SUMMARY
Virulence of the emerging Community-Associated Methicillin-Resistant Staphylococcus aureus (CA-MRSA) and other highly pathogenic S. aureus depends on the recently discovered phenol-soluble modulin (PSM) peptide toxins, which combine the capacities to attract and lyse neutrophils. The molecular basis of PSM-stimulated neutrophil recruitment has remained unknown. We demonstrate that the human formyl peptide receptor 2 (FPR2/ALX), which has previously been implicated in control of endogenous inflammatory processes, senses PSMs at nanomolar concentrations and initiates proinflammatory neutrophil responses to CA-MRSA. Specific blocking of FPR2/ALX or deletion of PSM genes in CA-MRSA led to severely diminished capacities of neutrophils to detect CA-MRSA. A specific inhibitor of FPR2/ALX and its functional mouse counterpart blocked PSM-mediated leukocyte infiltration in vivo in a mouse model. Thus, the innate immune system uses a new FPR2/ALX-dependent mechanism to sense bacterial peptide toxins and detect highly virulent bacterial pathogens. FPR2/ALX represents an attractive target for new anti-infective or anti-inflammatory strategies.
INTRODUCTION
Staphylococcus aureus is a frequent constituent of the human microflora and also a major cause of severe infectious diseases ranging from skin and wound infections to life-threatening disseminated infections (Foster, 2004). While methicillin-resistant S. aureus (MRSA) strains, which are particularly refractory to antibiotic treatment, used to be restricted to hospital settings without colonizing healthy individuals, new highly pathogenic MRSA clones such as USA300 (LAC) and USA400 (MW2) are increasingly emerging in the population (Chambers and DeLeo, 2009; Said-Salim et al., 2003). These community-associated (CA-) MRSA represent a major threat even for healthy persons and demand new efforts to understand, prevent, and treat S. aureus infections. CA-MRSA combine extraordinary virulence, resistance, and spreading capacities and are causing alarming epidemics in various parts of the world (Chambers and DeLeo, 2009; Kahl and Peters, 2007). Severe infections by CA-MRSA and other highly pathogenic S. aureus strains are associated with massive influx and subsequent lysis of neutrophils with concomitant tissue damage (Wang et al., 2007; Bubeck et al., 2007). A number of virulence factors including α-hemolysin and the phage-encoded Panton-Valentine toxin appear to contribute to the virulence of CA-MRSA (Diep et al., 2008; Bubeck et al., 2007; Kahl and Peters, 2007). In addition, we have recently shown that virulence of CA-MRSA depends critically on secretion of high amounts of phenol-soluble modulin (PSM) peptide toxins. The seven core genome-encoded S. aureus PSMs one of which is the δ-toxin, a long-known virulence factor, and the PSM-mec produced by a minority of hospital-acquired MRSA (HA-MRSA) strains have lengths of 20–44 amino acids and α-helical, amphipathic secondary structures (Wang et al., 2007; Queck et al., 2009). They destroy neutrophils very efficiently, most probably by disrupting the neutrophil membranes. In contrast, less pathogenic S. aureus such as most HA-MRSA produce only very low PSM levels (Wang et al., 2007). While the PSMs are cytolytic at micromolar concentrations, they are also chemoattractants for neutrophils and contribute to massive leukocyte infiltration (Wang et al., 2007; Queck et al., 2009); the molecular basis of this latter phenomenon has remained unknown.
Neutrophils are the most important immune cells in local infections with a high capacity to eliminate invading microbes and activate additional host defense mechanisms. Neutrophils migrate to sites of infection in response to chemotactic stimuli, which are recognized by G protein-coupled receptors (GPCRs). In addition to endogenous mediators such as chemokines, neutrophils can directly respond to specific bacterial molecules. Leukocytes recognize such ‘pathogen-associated molecular patterns’ (PAMPs) e.g. via Toll-like receptors (TLRs) (Uematsu and Akira, 2006) or via certain GPCRs such as the formyl peptide receptor 1 (FPR1) (Le et al., 2002; Fu et al., 2006). While TLRs can activate host cells, only the latter type of receptor can elicit chemotactic migration. FPR1 recognizes formylated bacterial peptides, which represent a hallmark of bacterial infections since only bacterial cells start protein biosynthesis with a formylated methionine, while the cytoplasmic ribosomes of human cells use an unmodified methionine residue. Accordingly, formylated peptides have been shown to represent major chemotactic PAMPs (Dürr et al., 2006; Mader et al., 2010). S. aureus produces a specific inhibitor of the human FPR1, the chemotaxis-inhibitory protein of S. aureus (CHIPS) to block formylated peptide recognition (de Haas et al., 2004). However, it has also become clear that bacteria produce further chemotactic molecules of recently unknown nature (Dürr et al., 2006).
S. aureus also secretes a specific inhibitor for the FPR1-related formyl peptide receptor 2 (FPR2/ALX; initially denoted formyl-peptide receptor-like 1, FPRL1) of human neutrophils, the FPR2/ALX-inhibitory protein (FLIPr) (Prat et al., 2006). The reasons for FLIPr secretion by S. aureus have remained mysterious and have pointed toward a previously unknown role of FPR2/ALX in S. aureus infections. While it is clear that FPR1 plays an important role in antibacterial host defence FPR2/ALX has only residual affinity for bacterial formylated peptides and has hardly been implicated in pathogen recognition (Murphy et al., 1992). Instead, FPR2/ALX has been associated with chronic inflammatory and degenerative diseases such as atherosclerosis and Alzheimer’s disease because it responds to a variety of amyloidogenic peptides (e.g serum amyloid A, the β-amyloid Aβ1–42 peptide, and the prion protein fragment PrP106–126) and other endogenous ligands (Su et al., 1999; Cui et al., 2002; Fu et al., 2006; Ye et al., 2009). Moreover, FPR2/ALX senses anti-inflammatory agonists such as the lipid lipoxin A4, endogenous annexin 1-derived peptides, and synthetic peptide compounds and has also been referred to as receptor for lipoxin A4 and aspirin-triggered lipoxins (ALX) (Perretti et al., 2002; Hecht et al., 2009). Nevertheless, the major functions of FPR2/ALX and the second human FPR1 ortholog, FPR3 that binds e.g. an N-terminally acetylated endogenous peptide (Migeotte et al., 2005), have remained elusive. FPR-like receptors are highly diverse in the various mammalian species indicating that they are subjected to strong selection pressures (Gao et al., 1998; Fu et al., 2006; Ye et al., 2009). In addition to different substrate specificities, the numbers of FPR paralogs vary widely between species with seven to eight paralogs in mice compared to three in humans, which represents a major obstacle for studying the role of FPR-like receptors in animal models.
Since staphylococcal PSM peptides have previously been shown to attract neutrophils (Wang et al., 2007; Queck et al., 2009) we studied the recognition of these molecules by receptors of the human FPR family and found the most potent PSMs to be active at nanomolar concentrations. We demonstrate that the activity of PSMs does not necessarily depend on N-terminal formylation and identify FPR2/ALX as the major receptor for all seven PSMs. Moreover, FPR2/ALX represents the dominant receptor mediating neutrophil chemotaxis in response to CA-MRSA while FPR2/ALX is not involved in the cytotoxic activity of PSMs. These data suggest that the immune system has developed FPR2/ALX for the detection of PSM-like virulence factors and indicate a critical role of FPR2/ALX in innate immunity.
RESULTS
Formylated and Nonformylated PSMs Induce Ca2+ Influx in Human Neutrophils, which Cannot be Blocked by the FPR1 Inhibitor CHIPS
S. aureus PSMs have been shown to attract neutrophils but structure-function relationships of PSMs with regard to their chemotactic potential are poorly defined (Wang et al., 2007). We found PSMα3, the most active of the PSMs to attract human neutrophils at nanomolar concentrations (Figure 1A,B). The decrease of chemotaxis at PSMα3 concentrations above 100 nM reflects the activation of neutrophils by high amounts of GPCR ligands leading to halt of migration (Christophe et al., 2001) and causing the typical bell-shaped dose-response curve (Figure 1A). PSMα3 also elicited intracellular calcium ion flux in neutrophils, which is known to accompany chemotactic migration (Hartt et al., 1999; Kalmar and Van Dyke, 1994) and can be measured very sensitively. In contrast, a PSM variant with scrambled amino acid sequence had no such activity (Figure S1). Since PSMs usually retain their N-terminal formyl-groups during secretion (Wang et al., 2007) we assumed them to be recognized by the human FPR1. Unexpectedly even the non-formylated PSMs, which are produced under certain environmental conditions depending on the varying activity of S. aureus peptide deformylase (Somerville et al., 2003), elicited chemotaxis and Ca2+ ion flux in neutrophils albeit at lower concentrations as the formylated peptides (Figure 1B). Moreover, the FPR1-inhibitory protein CHIPS blocked neutrophil response to the synthetic control ligand fMLF but had no impact on the activity of formylated or non-formylated PSMs (Figure 1C), which was another indication that a receptor different from FPR1 is responsible for the proinflammatory neutrophil responses to PSMs.
Figure 1. Formylated and Non-Formylated PSM Peptides Elicit Chemotaxis and Calcium Ion Fluxes in Human Neutrophils.
(A) PSMα3 stimulates chemotaxis in human neutrophils at nanomolar concentrations with a typical bell-shaped dose/response curve.
(B) Both, formylated and non-formylated core-genome encoded PSMs (PSMα1–4, PSMβ1–2, δ-toxin) and PSM-mec induce chemotaxis and chemotaxis-associated calcium fluxes in human neutrophils (values normalized to 1 µM)
(C) Neutrophil stimulation by PSMs is sensitive to pertussis toxin and the FPR2/ALX-specific inhibitor FLIPr but not the FPR1-specific inhibitor CHIPS. fMLF and MMK1 are synthetic control ligands of FPR1 and FPR2/ALX, respectively.
Data represent means ± SEM of at least three independent experiments. *, P < 0.05; **, P < 0.005; ***, P < 0.001 versus lowest peptide concentration (A), or no inhibition (C). All values were significant versus buffer control in (B) (P < 0.05 or lower). δ, δ-toxin; fl, fluorescence.
PSMs Are Specifically Detected by FPR2/ALX
We confirmed that PSMs are recognized by a G protein-coupled receptor as pertussis toxin (PTX), a general antagonist of this type of receptor blocked the calcium flux-inducing activities of representative formylated or non-formylated PSMs (Figure 1C). Notably, we found that FLIPr, a specific inhibitor of the FPR1 paralog FPR2/ALX leads to almost complete inhibition of neutrophil response to both, the formylated and non-formylated PSMs suggesting that these peptide toxins represent a new class of FPR2/ALX ligands. Control experiments with the synthetic FPR2/ALX ligand MMK1 confirmed the specificity of FLIPr (Figure 1C). Similar results were also obtained when the same set of inhibitors was tested for blocking neutrophil chemotaxis (data not shown).
In order to further evaluate the role of FPR2/ALX in recognizing PSM peptides, we analyzed the response to PSMs of HL60 cells stably transfected with the human FPR1, FPR2/ALX, or FPR3 receptors. While untransfected HL60 cells were unresponsive (data not shown), the FPR1 and FPR2/ALX-transfected cells responded to the corresponding control ligands fMLF and MMK1, respectively, as expected (Figure 2A). All eight formylated PSMs activated FPR2/ALX-transfected cells very efficiently and, depending on the individual peptide, led only to residual activation of FPR1 or FPR3-transfected cells. Similar results were obtained with the corresponding non-formylated PSMs. Dose-response curves of representative PSMs revealed effective concentrations in the low nanomolar range for formylated PSMα3 and δ-toxin (Figure 2B) and the other alpha PSMs (data not shown). pEC50 (negative logarithm of half-maximal effective concentration) values of formylated PSMs reached 8.67 (PSMα3), 7.37 (δ-toxin) or 6.47 (PSMβ2) (Table 1). These values were considerably lower for non-formylated peptides thereby confirming that the formyl groups are not essential but have a considerable impact on PSM activities. Neutrophils revealed similar or somewhat lower potencies for PSMs as FPR2/ALX-transfected HL60. (Figure 2B, Table 1). Thus, formylated PSM alpha peptides are among the most potent naturally occurring FPR2/ALX peptide ligands, which have reported pEC50 values between 4.60 and 9.00 (Ye et al., 2009).
Figure 2. PSMs Specifically Activate and Bind to FPR2/ALX.
(A) PSMs stimulate profound calcium fluxes in FPR2/ALX-transfected but not in FPR1 or FPR3-transfected cells. Untransfected HL60 cells exhibited no significant responses (mean fluorescence values below 1, data not shown). fMLF and MMK1 are synthetic control ligands of FPR1 and FPR2/ALX, respectively.
(B) Dose-response curves for Ca2+ fluxes induced by formylated PSMα3, β2, and δ-toxin in FPR2/ALX-transfected HL60 or neutrophils.
(C) 5-carboxytetramethylrhodamin-labeled PSMs bind specifically to FPR2/ALX-transfected HL60 cells. Maximum detected binding to FPR2/ALX-transfected cells was defined as 100%.
(D) PSMs prevent binding of a FPR2/ALX-specific phycoerythrin-labeled monoclonal antibody (αFPR2) from FPR2/ALX-transfected HL60 cells in a dose-dependent manner. Fluorescence of samples with the phycoerythrin-labeled Isotypecontrol was defined as the maximally achievable inhibition of binding (100% inhibition).
(E) PSMα3 displaces the synthetic 125I-labeld FPR2/ALX ligand WKYMVm from transiently FPR2/ALX-transfected cells. Unlabeled FPR2/ALX ligand CKβ8-1 was used as a positive control
Data represent mean ± SEM of at least three independent experiments. *, P < 0.05; ***, P < 0.001 versus untransfected cells (A, C) or versus no peptide (D). δ, δ-toxin.
Table 1. Potencies of PSM Peptides.
The calcium ion flux-inducing capacities of representative fomylated and non-formylated PSMs are expressed as pEC50 (negative logarithm of half-maximal effective concentration).
PSM peptide | pEC50
* |
|
---|---|---|
FPR2/ALX-transfected HL60 | Human neutrophils | |
PSMα3, formylated | 8.67 ± 0.29 | 7.61 ± 0.06 |
PSMα3, non-formylated | 6.59 ± 0.20 | 5.19 ± 0.24 |
δ-Toxin, formylated | 7.37 ± 0.17 | 7.59 ± 0.05 |
δ-Toxin, non-formylated | 6.32 ± 0.21 | 5.20 ± 0.14 |
PSMβ2, formylated | 6.41 ± 0.36 | 5.61 ± 0.26 |
Data represent means +/− SD of at least three independent experiments. pEC50 values were determined by nonlinear regression analysis of dose-response curves (11–18 points) generated using GraphPad Prism 4. A pEC50 value of non-formylated PSMβ2 could not bedetermined as the peptide reached a cytolytic concentration before the maximal calcium ion flux-inducing capacity was achieved.
Direct binding of representative PSMs to FPR2/ALX was confirmed by three different approaches. (i) PSM peptides C-terminally labeled with the fluorescent dye 5-carboxytetramethylrhodamin revealed only residual binding to untransfected or FPR1-transfected HL60 but bound strongly to FPR2/ALX-transfected cells and neutrophils (Figure 2C, Figure S2). (ii) PSM peptides prevented binding of the FPR2-specific monoclonal antibody #304405 to FPR2-transfected cells (Figure 2D). (iii) PSMα3 displaced the 125I-labelled synthetic FPR2/ALX ligand WKYMVm (Elagoz et al., 2004; Christophe et al., 2001) from FPR2/ALX-transfected cells (Figure 2E). From the latter experiment we calculated pIC50 and Ki values for PSMα3 of 7.3 and 13.3 nM, respectively. Taken together, these data confirm that FPR2/ALX is the major receptor on human neutrophils binding and recognizing S. aureus PSM peptides while FPR1 and FPR3 play only minor roles in recognizing PSM peptides.
FPR2/ALX Does not Affect the Ability of PSMs to Lyse Leukocytes
PSM peptides destroy leukocytes at micromolar concentrations, most probably by membrane disruption (Wang et al., 2007). However, it has remained unknown whether PSMs use a receptor on target cells to exert these cytotoxic activities. When release of cytoplasmic lactate dehydrogenase was monitored as a measure for PSM-mediated cytolysis, there were no notable differences in susceptibility of untransfected or FPR1, FPR2/ALX, or FPR3-transfected HL60 (Figure S3). Moreover, FLIPr had no considerable impact on PSM-mediated disruption of human neutrophils, indicating that PSMs lyse leukocytes independently of FPR2/ALX.
PSMs Induce Ca2+ Influx in Mouse Leukocytes, which Can be Specifically Blocked by an Inhibitor of Human FPR2/ALX or its Mouse Counterpart(s)
MMK1 and PSMs also induced calcium fluxes in mouse neutrophils (Figure 3A), which is in agreement with our previous finding that expression of PSM peptides contributes significantly to neutrophil recruitment by CA-MRSA in a mouse local infection model (Wang et al., 2007). Thus, mice probably also use FPR-like receptor(s) to sense PSM-like peptide toxins. However, mice have a different set of FPR paralogs with seven to eight members and it has remained unclear how many and which of these represent(s) the mouse ortholog(s) of human FPR2/ALX (Migeotte et al., 2006; Ye et al., 2009). FLIPr did not interfere with PSM-mediated activation of mouse neutrophils (data not shown) and is probably highly human-specific as previously described for CHIPS (Prat et al., 2006; de Haas et al., 2004). However, we noted that the synthetic peptide WRW4, an inhibitor of human FPR2/ALX and of at least one mouse FPR paralog (Fpr-rs2) (Karlsson et al., 2006; Onnheim et al., 2008), had a potent antagonistic effect on mouse neutrophil stimulation by MMK1 and PSMs while fMLF-mediated stimulation was not affected (Figure 3A).
Figure 3. In Vitro and In Vivo Inhibition of PSM-Mediated Neutrophil Activation and Mobilization in Mice by WRW4.
(A) WRW4 blocks calcium fluxes in response to MMK1 and PSMs but not to fMLF in mouse neutrophils.
(B) Intraperitoneal challenge of mice with S. aureus USA300 induces granulocyte infiltration, which can be blocked by WRW4. Granulocytes were identified by Gr1+ staining.
(C) PSMα2 leads to leukocyte infiltration into mouse air pouches and this process can be blocked by WRW4 but not by its scrambled congener wwrw3.
Data represent means ± SEM of at least three (A), 5–8 independent experiments with the same number of mice per group (B), or four independent experiments with 2–3 mice per group (C). *, P < 0.05 **, P < 0.005; ***, P < 0.001; ns, not significantly different as calculated by Student’s t-test (A, B) or Mann-Whitney u-test (C).
In order to evaluate the impact of FPR-like receptors on PSM-mediated leukocyte influx in vivo, we used two different mouse models. (i) Injection of PSMs-producing S. aureus USA300 into the mouse peritoneum caused a considerable increase in peritoneal neutrophil numbers (Figure 3B). When WRW4 was injected prior to infection the influx was inhibited indicating that the majority of the leukocyte-recruiting activity was dependent on the mouse counterpart(s) of FPR2/ALX. (ii) The capacity of synthetic PSMs to recruit leukocytes in a mouse air pouch model, which allows to monitor a 1.5 to 3-fold increased leukocyte influx upon local injection of chemoattractants into the air pouch (Perretti et al., 2002; Devosse et al., 2009) was evaluated. Injection of PSMα2, which was the most potent PSM peptide toward mouse neutrophils (data not shown), led to strong influx of leukocytes (Figure 3C). However, when WRW4 was injected into the air pouch prior to PSMα2, the influx of leukocytes was blocked. In contrast, a scrambled variant of WRW4 did not inhibit leukocyte influx into air pouches in response to PSMs (Figure 3C). These data confirm that PSMs are potent neutrophil attractants and that FPR2/ALX or its functional mouse ortholog(s) are crucial for neutrophils to migrate toward PSMs in vitro and in vivo.
FPR2/ALX Mediates very Strong Neutrophil Responses to CA-MRSA but not to HA-MRSA
We have recently described that CHIPS can block most of the chemotactic activity of neutrophils toward culture supernatants from the S. aureus laboratory strain RN4220 demonstrating that formylated peptides represent major neutrophil chemoattractants of this hardly pathogenic strain (Dürr et al., 2006). In contrast, we noted that CHIPS had no significant impact on neutrophil calcium fluxes induced by culture supernatants from USA300 or USA400 (Figure 4A), indicating that a receptor other than FPR1 is responsible for neutrophil recognition of CA-MRSA. Notably, CA-MRSA supernatants had about hundred-fold stronger proinflammatory activities compared to the HA-MRSA strains COL, Mu50, and N315, which corresponds to the extraordinary virulence of CA-MRSA (Wang et al., 2007) (Figure 4C). The potent neutrophil activation by CA-MRSA was clearly mediated by PSMs as deletion of the psmα gene cluster encoding the four most potent PSMs that together constitute the vast majority of the proinflammatory PSM activity produced by CA-MRSA (Wang et al., 2007) abrogated most of the neutrophil response (Figure 5A,B). Production of the remaining PSMs in this mutant has been shown to be unaltered (Wang et al., 2007; Queck et al., 2008) but their comparatively low expression and activity levels appears to be the reason for their marginal contribution to the FPR2/ALX-dependent stimulatory capacity of CA-MRSA. Moreover, the strong calcium flux-inducing capacity of CA-MRSA was almost entirely mediated by FPR2/ALX as FLIPr inhibited this activity completely and FPR2/ALX-transfected HL60 responded magnitudes higher than FPR1-transfected HL60 to CA-MRSA (Figure 5A–C). In contrast, the two cell lines responded with similar efficiency to supernatants from the HA-MRSA strains COL, Mu50, and N315, albeit at moderate levels. USA300-induced chemotaxis and IL-8 secretion by neutophils were also largely FPR2/ALX-dependent (Figure 5D). Collectively, these date indicate that FPR2/ALX has a critical, unrecognized role in responding to PSM-like peptide toxins and that leukocytes use PSMs as potent proinflammatory signals from CA-MRSA.
Figure 4. FPR2/ALX Initiates Exuberant Proinflammatory Neutrophil Responses to PSMs in CA-MRSA Culture Supernatants.
Calcium flux stimulated by CA-MRSA culture supernatants is FPR2/ALX-dependent in neutrophils (A) and receptor-transfected HL60 cells (B). Untransfected HL60 cells exhibited no significant responses (mean fluorescence values below 1, data not shown).
(C) FPR2/ALX-transfected HL60 cells respond exuberantly to CA-MRSA (USA300, USA400) but only moderately to HA-MRSA (COL, Mu50, N315) while FPR1-mediated responses differ only slightly between CA-MRSA and HA-MRSA.
(D) FLIPr inhibits USA300-stimulated neutrophil chemotaxis and IL-8 release. The indicated S. aureus wild-type (WT) or PSM gene deletion mutants (Δα, Δβ, or Δδ) were used in A–D. USA400 supernatants yielded similar results as shown for USA300 in (A, B, D) (data not shown). Data represent means ± SEM of at least three independent experiments. **, P < 0.005; ***, P < 0.001 versus no inhibition (A, D) or untransfected HL60 (B).
Figure 5. Proposed Role of FPR2/ALX-Mediated Sensing of PSM Peptide Toxins.
FPR2/ALX allows neutrophils to distinguish between S. aureus with low and high virulence potential by sensing the concentration of PSMs and causes neutrophil infiltration in response to highly pathogenic S. aureus. In contrast, FPR1 mediates moderate neutrophil activation in response to S. aureus irrespective of virulence. CHIPS and FLIPr are thought to prevent neutrophil influx during certain stages of human colonization and infection when it is most critical for the bacteria to prevent recruitment of leukocytes (de Haas et al., 2004; Prat et al., 2006).
DISCUSSION
While the FPR1 receptor recognizes a defined microbial molecular pattern, an N-formylated methionine followed by hydrophobic amino acids (Fu et al., 2006), it remains unclear, which structural features define an FPR2/ALX ligand. We aligned the S. aureus PSM peptides with each other and with further known FPR2/ALX ligands but were unable to detect a consistently conserved sequence motif (data not shown) suggesting that distinct properties in the secondary structure define the affinity of FPR2/ALX ligands. While the α-helical amphipathic structure of PSMs has been noted and may play a role in recognition by FPR2/ALX (Wang et al., 2007), more sophisticated techniques such as three-dimensional NMR will be necessary to elucidate common features of FPR2/ALX ligands. Of note, difficulties in defining common features in a diverse set of ligands are reminiscent of other pattern recognition receptors such as the Toll-like receptors (Uematsu and Akira, 2006).
While most of the currently known PAMPs are shared by microbial commensals and pathogens, recognition of PSMs by FPR2/ALX offers the possibility for the innate immune system to distinguish between commensal and highly pathogenic microbes and adjust host responses appropriately (Figure 6). While non-peptide PAMPs such as most TLR ligands are largely invariant, peptidic PAMPs such as the PSMs can probably be altered more rapidly in structure. Accordingly, TLR receptors exhibit only moderate interspecies changes (Leulier and Lemaitre, 2008), while FPR-like receptors vary widely between mammalian species with respect to paralog numbers and ligand specificities (Gao et al., 1998) indicating a particularly strong evolutionary pressure on this type of receptor. This notion is further supported by the fact that primate homologs of FPR2/ALX show the highest inter-species differences in the ligand-binding extracellular loops suggesting positive selection of ligand recognition capacities (Alvarez et al., 1996). Moreover, mice, cows, pigs, dogs, and cats hardly respond to fMLF, the most potent ligand of the human FPR1 (Styrt, 1989) while mice prefer formylated peptides with other sequences (Southgate et al., 2008).
As another result of ongoing coevolution, many bacterial pathogens exhibit remarkable host specificity with highly human-specific virulence factors such as the chemotaxis inhibitory S. aureus proteins CHIPS and FLIPr, which are only functional on FPR1 and FPR2/ALX, respectively, of human leukocytes (de Haas et al., 2004; Prat et al., 2006). S. aureus appears to employ these inhibitors for preventing the influx of neutrophils during certain stages of human colonization and infection when it is most critical for the bacteria to prevent recruitment of leukocytes (de Haas et al., 2004; Prat et al., 2006) (Figure 5). Of note, CA-MRSA have been shown to express the FLIPr gene, albeit under different conditions as the PSM genes. PSM but not FLIPr production is tightly controlled by the agr quorum sensing system while FLIPr is expressed upon contact with neutrophil granule contents (Palazzolo-Ballance et al., 2008; Queck et al., 2008). agr turnes PSM expression largely off at early stages of infection when it may be advantageous for the bacteria to remain unrecognized by the innate immune system and FLIPr may help to inhibit the activity or residual PSM amounts produced even with inactive agr. S. aureus strains involved in chronic infection often disable the agr system and thereby PSM production (Shopsin et al., 2008) while CA-MRSA strains are often regarded as less well adapted new lineages with dysregulated, excessive toxin production. In concert with other toxins, PSM peptides appear to be so effective in destroying neutrophils with concomitant tissue damage that the active recruitment of neutrophils via FPR2/ALX can even provide an advantage for these emerging pathogens (Diep and Otto, 2008). Accordingly, CA-MRSA infections are notorious for extensive tissue destruction e.g. in necrotizing pneumonia, necrotizing fasciitis, and sepsis with Waterhouse-Friedrichsen syndrome (Chambers and DeLeo, 2009; Kahl and Peters, 2007). The regulation of peptide deformylase activity, which leads to deformylated PSM peptides with reduced capacity to stimulate FPR2/ALX in response to bacterial growth phase and iron availability (Somerville et al., 2003) may contribute to the ability of S. aureus to adjust a favourable balance between neutrophil attraction and lysis.
The implication of FPR2/ALX in both, severe bacterial infections and chronic degenerative diseases contributes to the notion that such virtually unrelated pathological processes may be more closely connected than previously anticipated (Karin et al., 2006). Future studies will show in which types of disease FPR2/ALX is beneficial or may even contribute to exacerbation. A detailed understanding of FPR2/ALX ligands and consequences of FPR2/ALX activation will help to develop new strategies for interfering with CA-MRSA infection and other major human diseases.
EXPERIMENTAL PROCEDURES
Synthetic Peptides
PSM peptides with the recently published sequences (Wang et al., 2007; Queck et al., 2009), the FPR2/ALX-specific control ligand MMK1 (LESIFRSLLFRVM-NH2) (Fu et al., 2006), and the FPR2/ALX inhibitor WRW4 (WRWWWW-NH2) (Karlsson et al., 2006; Onnheim et al., 2008) were synthesized (EMC Microcollections). PSM-mec was synthesized by American Peptide Company, Inc. (Sunnyvale, CA). PSMα1 (MGIIAGIIKVIKSLIEQFTGK), PSMα2 (MGIIAGIIKFIKGLIEKFTGK), PSMα3 (MEFVAKLFKFFKDLLGKFLGNN), PSMα4 (MAIVGTIIKIIKAIIDIFAK), PSMβ1 (MEGLFNAIKDTVTAAINNDGAKLGTSIVSIVENGVGLLGKLFGF), PSMβ2 (MTGLAEAIANTVQAAQQHDSVKLGTSIVDIANGVGLLGKLFGF), δ-toxin (MAQDIISTISDLVKWIIDTVNKFTKK), and PSM-mec (MDFTGVITSIIDLIKTCIQAFG) were synthesized in their formylated and non-formylated form. Some peptides were C-terminally modified with 5-carboxytetramethylrhodamin. fMLF was purchased from Sigma. Scrambled variant of WRW4 with all-D amino acids (wwrwww) and of alpha PSM with retrograde amino acid sequence (KAFIDIIAKIIKIITGVIAM) were obtained from EMC Microcollections.
Bacteria and Cell Lines
USA300 and USA400 or COL, Mu50, and N315 are prevalent CA-MRSA or HA-MRSA strains, respectively (Diep and Otto, 2008). Defined PSM mutants have recently been described in detail (Wang et al., 2007). Bacterial culture supernatants were obtained by centrifugation of overnight cultures grown in tryptic soy broth (TSB) and filtered through 0.2-µm pore size filters. No chemotactic or stimulatory activity was detected in non-inoculated medium at relevant concentrations.
HL60 cells stably transfected with human FPR1, FPR2/ALX, and FPR3 have been described recently (Christophe et al., 2001; Dahlgren et al., 2000). These cells were grown in RPMI medium (Biochrom) supplemented with 10% FCS (Sigma-Aldrich), 20 mM Hepes (Biochrom), penicillin (100 units/ml), streptomycin (100 µg/ml) (Gibco), and 1 × Glutamax (Gibco). Transfected cells were grown in the presence of G418 (Biochrom) at a final concentration of 1 mg/ml.
Neutrophil Chemotaxis
Human neutrophils were isolated by standard Ficoll/Histopaque gradient centrifugation (Dürr et al., 2006). Chemotaxis of neutrophils towards staphylococcal supernatants or synthetic peptides was determined by using fluorescence-labeled neutrophils that migrated through a 3-µm-pore size polycarbonate trans-well filter as described recently (Dürr et al., 2006). Synthetic chemoattractants were used at concentrations in the linear range of the dose/response curves. Initial control experiments were performed to verify that PSM peptides mobilize neutrophils by chemotactic rather than by undirected chemokinetic stimulation. Neutrophil migration from the upper to the lower trans-well chamber was only observed when the peptides were added to the lower chamber. When an even PSM concentration was present in the two chambers no such migration was observed therby confirming that PSMs induce in fact chemotactic migration in neutrophils. USA300 culture supernatants were used at 1% dilution. The relative fluorescence measured was corrected for the buffer control (only buffer added to lower compartment) and divided through thousand.
Measurement of Calcium Ion Fluxes in Human Neutrophils and HL60 Cells
Calcium fluxes were monitored in many experiments as a surrogate marker for chemotaxis since it can be measured more robustly and accurately than chemotaxis. Calcium fluxes were analyzed by stimulating cells loaded with Fluo-3-AM (Molecular Probes) and monitoring fluorescence with a FACSCalibur flow cytometer (Becton Dickinson) as described recently (de Haas et al., 2004). For EC50 determination, Ca2+ flux measurements were carried out at 37°C. In order to study the influence of pertussis toxin (PTX), cells were preincubated with 1 µg/ml PTX (List Biological Laboratories) for three hours at 37°C under 5% CO 2. For measuring the influence of CHIPS or FLIPr, 1 × 106 cells/ml were preincubated with CHIPS or FLIPr at final concentrations of 1.4 µg/ml or 0.5 µg/ml, respectively, for 20 min at room temperature under agitation. The CHIPS and FLIPr proteins were prepared as described recently (de Haas et al., 2004; Prat et al., 2006). Synthetic chemoattractants were used at concentrations in the linear range of the dose-response curves. In order to stimulate neutrophils, peptides were used at final concentrations of 250 nM, 2 µM for formylated PSMsα3, β1, or 1 µM for δ–toxin and PSM-mec, respectively; 10 µM for non-formylated PSMα3; and 10 nM or 50 nM for fMLF or MMK1, respectively. HL60 cells were stimulated with final peptide concentrations of 62.5 nM (formylated PSMα3), 125 nM (formylated PSMα2), 250 nM (formylated δ–toxin, PSM-mec), 500 nM (formylated PSMsα1, α4, β1, β2), or 2 µM (all non-formylated PSMs). fMLF and MMK1 were used for HL60 cells at final concentrations of 20 nM and 10 nM, respectively. USA300 culture supernatants were used at 0.047% dilution unless otherwise noted. Measurements of 2,000 events were performed and calcium flux was expressed as relative fluorescence corrected for buffer controls.
Peptide Binding and Displacement Assays
105 HL60 cells or 5 × 105 PMN were washed with PBS containing 0.1% HSA and incubated with increasing concentrations of 5-carboxytetramethylrhodamin-labeled peptides for 30 min (neutrophils) or 2 h (HL60) at 37°C under agitation. Cells were washed twice with ice-cold PBS containing 0.1% HSA and cell-associated fluorescence was measured using a FACSCalibur flow cytometer.
For displacement experiments with phycoerythrin-labeled monoclonal mouse anti-human FPR2/ALX antibody Clone #304405 (R and D Systems) FPR2/ALX-transfected HL60 cells were first blocked 15 min at room temperature with 1% HSA in PBS. Then, 1 × 105 cells were washed with PBS, sedimented by centrifugation, resuspended in PBS containing increasing concentrations of PSMα2, PSMα3 or δ-toxin, and incubated for 15 min at 300 rpm and 37°C. Then either anti-FPR2/ALX antibody or an appropriate isotype control (R and D Systems) was added and cells were incubated for further 45 min under agitation on ice. After washing with 200 µl PBS fluorescence was measured using a FACSCalibur flow cytometer. Control experiments confirmed that this antibody does not bind to untransfected or FPR1-transfected HL60 (data not shown).
The radioactively-labeld ligand competition assay was performed by MDS Pharma Services (Taipei, Taiwan) as described recently (Elagoz et al., 2004; Christophe et al., 2001). Briefly, purified membranes of CHO cells transiently transfected with FPR2/ALX were incubated at room temperature for 90 min with 0.025 nM of the synthetic 125I-labeled FPR2/ALX ligand WKYMVm in the absence or presence of increasing concentrations of either formylated PSMα3 or of the established FPR2/ALX control ligand peptide CKβ8-1 (Elagoz et al., 2004). Unbound tracer was washed and bound label was counted using a TopCount microplate scintillation and luminescence counter (PerkinElmer Life and Analytical Sciences, Waltham, MA).
Mouse Experiments
Mouse leukocytes were obtained from the peritoneum of 12-week old female CD1 mice (Charles River) upon inducing an intraperitoneal inflammatory reaction by repeated casein injection as described previously (Dürr et al., 2006). Calcium fluxes in mouse neutrophils were determined as described above for human neutrophils with final concentrations of 250, 12.5 and 50 nM of formylated PSMα3, α2, and δ–toxin, respectively, or 500 nM of fMLF and MMK1.
The mouse air pouch model was conducted essentially as described elsewhere (Perretti et al., 2002; Devosse et al., 2009). Briefly, air pouches were raised on the dorsum of 6 to 8-week old female BALB/c mice (Harlan Winkelmann) by injection of 5 ml (day one) and 3 ml (day 4) sterile air. The test peptides were dissolved in DMSO at 100 mM (WRW4 and the scrambled variant wwrw3), and 10 mM (PSMα2) and then diluted with PBS. On day seven 250 µl PBS with 0.27 mM WRW4 or wwrw3 (corresponding to ca. 7.5 mg/kg body weight) or the corresponding amount of DMSO were injected into the air pouch. After 15 min the mice received a second injection into the air pouch of 750 µl PBS with 27 µM PSMα2 or the corresponding amount of DMSO. Four hours later mice were sacrificed, air pouches were rinsed with 2 ml of ice-cold PBS, and leukocytes were counted.
In the mouse peritonitis model WRW4 at 7.5 mg/kg body weight or the vehicle control (2% DMSO) in 300 µl PBS was injected into the peritoneum of 6–8 weeks-old female BALB/c mice. 20 min later 300 µl PBS or PBS containing 2 × 107 live S. aureus USA300 were intraperitoneally injected. Two hours after infection the mice were euthanized with CO2. Subsequently, peritoneal exudates were collected and leukocytes were stained and counted as described previously (Wang et al., 2007). FITC-conjugated antibody to mouse Gr1+ and a corresponding isotype control was purchased from BD Bioscience. Samples were analyzed on a FACSCalibur flow cytometer. Initial control experiments showed that application of WRW4 prior to infection had no impact on numbers of life bacteria after the two-hours infection period.
Animal experiments were performed according to German law with permission of the responsible authorities (Regierungspräsidium Tübingen)
IL-8 Production and Cell Lysis
Human IL-8 was measured using an ELISA kit (R&D Systems) according to the manufacturer’s instructions. Lysis of neutrophils and HL60 cells by PSM peptides was determined by measuring the release of cytoplasmic lactate dehydrogenase (Cytotoxicity Detection Kit, Roche) as described recently (Wang et al., 2007).
Statistical Methods
Statistical analyses were performed with the Prism 4.0 package (GraphPad Software) and the between-group differences were analyzed for significance with the two-tailed Student’s t-test unless otherwise noted.
Supplementary Material
HIGHLIGHTS.
-
-
PSMs attract human neutrophils in the nano- to micromolar range; non-formylated PSMs have similar activities as formylated forms albeit with much less potency
-
-
PSMs are sensed by the human FPR2/ALX receptor whose potential role in innate immunity has remained elusive before
-
-
FPR2/ALX mediates strong neutrophil responses to CA-MRSA but not to S. aureus with low pathogenicity
ACKNOWLEDGMENTS
We thank Manuela Dürr for helpful discussion and Nele Nikola for excellent technical help. Our research is supported by grants from the German Research Foundation (SFB685, GRK685, TR34, SFB766, SPP1130), the German Ministry of Education and Research (NGFN2, SkinStaph), and the IZKF program of the Medical Faculty, University of Tübingen, to AP, from the European Union (LSHM-CT-2004-512093) to J.v.S. and A.P., from the Intramural Program of the National Institutes of Allergy and Infectious Diseases, U.S. National Institutes of Health, to M.O., and from the CEA, the CNRS, and the University Joseph-Fourier to M.R. and F.B. The authors have no conflicting financial interests.
REFERENCES
- Alvarez V, Coto E, Setien F, Gonzalez-Roces S, Lopez-Larrea C. Molecular evolution of the N-formyl peptide and C5a receptors in non-human primates. Immunogenetics. 1996;44:446–452. doi: 10.1007/BF02602806. [DOI] [PubMed] [Google Scholar]
- Bubeck WJ, Bae T, Otto M, DeLeo FR, Schneewind O. Poring over pores: alpha-hemolysin and Panton-Valentine leukocidin in Staphylococcus aureus pneumonia. Nat. Med. 2007;13:1405–1406. doi: 10.1038/nm1207-1405. [DOI] [PubMed] [Google Scholar]
- Chambers HF, DeLeo FR. Waves of resistance:Staphylococcus aureus in the antibiotic era. Nat. Rev. Microbiol. 2009;7:629–641. doi: 10.1038/nrmicro2200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christophe T, Karlsson A, Dugave C, Rabiet MJ, Boulay F, Dahlgren C. The synthetic peptide Trp-Lys-Tyr-Met-Val-Met-NH2 specifically activates neutrophils through FPRL1/lipoxin A4 receptors and is an agonist for the orphan monocyte-expressed chemoattractant receptor FPRL2. J. Biol. Chem. 2001;276:21585–21593. doi: 10.1074/jbc.M007769200. [DOI] [PubMed] [Google Scholar]
- Cui Y, Le Y, Yazawa H, Gong W, Wang JM. Potential role of the formyl peptide receptor-like 1 (FPRL1) in inflammatory aspects of Alzheimer's disease. J. Leukoc. Biol. 2002;72:628–635. [PubMed] [Google Scholar]
- Dahlgren C, Christophe T, Boulay F, Madianos PN, Rabiet MJ, Karlsson A. The synthetic chemoattractant Trp-Lys-Tyr-Met-Val-DMet activates neutrophils preferentially through the lipoxin A(4) receptor. Blood. 2000;95:1810–1818. [PubMed] [Google Scholar]
- de Haas CJ, Veldkamp KE, Peschel A, Weerkamp F, van Wamel WJ, Heezius EC, Poppelier MJ, Van Kessel KP, Van Strijp JA. Chemotaxis inhibitory protein of Staphylococcus aureus a bacterial antiinflammatory agent. J Exp. Med. 2004;199:687–695. doi: 10.1084/jem.20031636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Devosse T, Guillabert A, D'Haene N, Berton A, De NP, Noel S, Brait M, Franssen JD, Sozzani S, Salmon I, Parmentier M. Formyl peptide receptor-like 2 is expressed and functional in plasmacytoid dendritic cells, tissue-specific macrophage subpopulations, and eosinophils. J. Immunol. 2009;182:4974–4984. doi: 10.4049/jimmunol.0803128. [DOI] [PubMed] [Google Scholar]
- Diep BA, Otto M. The role of virulence determinants in community-associated MRSA pathogenesis. Trends Microbiol. 2008;16:361–369. doi: 10.1016/j.tim.2008.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diep BA, Palazzolo-Ballance AM, Tattevin P, Basuino L, Braughton KR, Whitney AR, Chen L, Kreiswirth BN, Otto M, DeLeo FR, Chambers HF. 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]
- Dürr MC, Kristian SA, Otto M, Matteoli G, Margolis PS, Trias J, Van Kessel KP, Van Strijp JA, Bohn E, Landmann R, Peschel A. Neutrophil chemotaxis by pathogen-associated molecular patterns--formylated peptides are crucial but not the sole neutrophil attractants produced by Staphylococcus aureus. Cell Microbiol. 2006;8:207–217. doi: 10.1111/j.1462-5822.2005.00610.x. [DOI] [PubMed] [Google Scholar]
- Elagoz A, Henderson D, Babu PS, Salter S, Grahames C, Bowers L, Roy MO, Laplante P, Grazzini E, Ahmad S, Lembo PM. A truncated form of CKbeta8-1 is a potent agonist for human formyl peptide-receptor-like 1 receptor. Br. J. Pharmacol. 2004;141:37–46. doi: 10.1038/sj.bjp.0705592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foster TJ. The Staphylococcus aureus "superbug". J Clin. Invest. 2004;114:1693–1696. doi: 10.1172/JCI23825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fu H, Karlsson J, Bylund J, Movitz C, Karlsson A, Dahlgren C. Ligand recognition and activation of formyl peptide receptors in neutrophils. J. Leukoc. Biol. 2006;79:247–256. doi: 10.1189/jlb.0905498. [DOI] [PubMed] [Google Scholar]
- Gao JL, Chen H, Filie JD, Kozak CA, Murphy PM. Differential expansion of the N-formylpeptide receptor gene cluster in human and mouse. Genomics. 1998;51:270–276. doi: 10.1006/geno.1998.5376. [DOI] [PubMed] [Google Scholar]
- Haas PJ, de Haas CJ, Kleibeuker W, Poppelier MJ, Van Kessel KP, Kruijtzer JA, Liskamp RM, Van Strijp JA. N-terminal residues of the chemotaxis inhibitory protein of Staphylococcus aureus are essential for blocking formylated peptide receptor but not C5a receptor. J. Immunol. 2004;173:5704–5711. doi: 10.4049/jimmunol.173.9.5704. [DOI] [PubMed] [Google Scholar]
- Hartt JK, Barish G, Murphy PM, Gao JL. N-formylpeptides induce two distinct concentration optima for mouse neutrophil chemotaxis by differential interaction with two N-formylpeptide receptor (FPR) subtypes. Molecular characterization of FPR2, a second mouse neutrophil FPR. J. Exp. Med. 1999;190:741–747. doi: 10.1084/jem.190.5.741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hecht I, Rong J, Sampaio AL, Hermesh C, Rutledge C, Shemesh R, Toporik A, Beiman M, Dassa L, Niv H, Cojocaru G, Zauberman A, Rotman G, Perretti M, Vinten-Johansen J, Cohen Y. A novel peptide agonist of formyl-peptide receptor-like 1 (ALX) displays anti-inflammatory and cardioprotective effects. J. Pharmacol. Exp. Ther. 2009;328:426–434. doi: 10.1124/jpet.108.145821. [DOI] [PubMed] [Google Scholar]
- Kahl BC, Peters G. Microbiology. Mayhem in the lung. Science. 2007;315:1082–1083. doi: 10.1126/science.1139628. [DOI] [PubMed] [Google Scholar]
- Kalmar JR, Van Dyke TE. Effect of bacterial products on neutrophil chemotaxis. Methods Enzymol. 1994;236:58–87. doi: 10.1016/0076-6879(94)36009-x. [DOI] [PubMed] [Google Scholar]
- Karin M, Lawrence T, Nizet V. Innate immunity gone awry: linking microbial infections to chronic inflammation and cancer. Cell. 2006;124:823–835. doi: 10.1016/j.cell.2006.02.016. [DOI] [PubMed] [Google Scholar]
- Karlsson J, Fu H, Boulay F, Bylund J, Dahlgren C. The peptide Trp-Lys-Tyr-Met-Val-D-Met activates neutrophils through the formyl peptide receptor only when signaling through the formylpeptide receptor like 1 is blocked. A receptor switch with implications for signal transduction studies with inhibitors and receptor antagonists. Biochem. Pharmacol. 2006;71:1488–1496. doi: 10.1016/j.bcp.2006.02.010. [DOI] [PubMed] [Google Scholar]
- Le Y, Murphy PM, Wang JM. Formyl-peptide receptors revisited. Trends Immunol. 2002;23:541–548. doi: 10.1016/s1471-4906(02)02316-5. [DOI] [PubMed] [Google Scholar]
- Leulier F, Lemaitre B. Toll-like receptors--taking an evolutionary approach. Nat. Rev. Genet. 2008;9:165–178. doi: 10.1038/nrg2303. [DOI] [PubMed] [Google Scholar]
- Mader D, Rabiet MJ, Boulay F, Peschel A. Formyl peptide receptor-mediated proinflammatory consequences of peptide deformylase inhibition in Staphylococcus aureus. Microbes. Infect. 2010;12:415–419. doi: 10.1016/j.micinf.2010.01.014. 2010. [DOI] [PubMed] [Google Scholar]
- Migeotte I, Communi D, Parmentier M. Formyl peptide receptors: a promiscuous subfamily of G protein-coupled receptors controlling immune responses. Cytokine Growth Factor Rev. 2006;17:501–519. doi: 10.1016/j.cytogfr.2006.09.009. [DOI] [PubMed] [Google Scholar]
- Migeotte I, Riboldi E, Franssen JD, Gregoire F, Loison C, Wittamer V, Detheux M, Robberecht P, Costagliola S, Vassart G, Sozzani S, Parmentier M, Communi D. Identification and characterization of an endogenous chemotactic ligand specific for FPRL2. J. Exp. Med. 2005;201:83–93. doi: 10.1084/jem.20041277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murphy PM, Ozcelik T, Kenney RT, Tiffany HL, McDermott D, Francke U. A structural homologue of the N-formyl peptide receptor. Characterization and chromosome mapping of a peptide chemoattractant receptor family. J. Biol. Chem. 1992;267:7637–7643. [PubMed] [Google Scholar]
- Onnheim K, Bylund J, Boulay F, Dahlgren C, Forsman H. Tumour necrosis factor (TNF)-alpha primes murine neutrophils when triggered via formyl peptide receptor-related sequence 2, the murine orthologue of human formyl peptide receptor-like 1, through a process involving the type I TNF receptor and subcellular granule mobilization. Immunology. 2008;125:591–600. doi: 10.1111/j.1365-2567.2008.02873.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palazzolo-Ballance AM, Reniere ML, Braughton KR, Sturdevant DE, Otto M, Kreiswirth BN, Skaar EP, DeLeo FR. Neutrophil microbicides induce a pathogen survival response in community-associated methicillin-resistant Staphylococcus aureus. J. Immunol. 2008;180:500–509. doi: 10.4049/jimmunol.180.1.500. [DOI] [PubMed] [Google Scholar]
- Perretti M, Chiang N, La M, Fierro IM, Marullo S, Getting SJ, Solito E, Serhan CN. Endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor. Nat. Med. 2002;8:1296–1302. doi: 10.1038/nm786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prat C, Bestebroer J, de Haas CJ, Van Strijp JA, Van Kessel KP. A new staphylococcal anti-inflammatory protein that antagonizes the formyl peptide receptor-like 1. J. Immunol. 2006;177:8017–8026. doi: 10.4049/jimmunol.177.11.8017. [DOI] [PubMed] [Google Scholar]
- Queck SY, Jameson-Lee M, Villaruz AE, Bach TH, Khan BA, Sturdevant DE, Ricklefs SM, 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. doi: 10.1016/j.molcel.2008.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Queck SY, Khan BA, Wang R, Bach TH, Kretschmer D, Chen L, Kreiswirth BN, Peschel A, DeLeo FR, Otto M. Mobile genetic element-encoded cytolysin connects virulence to methicillin resistance in MRSA. PLoS. Pathog. 2009;5:e1000533. doi: 10.1371/journal.ppat.1000533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Said-Salim B, Mathema B, Kreiswirth BN. Community-acquired methicillin-resistant Staphylococcus aureus: an emerging pathogen. Infect. Control Hosp. Epidemiol. 2003;24:451–455. doi: 10.1086/502231. [DOI] [PubMed] [Google Scholar]
- Shopsin B, Drlica-Wagner A, Mathema B, Adhikari RP, Kreiswirth BN, Novick RP. Prevalence of agr dysfunction among colonizing Staphylococcus aureus strains. J. Infect. Dis. 2008;198:1171–1174. doi: 10.1086/592051. [DOI] [PubMed] [Google Scholar]
- Somerville GA, Cockayne A, Dürr M, Peschel A, Otto M, Musser JM. Synthesis and deformylation of Staphylococcus aureus delta-toxin are linked to tricarboxylic acid cycle activity. J. Bacteriol. 2003;185:6686–6694. doi: 10.1128/JB.185.22.6686-6694.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Southgate EL, He RL, Gao JL, Murphy PM, Nanamori M, Ye RD. Identification of formyl peptides from Listeria monocytogenes and Staphylococcus aureus as potent chemoattractants for mouse neutrophils. J. Immunol. 2008;181:1429–1437. doi: 10.4049/jimmunol.181.2.1429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Styrt B. Species variation in neutrophil biochemistry and function. J. Leukoc. Biol. 1989;46:63–74. doi: 10.1002/jlb.46.1.63. [DOI] [PubMed] [Google Scholar]
- Su SB, Gong W, Gao JL, Shen W, Murphy PM, Oppenheim JJ, Wang JM. A seven-transmembrane, G protein-coupled receptor, FPRL1, mediates the chemotactic activity of serum amyloid A for human phagocytic cells. J. Exp. Med. 1999;189:395–402. doi: 10.1084/jem.189.2.395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uematsu S, Akira S. Toll-like receptors and innate immunity. J. Mol. Med. 2006;84:712–725. doi: 10.1007/s00109-006-0084-y. [DOI] [PubMed] [Google Scholar]
- Wang R, Braughton KR, Kretschmer D, Bach TH, Queck SY, Li M, Kennedy AD, Dorward DW, Klebanoff SJ, Peschel A, DeLeo FR, Otto M. Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat. Med. 2007;13:1510–1514. doi: 10.1038/nm1656. [DOI] [PubMed] [Google Scholar]
- Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM. International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol. Rev. 2009;61:119–161. doi: 10.1124/pr.109.001578. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.