Shocking superantigens promote establishment of bacterial infection.

نویسندگان

  • Nikolai Siemens
  • Anna Norrby-Teglund
چکیده

Streptococcus pyogenes, also referred to as group A Streptococcus (GAS), is an exclusive human pathogen causing diseases ranging from uncomplicated infections of the throat and skin to severe invasive infections, such as necrotizing fasciitis and streptococcal toxic shock syndrome (STSS). Asymptomatic carriage, particularly in the nasoand oropharyngeal mucosa, is common. GAS is equipped with an arsenal of virulence factors (e.g., surface-associated and secreted factors that contribute to disease pathogenesis) (1). Among the secreted virulence factors, superantigens (SAgs) have been recognized as key factors mediating the systemic excessive inflammatory response associated with STSS (2). SAgs belong to a family of proteins that activate T cells in an unconventional manner bypassing the normal rules for antigen processing and presentation. SAgs bind without prior cellular processing to the major histocompatibility complex (MHC) class II molecules on antigen-presenting cells and to the variable β-chains of the T-cell receptor on CD4 and CD8 T cells. In this way, the fine MHCpeptide specificity of T cells is bypassed, allowing for activation of numerous Vβ-specific T cells and an overzealous inflammatory response. This cytokine storm is harmful to the host and underlies conditions such as STSS (2). While the role of SAgs in STSS is well established, a lingering question has been why the bacteria secrete such powerful immunostimulatory factors. To date, 14 distinct SAgs have been identified and most clinical strains express several, thus showing a redundancy of these toxins (3). In PNAS, Zeppa et al. (4) provide evidence that SAgs promote GAS nasopharyngeal colonization and infection in a Vβ-specific T-cell–dependent manner. Passive immunization with antibodies against the SAg streptococcal pyrogenic exotoxin A (SpeA) or vaccination with a SpeA toxoid devoid of superantigenic activity provided antibody-dependent protection against nasopharyngeal infection. In addition, the authors discovered an antibody-independent protective mechanism involving SAg-triggered T-cell responses that enhanced GAS infection, thereby offering an explanation as to the bacterial benefit of a SAgtriggered T-cell response (ref. 4; Fig. 1). The concept that SAgs are critical in the establishment of GAS infection was first proposed by Kasper et al. (5), who showed that mice expressing human MHC class II molecules (e.g., mice susceptible to the action of SAgs) were easily colonized by GAS, whereas the wild-type mice readily cleared the infection. The enhanced colonization and infection was attributed to the SAg SpeA. In PNAS, the study by Zeppa et al. (4) provides additional mechanistic insight into the role of SAg as a colonizing factor. First, the authors show that passive immunization of humanized mice with anti-SpeA serum resulted in a reduced bacterial burden in the nasopharynx. Similarly, direct vaccination with inactive SpeA toxoid lacking SAg activity resulted in a strong anti-SpeA humoral response and protection against nasopharyngeal infection. However, vaccination with either SpeA or the staphylococcal SAg staphylococcal enterotoxin B (SEB) also afforded protection against GAS infection despite the absence of anti-SpeA antibodies, hence also suggesting an antibody-independent protective mechanism. The authors then explored whether T-cell responses contributed to the protective effect, and they provide evidence of the involvement of the SpeA-targeted T-cell subset, namely, CD3Vβ8 lymphocytes, which were reduced and less responsive in SpeAor SEB-vaccinated mice. Thus, the data indicate that the T-cell response elicited by the SAg contributed to the noted enhanced bacterial infection. This was further supported by the finding that depletion of either CD8 T cells alone or in combination with CD4 T cells resulted in a reduced nasopharyngeal GAS burden, although an exaggerated bacterial burden for Streptococcus pneumoniae, a non-SAg–producing bacteria. As an additional potential explanation, the authors link the observed effects to the inflammatory state during bacterial infection. Mice lacking CD4 T cells, CD8 T cells, or both showed reduced levels of inflammation during bacterial infection, which was linked to bacterial clearance. Similarly, SpeAand SEBvaccinated mice that were protected against GAS, infection showed a reduced inflammatory response (4). The authors propose that SAgs target and activate Vβspecific T cells, resulting in an inflammatory milieu and

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

Identification of bacterial antigens and super antigens in synovial fluid of patients with arthritis: a cross sectional study

  Background : An accurate and prompt diagnosis of bacterial arthritis is essential for earlier treatment and a good outcome. Superantigens produced by Staph. Aureus are among the most lethal toxins. The paper objective was Identification of common bacterial antigens and S.aureus superantigens in synovial fluid (SF) of children with negative culture and direct smear for other bacteria except fo...

متن کامل

Bacterial Superantigens Promote Acute Nasopharyngeal Infection by Streptococcus pyogenes in a Human MHC Class II-Dependent Manner

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 ...

متن کامل

Recombinant Staphylococcus aureus exfoliative toxins are not bacterial superantigens.

Staphylococcal scalded-skin syndrome is an exfoliative dermatitis characterized by the separation of the epidermis at the stratum granulosum. This disruption is mediated by one of two Staphylococcus aureus exotoxins, exfoliative toxins A and B (ETA and ETB). Both ETA and ETB have been reported to be bacterial superantigens. A controversy exists, however, as other data indicate that these exotox...

متن کامل

Administration of superantigens protects mice from lethal Listeria monocytogenes infection by enhancing cytotoxic T cells.

Superantigens stimulate T-cell-receptor Vbeta-selective T-cell proliferation accompanying the release of cytokines, which may eventually protect the host from microbial infections. We investigated here whether superantigens can rescue the host from lethal bacterial infection. Mice were pretreated with Staphylococcus aureus enterotoxin B (SEB) 1 and 2 days before bacterial infection, and the mor...

متن کامل

Major histocompatibility complex class II-associated peptides control the presentation of bacterial superantigens to T cells

Recent studies have shown that only a subset of major histocompatibility complex (MHC) class II molecules are able to present bacterial superantigens to T cells, leading to the suggestion that class-II associated peptides may influence superantigen presentation. Here, we have assessed the potential role of peptides on superantigen presentation by (a) analyzing the ability of superantigens to bl...

متن کامل

ذخیره در منابع من


  با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

عنوان ژورنال:
  • Proceedings of the National Academy of Sciences of the United States of America

دوره 114 38  شماره 

صفحات  -

تاریخ انتشار 2017