Skip to main content
Infection and Immunity logoLink to Infection and Immunity
. 2015 Aug 12;83(9):3428–3437. doi: 10.1128/IAI.00401-15

Staphlyococcus aureus Phenol-Soluble Modulins Stimulate the Release of Proinflammatory Cytokines from Keratinocytes and Are Required for Induction of Skin Inflammation

Adnan K Syed a,b, Tamra J Reed c, Kaitlyn L Clark c, Blaise R Boles b, J Michelle Kahlenberg c,
Editor: A Camilli
PMCID: PMC4534673  PMID: 26077761

Abstract

Staphylococcus aureus is a human commensal that colonizes the skin. While it is normally innocuous, it has strong associations with atopic dermatitis pathogenesis and has become the leading cause of skin and soft tissue infections in the United States. The factors that dictate the role of S. aureus in disease are still being determined. In this work, we utilized primary keratinocyte culture and an epidermal murine colonization model to investigate the role of S. aureus phenol-soluble modulins (PSMs) in proinflammatory cytokine release and inflammation induction. We demonstrated that many species of Staphylococcus are capable of causing release of interleukin 18 (IL-18) from keratinocytes and that S. aureus PSMs are necessary and sufficient to stimulate IL-18 release from keratinocytes independently of caspase 1. Further, after 7 days of epicutaneous exposure to wild-type S. aureus, but not S. aureus Δpsm, we saw dramatic changes in gross pathology, as well as systemic release of proinflammatory cytokines. This work demonstrates the importance of PSM peptides in S. aureus-mediated inflammatory cytokine release from keratinocytes in vitro and in vivo and further implicates PSMs as important contributors to pathogenesis.

INTRODUCTION

Staphylococcus aureus is a human commensal that lives in the nose, skin, and throat in approximately 30% of the human population (1, 2). While S. aureus is usually harmless, it is an opportunistic pathogen that has become a leading cause of nosocomial infections in the United States (3) and can manifest as skin and soft tissue infections, infective endocarditis, osteomyelitis, and sepsis (1). Further, it has a role in exacerbation of atopic dermatitis (AD), a chronic inflammatory disease of the skin that affects up to 20% of children and up to 3% of adults (4, 5). Chronic cutaneous inflammation during AD results in increased production of extracellular matrix components that permit attachment of S. aureus (6). Subsequently, S. aureus promotes AD by stimulating a strong proinflammatory response (7). S. aureus proteins, such as hemolysin α (HLA), staphylococcal protein A (SPA), and lipoteichoic acids (LTA), promote proinflammatory cytokine production from keratinocytes (711). However, this stimulation requires the concurrent presence of a surfactant, such as SDS (11).

Keratinocytes serve as the first line of defense against cutaneous pathogens and are able to stimulate an immune response by releasing cytokines, defensins, and antimicrobial cationic peptides, such as LL-37, to fight off pathogens (12). One cytokine that is produced by keratinocytes and has a role in promoting AD is interleukin 18 (IL-18) (13, 14). IL-18 is a proinflammatory cytokine that is cleaved and activated primarily by caspase 1 (15). Caspase 1 activation occurs following exposure to danger-associated molecular patterns (DAMPs) or pathogen-associated molecular patterns (PAMPs), which stimulate the activation of the inflammasome (16). Caspase 1 activation can also result in the activation of the proinflammatory cytokine IL-1β (17). These mature cytokines are then released from the cell and have varied effects, such as leukocyte recruitment, promotion of T and B cell activation, and upregulation of other inflammatory cytokines (18).

The phenol-soluble modulins (PSMs) of S. aureus have been shown to have many characteristics, including the ability to recruit leukocytes, lyse erythrocytes and neutrophils, act as antimicrobial peptides, modulate biofilm development, and exhibit surfactant-like properties (1928). PSMs are small peptides ranging from 22 to 44 amino acids in length that are predicted to form amphipathic helices and in some cases can form amyloid fibers to abrogate their toxic properties and stabilize biofilms (19, 23). Several staphylococcal species have been reported to produce PSM proteins, including S. aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis (19, 29, 30).

Here, we set out to investigate staphylococcal factors that contribute to the release of proinflammatory cytokines from keratinocytes. We found that some, but not all, species of staphylococci examined produce compounds that are able to cause the lytic release of active IL-18 from human keratinocytes independently of caspase 1 activation. We also showed that PSMs are necessary and sufficient for this release. Further, we utilized a mouse model of cutaneous colonization (31) to demonstrate that neutrophil recruitment and systemic inflammatory response are dependent on S. aureus PSMs.

MATERIALS AND METHODS

Bacterial strains and growth conditions.

The bacteria used in this study are summarized in Table 1. S. aureus strain LAC (32) was used as the wild-type (WT) strain for the study. To create a Δpsm deletion strain, the PSMα and PSMβ operons were deleted using allelic replacement, as previously described (23), and the start codon of δ-toxin was mutated to prevent translation without affecting the function of RNAIII, as was done previously (19). Genetic complementation of the PSMα and PSMβ operons in the Δpsm strain was done as previously described using pALC2073-PSMα and pRS-PSMβ (23). Bacterial supernatants were collected after 24 h of growth in tryptic soy broth (TSB) (MP Biosciences, Santa Ana, CA) at 37°C with orbital shaking at 200 rpm. The cells were centrifuged at 7,000 × g for 10 min, followed by filter sterilization of the supernatants through 0.22-μm filters. The supernatants were stored at −20°C until use. Bacteria used for colonization were grown overnight in TSB at 37°C with orbital shaking at 200 rpm. Before use, they were diluted 1:500 in fresh TSB and grown for 4 h to obtain cells in the exponential growth phase. The cells were resuspended in phosphate-buffered saline (PBS) (Gibco, Grand Island, NY) prior to use.

TABLE 1.

Bacterial strains used in this study

Strain Description or species Source or reference
S. aureus
    AH1181 LAC hla::erm This study
    AH1263 LAC (WT) 32
    AH1292 LAC agr::tet 47
    BB3047 LAC Δpsm; pALC2073 and pAH8 This study, 23
    BB3048 LAC Δpsm; pALC2073-PSMα and pRS-PSMβ This study, 23
Other staphylococci
    BB2153 S. saprophyticus This study
    BB2164 S. epidermidis This study
    BB2191 S. lugdunensis This study
    BB2201 S. warneri This study
    BB2203 S. hominis This study
    BB2205 S. haemolyticus This study
    BB2207 S. capitis This study

Peptides.

PSM peptides were synthesized by LifeTein (South Plainfield, NJ) and assayed to be >90% pure by high-performance liquid chromatography (HPLC). Synthetic peptides were prepared as previously described (33). Briefly, the peptides were resuspended in hexafluoroisopropanol (HFIP), and 0.5 mg was aliquoted into microcentrifuge tubes. The HFIP was then removed using a SpeedVac system at room temperature. The dried peptides were stored at −20°C. Immediately before use, the peptides were suspended in PBS to a concentration of 10 mg/ml and then diluted further in PBS to the desired concentration. LL-37 was purchased from AnaSpec Inc. (Fremont, CA).

Cell culture.

Primary human keratinocyte cultures were obtained from Johann Gudjonsson, University of Michigan. The cells were cultured in EpiLife medium (Gibco) supplemented with human keratinocyte growth supplement (Gibco), 1% Pen-Strep (100 U/ml penicillin and 100 μg/ml streptomycin; Gibco), and 0.25 μg/ml amphotericin B (Fungizone; Gibco). For all experiments, cells were used at passages 2 to 6, and 2 × 104 cells/well were plated in 24-well plates.

Cells were exposed to bacterial supernatants or peptides at confluence. The cells were pretreated for 30 min with 10 μM caspase inhibitors (Enzo) where indicated prior to addition of the indicated concentrations of bacterial supernatants or peptides. The cells were treated for 1 h prior to harvesting. The extracellular medium was removed after 1 h, and the remaining cells were lysed using RIPA buffer (300 mM NaCl, 50 mM Tris, 6.4 mM EDTA, 0.5% Triton X-100) containing protease inhibitor cocktail (Complete Mini, EDTA free; Roche, South San Francisco, CA) by incubation for 10 min on ice. The cell debris was removed by centrifugation at 13,000 × g for 5 min, and the lysates were frozen at −20°C until use.

For caspase 1 activation studies, keratinocytes were plated at 1 × 104 cells/well of a 96-well plate. The following day, the cells were exposed to S. aureus supernatants for 1 h, followed by addition of fluorescein isothiocyanate (FITC)-conjugated YVAD (ImmunoChemistry Technologies) for 1 h, and processed according to the manufacturer's recommendations. The cells were counterstained with DAPI (4′,6-diamidino-2-phenylindole) before fixation. Images of active caspase 1 were acquired on an Olympus IX70 inverted microscope (Olympus, Center Valley, PA) using a 40× objective at the Center for Live Cell Imaging (CLCI) at the University of Michigan Medical School.

To control for caspase inhibitor effectiveness, human peripheral blood mononuclear cells (PBMCs) were isolated via Ficoll gradient. The cells were plated at 1 × 106 per well of a 12-well plate; 10 μM YVAD or ZVAD was added 30 min prior to stimulation with 1 μg/ml lipopolysaccharide (LPS) for 24 h. The medium was collected, and IL-1β release was measured via enzyme-linked immunosorbent assay (ELISA).

Mice.

All animal studies were performed according to protocols approved by the University of Michigan Committee on the Use and Care of Animals, protocol 01823. The animals were obtained from Jackson Laboratory and housed at the University of Michigan in specific-pathogen-free housing, followed by biohazard containment after treatment with bacterial strains. To test the effects of exposure to WT S. aureus versus S. aureus Δpsm, the dorsal skin of 8- to 10-week-old C57BL/6 female mice was shaved and depilated with Veet (Reckitt Benckiser Group plc). The next day, the stratum corneum barrier was disrupted via mild stripping using three applications of Tegaderm (3M, St. Paul, MN). This technique exposes the keratinocyte layers without creating a wound (31). S. aureus (1 × 107 CFU) or PBS was placed with sterile gauze on the stripped skin and occluded with Tegaderm (3M). One week after S. aureus exposure, the animals were euthanized. Serum was collected via cardiac puncture, and skin was removed for cell count analysis via flow cytometry (see below) and RNA analysis.

Cytokine production.

In vitro quantification of IL-18 and IL-1β was done by ELISA (eBioscience, Vienna, Austria) according to the manufacturer's instructions. Active IL-18 release was assessed via Western blotting (primary antibody, rabbit anti-human IL-18 [Santa Cruz]; secondary antibody, goat anti-rabbit-horseradish peroxidase [HRP] [Abcam]). To investigate changes in the serum cytokine profiles of mice exposed to S. aureus, a Milliplex (EMD Millipore, Darmstadt, Germany) assay was performed.

Cytotoxicity.

To test for cell lysis following peptide treatments of keratinocytes, release of lactate dehydrogenase (LDH) was measured using a CytoTox 96 Non-Radioactive Cytotoxicity Assay (Promega, Madison, WI) according to the manufacturer's instructions.

Flow cytometry.

Lymphocytes were isolated from skin biopsy specimens as follows. Immediately after euthanasia, skin samples were removed, diced into 1- to 2-mm pieces using sterile razor blades, and then placed into a GentleMacs C Tube with 10 ml RPMI-10 (RPMI plus 10% fetal bovine serum) with 2 ml enzyme solution (100 mg/ml DNase type I [Sigma; DN-25], 100 mg/ml hyaluronidase type V [Sigma; H6254], 500 mg/ml collagenase [Sigma; C5138] in Hanks balanced salt solution [HBSS]). GentleMacs tissue dissociation was performed for 1 min, followed by 2 h of incubation at 37°C. After incubation, dissociation was repeated for 1 min. The cells were strained and pelleted by centrifugation at 800 × g for 7 min at 4°C. The supernatant was discarded, and the cells were washed once with 15 ml RPMI-10. After washing, the cells were divided into tubes for fluorescence-activated cell sorter (FACS) analysis. To quantify the inflammatory cellular infiltrate following S. aureus exposure, purified cells were stained for 1 h on ice with the following antibodies (all from BioLegend, San Diego, CA): CD19-phycoerythrin (PE)-Cy7, CD3-allophycocyanin (APC), CD4-FITC, CD8-PE, Ly6G-FITC, F4/80 Pacific Blue, CD11b-APC, CD11c-PE-Cy7, CD11c-PE, and Ly6C-PE-Cy7. Flow data were collected on a BD LSR II and analyzed with FlowJo V10 (Tree Star).

RT-PCR.

Skin biopsy specimens were homogenized in TriPure (Roche), and RNA was isolated with Direct-zol mini RNA prep (Zymo). RNA (100 ng) was transcribed into cDNA, and real-time (RT) PCR analysis was completed on an ABI Prism 7900HT (Applied Biosystems) using Sybr green (Life Technologies) (Table 2 lists the primers). Cycle times were normalized to β-actin, and the fold change (2−ΔΔCT) was calculated versus PBS-exposed mice.

TABLE 2.

Primers used for real-time PCR

Gene Primer (5′→3′)
Forward Reverse
IFN-α ATGGCTAGRCTCTGTGCTTTCCT AGGGCTCTCCAGAYTTCTGCTCTG
IFN-β AGCTCCAAGAAAGGACGAACAT ATTCTTGCTTCGGCAGTTAC
IFN-γ AGCGGCTGACTGAACTCAGATTGTA GTCACAGTTTTCAGCTGTATAGGG
IL-18 ACTGTACAACCGCAGTAATACGC AGTGAACATTACAGATTTATCCC
IL-1β CCCTGCAGCTGGAGAGTGTGGA CTGAGCGACCTGTCTTGGCCG
IL-6 CTGCAAGAGACTTCCATCCAG AGTGGTATAGACAGGTCTGTTGG
IL-1α CGGGTGACAGTATCAGCAAC GACAAACTTCTGCCTGACGA
TNF-α CCCACTCTGACCCCTTTACT TTTGAGTCCTTGATGGTGGT
CAMP TCAACCAGCAGTCCCTAGAC AAGGCACATTGCTCAGGTAG
NLRP3 ATGCTGCTTCGACATCTCCT AACCAATGCGAGATCCTGAC
Chemokine (C-C motif) receptor 1 (CCR1) TGGGCAATGTCCTAGTGATT GCATCACCAAAAATCCAGTC
CCL4 AGCAACACCATGAAGCTCTG CTGTCTGCCTCTTTTGGTCA
CCL5 CAATCTTGCAGTCGTGTTTG GGAGTGGGAGTAGGGGATTA
C-X-C motif chemokine 10 (CXCL10) CAAAAGTAACTGCCGAAGCA CTGAGCTAGGGAGGACAAGG
CXCL13 AGAGGTTTGCGAGATGGACT GAGCCTGGACCTTTAAGCTG
β-Actin TGGAATCCTGTGGCATCCTGAAAC TAAAACGCAGCTCAGTAACAGTCCG

Statistical analysis.

Statistical analysis was performed using GraphPad Prism 6 software. Comparisons between data were made via 2-sided Student unpaired t tests or Mann-Whitney U tests for nonnormalized data. A P value of <0.05 was considered significant.

RESULTS

Some staphylococci produce cytotoxic factors that affect human keratinocytes.

To investigate the effects that secreted factors from different staphylococci have on human keratinocytes, we exposed human keratinocytes to bacterial supernatants. The keratinocytes were monitored for the release of IL-18 because of its constitutive production in keratinocytes and its proinflammatory activity in the skin (11). We found that supernatants from several staphylococcal species, including S. aureus, S. lugdunensis, S. epidermidis, Staphylococcus capitis, Staphylococcus warneri, and, to a lesser extent but still significantly, Staphylococcus hominis, were able to trigger the release of IL-18 from keratinocytes (Fig. 1A). Supernatants from Staphylococcus saprophyticus and Staphylococcus haemolyticus did not induce release of IL-18 (Fig. 1A). Importantly, release of LDH, a marker of cell lysis, aligned with IL-18 release from the cells, with the exception of S. hominis, which showed no significant increase in LDH release (Fig. 1B).

FIG 1.

FIG 1

Various staphylococcal species induce lytic release of IL-18 from human keratinocytes. (A and B) Human keratinocytes were exposed to supernatants from several Staphylococcus species and tested for release of IL-18 (A) and LDH (B). (C to E) Human keratinocytes were also exposed to supernatants from several strains of S. aureus and tested for the release of IL-18 (C), LDH (D), and IL-1β (E). ****, P < 0.0001; **, P < 0.01; *, P < 0.05; n = 2 in duplicate for each study. The error bars represent standard errors of the mean.

S. aureus phenol-soluble modulins induce IL-18, IL-1β, and LDH release from human keratinocytes.

Because of its prominent role in skin infections and AD, we chose to focus the rest of our study on S. aureus. Using a candidate approach, we found that the quorum-sensing system in S. aureus known as the accessory gene regulator (agr) was necessary for cytotoxicity and IL-18 and IL-1β release (Fig. 1C, D, and E). Because the agr system is known to regulate the expression of many virulence factors produced by S. aureus, we investigated mutants for two well-studied cytotoxic factors: HLA and the PSM family. S. aureus supernatants lacking PSMs (PSMα [PSMα1 to -4], PSMβ [PSMβ1 and -2], and δ-toxin) (ΔPSM) showed a significant decrease in the ability to stimulate the release of IL-18, IL-1β, and LDH from keratinocytes, and the release was restored when the strains were complemented with PSMα and PSMβ operons (Fig. 1C, D, and E). Notably, lack of HLA, which has been reported to lyse human cells and activate the inflammasome (34), did not impact the release of IL-18, LDH, or IL-1β compared to our WT S. aureus supernatant (Fig. 1C, D, and E). These data suggest that PSMs are the primary S. aureus factors that induce lysis of keratinocytes.

To further investigate which PSMs contribute to IL-18 release and keratinocyte lysis, synthetic PSMs were used. PSM peptides were sufficient to stimulate the release of IL-18 from keratinocytes (Fig. 2A). Interestingly, all of the PSMα peptides, as well as δ-toxin, but neither of the PSMβ peptides, induced release of LDH from the keratinocytes (Fig. 2B). As PSMs are thought to have a helical structure similar to that of the known lytic antimicrobial peptide LL-37 (reviewed in reference 35), we also compared the dose response of the ability of PSMα1 to induce IL-18 release to that of LL-37. As shown in Fig. 2C, both peptides induced IL-18 release at 5 μg/ml, but LL-37 appears to be more potent. Taken together, these data demonstrate that the PSM peptides of S. aureus are necessary and sufficient for the release of IL-18, likely through lytic release from human keratinocytes.

FIG 2.

FIG 2

S. aureus PSMs are sufficient for lytic release of IL-18 from human keratinocytes. (A and B) Human keratinocytes were exposed to synthetic PSM peptides and tested for release of IL-18 (A) and LDH (B). (C) PSMs induce release of IL-18 in a dose-dependent mechanism similar to that of LL-37. ****, P < 0.0001; ***, P < 0.001; **, P < 0.01; *, P < 0.05; n = 3 in duplicate for each study. The error bars represent standard errors of the mean.

The release of mature IL-18 is independent of caspase activity.

Several S. aureus toxins have been reported to activate the inflammasome, so we next investigated whether inflammasome activation participated in the release of IL-18 from keratinocytes following S. aureus exposure (36). As shown in Fig. 3A, we confirmed that stimulation of keratinocytes with WT S. aureus supernatant resulted in detectable active caspase 1 within the keratinocytes. Western blotting confirmed that the IL-18 released was in its active form (Fig. 3B). To see if caspase 1 activation was required for the release of IL-18 or IL-1β, we pretreated primary keratinocytes with various cell-permeable caspase inhibitors, followed by stimulation with WT S. aureus supernatant. Inhibition of caspase 1 (YVAD), caspase 3 (DEVD), caspase 5 (WEHD), caspase 6 (VEID), caspase 8 (IEHD), or all caspases (ZVAD) did not impact S. aureus-induced lytic cytokine release (Fig. 3C, D, and F). However, identical concentrations of YVAD and ZVAD repressed inflammasome activation induced by LPS in PBMCs (Fig. 3E). Elevated extracellular potassium, another condition that inhibits NLR family, pyrin domain containing 3 (NLRP3) inflammasome activation, did not inhibit S. aureus-mediated cytokine release (data not shown). These data suggest that release of active IL-18 by PSMs is specific to disruption of cell membranes and is not due to specific activation of the inflammasome.

FIG 3.

FIG 3

The release of mature IL-18 from human keratinocytes in response to S. aureus is independent of caspase 1 function. (A) Human keratinocytes were exposed to TSB or WT S. aureus supernatants for 1 h, followed by addition of a fluorescence-labeled peptide that binds active caspase 1 and visualized by fluorescence microscopy (n = 3). (B) Representative Western blotting for full-length, or “pro,” form and active, cleaved IL-18 released into the extracellular media (ECM) from keratinocytes exposed to TSB or WT supernatants. Recombinant active IL-18 (rIL-18) was run as a positive control. (C, D, and F) Dimethyl sulfoxide (DMSO) or caspase inhibitors (10 mM) were added to human keratinocytes prior to treatment with S. aureus supernatants and then testing for the release of IL-18 (n = 3) (C), IL-1β (n = 3) (D), and LDH (n = 1) (F). (E) Human PBMCs were incubated with 10 mM YVAD or ZVAD for 30 min prior to overnight incubation with 1 mg/ml LPS. Released IL-1β was detected via ELISA. The error bars represent standard errors of the mean. **, P < 0.01; ****, P < 0.0001.

Phenol-soluble modulins are required for neutrophil infiltration following epidermal S. aureus exposure.

Given that PSMs induce lysis of and inflammatory cytokine release from keratinocytes, we hypothesized that they may be required for initiation of inflammation in response to cutaneous colonization by S. aureus. We thus challenged mice epicutaneously with the WT or Δpsm strain of S. aureus. As shown in Fig. 4A, application of WT S. aureus to the surface of the skin resulted in infectious changes, including erythema and ulceration, whereas the Δpsm strain produced minimal skin changes. Flow cytometry of the colonized skin demonstrated a significant decrease in neutrophil infiltration of the skin in the WT compared to the Δpsm strain. Other cell types, including macrophages, dendritic cells (DC), B-cells, and T cells, were not significantly altered following S. aureus exposure (Fig. 4B). Using C57BL/6 IL-18−/− mice, we found that IL-18 was not required for S. aureus-induced neutrophil infiltration into the skin (Fig. 4C). Absence of IL-18 enhanced macrophage, CD11b+ DC, and CD4+ T cell migration, suggesting that keratinocyte release of IL-18 may have repressive effects on chronic inflammation (Fig. 4C). These data suggest that PSMs are required for invasive infection when S. aureus is exposed to intact keratinocyte layers via epicutaneous application and that IL-18 is not required for this process.

FIG 4.

FIG 4

PSMs are critical for the recruitment of neutrophils into the skin during S. aureus epidermal exposure. (A) Representative photographs of dorsal surfaces from mice taken after 7 days of exposure. (B) FACS analysis of leukocytes obtained from skin samples from mice 7 days postinoculation with S. aureus. *, P < 0.05; n = 8 for each group. (C) FACS analysis of skin leukocytes from WT or IL-18 knockout (KO) mice exposed (n = 5) to WT S. aureus, as in panel B. Neutrophils, Ly6G+; macrophages, F4/80+ CD11b+; dendritic cells, Ly6c+ CD11c+; T cells, CD3+; B cells, CD19+. The error bars represent standard errors of the mean. ***, P < 0.001; ****, P < 0.0001.

Others have demonstrated that epicutaneous application of S. aureus results in upregulation of proinflammatory cytokines (31). In order to determine whether PSMs play a role in modulating inflammatory gene expression in the skin, we performed RT-PCR on skin sections following exposure to the WT or Δpsm strain. As shown in Fig. 5A, both the WT and Δpsm strains were able to dramatically upregulate proinflammatory genes within the skin, including alpha interferon (IFN-α), IFN-γ, IL-18, IL-1β, tumor necrosis factor alpha (TNF-α), and IL-6, compared to skin exposed to PBS. Both were also able to upregulate the antimicrobial peptide cathelicidin antimicrobial peptide (CAMP), the murine LL-37 ortholog. Chemokines, such as chemokine (C-C motif) ligand 4 (CCL4), were also upregulated by both strains. Given that the skin phenotype following exposure to the WT versus the Δpsm strain was so dramatically different, we then chose to examine systemic cytokine concentrations in mice exposed to both strains. As shown in Fig. 5B and Table 3, the concentrations of inflammatory cytokines, including granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-1β, IL-6, and IL-17, in serum are significantly diminished in Δpsm strain-exposed compared to WT-exposed mice. Together, these data suggest that PSMs are not required to stimulate inflammatory gene upregulation but are required to initiate release of proinflammatory cytokines into circulation and to permit the subsequent inflammatory response.

FIG 5.

FIG 5

Systemic inflammatory response, but not inflammatory gene upregulation, requires S. aureus PSMs. (A) Fold changes (FC) in the cutaneous expression of the listed genes compared to PBS-exposed mice as calculated by real-time PCR. (B) Concentrations of the indicated cytokines in serum as determined by Milliplex in WT S. aureus-exposed versus S. aureus Δpsm-exposed mice. **, P < 0.01; ***, P < 0.001; n = 8 for each group. The error bars represent standard errors of the mean.

TABLE 3.

Cytokine concentrations from mice exposed to WT S. aureus, S. aureus Δpsm, or vehicle control (PBS)

Cytokine Concn (pg/ml) after exposure to:
Significant difference between WT S. aureus- and S. aureus Δpsm-exposed mice (P value)
WT S. aureus S. aureus Δpsm PBS
G-CSF 3443.4 287.9 434.1 Yes (0.0011)
GM-CSF 34.5 12.1 14.8 Yes (0.00540)
IFNγ 2.5 2.0 2.1 No (0.2000)
IL-1α 249.6 266.4 275.4 No (0.0827)
IL-1β 16.6 7.9 22.7 Yes (0.0009)
IL-2 3.6 2.7 3.7 Yes (0.0256)
IL-4 3.0 3.0 3.4 No (1.0000)
IL-5 8.4 5.6 17.8 No (0.2887)
IL-6 71.6 9.0 23.5 Yes (0.0070)
IL-7 3.4 1.0 18.5 Yes (0.0002)
IL-10 3.8 2.1 3.0 Yes (0.0020)
IL-12p40 4.9 3.7 3.5 No (0.2822)
IL-12p70 10.2 3.7 13.0 Yes (0.00480)
IL-13 84.7 45.7 58.7 Yes (0.0047)
IL-15 6.8 2.5 2.5 Yes (0.0256)
IL-17 11.7 3.0 3.3 Yes (0.00280)
IL-18 193.8 201.1 190.9 No (0.769)
IP-10 142.7 101.9 80.4 Yes (0.0379)
KC 116.3 81.5 104.1 No (0.3754)
MCP-1 63.9 25.4 24.4 No (0.0692)
MIP-1a 39.5 13.6 19.5 Yes (0.0006)
MIP-1b 18.7 9.0 13.1 No (0.2124)
MIP-2 88.0 75.1 60.4 No (0.3958)
RANTES 14.9 9.4 7.1 No (0.2247)
TNF-α 5.2 3.4 3.6 No (0.0816)

DISCUSSION

S. aureus colonization is known to be highly prevalent in the U.S. population and also contributes to chronic skin conditions, such as AD. In this paper, we demonstrate that the PSMα family members, including δ-toxin, are necessary and sufficient for lytic activity on keratinocytes, which results in the release of inflammatory cytokines, such as IL-18 and IL-1β (Fig. 1 and 2). Additionally, we demonstrate that PSMs, while not required for transcriptional upregulation of proinflammatory genes within the skin, are required for instigation of skin inflammation and systemic cytokine changes in an epicutaneous colonization model (Fig. 4 and 5).

Several groups have investigated mechanisms by which S. aureus can elicit an immune response from keratinocytes; however, these factors have required the use of surfactants to promote membrane permeability (11). Our data presented here demonstrate that keratinocyte-mediated inflammation can be stimulated by S. aureus PSMs alone. Because of their amphipathic nature, PSMs have surfactant-like properties that allow membrane intercalation and pore formation, similar to the human cathelicidin LL-37 (Fig. 2C) (21). Interestingly, not all species of staphylococci induce keratinocyte lysis (Fig. 1). We observed strong lytic abilities in species known to contain PSMs, including S. aureus, S. epidermidis, and S. lugdunensis, but there has been no study, to our knowledge, investigating the presence of PSMs in other staphylococcal species. Further investigation of S. hominis, which is able to induce release of IL-18 without inducing cell lysis, will be of interest. Additionally, the PSMβ peptides were able to induce the release of IL-18 from human keratinocytes without causing a release of LDH (Fig. 2). It is possible that the PSMβ peptides are able to activate a receptor-mediated cytokine release or possibly trigger slow, nonlytic inflammasome activation.

Keratinocyte activation and release of IL-18 and IL-1β by caspase 1 as part of an active inflammasome have been reported (37). Additionally, an inflammasome-independent role for caspase 1 in apoptosis of keratinocytes has been described (38). Interestingly, in the presence of S. aureus supernatants, we detected activation of caspase 1, but this activation is not required for IL-18 release. This suggests that PSMs are promoting cytokine release via lysis (Fig. 3) and that other enzymes, such as cathepsin activation or granzyme B, may be important for IL-18 activation in keratinocytes (39). This PSM-induced IL-18 release may resolve the contradiction between the role of increased IL-18 signaling in atopic dermatitis and recent data that describe repressed inflammasome activity in AD (40, 41). Chronic colonization with PSM-producing S. aureus may circumvent the need for inflammasome-mediated IL-18 release.

In our epicutaneous model of S. aureus exposure, we found that absence of IL-18 did not impact neutrophil recruitment but enhanced chronic inflammatory cell infiltration. This is an interesting finding, as the actual function of IL-18 in the skin has not been systematically investigated. Many studies examining the role of IL-18 in cutaneous disease report up- or downregulation of the cytokine without reporting the consequence of these changes (42). Our data suggest that following S. aureus exposure, IL-18 may function to modulate the acute versus chronic inflammatory response. Given that absence of IL-18 promoted a large increase in CD4+ T cells in our study, further investigation into the Th1 versus Th2 skewing of these populations is warranted. Additionally, determining whether the effects of IL-18 are different in AD models exposed to S. aureus is an important area for future research.

PSMs produced by S. aureus have been shown to be critical determinants of pathogenesis in murine models of abscess and bacteremia (19). They are recognized by the human formyl peptide receptor 2, which stimulates neutrophil chemotaxis (24). Not only can PSMs recruit neutrophils, but they have been shown to intercalate into cell membranes, resulting in lysis and death (19). In our in vivo model, transcriptional changes of cytokines in the dermis in our mice did not differ between mice exposed to WT S. aureus and mice exposed to S. aureus Δpsm (Fig. 5A). This likely reflects intact Toll-like receptor (TLR) ligands, such as peptidoglycan, present on each strain. Keratinocytes express TLR2, which results in robust inflammatory gene upregulation after exposure to peptidoglycan (43). Thus, it is not surprising to see robust gene expression changes in the skin after exposure to the WT and Δpsm strains. Importantly, however, while a robust systemic inflammatory response was noted with WT S. aureus, the circulating concentrations of inflammatory cytokines were dramatically and significantly reduced after exposure to S. aureus Δpsm (Fig. 5B). The large difference in G-CSF agrees with previously reported effects of one of the PSMs, δ-toxin, on mast cell degranulation and G-CSF release (44). Differences in circulating IL-6 and IL-1β, both of which are released from keratinocytes, were also noted (Fig. 3) (37, 45). Systemic levels of IL-18 were not altered after S. aureus exposure (Table 3), but this does not rule out important localized effects, as other cytokines important in AD have been noted to be increased in the skin but not in serum (46). Taken together with our in vitro data, which demonstrate PSM-mediated release of IL-18 and IL-1β from keratinocytes (Fig. 1C and E and 5B), these results suggest that PSM-mediated keratinocyte lysis may be a required factor to convert S. aureus colonization into infection.

In summary, we have described a novel role for staphylococcal PSMs, which induce keratinocyte lysis and release of inflammatory cytokines. This translates into a role of PSMs in infection and the systemic inflammatory response in vivo. We propose that PSMs are important for initiating pathogenesis after colonization and may serve as important targets for treating infection or modulating inflammatory cutaneous disease.

ACKNOWLEDGMENTS

We thank the laboratory of Johann Gudjonsson at the University of Michigan for providing cells and protocols and allowing us the use of their equipment. We also thank the members of the Kahlenberg laboratory at the University of Michigan, as well as Matthew Brown in the Boles laboratory at the University of Iowa, for insightful conversations.

This work was funded by the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health under award number K08AR063668 to J.M.K., NIH grant NIAID AI081748 to B.R.B., and American Heart Association Fellowship 13PRE13810001 to A.K.S.

REFERENCES

  • 1.Klevens RM, Morrison MA, Nadle J, Petit S, Gershman K, Ray S, Harrison LH, Lynfield R, Dumyati G, Townes JM, Craig AS, Zell ER, Fosheim GE, McDougal LK, Carey RB, Fridkin SK, Active Bacterial Core Surveillance (ABCs) Investigators MRSA. 2007. Invasive methicillin-resistant Staphylococcus aureus infections in the United States. JAMA 298:1763–1771. doi: 10.1001/jama.298.15.1763. [DOI] [PubMed] [Google Scholar]
  • 2.Kuehnert MJ, Kruszon-Moran D, Hill HA, McQuillan G, McAllister SK, Fosheim G, McDougal LK, Chaitram J, Jensen B, Fridkin SK, Killgore G, Tenover FC. 2006. Prevalence of Staphylococcus aureus nasal colonization in the United States, 2001–2002. J Infect Dis 193:172–179. doi: 10.1086/499632. [DOI] [PubMed] [Google Scholar]
  • 3.Sievert DM, Ricks P, Edwards JR, Schneider A, Patel J, Srinivasan A, Kallen A, Limbago B, Fridkin S, National Healthcare Safety Network (NHSN) Team and Participating Facilities NHSN. 2013. Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2009–2010. Infect Control Hosp Epidemiol 34:1–14. doi: 10.1086/668770. [DOI] [PubMed] [Google Scholar]
  • 4.De Benedetto A, Agnihothri R, McGirt LY, Bankova LG, Beck LA. 2009. Atopic dermatitis: a disease caused by innate immune defects? J Investig Dermatol 129:14–30. doi: 10.1038/jid.2008.259. [DOI] [PubMed] [Google Scholar]
  • 5.Nutten S. 2015. Atopic dermatitis: global epidemiology and risk factors. Ann Nutr Metab 66(Suppl 1):S8–S16. doi: 10.1159/000370220. [DOI] [PubMed] [Google Scholar]
  • 6.Foster TJ, Hook M. 1998. Surface protein adhesins of Staphylococcus aureus. Trends Microbiol 6:484–488. doi: 10.1016/S0966-842X(98)01400-0. [DOI] [PubMed] [Google Scholar]
  • 7.Travers JB, Norris DA, Leung DY. 2001. The keratinocyte as a target for staphylococcal bacterial toxins. J Investig Dermatol Symp Proc 6:225–230. doi: 10.1046/j.0022-202x.2001.00045.x. [DOI] [PubMed] [Google Scholar]
  • 8.Sasaki T, Kano R, Sato H, Nakamura Y, Watanabe S, Hasegawa A. 2003. Effects of staphylococci on cytokine production from human keratinocytes. Br J Dermatol 148:46–50. doi: 10.1046/j.1365-2133.2003.05017.x. [DOI] [PubMed] [Google Scholar]
  • 9.Travers JB, Kozman A, Mousdicas N, Saha C, Landis M, Al-Hassani M, Yao W, Yao Y, Hyatt A-M, Sheehan MP, Haggstrom AN, Kaplan MH. 2010. Infected atopic dermatitis lesions contain pharmacologic amounts of lipoteichoic acid. J Allergy Clin Immunol 125:146–152.e1–2. doi: 10.1016/j.jaci.2009.09.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Yao Y, Kozman A, Al-Hassani M, Saha CK, Yi Q, Yao W, Mousdicas N, Kaplan MH, Travers JB. 2010. Identification of staphylococcal protein A in infected atopic dermatitis lesions. J Investig Dermatol 130:2502–2504. doi: 10.1038/jid.2010.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Terada M, Tsutsui H, Imai Y, Yasuda K, Mizutani H, Yamanishi K, Kubo M, Matsui K, Sano H, Nakanishi K. 2006. Contribution of IL-18 to atopic-dermatitis-like skin inflammation induced by Staphylococcus aureus product in mice. Proc Natl Acad Sci U S A 103:8816–8821. doi: 10.1073/pnas.0602900103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ommori R, Ouji N, Mizuno F, Kita E, Ikada Y, Asada H. 2013. Selective induction of antimicrobial peptides from keratinocytes by staphylococcal bacteria. Microb Pathog 56:35–39. doi: 10.1016/j.micpath.2012.11.005. [DOI] [PubMed] [Google Scholar]
  • 13.Higashi N, Gesser B, Kawana S, Thestrup-Pedersen K. 2001. Expression of IL-18 mRNA and secretion of IL-18 are reduced in monocytes from patients with atopic dermatitis. J Allergy Clin Immunol 108:607–614. doi: 10.1067/mai.2001.118601. [DOI] [PubMed] [Google Scholar]
  • 14.Yamanaka K, Tanaka M, Tsutsui H, Kupper TS, Asahi K, Okamura H, Nakanishi K, Suzuki M, Kayagaki N, Black RA, Miller DK, Nakashima K, Shimizu M, Mizutani H. 2000. Skin-specific caspase-1-transgenic mice show cutaneous apoptosis and pre-endotoxin shock condition with a high serum level of IL-18. J Immunol 165:997–1003. doi: 10.4049/jimmunol.165.2.997. [DOI] [PubMed] [Google Scholar]
  • 15.Fantuzzi G, Puren AJ, Harding MW, Livingston DJ, Dinarello CA. 1998. Interleukin-18 regulation of interferon gamma production and cell proliferation as shown in interleukin-1beta-converting enzyme (caspase-1)-deficient mice. Blood 91:2118–2125. [PubMed] [Google Scholar]
  • 16.Lamkanfi M, Dixit VM. 2012. Inflammasomes and their roles in health and disease. Annu Rev Cell Dev Biol 28:137–161. doi: 10.1146/annurev-cellbio-101011-155745. [DOI] [PubMed] [Google Scholar]
  • 17.Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR, Aunins J, Elliston KO, Ayala JM, Casano FJ, Chin J, Ding J-F, Egger LA, Gaffney EP, Limjuco G, Palyha OC, Raju SM, Rolando AM, Salley JP, Yamin TT, Lee TD, Shivey JE, MacCross M, Mumford RA, Schmidt JA, Tocci MJ. 1992. A novel heterodimeric cysteine protease is required for interleukin-1β processing in monocytes. Nature 356:768–774. doi: 10.1038/356768a0. [DOI] [PubMed] [Google Scholar]
  • 18.Sahoo M, Ceballos-Olvera I, del Barrio L, Re F. 2011. Role of the inflammasome, IL-1β, and IL-18 in bacterial infections. ScientificWorldJournal 11:2037–2050. doi: 10.1100/2011/212680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wang R, Braughton KR, Kretschmer D, Bach T-HL, Queck SY, Li M, Kennedy AD, Dorward DW, Klebanoff SJ, Peschel A, DeLeo FR, Otto M. 2007. Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat Med 13:1510–1514. doi: 10.1038/nm1656. [DOI] [PubMed] [Google Scholar]
  • 20.Tsompanidou E, Denham EL, Becher D, de Jong A, Buist G, van Oosten M, Manson WL, Back JW, van Dijl JM, Dreisbach A. 2013. Distinct roles of phenol-soluble modulins in spreading of Staphylococcus aureus on wet surfaces. Appl Environ Microbiol 79:886–895. doi: 10.1128/AEM.03157-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tsompanidou E, Sibbald MJJB, Chlebowicz MA, Dreisbach A, Back JW, van Dijl JM, Buist G, Denham EL. 2011. Requirement of the agr locus for colony spreading of Staphylococcus aureus. J Bacteriol 193:1267–1272. doi: 10.1128/JB.01276-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Periasamy S, Joo H-S, Duong AC, Bach T-HL, Tan VY, Chatterjee SS, Cheung GYC, Otto M. 2012. How Staphylococcus aureus biofilms develop their characteristic structure. Proc Natl Acad Sci U S A 109:1281–1286. doi: 10.1073/pnas.1115006109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Schwartz K, Syed AK, Stephenson RE, Rickard AH, Boles BR. 2012. Functional amyloids composed of phenol soluble modulins stabilize Staphylococcus aureus biofilms. PLoS Pathog 8:e1002744. doi: 10.1371/journal.ppat.1002744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kretschmer D, Gleske A-K, Rautenberg M, Wang R, Köberle M, Bohn E, Schöneberg T, Rabiet M-J, Boulay F, Klebanoff SJ, van Kessel KA, van Strijp JA, Otto M, Peschel A. 2010. Human formyl peptide receptor 2 senses highly pathogenic Staphylococcus aureus. Cell Host Microbe 7:463–473. doi: 10.1016/j.chom.2010.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Surewaard BGJ, Nijland R, Spaan AN, Kruijtzer JAW, de Haas CJC, van Strijp JAG. 2012. Inactivation of staphylococcal phenol soluble modulins by serum lipoprotein particles. PLoS Pathog 8:e1002606. doi: 10.1371/journal.ppat.1002606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cogen AL, Yamasaki K, Sanchez KM, Dorschner RA, Lai Y, MacLeod DT, Torpey JW, Otto M, Nizet V, Kim JE, Gallo RL. 2010. Selective antimicrobial action is provided by phenol-soluble modulins derived from Staphylococcus epidermidis, a normal resident of the skin. J Invest Dermatol 130:192–200. doi: 10.1038/jid.2009.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Joo HS, Cheung GYC, Otto M. 2011. Antimicrobial activity of community-associated methicillin-resistant Staphylococcus aureus is caused by phenol-soluble modulin derivatives. J Biol Chem 286:8933–8940. doi: 10.1074/jbc.M111.221382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gonzalez DJ, Okumura CY, Hollands A, Kersten R, Akong-Moore K, Pence MA, Malone CL, Derieux J, Moore BS, Horswill AR, Dixon JE, Dorrestein PC, Nizet V. 2012. Novel phenol-soluble modulin derivatives in community-associated methicillin-resistant Staphylococcus aureus identified through imaging mass spectrometry. J Biol Chem 287:13889–13898. doi: 10.1074/jbc.M112.349860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mehlin C, Headley CM, Klebanoff SJ. 1999. An inflammatory polypeptide complex from Staphylococcus epidermidis: isolation and characterization. J Exp Med 189:907–918. doi: 10.1084/jem.189.6.907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Donvito B, Etienne J, Denoroy L, Greenland T, Benito Y, Vandenesch F. 1997. Synergistic hemolytic activity of Staphylococcus lugdunensis is mediated by three peptides encoded by a non-agr genetic locus. Infect Immun 65:95–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wanke I, Skabytska Y, Kraft B, Peschel A, Biedermann T, Schittek B. 2013. Staphylococcus aureus skin colonization is promoted by barrier disruption and leads to local inflammation. Exp Dermatol 22:153–155. doi: 10.1111/exd.12083. [DOI] [PubMed] [Google Scholar]
  • 32.Boles BR, Thoendel M, Roth AJ, Horswill AR. 2010. Identification of genes involved in polysaccharide-independent Staphylococcus aureus biofilm formation. PLoS One 5:e10146. doi: 10.1371/journal.pone.0010146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Schwartz K, Sekedat MD, Syed AK, O'Hara B, Payne DE, Lamb A, Boles BR. 2014. The AgrD N-terminal leader peptide of Staphylococcus aureus has cytolytic and amyloidogenic properties. Infect Immun 82:3837–3844. doi: 10.1128/IAI.02111-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kebaier C, Chamberland RR, Allen IC, Gao X, Broglie PM, Hall JD, Jania C, Doerschuk CM, Tilley SL, Duncan JA. 2012. Staphylococcus aureus α-hemolysin mediates virulence in a murine model of severe pneumonia through activation of the NLRP3 inflammasome. J Infect Dis 205:807–817. doi: 10.1093/infdis/jir846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kahlenberg JM, Kaplan MJ. 2013. Little peptide, big effects: the role of LL-37 in inflammation and autoimmune disease. J Immunol 191:4895–4901. doi: 10.4049/jimmunol.1302005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Muñoz-Planillo R, Franchi L, Miller LS, Núñez G. 2009. A critical role for hemolysins and bacterial lipoproteins in Staphylococcus aureus-induced activation of the Nlrp3 inflammasome. J Immunol 183:3942–3948. doi: 10.4049/jimmunol.0900729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Dai X, Sayama K, Tohyama M, Shirakata Y, Hanakawa Y, Tokumaru S, Yang L, Hirakawa S, Hashimoto K. 2011. Mite allergen is a danger signal for the skin via activation of inflammasome in keratinocytes. J Allergy Clin Immunol 127:806–814.e1–4. doi: 10.1016/j.jaci.2010.12.006. [DOI] [PubMed] [Google Scholar]
  • 38.Sollberger G, Strittmatter GE, Grossi S, Garstkiewicz M, Auf dem Keller U, French LE, Beer H-D. 2015. Caspase-1 activity is required for UVB-induced apoptosis of human keratinocytes. J Investig Dermatol 135:1395–1404. doi: 10.1038/jid.2014.551. [DOI] [PubMed] [Google Scholar]
  • 39.Akeda T, Yamanaka K, Tsuda K, Omoto Y, Gabazza EC, Mizutani H. 2014. CD8+ T cell granzyme B activates keratinocyte endogenous IL-18. Arch Dermatol Res 306:125–130. doi: 10.1007/s00403-013-1382-1. [DOI] [PubMed] [Google Scholar]
  • 40.Inoue Y, Aihara M, Kirino M, Harada I, Komori-Yamaguchi J, Yamaguchi Y, Nagashima Y, Ikezawa Z. 2011. Interleukin-18 is elevated in the horny layer in patients with atopic dermatitis and is associated with Staphylococcus aureus colonization. Br J Dermatol 164:560–567. doi: 10.1111/j.1365-2133.2010.10145.x. [DOI] [PubMed] [Google Scholar]
  • 41.Niebuhr M, Baumert K, Heratizadeh A, Satzger I, Werfel T. 2014. Impaired NLRP3 inflammasome expression and function in atopic dermatitis due to Th2 milieu. Allergy 69:1058–1067. doi: 10.1111/all.12428. [DOI] [PubMed] [Google Scholar]
  • 42.Puxeddu I, Italiani P, Giungato P, Pratesi F, Panza F, Bartaloni D, Rocchi V, Del Corso I, Boraschi D, Migliorini P. 2013. Free IL-18 and IL-33 cytokines in chronic spontaneous urticaria. Cytokine 61:741–743. doi: 10.1016/j.cyto.2013.01.015. [DOI] [PubMed] [Google Scholar]
  • 43.Yoshimura A, Lien E, Ingalls RR, Tuomanen E, Dziarski R, Golenbock D. 1999. Cutting edge: recognition of gram-positive bacterial cell wall components by the innate immune system occurs via Toll-like receptor 2. J Immunol 163:1–5. [PubMed] [Google Scholar]
  • 44.Nakamura Y, Oscherwitz J, Cease KB, Chan SM, Muñoz-Planillo R, Hasegawa M, Villaruz AE, Cheung GYC, McGavin MJ, Travers JB, Otto M, Inohara N, Núñez G. 2013. Staphylococcus δ-toxin induces allergic skin disease by activating mast cells. Nature 503:397–401. doi: 10.1038/nature12655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wozniacka A, Lesiak A, Boncela J, Smolarczyk K, McCauliffe DP, Sysa-Jedrzejowska A. 2008. The influence of antimalarial treatment on IL-1beta, IL-6 and TNF-alpha mRNA expression on UVB-irradiated skin in systemic lupus erythematosus. Br J Dermatol 159:1124–1130. doi: 10.1111/j.1365-2133.2008.08804.x. [DOI] [PubMed] [Google Scholar]
  • 46.Chen Y, Lind Enoksson S, Johansson C, Karlsson MA, Lundeberg L, Nilsson G, Scheynius A, Karlsson MCI. 2011. The expression of BAFF, APRIL and TWEAK is altered in eczema skin but not in the circulation of atopic and seborrheic eczema patients. PLoS One 6:e22202. doi: 10.1371/journal.pone.0022202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kiedrowski MR, Kavanaugh JS, Malone CL, Mootz JM, Voyich JM, Smeltzer MS, Bayles KW, Horswill AR. 2011. Nuclease modulates biofilm formation in community-associated methicillin-resistant Staphylococcus aureus. PLoS One 6:e26714. doi: 10.1371/journal.pone.0026714. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Infection and Immunity are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES