Next Article in Journal
Inflammation and Oxidative Stress in Snakebite Envenomation: A Brief Descriptive Review and Clinical Implications
Next Article in Special Issue
Editorial of the Special Issue “Toxins: Mr Hyde or Dr Jekyll?”
Previous Article in Journal
Detoxification of the Mycotoxin Citrinin by a Manganese Peroxidase from Moniliophthora roreri
Previous Article in Special Issue
Effector-Triggered Trained Immunity: An Innate Immune Memory to Microbial Virulence Factors?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Superantigens, a Paradox of the Immune Response

by
Sofia Noli Truant
,
Daniela María Redolfi
,
María Belén Sarratea
,
Emilio Luis Malchiodi
and
Marisa Mariel Fernández
*
Cátedra de Inmunología—IDEHU (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956 4 Piso, Ciudad Autónoma de Buenos Aires 1113, Argentina
*
Author to whom correspondence should be addressed.
Toxins 2022, 14(11), 800; https://doi.org/10.3390/toxins14110800
Submission received: 14 September 2022 / Revised: 27 October 2022 / Accepted: 27 October 2022 / Published: 18 November 2022
(This article belongs to the Special Issue Toxins: Mr Hyde or Dr Jekyll?)

Abstract

:
Staphylococcal enterotoxins are a wide family of bacterial exotoxins with the capacity to activate as much as 20% of the host T cells, which is why they were called superantigens. Superantigens (SAgs) can cause multiple diseases in humans and cattle, ranging from mild to life-threatening infections. Almost all S. aureus isolates encode at least one of these toxins, though there is no complete knowledge about how their production is triggered. One of the main problems with the available evidence for these toxins is that most studies have been conducted with a few superantigens; however, the resulting characteristics are attributed to the whole group. Although these toxins share homology and a two-domain structure organization, the similarity ratio varies from 20 to 89% among different SAgs, implying wide heterogeneity. Furthermore, every attempt to structurally classify these proteins has failed to answer differential biological functionalities. Taking these concerns into account, it might not be appropriate to extrapolate all the information that is currently available to every staphylococcal SAg. Here, we aimed to gather the available information about all staphylococcal SAgs, considering their functions and pathogenicity, their ability to interact with the immune system as well as their capacity to be used as immunotherapeutic agents, resembling the two faces of Dr. Jekyll and Mr. Hyde.
Key Contribution: Bacterial superantigens manipulate the host immune response favoring the spread and colonization of Staphylococcus aureus, causing multiple affections. This review summarizes the current knowledge on every superantigen described, focusing on the dual nature of these toxins.

1. Introduction

The term superantigen (SAg) was introduced by White et al. (1989) to describe a group of bacterial proteins targeting, as unprocessed molecules, the variable portion of the β chain of the T cell receptor (TCR) and the major histocompatibility complex type II molecules (MHC-II) expressed on antigen-presenting cells (APC). As a result of this simultaneous interaction, there is a massive activation of the immune system along with an intense proliferation of T cells, either CD4+ or CD8+ [1,2]. Although the term superantigen was introduced in 1989, these toxins had been described in the early 1980s as the causative agent of the highly lethal Toxic Shock Syndrome (TSS) associated with tampons [3,4,5], which was later explained by the deregulation of the immune system caused by SAgs characterized by generalized multiple organ failure caused by a pro-inflammatory cytokine storm. SAgs are also responsible for food poisoning and triggering autoimmune diseases in sensitive hosts, among other conditions. They can promote immunosuppression in the infected host, allowing bacterial spread and further sepsis [6,7,8].
SAgs are produced by Gram-positive bacteria such as S. aureus and Streptococcus pyogenes, but they have also been found in other species such as β-hemolytic streptococci, coagulase-negative staphylococci, Gram-negative Yersinia pseudotuberculosis, Pseudomonas fluorescens and cell wall-less bacteria Mycoplasma arthritidis [9,10,11,12]. Furthermore, SAgs encoded by murine and human retroviruses have also been described [13,14,15].
Until now, the best characterized SAgs were produced by S. aureus and S. pyogenes. Although these two bacteria are not phylogenetically related species, staphylococcal and streptococcal SAgs share sequence homology and biological similarities. Some SAgs from these two species are so close that they belong to the same group or family, as is very well described for the SE family or Group II, where the streptococcal superantigen SpeA is more similar to staphylococcal enterotoxin B (SEB) than to staphylococcal enterotoxin G (SEG). SpeA displays a low degree of similarity with the other streptococcal SAgs. This finding supports the horizontal transmission of genes between species. This theory is also reinforced by the encoding of sags in genetic mobile elements, such as plasmids or transposons [16].
Interestingly, the intense immune stimulation induced by SAgs could allow the spread and further colonization of the infected host by the pathogen instead of favoring its eradication. This characteristic would turn superantigen activity into a paradox of the immune response.

2. Staphylococcal SAgs

2.1. An Overall Description

S. aureus is one of the major pathogens responsible for human and veterinarian diseases causing mild to life-threatening infections. Human beings are the primary reservoir for this bacterium. It is well known that about 20–30% of the population persistently carries this microorganism in the anterior nares, while about 50–60% are intermittently colonized [17,18,19,20]. The colonization of other extra nasal sites, such as the skin and gastrointestinal tract [21,22,23,24] has also been reported. It should be mentioned that colonization is considered one of the main risk factors for S. aureus diseases [25].
This pathogen harbors several virulence factors among which some are surface proteins and many others are secreted. Staphylococcal enterotoxins (SEs) or SAgs are some of the most important and are associated with both colonization and pathogenicity of S. aureus.
The first staphylococcal SAgs described were the classical SAgs, including staphylococcal enterotoxins A to E and the non-emetic toxin TSST-1. Due to its strong association with TSS, the toxin formerly called SEF was later named Toxic Shock Syndrome Toxin number 1 or TSST-1 [26,27,28,29,30,31,32,33]. Since that first group was described, 24 more SAgs were identified, and were considered non-classical or new SAgs.
The first non-classical SAg, SEH, was described in 1994 [34]. Later, SElJ was acknowledged [35] at the same time that Munson et al. described two new SEs, G and I [36]. In 2001, Jarraud et al. defined the egc operon for the first time, as an enterotoxin gene nursery encoding for the proteins SEO, SEM, SEI, SEN and SEG (firstly named SEL, SEM, SEI, SEK and SEG) and two pseudogenes, ψ ent1 and ψ ent2 [37]. Moreover, SEK and SEL were simultaneously described [38,39,40]. In the early 2000s, genetic variations within the egc cluster were described, which allowed the identification of SElU and SElV [41,42].
Later on, SEP [43], SES and SET [44], SEQ [45], SER [46], SElW [47], SElX [48] SElY [49] and SElZ [50] were described. Lastly, SE01 [51], SE02 [52], and SEl26 and SEl27 were identified and characterized among the complex form by S. aureus, S. argenteus and S. schweitzeri [53]. Table 1 summarizes the information described above. Phylogenetic group correspondence is explained below in Section 2.4.
According to the International Nomenclature for Staphylococcal Superantigens, S. aureus enterotoxins (SEs) are defined by their emetic ability when ingested, while the term SAg considers the effects on the immune system. Taking that into account, only the toxins that induce emesis after oral administration in a primate model are designated as SEs. Other related toxins that either lack emetic properties in this model or have not been tested are defined as “staphylococcal enterotoxin–like” (SEl) SAgs, to indicate that their potential role in staphylococcal food poisoning has not been confirmed [54].
To date, TSST-1 and SElJ are the only tested superantigens with non-emetic properties, along with a report of a truncated SElX [55].
Furthermore, some of the so-called SEl and SE proteins have already been tested for their emetic properties in a proposed model using house musk shrews (suncus murinus) [56,57]. In this model, in addition to all the other SEs, SElY has been tested and shown to have emetic properties [44,49,56,58,59]. Recently, a new emetic animal model was established using common marmosets and, in this case, SEA, SEB, SEC, SEI, SElY and SE02 showed emesis-inducing activity, while TSST-1 did not [52,60].
It has been shown that almost every isolated strain of S. aureus carries at least one sag gene in its genome [61,62,63,64]. Although there are some differences in the percentages of total gene prevalence reported, a lower prevalence of sag genes (70–85%) was found when a few sag genes were analyzed [61,65], whereas in other studies evaluating a larger number of genes, the percentages were almost 98% [66,67]. Noteworthy, as mentioned before, 30 SAgs have been hitherto described, which suggests some underestimation of SAg prevalence. Additionally, more than 50% of the isolates encode for the egc operon—with eight different forms described—being that the egc SAgs are the most prevalent genes [37,62,65,68,69,70,71]. However, there are some differences in the methodology used to define the presence of the operon; while some papers considered all egc SAgs, others picked two (such as seg and sei or sem and seo) without considering the operon variants, which in some cases may underestimate the prevalence of the operon itself. With regard to this issue, a consensus method should be established to assess the presence of the egc operon, which should definitely consider all the toxins involved in the operon, taking into account possible DNA mutations in their genes.

2.2. Production and Detection of Staphylococcal SAgs

Although there are some statements about the production of almost every SAg, in addition to the 6 classical ones (SEA-SEE and TSST-1), there is not much information regarding the individual gene expression and the mechanism of regulation involved in their production. However, many factors and pathways have been described to impact positively and negatively in the production of SAgs and other virulence factors [72,73,74].
Considering the classical SAgs, early reports using mainly single or double-gel diffusion indicated that SEB and SEA are produced in all the growing phases [75,76,77,78]. In contrast, SEC seems to be produced during the exponential growth phase or at the early stationary phase [79,80]. In regard to SED, mRNA expression appears to be higher in the stationary growth phase using qPCR [81], and evidence of its production with SEE at the stationary phase has also been provided [82].
With respect to TSST-1, although its production occurs principally at the stationary phase, it was found that several environmental elements are capable of triggering or inhibiting its production, including temperature, carbon dioxide atmosphere and antibiotic pressure [16,83,84,85].
Many authors rely on the conclusions reported by Derzelle and collaborators to describe the patterns of staphylococcal enterotoxins expression [86]. These authors found a differential expression behavior between classical and new SAgs. Their work is limited to mRNA, as they performed RT-qPCR assays; however, protein secretion was not evaluated. Furthermore, although their analysis was performed in a total of 28 strains, the number of strains used per gene was variable and dependable on the sag gene profile of each strain, between one and thirteen. In the case of the egc operon, only seg and seu were evaluated in all egc positive strains (thirteen), and the remaining genes (sei, sen, sem and seo) were only evaluated in three strains. Therefore, these results should be validated using other strategies for reliable conclusions. A similar analysis for SE02 showed the production of this SAg in vitro in the early exponential phase and to a lesser extent in the stationary phase, with these results confirmed by Western blotting [52].
In vitro assays indicate that egc superantigens are produced at much lower concentrations than classical SAgs TSST-1, SEB and SEC, which may explain the relevance of these toxins in pathogenesis [87,88,89]. Particularly, it was proved that TSST-1 was produced in a high level in biofilms, which could be transferred to production on mucosal and skin surfaces [89].
Some evidence suggests that the regulation of gene expression is not the same for S. aureus in vitro than during infection in vivo [90,91], showing differences in SAg expression even between tissues or isolation sources [82,92]. As a result, there is a different SAg expression despite the evaluation of DNAs encoding the same sag genes. In addition, it has been proposed that the detection of SAg mRNA does not correlate with the presence of toxins [93].
Proteomics approaches confirmed the production of some toxins such as SEB, SEC, SEH, SEK and SEQ both in exponential and stationary phases, and TTST-1, SEL, SElU and SEP in the stationary phase [84,94,95,96], but the whole exo-proteome was analyzed in vitro; therefore, relevance in vivo has not been clarified [97]. In addition, a wide heterogeneity between the proteins identified amongst the different isolates analyzed in vitro [98,99] has been reported.
Although PCR is the chosen method to identify the presence of sag genes in bacterial DNA, it is not effective to evaluate the presence of SAgs in patients, food or in vitro-generated samples [100,101,102].
At present, ELISA-based methods are considered the best option for the detection and quantification of SEs/SEls in several samples because of their robust and sensitive results, with reported detection limits between 0.25 and 1 ng of SAgs per g of sample [102]. However, there are no available ELISA kits for each of the 30 SAgs identified, which complicates the evaluation of the full profile of SAgs potentially produced by S. aureus. An approach based on a combination of chromatography and MS measures simultaneously a group of toxins; however, it is not easy to conduct and does not cover all existing SAgs [103].
In addition, Surface Plasmon Resonance (SPR) was also postulated as a method to detect SAgs in different biological samples, showing a limit of detection in the picomolar range, with the advantage to measure simultaneously three or more SAgs present in the same assay depending on the biosensor used [104,105,106,107,108,109].
All in all, each method has its advantages and disadvantages and there is no ideal technique to assess the production of all toxins; however, it is important to bear in mind the limitations of the method used at the time of reaching conclusions.

2.3. Superantigens and Human Diseases

2.3.1. Toxic Shock Syndrome

Superantigens have been associated with many illnesses caused by S. aureus infections. One of the most well-recognized diseases is Toxic Shock Syndrome (TSS), a potentially lethal febrile illness related to multiorgan dysfunction that occurs as a consequence of the cytokine storm produced by SAgs. TTS is characterized by hypotension; diarrhea; labored breathing; and changes in heart, liver and kidney function [110,111,112]. This pathology has a high incidence in women during their menstrual periods, and is particularly called menstrual TSS (mTSS) [112]. TSST-1 is associated with all menstrual cases, in some instances, coproduced with SEA or SEC-1; conversely, SEB production has been negatively related to this syndrome [111,113,114,115,116]. Regarding non-classical SAgs, Jarraud and collaborators suggested that the production of SEG and SEI by S. aureus strains that do not produce TSST-1, SEA to SEE and SEH and isolated from mTSS patients may explain these clinical manifestations [117].
Studies in porcine vaginal tissue have proved that, initially, the presence of S. aureus triggers the innate immune system activation and increases TSST-1 flux across the mucosa [118]. Furthermore, it has been demonstrated that TSST-1 is essential to stimulate systemic inflammation by inducing the production of IFN-γ and suppressing autophagy [119,120].
TSST-1 biochemical and biological properties have been studied, and among them, its capacity to interact with CD40 on epithelial cells. This interaction promotes chemokine production and attracts other components of the adaptive immune response. Consequently, local inflammation occurs and provokes mucosal disruption, facilitating TSST-1 penetration and enhancing mTSS [121].
Moreover, mTSS progress has been associated with oxygen introduction into the anaerobic vagina. This phenomenon was related to the use of medical devices to control menstrual flow, especially certain tampons whose composition enhanced their ability to trap oxygen within its fibers. Remarkably, device insertion does not cause significant vaginal oxygenation. In addition to the introduction of oxygen, the increase in body temperature, as well as protein, low glucose levels and neutral pH generate an environment that is favorable for TSST-1 production. In addition, the device’s capability to increase the permeability of the mucosa promotes SAg penetration [114,122,123,124,125].
Moreover, it has been reported that tampon wearing time influences the development of mTSS [122]. Some studies propose that the tampon material also affects S. aureus proliferation and TSST-1 production, while others suggest that the tampon structure and fiber density have a higher impact on colonization and toxin production than the nature of its material [124,126]. It has been described that the application of menstrual cups could promote S. aureus growth and biofilm formation with toxin production. Although those conditions are affected by cup composition, toxin production induced by cup use is lower than that caused by tampon use. This fact has to be related to its incapacity to trap oxygen [124,125].
Non-menstrual TSS has also been associated with TSST-1 and other classical superantigens such as SEB, SEC1 and SEA [61,113,114,115,127,128,129]. A few studies suggest that SEB production is prevalent in S. aureus isolates that are negative for TSST-1 in non-menstrual TSS cases [113,129]. Furthermore, in case-control investigations, it was demonstrated that sea gene is correlated with TSS, whereas the presence of sem and seo genes is correlated with sepsis [61].

2.3.2. Infection Endocarditis

Infection endocarditis (IE) is a disease of the endocardium and cardiac valves, that causes cardiac and multi-organic symptoms usually as a consequence of S. aureus bacteremia [130,131]. Classical superantigens, such as TSST-1, SEC, SEB, SEA, SED and SEE are related to IE, as well as non-classical, from the egc operon [87,132,133,134,135]. It has been shown that TSST-1 and egc operon gene deletion raised mortality in a rabbit model. Many studies have associated disease development with vegetation formation in aortic valves, whose formation decreases with the deletion of egc, SEC and TSST-1 genes [87,133].
Another action mechanism that could influence the inflammation process is the secretion of IL-8 by aortic endothelial cells induced by SEC [134]. In addition, it has been demonstrated that SEC inhibits the pro-angiogenic factor serpin E1 impeding reendothelialization and promoting disease [133]. On the other hand, TSST-1 also acts on the endothelium, producing upregulation of vascular and intercellular adhesion molecules (VCAM-1 and ICAM-1), raising permeability and affecting tissue re-composition [135].

2.3.3. Pneumonia

Pneumonia is an infectious respiratory disease frequently related to S. aureus infections in both community and hospital settings [136]. Clinical features include cough, fever, tachypnea, tachycardia and pulmonary crackles, leading to severe pulmonary infections [137,138]. Staphylococcal pneumonia caused by methicillin-resistant Staphylococcus aureus (MRSA) has been of growing concern due to its resistance to several antibiotics and its higher risk of morbidity and mortality [139,140]. It has been observed that several isolates obtained from patients with lung damage produced TSST-1, SEB and SEC in vitro [141,142]. Spauding et al. [33] proposed that SAgs contribute to pneumonia development once they are produced by S. aureus in the respiratory tract, generating an intrapulmonary inflammation. In addition, the cytotoxic effect on endothelial cells of the alveolar–capillary barrier may cause edema and respiratory distress. Furthermore, T cell activation and the release of cytokines may contribute to the pathogenesis and would be associated with the clinical symptoms [143,144]. However, there is no solid evidence proving that SAgs are the cause of pneumonia in humans.

2.3.4. Staphylococcal Food Poisoning

Staphylococcal food poisoning (SFP) is a foodborne disease that provokes abdominal pain, intense diarrhea and vomiting [2]. This pathology has been related to many SAgs in investigations from all over the world. The presence of classical SAg genes, such as sea, seb, sec, sed and see, in S. aureus isolates from dairy products, animals and infected patients has been associated with SFP development [101,145,146,147,148,149,150]. In those samples, not only classical SAg genes, but also egc genes such as sei, seg, sem, sen, seo and seu have been detected [101,145,147,148,150,151,152,153]. Moreover, other non-classical SAg genes, such as seh, sej, sek, sep, seq and ser, have been identified in isolations related to SFP [101,145,147,149,150,154,155,156,157].
While genetic studies and genomic patterns are relevant to the study of this illness, the confirmation of the physical presence of toxins in food products suspected of contamination or an analysis of isolation capability of expressing SEs is needed to verify their contribution to SFP. Notably, some isolations from SFP patients and milk samples containing seg and sei genes did not produce detectable levels of SEG and SEI. On the other hand, isolations harboring seh were able to produce significant levels of this SAg. Moreover, some positive isolations for sea, seb and sec did not express those toxins in vitro [93,158]. Other studies showed the presence of SEC, SED, SEA, SEB and SEE in samples related to SFP cases and their expression in isolates from sick people or food [159]. SEH and SEA expressions were also shown in isolates from foodstuffs causing poisoning events [154,155]. SEI and SEM production in isolates associated with food poisoning was also corroborated in several cases [160].
A variable distribution of S. aureus genotypes and SEs expression has been shown worldwide, suggesting that the distribution of SAg genotypes may be related to geographical distribution [156,161].
Some mechanisms of action for vomiting and diarrhea have been proposed in several animal models. Many studies suggested that enterotoxins stimulate the vagus nerve in the gut, which transmits the signal to the vomiting center in the brain [162,163]. Supporting this idea, a few studies have shown that SEA increases 5-hydroxytryptamine (5-HT) and histamine release in the intestinal tract, which would increase the discharge of vagal afferent fibers and trigger an emetic effect; it was also shown that this SAg needs vagal 5-HT3 receptors to stimulate vomiting [60,162]. Additionally, it has been described that some SAgs, such as SEA, SEB, SEE and TSST-1, can penetrate the gut lining. Interestingly, it is proposed that transcytosis is mediated by an amino acid sequence highly conserved across superantigens [164]. After transcytosis, the activation of the local immune system occurs. Using a mice model, it was shown that SEB administration resulted in an early expansion of peripheral T cells and Th1 cytokine secretion [165], and that CD4+ T cell stimulation by SEB provoked a disruption of the jejune tonic and stimulated ion transport, creating water movement and contributing to a diarrheal response [166]. Furthermore, SEA and SEB could interact with the MH C II of human subepithelial intestinal myofibroblasts, triggering the secretion of pro-inflammatory cytokines. These effects may play a role in inflammatory injury [167].

2.3.5. Autoimmune Pathologies

SAgs have been correlated with the onset of autoimmune pathologies [168,169]. SAgs may activate autoreactive T and B cells. In addition, APC activation may lead to alterations in antigen processing, with the production and presentation of autoantigens as a consequence [170].

Kawasaki Disease

Kawasaki disease (KD) is an autoimmune, febrile and inflammatory syndrome that involves coronary vasculitis. It is reported that this illness may have different etiologies such as viral and bacterial infections, imbalance in immune response and genetic factors [171,172,173,174].
Although, individuals that develop KD may have a genetic predisposition, some theories propose that SAgs could be implicated in massive immune stimulation, activating T cells, B cells and macrophages, and promoting a considerable pro-inflammatory cytokine secretion, allowing vasculitis development [173,174]. A proposed mechanism of action suggests that superantigens, secreted by S. aureus in the gut microbiota, may penetrate the mucosal membrane and activate Th1, T cytotoxic and B cells, generating autoreactive cells capable of enhancing illness development [173,175].
SAgs secretion by S. aureus isolations from patients with KD has been demonstrated in several cases [176,177,178]. Moreover, IgM antibodies against TSST-1, SEA, SEB and SEC were detected in KD patients’ sera, and titers were significantly higher than in the control group [179,180].
In contrast, some authors state that there is no evidence of SAg involvement in the pathogenesis of KD, or that it remains unclear [171,181,182].

Diabetes Mellitus

Some animal models and in vitro experiments suggest that SAgs could have a role in diabetes mellitus II (DMII) progression. Chronic exposure to sublethal doses of TSST-1 in a rabbit model showed that this SAg induced glucose tolerance in vivo and systemic inflammation. In addition, SAg-treated adipocytes produced a higher level of pro-inflammatory cytokines (IL-6, IL-8 and TNF) in comparison to the control group. Moreover, TSST-1 induced lipolysis and insulin resistance in adipocytes. In addition, the liver damage shown in the TSST-1 group could lead to an increase in circulating endotoxin levels, which could impact the development of insulin resistance in DMII. Altogether, it was postulated that SAgs may promote insulin resistance and systemic inflammation, leading to disease worsening [183].
Other investigations on adipocyte metabolism showed a possible relationship between SAgs and insulin resistance and cytokine production. A study showed that SEA interacts with the human cytokine receptor gp130, which is present ubiquitously in human cells, and particularly in adipocytes, and consequently reduces insulin signaling and modulates glucose uptake. Binding was mediated by a structure that is well-conserved in several other SAgs [184]. In addition, it was demonstrated that egc SAgs, SEI, SEG, SEM and SEO could bind gp130, by SPR [62]. Furthermore, the effect of TSST-1 and SEB in adipocyte cytokine production was analyzed, showing that they provoke IL-6 and IL-8 secretion, which could contribute to inflammation and diabetes persistence [185].
Despite the previous reports, there are no significant studies in humans that demonstrate that there is a significant correlation between DMII development and chronic SAg exposure; thus, further investigations should be conducted.

Rheumatoid Arthritis

Rheumatoid arthritis (RA) is an autoimmune disease characterized by inflammation and infiltration of synovial tissues that can lead to important damage in articulations [186]. It has been suggested that SAgs could trigger the disease by activating autoreactive T cells, could have cross-reactivity with self-antigens and may contribute to long-term inflammation [186,187].
Numerous studies in RA patients demonstrated the presence of SEC, SEA and SED and their genes in blood samples, synovial fluids and DNA extracts from those samples [188,189,190]. The see gene was present in several samples of synovial fluid of RA patients, but the expression of the toxin was not analyzed [191]. SEA, SEB, SEC and TSST-1 were also identified in the synovial fluids of a group of pediatric RA patients [192]. All these studies have a limitation in common: they did not assess other SAgs.
In addition, some studies found antibodies against SEB and TSST-1, with significantly higher titers in RA patients than in healthy groups [193,194].
Several animal models were used to investigate the correlation between SAgs and RA development and, as mentioned earlier, it was proposed that the inflammatory response and the reactive cells generated by SAgs play an important role in the development of the illness and its reactivations and severity [195,196,197,198,199]. It was suggested that pro-inflammatory cytokine secretion induced by SAgs may enhance the expression of MHC II in synoviocytes, promoting the release of chemotactic factors and cell infiltration. In addition, SEA and SEB significantly increase the proliferation of T cells in co-culture with synoviocytes from RA patients, compared to the control group. These results suggest that the interaction between both cells and SAgs may be important in disease pathogenicity [200].

Atopic Dermatitis

Atopic dermatitis (AD) is a pruritic skin disorder that presents with eczema, inflammation and immune cell infiltration into the local skin lesion [201]. Chronic exposure to SAgs may be a factor that promotes disease incidence, as S. aureus colonization is usual in AD patients [202,203]. It is proposed that the disease is caused by inflammatory and allergic factors, and SAgs may be implicated in both. On the one hand, SAgs can activate T cells promoting pro-inflammatory cytokine release and contributing to skin injury, enhancing the Th1 profile. On the other hand, SAgs may activate B cells promoting SAg-specific IgE which can stimulate basophile and mast cell degranulation, leading to a Th2 response [204].
The relationship between SAgs and AD has been based on several factors, such as the presence of anti-SAgs IgE in sera from patients with AD. Anti-SEA, SEB and TSST-1 IgE has been documented in several AD patients with significantly higher titers than in the control groups [201,205,206,207]. Moreover, a total IgE increase has been positively correlated with a SAg-specific IgE increment in patient sera, and it was demonstrated that SAg-specific IgE titers increased with the severity of the condition [206].
In addition, SEA, SEB and TSST-1 provoked a SAg-specific histamine release by basophils from AD patients after being sensitized with IgE-SAg specific serum, in contrast to the control group. Notably, basophil samples from patients lacking these antibodies did not secrete histamine. These results showed that SAgs and their interaction with basophils mediated by IgE could exacerbate AD, promoting an allergic response [201].
Another important fact, which relates SAgs and AD development, is the production of SAgs by S. aureus isolated from patients. Compared with control groups, these isolates had a significantly higher production of SAgs and greater heterogeneity of encoded genes. Moreover, they were more likely to produce TSST-1, SEB and SEC [208]. In addition, S. aureus isolated from AD patients produced TSST-1, SEA, SEB, SEC and SED [201,209,210].
SAgs are able to induce these pathologies due to their ability to promote a pro-inflammatory environment by upregulating co-stimulatory molecules, which are usually expressed at low levels, preventing the stimulation of autoreactive lymphocytes. Another important point to note is the globular and compact structure shared by these toxins, which is the key to remaining as a whole and active molecule during the passage through the lysosome vesicle and reaching again the dendritic cell surface in the lymph nodes, where they are able to activate different lymphocyte T populations by the crosslinking of the TCRs in association with the MHC-II molecules and other recently described molecular targets, such as CD28 and B7 [211,212,213,214]. In addition, their ability to interact with diverse molecular targets in different types of cells opens up a new avenue to study their functions in new pathologies.

2.4. Staphylococcal Classification, Structure and Molecular Targets

Staphylococcal SAgs are homolog proteins that share from 20 to 89% similarity in their amino acid sequence (Supplementary Table S1). Based on their sequence of nucleotides and amino acids, SAgs were separated into five evolutive groups or families: Group I, which includes TSST-1, SET, SElX, SElY and members of the superantigen-like toxins; Group II, including SEB, SECs, SEG, SER, SElU, SElW, SElZ, SEl27 and SE02; Group III, comprising SEA, SED, SEE, SElJ, SEH, SEN, SEO, SEP, SES and SE01; Group V, gathering SEI, SEK, SEL, SEQ, SEM SElV and the recently described SEl26. Group IV is represented by another group of toxins produced by S. pyogenes [33] (Table 1).
Staphylococcal and streptococcal superantigens share a common three-dimensional arrangement comprising an N-terminal β-barrel domain (OB-fold) and a C-terminal β-grasp domain with β-sheets that wrap around a long α-helix. TSST-1 is the only SE described that is less related to the other SEs from a structural point of view. Despite this fact, the basic tertiary structure present in TSST-1 is also found in the other SEs, even though these toxins have more structural complexity. This overall structure is represented in Figure 1. The loop known as the disulfide loop is located between the β4 and β5 strands. Due to its flexibility, no electron density from the crystallographic data is recorded. Experimental data suggest that this loop is responsible for the emetic properties of the SEs. The direct mutagenesis of the residues involved in the disulfide loop eliminates the emetic capacity in SEC1 [215,216].
The major and best-described molecular targets of SAgs are the MHC-II molecules and the TCR. Different regions of the toxins participate in the interaction with these targets. The interaction with the MHC-II is well documented for SAgs of different groups. SAgs mostly interact with the DR isoform of the MHC-II molecule, showing less interaction with DQ and DP. SAgs–MHC-II structures have been already solved, showing the binding surface between the SAg and the DR1 isoform of the MHC-II molecule (Figure 2).
SAgs can interact with the α and/or β chain of the MHC-II molecule, alone or in a complex with a peptide. The interaction with the polymorphic β chain is characterized by strong affinity which is mediated by zinc. SAgs involve highly conserved His and Asp residues, in positions 207 and 209, respectively, to establish coordination with this metal. As Zn2+ needs four atoms to complete the interaction, the interaction could involve one more amino acid in position 169, mainly a His or Asp, and the β chain completes the interaction with a His in position 81. SEH, which also displays this kind of interaction with the MHC-II molecule, only contributes with two residues to the union. A water molecule contributes to completing the coordination required by Zn2+. All the studied SAgs that contact the MHC-II β chain also contact the bound peptide, which provides more specificity to the binding. On the contrary, the interaction with the invariant α chain usually displays low affinity. SAgs such as SEB and SEC3 that bind to the low-affinity site on the MHC-II α-domain can recognize multiple HLA types (DR, DQ, DP). This is easily explained because the DR α-chain is nonpolymorphic and the SAg makes no contact with the bound peptide [217,218,219].
The interaction with the TCR is very well studied for Group II SAgs. The SE members of this group, SEB, SEC1-3 and SEG are crystallized in complex with the murine Vβ8.2 chain. In addition, the streptococcal SAg, SpeA, which also belongs to this group, is crystallized in complex with this TCR. All the SAgs of this group involve the Asn 25 (SEB numbering) or Asn 24 (SEG numbering) as the hottest spot to interact with the TCR Vβ chain [220]. The SAgs contact the region CDR2, FR2 and 3 and the hypervariable region HV4 of the TCR, except SEG, which does not bind HV4. Despite this fact, SEG shows the highest affinity to the TCR already documented by wild-type SAg. SEG interacts with mVβ8.2 with an affinity in the nanomolar range (500 nM). An analysis of the mVβ8.2-SEG interface elucidates the higher affinity of this complex compared to others as it establishes the highest number of hydrogen bonds in the smallest binding surface [221] (Figure 3). This phenomenon may at least partially explain the early activation of T cells bearing mVβ8.2 by SEG compared to other members of Group II. SEG induces the increase in Vβ8.1+2 T cells in mice 48 h after inoculation, followed by the apoptosis of this cell population. The other members of this group cause this same effect at 96 h, showing a temporary delay compared with SEG biological function. Considering this early immunosuppression, SEG-mVβ8.2 interaction could be advantageous to the pathogen as it may facilitate the colonization of the host [222].
In 2010, Saline et al. solved the crystal structure of the ternary complex between the human MHC class II molecule DR1 loaded with the Influenza hemagglutinin (HA) (306–318) peptide (PKYVKQNTLKLAT), the human TCR (JM22:TRAV27/TRBV19) and the enterotoxin SEH at 2.3 Å of the resolution. The structure reveals that SEH mostly contacts the variable portion of the TCR α chain and only 6% of the variable portion of the β chain. Until now, SEH was the only SE described which presents this kind of binding [223].
With the aim to investigate if other staphylococcal enterotoxins interacting with the DR1 β chain and displaying a similar orientation than SEH over the MHC-II molecule could contact the TCR α chain, we superimposed the structure of SEI-DR1 over the structure of the ternary complex using the DR1 β chain as the template. SEI and SEH show a displacement that puts SEI away from the sphere of the interaction with the variable portion of the TCR α chain (Figure 3). Furthermore, a sequence analysis that took into account all the staphylococcal enterotoxins contacting the DR1 β chain [219,223] shows that none of them conserve the crucial residues to contact the variable portion of the TCR α chain, nor hydrogen bonds or Van der Waals contacts. Considering all the available structural information, SEH would be the only described SE that presents that kind of binding.
The interaction between staphylococcal SAgs and other molecular targets, such as CD1, B7, CD28 and gp130, is well documented. Even though these interactions were not tested in all the staphylococcal SAgs, the conserved residues in their sequence strongly suggest that all of them could bind these molecular targets. Nevertheless, more studies should be conducted to determine the physiological consequences of these bindings. Kaempfer et al. reported in deep detail the interaction between SAgs and B7/CD28 molecules, and as a consequence of this binding, SAgs would improve the contact between B7-2 and CD28, inducing T cell hyperactivation [211,212,213,224].
The interaction between CD1 and staphylococcal SAgs was described by Gregory and collaborators in 2000, and the engagement between SAgs and the CD1a isoform on the monocyte surface disturbs the intracellular calcium flux, altering the biological functions of the target cell [225].
The new molecular targets described increase the cell populations affected by SAgs, implying direct effects outside the immune system.
Latterly, the interaction between gp130, the interleukin 6 receptor and SEA was described by Banke [184]. Gp130 binds SEA with medium affinity inducing phosphorylation of STAT3 in adipocytes. They suggested that an Asp in position 227 would be necessary to stabilize the interaction. Nonetheless, more recently, Noli Truant and collaborators demonstrated that non-classical SAgs lacking Asp227 also interact with gp130 with micromolar affinity [62]. Later, it was shown that SEE can bind human gp130 with a similar affinity to SEA, whereas SEH displays a ten-fold lower affinity [226].
In recent years, Schlievert and collaborators showed that TSST-1, SEB and SEC induce pro-inflammatory chemokine production from human vaginal epithelial cells, being that TSST-1 is much more potent than the others. In addition, using CRISPR-Cas9 knockout CD40 cells, it was shown that this receptor is essential for the chemokine response and that the specificity of TSST-1 for menstrual TSS is in part dependent on the higher-affinity interaction with CD40 than other SAgs, such as SEB and SEC. Furthermore, CD40 seems crucial for the disruption of the human vaginal epithelial barrier by pathogens such as S. aureus and might be a potential therapeutic target for drug development [227,228,229].

2.5. Actions of SAgs on the Immune System Cells

Conventional immunity mediated by T cells is given by the interaction of an αβ TCR and a complex MHC II-peptide [230,231]. If the TCR recognizes specifically the foreign antigen, the interactions will trigger signaling pathways that will result in the proliferation and differentiation of several clones of T cells [232]. As TCRs are highly diverse molecules, only ~0.01% of the näive T cells will be capable of recognizing a particular antigen [233].
T cell activation by SAgs is distinctive from conventional T cell activation both qualitatively and quantitatively [234]. Given that the characteristic feature of SAgs is their ability to activate T cells in a variable beta chain-dependent manner [2], between 5 and 20% of the total T cell population is activated as a consequence of SAg exposure. TCR diversity relies on the CD3 loops due to the V (D) J recombination during T cell development; however, there is a limited number of Vβ possible regions of the TCR, around 50 in humans, and therefore, SAgs can activate many more T cells than conventional antigens. In addition, SAgs can act in the order of picograms per mL [235], inducing a CPA-dependent production of cytokines by T cells characterized by a Th1/Th17 profile [16].
Furthermore, T cell anergy, a phenomenon whereby T cells stop responding to stimuli, has been proposed as an S. aureus immune evasion tactic. While the first encounter with classical SAgs induces a rapid clonal expansion of CD4+ and CD8+ T cells with a strong production of Th1/Th17 cytokines, repeated SEA or SEB challenges transduce a hyporesponsive state characterized by T cell deletions and anergy in the remaining SEA/SEB-reactive T cells [236,237]. As a consequence of repeated exposure to SEA, SEB and some streptococcal SAgs, there is an induction of CD25+ FOXP3+ CD4+ T cells Vβ specific with a regulatory profile that expresses IL-10 [238,239] as well as a promotion of CD4+ CD25- T cells [240]. This anergy has been attributed at least in part to CD8+ regulatory suppressive T cells in the case of SEA, SEB, TSST-1 and SEC, and may depend on the concentration of SAgs used to stimulate T cells [241,242,243]. Nonetheless, these regulatory T cells appear to turn ineffective in the massive inflammatory context of TSS [244].
Using SPEA, Sahr and collaborators showed that SAg-stimulated APCs induce pro-inflammatory responses but also promote the initiation of co-inhibitory circuits such as anti-inflammatory cytokines (IL-10), co-inhibitory molecules (PD-L1) and the induction of inhibitory effector programs (IDO) [245]. However, they performed all the assays using only one streptococcal SAg, which seems not enough to generalize for all SAgs.
No reports of T cell anergy using non classical SAgs were found to date; nevertheless, almost every publication using one or two classical staphylococcal SAgs concluded above all SAgs, which may not be true for each of the 30 toxins identified. Although they all are likely to induce anergy, considering that all SAgs stimulate T cells with low specificity, confirmation is required.
Although it is still unclear how exactly anergy occurs, it is clear that not only T cells participate in the reported immunosuppression but also APCs and other immune cells involving several mechanisms, such as anergy, deletion of effector T cells, development of Tregs and induction of tolerogenic profiles in different cell types that should be addressed.
While the superantigenic and enterotoxic effects of SAgs are the most studied mechanisms of the pathogenicity of these toxins, several new works have shown that SAgs have additional capacities.
Dendritic cells (DCs) can uptake SAgs, without inducing maturation, having the ability to maintain themselves as biologically active inside this cell type. Ganem and collaborators reported that DCs effectively incorporated SAgs in a dose-dependent manner by macropinocytosis, retaining unmodified SAgs in endosomes to finally release them to the extracellular medium [214]. Moreover, it was demonstrated that solely the uptake of SEG, SEI, SSA, SEB and SEA did not induce the maturation of DCs [246]. Concerning the retention of biological activity, in this study, SEG conserved its three-dimensional structure since it was recognized by polyclonal antisera during the entire uptake process, inclusion in diverse vesicles and exposure on the membrane, in addition to being able to stimulate T cells in vivo and in vitro. This strongly suggests that SAgs traffic through DCs in intact form. This is not surprising, given that SAgs are known to be very stable and highly resistant to proteases and can resist temperatures of 60 °C or higher, as well as extreme pHs. Earlier studies showed that DCs are more efficient than other APCs, such as B cells or monocytes, to initiate T cell proliferation by presenting TSST-1, SEA, SEB and SEE. Results suggested that picomolar levels of these SAgs are required on DCs to maximally stimulate T cells. This would be related to the high amount of MHC II expressed on DC surface [247].
An ex vivo study in mice showed that DC maturation and migration induced by SEB and TSST-1 require T cell activation. Furthermore, it was evidenced that SEB is also able to upregulate MHC II, CD40, CD205 and CD86 markers [248]. Human DC capability to upregulate MHC II, CD86, CD80, CD83 and CD54 in the presence of SEB was also demonstrated. In contrast with previous data, Coutant et al. found no effect on CD40 expression. An increase in the production of TNF-α by human DCs induced by SEB was shown, compared to other compounds such as LPS. Enhancement of IL-12p270 human DC production by SEB remains controversial because some suggest that it occurs, and others affirm that it does not affect its production [249,250].
As mentioned before, SAgs interact simultaneously with TCR on T cells and MHC II on APC and they have to be in close contact, which occurs in the lymphoid tissues. It was shown in mice DCs that they can internalize SEG, SEI and SSA as biologically active molecules and recycle them into the cell membrane. This fact does not cause DC maturation. SAgs were found intact in the acidic cells compartment and remained active. These results suggest that intracellular trafficking of SAgs in DCs improves their local concentration and promotes their encounter with T cells in lymph tissues [214].
SAgs induce cytokine production in APCs, which promote Th1/Th17 profiles [16]. However, an in vitro study in human DCs suggested that SEB may induce T cell immunoglobulin mucin domain (TIM) 4 expression in these cells when it is processed via TLR2 and NOD1 pathways. The interaction between TIM4 in APCs and TIM1 in CD4+ naïve T cells induces a Th2 profile [250]. These results agree with the idea that SAgs may have diverse effects on immune system cells depending on the interaction pathway.
Plasmacytoid dendritic cells (pDCs) are another innate cell subpopulation that may interact with SAgs. An assay in mice demonstrated that SEA increases the number of pDCs in lymphoid organs and promotes the expression of CD86 and CD40. Furthermore, pDCs would strictly require the presence of IFN-γ, in addition to the interaction with T cells, for maturation. In contrast, DCs do not depend on this cytokine for its activation by SAgs [251].
It has been reported that the interaction of SEB with monocytes enriched from PBMCs induced cell death and IFN-γ production [252]; however, the presence of the remaining T cells, a possible source of IFN-γ, cannot be discarded in those studies. Furthermore, egc SAgs inhibited monocytic proliferation in a dose-dependent manner, promoting cell death by apoptosis, and to a lesser extent, by necrosis. The significant production of TNF-α, IL-6 and IL-12 but not of IFN-γ was detected in this scenario [62]. Only SEI induced early apoptosis at the times assayed, similar to SEB that would induce TNF-α dependent apoptosis in THP-1 cells (a cytokine that the classical SAg induces to a greater extent than egc SAgs [253]), suggesting that other intracellular pathways might be involved in the death processes promoted by different SAgs. In concordance, Ulett and Anderson [254] proposed that death pathways could vary not only between different cell types but considering the toxin and the molecular conditions of the stimuli. Furthermore, the absence of CD4+ T cells in the cultures of THP-1 may explain the lack of IFN-γ production [255].
Macrophagic cells obtained by PMA-differentiated THP-1 cells inhibited their proliferation and produced pro-inflammatory cytokines in response to egc SAgs. In addition, SEI induced significant cell death by apoptosis in this cell type, while SEG, SEM and SEO induced death by apoptosis as well as by necrosis. SEB is the only classical SAg reported to induce apoptosis in THP-1 cells PMA-differentiated to macrophages by caspases 3 and 8 [252], but this does not necessarily explain the death mechanism of egc SAgs. All these results not only demonstrate that damage mechanisms are variable according to each toxin, between other factors, but that the response triggered by different SAgs simultaneously may be more diverse than expected.
Despite visible differences, several studies have shown evidence that immune activation generated by classical and new SAgs is similar [256], inducing an intense profile of the Th1/Th17 immune response [235,257,258,259,260].
Although Dauwalder (2006) suggested that egc SAgs would have a different inflammatory potential than classical SAgs, which may explain the different severity of the symptoms caused by S. aureus carriers of one or another type of SAg, this has not been confirmed by other authors. Possibly, their results could be attributed to differences in the production or the purification of SAgs, considering that they used a purchased SEA and a lab-produced SEG, or even to the concentrations used. The most accepted hypothesis is that the differences between the effects of classical and egc SAgs lie in a complex network of regulatory pathways that determine the moment and the conditions in which S. aureus produces them [256,261], resulting in much lower concentrations of egc SAgs compared to classical SAgs [87,88,89], as was mentioned in Section 2.2.
In contrast with αβ T cells, γδ T cells constitute between 1 and 5% of PBMCs and are mainly concentrated in skin and mucosa. Eighty percent of γδ T cells in peripheral blood co-express the chains Vγ9 (alternatively Vγ2) or Vδ2, and it has been demonstrated that when activated, they can act as antigen-presenting cells [262], in addition to having the cytotoxic capacity and a great source of cytokine production. While some works stated that some classic SAgs, such as SEA, SEB and TSST-1, can stimulate directly or indirectly different subsets of γδ T cells, differences are referring to each SAg [260,263,264]. These suggest that the activation of this specific cell type could be related to a characteristic affinity of each subset of γδ TCR and its capacity to interact with one or another SAg, indicating that the residues involved in the binding with the αβ and γδ TCR are not shared. The capacity to stimulate this subpopulation of T cells has been extended to other SAgs; however, no evidence supports this fact for non-classical SAgs.
Furthermore, it has been reported that SEB activates NKT cells, and that SEA and SEB can interact with invariant NKT cells (iNKT) [265,266,267], inducing a pathogenic role of this cell type in SST. Moreover, classical SAgs interact with B cells [268,269,270], and IgE antibodies against superantigens have been detected [271,272].
It is demonstrated as well that SEA, SEB, SEE and TSST-1 can interact with mast cells [60,273,274,275], and the novelty that SEB activates mucosal-associated invariant T cells (MAIT cells) to produce high levels of INF-y, TNF-α and IL-2 and then induces anergy in this cell type [276]. MAIT cells are an unconventional T cell subset with a semi-invariant T cell receptor that recognizes antigens presented in the context of classical MHC-like molecules [277] and are involved in microbial immunity displaying protective and pathogenic responses [278,279]. Again, there is no information about the effect of non-classical Sags on these cells.
SSL toxins are another group of exotoxins of S. aureus that are unable to induce T cell proliferation but can contribute to immune evasion [48], interfering with complement and neutrophil function [280]. Similarly, SelX can bind both monocytes and neutrophils, impairing neutrophil activation [281,282], and is considered the first line of defense against S. aureus [283].
Although the activation of other cell types could have a considerable impact on the immune system response, almost every study published has been developed using the less prevalent classical SAgs, and it is not clear if their effects can be extended to the newer SAgs, which are widely distributed, such as the egc operon SAgs.
Due to their ability to interact with several components of the immune system, SAgs have been clinically used as immune modifiers in neoplasm treatment and other pathologies.

2.6. Superantigens as Therapeutic Tools

Considering the complexity of oncological processes, altered superantigens with reducing systemic toxicity that conserves the ability to eliminate tumoral cells have being considered as alternative therapies. Indeed, several cancer cell lines are effectively attacked by PBMCs activated by classical SAgs. Furthermore, various SAgs mutated in the MHC II contact region have been created willing to keep the antitumoral effects of SAgs, thus reducing systemic toxicity. Considering this strategy to diminish side effects, SAgs continue being extensively investigated for oncological applications either alone or in combination with classical anticancer drugs [284]. It was described that PBMCs stimulated with SEA display the ability to promote the death of human lung carcinoma A549 cells [285]. Similar results were observed when SEB-stimulated PBMCs induced apoptosis on transitional cell carcinoma cells [286]. These first studies were carried out with wild-type superantigens, which conserved intact their potent inflammatory activity. With the aim to avoid SAg systemic toxicity, various SAgs altered in hot spot regions have been created. That is the case for SEA D227A, which was conjugated with an antitumor Fab antibody; the fusion protein conserved the capacity to activate T cells but reduced the binding to the MHC-II molecule 500 times, compared to wild-type SEA [287]. Moreover, SEC2 double mutant, T20L/G22E, inhibited the growth of S180 sarcoma with less toxicity in mice [288].
Other alternatives to reduce systemic toxicity in using SAg as therapeutic tools include mutant SAgs that were designed as a part of chimeric single chain antibodies (scFv) specific to tumoral antigens. These SAgs have been mutated in the binding site to the MHC-II molecule with the aim of reducing their superantigenicity. These mutations reduced their affinity to the MHC-II molecule, and the cytotoxic effect on MHC class II- expressing cells [289]. Recently, a new generation of antibody–superantigen fusion proteins was designed, in which the SAg, in this case SEA, was split into two fragments. Individually, each fragment remains inactive, but when the biological formulation reaches the target cell by binding to cell surface antigens, it dimerizes and the SAg regains its biological functionality activating T cells. The effective split SEA design would not affect MHC class II-expressing cells, but when bound to a tumor antigen via a targeting moiety it would activate a T cell response [290].
Non-classical SAgs were also investigated in the immunotherapy field. The egc operon superantigens SEG and SEI were linked to the endogenous human MHCII HLA-DQ8 allele in humanized mice inducing a potent antitumor response and extending life in an established melanoma mice model [291].
In addition, the combination of mutant SAgs with other molecules to create chimeras that specifically stimulate the immune system with low toxicity is another strategy not fully studied yet, but with promising results, as in the case of mutant SEG used as a candidate for Chagas vaccination [292]. In that work, a chimeric molecule comprising a T. cruzi antigen and a non-toxic form of SEG was used as a novel immunogen to confer protection against T. cruzi infection. The mutant SAg used retained its ability to trigger classical activation of macrophages without affecting T cells, because this Th1 profile was adequate to eradicate intracellular protozoa such as T. cruzi, proving to be an effective immune modulator against this parasite.
These uses of SAgs as therapeutic agents that permit taking advantage of these toxins allow us to compare them to Dr. Jekyll, but it should not be forgotten that SAgs usually also act as Mr. Hyde, causing several affections and worsening disease conditions.
Unfortunately, everything in nature has its Dr. Jekyll and Mr. Hyde side, and superantigens are one of the most crucial toxin threats in warfare or bioterrorism as they are resistant to heat and can be administrated in contaminated water or as aerosols in the air.
It is our choice to use these toxins either like Dr. Jekyll or Mr. Hyde.

3. Concluding Remarks

Microorganisms evolve to evade the host immune system. All mammal pathogens have to face a strong and complex immune response, designed to fight them back. Manipulating this kind of response could be the most effective mechanism to spread the infection. This situation is the ideal scenario to develop a chronic infection and reach an equilibrium between the effector mechanisms of the immune system and the war for survival of the pathogen.
Staphylococcus aureus causes an acute infection where quick spread and colonization are essential to reach its goal. Considering that inflammation is crucial for the resolution of most bacterial infections, it is not clear how a pro-inflammatory state may help bacteria succeed. However, the inflammation induced by SAgs seems to impair activation and recruitment of important effector cells, promoting a temporal host immune suppression and the survival of S. aureus. In view of the numerous diseases in which SAgs participate or are implicated, it can be ascertained that Mr. Hyde’s side of SAgs is the easily visible face of these toxins.
For many years, it was considered that SAgs only affected T cell function, driving them to anergy or apoptosis, and promoting host immune suppression. As described throughout this work, SAgs would not only exert their function on T lymphocytes in the presence of antigen-presenting cells, but they would also be capable of eliciting different effects on monocyte/macrophage effector cells, ɣδ T lymphocytes, lymphocytes B, neutrophils and other cells not belonging to the immune system, such as adipocytes. In general, this action would lead to nonspecific pro-inflammatory conditions that would deplete effector cell populations, thus being inhibited to eradicate the bacteria that produce them, promoting a state of host immunosuppression from the first to the last stages of the infection, which would favor bacterial propagation. However, this intricate scenario, which is gradually being cleared, can be reversed, by taking advantage of the action of SAgs while restricting their effects for human benefit, thus revealing the protective yet still unclear Dr Jekyll side of SAgs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/toxins14110800/s1, Table S1: Rate of sequence similarity between SAgs.

Author Contributions

Conceptualization, writing the original draft preparation, S.N.T.; writing the original draft, D.M.R., writing special section, M.B.S.; editing, E.L.M.; writing, review and supervision, M.M.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. White, J.; Herman, A.; Pullen, A.M.; Kubo, R.; Kappler, J.W.; Marrack, P. The Vβ-specific superantigen staphylococcal enterotoxin B: Stimulation of mature T cells and clonal deletion in neonatal mice. Cell 1989, 56, 27–35. [Google Scholar] [CrossRef]
  2. Marrack, P.; Kappler, J. The Staphylococcal Enterotoxins and Their Relatives. Science 1990, 248, 705–711. [Google Scholar] [CrossRef] [PubMed]
  3. Schutzer, S.E.; Fischetti, V.A.; Zabriskie, J.B. Toxic Shock Syndrome and Lysogeny in Staphylococcus aureus. Obstet. Gynecol. Surv. 1983, 220, 316–318. [Google Scholar]
  4. Quimby, F.; Nguyen, H.T.; Bergdoll, M.S. Animal studies of toxic shock syndrome. Crit. Rev. Microbiol. 1985, 12, 1–44. [Google Scholar] [CrossRef]
  5. Gaventa, S.; Reingold, A.L.; Hightower, A.W.; Broome, C.V.; Schwartz, B.; Hoppe, C.; Harwell, J.; Lefkowitz, L.K.; Makintubee, S.; Cundiff, D.R.; et al. Active surveillance for toxic shock syndrome in the united states, 1986. Rev. Infect. Dis. 1989, 11, S28–S34. [Google Scholar] [CrossRef]
  6. Blank, C.; Luz, A.; Bendigs, S.; Erdmann, A.; Wagner, H.; Heeg, K. Superantigen and endotoxin synergize in the induction of lethal shock. Eur. J. Immunol. 1997, 27, 825–833. [Google Scholar] [CrossRef]
  7. Dalpke, A.H.; Heeg, K. Synergistic and antagonistic interactions between LPS and superantigens. J. Endotoxin Res. 2003, 9, 51–54. [Google Scholar] [CrossRef] [Green Version]
  8. MacIsaac, C.M.; Curtis, N.; Cade, J.; Visvanathan, K. Superantigens in sepsis. Int. Congr. Ser. 2006, 1289, 121–124. [Google Scholar] [CrossRef]
  9. Cole, B.C.; Atkin, C.L. The Mycoplasma arthritidis T-cell mitogen MAM: A model superantigen. Immunol. Today 1991, 12, 271–276. [Google Scholar] [CrossRef]
  10. Igwe, E.I.; Shewmaker, P.L.; Facklam, R.R.; Farley, M.M.; Van Beneden, C.; Beall, B. Identification of superantigen genes speM, ssa, and smeZ in invasive strains of beta-hemolytic group C and G streptococci recovered from humans. FEMS Microbiol. Lett. 2003, 229, 259–264. [Google Scholar] [CrossRef] [Green Version]
  11. Abe, J.; Takeda, T.; Watanabe, Y.; Nakao, H.; Kobayashi, N.; Leung, D.Y.; Kohsaka, T. Evidence for superantigen production by Yersinia pseudotuberculosis. J. Immunol. 1993, 151, 4183. [Google Scholar] [PubMed]
  12. Wei, B.; Huang, T.; Dalwadi, H.; Sutton, C.L.; Bruckner, D.; Braun, J. Pseudomonas fluorescens encodes the Crohn’s disease-associated I2 sequence and T-cell superantigen. Infect. Immun. 2002, 70, 6567–6575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Huber, B.T.; Hsu, P.N.; Sutkowski, N. Virus-encoded superantigens. Microbiol. Rev. 1996, 60, 473–482. [Google Scholar] [CrossRef] [PubMed]
  14. Sutkowski, N.; Conrad, B.; Thorley-Lawson, D.A.; Huber, B.T. Epstein-Barr virus transactivates the human endogenous retrovirus HERV-K18 that encodes a superantigen. Immunity 2001, 15, 579–589. [Google Scholar] [CrossRef] [Green Version]
  15. Azar, G.A.; Thibodeau, J. Human endogenous retrovirus IDDMK1,222 and mouse mammary tumor virus superantigens differ in their ability to stimulate murine T cell hybridomas. Immunol. Lett. 2002, 81, 87–91. [Google Scholar] [CrossRef]
  16. Tuffs, S.; Haeryfar, S.; McCormick, J. Manipulation of Innate and Adaptive Immunity by Staphylococcal Superantigens. Pathogens 2018, 7, 53. [Google Scholar] [CrossRef] [Green Version]
  17. Kluytmans, J.; Van Belkum, A.; Verbrugh, H. Nasal Carriage of Staphylococcus aureus: Epidemiology, Underlying Mechanisms, and Associated Risks. Clin. Microbiol. Rev. 1997, 10, 505–520. [Google Scholar] [CrossRef]
  18. Laux, C.; Peschel, A.; Krismer, B. Staphylococcus aureus Colonization of the Human Nose and Interaction with Other Microbiome Members. Microbiol. Spectr. 2019, 7, GPP3-0029-2018. [Google Scholar] [CrossRef]
  19. Verhoeven, P.O.; Gagnaire, J.; Botelho-Nevers, E.; Grattard, F.; Carricajo, A.; Lucht, F.; Pozzetto, B.; Berthelot, P. Detection and clinical relevance of Staphylococcus aureus nasal carriage: An update. Expert Rev. Anti. Infect. Ther. 2014, 12, 75–89. [Google Scholar] [CrossRef]
  20. Williams, R.E. Healthy carriage of Staphylococcus aureus: Its prevalence and importance. Bacteriol. Rev. 1963, 27, 56–71. [Google Scholar] [CrossRef]
  21. Armstrong-Esther, C.A.; Smith, J.E. Carriage patterns of staphylococcus aureus in a healthy non-hospital population of adults and children. Ann. Hum. Biol. 1976, 3, 221–227. [Google Scholar] [CrossRef] [PubMed]
  22. Gagnaire, J.; Verhoeven, P.O.; Grattard, F.; Rigaill, J.; Lucht, F.; Pozzetto, B.; Berthelot, P.; Botelho-Nevers, E. Epidemiology and clinical relevance of Staphylococcus aureus intestinal carriage: A systematic review and meta-analysis. Expert Rev. Anti. Infect. Ther. 2017, 15, 767–785. [Google Scholar] [CrossRef] [PubMed]
  23. Ridley, M. Perineal carriage of staph. aureus. Br. Med. J. 1959, 1, 270–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Wertheim, H.F.L.; Verveer, J.; Boelens, H.A.M.; Van Belkum, A.; Verbrugh, H.A.; Vos, M.C. Effect of mupirocin treatment on nasal, pharyngeal, and perineal carriage of Staphylococcus aureus in healthy adults. Antimicrob. Agents Chemother. 2005, 49, 1465–1467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. von Eiff, C.; Becker, K.; Machka, K.; Stammer, H.; Peters, G. Nasal Carriage as a Source of Staphylococcus aureus Bacteremia. N. Engl. J. Med. 2001, 344, 11–16. [Google Scholar] [CrossRef]
  26. Bergdoll, M.S.; Borja, C.R.; Avena, R.M. Identification of a new enterotoxin as enterotoxin C. J. Bacteriol. 1965, 90, 1481–1485. [Google Scholar] [CrossRef] [Green Version]
  27. Bergdoll, M.S.; Borja, C.R.; Robbins, R.N.; Weiss, K.F. Identification of enterotoxin E. Infect. Immun. 1971, 4, 593–595. [Google Scholar] [CrossRef] [Green Version]
  28. Casman, E.P.; Bennett, R.W.; Dorsey, A.E.; Issa, J.A. Identification of a fourth staphylococcal enterotoxin, enterotoxin D. J. Bacteriol. 1967, 94, 1875–1882. [Google Scholar] [CrossRef] [Green Version]
  29. Casman, E.P.; Bennett, R.W. Culture medium for the production of staphylococcal enterotoxin A. J. Bacteriol. 1963, 86, 18–23. [Google Scholar] [CrossRef] [Green Version]
  30. Casman, E.P.; Bergdoll, M.S.; Robinson, J. Designation of staphylococcal enterotoxins. J. Bacteriol. 1963, 85, 715–716. [Google Scholar] [CrossRef] [Green Version]
  31. Jones, C.L.; Khan, S.A. Nucleotide Sequence of the Enterotoxin B Gene from Staphylococcus aureus. J. Bacteriol. 1986, 166, 29–33. [Google Scholar] [CrossRef] [PubMed]
  32. Peavy, D.L.; Adler, W.H.; Smith, R.T. The mitogenic effects of endotoxin and staphylococcal enterotoxin B on mouse spleen cells and human peripheral lymphocytes. J. Immunol. 1970, 105, 1453–1458. [Google Scholar] [PubMed]
  33. Spaulding, A.R.; Salgado-Pabón, W.; Kohler, P.L.; Horswill, A.R.; Leung, D.Y.M.; Schlievert, P.M. Staphylococcal and streptococcal superantigen exotoxins. Clin. Microbiol. Rev. 2013, 26, 422–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ren, K.; Bannan, J.D.; Pancholi, V.; Cheung, A.L.; Robbins, J.C.; Fischetti, V.A.; Zabriskie, J.B. Characterization and Biological Properties of a New Staphylococcal Exotoxin. J. Exp. Med. 1994, 180, 1675–1683. [Google Scholar] [CrossRef] [Green Version]
  35. Zhang, S.; Iandolo, J.J.; Stewart, G.C. The enterotoxin D plasmid of Staphylococcus aureus encodes a second enterotoxin determinant (sej). FEMS Microbiol. Lett. 1998, 168, 227–233. [Google Scholar] [CrossRef] [Green Version]
  36. Munson, S.H.; Tremaine, M.T.; Betley, M.J.; Welch, R.A. Identification and Characterization of Staphylococcal Enterotoxin Types G and I from Staphylococcus aureus Identification and Characterization of Staphylococcal Enterotoxin Types G and I from Staphylococcus aureus. Infect. Immun. 1998, 66, 3337–3348. [Google Scholar] [CrossRef] [Green Version]
  37. Jarraud, S.; Peyrat, M.A.; Lim, A.; Tristan, A.; Bes, M.; Mougel, C.; Etienne, J.; Vandenesch, F.; Bonneville, M.; Lina, G. egc, a highly prevalent operon of enterotoxin gene, forms a putative nursery of superantigens in Staphylococcus aureus. J. Immunol. 2001, 166, 669–677. [Google Scholar] [CrossRef] [Green Version]
  38. Fitzgerald, J.R.; Monday, S.R.; Foster, T.J.; Bohach, G.A.; Hartigan, P.J.; Meaney, W.J.; Smyth, C.J. Characterization of a Putative Pathogenicity Island from Bovine Staphylococcus aureus Encoding Multiple Superantigens. J. Bacteriol. 2001, 183, 63–70. [Google Scholar] [CrossRef] [Green Version]
  39. Orwin, P.M.; Leung, D.Y.M.; Donahue, H.L.; Novick, R.P.; Schlievert, P.M. Biochemical and Biological Properties of Staphylococcal Enterotoxin K. Infect. Immun. 2001, 69, 360–366. [Google Scholar] [CrossRef] [Green Version]
  40. Correction. J. Immunol. 2001, 166, 4260. [CrossRef] [Green Version]
  41. Letertre, C.; Perelle, S.; Dilasser, F.; Fach, P. Identification of a new putative enterotoxin SEU encoded by the egc cluster of Staphylococcus aureus. J. Appl. Microbiol. 2003, 95, 38–43. [Google Scholar] [CrossRef] [PubMed]
  42. Thomas, D.Y.; Jarraud, S.; Lemercier, B.; Cozon, G.; Echasserieau, K.; Etienne, J.; Gougeon, M.L.; Lina, G.; Vandenesch, F. Staphylococcal enterotoxin-like toxins U2 and V, two new staphylococcal superantigens arising from recombination within the enterotoxin gene cluster. Infect. Immun. 2006, 74, 4724–4734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Omoe, K.; Ichi Imanishi, K.; Hu, D.L.; Kato, H.; Fugane, Y.; Abe, Y.; Hamaoka, S.; Watanabe, Y.; Nakane, A.; Uchiyama, T.; et al. Characterization of Novel Staphylococcal Enterotoxin-Like Toxin Type P. Infect. Immun. 2005, 73, 5540–5546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ono, H.K.; Omoe, K.; Imanishi, K.; Iwakabe, Y.; Hu, D.L.; Kato, H.; Saito, N.; Nakane, A.; Uchiyama, T.; Shinagawa, K. Identification and Characterization of Two Novel Staphylococcal Enterotoxins, Types S and T. Infect. Immun. 2008, 76, 4999–5005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Orwin, P.M.; Leung, D.Y.M.; Tripp, T.J.; Bohach, G.A.; Earhart, C.A.; Ohlendorf, D.H.; Schlievert, P.M. Characterization of a novel staphylococcal enterotoxin-like superantigen, a member of the group V subfamily of pyrogenic toxins. Biochemistry 2002, 41, 14033–14040. [Google Scholar] [CrossRef] [PubMed]
  46. Omoe, K.; Ichi Imanishi, K.; Hu, D.L.; Kato, H.; Takahashi-Omoe, H.; Nakane, A.; Uchiyama, T.; Shinagawa, K. Biological Properties of Staphylococcal Enterotoxin-Like Toxin Type R. Infect. Immun. 2004, 72, 3664–3667. [Google Scholar] [CrossRef] [Green Version]
  47. Okumura, K.; Shimomura, Y.; Yamagata Murayama, S.; Yagi, J.; Ubukata, K.; Kirikae, T.; Miyoshi-Akiyama, T. Evolutionary paths of streptococcal and staphylococcal superantigens. BMC Genom. 2012, 13, 404. [Google Scholar] [CrossRef] [Green Version]
  48. Wilson, G.J.; Seo, K.S.; Cartwright, R.A.; Connelley, T.; Chuang-Smith, O.N.; Merriman, J.A.; Guinane, C.M.; Park, J.Y.; Bohach, G.A.; Schlievert, P.M.; et al. A Novel Core Genome-Encoded Superantigen Contributes to Lethality of Community-Associated MRSA Necrotizing Pneumonia. PLoS Pathog. 2011, 7, e1002271. [Google Scholar] [CrossRef] [Green Version]
  49. Ono, H.K.; Sato’o, Y.; Narita, K.; Naito, I.; Hirose, S.; Hisatsune, J.; Asano, K.; Hu, D.L.; Omoe, K.; Sugai, M.; et al. Identification and characterization of a novel staphylococcal emetic toxin. Appl. Environ. Microbiol. 2015, 81, 7034–7040. [Google Scholar] [CrossRef] [Green Version]
  50. Spoor, L.E.; Richardson, E.; Richards, A.C.; Wilson, G.J.; Mendonca, C.; Gupta, R.K.; Mcadam, P.R.; Nutbeam-Tuffs, S.; Black, N.S.; O’gara, J.P.; et al. Recombination-mediated remodelling of host-pathogen interactions during Staphylococcus aureus niche adaptation. Microb. Genom. 2015, 1, e000036. [Google Scholar] [CrossRef]
  51. Hisatsune, J.; Hagiya, H.; Shiota, S.; Sugai, M. Complete Genome Sequence of Systemically Disseminated Sequence Type 8 Staphylococcal Cassette Chromosome mec Type IVl Community-Acquired Methicillin-Resistant Staphylococcus aureus. Genome Announc. 2017, 5, e00852-17. [Google Scholar] [CrossRef] [PubMed]
  52. Suzuki, Y.; Ono, H.K.; Shimojima, Y.; Kubota, H.; Kato, R.; Kakuda, T.; Hirose, S.; Hu, D.L.; Nakane, A.; Takai, S.; et al. A novel staphylococcal enterotoxin SE02 involved in a staphylococcal food poisoning outbreak that occurred in Tokyo in 2004. Food Microbiol. 2020, 92, 103588. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, D.F.; Yang, X.Y.; Zhang, J.; Qin, X.; Huang, X.; Cui, Y.; Zhou, M.; Shi, C.; French, N.P.; Shi, X. Identification and characterization of two novel superantigens among Staphylococcus aureus complex. Int. J. Med. Microbiol. 2018, 308, 438–446. [Google Scholar] [CrossRef] [PubMed]
  54. Lina, G.; Bohach, G.A.; Nair, S.P.; Hiramatsu, K.; Jouvin-Marche, E.; Mariuzza, R. Standard Nomenclature for the Superantigens Expressed by Staphylococcus. J. Infect. Dis. 2004, 189, 2334–2336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wang, T.; Tao, X.X.; Meng, F.L.; Li, X.P.; Ono-Hisaya, O.H.; Wang, D.; Hu, D.L.; Zhang, J.Z.; Wang, G.Q.; Yan, X.M. Cloning and expression of recombinant truncated SElX protein and evaluation on the related emetic activities. Zhonghua Liu Xing Bing Xue Za Zhi 2020, 41, 567–570. [Google Scholar] [CrossRef] [PubMed]
  56. Hu, D.L.; Omoe, K.; Shimoda, Y.; Nakane, A.; Shinagawa, K. Induction of Emetic Response to Staphylococcal Enterotoxins in the House Musk Shrew (Suncus murinus). Infect. Immun. 2003, 71, 567–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ueno, S.; Matsuki, N.; Saito, H. Suncus murinus as a new experimental model for motion sickness. Life Sci. 1988, 43, 413–420. [Google Scholar] [CrossRef]
  58. Omoe, K.; Hu, D.L.; Takahashi-Omoe, H.; Nakane, A.; Shinagawa, K. Comprehensive analysis of classical and newly described staphylococcal superantigenic toxin genes in Staphylococcus aureus isolates. FEMS Microbiol. Lett. 2005, 246, 191–198. [Google Scholar] [CrossRef] [Green Version]
  59. Hu, D.L.; Ono, H.K.; Isayama, S.; Okada, R.; Okamura, M.; Lei, L.C.; Liu, Z.S.; Zhang, X.C.; Liu, M.Y.; Cui, J.C.; et al. Biological characteristics of staphylococcal enterotoxin Q and its potential risk for food poisoning. J. Appl. Microbiol. 2017, 122, 1672–1679. [Google Scholar] [CrossRef]
  60. Ono, H.K.; Hirose, S.; Narita, K.; Sugiyama, M.; Asano, K.; Hu, D.L.; Nakane, A. Histamine release from intestinal mast cells induced by staphylococcal enterotoxin A (SEA) evokes vomiting reflex in common marmoset. PLoS Pathog. 2019, 15, e1007803. [Google Scholar] [CrossRef] [Green Version]
  61. Ferry, T.; Thomas, D.; Genestier, A.L.; Bes, M.; Lina, G.; Vandenesch, F.; Etienne, J. Comparative Prevalence of Superantigen Genes in Staphylococcus aureus Isolates Causing Sepsis with and without Septic Shock. Clin. Infect. Dis. 2005, 41, 771–777. [Google Scholar] [CrossRef] [PubMed]
  62. Noli Truant, S.; De Marzi, M.C.; Sarratea, M.B.; Antonoglou, M.B.; Meo, A.P.; Iannantuono López, L.V.; Fernández Lynch, M.J.; Todone, M.; Malchiodi, E.L.; Fernández, M.M. egc Superantigens Impair Monocytes/Macrophages Inducing Cell Death and Inefficient Activation. Front. Immunol. 2020, 10, 3008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zhang, P.; Liu, X.; Zhang, M.; Kou, M.; Chang, G.; Wan, Y.; Xu, X.; Ruan, F.; Wang, Y.; Wang, X. Prevalence, Antimicrobial Resistance, and Molecular Characteristics of Staphylococcus aureus and Methicillin-Resistant Staphylococcus aureus from Retail Ice Cream in Shaanxi Province, China. Foodborne Pathog. Dis. 2022, 19, 217–225. [Google Scholar] [CrossRef] [PubMed]
  64. Aung, M.S.; Urushibara, N.; Kawaguchiya, M.; Ito, M.; Habadera, S.; Kobayashi, N. Prevalence and Genetic Diversity of Staphylococcal Enterotoxin (-Like) Genes sey, selw, selx, selz, sel26 and sel27 in Community-Acquired Methicillin-Resistant Staphylococcus aureus. Toxins 2020, 12, 347. [Google Scholar] [CrossRef] [PubMed]
  65. Becker, K.; Friedrich, A.W.; Lubritz, G.; Weilert, M.; Peters, G.; Von Eiff, C. Prevalence of Genes Encoding Pyrogenic Toxin Superantigens and Exfoliative Toxins among Strains of Staphylococcus aureus Isolated from Blood and Nasal Specimens. J. Clin. Microbiol. 2003, 41, 1434–1439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Fischer, A.J.; Kilgore, S.H.; Singh, S.B.; Allen, P.D.; Hansen, A.R.; Limoli, D.H.; Schlievert, P.M. High Prevalence of Staphylococcus aureus Enterotoxin Gene Cluster Superantigens in Cystic Fibrosis Clinical Isolates. Genes 2019, 10, 1036. [Google Scholar] [CrossRef] [Green Version]
  67. Park, K.H.; Greenwood-Quaintance, K.E.; Cunningham, S.A.; Chia, N.; Jeraldo, P.R.; Sampathkumar, P.; Nelson, H.; Patel, R. Lack of correlation of virulence gene profiles of Staphylococcus aureus bacteremia isolates with mortality. Microb. Pathog. 2019, 133, 103543. [Google Scholar] [CrossRef]
  68. Garbacz, K.; Piechowicz, L.; Podkowik, M.; Mroczkowska, A.; Empel, J.; Bania, J. Infection and Drug Resistance Dovepress emergence and spread of worldwide Staphylococcus aureus clones among cystic fibrosis patients. Infect. Drug Resist. 2018, 11, 247. [Google Scholar] [CrossRef] [Green Version]
  69. Mempel, M.; Lina, G.; Hojka, M.; Schnopp, C.; Seidl, H.P.; Schäfer, T.; Ring, J.; Vandenesch, F.; Abeck, D. High prevalence of superantigens associated with the egc locus in Staphylococcus aureus isolates from patients with atopic eczema. Eur. J. Clin. Microbiol. Infect. Dis. 2003, 22, 306–309. [Google Scholar] [CrossRef]
  70. Merriman, J.A.; Mueller, E.A.; Cahill, M.P.; Beck, L.A.; Paller, A.S.; Hanifin, J.M.; Ong, P.Y.; Schneider, L.; Babineau, D.C.; David, G.; et al. Temporal and Racial Differences Associated with Atopic Dermatitis Staphylococcus aureus and Encoded Virulence Factors. Msphere 2016, 1, e00295-16. [Google Scholar] [CrossRef] [Green Version]
  71. Van Belkum, A.; Melles, D.C.; Snijders, S.V.; Van Leeuwen, W.B.; Wertheim, H.F.L.; Nouwen, J.L.; Verbrugh, H.A.; Etienne, J. Clonal Distribution and Differential Occurrence of the Enterotoxin Gene Cluster, egc, in Carriage-versus Bacteremia-Associated Isolates of Staphylococcus aureus. J. Clin. Microbiol. 2006, 44, 1555–1557. [Google Scholar] [CrossRef] [PubMed]
  72. Bronner, S.; Monteil, H.; Prévost, G. Regulation of virulence determinants in Staphylococcus aureus: Complexity and applications. FEMS Microbiol. Rev. 2006, 28, 183–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Cheung, A.L.; Bayer, A.S.; Zhang, G.; Gresham, H.; Xiong, Y.Q. Regulation of virulence determinants in vitro and in vivo in Staphylococcus aureus. FEMS Immunol. Med. Microbiol. 2006, 40, 1–9. [Google Scholar] [CrossRef] [Green Version]
  74. Jenul, C.; Horswill, A.R. Regulation of Staphylococcus aureus virulence. Microbiol. Spectr. 2019, 7, 7-2. [Google Scholar] [CrossRef]
  75. Bergdoll, M.S.; Czop, J.K.; Gould, S.S. Enterotoxin Synthesis by the Staphylococci. Ann. N. Y. Acad. Sci. 1974, 236, 307–316. [Google Scholar] [CrossRef]
  76. Bernardo, K.; Pakulat, N.; Fleer, S.; Schnaith, A.; Utermöhlen, O.; Krut, O.; Müller, S.; Krönke, M. Subinhibitory Concentrations of Linezolid Reduce Staphylococcus aureus Virulence Factor Expression. Antimicrob. Agents Chemother. 2004, 48, 546–555. [Google Scholar] [CrossRef] [Green Version]
  77. Czop, J.K.; Bergdoll, M.S. Staphylococcal Enterotoxin Synthesis during the Exponential, Transitional, and Stationary Growth Phases. Infect. Immun. 1974, 9, 229–235. [Google Scholar] [CrossRef] [Green Version]
  78. Gaskill, M.E.; Khan, S.A. Regulation of the enterotoxin B gene in Staphylococcus aureus. J. Biol. Chem. 1988, 263, 6276–6280. [Google Scholar] [CrossRef]
  79. Otero, A.; Garcia, M.L.; Garcia, M.C.; Moreno, B.; Bergdoll, M.S. Production of Staphylococcal Enterotoxins C1 and C2 and Thermonuclease throughout the Growth Cycle. Appl. Environ. Microbiol. 1990, 56, 555–559. [Google Scholar] [CrossRef] [Green Version]
  80. Regassa, L.B.; Couch, J.L.; Betley, M.J. Steady-State Staphylococcal Enterotoxin Type C mRNA Is Affected by a Product of the Accessory Gene Regulator (agr) and by Glucose. Infect. Immun. 1991, 59, 955–962. [Google Scholar] [CrossRef] [Green Version]
  81. Sihto, H.M.; Tasara, T.; Stephan, R.; Johler, S. Temporal expression of the staphylococcal enterotoxin D gene under NaCl stress conditions. FEMS Microbiol. Lett. 2015, 362, 24. [Google Scholar] [CrossRef] [PubMed]
  82. Macori, G.; Bellio, A.; Bianchi, D.M.; Chiesa, F.; Gallina, S.; Romano, A.; Zuccon, F.; Cabrera-Rubio, R.; Cauquil, A.; Merda, D.; et al. Genome-wide profiling of enterotoxigenic Staphylococcus aureus strains used for the production of naturally contaminated cheeses. Genes 2020, 11, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Andrey, D.O.; Jousselin, A.; Villanueva, M.; Renzoni, A.; Monod, A.; Barras, C.; Rodriguez, N.; Kelley, W.L. Impact of the regulators SigB, rot, SarA and sarS on the toxic shock tst promoter and TSST-1 expression in Staphylococcus aureus. PLoS ONE 2015, 10, e0135579. [Google Scholar] [CrossRef] [PubMed]
  84. Baroja, M.L.; Herfst, C.A.; Kasper, K.J.; Xu, S.X.; Gillett, D.A.; Li, J.; Reid, G.; McCormick, J.K. The SaeRS two-component system is a direct and dominant transcriptional activator of toxic shock syndrome toxin 1 in Staphylococcus aureus. J. Bacteriol. 2016, 198, 2732–2742. [Google Scholar] [CrossRef] [Green Version]
  85. Nagao, M.; Okamoto, A.; Yamada, K.; Hasegawa, T.; Hasegawa, Y.; Ohta, M. Variations in amount of TSST-1 produced by clinical methicillin resistant Staphylococcus aureus (MRSA) isolates and allelic variation in accessory gene regulator (agr) locus. BMC Microbiol. 2009, 9, 52. [Google Scholar] [CrossRef] [Green Version]
  86. Derzelle, S.; Dilasser, F.; Duquenne, M.; Deperrois, V. Differential temporal expression of the staphylococcal enterotoxins genes during cell growth. Food Microbiol. 2009, 26, 896–904. [Google Scholar] [CrossRef]
  87. Stach, C.S.; Vu, B.G.; Merriman, J.A.; Herrera, A.; Cahill, M.P.; Schlievert, P.M.; Salgado-Pabón, W. Novel Tissue Level Effects of the Staphylococcus aureus Enterotoxin Gene Cluster Are Essential for Infective Endocarditis. PLoS ONE 2016, 11, e0154762. [Google Scholar] [CrossRef]
  88. Strandberg, K.L.; Rotschafer, J.H.; Vetter, S.M.; Buonpane, R.A.; Kranz, D.M.; Schlievert, P.M. Staphylococcal Superantigens Cause Lethal Pulmonary Disease in Rabbits. J. Infect. Dis. 2010, 202, 1690–1697. [Google Scholar] [CrossRef] [Green Version]
  89. Schlievert, P.M.; Peterson, M.L. Glycerol Monolaurate Antibacterial Activity in Broth and Biofilm Cultures. PLoS ONE 2012, 7, e40350. [Google Scholar] [CrossRef] [Green Version]
  90. Goerke, C.; Campana, S.; Bayer, M.G.; Döring, G.; Botzenhart, K.; Wolz, C. Direct quantitative transcript analysis of the agr regulon of Staphylococcus aureus during human infection in comparison to the expression profile in vitro. Infect. Immun. 2000, 68, 1304–1311. [Google Scholar] [CrossRef] [Green Version]
  91. Goerke, C.; Wolz, C. Regulatory and genomic plasticity of Staphylococcus aureus during persistent colonization and infection. Int. J. Med. Microbiol. 2004, 294, 195–202. [Google Scholar] [CrossRef] [PubMed]
  92. Banks, M.C.; Kamel, N.S.; Zabriskie, J.B.; Larone, D.H.; Ursea, D.; Posnett, D.N. Staphylococcus aureus Express Unique Superantigens Depending on the Tissue Source. J. Infect. Dis. 2003, 187, 77–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Omoe, K.; Ishikawa, M.; Shimoda, Y.; Hu, D.L.; Ueda, S.; Shinagawa, K. Detection of seg, seh, and sei genes in Staphylococcus aureus Isolates and Determination of the Enterotoxin Productivities of S. aureus Isolates Harboring seg, seh, or sei Genes. J. Clin. Microbiol. 2002, 40, 857–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Burlak, C.; Hammer, C.H.; Robinson, M.A.; Whitney, A.R.; Mcgavin, M.J.; Kreiswirth, B.N.; Deleo, F.R. Global analysis of community-associated methicillin-resistant Staphylococcus aureus exoproteins reveals molecules produced in vitro and during infection. Cell. Microbiol. 2007, 9, 1172–1190. [Google Scholar] [CrossRef] [Green Version]
  95. Enany, S.; Yoshida, Y.; Magdeldin, S.; Zhang, Y.; Bo, X.; Yamamoto, T. Extensive proteomic profiling of the secretome of European community acquired methicillin resistant Staphylococcus aureus clone. Peptides 2012, 37, 128–137. [Google Scholar] [CrossRef]
  96. Pocsfalvi, G.; Cacace, G.; Cuccurullo, M.; Serluca, G.; Sorrentino, A.; Schlosser, G.; Blaiotta, G.; Malorni, A. Proteomic analysis of exoproteins expressed by enterotoxigenic Staphylococcus aureus strains. Proteomics 2008, 8, 2462–2476. [Google Scholar] [CrossRef]
  97. Enany, S.; Yoshida, Y.; Magdeldin, S.; Bo, X.; Zhang, Y.; Enany, M.; Yamamoto, T. Two dimensional electrophoresis of the exo-proteome produced from community acquired methicillin resistant Staphylococcus aureus belonging to clonal complex 80. Microbiol. Res. 2013, 168, 504–511. [Google Scholar] [CrossRef] [PubMed]
  98. Smith, D.S.; Siggins, M.K.; Gierula, M.; Pichon, B.; Turner, C.E.; Lynskey, N.N.; Mosavie, M.; Kearns, A.M.; Edwards, R.J.; Sriskandan, S. Identification of commonly expressed exoproteins and proteolytic cleavage events by proteomic mining of clinically relevant UK isolates of Staphylococcus aureus. Microb. Genom. 2016, 2, e000049. [Google Scholar] [CrossRef]
  99. Ziebandt, A.K.; Kusch, H.; Degner, M.; Jaglitz, S.; Sibbald, M.J.J.B.; Arends, J.P.; Chlebowicz, M.A.; Albrecht, D.; Pantuček, R.; Doškar, J.; et al. Proteomics uncovers extreme heterogeneity in the Staphylococcus aureus exoproteome due to genomic plasticity and variant gene regulation. Proteomics 2010, 10, 1634–1644. [Google Scholar] [CrossRef]
  100. Brizzio, A.A.; Tedeschi, F.A.; Zalazar, F.E. Multiplex PCR strategy for the simultaneous identification of Staphylococcus aureus and detection of staphylococcal enterotoxins in isolates from food poisoning outbreaks. Biomed. Rev. Inst. Nac. Salud 2013, 33, 122–127. [Google Scholar] [CrossRef]
  101. Chiang, Y.C.; Liao, W.W.; Fan, C.M.; Pai, W.Y.; Chiou, C.S.; Tsen, H.Y. PCR detection of Staphylococcal enterotoxins (SEs) N, O, P, Q, R, U, and survey of SE types in Staphylococcus aureus isolates from food-poisoning cases in Taiwan. Int. J. Food Microbiol. 2008, 121, 66–73. [Google Scholar] [CrossRef] [PubMed]
  102. Hait, J.M.; Tallent, S.M.; Bennett, R.W. Screening, Detection, and Serotyping Methods for Toxin Genes and Enterotoxins in Staphylococcus Strains. J. AOAC Int. 2014, 97, 1078–1083. [Google Scholar] [CrossRef] [PubMed]
  103. Lefebvre, D.; Blanco-Valle, K.; Hennekinne, J.A.; Simon, S.; Fenaille, F.; Becher, F.; Nia, Y.; Lefebvre, D.; Blanco-Valle, K.; Hennekinne, J.-A.; et al. Multiplex Detection of 24 Staphylococcal Enterotoxins in Culture Supernatant Using Liquid Chromatography Coupled to High-Resolution Mass Spectrometry. Toxins 2022, 14, 249. [Google Scholar] [CrossRef] [PubMed]
  104. Sarratea, M.B.; Noli, T.S.; Mitarotonda, R.; Antonoglou, M.B.; Chiappini, S.; Fernández Lynch, M.J.; Romasanta, P.; Vescina, C.; Desimone, M.; De Marzi, M.; et al. Optimized surface plasmon resonance immunoassay for staphylococcal enterotoxin G detection using silica nanoparticles. Biochem. Biophys. Res. Commun. 2021, 558, 168–174. [Google Scholar] [CrossRef]
  105. Gupta, G.; Singh, P.K.; Boopathi, M.; Kamboj, D.V.; Singh, B.; Vijayaraghavan, R. Surface plasmon resonance detection of biological warfare agent Staphylococcal enterotoxin B using high affinity monoclonal antibody. Thin Solid Film 2010, 519, 1171–1177. [Google Scholar] [CrossRef]
  106. Hait, J.M.; Nguyen, A.T.; Tallent, S.M. Analysis of the VIDAS® Staph Enterotoxin III (SET3) for detection of staphylococcal enterotoxins G, H, and I in foods. J. AOAC Int. 2018, 101, 1482–1489. [Google Scholar] [CrossRef]
  107. Nagaraj, S.; Ramlal, S.; Kingston, J.; Batra, H.V. Development of IgY based sandwich ELISA for the detection of staphylococcal enterotoxin G (SEG), an egc toxin. Int. J. Food Microbiol. 2016, 237, 136–141. [Google Scholar] [CrossRef]
  108. Wu, S.; Duan, N.; Gu, H.; Hao, L.; Ye, H.; Gong, W.; Wang, Z. A Review of the Methods for Detection of Staphylococcus aureus Enterotoxins. Toxins 2016, 8, 176. [Google Scholar] [CrossRef] [Green Version]
  109. Rasooly, L.; Rasooly, A. Real time biosensor analysis of Staphylococcal enterotoxin A in food. Int. J. Food Microbiol. 1999, 49, 119–127. [Google Scholar] [CrossRef]
  110. Schlievert, P.M.; Blomster, D.A. Production of Staphylococcal Pyrogenic Exotoxin Type C: Influence of Physical and Chemical Factors. J. Infect. Dis. 1983, 147, 236–242. [Google Scholar] [CrossRef]
  111. Hajjeh, R.A.; Reingold, A.; Weil, A.; Shutt, K.; Schuchat, A.; Perkins, B.A. Hajjeh Toxic Shock Syndrome in the United States: Surveillance Update, 1979–1996. Emerg. Infect. Dis. 1996, 5, 807. [Google Scholar] [CrossRef] [PubMed]
  112. Davis, J.P. Toxic-Shock Syndrome Epidemiologic Features, Recurrence, Risk Factors, and Prevention. N. Engl. J. Med. 1986, 314, 144–149. [Google Scholar] [CrossRef]
  113. Lee, V.T.P.; Chang, A.H.; Chow, A.W. Detection of Staphylococcal Enterotoxin B among Toxic Shock Syndrome (TSS)-and Non-TSS-Associated Staphylococcus aureus Isolates. J. Infect. Dis. 1992, 166, 911–915. [Google Scholar] [CrossRef] [PubMed]
  114. De Vries, A.S.; Lesher, L.; Schlievert, P.M.; Rogers, T.; Villaume, L.G.; Danila, R.; Lynfield, R. Staphylococcal toxic shock syndrome 2000–2006: Epidemiology, clinical features, and molecular characteristics. PLoS ONE 2011, 6, e22997. [Google Scholar] [CrossRef]
  115. Bohach, G.A.; Fast, D.J.; Nelson, R.D.; Schlievert, P.M. Staphylococcal and Streptococcal Pyrogenic Toxins Involved in Toxic Shock Syndrome and Related Illnesses. Crit. Rev. Microbiol. 1990, 17, 251–272. [Google Scholar] [CrossRef]
  116. Chang, A.; Musser, J.; Chow, A.W. A single clone of S. aureus which produces both TSST-1 and SEA causes the majority of menstrual toxic shock syndrome. In Clinical Research; Slack Inc.: Thorofare, NJ, USA, 1991. [Google Scholar]
  117. Jarraud, S.; Cozon, G.; Vandenesch, O.I.S.; Etienne, J.; Lina, G. Involvement of Enterotoxins G and I in Staphylococcal Toxic Shock Syndrome and Staphylococcal Scarlet Fever. J. Clin. Microbiol. 1999, 37, 2446–2449. [Google Scholar] [CrossRef] [Green Version]
  118. Peterson, M.L.; Ault, K.; Kremer, M.J.; Klingelhutz, A.J.; Davis, C.C.; Squier, C.A.; Schlievert, P.M. The innate immune system is activated by stimulation of vaginal epithelial cells with Staphylococcus aureus and toxic shock syndrome toxin 1. Infect. Immun. 2005, 73, 2164–2174. [Google Scholar] [CrossRef] [Green Version]
  119. Asano, K.; Narita, K.; Hirose, S.; Nakane, A. Contribution of toxic shock syndrome toxin-1 to systemic inflammation investigated by a mouse model of cervicovaginal infection with Staphylococcus aureus. Med. Microbiol. Immunol. 2018, 207, 297–306. [Google Scholar] [CrossRef]
  120. Asano, K.; Asano, Y.; Ono, H.K.; Nakane, A. Suppression of starvation-induced autophagy by recombinant toxic shock syndrome toxin-1 in epithelial cells. PLoS ONE 2014, 9, e113018. [Google Scholar] [CrossRef] [Green Version]
  121. Schlievert, P.M.; Davis, C.C. Device-Associated Menstrual Toxic Shock Syndrome. Clin. Microbiol. Rev. 2020, 33, e00032-19. [Google Scholar] [CrossRef]
  122. Billon, A.; Gustin, M.-P.; Tristan, A.; Bénet, T.; Berthiller, J.; Gustave, C.A.; Vanhems, P.; Lina, G. Association of characteristics of tampon use with menstrual toxic shock syndrome in France. EClinicalMedicine 2020, 21, 100308. [Google Scholar] [CrossRef] [PubMed]
  123. Davis, C.C.; Baccam, M.; Mantz, M.J.; Osborn, T.W.; Hill, D.R.; Squier, C.A. Use of porcine vaginal tissue ex-vivo to model environmental effects on vaginal mucosa to toxic shock syndrome toxin-1. Toxicol. Appl. Pharmacol. 2014, 274, 240–248. [Google Scholar] [CrossRef] [PubMed]
  124. Nonfoux, L.; Chiaruzzi, M.; Badiou, C.; Baude, J.; Tristan, A.; Thioulouse, J.; Muller, D.; Prigent-Combaret, C.; Lina, G. Impact of currently marketed tampons and menstrual cups on Staphylococcus aureus growth and toxic shock syndrome toxin 1 production in vitro. Appl. Environ. Microbiol. 2018, 84, e00351-18. [Google Scholar] [CrossRef] [Green Version]
  125. Schlievert, P.M. Effect of non-absorbent intravaginal menstrual/contraceptive products on Staphylococcus aureus and production of the superantigen TSST-1. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 31–38. [Google Scholar] [CrossRef] [PubMed]
  126. Tierno, P.M.; Hanna, B.A. Propensity of Tampons and Barrier Contraceptives to Amplify Staphylococcus aureus Toxic Shock Syndrome Toxin-1. Infect. Dis. Obstet. Gynecol. 1994, 2, 140–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Herrmann, T.; Accolla, R.S.; MacDonald, H.R. Different staphylococcal enterotoxins bind preferentially to distinct major histocompatibility complex class ii isotypes. Eur. J. Immunol. 1989, 19, 2171–2174. [Google Scholar] [CrossRef] [PubMed]
  128. Reingold, A.L.; Hargrett, N.T.; Dan, B.B.; Shands, K.N.; Strickland, B.Y.; Broome, C.V.; Atlanta, G. Nonmenstrual Toxic Shock Syndrome A Review of 130 Cases. Ann. Intern. Med. 1982, 96, 871–874. [Google Scholar] [CrossRef]
  129. Whiting, J.L.; Rosten, P.M.; Chow, A.W. Determination by Western blot (immunoblot) of seroconversions to toxic shock syndrome (TSS) toxin 1 and enterotoxin A, B, or C during infection with TSS- and Non-TSS-associated Staphylococcus aureus. Infect. Immun. 1989, 57, 231–234. [Google Scholar] [CrossRef] [Green Version]
  130. Ferro, J.M.; Fonseca, A.C. Infective endocarditis. Handb. Clin. Neurol. 2014, 119, 75–91. [Google Scholar] [CrossRef]
  131. Fowler, V.G.; Olsen, M.K.; Corey, G.R.; Cheng, A.C.; Dudley, T.; Oddone, E.Z. Clinical Identifiers of Complicated S. aureus bacteremia. Arch. Intern. Med. 2003, 163, 2066–2072. [Google Scholar] [CrossRef] [Green Version]
  132. Chung, J.W.; Karau, M.J.; Greenwood-Quaintance, K.E.; Ballard, A.D.; Tilahun, A.; Khaleghi, S.R.; David, C.S.; Patel, R.; Rajagopalan, G. Superantigen profiling of Staphylococcus aureus infective endocarditis isolates. Diagn. Microbiol. Infect. Dis. 2014, 79, 119–124. [Google Scholar] [CrossRef] [PubMed]
  133. Kinney, K.J.; Tran, P.M.; Gibson-Corley, K.N.; Forsythe, A.N.; Kulhankova, K.; Salgado-Pabón, W. Staphylococcal Enterotoxin C promotes Staphylococcus aureus Infective Endocarditis Independent of Superantigen Activity. bioRxiv 2019, 14. [Google Scholar] [CrossRef] [Green Version]
  134. Salgado-Pabón, W.; Injury, A.K.; Peterson, M.L.; Schlievert, M. Superantigens Are Critical for Staphylococcus aureus Infective Endocarditis, Sepsis, and Acute Kidney Injury. MBio 2013, 4, e00494-13. [Google Scholar] [CrossRef] [Green Version]
  135. Kulhankova, K. The Superantigen Toxic Shock Syndrome Toxin 1 Alters Human Aortic Endothelial Cell Function. Infect. Immun. 2018, 86, e00848-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Becker, A.; Kerr, E. Staphylococcus aureus pneumonia: A “superbug” infection in community and hospital settings. Chest 2005, 128, 99–111. [Google Scholar] [CrossRef]
  137. Torres, A.; Menéndez, R.; Wundernik, R.G. Bacterial pneumonia and lung abscess. Murray Nadel’s Textb. Respir. Med. 2020, 557–582. [Google Scholar] [CrossRef]
  138. Hyvernat, H.; Pulcini, C.; Carles, D.; Roques, A.; Lucas, P.; Hofman, V.; Hofman, P.; Bernardin, G. Fatal Staphylococcus aureus haemorrhagic pneumonia producing Panton-Valentine leucocidin. Scand. J. Infect. Dis. 2007, 39, 183–185. [Google Scholar] [CrossRef]
  139. Ippolito, G.; Leone, S.; Lauria, F.N.; Nicastri, E.; Wenzel, R.P. Methicillin-resistant Staphylococcus aureus: The superbug. Int. J. Infect. Dis. 2010, 14, S7–S11. [Google Scholar] [CrossRef] [Green Version]
  140. Aliberti, S.; Reyes, L.F.; Faverio, P.; Sotgiu, G.; Dore, S.; Rodriguez, A.H.; Soni, N.J.; Restrepo, M.I.; Aruj, P.K.; Attorri, S.; et al. Global initiative for meticillin-resistant Staphylococcus aureus pneumonia (GLIMP): An international, observational cohort study. Lancet Infect. Dis. 2016, 16, 1364–1376. [Google Scholar] [CrossRef]
  141. Centers for Disease Control and Prevention. Four pediatric deaths from community-acquired methicillin-resistant Staphylococcus aureus—Minnesota and North Dakota, 1997–1999. JAMA 1999, 282, 1123–1125. [Google Scholar]
  142. Strandberg, K.L.; Rotschafer, J.H.; Schlievert, P.M. Extreme Pyrexia and Rapid Death Due to Staphylococcus aureus Infection: Analysis of 2 Cases. Clin. Infect. Dis. 2009, 48, 612–614. [Google Scholar] [CrossRef] [Green Version]
  143. Kulhankova, K.; King, J.; Salgado-Pabón, W. Staphylococcal toxic shock syndrome: Superantigen-mediated enhancement of endotoxin shock and adaptive immune suppression. Immunol. Res. 2014, 59, 182–187. [Google Scholar] [CrossRef] [PubMed]
  144. Parker, D.; Ryan, C.L.; Alonzo, F.; Torres, V.J.; Planet, P.J.; Prince, A.S. CD4+ T cells promote the pathogenesis of Staphylococcus aureus pneumonia. J. Infect. Dis. 2015, 211, 835–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Chao, G.; Bao, G.; Cao, Y.; Yan, W.; Wang, Y.; Zhang, X.; Zhou, L.; Wu, Y. Prevalence and diversity of enterotoxin genes with genetic background of Staphylococcus aureus isolates from different origins in China. Int. J. Food Microbiol. 2015, 211, 142–147. [Google Scholar] [CrossRef] [PubMed]
  146. Johler, S.; Giannini, P.; Jermini, M.; Hummerjohann, J.; Baumgartner, A.; Stephan, R. Further evidence for staphylococcal food poisoning outbreaks caused by egc-Encoded enterotoxins. Toxins 2015, 7, 997–1004. [Google Scholar] [CrossRef]
  147. Lv, G.P.; Xu, B.H.; Wei, P.N.; Song, J.; Zhang, H.Y.; Zhao, C.; Qin, L.Y.; Zhao, B.H. Molecular characterization of foodborne-associated Staphylococcus aureus strains isolated in Shijiazhuang, China, from 2010 to 2012. Diagn. Microbiol. Infect. Dis. 2014, 78, 462–468. [Google Scholar] [CrossRef]
  148. Shen, M.; Li, Y.; Zhang, L.; Dai, S.; Wang, J.; Li, Y.; Zhang, L.; Huang, J. Staphylococcus enterotoxin profile of China isolates and the superantigenicity of some novel enterotoxins. Arch. Microbiol. 2017, 199, 723–736. [Google Scholar] [CrossRef]
  149. Wattinger, L.; Stephan, R.; Layer, F.; Johler, S. Comparison of staphylococcus aureus isolates associated with food intoxication with isolates from human nasal carriers and human infections. Eur. J. Clin. Microbiol. Infect. Dis. 2012, 31, 455–464. [Google Scholar] [CrossRef] [Green Version]
  150. Yan, X.; Wang, B.; Tao, X.; Hu, Q.; Cui, Z.; Zhang, J.; Lin, Y.; You, Y.; Shi, X.; Grundmann, H. Characterization of Staphylococcus aureus strains associated with food poisoning in Shenzhen, China. Appl. Environ. Microbiol. 2012, 78, 6637–6642. [Google Scholar] [CrossRef] [Green Version]
  151. Blaiotta, G.; Ercolini, D.; Pennacchia, C.; Fusco, V.; Casaburi, A.; Pepe, O.; Villani, F. PCR detection of staphylococcal enterotoxin genes in Staphylococcus spp. strains isolated from meat and dairy products. Evidence for new variants of seG and seI in S. aureus AB-8802. J. Appl. Microbiol. 2004, 97, 719–730. [Google Scholar] [CrossRef]
  152. McLauchlin, J.; Narayanan, G.L.; Mithani, V.; O’Neill, G. The detection of enterotoxins and toxic shock syndrome toxin genes in Staphylococcus aureus by polymerase chain reaction. J. Food Prot. 2000, 63, 479–488. [Google Scholar] [CrossRef] [PubMed]
  153. Viçosa, G.N.; Le Loir, A.; Le Loir, Y.; de Carvalho, A.F.; Nero, L.A. Egc characterization of enterotoxigenic Staphylococcus aureus isolates obtained from raw milk and cheese. Int. J. Food Microbiol. 2013, 165, 227–230. [Google Scholar] [CrossRef] [PubMed]
  154. Ikeda, T.; Tamate, N.; Yamaguchi, K.; Makino, S.I. Mass outbreak of food poisoning disease caused by small amounts of staphylococcal enterotoxins A and H. Appl. Environ. Microbiol. 2005, 71, 2793–2795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Jørgensen, H.J.; Mathisen, T.; Løvseth, A.; Omoe, K.; Qvale, K.S.; Loncarevic, S. An outbreak of staphylococcal food poisoning caused by enterotoxin H in mashed potato made with raw milk. FEMS Microbiol. Lett. 2005, 252, 267–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. de Freitas, M.F.; Luz, I.D.S.; Silveira-Filho, V.D.M.; Júnior, J.W.P.; Stamford, T.L.; Mota, R.A.; de Sena, M.J.; de Almeida, A.M.P.; de Q. Balbino, V.; Leal-Balbino, T.C. Staphylococcal toxin genes in strains isolated from cows with subclinical mastitis. Pesqui. Veterinária Bras. 2008, 28, 617–621. [Google Scholar] [CrossRef] [Green Version]
  157. Bianchi, D.M.; Gallina, S.; Bellio, A.; Chiesa, F.; Civera, T.; Decastelli, L. Enterotoxin gene profiles of Staphylococcus aureus isolated from milk and dairy products in Italy. Lett. Appl. Microbiol. 2014, 58, 190–196. [Google Scholar] [CrossRef]
  158. Carfora, V.; Caprioli, A.; Marri, N.; Sagrafoli, D.; Boselli, C.; Giacinti, G.; Giangolini, G.; Sorbara, L.; Dottarelli, S.; Battisti, A.; et al. Enterotoxin genes, enterotoxin production, and methicillin resistance in Staphylococcus aureus isolated from milk and dairy products in Central Italy. Int. Dairy J. 2015, 42, 12–15. [Google Scholar] [CrossRef] [Green Version]
  159. Rosec, J.P.; Guiraud, J.P.; Dalet, C.; Richard, N. Enterotoxin production by staphylococci isolated from foods in France. Int. J. Food Microbiol. 1997, 35, 213–221. [Google Scholar] [CrossRef]
  160. Zhao, Y.; Zhu, A.; Tang, J.; Tang, C.; Chen, J.; Liu, J. Identification and measurement of staphylococcal enterotoxin-like protein I (SEll) secretion from Staphylococcus aureus clinical isolate. J. Appl. Microbiol. 2016, 121, 539–546. [Google Scholar] [CrossRef]
  161. Cheng, F.H.M.; Andrews, P.L.R.; Moreaux, B.; Ngan, M.P.; Rudd, J.A.; Sam, T.S.W.; Wai, M.K.; Wan, C. Evaluation of the anti-emetic potential of anti-migraine drugs to prevent resiniferatoxin-induced emesis in Suncus murinus (house musk shrew). Eur. J. Pharmacol. 2005, 508, 231–238. [Google Scholar] [CrossRef]
  162. Hu, D.L.; Zhu, G.; Mori, F.; Omoe, K.; Okada, M.; Wakabayashi, K.; Kaneko, S.; Shinagawa, K.; Nakane, A. Staphylococcal enterotoxin induces emesis through increasing serotonin release in intestine and it is downregulated by cannabinoid receptor 1. Cell. Microbiol. 2007, 9, 2267–2277. [Google Scholar] [CrossRef] [PubMed]
  163. Sugiyama, H.; Hayama, T. Abdominal viscera as site of emetic action for staphylococcal enterotoxin in the monkey. J. Infect. Dis. 1965, 115, 330–336. [Google Scholar] [CrossRef] [PubMed]
  164. Shupp, J.W.; Jett, M.; Pontzer, C.H. Identification of a transcytosis epitope on staphylococcal enterotoxins. Infect. Immun. 2002, 70, 2178–2186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Spiekermann, G.M.; Anderson, C.N. Oral Administration of the Bacterial Superantigen Staphylococcal Enterotoxin B Induces Activation and Cytokine Production by T Cells in Murine Gut-Associated Lymphoid Tissue. J. Immunol. 1998, 161, 5825–5831. [Google Scholar] [CrossRef] [Green Version]
  166. McKay, D.M.; Benjamin, M.A.; Lu, J. CD4+ T cells mediate superantigen-induced abnormalities in murine jejunal ion transport. Am. J. Physiol.-Gastrointest. Liver Physiol. 1998, 275, 29–38. [Google Scholar] [CrossRef]
  167. Pinchuk, I.V.; Beswick, E.J.; Reyes, V.E. Staphylococcal enterotoxins. Toxins 2010, 2, 2177–2197. [Google Scholar] [CrossRef] [Green Version]
  168. Friedman, S.M.; Posnett, D.N.; Tumang, J.R.; Crow, M.K.; Cole, B.C. A potential role for microbial superantigens in the pathogenesis of systemic autoimmune disease. Arthritis Rheum. 1991, 34, 468–480. [Google Scholar] [CrossRef]
  169. Wucherpfennig, K.W. Mechanisms for the induction of autoimmunity by infectious agents. J. Clin. Investig. 2001, 108, 1097–1104. [Google Scholar] [CrossRef]
  170. Schiffenbauer, J.; Soos, J.; Johnson, H. The possible role of bacterial superantigens in the pathogenesis of autoimmune disorders. Immunol. Today 1998, 19, 117–120. [Google Scholar] [CrossRef]
  171. Dietz, S.M.; van Stijn, D.; Burgner, D.; Levin, M.; Kuipers, I.M.; Hutten, B.A.; Kuijpers, T.W. Dissecting Kawasaki disease: A state-of-the-art review. Eur. J. Pediatr. 2017, 176, 995–1009. [Google Scholar] [CrossRef] [Green Version]
  172. Matsubara, K.; Fukaya, T. The role of superantigens of group A Streptococcus and Staphylococcus aureus in Kawasaki disease. Curr. Opin. Infect. Dis. 2007, 20, 298–303. [Google Scholar] [CrossRef] [PubMed]
  173. Nagata, S. Causes of Kawasaki disease—From past to present. Front. Pediatr. 2019, 7, 1–7. [Google Scholar] [CrossRef] [PubMed]
  174. Yeung, R.S.M. The etiology of Kawasaki disease: A superantigen-mediated process. Prog. Pediatr. Cardiol. 2004, 19, 115–122. [Google Scholar] [CrossRef]
  175. Nagata, S.; Yamashiro, Y.; Ohtsuka, Y.; Shimizu, T.; Sakurai, Y.; Misawa, S.; Ito, T. Heat shock proteins and superantigenic properties of bacteria from the gastrointestinal tract of patients with Kawasaki disease. Immunology 2009, 128, 511–520. [Google Scholar] [CrossRef] [Green Version]
  176. Hall, M.; Schlievert, P.M. Kawasaki Syndrome-Like Illness Associated with Infection Caused by Enterotoxin B-Secreting Staphylococcus aureus. Clin. Infect. Dis. 1999, 29, 586–589. [Google Scholar] [CrossRef] [Green Version]
  177. Leung, D.Y.M.; Meissner, H.C.; Fulton, D.; Murray, D.L.; Kotzin, B.; Schlievert, P.M. Toxic shock syndrome toxin-secreting Staphylococcus aureus in Kawasaki syndrome. Lancet 1994, 343, 300. [Google Scholar] [CrossRef]
  178. Leung, D.Y.M.; Meissner, H.C.; Shulman, S.T.; Mason, W.H.; Gerber, M.A.; Glode, M.P.; Myones, B.L.; Wheeler, J.G.; Ruthazer, R.; Schlievert, P.M. Prevalence of superantigen-secreting bacteria in patients with Kawasaki disease. J. Pediatr. 2002, 140, 742–746. [Google Scholar] [CrossRef]
  179. Matsubara, K.; Fukaya, T.; Miwa, K.; Shibayama, N.; Nigami, H.; Harigaya, H.; Nozaki, H.; Hirata, T.; Baba, K.; Suzuki, T.; et al. Development of serum IgM antibodies against superantigens of Staphylococcus aureus and Streptococcus pyogenes in Kawasaki disease. Clin. Exp. Immunol. 2006, 143, 427–434. [Google Scholar] [CrossRef]
  180. Nomura, Y.; Yoshinaga, M.; Masuda, K.; Takei, S.; Miyata, K. Maternal Antibody against Toxic Shock Syndrome Toxin–1 May Protect Infants Younger than 6 Months of Age from Developing Kawasaki Syndrome. J. Infect. Dis. 2002, 185, 1677–1680. [Google Scholar] [CrossRef] [Green Version]
  181. Terai, M.; Miwa, K.; Williams, T.; Kabat, W.; Fukuyama, M.; Okajima, Y.; Igarashi, H.; Shulman, S.T. The absence of evidence of staphylococcal toxin involvement in the pathogenesis of kawasaki disease. J. Infect. Dis. 1995, 172, 558–561. [Google Scholar] [CrossRef]
  182. Yim, D.; Ramsay, J.; Kothari, D.; Burgner, D. Coronary artery dilatation in toxic shock-like syndrome: The kawasaki disease shock syndrome. Pediatr. Cardiol. 2010, 31, 1232–1235. [Google Scholar] [CrossRef] [PubMed]
  183. Vu, B.G.; Stach, C.S.; Kulhankova, K.; Salgado-Pabón, W.; Klingelhutz, A.J.; Schlievert, P.M. Chronic superantigen exposure induces systemic inflammation, elevated bloodstream endotoxin, and abnormal glucose tolerance in rabbits: Possible role in diabetes. MBio 2015, 6, e02554-14. [Google Scholar] [CrossRef] [PubMed]
  184. Banke, E.; Rödström, K.; Ekelund, M.; Dalla-Riva, J.; Lagerstedt, J.O.; Nilsson, S.; Degerman, E.; Lindkvist-Petersson, K.; Nilson, B. Superantigen activates the gp130 receptor on adipocytes resulting in altered adipocyte metabolism. Metabolism 2014, 63, 831–840. [Google Scholar] [CrossRef] [PubMed]
  185. Vu, B.G.; Gourronc, F.A.; Bernlohr, D.A.; Schlievert, P.M.; Klingelhutz, A.J. Staphylococcal superantigens stimulate immortalized human adipocytes to produce chemokines. PLoS ONE 2013, 8, e77988. [Google Scholar] [CrossRef] [Green Version]
  186. Paliard, X.; West, S.G.; Lafferty, J.A.; Clements, J.R.; Kappler, J.W.; Marrack, P.; Kotzin, B.L. Evidence for the effects of a superantigen in rheumatoid arthritis. Science 1991, 253, 325–329. [Google Scholar] [CrossRef]
  187. Goodacre, J.A.; Brownlie, C.E.D.; Ross, D.A. Bacterial superantigens in autoimmune arthritis. Rheumatology 1994, 33, 413–419. [Google Scholar] [CrossRef]
  188. Ataee, R.A.; Ataee, M.H.; Alishiri, G.H.; Esmaeili, D. Staphylococcal enterotoxin C in synovial fluid of patients with rheumatoid arthritis. Iran. Red Crescent Med. J. 2014, 16, e16075. [Google Scholar] [CrossRef] [Green Version]
  189. Ataee, R.A.; Kahani, M.S.; Alishiri, G.H.; Ahamadi, Z. Staphylococcal Enterotoxin A Detection from Rheumatoid Arthritis Patients’ Blood and Synovial Fluid Ramezan. Electron. Physician 2016, 8, 1850. [Google Scholar] [CrossRef] [Green Version]
  190. Ataee, R.A.; Kashefi, R.; Alishiri, G.H.; Esmaieli, D. Staphylococcus aureus enterotoxin D: Absence of bacteria but presence of its toxin. Jundishapur J. Microbiol. 2015, 8, e28395. [Google Scholar] [CrossRef] [Green Version]
  191. Rashki, M.; Ataee, R.A.; Alishiri, G.H.; Esmaeili, D. Molecular Assay of Staphylococcal Enterotoxin E in Synovial Fluid of Patients with Rheumatoid Arthritis. Int. J. Infect. 2018, 5, 1–4. [Google Scholar] [CrossRef] [Green Version]
  192. Shokrollahi, M.R.; Noorbakhsh, S.; Aliakbari, M.; Tabatabaei, A. Searching the staphylococcal superantigens: Enterotoxins A, B, C, and TSST1 in synovial fluid of cases with negative culture inflammatory arthritis. Jundishapur J. Microbiol. 2014, 7, e11647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Tabarya, D.; Hoffman, W.L. Staphylococcus aureus nasal carriage in rheumatoid arthritis: Antibody response to toxic shock syndrome toxin-1. Ann. Rheum. Dis. 1996, 55, 823–828. [Google Scholar] [CrossRef] [PubMed]
  194. Origuchi, T.; Eguchi, K.; Kawabe, Y.; Yamashita, I.; Mizokami, A.; Ida, H.; Nagataki, S. Increased levels of serum IgM antibody to staphylococcal enterotoxin B in patients with rheumatoid arthritis. Ann. Rheum. Dis. 1995, 54, 713–720. [Google Scholar] [CrossRef] [PubMed]
  195. Gerlach, K.; Tomuschat, C.; Finke, R.; Staege, M.S.; Brütting, C.; Brandt, J.; Jordan, B.; Schwesig, R.; Rosemeier, A.; Delank, K.S.; et al. Experimental Arthritis in the Rat Induced by the Superantigen Staphylococcal Enterotoxin A. Scand. J. Immunol. 2017, 85, 191–196. [Google Scholar] [CrossRef] [Green Version]
  196. Banaei, M.; Alishiri, G.H.; Ataee, R.A.; Mahalati, A.H. Rheumatoid arthritis mediator CD18 expression by Staphylococcus aureus superantigen C in rats. Iran. J. Microbiol. 2019, 11, 337–344. [Google Scholar] [CrossRef]
  197. Abdelnour, A.; Bremell, T.; Holmdahl, R.; Tarkowski, A. Clonal expansion of T lymphocytes causes arthritis and mortality in mice infected with toxic shock syndrome toxin-1-producing staphylococci. Eur. J. Immunol. 1994, 24, 1161–1166. [Google Scholar] [CrossRef] [PubMed]
  198. Wooley, P.H.; Cingel, B. Staphylococcal enterotoxin B increases the severity of type II collagen induced arthritis in mice. Ann. Rheum. Dis. 1995, 54, 298–304. [Google Scholar] [CrossRef] [Green Version]
  199. Schwab, J.H.; Brown, R.R.; Anderle, S.K.; Schlievert, P.M. Superantigen can reactivate bacterial cell wall-induced arthritis. J. Immunol. 1993, 150, 4151–4159. [Google Scholar]
  200. Tsai, C.; Diaz, L.A.; Singer, N.G.; Li, L.L.; Kirsch, A.H.; Mitra, R.; Nickoloff, B.J.; Crofford, L.J.; Fox, D.A. Responsiveness of human T lymphocytes to bacterial superantigens presented by cultured rheumatoid arthritis synoviocytes. Arthritis Rheum. 1996, 39, 125–136. [Google Scholar] [CrossRef] [Green Version]
  201. Leung, D.Y.M.; Harbeck, R.; Bina, P.; Reiser, R.F.; Yang, E.; Norris, D.A.; Hanifin, J.M.; Sampson, H.A. Presence of IgE antibodies to staphylococcal exotoxins on the skin of patients with atopic dermatitis evidence for a new group of allergens. J. Clin. Investig. 1993, 92, 1374–1380. [Google Scholar] [CrossRef] [Green Version]
  202. Leyden, J.J.; Marples, R.R.; Kligman, A.M. Staphylococcus aureus in the lesions of atopic dermatitis. Br. J. Dermatol. 1974, 90, 525. [Google Scholar] [CrossRef] [PubMed]
  203. Skov, L.; Baadsgaard, O. Bacterial superantigens and inflammatory skin diseases. Clin. Exp. Dermatol. 2000, 25, 57–61. [Google Scholar] [CrossRef] [PubMed]
  204. Jappe, U. Superantigens and their association with dermatological inflammatory diseases: Facts and hypotheses. Acta Derm. Venereol. 2000, 80, 321–328. [Google Scholar] [CrossRef] [PubMed]
  205. Bunikowski, R.; Mielke, M.; Skarabis, H.; Herz, U.; Bergmann, R.L.; Wahn, U.; Renz, H. Prevalence and role of serum IgE antibodies to the Staphylococcus aureus-derived superantigens SEA and SEB in children with atopic dermatitis. J. Allergy Clin. Immunol. 1999, 103, 119–124. [Google Scholar] [CrossRef]
  206. Lin, Y.T.; Yang, Y.H.; Hwang, Y.W.; Tsai, M.J.; Tsao, P.N.; Chiang, B.L.; Shau, W.Y.; Wang, L.F. Comparison of serum specific IgE antibodies to staphylococcal enterotoxins between atopic children with and without atopic dermatitis. Allergy Eur. J. Allergy Clin. Immunol. 2000, 55, 641–646. [Google Scholar] [CrossRef]
  207. Tomczak, H.; Wróbel, J.; Jenerowicz, D.; Sadowska-Przytocka, A.; Wachal, M.; Adamski, Z.; Czarnecka-Operacz, M.M. The role of Staphylococcus aureus in atopic dermatitis: Microbiological and immunological implications. Postep. Dermatol. Alergol. 2019, 36, 485–491. [Google Scholar] [CrossRef]
  208. Schlievert, P.M.; Case, L.C.; Strandberg, K.L.; Abrams, B.B.; Leung, D.Y.M. Superantigen profile of Staphylococcus aureus isolates from patients with steroid-resistant atopic dermatitis. Clin. Infect. Dis. 2008, 46, 1562–1567. [Google Scholar] [CrossRef] [Green Version]
  209. Strickland, I.; Hauk, P.J.; Trumble, A.E.; Picker, L.J.; Leung, D.Y.M. Evidence for superantigen involvement in skin homing of T cells in atopic dermatitis. J. Investig. Dermatol. 1999, 112, 249–253. [Google Scholar] [CrossRef] [Green Version]
  210. McFadden, J.P.; Noble, W.C.; Camp, R.D.R. Superantigenic exotoxin-secreting potential of staphylococci isolated from atopic eczematous skin. Br. J. Dermatol. 1993, 128, 631–632. [Google Scholar] [CrossRef]
  211. Arad, G.; Levy, R.; Nasie, I.; Hillman, D.; Rotfogel, Z.; Barash, U.; Supper, E.; Shpilka, T.; Minis, A.; Kaempfer, R. Binding of superantigen toxins into the CD28 homodimer interface is essential for induction of cytokine genes that mediate lethal shock. PLoS Biol. 2011, 9, e1001149. [Google Scholar] [CrossRef]
  212. Ramachandran, G.; Tulapurkar, M.E.; Harris, K.M.; Arad, G.; Shirvan, A.; Shemesh, R.; Detolla, L.J.; Benazzi, C.; Opal, S.M.; Kaempfer, R.; et al. A peptide antagonist of CD28 signaling attenuates toxic shock and necrotizing soft-tissue infection induced by streptococcus pyogenes. J. Infect. Dis. 2013, 207, 1869–1877. [Google Scholar] [CrossRef] [PubMed]
  213. Levy, R.; Rotfogel, Z.; Hillman, D.; Popugailo, A.; Arad, G.; Supper, E.; Osman, F.; Kaempfer, R. Superantigens hyperinduce inflammatory cytokines by enhancing the B7-2/CD28 costimulatory receptor interaction. Proc. Natl. Acad. Sci. USA 2016, 113, E6437–E6446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Ganem, M.B.; De Marzi, M.C.; Fernández-Lynch, M.J.; Jancic, C.; Vermeulen, M.; Geffner, J.; Mariuzza, R.A.; Fernández, M.M.; Malchiodi, E.L. Uptake and Intracellular Trafficking of Superantigens in Dendritic Cells. PLoS ONE 2013, 8, e66244. [Google Scholar] [CrossRef] [PubMed]
  215. Hovde, C.J.; Marr, J.C.; Hoffmann, M.L.; Hackett, S.P.; Chi, Y.I.; Crum, K.K.; Stevens, D.L.; Stauffacher, C.V.; Bohach, G.A. Investigation of the role of the disulphide bond in the activity and structure of staphylococcal enterotoxin C1. Mol. Microbiol. 1994, 13, 897–909. [Google Scholar] [CrossRef]
  216. Baker, M.D.; Acharya, K.R. Superantigens: Structure-function relationships. Int. J. Med. Microbiol. 2004, 293, 529–537. [Google Scholar] [CrossRef]
  217. Petersson, K.; Håkansson, M.; Nilsson, H.; Forsberg, G.; Svensson, L.A.; Liljas, A.; Walse, B. Crystal structure of a superantigen bound to MHC class II displays zinc and peptide dependence. EMBO J. 2001, 20, 3306–3312. [Google Scholar] [CrossRef]
  218. Jardetzky, T.S.; Brown, J.H.; Gorga, J.C.; Stern, L.J.; Urban, R.G.; Chi, Y.I.; Stauffacher, C.; Strominger, J.L.; Wiley, D.C. Three-dimensional structure of a human class II histocompatibility molecule complexed with superantigen. Nature 1994, 368, 711–718. [Google Scholar] [CrossRef]
  219. Fernández, M.M.; Guan, R.; Swaminathan, C.P.; Malchiodi, E.L.; Mariuzza, R.A. Crystal structure of staphylococcal enterotoxin I (SEI) in complex with a human major histocompatibility complex class II molecule. J. Biol. Chem. 2006, 281, 25356–25364. [Google Scholar] [CrossRef] [Green Version]
  220. Leder, L.; Llera, A.; Lavoie, P.M.; Lebedeva, M.I.; Li, H.; Sékaly, R.P.; Bohach, G.A.; Gahr, P.J.; Schlievert, P.M.; Karjalainen, K.; et al. A Mutational Analysis of the Binding of Staphylococcal Enterotoxins B and C3 to the T Cell Receptor Beta Chain and Major Histocompatibility Complex Class II. J. Exp. Med. 1998, 187, 823–833. [Google Scholar] [CrossRef] [Green Version]
  221. Fernández, M.M.; Cho, S.; De Marzi, M.C.; Kerzic, M.C.; Robinson, H.; Mariuzza, R.A.; Malchiodi, E.L. Crystal structure of Staphylococcal Enterotoxin G (SEG) in complex with a mouse T-cell receptor β chain. J. Biol. Chem. 2011, 286, 1189–1195. [Google Scholar] [CrossRef] [Green Version]
  222. Fernández, M.M.; De Marzi, M.C.; Berguer, P.; Burzyn, D.; Langley, R.J.; Piazzon, I.; Mariuzza, R.A.; Malchiodi, E.L. Binding of natural variants of staphylococcal superantigens SEG and SEI to TCR and MHC class II molecule. Mol. Immunol. 2006, 43, 927–938. [Google Scholar] [CrossRef] [PubMed]
  223. Saline, M.; Rödström, K.E.J.; Fischer, G.; Orekhov, V.Y.; Karlsson, B.G.; Lindkvist-Petersson, K. The structure of superantigen complexed with TCR and MHC reveals novel insights into superantigenic T cell activation. Nat. Commun. 2010, 1, 119. [Google Scholar] [CrossRef] [PubMed]
  224. Popugailo, A.; Rotfogel, Z.; Supper, E.; Hillman, D.; Kaempfer, R. Staphylococcal and streptococcal superantigens trigger B7/CD28 costimulatory receptor engagement to hyperinduce inflammatory cytokines. Front. Immunol. 2019, 10, 942. [Google Scholar] [CrossRef]
  225. Gregory, S.; Zilber, M.T.; Charron, D.; Gelin, C. Human CD1a Molecule Expressed on Monocytes Plays an Accessory Role in the Superantigen-induced Activation of T Lymphocytes. Hum. Immunol. 2000, 61, 193–201. [Google Scholar] [CrossRef]
  226. Uzunçayır, S.; Vera-Rodriguez, A.; Regenthal, P.; Åbacka, H.; Emanuelsson, C.; Bahl, C.D.; Lindkvist-Petersson, K. Analyses of the complex formation of staphylococcal enterotoxin A and the human gp130 cytokine receptor. FEBS Lett. 2022, 596, 910–923. [Google Scholar] [CrossRef] [PubMed]
  227. Schlievert, P.M.; Cahill, M.P.; Hostager, B.S.; Brosnahan, A.J.; Klingelhutz, A.J.; Gourronc, F.A.; Bishop, G.A.; Leung, D.Y.M. Staphylococcal superantigens stimulate epithelial cells through CD40 to produce chemokines. MBio 2019, 10, e00214-19. [Google Scholar] [CrossRef] [Green Version]
  228. Schlievert, P.M.; Gourronc, F.A.; Leung, D.Y.M.; Klingelhutz, A.J. Human Keratinocyte Response to Superantigens. mSphere 2020, 5, e00803-20. [Google Scholar] [CrossRef]
  229. Schlievert, P.M.; Kilgore, S.H.; Benavides, A.; Klingelhutz, A.J. Pathogen Stimulation of Interleukin-8 from Human Vaginal Epithelial Cells through CD40. Microbiol. Spectr. 2022, 10, e00106-22. [Google Scholar] [CrossRef]
  230. Garcia, K.C.; Teyton, L.; Wilson, I.A. Structural basis of T cell recognition. Annu. Rev. Immunol. 1999, 17, 369–397. [Google Scholar] [CrossRef] [PubMed]
  231. Garcia, K.C.; Adams, E.J. How the T cell receptor sees antigen—A structural view. Cell 2005, 122, 333–336. [Google Scholar] [CrossRef] [Green Version]
  232. Smith-Garvin, J.E.; Koretzky, G.A.; Jordan, M.S. T cell activation. Annu. Rev. Immunol. 2009, 27, 592–619. [Google Scholar] [CrossRef] [PubMed]
  233. Givan, A.L.; Fisher, J.L.; Waugh, M.; Ernstoff, M.S.; Wallace, P.K. A flow cytometric method to estimate the precursor frequencies of cells proliferating in response to specific antigens. J. Immunol. Methods 1999, 230, 99–112. [Google Scholar] [CrossRef]
  234. Bueno, C.; Criado, G.; McCormick, J.K.; Madrenas, J. T Cell Signalling Induced by Bacterial Superantigens. In Superantigens and Superallergens; KARGER: Basel, Switzerland, 2007; pp. 161–180. [Google Scholar]
  235. Mccormick, J.K.; Yarwood, J.M.; Schlievert, P.M. Toxic shock syndrome and bacterial superantigens: An update. Annu. Rev. Microbiol. 2001, 55, 77. [Google Scholar] [CrossRef] [PubMed]
  236. Kawabe, Y.; Ochi, A. Programmed cell death and extrathymic reduction of Vβ8+ CD4+ T cells in mice tolerant to Staphylococcus aureus enterotoxin B. Nature 1991, 349, 245–248. [Google Scholar] [CrossRef]
  237. MacDonald, H.R.; Baschieri, S.; Lees, R.K. Clonal expansion precedes anergy and death of Vβ8+ peripheral T cells responding to staphylococcal enterotoxin B in vivo. Eur. J. Immunol. 1991, 21, 1963–1966. [Google Scholar] [CrossRef] [PubMed]
  238. Sundstedt, A.; Höiden, I.; Rosendahl, A.; Kalland, T.; van Rooijen, N.; Dohlsten, M. Immunoregulatory role of IL-10 during superantigen-induced hyporesponsiveness in vivo. J. Immunol. 1997, 158, 180–186. [Google Scholar]
  239. Taylor, A.L.; Llewelyn, M.J. Superantigen-Induced Proliferation of Human CD4 + CD25− T Cells Is Followed by a Switch to a Functional Regulatory Phenotype. J. Immunol. 2010, 185, 6591–6598. [Google Scholar] [CrossRef] [Green Version]
  240. Feunou, P.; Poulin, L.; Habran, C.; Le Moine, A.; Goldman, M.; Braun, M.Y. CD4+ CD25+ and CD4+ CD25− T Cells Act Respectively as Inducer and Effector T Suppressor Cells in Superantigen-Induced Tolerance. J. Immunol. 2003, 171, 3475–3484. [Google Scholar] [CrossRef] [Green Version]
  241. Lee, J.; Park, N.; Park, J.Y.; Kaplan, B.L.F.; Pruett, S.B.; Park, J.W.; Ho Park, Y.; Seo, K.S. Induction of Immunosuppressive CD8+ CD25+ FOXP3+ Regulatory T Cells by Suboptimal Stimulation with Staphylococcal Enterotoxin C1. J. Immunol. 2018, 200, 669–680. [Google Scholar] [CrossRef] [Green Version]
  242. Taylor, A.L.; Cross, E.L.A.; Llewelyn, M.J. Induction of contact-dependent CD8+ regulatory T cells through stimulation with staphylococcal and streptococcal superantigens*. Immunology 2012, 135, 158–167. [Google Scholar] [CrossRef]
  243. Wang, Z.Q.; Orlikowsky, T.; Dudhane, A.; Trejo, V.; Dannecker, G.E.; Pernist, B.; Hoffmann, M.K. Staphylococcal enterotoxin B-induced T-cell anergy is mediated by regulatory T cells. Immunology 1998, 94, 331–339. [Google Scholar] [CrossRef] [PubMed]
  244. Tilahun, A.Y.; Chowdhary, V.R.; David, C.S.; Rajagopalan, G. Systemic Inflammatory Response Elicited by Superantigen Destabilizes T Regulatory Cells Rendering them Ineffective during Toxic Shock Syndrome NIH Public Access. J. Immunol. 2014, 193, 2919–2930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Parker, D.; Karauzum, H.; Sähr, A.; Förmer, S.; Hildebrand, D.; Heeg, K. T-cell activation or tolerization: The Yin and Yang of bacterial superantigens. Front. Microbiol. 2015, 6, 1153. [Google Scholar] [CrossRef] [Green Version]
  246. Kato, M.; Nakamura, Y.; Suda, T.; Ozawa, Y.; Inui, N.; Seo, N.; Nagata, T.; Koide, Y.; Kalinski, P.; Nakamura, H.; et al. Enhanced anti-tumor immunity by superantigen-pulsed dendritic cells. Cancer Immunol. Immunother. 2011, 60, 1029–1038. [Google Scholar] [CrossRef] [PubMed]
  247. Bhardwaj, N.; Friedman, S.M.; Cole, B.C.; Nisanian, A.J. Dendritic Cells Are Potent Antigen-presenting Cells for Microbial Superantigens. J. Exp. Med. 1992, 175, 267–273. [Google Scholar] [CrossRef] [PubMed]
  248. Muraille, E.; De Trez, C.; Pajak, B.; Brait, M.; Urbain, J.; Leo, O. T Cell-Dependent Maturation of Dendritic Cells in Response to Bacterial Superantigens. J. Immunol. 2002, 168, 4352–4360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Coutant, K.D.; De Fraissinette, A.D.B.; Cordier, A.; Ulrich, P. Modulation of the activity of human monocyte-derived dendritic cells by chemical haptens, a metal allergen, and a staphylococcal superantigen. Toxicol. Sci. 1999, 52, 189–198. [Google Scholar] [CrossRef] [Green Version]
  250. Liu, T.; He, S.H.; Zheng, P.Y.; Zhang, T.Y.; Wang, B.Q.; Yang, P.C. Staphylococcal enterotoxin B increases TIM4 expression in human dendritic cells that drives naïve CD4 T cells to differentiate into Th2 cells. Mol. Immunol. 2007, 44, 3580–3587. [Google Scholar] [CrossRef]
  251. Muralimohan, G.; Vella, A.T. A role for IFNγ in differential superantigen stimulation of conventional versus plasmacytoid DCs. Cell. Immunol. 2006, 23, 9–22. [Google Scholar] [CrossRef] [Green Version]
  252. Takahashi, M.; Takahashi, M.; Shinohara, F.; Takada, H.; Rikiishi, H. Effects of superantigen and lipopolysaccharide on induction of CD80 through apoptosis of human monocytes. Infect. Immun. 2001, 69, 3652–3657. [Google Scholar] [CrossRef] [Green Version]
  253. Zhang, X.; Shang, W.; Yuan, J.; Hu, Z.; Peng, H.; Zhu, J.; Hu, Q.; Yang, Y.; Liu, H.; Jiang, B.; et al. Positive Feedback Cycle of TNFα Promotes Staphylococcal Enterotoxin B-Induced THP-1 Cell Apoptosis. Front. Cell. Infect. Microbiol. 2016, 6, 109. [Google Scholar] [CrossRef] [Green Version]
  254. Ulett, G.C.; Adderson, E.E. Regulation of Apoptosis by Gram-Positive Bacteria: Mechanistic Diversity and Consequences for Immunity. Curr. Immunol. Rev. 2006, 2, 119–141. [Google Scholar] [CrossRef] [Green Version]
  255. Krakauer, T. Staphylococcal Superantigens: Pyrogenic Toxins Induce Toxic Shock. Toxins 2019, 11, 178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Grumann, D.; Scharf, S.S.; Holtfreter, S.; Kohler, C.; Steil, L.; Engelmann, S.; Hecker, M.; Völker, U.; Bröker, B.M.; Volker, U.; et al. Immune Cell Activation by Enterotoxin Gene Cluster (egc)-Encoded and Non-egc Superantigens from Staphylococcus aureus. J. Immunol. 2008, 181, 5054–5061. [Google Scholar] [CrossRef] [PubMed]
  257. Islander, U.; Andersson, A.; Lindberg, E.; Adlerberth, I.; Wold, A.E.; Rudin, A. Superantigenic Staphylococcus aureus stimulates production of interleukin-17 from memory but not naive T cells. Infect. Immun. 2010, 78, 381–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Jupin, C.; Anderson, S.; Damais, C.; Alouf, J.E.; Parant, M. Toxic shock syndrome toxin 1 as an inducer of human tumor necrosis factors and λ interferon. J. Exp. Med. 1988, 167, 752–761. [Google Scholar] [CrossRef]
  259. Kum, W.W.S.; Cameron, S.B.; Hung, R.W.Y.; Kalyan, S.; Chow, A.W. Temporal Sequence and Kinetics of Proinflammatory and Anti-Inflammatory Cytokine Secretion Induced by Toxic Shock Syndrome Toxin 1 in Human Peripheral Blood Mononuclear Cells. Infect. Immun. 2001, 69, 7544–7549. [Google Scholar] [CrossRef] [Green Version]
  260. Rust, C.J.; Koning, F. Gamma delta T cell reactivity towards bacterial superantigens. Semin. Immunol. 1993, 5, 41–46. [Google Scholar] [CrossRef]
  261. Fisher, E.L.; Otto, M.; Cheung, G.Y.C. Basis of virulence in enterotoxin-mediated staphylococcal food poisoning. Front. Microbiol. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  262. Brandes, M.; Willimann, K.; Moser, B. Immunology: Professional antigen-presentation function by human γδ cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef]
  263. Maeurer, M.; Zitvogel, L.; Elder, E.; Storkus, W.J.; Lotze, M.T. Human intestinal V delta 1+ T cells obtained from patients with colon cancer respond exclusively to SEB but not to SEA. Nat. Immun. 1995, 14, 188–197. [Google Scholar] [PubMed]
  264. Morita, C.T.; Li, H.; Lamphear, J.G.; Rich, R.R.; Fraser, J.D.; Mariuzza, R.A.; Lee, H.K. Superantigen recognition by gamma delta T cells: SEA recognition site for human Vgamma2 T cell receptors. Immunity 2001, 14, 331–344. [Google Scholar] [CrossRef] [Green Version]
  265. Hayworth, J.L.; Mazzuca, D.M.; Vareki, S.M.; Welch, I.; McCormick, J.K.; Haeryfar, S.M. CD1d-independent activation of mouse and human i NKT cells by bacterial superantigens. Immunol. Cell Biol. 2012, 90, 699–709. [Google Scholar] [CrossRef] [PubMed]
  266. Rieder, S.A.; Nagarkatti, P.; Nagarkatti, M. CD1d-independent activation of invariant natural killer T cells by Staphylococcal enterotoxin B through major histocompatibility complex class II/T cell receptor interaction results in acute lung injury. Infect. Immun. 2011, 79, 3141–3148. [Google Scholar] [CrossRef] [PubMed]
  267. Szabo, P.A.; Rudak, P.T.; Choi, J.; Xu, S.X.; Schaub, R.; Singh, B.; McCormick, J.K.; Haeryfar, S.M.M. Invariant Natural Killer T Cells Are Pathogenic in the HLA-DR4-Transgenic Humanized Mouse Model of Toxic Shock Syndrome and Can Be Targeted to Reduce Morbidity. J. Infect. Dis. 2017, 215, 824–829. [Google Scholar] [CrossRef] [Green Version]
  268. Stohl, W.; Elliott, J.E.; Linsley, P.S. Human T cell-dependent B cell differentiation induced by staphylococcal superantigens. J. Immunol. 1994, 153, 117–127. [Google Scholar]
  269. Domiati-Saad, R.; Attrep, J.F.; Brezinschek, H.P.; Cherrie, A.H.; Karp, D.R.; Lipsky, P.E. Staphylococcal enterotoxin D functions as a human B cell superantigen by rescuing VH4-expressing B cells from apoptosis. J. Immunol. 1996, 156, 3608–3620. [Google Scholar]
  270. Domiati-Saad, R.; Lipsky, P.E. Staphylococcal Enterotoxin A Induces Survival of V H 3 Expressing Human B Cells by Binding to the V H Region with Low Affinity. J. Immunol. 1998, 161, 1257–1266. [Google Scholar]
  271. Gould, H.J.; Takhar, P.; Harries, H.E.; Chevretton, E.; Sutton, B.J. The allergic March from Staphylococcus aureus superantigens to immunoglobulin E. Chem. Immunol. Allergy 2007, 93, 106–136. [Google Scholar] [CrossRef]
  272. Shin, S.Y.; Choi, G.S.M.; Lee, K.H.; Kim, S.W.; Cho, J.S.; Park, H.S. IgE Response to Staphylococcal Enterotoxins in Adenoid Tissues from Atopic Children. Laryngoscope 2009, 119, 171–175. [Google Scholar] [CrossRef]
  273. Lotfi-Emran, S.; Ward, B.R.; Le, Q.T.; Pozez, A.L.; Manjili, M.H.; Woodfolk, J.A.; Schwartz, L.B. Human mast cells present antigen to autologous CD4+ T cells. J. Allergy Clin. Immunol. 2018, 141, 311–321.e10. [Google Scholar] [CrossRef] [Green Version]
  274. Ono, H.K.; Nishizawa, M.; Yamamoto, Y.; Hu, D.L.; Nakane, A.; Shinagawa, K.; Omoe, K. Submucosal mast cells in the gastrointestinal tract are a target of staphylococcal enterotoxin type A. FEMS Immunol. Med. Microbiol. 2012, 64, 392–402. [Google Scholar] [CrossRef] [Green Version]
  275. Scheuber, P.H.; Denzlinger, C.; Wilker, D.; Beck, G.; Keppler, D.; Hammer, D.K. Staphylococcal enterotoxin B as a nonimmunological mast cell stimulus in primates: The role of endogenous cysteinyl leukotrienes. Int. Arch. Allergy Immunol. 1987, 82, 289–291. [Google Scholar] [CrossRef]
  276. Shaler, C.R.; Choi, J.; Rudak, P.T.; Memarnejadian, A.; Szabo, P.A.; Tun-Abraham, M.E.; Rossjohn, J.; Corbett, A.J.; McCluskey, J.; McCormick, J.K.; et al. MAIT cells launch a rapid, robust and distinct hyperinflammatory response to bacterial superantigens and quickly acquire an anergic phenotype that impedes their cognate antimicrobial function: Defining a novel mechanism of superantigen-induced immunopathology and immunosuppression. PLoS Biol. 2017, 15, e2001930. [Google Scholar] [CrossRef]
  277. Godfrey, D.I.; Uldrich, A.P.; Mccluskey, J.; Rossjohn, J.; Moody, D.B. The burgeoning family of unconventional T cells. Nat. Publ. Gr. 2015, 16, 1114–1123. [Google Scholar] [CrossRef]
  278. D’Souza, C.; Chen, Z.; Corbett, A.J. Revealing the protective and pathogenic potential of MAIT cells. Mol. Immunol. 2018, 103, 46–54. [Google Scholar] [CrossRef]
  279. Godfrey, D.I.; Koay, H.F.; McCluskey, J.; Gherardin, N.A. The biology and functional importance of MAIT cells. Nat. Immunol. 2019, 20, 1110–1128. [Google Scholar] [CrossRef]
  280. Langley, R.; Patel, D.; Jackson, N.; Clow, F.; Fraser, J.D. Staphylococcal superantigen super-domains in immune evasion. Crit. Rev. Immunol. 2010, 30, 149–165. [Google Scholar] [CrossRef]
  281. Langley, R.J.; Ting, Y.T.; Clow, F.; Young, P.G.; Radcliff, F.J.; Choi, J.M.; Sequeira, R.P.; Holtfreter, S.; Baker, H.; Fraser, J.D. Staphylococcal enterotoxin-like X (SElX) is a unique superantigen with functional features of two major families of staphylococcal virulence factors. PLoS Pathog. 2017, 13, e1006549. [Google Scholar] [CrossRef] [Green Version]
  282. Tuffs, S.W.; James, D.B.A.; Bestebroer, J.; Richards, A.C.; Goncheva, M.I.; O’Shea, M.; Wee, B.A.; Seo, K.S.; Schlievert, P.M.; Lengeling, A.; et al. The Staphylococcus aureus superantigen SElX is a bifunctional toxin that inhibits neutrophil function. PLoS Pathog. 2017, 13, e1006461. [Google Scholar] [CrossRef] [Green Version]
  283. Spaan, A.N.; Surewaard, B.G.J.; Nijland, R.; van Strijp, J.A.G. Neutrophils versus Staphylococcus aureus: A Biological Tug of War. Annu. Rev. Microbiol. 2013, 67, 629–650. [Google Scholar] [CrossRef] [Green Version]
  284. Chen, J.Y. Superantigens, superantigen-like proteins and superantigen derivatives for cancer treatment. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 1622–1630. [Google Scholar]
  285. Liu, X.; Zeng, L.; Zhao, Z.; He, J.; Xie, Y.; Xiao, L.; Wang, S.; Zhang, J.; Zou, Z.; He, Y.; et al. PBMC activation via the ERK and STAT signaling pathways enhances the anti-tumor activity of Staphylococcal enterotoxin A. Mol. Cell. Biochem. 2017, 434, 75–87. [Google Scholar] [CrossRef]
  286. Perabo, F.G.E.; Willert, P.L.; Wirger, A.; Schmidt, D.H.; Von Ruecker, A.; Mueller, S.C. Superantigen-activated Mononuclear Cells Induce Apoptosis in Transitional Cell Carcinoma. Anticancer Res. 2005, 25, 3565–3573. [Google Scholar]
  287. Kodama, H.; Suzuki, M.; Katayose, Y.; Shinoda, M.; Sakurai, N.; Takemura, S.I.; Yoshida, H.; Saeki, H.; Ichiyama, M.; Tsumoto, K.; et al. Mutated SEA-D227A-conjugated antibodies greatly enhance antitumor activity against MUC1-expressing bile duct carcinoma. Cancer Immunol. Immunother. 2001, 50, 539–548. [Google Scholar] [CrossRef]
  288. Zhang, J.; Cai, Y.M.; Xu, M.K.; Song, Z.H.; Li, C.Y.; Wang, H.R.; Dai, H.H.; Zhang, Z.P.; Liu, C.X. Anti-tumor activity and immunogenicity of a mutated staphylococcal enterotoxin C2. Pharmazie 2013, 68, 359–364. [Google Scholar] [CrossRef]
  289. Forsberg, G.; Skartved, N.J.; Wallén-Öhman, M.; Nyhlén, H.C.; Behm, K.; Hedlund, G.; Nederman, T. Naptumomab estafenatox, an engineered antibody-superantigen fusion protein with low toxicity and reduced antigenicity. J. Immunother. 2010, 33, 492–499. [Google Scholar] [CrossRef]
  290. Golob-Urbanc, A.; Rajčević, U.; Strmšek, Ž.; Jerala, R. Design of split superantigen fusion proteins for cancer immunotherapy. J. Biol. Chem. 2019, 294, 6294–6305. [Google Scholar] [CrossRef] [Green Version]
  291. Knopick, P.; Terman, D.; Riha, N.; Alvine, T.; Larson, R.; Badiou, C.; Lina, G.; Ballantyne, J.; Bradley, D. Endogenous HLA-DQ8αβ programs superantigens (SEG/SEI) to silence toxicity and unleash a tumoricidal network with long-term melanoma survival. J. Immunother. Cancer 2020, 8, e001493. [Google Scholar] [CrossRef]
  292. Antonoglou, M.B.; Sánchez Alberti, A.; Redolfi, D.M.; Bivona, A.E.; Fernández Lynch, M.J.; Noli Truant, S.; Sarratea, M.B.; López, L.V.I.; Malchiodi, E.L.; Fernández, M.M. Heterologous Chimeric Construct Comprising a Modified Bacterial Superantigen and a Cruzipain Domain Confers Protection Against Trypanosoma cruzi Infection. Front. Immunol. 2020, 1, 1279. [Google Scholar] [CrossRef]
Figure 1. Structural features of Staphylococcal superantigens. (A) Overall structure of staphylococcal enterotoxin G (SEG) PDB accession number 1XXG. The general structure of SEG is displayed as a cartoon, and the secondary structures are colored yellow, β strands; red, α helixes; and flexible loops, green. The N-terminal domain and the C-terminal extreme are both located in the same domain of the molecule. (B) Superimposition of SEG and SEB. SEG overall structure (red) was superimposed over SEB structure (green) with an RMS of 0.714 Å, suggesting high similarity. (C) Overall structure of TSST-1, showing a simpler general structure than that of other staphylococcal enterotoxins. All the figures were performed using the PyMOL software.
Figure 1. Structural features of Staphylococcal superantigens. (A) Overall structure of staphylococcal enterotoxin G (SEG) PDB accession number 1XXG. The general structure of SEG is displayed as a cartoon, and the secondary structures are colored yellow, β strands; red, α helixes; and flexible loops, green. The N-terminal domain and the C-terminal extreme are both located in the same domain of the molecule. (B) Superimposition of SEG and SEB. SEG overall structure (red) was superimposed over SEB structure (green) with an RMS of 0.714 Å, suggesting high similarity. (C) Overall structure of TSST-1, showing a simpler general structure than that of other staphylococcal enterotoxins. All the figures were performed using the PyMOL software.
Toxins 14 00800 g001
Figure 2. Structure of the SAg-HA-HLA DR1 complex. (A) Ribbon diagram of SEB-HA-HLA DR1 complex. (B) The interface of the interaction is shown in detail. SEB compromises residues in positions 43, 44, 45, 46, 47, 67, 89, 92, 94, 95, 115 and 209. HLA DR1α chain, involves the residues: 13, 17, 18, 36, 37, 39, 57, 60, 61, 63, 67 and 68. Non-contacts are found between the peptide and SEB or SEB with the DR1β chain. (C) Ribbon diagram of the SEI-HA-HLA DR1 complex. (D) The interface of the interaction is shown in detail. The interaction is coordinated by Zn2+. This metal ion interacts with His81 of the DR1β chain and His169, His207 and Asp209 of SEI. SEI compromises residues in positions 98, 100, 105 and 211 to contact the residues 307 and 309 of the hemagglutinin (HA). No contacts are found between SEI and the DR1α chain. In all panels, the superantigen is colored red; the HLAD1α chain, blue; and the DR1β chain, orange. Zn2+ is represented as a sphere in cyan and the HA peptide, green. The residues conforming the interaction surface are represented as balls and sticks and colored pink (SEB or SEI); cyan, HLAD1α chain; yellow, DR1β chain; and light green, HA peptide. The figures were performed using PyMOL and the analysis of the structures was carried out using the CCP4i suite program.
Figure 2. Structure of the SAg-HA-HLA DR1 complex. (A) Ribbon diagram of SEB-HA-HLA DR1 complex. (B) The interface of the interaction is shown in detail. SEB compromises residues in positions 43, 44, 45, 46, 47, 67, 89, 92, 94, 95, 115 and 209. HLA DR1α chain, involves the residues: 13, 17, 18, 36, 37, 39, 57, 60, 61, 63, 67 and 68. Non-contacts are found between the peptide and SEB or SEB with the DR1β chain. (C) Ribbon diagram of the SEI-HA-HLA DR1 complex. (D) The interface of the interaction is shown in detail. The interaction is coordinated by Zn2+. This metal ion interacts with His81 of the DR1β chain and His169, His207 and Asp209 of SEI. SEI compromises residues in positions 98, 100, 105 and 211 to contact the residues 307 and 309 of the hemagglutinin (HA). No contacts are found between SEI and the DR1α chain. In all panels, the superantigen is colored red; the HLAD1α chain, blue; and the DR1β chain, orange. Zn2+ is represented as a sphere in cyan and the HA peptide, green. The residues conforming the interaction surface are represented as balls and sticks and colored pink (SEB or SEI); cyan, HLAD1α chain; yellow, DR1β chain; and light green, HA peptide. The figures were performed using PyMOL and the analysis of the structures was carried out using the CCP4i suite program.
Toxins 14 00800 g002
Figure 3. Crystallographic structures of the SAg-TCR interaction. (A) Ribbon diagram of SEG-mVβ8.2 complex. SEG is colored red and the TCR β chain, yellow (B) as shown in detail. SEG residues are colored pink and mVβ8.2 residues, wheat. Residues are indicated with a one letter code and numbered. (C) Ribbon diagram of the SEH–human TCR complex. SEH is colored red and the TCR, light blue (α chain) and yellow (β chain). (D) The interface of the interaction is shown in detail. SEH residues are colored hot pink, hVα27 chain residues are colored violet and hVβ19 residues are colored wheat. Residues are indicated with a one letter code and numbered. (E) Ribbon diagram of the SEH-TCR-MHC-II tri molecular complex. SEH is colored red, HLA-HA-DR1 is colored blue (α chain) and orange (β chain) and the TCR as indicated in C. (F) Superimposition of the SEI-HA-HLADR1 complex over the trimolecular complex using the DR1 as template. SEI shown in pink is clearly away from the interaction surface with the TCR α chain. The figures were performed using PyMOL and the analyses of the structures were carried out using the CCP4i suite program.
Figure 3. Crystallographic structures of the SAg-TCR interaction. (A) Ribbon diagram of SEG-mVβ8.2 complex. SEG is colored red and the TCR β chain, yellow (B) as shown in detail. SEG residues are colored pink and mVβ8.2 residues, wheat. Residues are indicated with a one letter code and numbered. (C) Ribbon diagram of the SEH–human TCR complex. SEH is colored red and the TCR, light blue (α chain) and yellow (β chain). (D) The interface of the interaction is shown in detail. SEH residues are colored hot pink, hVα27 chain residues are colored violet and hVβ19 residues are colored wheat. Residues are indicated with a one letter code and numbered. (E) Ribbon diagram of the SEH-TCR-MHC-II tri molecular complex. SEH is colored red, HLA-HA-DR1 is colored blue (α chain) and orange (β chain) and the TCR as indicated in C. (F) Superimposition of the SEI-HA-HLADR1 complex over the trimolecular complex using the DR1 as template. SEI shown in pink is clearly away from the interaction surface with the TCR α chain. The figures were performed using PyMOL and the analyses of the structures were carried out using the CCP4i suite program.
Toxins 14 00800 g003
Table 1. Staphylococcal SAgs.
Table 1. Staphylococcal SAgs.
SAgTypeYear of DiscoveryPhylogenetic Group
SEAClassical1962III
SEBClassical1962II
SECClassical1965II
SEDClassical1967III
SEEClassical1971III
TSST-1 (SEF)Classical1981II
SEGNew1998I
SEHNew1994III
SEINew1998V
SElJNew1998III
SEKNew2001V
SELNew2001V
SEMNew2001V
SENNew2001III
SEONew2001III
SEPNew2005III
SEQNew2002V
SERNew2004II
SESNew2008III
SETNew2008I
SElUNew2003II
SElVNew2006V
SElWNew2012III
SElXNew2011I
SElYNew2015I
SElZNew2015I
SEl26New2018V
SEl27New2018II
SE01New2017III
SE02New2020II
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Noli Truant, S.; Redolfi, D.M.; Sarratea, M.B.; Malchiodi, E.L.; Fernández, M.M. Superantigens, a Paradox of the Immune Response. Toxins 2022, 14, 800. https://doi.org/10.3390/toxins14110800

AMA Style

Noli Truant S, Redolfi DM, Sarratea MB, Malchiodi EL, Fernández MM. Superantigens, a Paradox of the Immune Response. Toxins. 2022; 14(11):800. https://doi.org/10.3390/toxins14110800

Chicago/Turabian Style

Noli Truant, Sofia, Daniela María Redolfi, María Belén Sarratea, Emilio Luis Malchiodi, and Marisa Mariel Fernández. 2022. "Superantigens, a Paradox of the Immune Response" Toxins 14, no. 11: 800. https://doi.org/10.3390/toxins14110800

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop