be classified into five distinct evolutionary groups. Group V SAGs are characterized by the alpha3-beta8 loop, a unique approximately 15 amino acid residue extension that is required for optimal T cell activation. Here, we report the X-ray crystal structures of the group V SAG staphylococcal enterotoxin K (SEK) alone and in complex with the TCR hVbeta5.1 domain. SEK adopts a unique TCR binding orientation relative to other SAG-TCR complexes, which results in the alpha3-beta8 loop contacting the apical loop of framework region 4, thereby extending the known TCR recognition site of SAGs. These interactions are absolutely required for TCR binding and T cell activation by SEK, and dictate the TCR Vbeta domain specificity of SEK and other group V SAGs.


Subject(s)
Bacterial Proteins/chemistry , Enterotoxins/chemistry , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Staphylococcus aureus/immunology , Superantigens/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Crystallography, X-Ray , Enterotoxins/immunology , Humans , Models, Molecular , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Signal Transduction/physiology , Superantigens/genetics , Superantigens/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...