Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Feb 1.
Published in final edited form as: Curr Opin Microbiol. 2013 Feb;16(1):63–69. doi: 10.1016/j.mib.2013.01.012

The effects of Staphylococcus aureus leukotoxins on the host: cell lysis and beyond

Pauline Yoong 1, Victor J Torres 1
PMCID: PMC3670676  NIHMSID: NIHMS443541  PMID: 23466211

Abstract

The success of Staphylococcus aureus as a leading cause of deadly hospital-acquired and community-acquired infections is attributed to its high-level resistance to most antibiotics, and the multitude of virulence factors it elaborates. Most clinical isolates produce up to four bi-component pore-forming toxins capable of lysing cells of the immune system. Subtle differences in activity and target range of each leukotoxin suggest that these toxins are not redundant, but instead may have specialized functions in attacking and/or evading host defenses. In turn, the host has developed countermeasures recognizing sublytic levels of leukotoxins as signals to activate protective immune defenses. The opposing cytotoxic and immune-activating effects of leukotoxins on host cells make for a complex dynamic between S. aureus and the host.

Introduction

Staphylococcus aureus is a highly successful bacterial pathogen that causes significant hospital-acquired infections [1]. Its success is attributed in part to its rapid acquisition of resistance to most available antimicrobial therapies. Even more alarming, it has recently begun causing deadly infections outside of healthcare settings, in relatively healthy and young individuals [2,3]. Strains isolated from both community-acquired and hospital-acquired infections are predominantly methicillin-resistant (MRSA), making successful treatment highly challenging [2]. In addition to its resistance to almost every optimally effective antibiotic, the tenacity of S. aureus as a pathogen can be attributed to its arsenal of virulence factors designed to evade or attack host defenses at every level [4,5]. A subset of its virulence factors includes a collection of pore-forming toxins capable of targeted killing of select host cells by creating channels in the plasma membrane. The resulting osmotic dysregulation eventually leads to cell lysis. The pore-forming leukotoxins produced and secreted into the extracellular milieu by S. aureus are unique in that they consist of two different protein components that assemble together to form β-barrel pores [6,7••]. S. aureus strains associated with human infections produce four leukotoxins with considerable sequence similarity: the Panton-Valentine Leukocidin (PVL), gamma (γ)-hemolysin (HlgACB), Leukotoxin ED (LukED), and Leukotoxin AB/GH (LukAB/GH). This review outlines the activities of each leukotoxin, as well as highlights the differences between them (Table 1 and Figure 1).

Table 1.

List of Staphylococcus aureus bicomponent leukotoxins, and the ‘S’ and ‘F’ subunits that encompass each leukotoxin

Leukotoxin S subunit F subunit
PVL LukS-PV LukF-PV
LukAB/LukGH LukA/LukH LukB/LukG
LukED LukE LukD
γ-Hemolysin HlgA, HlgC HlgB

Figure 1.

Figure 1

Amino acid sequence alignments of S. aureus leukotoxin subunits, and percentage identities between them. Panel (a) shows a comparison between ‘S’ subunits of leukotoxins, while panel (b) shows a comparison between ‘F’ subunits. Amino acid sequences from the predicted secreted toxin subunits from MRSA USA300 strain FPR3757 were aligned using the Clustal Omega program powered by the European Molecular Biology Laboratory-European Bioinformatics Institute. Red shading signifies fully conserved residues, gray shading indicates conservation between groups of strongly similar properties, and blue letters denote conservation between groups of weakly similar residues. Tables denote the percentage identity between amino acid sequences of the respective leukotoxin subunits.

Leukotoxin pore formation

The two protein subunits of each leukotoxin are classified as ‘S’ and ‘F’. This nomenclature corresponds to the initial purification of PVL where the first eluted subunit was termed fast (F), and the subunit that eluted subsequently was termed slow (S) [8]. The mechanism by which these toxins bind and/or interact with the target host cells has not been completely elucidated. Initial studies with PVL gave rise to a model whereby LukS-PV serves as the homing component that binds specifically to a host cell receptor, after which LukF-PV is recruited by LukS-PV already docked to its receptor [9]. Conversely, work carried out with γ-hemolysin, indicated that HlgB (the ‘F’ component of the toxin) binds to its target cell first, followed by the binding of HlgA (the ‘S’ component) [10]. More recent work suggests the possibility of distinct cellular receptors for both LukS-PV and LukF-PV [11].

Once localized on the cell surface, the subunits assemble into an octameric prepore with four of each alternating F and S components [6,7••,12]. Pore formation culminates when the stem domains of the assembled subunits unfold and insert into the cell membrane to form β-barrel pores (Figure 2, steps in pore formation outlined in gray).

Figure 2.

Figure 2

Schematic of S. aureus leukotoxin pore-formation, and the different cellular pathways it uses to activate host cells. Steps in pore formation are outlined in gray, while steps involved in immune activation are outlined with red dashed boxes and shapes. This model is based predominantly on research and data from PVL [6,9,11,38,39,49]. Activation of the inflammasome and Caspase-1 is based on work studying γ-hemolysin [44••,45].

This figure was adapted from [50].

Target cells of S. aureus leukotoxins

As their names would suggest, these leukotoxins can lyse cells of the leukocytic lineage; however, subtle differences in the susceptibilities of various host cells may explain why S. aureus produces multiple highly related molecules that seemingly perform the same function. For instance, all four leukotoxins have been demonstrated to kill human neutrophils [1315,16••,17••,18], but only γ-hemolysin, and to a lesser extent, LukED, exhibit lytic activity against red blood cells [10,13,18]. Further details of cellular targets of each leukotoxin will be expanded on in the following section.

Sensitivity to leukotoxins also differs between species. For example, human neutrophils are highly susceptible to PVL and LukAB/GH, while neutrophils from mice are largely resistant to lysis by both toxins [16••,19]. Susceptibility of cells from any one species also varies depending on the toxin, as in the case of monkey neutrophils, which are highly resistant to killing by PVL [16••] but susceptible to LukAB/GH [19]. In addition, LukED appears to be the only leukotoxin that can kill mouse neutrophils efficiently [20••].

Leukotoxin involvement in S. aureus pathogenesis

γ-Hemolysin (HlgAB/HlgCB)

Two protein subunit combinations comprise γ-hemolysin, both sharing the same F subunit (HlgB), but differing in S subunit composition (HlgA or HlgC) [21]. Given that γ-hemolysin is the only leukotoxin within this family that can lyse red blood cells with high efficiency, it was not surprising to find that the hlg genes are highly upregulated in human blood. In turn, γ-hemolysin promotes the survival of S. aureus in blood [22]. Consistent with these ex vivo findings, γ-hemolysin conferred an advantage to S. aureus in mouse models of bloodstream infections, including bacteremia and septic arthritis [22,23]. The presence of hlg genes in virtually all S. aureus strains (99%), not only in isolates from bloodborne infections [24], would suggest that γ-hemolysin could also be an important factor in multiple modes of infection.

LukED

Despite the initial report of this toxin over a decade ago [14,18], research on LukED has remained minimal. As mentioned in the preceding section, in addition to its toxicity toward human neutrophils and rabbit red blood cells, LukED is unique in being highly effective at killing mouse phagocytes [20]. Accordingly, LukED was found to contribute to the lethality observed in a mouse model of S. aureus bacteremia. It did so by targeting and killing phagocytes, thereby allowing LukED+ S. aureus to proliferate. The lukED genes have been found in 87% of clinical strains [18], including many MRSA strains responsible for current pandemics [20], suggesting a potentially significant contribution of this leukotoxin to the pathogenesis of S. aureus.

LukAB/GH

LukAB/GH is the most recently identified leukotoxin [17••,25], and is also the most divergent among those produced by S. aureus. Sequence identities with other Staphylococcal leukotoxins are below 40% (Figure 1). Despite this low level of sequence identity, LukAB/ GH can efficiently kill human neutrophils, monocytes, macrophages, and dendritic cells [17••]. Its toxicity is enhanced further by synergism with PVL [25]. Interestingly, LukAB/GH is the only leukotoxin reported to associate with the cell surface of S. aureus, in addition to being secreted like the other leukotoxins [25]. The mechanism of LukAB/GH association with the bacterial surface is yet to be investigated. It is also presently unclear if surface-associated LukAB/GH can function as an active toxin. So far LukAB/GH is the only bi-component leukotoxin known to enhance S. aureus survival upon phagocytosis by human neutrophils [17••], implying that in addition to ‘conventional’ cell lysis from outside the neutrophil, this toxin may also facilitate the escape of S. aureus from the phagosome. Toxicity of LukAB/GH toward neutrophils is the likely mechanism by which the toxin promotes the survival of S. aureus in whole blood, as it has no detectable hemolytic activity [17••]. The contribution of LukAB/GH to virulence was recently demonstrated using a low infectious dose systemic infection model, where presence of the toxin was associated with higher bacterial burden in infected kidneys [17••]. While exhaustive analyses identifying the presence of lukAB/GH genes in clinical strains of S. aureus are not presently available, all sequenced S. aureus strains and all clinical isolates examined in our laboratory are positive for lukAB/GH, further suggesting that this toxin is also a highly relevant virulence factor produced by S. aureus.

PVL

PVL is present in a small percentage (~5%) of overall S. aureus clinical strains, although it is strongly associated with community-acquired MRSA strains (~85%), in particular those causing pneumonia, and skin and soft tissue infections [2,26]. In spite of its apparent linkage to virulent strains capable of causing deadly infections in healthy people, its contribution to virulence has not been conclusively proven. Several multinational studies have concluded that S. aureus infection outcomes are not linked to PVL production [2729], while in some other cases, infections with PVL+ strains are even associated with better outcome [3032]. As such, while PVL is the most studied Staphylococcal leukotoxin, it is also arguably the most contentious.

Because of the resistance of mouse cells to lysis by PVL [16••], murine models of infection examining the contribution of PVL’s pore-forming ability yielded inconsistent conclusions by various research groups. Instead, the use of rabbit infection models was considered more appropriate, given that rabbit neutrophils are highly sensitive to PVL [16••]. Nonetheless, data compiled from several rabbit models provided little new insight. One study showed that PVL conferred a modest transient advantage to S. aureus in the early stages of bacteremia and in a pneumonia model, but only at very high inocula (>5 × 109 CFU) [33,34]. Using rabbit skin infection models, divergent conclusions were reached by two independent groups, despite both using the same MRSA strain. Lipinska et al. showed the PVL+ strain to cause more significant skin necrosis compared to the isogenic Δpvl strain [35]. On the contrary, Kobayashi et al. reported that larger abscesses were caused by the Δpvl strain [36]. It should be noted however, that there were subtle variations in experimental design, including the species of rabbit used, slight differences in inocula and site of injection. There was also a study in nonhuman primates (cynomolgus macaques) that compared an isogenic WT and Δpvl MRSA strain in causing lower respiratory tract disease [37]. Outcomes of infection were comparable between WT and Δpvl infected animals, however, the sample sizes were small. Thus, after decades of research by multiple groups, the contribution of PVL to S. aureus and MRSA pathogenesis remains inconclusive.

Immune-activating properties of Staphylococcal leukotoxins

In addition to the pore-forming capabilities of these leukotoxins, it has become increasingly apparent that these molecules are also potent activators of innate immunity. Early studies titrating PVL below the threshold of cytotoxicity on its target cells (sublytic concentrations), revealed that PVL activates cells to amplify host immune defenses without causing cell damage. At sublytic concentrations, PVL can cause human neutrophils to secrete proinflammatory mediators (including interleukin (IL)-8 and leukotriene B4) [38,39] and antimicrobial factors [40,41], as well as enhance phagocytosis and killing of S. aureus [42••]. Additionally, PVL can also directly activate cells that are resistant to its lytic activity, including mouse cells and non-immune human cells [40]. Inflammatory cytokines elicited by PVL may in turn activate other cells to secrete additional immune mediators, potentially augmenting inflammation [43••]. As such, host cell activation by PVL has the potential to outweigh its lytic action, possibly explaining why PVL is sometimes associated with better outcomes for the host.

There is ample evidence that other Staphylococcal leukotoxins are also highly effective immune-activators. Like PVL, LukAB/GH can elicit an inflammatory response in rabbits when injected intradermally [19]. γ-Hemolysin can also synergize with α-hemolysin and β-hemolysin to activate the NLRP3 inflammasome and Caspase-1 [44••]. Interestingly, pore-formation on immune cells can trigger activation of Caspase-1, suggesting that all leukotoxins are capable of doing so [45]. Caspase-1 in turn initiates inflammatory programmed cell death, termed pyroptosis, coupled with the release of large amounts of proinflammatory cytokines IL-1β, IL-18 and IL-33 [45,46]. Cytokine activation of surrounding immune cells and Caspase-1 triggered pathways involved in membrane biogenesis are also linked to increased cell survival [45] (Figure 2, steps involved in immune activation are outlined with red dashed boxes). Taken together, the contribution of leukotoxins to S. aureus infection is likely to be multifactorial, contingent upon toxin concentration.

Because of the potent lytic activity of the Staphylococcal leukotoxins, there is little doubt that they have tremendous potential to cause cellular damage, which in turn will potentiate S. aureus pathogenesis. In an attempt to counter this response, the mammalian host recognizes low concentrations of these leukotoxins to amplify immune defenses. There are multiple factors involved in establishing an infection, including bacterial load, bacterial factors elaborated during infection, site of infection, immune status of the host, and antimicrobial therapies, to name a few. Interplay between host defenses and bacterial factors can dynamically tilt the balance between the host response and the pathogenic process, impacting the outcome of infection.

Complexities of multiple leukotoxin production by S. aureus

Despite production of numerous related leukotoxins by S. aureus, each leukotoxin has a unique repertoire of targets cells, indicating a distinct role for each during the infection process. Additionally, expression and production of each leukotoxin varies independently of each other when S. aureus is grown under different conditions [47,48]. This highlights the possibility for differential toxin production at different tissue sites, as the unique environment at each site could alter bacterial growth and toxin production. Adding to the complexity, S. aureus clinical isolates also differ in the combination of leukotoxins that they produce [24]. This variability could have major consequences for disease progression when the potential for synergism between leukotoxins is considered [25]. The non-lytic, immune-activating effects of these leukotoxins further confound the actual sequence of events during an infection. Further comprehensive study and understanding of the Staphylococcal leukotoxins are needed, as many thought provoking questions remain.

Acknowledgments

We thank Francis Alonzo III and Ashley L. DuMont for critical reading of this manuscript. Work in the Torres laboratory is supported by funds from the National Institute of Allergy and Infectious Diseases (R56AI091856-01A1), the American Heart Association (Scientist Development Grant 09SDG2060036), and New York University School of Medicine Development Funds to VJT. PY was supported in part by a The Vilcek Endowed Fund fellowship from New York University School of Medicine.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

• of special interest

•• of outstanding interest

  • 1.Fridkin SK, Hageman JC, Morrison M, Sanza LT, Como-Sabetti K, Jernigan JA, Harriman K, Harrison LH, Lynfield R, Farley MM. Methicillin-resistant Staphylococcus aureus disease in three communities. N Engl J Med. 2005;352:1436–1444. doi: 10.1056/NEJMoa043252. [DOI] [PubMed] [Google Scholar]
  • 2.Naimi TS, LeDell KH, Como-Sabetti K, Borchardt SM, Boxrud DJ, Etienne J, Johnson SK, Vandenesch F, Fridkin S, O’Boyle C, et al. Comparison of community- and health care-associated methicillin-resistant Staphylococcus aureus infection. JAMA. 2003;290:2976–2984. doi: 10.1001/jama.290.22.2976. [DOI] [PubMed] [Google Scholar]
  • 3.Herold BC, Immergluck LC, Maranan MC, Lauderdale DS, Gaskin RE, Boyle-Vavra S, Leitch CD, Daum RS. Community-acquired methicillin-resistant Staphylococcus aureus in children with no identified predisposing risk. JAMA. 1998;279:593–598. doi: 10.1001/jama.279.8.593. [DOI] [PubMed] [Google Scholar]
  • 4.Otto M. Basis of virulence in community-associated methicillin-resistant Staphylococcus aureus. Annu Rev Microbiol. 2010;64:143–162. doi: 10.1146/annurev.micro.112408.134309. [DOI] [PubMed] [Google Scholar]
  • 5.Bartlett AH, Hulten KG. Staphylococcus aureus pathogenesis: secretion systems, adhesins, and invasins. Pediatr Infect Dis J. 2010;29:860–861. doi: 10.1097/INF.0b013e3181ef2477. [DOI] [PubMed] [Google Scholar]
  • 6•.Aman MJ, Karauzum H, Bowden MG, Nguyen TL. Structural model of the pre-pore ring-like structure of Panton-Valentine leukocidin: providing dimensionality to biophysical and mutational data. J Biomol Struct Dyn. 2010;28:1–12. doi: 10.1080/073911010010524952. The authors present a detailed structural model of pore-formation by PVL, which provided a guide as to how PVL and similar toxins assemble and disrupt host cell membranes. [DOI] [PubMed] [Google Scholar]
  • 7••.Yamashita K, Kawai Y, Tanaka Y, Hirano N, Kaneko J, Tomita N, Ohta M, Kamio Y, Yao M, Tanaka I. Crystal structure of the octameric pore of staphylococcal gamma-hemolysin reveals the beta-barrel pore formation mechanism by two components. Proc Natl Acad Sci USA. 2011;108:17314–17319. doi: 10.1073/pnas.1110402108. The authors provide the first high resolution, three-dimensional structure of a S. aureus leukocidin pore. This landmark work provides a detailed view of how the S and F subunits assemble into an octameric pore. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Finck-Barbancon V, Prevost G, Piemont Y. Improved purification of leukocidin from Staphylococcus aureus and toxin distribution among hospital strains. Res Microbiol. 1991;142:75–85. doi: 10.1016/0923-2508(91)90099-v. [DOI] [PubMed] [Google Scholar]
  • 9.Colin DA, Mazurier I, Sire S, Finck-Barbancon V. Interaction of the two components of leukocidin from Staphylococcus aureus with human polymorphonuclear leukocyte membranes: sequential binding and subsequent activation. Infect Immun. 1994;62:3184–3188. doi: 10.1128/iai.62.8.3184-3188.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kaneko J, Ozawa T, Tomita T, Kamio Y. Sequential binding of Staphylococcal gamma-hemolysin to human erythrocytes and complex formation of the hemolysin on the cell surface. Biosci Biotechnol Biochem. 1997;61:846–851. doi: 10.1271/bbb.61.846. [DOI] [PubMed] [Google Scholar]
  • 11•.Meyer F, Girardot R, Piemont Y, Prevost G, Colin DA. Analysis of the specificity of Panton-Valentine leucocidin and gamma-hemolysin F component binding. Infect Immun. 2009;77:266–273. doi: 10.1128/IAI.00402-08. The authors provide evidence that the leukocidin F subunits can also bind to the plasma membrane of host cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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. doi: 10.1271/bbb.68.981. [DOI] [PubMed] [Google Scholar]
  • 13.Prevost G, Cribier B, Couppie P, Petiau P, Supersac G, Finck-Barbancon V, Monteil H, Piemont Y. Panton-Valentine leucocidin and gamma-hemolysin from Staphylococcus aureus ATCC 49775 are encoded by distinct genetic loci and have different biological activities. Infect Immun. 1995;63:4121–4129. doi: 10.1128/iai.63.10.4121-4129.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gravet A, Colin DA, Keller D, Girardot R, Monteil H, Prevost G. Characterization of a novel structural member, LukE-LukD, of the bi-component staphylococcal leucotoxins family. FEBS Lett. 1998;436:202–208. doi: 10.1016/s0014-5793(98)01130-2. [DOI] [PubMed] [Google Scholar]
  • 15.Gauduchon V, Werner S, Prevost G, Monteil H, Colin DA. Flow cytometric determination of Panton-Valentine leucocidin S component binding. Infect Immun. 2001;69:2390–2395. doi: 10.1128/IAI.69.4.2390-2395.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16••.Loffler B, Hussain M, Grundmeier M, Bruck M, Holzinger D, Varga G, Roth J, Kahl BC, Proctor RA, Peters G. Staphylococcus aureus Panton-Valentine leukocidin is a very potent cytotoxic factor for human neutrophils. PLoS Pathog. 2010;6:e100715. doi: 10.1371/journal.ppat.1000715. This is a comprehensive study where the authors examine the cytotoxic activity of PVL toward neutrophils isolated from different species. The authors provide strong evidence that question the value of infection-models in mice and non-human primates to elucidate the contribution of PVL to S. aureus pathogenesis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17••.Dumont AL, Nygaard TK, Watkins RL, Smith A, Kozhaya L, Kreiswirth BN, Shopsin B, Unutmaz D, Voyich JM, Torres VJ. Characterization of a new cytotoxin that contributes to Staphylococcus aureus pathogenesis. Mol Microbiol. 2010;79:814–825. doi: 10.1111/j.1365-2958.2010.07490.x. This study describes the identification of LukAB (also known as LukGH) as a critical virulence factor involved in the cytotoxicity of both MSSA and MRSA toward human neutrophils, monocytes, macrophages, and dendritic cells. In addition, this study demonstrates for the first time that LukAB enhances S. aureus survival upon phagocytosis by neutrophils. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Morinaga N, Kaihou Y, Noda M. Purification, cloning and characterization of variant LukE-LukD with strong leukocidal activity of staphylococcal bi-component leukotoxin family. Microbiol Immunol. 2003;47:81–90. doi: 10.1111/j.1348-0421.2003.tb02789.x. [DOI] [PubMed] [Google Scholar]
  • 19•.Malachowa N, Kobayashi SD, Braughton KR, Whitney AR, Parnell MJ, Gardner DJ, Deleo FR. Staphylococcus aureus leukotoxin GH promotes inflammation. J Infect Dis. 2012;206:1185–1193. doi: 10.1093/infdis/jis495. The authors demonstrate that LukAB/GH cytotoxicity toward neutrophils is species dependent. They report that neutrophils from human and cynomolgus monkey are the most susceptible to this toxin followed by rabbit and then murine PMNs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20••.Alonzo F, 3rd, Benson MA, Chen J, Novick RP, Shopsin B, Torres VJ. Staphylococcus aureus leucocidin ED contributes to systemic infection by targeting neutrophils and promoting bacterial growth in vivo. Mol Microbiol. 2012;83:423–435. doi: 10.1111/j.1365-2958.2011.07942.x. This study demonstrates for the first time that LukED is critical for S. aureus bloodstream infection in mice. The authors found that this toxin promotes bacterial replication in vivo by killing phagocytes at the sites of infection. The activity of LukED toward murine phagocytes highlights the utility of murine models and LukED to unmask the contribution of leukotoxins to S. aureus pathobiology. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kamio Y, Rahman A, Nariya H, Ozawa T, Izaki K. The two staphylococcal bi-component toxins, leukocidin and gamma-hemolysin, share one component in common. FEBS Lett. 1993;321:15–18. doi: 10.1016/0014-5793(93)80611-w. [DOI] [PubMed] [Google Scholar]
  • 22.Malachowa N, Whitney AR, Kobayashi SD, Sturdevant DE, Kennedy AD, Braughton KR, Shabb DW, Diep BA, Chambers HF, Otto M, et al. Global changes in Staphylococcus aureus gene expression in human blood. PLoS ONE. 2011;6:e18617. doi: 10.1371/journal.pone.0018617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Nilsson IM, Hartford O, Foster T, Tarkowski A. Alpha-toxin and gamma-toxin jointly promote Staphylococcus aureus virulence in murine septic arthritis. Infect Immun. 1999;67:1045–1049. doi: 10.1128/iai.67.3.1045-1049.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.von Eiff C, Friedrich AW, Peters G, Becker K. Prevalence of genes encoding for members of the staphylococcal leukotoxin family among clinical isolates of Staphylococcus aureus. Diagn Microbiol Infect Dis. 2004;49:157–162. doi: 10.1016/j.diagmicrobio.2004.03.009. [DOI] [PubMed] [Google Scholar]
  • 25•.Ventura CL, Malachowa N, Hammer CH, Nardone GA, Robinson MA, Kobayashi SD, DeLeo FR. Identification of a novel Staphylococcus aureus two-component leukotoxin using cell surface proteomics. PLoS ONE. 2010;5:e11634. doi: 10.1371/journal.pone.0011634. This study describes the identification of LukGH (also known as LukAB) as a major leukotoxin involved in the cytotoxicity of CA-MRSA clone USA300. In addition, this study revealed that this toxin is both secreted and can be found associated with the bacterial surface. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lina G, Piemont Y, Godail-Gamot F, Bes M, Peter MO, Gauduchon V, Vandenesch F, Etienne J. Involvement of Panton-Valentine leukocidin-producing Staphylococcus aureus in primary skin infections and pneumonia. Clin Infect Dis. 1999;29:1128–1132. doi: 10.1086/313461. [DOI] [PubMed] [Google Scholar]
  • 27.Sharma-Kuinkel BK, Ahn SH, Rude TH, Zhang Y, Tong SY, Ruffin F, Genter FC, Braughton KR, Deleo FR, Barriere SL, et al. Presence of genes encoding panton-valentine leukocidin is not the primary determinant of outcome in patients with hospital-acquired pneumonia due to Staphylococcus aureus. J Clin Microbiol. 2012;50:848–856. doi: 10.1128/JCM.06219-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boakes E, Kearns AM, Badiou C, Lina G, Hill RL, Ellington MJ. Do differences in Panton-Valentine leukocidin production among international methicillin-resistant Staphylococcus aureus clones affect disease presentation and severity? J Clin Microbiol. 2012;50:1773–1776. doi: 10.1128/JCM.06421-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hamilton SM, Bryant AE, Carroll KC, Lockary V, Ma Y, McIndoo E, Miller LG, Perdreau-Remington F, Pullman J, Risi GF, et al. In vitro production of panton-valentine leukocidin among strains of methicillin-resistant Staphylococcus aureus causing diverse infections. Clin Infect Dis. 2007;45:1550–1558. doi: 10.1086/523581. [DOI] [PubMed] [Google Scholar]
  • 30.Campbell SJ, Deshmukh HS, Nelson CL, Bae IG, Stryjewski ME, Federspiel JJ, Tonthat GT, Rude TH, Barriere SL, Corey R, et al. Genotypic characteristics of Staphylococcus aureus isolates from a multinational trial of complicated skin and skin structure infections. J Clin Microbiol. 2008;46:678–684. doi: 10.1128/JCM.01822-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bae IG, Tonthat GT, Stryjewski ME, Rude TH, Reilly LF, Barriere SL, Genter FC, Corey GR, Fowler VG., Jr Presence of genes encoding the panton-valentine leukocidin exotoxin is not the primary determinant of outcome in patients with complicated skin and skin structure infections due to methicillin-resistant Staphylococcus aureus: results of a multinational trial. J Clin Microbiol. 2009;47:3952–3957. doi: 10.1128/JCM.01643-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lalani T, Federspiel JJ, Boucher HW, Rude TH, Bae IG, Rybak MJ, Tonthat GT, Corey GR, Stryjewski ME, Sakoulas G, et al. Associations between the genotypes of Staphylococcus aureus bloodstream isolates and clinical characteristics and outcomes of bacteremic patients. J Clin Microbiol. 2008;46:2890–2896. doi: 10.1128/JCM.00905-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Diep BA, Chan L, Tattevin P, Kajikawa O, Martin TR, Basuino L, Mai TT, Marbach H, Braughton KR, Whitney AR, et al. Polymorphonuclear leukocytes mediate Staphylococcus aureus Panton-Valentine leukocidin-induced lung inflammation and injury. Proc Natl Acad Sci USA. 2010;107:5587–5592. doi: 10.1073/pnas.0912403107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Diep BA, Palazzolo-Ballance AM, Tattevin P, Basuino L, Braughton KR, Whitney AR, Chen L, Kreiswirth BN, Otto M, DeLeo FR, et al. Contribution of Panton-Valentine leukocidin in community-associated methicillin-resistant Staphylococcus aureus pathogenesis. PLoS ONE. 2008;3:e3198. doi: 10.1371/journal.pone.0003198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lipinska U, Hermans K, Meulemans L, Dumitrescu O, Badiou C, Duchateau L, Haesebrouck F, Etienne J, Lina G. Panton-Valentine leukocidin does play a role in the early stage of Staphylococcus aureus skin infections: a rabbit model. PLoS ONE. 2011;6:e22864. doi: 10.1371/journal.pone.0022864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kobayashi SD, Malachowa N, Whitney AR, Braughton KR, Gardner DJ, Long D, Bubeck Wardenburg J, Schneewind O, Otto M, Deleo FR. Comparative analysis of USA300 virulence determinants in a rabbit model of skin and soft tissue infection. J Infect Dis. 2011;204:937–941. doi: 10.1093/infdis/jir441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Olsen RJ, Kobayashi SD, Ayeras AA, Ashraf M, Graves SF, Ragasa W, Humbird T, Greaver JL, Cantu C, Swain JL, et al. Lack of a major role of Staphylococcus aureus Panton-Valentine leukocidin in lower respiratory tract infection in nonhuman primates. Am J Pathol. 2010;176:1346–1354. doi: 10.2353/ajpath.2010.090960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Konig B, Prevost G, Piemont Y, Konig W. Effects of Staphylococcus aureus leukocidins on inflammatory mediator release from human granulocytes. J Infect Dis. 1994;171:607–613. doi: 10.1093/infdis/171.3.607. [DOI] [PubMed] [Google Scholar]
  • 39.Staali L, Monteil H, Colin DA. The staphylococcal pore-forming leukotoxins open Ca2+ channels in the membrane of human polymorphonuclear neutrophils. J Membr Biol. 1998;162:209–216. doi: 10.1007/s002329900358. [DOI] [PubMed] [Google Scholar]
  • 40.Yoong P, Pier GB. Immune-activating properties of Panton-Valentine leukocidin improve the outcome in a model of methicillin-resistant Staphylococcus aureus pneumonia. Infect Immun. 2012;80:2894–2904. doi: 10.1128/IAI.06360-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Yoong P, Pier GB. Antibody-mediated enhancement of community-acquired methicillin-resistant Staphylococcus aureus infection. Proc Natl Acad Sci USA. 2010;107:2241–2246. doi: 10.1073/pnas.0910344107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42••.Graves SF, Kobayashi SD, Braughton KR, Whitney AR, Sturdevant DE, Rasmussen DL, Kirpotina LN, Quinn MT, DeLeo FR. Sublytic concentrations of Staphylococcus aureus Panton-Valentine leukocidin alter human PMN gene expression and enhance bactericidal capacity. J Leukoc Biol. 2012;92:361–374. doi: 10.1189/jlb.1111575. This comprehensive study investigates the effect of sublytic concentrations of PVL on neutrophil function. The authors provide strong evidence that exposure of human PMNs to sublytic concentrations of PVL elicits a proinflammatory response that enhances the host innate immune response against S. aureus. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43••.Perret M, Badiou C, Lina G, Burbaud S, Benito Y, Bes M, Cottin V, Couzon F, Juruj C, Dauwalder O, et al. Cross-talk between Staphylococcus aureus leukocidins-intoxicated macrophages and lung epithelial cells triggers chemokine secretion in an inflammasome-dependent manner. Cell Microbiol. 2012;14:1019–1036. doi: 10.1111/j.1462-5822.2012.01772.x. This comprehensive study demonstrates that PVL is a major virulence factor involved in the activation of the inflammasome and IL-1β production in human macrophages. In addition, this study nicely highlights the effect of PVL-induced inflammation on the amplification of proinflammatory cascade by cells that are not susceptible to PVL lysis. [DOI] [PubMed] [Google Scholar]
  • 44••.Munoz-Planillo R, Franchi L, Miller LS, Nunez G. A critical role for hemolysins and bacterial lipoproteins in Staphylococcus aureus-induced activation of the Nlrp3 inflammasome. J Immunol. 2009;183:3942–3948. doi: 10.4049/jimmunol.0900729. This study demonstrates that Gamma hemolysin synergizes with other bacterial toxins to activate inflammasomes and proinflammatory cascades of the host. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gurcel L, Abrami L, Girardin S, Tschopp J, van der Goot FG. Caspase-1 activation of lipid metabolic pathways in response to bacterial pore-forming toxins promotes cell survival. Cell. 2006;126:1135–1145. doi: 10.1016/j.cell.2006.07.033. [DOI] [PubMed] [Google Scholar]
  • 46.Bersbaken TFS, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7:99–109. doi: 10.1038/nrmicro2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Graves SF, Kobayashi SD, Braughton KR, Diep BA, Chambers HF, Otto M, Deleo FR. Relative contribution of Panton-Valentine leukocidin to PMN plasma membrane permeability and lysis caused by USA300 and USA400 culture supernatants. Microbes Infect. 2010;12:446–456. doi: 10.1016/j.micinf.2010.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Li M, Cheung GY, Hu J, Wang D, Joo HS, Deleo FR, Otto M. Comparative analysis of virulence and toxin expression of global community-associated methicillin-resistant Staphylococcus aureus strains. J Infect Dis. 2010;202:1866–1876. doi: 10.1086/657419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49•.Pietrocola G, Arciola CR, Rindi S, Di Poto A, Missineo A, Montanaro L, Speziale P. Toll-like receptors (TLRs) in innate immune defense against Staphylococcus aureus. Int J Artif Organs. 2011;34:799–810. doi: 10.5301/ijao.5000030. This concise report summarizes how toll-like receptors recognize staphylococcal components, including PVL, as a trigger for host defense. [DOI] [PubMed] [Google Scholar]
  • 50.Yoong P. Immunomodulation by the Panton-Valentine leukocidin can benefit the host during Staphylococcus aureus infections. Virulence. 2013;4:92–96. doi: 10.4161/viru.23165. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES