Next Article in Journal
A Rabbit Model for the Evaluation of Drugs for Treating the Chronic Phase of Botulism
Previous Article in Journal
Formulation and Safety Tests of a Wickerhamomyces anomalus–Based Product: Potential Use of Killer Toxins of a Mosquito Symbiotic Yeast to Limit Malaria Transmission
Previous Article in Special Issue
In-Host Emergence of Linezolid Resistance in a Complex Pattern of Toxic Shock Syndrome Toxin-1-Positive Methicillin-Resistant Staphylococcus aureus Colonization in Siblings with Cystic Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments

1
Laboratory of Pathogen Host Interactions, CNRS UMR5235, Université de Montpellier, 34000 Montpellier, France
2
Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Infectious and Tropical Diseases, Université de Montpellier, 30908 Nîmes, France
3
Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, 30908 Nîmes, France
4
Virulence Bactérienne et Infections Chroniques, INSERM U1047, Department of Microbiology and Hospital Hygiene, Université de Montpellier, 30908 Nîmes, France
*
Author to whom correspondence should be addressed.
Submission received: 16 June 2021 / Revised: 18 September 2021 / Accepted: 21 September 2021 / Published: 23 September 2021
(This article belongs to the Special Issue Resistance to Staphylococcus aureus Toxins)

Abstract

:
Staphylococcus aureus is a clinically important pathogen that causes a wide range of human infections, from minor skin infections to severe tissue infection and sepsis. S. aureus has a high level of antibiotic resistance and is a common cause of infections in hospitals and the community. The rising prevalence of community-acquired methicillin-resistant S. aureus (CA-MRSA), combined with the important severity of S. aureus infections in general, has resulted in the frequent use of anti-staphylococcal antibiotics, leading to increasing resistance rates. Antibiotic-resistant S. aureus continues to be a major health concern, necessitating the development of novel therapeutic strategies. S. aureus uses a wide range of virulence factors, such as toxins, to develop an infection in the host. Recently, anti-virulence treatments that directly or indirectly neutralize S. aureus toxins have showed promise. In this review, we provide an update on toxin pathogenic characteristics, as well as anti-toxin therapeutical strategies.
Key Contribution: This review described the main toxins produced by Staphylococcus aureus and discussed anti-toxin strategies to fight these bacteria.

1. Introduction

Staphylococcus aureus continues to be one of the most involved bacteria in human diseases. This bacteria is found in the normal skin microbiota of both animals and humans, with a carriage rate between 20 and 30% in the healthy human population [1,2]. Abscesses, lung infections, bacteremia, endocarditis, and osteomyelitis are all caused by S. aureus infections in humans [3]. With the appearance of methicillin-resistant S. aureus (MRSA) strains, the pathogenicity of S. aureus has become a problem in both health institutions and community settings. MRSA is on the rise since its discovery in the early 1960s, although there has been some stabilization or decline in European countries [4]. However, MRSA remains an important opportunistic pathogen in Europe and the most frequently identified worldwide [5]. MRSA is prevalent in several hospitals, especially those in Europe, Asia, and the United States. The prevalence of CA-MRSA strains from community-acquired (CA) infections among previously healthy individuals with few or no traditional healthcare-associated (HA) risk factors for MRSA increased in the late 1990s.
S. aureus infections rely on the production of surface proteins that initiate bacterial adherence to host tissues, the secretion of extracellular toxins and enzymes that destroy host cells and tissues, the avoidance or inactivation of the host immune system, and the growth and expansion of bacteria in host cells and tissue [6]. Coagulase, hyaluronidase, deoxyribonuclease, and lipase are some of the enzymes that S. aureus can synthesize to enhance its pathogenicity and disseminate within the host [7]. Moreover, enterotoxins, toxic shock syndrome toxin 1 (TSST-1), exfoliative toxins (ETs), hemolysins, epidermal cell differentiation inhibitors (EDINs), and Panton–Valentine leukocidin (PVL) have all been identified as extracellular protein toxins that enhance pathogenicity [8]. Interestingly, some of these toxins were detected in MRSA infections more frequently than non-MRSA cases [9,10,11].
Hospitalizations related to staphylococcal infections are frequent, increasing mortality and health costs [12,13]. Moreover, S. aureus’ capacity to produce antibiotic-neutralizing enzymes has exacerbated the issues associated with antimicrobial therapy, resulting in numerous resistances to these drugs [14]. Antibiotic resistance enzymes play a significant role in bacterial resistance to antibiotic pressure regarding diversity, evolution, and spread. Antibiotic-producing bacteria need strategies to counteract the chemicals’ deadly effects, by the production of degradative enzymes [14,15]. However, the selection pressure caused by the widespread use of antibiotics in humans and animals propagated resistant bacterial clones.
Antibiotic resistance develops quickly in S. aureus, and the rise of multidrug-resistant forms is a major problem. It has been reported that the annual mortality toll from antibiotic-resistant diseases has surpassed 10 million and that by 2050, it will outnumber cancer deaths [16]. The morbidity and mortality consequences reinforce the need to urgently discover new effective solutions due to the inefficiency of traditional antibiotics. Therefore, alternative treatments represent a promising field of investigation due to the lack of new antibiotic classes. Different strategies have been conducted, notably based on drug design with synthetic analogs, that could inhibit virulence factors. However, these studies have not yet generated promising results due to toxicity and/or low bioavailability. New options are now under study with a focus on biological molecules or compounds to interfere with toxins or toxin-regulator genes, constituting a new generation of promising anti-staphylococcal treatments [17,18,19,20,21].
This review outlines key properties related to the pathogenic roles of numerous S. aureus toxins (Table 1), as well as up to date anti-toxin treatments (Table 2).

2. Toxins Involved in the Pathogenicity of S. aureus

2.1. Staphylococcal Pore-Forming Toxins (PFTs)

PFTs are a type of bacterial virulence factor found in a wide range of human diseases, including S. aureus, which uses a variety of pore-forming cytotoxins (i.e., hemolysins, leukotoxins, and phenol-soluble modulins) to create pores in the host cell membrane causing cell lysis or to disrupt host cell actin cytoskeleton creating breaches in endothelial cells (EDIN exotoxin).

2.1.1. Hemolysins

S. aureus encodes α-, β-, γ-, and δ-hemolysins, which are regulated by the accessory gene regulator (Agr) and, principally, lyse erythrocytes by creating pores in host cell membranes or dissolving cell wall components [22]. The best-studied virulence factor of S. aureus is α-hemolysin, encoded by the hla gene, causing damage to a large variety of host cells, such as epithelial cells, endothelial cells, erythrocytes, monocytes, and keratinocytes, as well as causing cell membrane damage and apoptosis [22]. It is the prototype for the small β-barrel class pore-forming cytotoxins that is secreted as a 33 kDa water-soluble monomer that forms a prepore by assembling into one homoheptamer. Then, this prepore matures as a β-barrel transmembrane aqueous channel [23]. Finally, the binding of α-hemolysin to its host receptor ADAM10 stimulates ADAM10’s metalloprotease activity, allowing it to cleave endothelial cadherin, compromising endothelial barrier function [24]. Cellular reactions, including the release of potent lipid mediators originating from the arachidonate cascade, are then activated by the transport of ions such as Ca2+ through the pore, resulting in the target cells apoptosis [25].
The large majority of S. aureus strains (95%) possess the hla gene, irrespective of their resistance to methicillin, without showing a specific repartition in S. aureus clones nor a higher prevalence in certain regions of the world [26]. The role of the α-hemolysin toxin in the development of severe infections, such as pneumonia, osteomyelitis, and bacteremia, has been established in studies employing different experimental models infected with the S. aureus USA300 strain [27,28]. Interestingly, even though most recent S. aureus strains encode hla, data suggest that greater hla expression promotes pathogenicity. For instance, in a rat model of pneumonia, the epidemic strain USA300, which supplanted USA400 to become the dominant community-acquired methicillin-resistant S. aureus (CA MRSA) strain in the United States during the early 2000s, was reported to be significantly more virulent and fatal than USA400 and was strongly correlated with a substantial increase in hla expression [27,29].
Therefore, based on its crucial role in virulence, the α-hemolysin toxin is an ideal target for the development of anti-toxin treatments against S. aureus.

2.1.2. Panton-Valentine Leukocidin (PVL)

Leukotoxins target white blood cells, such as neutrophils, monocytes, or macrophages [30]. Panton–Valentine leukocidin (PVL), LukDE, and LukAB (sometimes known as LukGH) are all members of the bi-component Luk toxin family, with PVL presenting a 100-fold higher leukocytotic activity than the others. This Luk toxin family includes 32–35 kDa leukotoxins, which are encoded on the core genome or phage and oligomerize to form a pore structure [31]. Leukotoxins’ leukocytotic activity is based on receptor interaction. CCR5 on immune cells is the receptor for LukDE, whereas C5aR, C5L2, and CD11b are the receptors for PVL and LukAB [32,33,34]. PVL is a toxin that is made of two parts: LukS-PV and LukF-PV. These two components are excreted before assembling into a pore-forming heptamer on neutrophil membranes, resulting in their lysis [35].
PVL is primarily linked to skin and soft tissue disease, with other types of invasive disease, such as pneumonia, musculoskeletal disease, and bacteremia, being far less common. Infection with a PVL-positive strain does not appear to predict a poor clinical outcome for staphylococcal pneumonia, musculoskeletal disease, or bacteremia in adults, but patients with PVL-positive skin and soft tissue disease seem to be more likely to require surgical intervention [36]. PVL is linked to skin and soft tissue infections in both MRSA and MSSA strains, irrespective of the strain type [36,37,38].
Therefore, new treatments are required because the likelihood of infection with PVL-positive S. aureus strains is rising, and some of these strains are MRSA that already have limited treatment options.

2.1.3. Phenol-Soluble Modulins (PSMs)

PSMs are one of the most important and aggressive virulence factors in S. aureus involved in a variety of staphylococcal pathogenesis, such as red and white blood cell lysis, inflammatory response induction, and antimicrobial activities [39,40,41]. Moreover, while PSMs are reported to be the most cytolytic and immunological modulating factors, they all play a function in epithelial surface spreading and have also been associated with the structuring and detachment of biofilms [42,43]. S. aureus produces a variety of PSMs, each with unique cytolytic and antibacterial characteristics [41]. These toxins are a class of small peptides with an amphipathic α-helical structure and surfactant-like characteristics [41]. PSMs are categorized into two subfamilies: (i) PSMα peptides that are 20–26 amino acids long and contain PSMα1–PSMα4 and the δ-toxin and (ii) PSMβ peptides that are 43–44 amino acids in length and contain PSMβ1 and PSMβ2 [39]. The PSMα and β peptides are encoded in the psmα and psmβ operon, while the δ-toxin gene is within the sequence of RNAIII, the effector molecule of the Agr (accessory gene regulator) quorum-sensing pathway [44,45]. PSMs attach to the formyl peptide receptor 2 (FPR2), which attracts innate immune cells, such as neutrophils, macrophages, and dendritic cells [46,47]. As a result, holes formed in the host cell membrane cause osmotic instability and cell lysis. PSMα peptides have shown their great capacity to lyse human leukocytes and erythrocytes, with PSMα3 having the most important activity. The δ-toxin, on the other hand, has a mild cytolytic activity, while the PSMβ peptides are non-cytolytic [40].
PSMs are mainly present in highly virulent S. aureus, notably CA-MRSA. In vitro, these strains show a greater expression of PSMs, particularly cytolytic PSMα peptides, than that of hospital-acquired MRSA (HA-MRSA) strains [48]. In animal infection models, the PSMα peptides generated by the CA-MRSA USA300 and USA400 have a significant impact on the ability of virulent S. aureus to generate cutaneous infection and bacteremia [49,50,51].
Therefore, targeting PSMs for anti-staphylococcal treatment and drug development would be beneficial since eliminating all PSMs’ cytolytic and pro-inflammatory activities would lower their potency against host cells and possibly their overall contribution to S. aureus disease progression.

2.1.4. Epidermal Cell Differentiation Inhibitor (EDIN) Exotoxins

To date, three forms of EDIN toxins have been identified: EDIN-A, EDIN-B, and EDIN-C [52,53]. EDINs enter host cells and induce macroapertures, which are large and temporary transcellular tunnels within endothelial cells, thus compromising the integrity of the endothelium barrier, and then target and inhibit the small host protein RhoA [54,55]. This small GTPase is a critical regulator of the actin cytoskeleton in the host cell [56]. The inhibition of RhoA has been shown in several cell biology studies to have a negative effect on the cohesiveness of the epithelial and endothelium barrier, thus favoring bacterial spread [54,57]. In addition, RhoA inhibition suppresses complement-mediated phagocytosis [58]. Overall, a significant number of research exploring the effects of RhoA inhibition indicate that EDINs secreted factors play an important role in S. aureus colonization and bacterial host tissue invasion [59,60].
Numerous pathogenic strains of S. aureus, especially those from the European MRSA lineage (ST80-MRSA-IV) [53], express EDIN or EDIN-like exotoxins [61,62,63]. The prevalence of these genes in S. aureus is poorly described. Though, in diabetic foot ulcers, S. aureus isolates were positive for edin (A and B) genes in 14 (7.2%) out of 195 patients [53]. A prevalence of 14 % of EDIN-encoding genes (primarily edinC) was found in 256 S. aureus isolates from diverse clinical sites of infection in Nice (France) [64]. Interestingly, the association between PVL and EDIN among MRSA has been observed with a prevalence varying between 12 and 100% [65].
EDIN exotoxins are thus important virulence factors in promoting bacterial colonization and host tissue invasion, such as in diabetic foot infections, bacteremia, and pneumonia [53,66,67].

2.2. Exfoliative Toxins (ETs)

Staphylococcal exfoliative toxins (ETs) are responsible for staphylococcal scalded skin syndrome (SSSS), also known as Ritter’s disease, and characterized by dehydration, the loss of superficial skin layers, and secondary infections [68]. Large areas of the body are affected by SSSS, and the lesions are frequently sterile. Bullous impetigo is a skin disease caused by the same exfoliative toxin that causes SSSS, generated by the same underlying infection, and most commonly affects the face, hands, trunk, and buttocks. Pustules and blisters grow near the original site of infection in bullous impetigo but not elsewhere on the body as in SSSS. Because the blisters and pustules originate so close to the epidermis’ surface, they never grow larger than a few millimeters before perforating and expanding at the border, where oozing and yellow crusting develop, and the infection might disseminate to the surrounding skin when patients rub the rash [69]. Therefore, the only difference between the two disorders is the level of skin damage. Bullous impetigo usually affects young children and infants, although redness and rashes do not develop as they would in SSSS because older children and adults possess neutralizing antibodies that inactivate the toxin [69]. SSSS also primarily affects newborns and infants, although it can also impact adults with renal insufficiency or immunological deficiencies [68]
ETA, ETB, ETC, and ETD are the most common ETs, while ETA and ETB have received most of the attention due to their link to SSSS [68]. ETs are encoded on various genetic elements, and their expression is controlled by the accessory gene regulator (Agr) [31,70]. ETs exhibit glutamate-specific serine protease activity and target desmoglein 1 (Dsg1; desmosomal intercellular adhesion molecule), a keratinocyte cell–cell adhesion protein [71]. ETs bind to Dsg1, destroying desmosomal cell attachments and causing epidermal dissociation of the human epidermis [72]. The rupture of epidermal layers allows bacteria to penetrate the skin and induce blistering disorders, such as bullous impetigo and SSSS [73].
The prevalence of ETA in methicillin-resistant (MRSA) and methicillin-susceptible (MSSA) strains does not differ considerably. According to recent studies, 4% of MSSA strains possess the eta or etb gene, while about 10% of MRSA strains are eta positive [74,75]. Resistant strains, however, may pose a problem in the future. For instance, in Japan, issues with treating etb-positive CA-MRSA infections that causes SSSS in healthy persons have already been described [76,77].

2.3. Superantigens (SAgs)

S. aureus superantigens (SAgs) are the most effective T-cell mitogens. The mechanism of action of SAgs varies from those of traditional peptide antigens. Antigen-presenting cells (APCs) ingest and process conventional antigens [78]. T-cells are able to identify an MHC class II-restricted antigenic peptide exposed on APC surfaces utilizing hypervariable regions of T-cell receptor (TCR) α- and β-chains [78]. However, SAgs can directly link TCR β-domains by exploiting conserved MHC class II structures displayed on APCs, then triggering T-cell activation and proliferation without the use of antigen processing [79]. This causes pro-inflammatory cytokines, including IL-2, IFN-γ, and TNF-α, to become overactive and release causing a multitude of side effects and symptoms, including the possibility of multi-system organ failure that is specific to each superantigen [79].
SAgs include staphylococcal enterotoxins (SEs) that have emetic effects after oral administration and the toxic shock syndrome toxin 1 (TSST-1) that does not have emetic properties [80].

2.3.1. Staphylococcal Enterotoxins (SEs)

SEs are 20–30 kDa released toxins that disrupt intestinal activity and induce staphylococcal food poisoning (SFP), which is characterized by nausea, vomiting, abdominal pain, and diarrhea without indications of toxic effects, such as fever or hypotension [81,82,83]. Based on antigenic heterogeneity, more than 20 SEs (SEA—SElV) have been discovered [81,84,85]. Although the receptors involved in the emetic response to SEs have not been discovered, clinical signs of SFP have been linked to inflammatory mediators, such as leukotriene B4 and prostaglandin E2, both of which are produced in response to SEs [86,87]. The stomach and upper small intestine present the most significant mucosa lesions, which are associated with neutrophil infiltrates in the epithelium and lamina propria, whereas the jejunum exhibits broken brush boundaries and enlarged crypts [88].
In some CA-MRSA infections, lethal sepsis, infective endocarditis, and kidney infections are critically dependent on a high level of staphylococcal enterotoxin C (SEC) [89]. While staphylococcal enterotoxin B (SEB) is associated with food poisoning, it has been studied for potential utilization as an inhaled biological weapon [88].

2.3.2. Toxic Shock Syndrome Toxin 1 (TSST-1)

Unlike SEs, TSST-1 (22-kD) does not trigger emesis but stimulates the release of substantial amounts of pro-inflammatory cytokines from the host T-cells and macrophages [90]. This cytokine outburst causes toxic shock syndrome (TSS) symptoms, such as high fever, rash, desquamation, hypotension, and hypovolemic shock, which can progress to multiorgan failure [91].
Given the growing development of MRSA infections associated with TSST-1 expression, it is therefore becoming more difficult to treat and may ultimately lead to death.
Table 1. Toxins secreted by S. aureus.
Table 1. Toxins secreted by S. aureus.
ToxinBiological Properties and FunctionAssociated DiseaseReferences
α-hemolysin
-
Pore-forming activity
-
Lysis of erythrocytes, leukocytes, epithelial cells, and fibroblasts
-
Pro-inflammatory properties
-
Pneumonia
-
Sepsis
[23,24,92,93]
Panton–Valentine
Leukocidin (PVL)
-
Pore-forming activity
-
Lysis of neutrophils, monocytes, macrophages
-
Pro-inflammatory properties
-
Pneumonia
-
Bacteremia
-
Necrotizing fasciitis
-
Skin and soft tissue infections
[30,31,94]
Phenol-Soluble Modulins
(PSMs)
-
Pore-forming activity
-
Lysis of erythrocytes, neutrophils, monocytes, bacterial protoplasts, spheroplasts
-
Pro-inflammatory properties
-
Promote biofilm formation
-
Bacteremia
-
Skin infection
[40,82,95,96]
Epidermal Cell Differentiation Inhibitor (EDIN)
-
Transcellular tunnel activity
-
Breaches in endothelial cells
-
Pneumonia
-
Bacteremia
-
Diabetic foot ulcer
[53,54,66]
Exfoliative Toxins
(ETs)
-
Serine protease activity
-
Disruption of the cell–cell adhesions and junctions of the epidermis cells
-
Staphylococcal scalded skin syndrome (SSSS)
[68,97]
Staphylococcal Enterotoxins (SEs)
-
Superantigen activity
-
Pro-inflammatory activity
-
Staphylococcal food poisoning
-
Toxic shock syndrome
[19,81,85]
Toxic Shock Syndrome Toxin 1 (TSST-1)
-
Superantigen activity
-
Pro-inflammatory activity
-
Toxic shock syndrome
[82,90,91]

3. Anti-Toxin Treatments

The growth and spread of antibiotic resistance among S. aureus strains emphasize the imperative need for the development of alternative treatments that do not exert selective pressure in order to avoid evolution toward multi-resistance, such as that experienced with antibiotics. Interestingly, toxin-targeting therapy has already been effective against a variety of pathogenic bacteria, including S. aureus [17]. The therapeutic treatments that neutralize or interfere with the expression of staphylococcal toxins are detailed in this section.

3.1. Antibodies

Antibodies are one of the most important anti-virulence strategies for neutralizing toxins. Unlike active immunization, which would necessitate multiple boosters and a lengthy time to produce optimal immune responses, passive immunization would give prompt treatment for infected patients, thereby reducing the severity of S. aureus infections.
For instance, attempts to neutralize the α-hemolysin toxin during the course of human infection are underway and are based on substantial evidence for its participation in pathogenesis in murine models, as well as its putative importance in human disease. MEDI4893 (suvratoxumab), an α-hemolysin-neutralizing monoclonal antibody (mAb) formerly called LC10, is among the best-studied anti-virulence treatments against S. aureus infections [21]. As described above, α-hemolysin interacts with the metalloprotease ADAM10, which promotes oligomerization and pore formation [98]. By binding to a highly conserved area of the α-hemolysin toxin, MEDI4893 inhibits its interaction with ADAM10 as well as it self-oligomerization, thus neutralizing its action [98,99,100]. The treatment of rabbits with MEDI4893 resulted in a considerable reduction in clinical outcomes, according to Le et al. [101]. Similarly, Ortines et al. found that S. aureus-infected mice previously passively immunized with MEDI4893 showed fewer wounds and decreased bacterial counts than those of untreated controls in non-diabetic and diabetic mice [102]. Surewaard et al. recently revealed that α-hemolysin causes rapid platelet aggregation and liver injury, resulting in multi-organ failure during S. aureus sepsis, but these consequences could be avoided in mice treated with MEDI4893 [103]. MEDI4893 has passed Phase 2 clinical studies for the prevention of S. aureus pneumonia in high-risk critical care unit patients in 2020, and more recently, MEDI4893 showed efficacy and safety in preventing S. aureus ventilator-associated pneumonia [104,105]. In addition, AR-301 and ASN100 are two other neutralizing antibodies that have entered clinical trials. AR-301 is an α-hemolysin-targeting monoclonal antibody that has recently entered Phase 3 tests as an adjuvant therapy for S. aureus pneumonia [106], and ASN100 is a combination of two monoclonal antibodies that neutralize six cytolytic toxins corresponding to α-hemolysin, PVL, LukAB, γ-hemolysin AB (HlgAB), γ -hemolysin CB (HlgCB), and leukocidin ED (LukED) [107]. Unfortunately, while ASN100 passed the Phase 1 clinical safety testing by reducing tissue damage in a rabbit model of S. aureus pneumonia, the Phase 2 trial was stopped due to inefficiency [107]. Although these trials involving MEDI4893 [108], AR-301 [106], and the multivalent antitoxin ASN100 [107] were not statistically significant, passive immunization was found to have some protective potential. For example, AR-30 shortened the time spent on mechanical ventilation, whilst MEDI4893 decreased hospital and intensive care unit stay, as well as antibiotic treatment duration, while remaining safe and well tolerated [106,107,108]. Foletti et al. identified antibodies against α-hemolysin from a human donor-derived single-chain variable fragment (scFv) phage library [99]. LTM14, a notable clone in this family, was transformed to a complete IgG and showed an unusually high affinity for α-hemolysin. LMT14 offered protection against S. aureus cutaneous and bacteremia mice models of infection and also demonstrated therapeutic potential in a pneumonia model [99]. In addition, when combined with the antibiotic linezolid, LTM14 showed improved efficacy. This is essential because, in a therapeutic situation, passive immune treatment will almost certainly be delivered in conjunction with the most suitable antibiotic [99].
While some studies have found an epidemiological link between PVL and CA-MRSA, the presence of large levels of neutralizing antibodies did not provide resistance to PVL-positive MRSA skin and soft tissue infections [109,110,111]. Despite this, PVL is still one of the most important targets for anti-toxin drug research. PVL-specific antibodies are present in available commercial human intravenous polyclonal immunoglobulin preparations (IVIg), which decrease the cytopathic effects of PVL in a dose-dependent manner, most likely through interfering with PVL–neutrophil interactions [112]. In 2015, Mairpady Shambat et al. showed that IVIg abolished PVL and α-hemolysin-mediated cytotoxicity in epithelial cells in a human lung tissue model [113]. Moreover, antibiotic therapy combined with IVIg anti-toxin treatment significantly improved the situation of patients with acute necrotizing pneumonia caused by PVL-positive S. aureus strains, demonstrating the effectiveness of IVIg in limiting disease progression, particularly in highly lethal S. aureus infections linked to PVL production [114]. Moreover, in vitro humanized antibodies developed by Leventie et al. are able to disrupt PVL binding to polymorphonuclear leukocytes and impede the development of new pores [115]. A reduction in inflammatory reactions and tissue damage was also found in this non-infectious rabbit model of endophthalmitis, with the tetravalent anti-PVL antibody [115].
In various in vivo models, antibodies targeting superantigens were shown to neutralize these toxins and have been linked to protection [116]. The staphylococcal enterotoxin B (SEB) is one of the most studied enterotoxins, and its designation as a bioweapon makes it an interesting target to produce anti-toxin-neutralizing antibodies. Drozdowski et al. generated and selected human monoclonal antibodies (HuMAbs) specific for SEB from human B-cell hybridomas [117]. In vitro, these antibodies exhibited biological activity against SEB, and, HuMAb-154, which had the highest anti-SEB affinity, demonstrated both preventive and therapeutic action in a mouse model of SEB-induced mortality [117]. In addition, in numerous mouse models, including sepsis and cutaneous and deep tissue abscesses, another monoclonal antibody against SEB, named 20B1, was demonstrated to be protective against MRSA infection [118].
Therefore, all these therapy examples highlight the usefulness of antibodies as anti-virulence treatments able to neutralize staphylococcal toxins.

3.2. Nanoparticles

Aside from the use of anti-toxin antibodies in anti-virulence therapies, researchers have also shown the effectiveness of manufactured nanoparticles that imitate cell membranes, such as liposomes, in sequestering bacterial toxins in vitro and in vivo [119]. For example, Henry et al. demonstrated the ability of artificial liposomes to trap bacterial toxins in vitro while maintaining the integrity of mammalian cells [120]. They also discovered that administering artificial liposomes to mice during in vivo studies helped them recover from septicemia induced by S. aureus, as well as protect them against pneumonia [120]. Because customized liposomes are made entirely of naturally occurring lipids, they are not bactericidal and could be employed alone or in combination with antibiotics to treat bacterial infections and reduce toxin-induced tissue damage [120]. Interestingly, Wolfmeier et al. employed sphingomyelin liposomes, with or without cholesterol, to neutralize secreted PSMs and other virulence factors in vitro during human blood or epithelial cell staphylococcal infections, as well as in a murine dermonecrosis model [121]. Sphingomyelin liposomes blocked cell lysis by PSMs, particularly PSM3, whereas cholesterol-containing sphingomyelin liposomes preferentially trapped α-hemolysin [121]. A combination of both liposome types was recently evaluated in a Phase I clinical trial against severe pneumococcal pneumonia, although its utility in S. aureus pneumonia remains unknown. Furthermore, targeting both PSMs and α-hemolysin at the same time remains a possibility, as PSMs have been demonstrated to regulate α-hemolysin expression both in vitro and in vivo [23].
Recently, in response to S. aureus infection, exosomes (called “defensosomes”) with increased ADAM10 receptors were discovered to be produced from host cells in a TLR-dependent mode, resulting in α-hemolysin retention and a reduction in disease mortality [122]. Then, therapeutic poly (lactic-co-glycolic acid) (PLGA)-based nanoparticles covered with natural human erythrocyte membranes performed a comparable decoy effect to fight infection and reduce the activity of α-hemolysin [123].
Moreover, similar to other anti-virulence strategies, the nanoparticle-based neutralization and administration not only help to avoid severe bacterial infections but can also participate in reducing the development of antibiotic resistance [124].

3.3. RNAIII-Inhibiting Peptides

In addition to the direct neutralization strategy outlined in the previous sections, targeting S. aureus toxins can be carried out indirectly by affecting the regulatory processes that govern virulence genes’ expression. This approach is based on the utilization of small molecules, such as peptides, to target global regulators, such as the accessory gene regulator Agr in S. aureus. Agr regulates the quorum-sensing pathway that controls whether S. aureus develops a biofilm or remains planktonic, as well as toxin gene synthesis [44,125,126]. The P3 promoter of the S. aureus quorum-sensing Agr system transcribes RNAIII, a stable regulatory RNA that regulates the expression of a large variety of virulence factors [127]. Therefore, inhibiting RNAIII represents a promising strategy for reducing toxin expression as well as other virulence factors. When evaluated in cellulitis in in vivo models, RNAIII-inhibiting peptides (RIP) were found to block agr RNA transcripts and impede staphylococcal adhesion to mammalian cells, resulting in a decrease in S. aureus pathogenicity [128,129,130]. In an MRSA sepsis model, two new RIP derivatives were recently discovered to substantially extend mouse survival and reduce pathological damage without impacting bacterial viability [131]. Interestingly, in an S. aureus-induced sepsis mouse model, and in association with clinically prescribed antibiotics, RIP increased the healing of wounds and reduced mortality in comparison to antibiotics alone, thus confirming the potential of combined therapies [132,133]. Moreover, PSMs have been specifically targeted using a variety of methods to neutralize their pathogenic effect. In a mouse pneumonia model, targeting PSMs indirectly by inhibiting the Agr system with an RNAIII-inhibiting peptide resulted in a lower bacterial load and mortality [134].

3.4. Antimicrobial Peptides (AMPs)

Antimicrobial peptides (AMPs) have been known for several decades and are part of the innate immunity of practically all living organisms, ranging from bacteria, insects, and plants to vertebrates [135]. As of August 2021, the antimicrobial peptide database (https://aps.unmc.edu/ (accessed on 23 August 2021)) contains 3273 antimicrobial peptides from six kingdoms (369 bacteriocins/peptide antibiotics from bacteria, 5 from archaea, 8 from protists, 22 from fungi, 361 from plants, and 2424 from animals, including some synthetic peptide records). AMPs have a wide spectrum of antibacterial, antifungal, antiparasitic, and antiviral properties [135]. AMPs not only possess a large spectrum of antibacterial activity but can also display anti-toxin activities [136,137]. Cathelicidins and defensins are the two major categories of AMPs in humans. Human defensins are amphipathic cationic peptides that are divided into two types: α- and β -defensins, and, to date, four α-defensins have been identified from polymorphonuclear neutrophils (PMNs) [138]. Human neutrophil peptides (HNP1–HNP4) are part of the phagolysosome’s microbicidal machinery that can be detected in the extracellular environment after degranulation [139]. PVL is thought to have a role in CA-MRSA pathogenesis by attracting and lysing PMNs at the infection site, which induces tissue damage caused by the release of cytotoxic granule constituents [140]. Interestingly, Cardot-Martin et al. found that HNP3 defensins, but not HNP-1 or -2, substantially protect neutrophils from PVL-induced lysis by interacting with LukS-PV and LukF-PV, which disables PVL’s pore creation function and reduces PVL cytotoxic effects [141].

3.5. Natural Compounds

Natural product-based compounds that present anti-toxin properties correspond to a promising therapeutical approach to treat S. aureus infections [18]. A modified cyclodextrin compound, named IB201, is used to treat pneumonia. Cyclodextrins are cyclic oligosaccharides that are made from starch or starch derivatives. Because of its spatial resemblance to α-hemolysin, this compound was identified based on the prediction that it would prevent α-hemolysin action with a high affinity [142]. Moreover, in mice S. aureus pneumonia models, aloe-emodin, an active component from aloe vera, and apigenin, an active compound from parsley, both exhibited sufficient protection [143,144]. Other compounds, such as morin hydrate (also known as 2′,3,4′,5,7-pentahydroxyflavone), which is a flavonoid present in Maclura pomifera (Osage orange), in Maclura tinctoria (old fustic), and in the leaves of Psidium guajava (common guava), was discovered to disrupt the self-assembly of the transmembrane pore of α-hemolysin in a mouse model of pneumonia and then to decrease its hemolytic activity [145].
Oroxin A (ORA), oroxin B (ORB), and oroxylin A 7-O-glucuronide (OLG), three oroxylin glycosides, are natural flavonoids found in strawberries, grapes, onions, apples, Bignoniaceae plants, and other vegetables and fruits. These substances possess structural similarities and bind to hemolysin’s stem domain, preventing it from transitioning from monomer to oligomer in vitro and inhibiting its hemolytic action [146,147].
Friedman et al. showed that the pure olive chemical 4-hydroxytyrosol and the commercialized olive powder Hidrox-12, containing 6% of 4-hydroxytyrosol and 6% of additional phenolic compounds, were able to suppress the biological action of the superantigen enterotoxin A (SEA) [148]. However, this effect still remains to be validated in animal models.
Solonamide B, a cyclodepsipeptide from the halotolerant bacterium Photobacterium halotolerans, was one of the first natural inhibitors of the Agr signaling pathway to be reported [149]. Solonamide B and its derivatives prevent the quorum-sensing peptide AIP from interacting with AgrC. Interestingly, in CA-MRSA strains, such as USA300, solonamide B strongly reduced the activity of α-hemolysin and the transcription of psma-encoding PSMs resulting in an 80% reduction in toxicity of supernatants toward human neutrophils and rabbit erythrocytes [150,151,152].
Isorhamnetin, chrysin, and puerarin have been shown to inhibit RNAIII transcription and, hence, α-hemolysin expression, providing protection against MRSA and MSSA-induced pneumonia [153,154,155]. Isorhamnetin (also known as 3′-methoxy-3,4′,5,7-tetrahydroxyflavone) is an O-methylated flavanol found in apples, blackberries, cherries, and pears, as well as in medicinal plants and herbs [155]. Honey, propolis, the passion flowers Passiflora caerulea and Passiflora incarnata, and Oroxylum indicum all contain chrysin (5, 7-dihydroxyflavone) [156]. Puerarin is the main bioactive compound obtained from the root plant Pueraria lobata Ohwi, also called Gegen in traditional Chinese medicine [157].
Naringenin, a flavanone found primarily in grapefruit, but also in a range of fruits and herbs, has been shown to drastically lower the amounts of agrA and hla transcripts in S. aureus culture as well as to inhibit hemolysin synthesis and protect mice from S. aureus-induced pneumonia [158,159].
In vitro, Castanea sativa leaf extract 224C-F2 [160] and Schinus terebinthifolia berry extract 430D-F5 [161] were found to inhibit agr expression, thus resulting in decreased hemolysin synthesis and hemolytic activity. A pretreatment with 224C-F2 diminished infection-induced ulcer sizes and substantially lowered morbidity in an in vivo model of MRSA infection [160]. In addition, pretreatment with a single dose of 430D-F5 massively reduced skin ulcer formation and mortality in a mouse model of MRSA skin infection [161].
Ambuic acid is a fungal product that targets AgrB activity and has been shown to prevent hemolysin and RNAIII production in MRSA infections [162]. A single preventive administration of ambuic acid totally prevented skin ulcer formation in a murine mouse model [162]. Another fungal product, named omega-hydroxyemodin (OHM) and produced by Penicillium restrictum, decreases Agr activity by disrupting AgrA binding to its promoter. Importantly, OHM particularly inhibited the Agr pathway activation in a mouse model of MRSA cutaneous infection without affecting the host [163,164].
Even though some of the molecular mechanisms of these natural compounds have yet to be determined, they provide possible novel scaffolds for the development of successful anti-virulence therapeutics toward S. aureus infection.

3.6. Vaccines

Despite numerous attempts, there is currently no vaccination against S. aureus. As mentioned in the first section, S. aureus secretes a broad range of toxins during colonization and infection of the host, which poses a challenge for vaccine development.
There have been several studies to investigate the efficacy of α-hemolysin as a vaccine agent. Indeed, H35L, a mutant isoform of α-hemolysin, was discovered to have minimal hemolytic action [165]. In a mouse pneumonia model, this inactivated toxin (toxoid) was studied in two separate models and found to be efficacious through both active vaccination and the development of protective rabbit anti α-hemolysin antibodies; despite this fact, no human trials have been conducted to date [166].
IBT-VO2 is a multivalent vaccine currently under investigation. α-hemolysin, PVL LukS, LukF, LukAB, enterotoxins A and B, and toxic shock syndrome toxin 1 toxoids are all included in this heptavalent vaccine [167]. The multi-subunit vaccine generates an antibody response that is cross-reactive with 12 to 15 S. aureus toxins and gives protection in different mice and rabbit infection models due to structural similarities [167]. Recently, IBT-VO2 entered a Phase I clinical study after completing the encouraging pre-clinical phase, and as a result, received additional funding to help it progress.
Previous studies in animal models suggested that PVL subunits could be useful vaccines, but these attempts have yet to be converted into human trials [168,169]. However, for the creation of the StaphVax vaccine, recombinant PVL subunits were exploited (Nabi Biopharmaceuticals, Alpharetta, GA), but this vaccine failed in Phase 3 clinical testing [170]. However, some of the vaccine antigens, including PVL, were recycled into a new vaccine called PentaStaph, which was acquired by GlaxoSmithKline Biologicals (GSK) [171,172].
In mice, an inactive isoform of the staphylococcal enterotoxin B (SEB) was cloned into an Lactococcus lactis strain and tested as an oral vaccine. The vaccination was able to generate a significant antibody response, thus resulting in improved survival of infected mice [173]. Another vaccine against this SEB toxin, named STEBVax, was also generated and corresponds to a recombinant isoform that impedes the toxin from interacting with the major histocompatibility complex (MHC) class II [174]. If effective, this vaccine could be useful as a polyvalent S. aureus vaccine in general.
In another study, the injection of TSST-1-specific antibodies to treat toxic shock syndrome has been found to reduce mortality in a septic mouse model of infection [175]. Moreover, a modified TSST-1 antigen was also exploited to create an attenuated TSST-1 vaccine able to prevent infection during sepsis in mice [175]. Subsequently, a recombinant TSST-1 variant vaccine was created and tested in a Phase I clinical study, which was quite well tolerated, and a Phase II trial was engaged [176,177].
Even though numerous vaccine candidates have demonstrated protective efficacy in preclinical or early clinical investigations as detailed above, no vaccine has been authorized to date for human use.

3.7. Others

Eukaryotes, archaea, and bacteria create extracellular vesicles (EVs), which are lipid bilayers that form lumen-containing spheres with diameters ranging from 20 to 500 nm. EVs contain a variety of proteins, polysaccharides, nucleic acids, and lipids. EVs from Gram-positive bacteria carry physiologically active toxins, display cytotoxicity, and stimulate proinflammatory mediators, thus having a significant role in host–pathogen interactions [178]. Unfortunately, the toxicity of staphylococcal EVs limited their use as a vaccine platform. However, in a recent study, Wang et al. engineered EVs with unique features in the S. aureus USA300 strain, representative of the dominant CA-MRSA clone in the United States [179]. The originality of this study was to consider that S. aureus EVs could be used as a vaccine platform if their cytotoxicity was reduced. Therefore, EVs over producing Hla- and LukE-modified toxins that possess the capacity to be immunogenic without being toxic were engineered in order to stimulate the production of toxin-neutralizing antibodies. Immunization with engineered EVs showed considerable protection in an S. aureus lethal sepsis model [179]. Though the efficiency of these vesicles as a novel vaccine platform against different S. aureus strains and in additional infection models has yet to be determined, they do represent an attractive promise.
Moreover, novel approaches have been developed to combat enterotoxins. In an in vitro T-cell experiment, Mattis et al. developed a yeast display technology to create a soluble T-cell receptor variable domain variation capable of neutralizing both SEC and SEB enterotoxins. In different rabbit models, including endocarditis and necrotizing pneumonia, this variation was proven to be effective in reducing the infection [180].
Table 2. Summary of the anti-toxin treatments strategies.
Table 2. Summary of the anti-toxin treatments strategies.
TreatmentNameTargetReferences
AntibodiesMEDI4893 (suvratoxumab)α-hemolysin [98,99,100]
AR-301α-hemolysin[106]
ASN100α-hemolysin, Panton–Valentine leukocidin (PVL), LukAB, Ɣ-hemolysin AB (HlgAB), Ɣ-hemolysin CB (HlgCB), leukocidin ED (LukED)[107]
LTM14α-hemolysin[99]
IVIgα-hemolysin, PVL[112,113,114]
HuMAb-154Staphylococcal enterotoxin B (SEB)[117]
20B1Staphylococcal enterotoxin B (SEB)[118]
NanoparticlesSphingomyelin liposomesPhenol-soluble modulins (PSMs)[121]
Cholesterol-containing sphingomyelin liposomesα-hemolysin[121]
Poly (lactic-co-glycolic acid) (PLGA)-based nanoparticles covered with natural human erythrocyte membranesα-hemolysin[123]
RNAIII-inhibiting peptidesRNAIII-inhibiting peptides (RIP)agr RNA transcripts[128,129,130]
Antimicrobial peptidesHNP3PVL[141]
Natural compoundsCyclodextrin IB201α-hemolysin[142]
Aloe-emodinα-hemolysin[144]
Apigeninα-hemolysin[143]
Morin hydrate
(2′,3,4′,5,7-pentahydroxyflavone)
α-hemolysin[145]
Oroxylin glycosides
(oroxin A (ORA), oroxin B (ORB), and oroxylin A 7-O-glucuronide (OLG))
α-hemolysin[146,147]
4-hydroxytyrosol
Hidrox-12
Staphylococcal enterotoxin A (SEA)[148]
Solonamide BQuorum-sensing peptide
(AIP)
[149,150,151,152]
Isorhamnetin
(3′-methoxy-3,4′,5,7-tetrahydroxyflavone)
α-hemolysin[155]
Chrysin
(5, 7-dihydroxyflavone)
α-hemolysin[156]
Puerarinα-hemolysin[157]
NaringeninagrA and hla expression[158,159]
224C-F2 (Castanea sativa leaf)
430D-F5 (Schinus terebinthifolia berry)
agr expression[161]
Ambuic acidAgrB activity, RNAIII expression[162]
Omega-hydroxyemodin (OHM)AgrA[163,164]
VaccinesH35Lα-hemolysin[165,166]
IBT-VO2α-hemolysin, PVL, enterotoxins A and B, toxic shock syndrome toxin 1 (TSST-1)[167]
StaphVaxPVL[170]
STEBVaxSEB[174]
Attenuated TSST-1 vaccineTSST-1[175,176,177]
OthersExtracellular vesicles (EVs)α-hemolysin, LukE[181]
Yeast display technology to create a soluble T-cell receptorSEC, SEB[180]

4. Conclusions and Future Directions

The problem of antibiotic resistance has prompted scientists around the world to explore alternatives for effective treatments. Because antimicrobial resistance is a complex phenomenon, the solution to this problem comprises a variety of techniques aimed at reducing the factors that contribute to the establishment of resistance and spread. These strategies require the development of novel therapeutic drugs that work on principles distinct from those currently available for antibiotics. Bacterial toxins, as detailed in this review, are directly involved in disease outcomes. Anti-toxin therapies have been proposed as a promising alternative in this regard, with the intention of reducing pathogen virulence without exposing pathogens to selective pressure.
Anti-toxin therapies target diseases that are the most dangerous to patients, such as hospital-acquired bacterial pneumonia and ventilator-associated bacterial pneumonia, osteomyelitis, sepsis, and endocarditis, and have the capacity to enhance chances of survival. One of the benefits of these anti-toxin treatments is their use in conjunction with antibiotics to help fight the most dangerous infections. Furthermore, anti-toxin therapies contribute to significantly reduce the bacterial load, most likely by interfering with the bacterial strategies used to multiply involving secreted toxins [182]. Moreover, anti-toxin treatments do not place selective pressure on bacterial growth since they neutralize the pathogen rather than killing it, which could provide a long-term solution to the resistance issue. However, the potential of anti-toxin treatments to counter drug resistance without putting severe selective pressure on the bacterial population needs more investigation.
Even though efforts to develop innovative anti-toxin compounds have had varying levels of success, the potential leads need pharmacology and toxicology evidence. For extensive mechanistic study, additional studies should concentrate on a few very promising candidates. The anti-toxin treatments potential negative effects must also be considered. Because of the toxins’ extensive combination and cross-reactivity, efforts to interfere at the host–toxin level necessitate robust anti-toxin efficacy. Therefore, staphylococcal toxin biology requires more research to decipher the specific toxin roles, the differences in expression or genetic existence of toxins all over strain lineages, and the importance of specific toxins along various clinical strains. Another drawback of anti-toxin compounds is their possible limited spectrum efficacy, as these treatment candidates only specifically target virulence-mediated pathways in certain S. aureus strains, thereby limiting their general clinical use.
Moreover, it is clear that evaluating the effectiveness of anti-toxin compounds is delayed by the lack of therapy models, which may more precisely mimic the clinical condition in humans. Thus, developing such models represents an essential future direction. Human clinical trials will always be required to prove the success of a treatment. However, even though animal models remain necessary to decipher fundamental host–pathogen interactions and even though many potentially promising S. aureus anti-toxin therapeutics exist, most have failed in human trials or have not been tested. Therefore, the development of humanized mice with engrafted human immune cells for instance could help improve the translatability of animal investigations to human trials in the future [183,184,185]. This strategy will improve animal models, thus helping in deciding which treatments should proceed to clinical trials.
It is also necessary to specify which criteria will be used to assess the anti-virulence therapy’s efficacy, as well as which types of infections the treatment is most suited for. For instance, a defective Agr–quorum sensing system appears to be favorable for the pathogen in S. aureus chronic infections or bacteremia [186]. Furthermore, it was recently demonstrated that a dysfunctional Agr system could facilitate antibiotic resistance to gentamicin and ciprofloxacin [187]. Moreover, phenol-soluble modulin toxins are known to be implicated in the regulation of S. aureus persister cell populations [188]. Then, as the Agr system oversees PSMs’ production, a misfunctioning system is likely to suppress PSM expression, favoring the formation of persister cells resistant to antibiotics.
To summarize, a tremendous amount of work has investigated S. aureus toxins, expanding our understanding of their mode of action and involvement in pathogenesis, and several promising therapies have resulted from various treatment strategies. However, improved therapeutical models need to be developed to validate most of these anti-toxin treatments.

Author Contributions

Conceptualization J.-P.L. and V.M.; investigation, N.A.-M. and V.M.; resources, P.L., C.P., and C.D.-R.; writing—original draft preparation, N.A.-M., J.-P.L., and V.M.; writing—review and editing, P.L., C.P., C.D.-R., and A.S.; visualization, N.A.-M., P.L., C.P., C.D.-R., and A.S.; supervision, J.-P.L. and V.M.; project administration, J.-P.L. and V.M.; funding acquisition, J.-P.L. and V.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Fondation de Coopération Scientifique, Méditerranée-Infection (Marseille IHU grant).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We thank the Nîmes University hospital for its structural, human, and financial support through the award obtained by our team during the internal call for tenders “Thématiques phares”. The authors belong to the FHU InCh (Federation Hospitalo Universitaire Infections Chroniques, Aviesan).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wertheim, H.F.; Melles, D.C.; Vos, M.C.; van Leeuwen, W.; van Belkum, A.; Verbrugh, H.A.; Nouwen, J.L. The Role of Nasal Carriage in Staphylococcus aureus. Lancet Infect. Dis. 2005, 5, 751–762. [Google Scholar] [CrossRef]
  2. Hanselman, B.A.; Kruth, S.A.; Rousseau, J.; Weese, J.S. Coagulase Positive Staphylococcal Colonization of Humans and Their Household Pets. Can. Vet. J. 2009, 50, 954–958. [Google Scholar] [PubMed]
  3. Tong, S.Y.C.; Davis, J.S.; Eichenberger, E.; Holland, T.L.; Fowler, V.G. Staphylococcus aureus Infections: Epidemiology, Pathophysiology, Clinical Manifestations, and Management. Clin. Microbiol. Rev. 2015, 28, 603–661. [Google Scholar] [CrossRef] [Green Version]
  4. Antimicrobial Resistance in the EU/EEA (EARS-Net)—Annual Epidemiological Report for 2019. Available online: https://www.ecdc.europa.eu/en/publications-data/surveillance-antimicrobial-resistance-europe-2019 (accessed on 23 August 2021).
  5. Borg, M.A.; Camilleri, L. What Is Driving the Epidemiology of Methicillin-Resistant Staphylococcus aureus Infections in Europe? Microb. Drug Resist. 2020, 27, 889–894. [Google Scholar] [CrossRef] [PubMed]
  6. Lowy, F.D. Staphylococcus aureus Infections. N. Engl. J. Med. 1998, 339, 520–532. [Google Scholar] [CrossRef] [PubMed]
  7. Tam, K.; Torres, V.J. Staphylococcus aureus Secreted Toxins and Extracellular Enzymes. Microbiol. Spectr. 2019, 7, 16. [Google Scholar] [CrossRef]
  8. Oliveira, D.; Borges, A.; Simões, M. Staphylococcus aureus Toxins and Their Molecular Activity in Infectious Diseases. Toxins 2018, 10, 252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Shimaoka, M.; Yoh, M.; Takarada, Y.; Yamamoto, K.; Honda, T. Detection of the Gene for Toxic Shock Syndrome Toxin 1 in Staphylococcus aureus by Enzyme-Labelled Oligonucleotide Probes. J. Med. Microbiol. 1996, 44, 215–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Liu, M.; Liu, J.; Guo, Y.; Zhang, Z. Characterization of Virulence Factors and Genetic Background of Staphylococcus aureus Isolated from Peking University People’s Hospital between 2005 and 2009. Curr. Microbiol. 2010, 61, 435–443. [Google Scholar] [CrossRef]
  11. Ezeamagu, C.; Imanatue, I.; Dosunmu, M.; Odeseye, A.; Baysah, G.; Aina, D.; Odutayo, F.; Mensah-Agyei, G. Detection of Methicillin Resistant and Toxin-Associated Genes in Staphylococcus aureus. Beni-Suef Univ. J. Basic Appl. Sci. 2018, 7, 92–97. [Google Scholar] [CrossRef]
  12. Suaya, J.A.; Mera, R.M.; Cassidy, A.; O’Hara, P.; Amrine-Madsen, H.; Burstin, S.; Miller, L.G. Incidence and Cost of Hospitalizations Associated with Staphylococcus aureus Skin and Soft Tissue Infections in the United States from 2001 through 2009. BMC Infect. Dis. 2014, 14, 296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zhen, X.; Lundborg, C.S.; Zhang, M.; Sun, X.; Li, Y.; Hu, X.; Gu, S.; Gu, Y.; Wei, J.; Dong, H. Clinical and Economic Impact of Methicillin-Resistant Staphylococcus aureus: A Multicentre Study in China. Sci. Rep. 2020, 10, 3900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Egorov, A.M.; Ulyashova, M.M.; Rubtsova, M.Y. Bacterial Enzymes and Antibiotic Resistance. Acta Naturae 2018, 10, 33–48. [Google Scholar] [CrossRef] [Green Version]
  15. Varela, M.F.; Stephen, J.; Lekshmi, M.; Ojha, M.; Wenzel, N.; Sanford, L.M.; Hernandez, A.J.; Parvathi, A.; Kumar, S.H. Bacterial Resistance to Antimicrobial Agents. Antibiotics 2021, 10, 593. [Google Scholar] [CrossRef] [PubMed]
  16. Tackling Drug-Resistant Infections Globally: Final Report and Recommendations—The Review on Antimicrobial Resistance. Available online: https://amr-review.org/sites/default/files/160525_Final%20paper_with%20cover.pdf (accessed on 23 August 2021).
  17. Fleitas Martínez, O.; Cardoso, M.H.; Ribeiro, S.M.; Franco, O.L. Recent Advances in Anti-Virulence Therapeutic Strategies with a Focus on Dismantling Bacterial Membrane Microdomains, Toxin Neutralization, Quorum-Sensing Interference and Biofilm Inhibition. Front. Cell. Infect. Microbiol. 2019, 9, 74. [Google Scholar] [CrossRef]
  18. Kim, M.-K. Staphylococcus aureus Toxins: From Their Pathogenic Roles to Anti-Virulence Therapy Using Natural Products. Biotechnol. Bioprocess Eng. 2019, 24, 424–435. [Google Scholar] [CrossRef]
  19. Bennett, M.R.; Thomsen, I.P. Epidemiological and Clinical Evidence for the Role of Toxins in Staphylococcus aureus Human Disease. Toxins 2020, 12, 408. [Google Scholar] [CrossRef] [PubMed]
  20. Vlaeminck, J.; Raafat, D.; Surmann, K.; Timbermont, L.; Normann, N.; Sellman, B.; van Wamel, W.J.B.; Malhotra-Kumar, S. Exploring Virulence Factors and Alternative Therapies against Staphylococcus aureus Pneumonia. Toxins 2020, 12, 721. [Google Scholar] [CrossRef]
  21. Ford, C.A.; Hurford, I.M.; Cassat, J.E. Antivirulence Strategies for the Treatment of Staphylococcus aureus Infections: A Mini Review. Front. Microbiol. 2021, 11, 3568. [Google Scholar] [CrossRef]
  22. Divyakolu, S.; Chikkala, R.; Ratnakar, K.S.; Sritharan, V. Hemolysins of Staphylococcus aureus—An Update on Their Biology, Role in Pathogenesis and as Targets for Anti-Virulence Therapy. Adv. Infect. Dis. 2019, 9, 80–104. [Google Scholar] [CrossRef] [Green Version]
  23. Berube, B.J.; Wardenburg, J.B. Staphylococcus aureus α-Toxin: Nearly a Century of Intrigue. Toxins 2013, 5, 1140–1166. [Google Scholar] [CrossRef] [Green Version]
  24. Hernandez, S.L.; Nelson, M.; Sampedro, G.R.; Bagrodia, N.; Defnet, A.M.; Lec, B.; Emolo, J.; Kirschner, R.; Wu, L.; Biermann, H.; et al. Staphylococcus aureus Alpha Toxin Activates Notch in Vascular Cells. Angiogenesis 2019, 22, 197–209. [Google Scholar] [CrossRef]
  25. Bhakdi, S.; Tranum-Jensen, J. Alpha-Toxin of Staphylococcus aureus. Microbiol. Rev. 1991, 55, 733–751. [Google Scholar] [CrossRef]
  26. Pereira-Franchi, E.P.L.; Barreira, M.R.N.; de Costa, N.d.S.L.M.; Riboli, D.F.M.; Abraão, L.M.; Martins, K.B.; Victória, C.; da Cunha, M.d.L.R.d.S. Molecular Epidemiology of Methicillin-Resistant Staphylococcus aureus in the Brazilian Primary Health Care System. Trop. Med. Int. Health 2019, 24, 339–347. [Google Scholar] [CrossRef] [PubMed]
  27. Bubeck Wardenburg, J.; Palazzolo-Ballance, A.M.; Otto, M.; Schneewind, O.; DeLeo, F.R. Panton-Valentine Leukocidin Is Not a Virulence Determinant in Murine Models of Community-Associated Methicillin-Resistant Staphylococcus aureus Disease. J. Infect. Dis. 2008, 198, 1166–1170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Crémieux, A.-C.; Saleh-Mghir, A.; Danel, C.; Couzon, F.; Dumitrescu, O.; Lilin, T.; Perronne, C.; Etienne, J.; Lina, G.; Vandenesch, F. α-Hemolysin, Not Panton-Valentine Leukocidin, Impacts Rabbit Mortality from Severe Sepsis with Methicillin-Resistant Staphylococcus aureus Osteomyelitis. J. Infect. Dis. 2014, 209, 1773–1780. [Google Scholar] [CrossRef] [Green Version]
  29. Montgomery, C.P.; Boyle-Vavra, S.; Adem, P.V.; Lee, J.C.; Husain, A.N.; Clasen, J.; Daum, R.S. Comparison of Virulence in Community-Associated Methicillin-Resistant Staphylococcus aureus Pulsotypes USA300 and USA400 in a Rat Model of Pneumonia. J. Infect. Dis. 2008, 198, 561–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Spaan, A.N.; van Strijp, J.A.G.; Torres, V.J. Leukocidins: Staphylococcal Bi-Component Pore-Forming Toxins Find Their Receptors. Nat. Rev. Microbiol. 2017, 15, 435–447. [Google Scholar] [CrossRef] [PubMed]
  31. Grumann, D.; Nübel, U.; Bröker, B.M. Staphylococcus aureus Toxins–Their Functions and Genetics. Infect. Genet. Evol. 2014, 21, 583–592. [Google Scholar] [CrossRef] [Green Version]
  32. Alonzo, F.; Kozhaya, L.; Rawlings, S.A.; Reyes-Robles, T.; DuMont, A.L.; Myszka, D.G.; Landau, N.R.; Unutmaz, D.; Torres, V.J. CCR5 Is a Receptor for Staphylococcus aureus Leukotoxin ED. Nature 2013, 493, 51–55. [Google Scholar] [CrossRef] [Green Version]
  33. Spaan, A.N.; Henry, T.; van Rooijen, W.J.M.; Perret, M.; Badiou, C.; Aerts, P.C.; Kemmink, J.; de Haas, C.J.C.; van Kessel, K.P.M.; Vandenesch, F.; et al. The Staphylococcal Toxin Panton-Valentine Leukocidin Targets Human C5a Receptors. Cell Host Microbe 2013, 13, 584–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. DuMont, A.L.; Yoong, P.; Day, C.J.; Alonzo, F.; McDonald, W.H.; Jennings, M.P.; Torres, V.J. Staphylococcus aureus LukAB Cytotoxin Kills Human Neutrophils by Targeting the CD11b Subunit of the Integrin Mac-1. Proc. Natl. Acad. Sci. USA 2013, 110, 10794–10799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kaneko, J.; Kamio, Y. Bacterial Two-Component and Hetero-Heptameric Pore-Forming Cytolytic Toxins: Structures, Pore-Forming Mechanism, and Organization of the Genes. Biosci. Biotechnol. Biochem. 2004, 68, 981–1003. [Google Scholar] [CrossRef] [Green Version]
  36. Shallcross, L.J.; Fragaszy, E.; Johnson, A.M.; Hayward, A.C. The Role of the Panton-Valentine Leucocidin Toxin in Staphylococcal Disease: A Systematic Review and Meta-Analysis. Lancet Infect. Dis. 2013, 13, 43–54. [Google Scholar] [CrossRef] [Green Version]
  37. Otter, J.A.; French, G.L. Molecular Epidemiology of Community-Associated Meticillin-Resistant Staphylococcus aureus in Europe. Lancet Infect. Dis. 2010, 10, 227–239. [Google Scholar] [CrossRef]
  38. Mesrati, I.; Saïdani, M.; Ennigrou, S.; Zouari, B.; Ben Redjeb, S. Clinical Isolates of Pantone-Valentine Leucocidin- and Gamma-Haemolysin-Producing Staphylococcus aureus: Prevalence and Association with Clinical Infections. J. Hosp. Infect. 2010, 75, 265–268. [Google Scholar] [CrossRef] [PubMed]
  39. Otto, M. Phenol-Soluble Modulins. Int. J. Med. Microbiol. 2014, 304, 164–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Peschel, A.; Otto, M. Phenol-Soluble Modulins and Staphylococcal Infection. Nat. Rev. Microbiol. 2013, 11, 667–673. [Google Scholar] [CrossRef] [PubMed]
  41. Cheung, G.Y.C.; Joo, H.-S.; Chatterjee, S.S.; Otto, M. Phenol-Soluble Modulins–Critical Determinants of Staphylococcal Virulence. FEMS Microbiol. Rev. 2014, 38, 698–719. [Google Scholar] [CrossRef] [PubMed]
  42. Baldry, M.; Bojer, M.S.; Najarzadeh, Z.; Vestergaard, M.; Meyer, R.L.; Otzen, D.E.; Ingmer, H. Phenol-Soluble Modulins Modulate Persister Cell Formation in Staphylococcus aureus. Front. Microbiol. 2020, 11, 573253. [Google Scholar] [CrossRef] [PubMed]
  43. Zaman, M.; Andreasen, M. Cross-Talk between Individual Phenol-Soluble Modulins in Staphylococcus aureus Biofilm Enables Rapid and Efficient Amyloid Formation. eLife 2020, 9, e59776. [Google Scholar] [CrossRef]
  44. Queck, S.Y.; Jameson-Lee, M.; Villaruz, A.E.; Bach, T.-H.L.; Khan, B.A.; Sturdevant, D.E.; Ricklefs, S.M.; Li, M.; Otto, M. RNAIII-Independent Target Gene Control by the Agr Quorum-Sensing System: Insight into the Evolution of Virulence Regulation in Staphylococcus aureus. Mol. Cell 2008, 32, 150–158. [Google Scholar] [CrossRef] [Green Version]
  45. Zapf, R.L.; Wiemels, R.E.; Keogh, R.A.; Holzschu, D.L.; Howell, K.M.; Trzeciak, E.; Caillet, A.R.; King, K.A.; Selhorst, S.A.; Naldrett, M.J.; et al. The Small RNA Teg41 Regulates Expression of the Alpha Phenol-Soluble Modulins and Is Required for Virulence in Staphylococcus aureus. MBio 2019, 10, e02484-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kretschmer, D.; Gleske, A.-K.; Rautenberg, M.; Wang, R.; Köberle, M.; Bohn, E.; Schöneberg, T.; Rabiet, M.-J.; Boulay, F.; Klebanoff, S.J.; et al. Human Formyl Peptide Receptor 2 Senses Highly Pathogenic Staphylococcus aureus. Cell Host Microbe 2010, 7, 463–473. [Google Scholar] [CrossRef] [Green Version]
  47. Schreiner, J.; Kretschmer, D.; Klenk, J.; Otto, M.; Bühring, H.-J.; Stevanovic, S.; Wang, J.M.; Beer-Hammer, S.; Peschel, A.; Autenrieth, S.E. Staphylococcus aureus Phenol-Soluble Modulin Peptides Modulate Dendritic Cell Functions and Increase In Vitro Priming of Regulatory T Cells. J. Immunol. 2013, 190, 3417–3426. [Google Scholar] [CrossRef] [Green Version]
  48. Wang, R.; Braughton, K.R.; Kretschmer, D.; Bach, T.-H.L.; Queck, S.Y.; Li, M.; Kennedy, A.D.; Dorward, D.W.; Klebanoff, S.J.; Peschel, A.; et al. Identification of Novel Cytolytic Peptides as Key Virulence Determinants for Community-Associated MRSA. Nat. Med. 2007, 13, 1510–1514. [Google Scholar] [CrossRef] [PubMed]
  49. Gonzalez, D.J.; Vuong, L.; Gonzalez, I.S.; Keller, N.; McGrosso, D.; Hwang, J.H.; Hung, J.; Zinkernagel, A.; Dixon, J.E.; Dorrestein, P.C.; et al. Phenol Soluble Modulin (PSM) Variants of Community-Associated Methicillin-Resistant Staphylococcus aureus (MRSA) Captured Using Mass Spectrometry-Based Molecular Networking. Mol. Cell. Proteom. 2014, 13, 1262–1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Davido, B.; Saleh-Mghir, A.; Laurent, F.; Danel, C.; Couzon, F.; Gatin, L.; Vandenesch, F.; Rasigade, J.-P.; Crémieux, A.-C. Phenol-Soluble Modulins Contribute to Early Sepsis Dissemination Not Late Local Usa300-Osteomyelitis Severity in Rabbits. PLoS ONE 2016, 11, e0157133. [Google Scholar] [CrossRef] [Green Version]
  51. Richardson, J.R.; Armbruster, N.S.; Günter, M.; Biljecki, M.; Klenk, J.; Heumos, S.; Autenrieth, S.E. PSM Peptides from Community-Associated Methicillin-Resistant Staphylococcus aureus Impair the Adaptive Immune Response via Modulation of Dendritic Cell Subsets in Vivo. Front. Immunol. 2019, 10, 995. [Google Scholar] [CrossRef]
  52. Czech, A.; Yamaguchi, T.; Bader, L.; Linder, S.; Kaminski, K.; Sugai, M.; Aepfelbacher, M. Prevalence of Rho-Inactivating Epidermal Cell Differentiation Inhibitor Toxins in Clinical Staphylococcus aureus Isolates. J. Infect. Dis. 2001, 184, 785–788. [Google Scholar] [CrossRef] [Green Version]
  53. Messad, N.; Landraud, L.; Canivet, B.; Lina, G.; Richard, J.-L.; Sotto, A.; Lavigne, J.-P.; Lemichez, E. Distribution of Edin in Staphylococcus aureus Isolated from Diabetic Foot Ulcers. Clin. Microbiol. Infect. 2013, 19, 875–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Boyer, L.; Doye, A.; Rolando, M.; Flatau, G.; Munro, P.; Gounon, P.; Clément, R.; Pulcini, C.; Popoff, M.R.; Mettouchi, A.; et al. Induction of Transient Macroapertures in Endothelial Cells through RhoA Inhibition by Staphylococcus aureus Factors. J. Cell Biol. 2006, 173, 809–819. [Google Scholar] [CrossRef] [PubMed]
  55. Aktories, K.; Lang, A.E.; Schwan, C.; Mannherz, H.G. Actin as Target for Modification by Bacterial Protein Toxins. FEBS J. 2011, 278, 4526–4543. [Google Scholar] [CrossRef]
  56. Jaffe, A.B.; Hall, A. Rho GTPases: Biochemistry and Biology. Annu. Rev. Cell Dev. Biol. 2005, 21, 247–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Nusrat, A.; Giry, M.; Turner, J.R.; Colgan, S.P.; Parkos, C.A.; Carnes, D.; Lemichez, E.; Boquet, P.; Madara, J.L. Rho Protein Regulates Tight Junctions and Perijunctional Actin Organization in Polarized Epithelia. Proc. Natl. Acad. Sci. USA 1995, 92, 10629–10633. [Google Scholar] [CrossRef] [Green Version]
  58. Caron, E.; Hall, A. Identification of Two Distinct Mechanisms of Phagocytosis Controlled by Different Rho GTPases. Science 1998, 282, 1717–1721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Lemichez, E.; Lecuit, M.; Nassif, X.; Bourdoulous, S. Breaking the Wall: Targeting of the Endothelium by Pathogenic Bacteria. Nat. Rev. Microbiol. 2010, 8, 93–104. [Google Scholar] [CrossRef]
  60. Edwards, A.M.; Massey, R.C. How Does Staphylococcus aureus Escape the Bloodstream? Trends Microbiol. 2011, 19, 184–190. [Google Scholar] [CrossRef]
  61. Sugai, M.; Enomoto, T.; Hashimoto, K.; Matsumoto, K.; Matsuo, Y.; Ohgai, H.; Hong, Y.-M.; Inoue, S.; Yoshikawa, K.; Suginaka, H. A Novel Epidermal Cell Differentiation Inhibitor (EDIN): Purification and Characterization from Staphylococcus aureus. Biochem. Biophys. Res. Commun. 1990, 173, 92–98. [Google Scholar] [CrossRef]
  62. Yamaguchi, T.; Hayashi, T.; Takami, H.; Ohnishi, M.; Murata, T.; Nakayama, K.; Asakawa, K.; Ohara, M.; Komatsuzawa, H.; Sugai, M. Complete Nucleotide Sequence of a Staphylococcus aureus Exfoliative Toxin B Plasmid and Identification of a Novel ADP-Ribosyltransferase, EDIN-C. Infect. Immun. 2001, 69, 7760–7771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yamaguchi, T.; Nishifuji, K.; Sasaki, M.; Fudaba, Y.; Aepfelbacher, M.; Takata, T.; Ohara, M.; Komatsuzawa, H.; Amagai, M.; Sugai, M. Identification of the Staphylococcus aureus Etd Pathogenicity Island Which Encodes a Novel Exfoliative Toxin, ETD, and EDIN-B. Infect. Immun. 2002, 70, 5835–5845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Munro, P.; Benchetrit, M.; Nahori, M.-A.; Stefani, C.; Clément, R.; Michiels, J.-F.; Landraud, L.; Dussurget, O.; Lemichez, E. The Staphylococcus aureus Epidermal Cell Differentiation Inhibitor Toxin Promotes Formation of Infection Foci in a Mouse Model of Bacteremia. Infect. Immun. 2010, 78, 3404–3411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Antri, K.; Rouzic, N.; Boubekri, I.; Dauwalder, O.; Beloufa, A.; Ziane, H.; Djennane, F.; Neggazi, M.; Benhabyles, B.; Bes, M.; et al. High prevalence of community and hospital acquired infections of methicillin-resistant Staphylococcus aureus containing Panton-Valentine leukocidin gene in Algiers. Pathol. Biol. 2010, 58, e15–e20. [Google Scholar] [CrossRef] [PubMed]
  66. Courjon, J.; Munro, P.; Benito, Y.; Visvikis, O.; Bouchiat, C.; Boyer, L.; Doye, A.; Lepidi, H.; Ghigo, E.; Lavigne, J.-P.; et al. EDIN-B Promotes the Translocation of Staphylococcus aureus to the Bloodstream in the Course of Pneumonia. Toxins 2015, 7, 4131–4142. [Google Scholar] [CrossRef] [Green Version]
  67. Pérez-Montarelo, D.; Viedma, E.; Larrosa, N.; Gómez-González, C.; Ruiz de Gopegui, E.; Muñoz-Gallego, I.; San Juan, R.; Fernández-Hidalgo, N.; Almirante, B.; Chaves, F. Molecular Epidemiology of Staphylococcus aureus Bacteremia: Association of Molecular Factors With the Source of Infection. Front. Microbiol. 2018, 9, 2210. [Google Scholar] [CrossRef] [PubMed]
  68. Bukowski, M.; Wladyka, B.; Dubin, G. Exfoliative Toxins of Staphylococcus aureus. Toxins 2010, 2, 1148–1165. [Google Scholar] [CrossRef] [Green Version]
  69. Stanley, J.R.; Amagai, M. Pemphigus, Bullous Impetigo, and the Staphylococcal Scalded-Skin Syndrome. N. Engl. J. Med. 2006, 355, 1800–1810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Kato, F.; Kadomoto, N.; Iwamoto, Y.; Bunai, K.; Komatsuzawa, H.; Sugai, M. Regulatory Mechanism for Exfoliative Toxin Production in Staphylococcus aureus. Infect. Immun. 2011, 79, 1660–1670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Eyre, R.W.; Stanley, J.R. Human Autoantibodies against a Desmosomal Protein Complex with a Calcium-Sensitive Epitope Are Characteristic of Pemphigus Foliaceus Patients. J. Exp. Med. 1987, 165, 1719–1724. [Google Scholar] [CrossRef] [Green Version]
  72. Hanakawa, Y.; Schechter, N.M.; Lin, C.; Garza, L.; Li, H.; Yamaguchi, T.; Fudaba, Y.; Nishifuji, K.; Sugai, M.; Amagai, M.; et al. Molecular Mechanisms of Blister Formation in Bullous Impetigo and Staphylococcal Scalded Skin Syndrome. J. Clin. Investig. 2002, 110, 53–60. [Google Scholar] [CrossRef] [PubMed]
  73. Nishifuji, K.; Sugai, M.; Amagai, M. Staphylococcal Exfoliative Toxins: “Molecular Scissors” of Bacteria That Attack the Cutaneous Defense Barrier in Mammals. J. Dermatol. Sci. 2008, 49, 21–31. [Google Scholar] [CrossRef] [PubMed]
  74. Sila, J.; Sauer, P.; Kolar, M. Comparison of the Prevalence of Genes Coding for Enterotoxins, Exfoliatins, Panton-Valentine Leukocidin and Tsst-1 between Methicillin-Resistant and Methicillin-Susceptible Isolates of Staphylococcus aureus at the University Hospital in Olomouc. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czechoslov. 2009, 153, 215–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Mégevand, C.; Gervaix, A.; Heininger, U.; Berger, C.; Aebi, C.; Vaudaux, B.; Kind, C.; Gnehm, H.-P.; Hitzler, M.; Renzi, G.; et al. Molecular Epidemiology of the Nasal Colonization by Methicillin-Susceptible Staphylococcus aureus in Swiss Children. Clin. Microbiol. Infect. 2010, 16, 1414–1420. [Google Scholar] [CrossRef]
  76. Yamaguchi, T.; Yokota, Y.; Terajima, J.; Hayashi, T.; Aepfelbacher, M.; Ohara, M.; Komatsuzawa, H.; Watanabe, H.; Sugai, M. Clonal Association of Staphylococcus aureus Causing Bullous Impetigo and the Emergence of New Methicillin-Resistant Clonal Groups in Kansai District in Japan. J. Infect. Dis. 2002, 185, 1511–1516. [Google Scholar] [CrossRef] [Green Version]
  77. Noguchi, N.; Nakaminami, H.; Nishijima, S.; Kurokawa, I.; So, H.; Sasatsu, M. Antimicrobial Agent of Susceptibilities and Antiseptic Resistance Gene Distribution among Methicillin-Resistant Staphylococcus aureus Isolates from Patients with Impetigo and Staphylococcal Scalded Skin Syndrome. J. Clin. Microbiol. 2006, 44, 2119–2125. [Google Scholar] [CrossRef] [Green Version]
  78. Wieczorek, M.; Abualrous, E.T.; Sticht, J.; Álvaro-Benito, M.; Stolzenberg, S.; Noé, F.; Freund, C. Major Histocompatibility Complex (MHC) Class I and MHC Class II Proteins: Conformational Plasticity in Antigen Presentation. Front. Immunol. 2017, 8, 292. [Google Scholar] [CrossRef] [Green Version]
  79. Fraser, J.D.; Proft, T. The Bacterial Superantigen and Superantigen-like Proteins. Immunol. Rev. 2008, 225, 226–243. [Google Scholar] [CrossRef]
  80. 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]
  81. Hennekinne, J.-A.; De Buyser, M.-L.; Dragacci, S. Staphylococcus aureus and Its Food Poisoning Toxins: Characterization and Outbreak Investigation. FEMS Microbiol. Rev. 2012, 36, 815–836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Otto, M. Staphylococcus aureus Toxins. Curr. Opin. Microbiol. 2014, 17, 32–37. [Google Scholar] [CrossRef] [Green Version]
  83. Hu, D.-L.; Li, S.; Fang, R.; Ono, H.K. Update on Molecular Diversity and Multipathogenicity of Staphylococcal Superantigen Toxins. Anim. Dis. 2021, 1, 7. [Google Scholar] [CrossRef]
  84. Kong, C.; Neoh, H.; Nathan, S. Targeting Staphylococcus aureus Toxins: A Potential Form of Anti-Virulence Therapy. Toxins 2016, 8, 72. [Google Scholar] [CrossRef] [Green Version]
  85. Fisher, E.L.; Otto, M.; Cheung, G.Y.C. Basis of Virulence in Enterotoxin-Mediated Staphylococcal Food Poisoning. Front. Microbiol. 2018, 9, 436. [Google Scholar] [CrossRef] [PubMed]
  86. Uchiyama, T.; Araake, M.; Yan, X.J.; Miyanaga, Y.; Igarashi, H. Involvement of HLA Class II Molecules in Acquisition of Staphylococcal Enterotoxin A-Binding Activity and Accessory Cell Activity in Activation of Human T Cells by Related Toxins in Vascular Endothelial Cells. Clin. Exp. Immunol. 1992, 87, 322–328. [Google Scholar] [CrossRef]
  87. Pezato, R.; Świerczyńska-Krępa, M.; Niżankowska-Mogilnicka, E.; Derycke, L.; Bachert, C.; Pérez-Novo, C.A. Role of Imbalance of Eicosanoid Pathways and Staphylococcal Superantigens in Chronic Rhinosinusitis. Allergy 2012, 67, 1347–1356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Pinchuk, I.V.; Beswick, E.J.; Reyes, V.E. Staphylococcal Enterotoxins. Toxins 2010, 2, 2177–2197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Salgado-Pabón, W.; Breshears, L.; Spaulding, A.R.; Merriman, J.A.; Stach, C.S.; Horswill, A.R.; Peterson, M.L.; Schlievert, P.M. Superantigens Are Critical for Staphylococcus aureus Infective Endocarditis, Sepsis, and Acute Kidney Injury. MBio 2013, 4, e00494-13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Stach, C.S.; Herrera, A.; Schlievert, P.M. Staphylococcal Superantigens Interact with Multiple Host Receptors to Cause Serious Diseases. Immunol. Res. 2014, 59, 177–181. [Google Scholar] [CrossRef] [PubMed]
  91. McCormick, J.K.; Yarwood, J.M.; Schlievert, P.M. Toxic Shock Syndrome and Bacterial Superantigens: An Update. Annu. Rev. Microbiol. 2001, 55, 77–104. [Google Scholar] [CrossRef]
  92. Adhikari, R.P.; Ajao, A.O.; Aman, M.J.; Karauzum, H.; Sarwar, J.; Lydecker, A.D.; Johnson, J.K.; Nguyen, C.; Chen, W.H.; Roghmann, M.-C. Lower Antibody Levels to Staphylococcus aureus Exotoxins Are Associated With Sepsis in Hospitalized Adults With Invasive S. aureus Infections. J. Infect. Dis. 2012, 206, 915–923. [Google Scholar] [CrossRef] [PubMed]
  93. Sharma-Kuinkel, B.K.; Tkaczyk, C.; Bonnell, J.; Yu, L.; Tovchigrechko, A.; Tabor, D.E.; Park, L.P.; Ruffin, F.; Esser, M.T.; Sellman, B.R.; et al. Associations of Pathogen-Specific and Host-Specific Characteristics with Disease Outcome in Patients with Staphylococcus aureus Bacteremic Pneumonia. Clin. Transl. Immunol. 2019, 8, e01070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Gillet, Y.; Issartel, B.; Vanhems, P.; Fournet, J.-C.; Lina, G.; Bes, M.; Vandenesch, F.; Piémont, Y.; Brousse, N.; Floret, D.; et al. Association between Staphylococcus aureus Strains Carrying Gene for Panton-Valentine Leukocidin and Highly Lethal Necrotising Pneumonia in Young Immunocompetent Patients. Lancet 2002, 359, 753–759. [Google Scholar] [CrossRef]
  95. Verdon, J.; Girardin, N.; Lacombe, C.; Berjeaud, J.-M.; Héchard, Y. Delta-Hemolysin, an Update on a Membrane-Interacting Peptide. Peptides 2009, 30, 817–823. [Google Scholar] [CrossRef]
  96. Surewaard, B.G.J.; de Haas, C.J.C.; Vervoort, F.; Rigby, K.M.; DeLeo, F.R.; Otto, M.; van Strijp, J.A.G.; Nijland, R. Staphylococcal Alpha-Phenol Soluble Modulins Contribute to Neutrophil Lysis after Phagocytosis. Cell. Microbiol. 2013, 15, 1427–1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Mishra, A.K.; Yadav, P.; Mishra, A. A Systemic Review on Staphylococcal Scalded Skin Syndrome (SSSS): A Rare and Critical Disease of Neonates. Open Microbiol. J. 2016, 10, 150. [Google Scholar] [CrossRef] [Green Version]
  98. Wilke, G.A.; Bubeck Wardenburg, J. Role of a Disintegrin and Metalloprotease 10 in Staphylococcus aureus Alpha-Hemolysin-Mediated Cellular Injury. Proc. Natl. Acad. Sci. USA 2010, 107, 13473–13478. [Google Scholar] [CrossRef] [Green Version]
  99. Foletti, D.; Strop, P.; Shaughnessy, L.; Hasa-Moreno, A.; Casas, M.G.; Russell, M.; Bee, C.; Wu, S.; Pham, A.; Zeng, Z.; et al. Mechanism of Action and in Vivo Efficacy of a Human-Derived Antibody against Staphylococcus aureus α-Hemolysin. J. Mol. Biol. 2013, 425, 1641–1654. [Google Scholar] [CrossRef]
  100. Oganesyan, V.; Peng, L.; Damschroder, M.M.; Cheng, L.; Sadowska, A.; Tkaczyk, C.; Sellman, B.R.; Wu, H.; Dall’Acqua, W.F. Mechanisms of Neutralization of a Human Anti-α-Toxin Antibody. J. Biol. Chem. 2014, 289, 29874–29880. [Google Scholar] [CrossRef] [Green Version]
  101. Le, V.T.M.; Tkaczyk, C.; Chau, S.; Rao, R.L.; Dip, E.C.; Pereira-Franchi, E.P.; Cheng, L.; Lee, S.; Koelkebeck, H.; Hilliard, J.J.; et al. Critical Role of Alpha-Toxin and Protective Effects of Its Neutralization by a Human Antibody in Acute Bacterial Skin and Skin Structure Infections. Antimicrob. Agents Chemother. 2016, 60, 5640–5648. [Google Scholar] [CrossRef] [Green Version]
  102. Ortines, R.V.; Liu, H.; Cheng, L.I.; Cohen, T.S.; Lawlor, H.; Gami, A.; Wang, Y.; Dillen, C.A.; Archer, N.K.; Miller, R.J.; et al. Neutralizing Alpha-Toxin Accelerates Healing of Staphylococcus aureus-Infected Wounds in Nondiabetic and Diabetic Mice. Antimicrob. Agents Chemother. 2018, 62, e02288-17. [Google Scholar] [CrossRef] [Green Version]
  103. Surewaard, B.G.J.; Thanabalasuriar, A.; Zeng, Z.; Tkaczyk, C.; Cohen, T.S.; Bardoel, B.W.; Jorch, S.K.; Deppermann, C.; Bubeck Wardenburg, J.; Davis, R.P.; et al. α-Toxin Induces Platelet Aggregation and Liver Injury during Staphylococcus aureus Sepsis. Cell Host Microbe 2018, 24, 271–284.e3. [Google Scholar] [CrossRef] [PubMed]
  104. Yu, X.-Q.; Robbie, G.J.; Wu, Y.; Esser, M.T.; Jensen, K.; Schwartz, H.I.; Bellamy, T.; Hernandez-Illas, M.; Jafri, H.S. Safety, Tolerability, and Pharmacokinetics of MEDI4893, an Investigational, Extended-Half-Life, Anti-Staphylococcus aureus Alpha-Toxin Human Monoclonal Antibody, in Healthy Adults. Antimicrob. Agents Chemother. 2017, 61, e01020-16. [Google Scholar] [CrossRef] [Green Version]
  105. François, B.; Jafri, H.S.; Chastre, J.; Sánchez-García, M.; Eggimann, P.; Dequin, P.-F.; Huberlant, V.; Viña Soria, L.; Boulain, T.; Bretonnière, C.; et al. Efficacy and Safety of Suvratoxumab for Prevention of Staphylococcus aureus Ventilator-Associated Pneumonia (SAATELLITE): A Multicentre, Randomised, Double-Blind, Placebo-Controlled, Parallel-Group, Phase 2 Pilot Trial. Lancet Infect. Dis. 2021, 21, 1313–1323. [Google Scholar] [CrossRef]
  106. François, B.; Mercier, E.; Gonzalez, C.; Asehnoune, K.; Nseir, S.; Fiancette, M.; Desachy, A.; Plantefève, G.; Meziani, F.; de Lame, P.-A.; et al. Safety and Tolerability of a Single Administration of AR-301, a Human Monoclonal Antibody, in ICU Patients with Severe Pneumonia Caused by Staphylococcus aureus: First-in-Human Trial. Intensive Care Med. 2018, 44, 1787–1796. [Google Scholar] [CrossRef]
  107. Magyarics, Z.; Leslie, F.; Bartko, J.; Rouha, H.; Luperchio, S.; Schörgenhofer, C.; Schwameis, M.; Derhaschnig, U.; Lagler, H.; Stiebellehner, L.; et al. Randomized, Double-Blind, Placebo-Controlled, Single-Ascending-Dose Study of the Penetration of a Monoclonal Antibody Combination (ASN100) Targeting Staphylococcus aureus Cytotoxins in the Lung Epithelial Lining Fluid of Healthy Volunteers. Antimicrob. Agents Chemother. 2019, 63, e00350-19. [Google Scholar] [CrossRef] [Green Version]
  108. Ruzin, A.; Yu, L.; Barraud, O.; François, B.; Sánchez Garcia, M.; Eggimann, P.; Dequin, P.-F.; Laterre, P.-F.; Huberlant, V.; Viña, L.; et al. 2160. Performance of the Cepheid Rapid PCR Test for Patient Screening and Association with Efficacy of Suvratoxumab, A Novel Anti-Staphylococcus aureus Monoclonal Antibody, During the Phase 2 SAATELLITE Study. Open Forum Infect. Dis. 2019, 6, S733. [Google Scholar] [CrossRef]
  109. Vandenesch, F.; Naimi, T.; Enright, M.C.; Lina, G.; Nimmo, G.R.; Heffernan, H.; Liassine, N.; Bes, M.; Greenland, T.; Reverdy, M.-E.; et al. Community-Acquired Methicillin-Resistant Staphylococcus aureus Carrying Panton-Valentine Leukocidin Genes: Worldwide Emergence. Emerg. Infect. Dis. 2003, 9, 978–984. [Google Scholar] [CrossRef] [PubMed]
  110. Diep, B.A.; Otto, M. The Role of Virulence Determinants in Community-Associated MRSA Pathogenesis. Trends Microbiol. 2008, 16, 361–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Hermos, C.R.; Yoong, P.; Pier, G.B. High Levels of Antibody to Panton-Valentine Leukocidin Are Not Associated with Resistance to Staphylococcus aureus-Associated Skin and Soft-Tissue Infection. Clin. Infect. Dis. 2010, 51, 1138–1146. [Google Scholar] [CrossRef] [Green Version]
  112. Gauduchon, V.; Cozon, G.; Vandenesch, F.; Genestier, A.-L.; Eyssade, N.; Peyrol, S.; Etienne, J.; Lina, G. Neutralization of Staphylococcus aureus Panton Valentine Leukocidin by Intravenous Immunoglobulin In Vitro. J. Infect. Dis. 2004, 189, 346–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Mairpady Shambat, S.; Chen, P.; Nguyen Hoang, A.T.; Bergsten, H.; Vandenesch, F.; Siemens, N.; Lina, G.; Monk, I.R.; Foster, T.J.; Arakere, G.; et al. Modelling Staphylococcal Pneumonia in a Human 3D Lung Tissue Model System Delineates Toxin-Mediated Pathology. Dis. Model. Mech. 2015, 8, 1413–1425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Rouzic, N.; Janvier, F.; Libert, N.; Javouhey, E.; Lina, G.; Nizou, J.-Y.; Pasquier, P.; Stamm, D.; Brinquin, L.; Pelletier, C.; et al. Prompt and Successful Toxin-Targeting Treatment of Three Patients with Necrotizing Pneumonia Due to Staphylococcus aureus Strains Carrying the Panton-Valentine Leukocidin Genes. J. Clin. Microbiol. 2010, 48, 1952–1955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Laventie, B.-J.; Rademaker, H.J.; Saleh, M.; de Boer, E.; Janssens, R.; Bourcier, T.; Subilia, A.; Marcellin, L.; van Haperen, R.; Lebbink, J.H.G.; et al. Heavy Chain-Only Antibodies and Tetravalent Bispecific Antibody Neutralizing Staphylococcus aureus Leukotoxins. Proc. Natl. Acad. Sci. USA 2011, 108, 16404–16409. [Google Scholar] [CrossRef] [Green Version]
  116. Aman, M.J. Superantigens of a Superbug: Major Culprits of Staphylococcus aureus Disease? Virulence 2017, 8, 607–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Drozdowski, B.; Zhou, Y.; Kline, B.; Spidel, J.; Chan, Y.Y.; Albone, E.; Turchin, H.; Chao, Q.; Henry, M.; Balogach, J.; et al. Generation and Characterization of High Affinity Human Monoclonal Antibodies That Neutralize Staphylococcal Enterotoxin B. J. Immune Based Ther. Vaccines 2010, 8, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Varshney, A.K.; Wang, X.; Scharff, M.D.; MacIntyre, J.; Zollner, R.S.; Kovalenko, O.V.; Martinez, L.R.; Byrne, F.R.; Fries, B.C. Staphylococcal Enterotoxin B-Specific Monoclonal Antibody 20B1 Successfully Treats Diverse Staphylococcus aureus Infections. J. Infect. Dis. 2013, 208, 2058–2066. [Google Scholar] [CrossRef] [Green Version]
  119. Fang, R.H.; Luk, B.T.; Hu, C.-M.J.; Zhang, L. Engineered Nanoparticles Mimicking Cell Membranes for Toxin Neutralization. Adv. Drug Deliv. Rev. 2015, 90, 69–80. [Google Scholar] [CrossRef] [Green Version]
  120. Henry, B.D.; Neill, D.R.; Becker, K.A.; Gore, S.; Bricio-Moreno, L.; Ziobro, R.; Edwards, M.J.; Mühlemann, K.; Steinmann, J.; Kleuser, B.; et al. Engineered Liposomes Sequester Bacterial Exotoxins and Protect from Severe Invasive Infections in Mice. Nat. Biotechnol. 2015, 33, 81–88. [Google Scholar] [CrossRef] [Green Version]
  121. Wolfmeier, H.; Mansour, S.C.; Liu, L.T.; Pletzer, D.; Draeger, A.; Babiychuk, E.B.; Hancock, R.E.W. Liposomal Therapy Attenuates Dermonecrosis Induced by Community-Associated Methicillin-Resistant Staphylococcus aureus by Targeting α-Type Phenol-Soluble Modulins and α-Hemolysin. EBioMedicine 2018, 33, 211–217. [Google Scholar] [CrossRef]
  122. Keller, M.D.; Ching, K.L.; Liang, F.-X.; Dhabaria, A.; Tam, K.; Ueberheide, B.M.; Unutmaz, D.; Torres, V.J.; Cadwell, K. Decoy Exosomes Provide Protection against Bacterial Toxins. Nature 2020, 579, 260–264. [Google Scholar] [CrossRef]
  123. Chen, Y.; Chen, M.; Zhang, Y.; Lee, J.H.; Escajadillo, T.; Gong, H.; Fang, R.H.; Gao, W.; Nizet, V.; Zhang, L. Broad-Spectrum Neutralization of Pore-Forming Toxins with Human Erythrocyte Membrane-Coated Nanosponges. Adv. Healthc. Mater. 2018, 7, e1701366. [Google Scholar] [CrossRef] [PubMed]
  124. Wei, X.; Gao, J.; Wang, F.; Ying, M.; Angsantikul, P.; Kroll, A.V.; Zhou, J.; Gao, W.; Lu, W.; Fang, R.H.; et al. In Situ Capture of Bacterial Toxins for Antivirulence Vaccination. Adv. Mater. 2017, 29, 1701644. [Google Scholar] [CrossRef] [PubMed]
  125. Yarwood, J.M.; Bartels, D.J.; Volper, E.M.; Greenberg, E.P. Quorum Sensing in Staphylococcus aureus Biofilms. J. Bacteriol. 2004, 186, 1838–1850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Cheung, G.Y.C.; Wang, R.; Khan, B.A.; Sturdevant, D.E.; Otto, M. Role of the Accessory Gene Regulator Agr in Community-Associated Methicillin-Resistant Staphylococcus aureus Pathogenesis. Infect. Immun. 2011, 79, 1927–1935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Novick, R.P.; Ross, H.F.; Projan, S.J.; Kornblum, J.; Kreiswirth, B.; Moghazeh, S. Synthesis of Staphylococcal Virulence Factors Is Controlled by a Regulatory RNA Molecule. EMBO J. 1993, 12, 3967–3975. [Google Scholar] [CrossRef] [PubMed]
  128. Balaban, N.; Collins, L.V.; Cullor, J.S.; Hume, E.B.; Medina-Acosta, E.; Vieira da Motta, O.; O’Callaghan, R.; Rossitto, P.V.; Shirtliff, M.E.; Serafim da Silveira, L.; et al. Prevention of Diseases Caused by Staphylococcus aureus Using the Peptide RIP. Peptides 2000, 21, 1301–1311. [Google Scholar] [CrossRef]
  129. Gov, Y.; Bitler, A.; Dell’Acqua, G.; Torres, J.V.; Balaban, N. RNAIII Inhibiting Peptide (RIP), a Global Inhibitor of Staphylococcus aureus Pathogenesis: Structure and Function Analysis. Peptides 2001, 22, 1609–1620. [Google Scholar] [CrossRef]
  130. Vieira-da-Motta, O.; Damasceno Ribeiro, P.; Dias da Silva, W.; Medina-Acosta, E. RNAIII Inhibiting Peptide (RIP) Inhibits Agr-Regulated Toxin Production. Peptides 2001, 22, 1621–1627. [Google Scholar] [CrossRef]
  131. Ma, B.; Zhou, Y.; Li, M.; Yu, Q.; Xue, X.; Li, Z.; Da, F.; Hou, Z.; Luo, X. RIP-V Improves Murine Survival in a Sepsis Model by down-Regulating RNAIII Expression and α-Hemolysin Release of Methicillin-Resistant Staphylococcus aureus. Pharmazie 2015, 70, 81–87. [Google Scholar]
  132. Giacometti, A.; Cirioni, O.; Ghiselli, R.; Dell’Acqua, G.; Orlando, F.; D’Amato, G.; Mocchegiani, F.; Silvestri, C.; Del Prete, M.S.; Rocchi, M.; et al. RNAIII-Inhibiting Peptide Improves Efficacy of Clinically Used Antibiotics in a Murine Model of Staphylococcal Sepsis. Peptides 2005, 26, 169–175. [Google Scholar] [CrossRef]
  133. Simonetti, O.; Cirioni, O.; Ghiselli, R.; Goteri, G.; Scalise, A.; Orlando, F.; Silvestri, C.; Riva, A.; Saba, V.; Madanahally, K.D.; et al. RNAIII-Inhibiting Peptide Enhances Healing of Wounds Infected with Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2008, 52, 2205–2211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zhou, Y.; Niu, C.; Ma, B.; Xue, X.; Li, Z.; Chen, Z.; Li, F.; Zhou, S.; Luo, X.; Hou, Z. Inhibiting PSMα-Induced Neutrophil Necroptosis Protects Mice with MRSA Pneumonia by Blocking the Agr System. Cell Death Dis. 2018, 9, 362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Huan, Y.; Kong, Q.; Mou, H.; Yi, H. Antimicrobial Peptides: Classification, Design, Application and Research Progress in Multiple Fields. Front. Microbiol. 2020, 11, 2559. [Google Scholar] [CrossRef] [PubMed]
  136. Kudryashova, E.; Seveau, S.M.; Kudryashov, D.S. Targeting and Inactivation of Bacterial Toxins by Human Defensins. Biol. Chem. 2017, 398, 1069–1085. [Google Scholar] [CrossRef] [PubMed]
  137. Patrulea, V.; Borchard, G.; Jordan, O. An Update on Antimicrobial Peptides (AMPs) and Their Delivery Strategies for Wound Infections. Pharmaceutics 2020, 12, 840. [Google Scholar] [CrossRef]
  138. Lehrer, R.I.; Lu, W. α-Defensins in Human Innate Immunity. Immunol. Rev. 2012, 245, 84–112. [Google Scholar] [CrossRef] [PubMed]
  139. Lillard, J.W.; Boyaka, P.N.; Chertov, O.; Oppenheim, J.J.; McGhee, J.R. Mechanisms for Induction of Acquired Host Immunity by Neutrophil Peptide Defensins. Proc. Natl. Acad. Sci. USA 1999, 96, 651–656. [Google Scholar] [CrossRef] [Green Version]
  140. Diep, B.A.; Chan, L.; Tattevin, P.; Kajikawa, O.; Martin, T.R.; Basuino, L.; Mai, T.T.; Marbach, H.; Braughton, K.R.; Whitney, A.R.; et al. Polymorphonuclear Leukocytes Mediate Staphylococcus aureus Panton-Valentine Leukocidin-Induced Lung Inflammation and Injury. Proc. Natl. Acad. Sci. USA 2010, 107, 5587–5592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Cardot-Martin, E.; Casalegno, J.S.; Badiou, C.; Dauwalder, O.; Keller, D.; Prévost, G.; Rieg, S.; Kern, W.V.; Cuerq, C.; Etienne, J.; et al. α-Defensins Partially Protect Human Neutrophils against Panton-Valentine Leukocidin Produced by Staphylococcus aureus. Lett. Appl. Microbiol. 2015, 61, 158–164. [Google Scholar] [CrossRef]
  142. Karginov, V.A.; Nestorovich, E.M.; Schmidtmann, F.; Robinson, T.M.; Yohannes, A.; Fahmi, N.E.; Bezrukov, S.M.; Hecht, S.M. Inhibition of S. aureus α-Hemolysin and B. anthracis Lethal Toxin by β-Cyclodextrin Derivatives. Bioorg. Med. Chem. 2007, 15, 5424–5431. [Google Scholar] [CrossRef] [Green Version]
  143. Dong, J.; Qiu, J.; Wang, J.; Li, H.; Dai, X.; Zhang, Y.; Wang, X.; Tan, W.; Niu, X.; Deng, X.; et al. Apigenin Alleviates the Symptoms of Staphylococcus aureus Pneumonia by Inhibiting the Production of Alpha-Hemolysin. FEMS Microbiol. Lett. 2013, 338, 124–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Jiang, L.; Yi, T.; Shen, Z.; Teng, Z.; Wang, J. Aloe-Emodin Attenuates Staphylococcus aureus Pathogenicity by Interfering with the Oligomerization of α-Toxin. Front. Cell. Infect. Microbiol. 2019, 9, 157. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, J.; Zhou, X.; Liu, S.; Li, G.; Shi, L.; Dong, J.; Li, W.; Deng, X.; Niu, X. Morin Hydrate Attenuates Staphylococcus aureus Virulence by Inhibiting the Self-Assembly of α-Hemolysin. J. Appl. Microbiol. 2015, 118, 753–763. [Google Scholar] [CrossRef]
  146. Dong, J.; Qiu, J.; Zhang, Y.; Lu, C.; Dai, X.; Wang, J.; Li, H.; Wang, X.; Tan, W.; Luo, M.; et al. Oroxylin A Inhibits Hemolysis via Hindering the Self-Assembly of α-Hemolysin Heptameric Transmembrane Pore. PLoS Comput. Biol. 2013, 9, e1002869. [Google Scholar] [CrossRef]
  147. Qiu, J.; Wang, D.; Zhang, Y.; Dong, J.; Wang, J.; Niu, X. Molecular Modeling Reveals the Novel Inhibition Mechanism and Binding Mode of Three Natural Compounds to Staphylococcal α-Hemolysin. PLoS ONE 2013, 8, e80197. [Google Scholar] [CrossRef]
  148. Friedman, M.; Rasooly, R.; Do, P.M.; Henika, P.R. The Olive Compound 4-Hydroxytyrosol Inactivates Staphylococcus aureus Bacteria and Staphylococcal Enterotoxin A (SEA). J. Food Sci. 2011, 76, M558–M563. [Google Scholar] [CrossRef] [PubMed]
  149. Mansson, M.; Nielsen, A.; Kjærulff, L.; Gotfredsen, C.H.; Wietz, M.; Ingmer, H.; Gram, L.; Larsen, T.O. Inhibition of Virulence Gene Expression in Staphylococcus aureus by Novel Depsipeptides from a Marine Photobacterium. Mar. Drugs 2011, 9, 2537–2552. [Google Scholar] [CrossRef] [Green Version]
  150. Nielsen, A.; Månsson, M.; Bojer, M.S.; Gram, L.; Larsen, T.O.; Novick, R.P.; Frees, D.; Frøkiær, H.; Ingmer, H. Solonamide B Inhibits Quorum Sensing and Reduces Staphylococcus aureus Mediated Killing of Human Neutrophils. PLoS ONE 2014, 9, e84992. [Google Scholar] [CrossRef] [Green Version]
  151. Baldry, M.; Kitir, B.; Frøkiær, H.; Christensen, S.B.; Taverne, N.; Meijerink, M.; Franzyk, H.; Olsen, C.A.; Wells, J.M.; Ingmer, H. The Agr Inhibitors Solonamide B and Analogues Alter Immune Responses to Staphylococcus aureus but Do Not Exhibit Adverse Effects on Immune Cell Functions. PLoS ONE 2016, 11, e0145618. [Google Scholar] [CrossRef] [PubMed]
  152. Baldry, M.; Nakamura, Y.; Nakagawa, S.; Frees, D.; Matsue, H.; Núñez, G.; Ingmer, H. Application of an Agr-Specific Antivirulence Compound as Therapy for Staphylococcus aureus-Induced Inflammatory Skin Disease. J. Infect. Dis. 2018, 218, 1009–1013. [Google Scholar] [CrossRef]
  153. Wang, J.; Qiu, J.; Dong, J.; Li, H.; Luo, M.; Dai, X.; Zhang, Y.; Leng, B.; Niu, X.; Zhao, S.; et al. Chrysin Protects Mice from Staphylococcus aureus Pneumonia. J. Appl. Microbiol. 2011, 111, 1551–1558. [Google Scholar] [CrossRef]
  154. Tang, F.; Li, W.-H.; Zhou, X.; Liu, Y.-H.; Li, Z.; Tang, Y.-S.; Kou, X.; Wang, S.-D.; Bao, M.; Qu, L.-D.; et al. Puerarin Protects against Staphylococcus aureus-Induced Injury of Human Alveolar Epithelial A549 Cells via Downregulating Alpha-Hemolysin Secretion. Microb. Drug Resist. 2014, 20, 357–363. [Google Scholar] [CrossRef] [PubMed]
  155. Jiang, L.; Li, H.; Wang, L.; Song, Z.; Shi, L.; Li, W.; Deng, X.; Wang, J. Isorhamnetin Attenuates Staphylococcus aureus-Induced Lung Cell Injury by Inhibiting Alpha-Hemolysin Expression. J. Microbiol. Biotechnol. 2016, 26, 596–602. [Google Scholar] [CrossRef] [Green Version]
  156. Nabavi, S.F.; Braidy, N.; Habtemariam, S.; Orhan, I.E.; Daglia, M.; Manayi, A.; Gortzi, O.; Nabavi, S.M. Neuroprotective Effects of Chrysin: From Chemistry to Medicine. Neurochem. Int. 2015, 90, 224–231. [Google Scholar] [CrossRef]
  157. Zhou, Y.-X.; Zhang, H.; Peng, C. Puerarin: A Review of Pharmacological Effects. Phytother. Res. PTR 2014, 28, 961–975. [Google Scholar] [CrossRef] [PubMed]
  158. Zhang, Y.; Wang, J.-F.; Dong, J.; Wei, J.-Y.; Wang, Y.-N.; Dai, X.-H.; Wang, X.; Luo, M.-J.; Tan, W.; Deng, X.-M.; et al. Inhibition of α-Toxin Production by Subinhibitory Concentrations of Naringenin Controls Staphylococcus aureus Pneumonia. Fitoterapia 2013, 86, 92–99. [Google Scholar] [CrossRef] [PubMed]
  159. Salehi, B.; Fokou, P.V.T.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The Therapeutic Potential of Naringenin: A Review of Clinical Trials. Pharmaceuticals 2019, 12, 11. [Google Scholar] [CrossRef] [Green Version]
  160. Quave, C.L.; Plano, L.R.W.; Bennett, B.C. Quorum Sensing Inhibitors of Staphylococcus aureus from Italian Medicinal Plants. Planta Med. 2011, 77, 188–195. [Google Scholar] [CrossRef] [Green Version]
  161. Muhs, A.; Lyles, J.T.; Parlet, C.P.; Nelson, K.; Kavanaugh, J.S.; Horswill, A.R.; Quave, C.L. Virulence Inhibitors from Brazilian Peppertree Block Quorum Sensing and Abate Dermonecrosis in Skin Infection Models. Sci. Rep. 2017, 7, 42275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Todd, D.A.; Parlet, C.P.; Crosby, H.A.; Malone, C.L.; Heilmann, K.P.; Horswill, A.R.; Cech, N.B. Signal Biosynthesis Inhibition with Ambuic Acid as a Strategy To Target Antibiotic-Resistant Infections. Antimicrob. Agents Chemother. 2017, 61, e00263-17. [Google Scholar] [CrossRef] [Green Version]
  163. Geisinger, E.; Muir, T.W.; Novick, R.P. Agr Receptor Mutants Reveal Distinct Modes of Inhibition by Staphylococcal Autoinducing Peptides. Proc. Natl. Acad. Sci. USA 2009, 106, 1216–1221. [Google Scholar] [CrossRef] [Green Version]
  164. Malachowa, N.; Kobayashi, S.D.; Braughton, K.R.; DeLeo, F.R. Mouse Model of Staphylococcus aureus Skin Infection. In Mouse Models of Innate Immunity; Humana Press: Totowa, NJ, USA, 2013; Volume 1031, pp. 109–116. [Google Scholar] [CrossRef]
  165. Menzies, B.E.; Kernodle, D.S. Site-Directed Mutagenesis of the Alpha-Toxin Gene of Staphylococcus aureus: Role of Histidines in Toxin Activity in Vitro and in a Murine Model. Infect. Immun. 1994, 62, 1843–1847. [Google Scholar] [CrossRef] [Green Version]
  166. Bubeck Wardenburg, J.; Schneewind, O. Vaccine Protection against Staphylococcus aureus Pneumonia. J. Exp. Med. 2008, 205, 287–294. [Google Scholar] [CrossRef]
  167. Karauzum, H.; Venkatasubramaniam, A.; Adhikari, R.P.; Kort, T.; Holtsberg, F.W.; Mukherjee, I.; Mednikov, M.; Ortines, R.; Nguyen, N.T.Q.; Doan, T.M.N.; et al. IBT-V02: A Multicomponent Toxoid Vaccine Protects Against Primary and Secondary Skin Infections Caused by Staphylococcus aureus. Front. Immunol. 2021, 12, 624310. [Google Scholar] [CrossRef]
  168. Brown, E.L.; Dumitrescu, O.; Thomas, D.; Badiou, C.; Koers, E.M.; Choudhury, P.; Vazquez, V.; Etienne, J.; Lina, G.; Vandenesch, F.; et al. The Panton-Valentine Leukocidin Vaccine Protects Mice against Lung and Skin Infections Caused by Staphylococcus aureus USA300. Clin. Microbiol. Infect. 2009, 15, 156–164. [Google Scholar] [CrossRef] [Green Version]
  169. Karauzum, H.; Adhikari, R.P.; Sarwar, J.; Devi, V.S.; Abaandou, L.; Haudenschild, C.; Mahmoudieh, M.; Boroun, A.R.; Vu, H.; Nguyen, T.; et al. Structurally Designed Attenuated Subunit Vaccines for S. aureus LukS-PV and LukF-PV Confer Protection in a Mouse Bacteremia Model. PLoS ONE 2013, 8, e65384. [Google Scholar] [CrossRef]
  170. Landrum, M.L.; Lalani, T.; Niknian, M.; Maguire, J.D.; Hospenthal, D.R.; Fattom, A.; Taylor, K.; Fraser, J.; Wilkins, K.; Ellis, M.W.; et al. Safety and Immunogenicity of a Recombinant Staphylococcus aureus α-Toxoid and a Recombinant Panton-Valentine Leukocidin Subunit, in Healthy Adults. Hum. Vaccines Immunother. 2017, 13, 791–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Jones, T. StaphVAX (Nabi). Curr. Opin. Investig. Drugs Lond. Engl. 2000 2002, 3, 48–50. [Google Scholar]
  172. Staphylococcus aureus Vaccine Conjugate–Nabi: Nabi-StaphVAX, StaphVAX. Drugs RD 2003, 4, 383–385. [CrossRef]
  173. Asensi, G.F.; de Sales, N.F.F.; Dutra, F.F.; Feijó, D.F.; Bozza, M.T.; Ulrich, R.G.; Miyoshi, A.; de Morais, K.; Azevedo, V.A.d.C.; Silva, J.T.; et al. Oral Immunization with Lactococcus lactis Secreting Attenuated Recombinant Staphylococcal Enterotoxin B Induces a Protective Immune Response in a Murine Model. Microb. Cell Fact. 2013, 12, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Chen, W.H.; Pasetti, M.F.; Adhikari, R.P.; Baughman, H.; Douglas, R.; El-Khorazaty, J.; Greenberg, N.; Holtsberg, F.W.; Liao, G.C.; Reymann, M.K.; et al. Safety and Immunogenicity of a Parenterally Administered, Structure-Based Rationally Modified Recombinant Staphylococcal Enterotoxin B Protein Vaccine, STEBVax. Clin. Vaccine Immunol. 2016, 23, 918–925. [Google Scholar] [CrossRef] [Green Version]
  175. Hu, D.-L.; Omoe, K.; Sasaki, S.; Sashinami, H.; Sakuraba, H.; Yokomizo, Y.; Shinagawa, K.; Nakane, A. Vaccination with Nontoxic Mutant Toxic Shock Syndrome Toxin 1 Protects against Staphylococcus aureus Infection. J. Infect. Dis. 2003, 188, 743–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Schwameis, M.; Roppenser, B.; Firbas, C.; Gruener, C.S.; Model, N.; Stich, N.; Roetzer, A.; Buchtele, N.; Jilma, B.; Eibl, M.M. Safety, Tolerability, and Immunogenicity of a Recombinant Toxic Shock Syndrome Toxin (RTSST)-1 Variant Vaccine: A Randomised, Double-Blind, Adjuvant-Controlled, Dose Escalation First-in-Man Trial. Lancet Infect. Dis. 2016, 16, 1036–1044. [Google Scholar] [CrossRef]
  177. Roetzer, A.; Jilma, B.; Eibl, M.M. Vaccine against Toxic Shock Syndrome in a First-in-Man Clinical Trial. Expert Rev. Vaccines 2017, 16, 81–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Brown, L.; Wolf, J.M.; Prados-Rosales, R.; Casadevall, A. Through the Wall: Extracellular Vesicles in Gram-Positive Bacteria, Mycobacteria and Fungi. Nat. Rev. Microbiol. 2015, 13, 620–630. [Google Scholar] [CrossRef] [Green Version]
  179. Wang, X.; Koffi, P.F.; English, O.F.; Lee, J.C. Staphylococcus aureus Extracellular Vesicles: A Story of Toxicity and the Stress of 2020. Toxins 2021, 13, 75. [Google Scholar] [CrossRef] [PubMed]
  180. Mattis, D.M.; Spaulding, A.R.; Chuang-Smith, O.N.; Sundberg, E.J.; Schlievert, P.M.; Kranz, D.M. Engineering a Soluble High-Affinity Receptor Domain That Neutralizes Staphylococcal Enterotoxin C in Rabbit Models of Disease. Protein Eng. Des. Sel. PEDS 2013, 26, 133–142. [Google Scholar] [CrossRef] [Green Version]
  181. Wang, X.; Thompson, C.D.; Weidenmaier, C.; Lee, J.C. Release of Staphylococcus aureus Extracellular Vesicles and Their Application as a Vaccine Platform. Nat. Commun. 2018, 9, 1379. [Google Scholar] [CrossRef] [Green Version]
  182. da Silveira, S.A.; Perez, A. Improving the Fate of Severely Infected Patients: The Promise of Anti-Toxin Treatments and Superiority Trials. Expert Rev. Anti Infect. Ther. 2017, 15, 973–975. [Google Scholar] [CrossRef] [Green Version]
  183. Parker, D. Humanized Mouse Models of Staphylococcus aureus Infection. Front. Immunol. 2017, 8, 512. [Google Scholar] [CrossRef]
  184. Allen, T.M.; Brehm, M.A.; Bridges, S.; Ferguson, S.; Kumar, P.; Mirochnitchenko, O.; Palucka, K.; Pelanda, R.; Sanders-Beer, B.; Shultz, L.D.; et al. Humanized Immune System Mouse Models: Progress, Challenges and Opportunities. Nat. Immunol. 2019, 20, 770–774. [Google Scholar] [CrossRef] [PubMed]
  185. Mrochen, D.M.; Fernandes de Oliveira, L.M.; Raafat, D.; Holtfreter, S. Staphylococcus aureus Host Tropism and Its Implications for Murine Infection Models. Int. J. Mol. Sci. 2020, 21, 7061. [Google Scholar] [CrossRef] [PubMed]
  186. Khan, B.A.; Yeh, A.J.; Cheung, G.Y.; Otto, M. Investigational Therapies Targeting Quorum-Sensing for the Treatment of Staphylococcus aureus Infections. Expert Opin. Investig. Drugs 2015, 24, 689–704. [Google Scholar] [CrossRef] [PubMed]
  187. Kumar, K.; Chen, J.; Drlica, K.; Shopsin, B. Tuning of the Lethal Response to Multiple Stressors with a Single-Site Mutation during Clinical Infection by Staphylococcus aureus. MBio 2017, 8, e01476-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Bojer, M.S.; Lindemose, S.; Vestergaard, M.; Ingmer, H. Quorum Sensing-Regulated Phenol-Soluble Modulins Limit Persister Cell Populations in Staphylococcus aureus. Front. Microbiol. 2018, 9, 255. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ahmad-Mansour, N.; Loubet, P.; Pouget, C.; Dunyach-Remy, C.; Sotto, A.; Lavigne, J.-P.; Molle, V. Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments. Toxins 2021, 13, 677. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins13100677

AMA Style

Ahmad-Mansour N, Loubet P, Pouget C, Dunyach-Remy C, Sotto A, Lavigne J-P, Molle V. Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments. Toxins. 2021; 13(10):677. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins13100677

Chicago/Turabian Style

Ahmad-Mansour, Nour, Paul Loubet, Cassandra Pouget, Catherine Dunyach-Remy, Albert Sotto, Jean-Philippe Lavigne, and Virginie Molle. 2021. "Staphylococcus aureus Toxins: An Update on Their Pathogenic Properties and Potential Treatments" Toxins 13, no. 10: 677. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins13100677

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