Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Dec 1.
Published in final edited form as: Curr Dermatol Rep. 2018 Sep 22;7(4):338–349. doi: 10.1007/s13671-018-0235-8

Immune and Inflammatory Reponses to Staphylococcus aureus Skin Infections

Qi Liu 1, Momina Mazhar 1, Lloyd S Miller 1,2,3,4
PMCID: PMC6461387  NIHMSID: NIHMS1507796  PMID: 30989002

Abstract

Purpose of Review

There have been recent advances in our understanding of cutaneous immune responses to the important human skin pathogen, Staphylococcus aureus (S. aureus). This review will highlight these insights into innate and adaptive immune mechanisms in host defense and cutaneous inflammation in response to S. aureus skin infections.

Recent Findings

Antimicrobial peptides, pattern recognition receptors and inflammasome activation function in innate immunity as well as T cells and their effector cytokines play a key role in adaptive immunity against S. aureus skin infections. In addition, certain mechanisms by which S. aureus contributes to aberrant cutaneous inflammation, such as in flares of the inflammatory skin disease atopic dermatitis have also been identified.

Summary

These cutaneous immune mechanisms could provide new targets for future vaccines and immune-based therapies to combat skin infections and cutaneous inflammation caused by S. aureus.

Keywords: Staphylococcus aureus, antimicrobial peptides, Toll-like receptors, inflammasome, T cells, neutrophils

Introduction

S. aureus is the most common cause of bacterial skin infections in humans, including infections such as impetigo (superficial infection of the epidermis), cellulitis (infection spreading through dermal and subcutaneous tissue planes), ecthyma (deep ulcerative skin infections), folliculitis (infection of hair follicles), furunculosis (deep hair follicle infections also known as boils), and carbuncles (deep communicating furuncles) as well as abscesses, wounds, and ulcers (1-3). Such infections cause between 11 and 14 million outpatient and emergency room visits and nearly 500,000 hospital admissions per year in the U.S. (4, 5). In addition, the inpatient costs for S. aureus skin infections alone range from $3.2-$4.2 billion each year in the U.S. (6). Moreover, community-acquired methicillin-resistant S. aureus (CA-MRSA) strains are causing severe skin infections in healthy people outside of hospitals and becoming increasingly resistant to antibiotics, which is creating a serious public health threat (7, 8).

An important risk factor for S. aureus skin infections is the colonization of mucosa or skin surfaces by S. aureus. By microbiologic culture techniques, S. aureus colonization is found in the anterior nares in ῀30% of the population (and transiently in up to 60-80% of the population), but other sites of colonization (such as the pharynx, rectal mucosa, or the skin in the inguinal region, axillae and peri-rectum) are also common (9, 10). Increased S. aureus skin colonization is also associated with the marked skin inflammation in disease flares of atopic dermatitis (AD) (11, 12), which is a chronic and relapsing inflammatory skin disease affecting 15-30% of children and 5% of adults, resulting in annual healthcare costs of $5.2 billion in the U.S. (13-15). However, the skin inflammation induced by S. aureus in AD was previously thought to be primarily caused by many bacterial-derived factors, such as S. aureus cytolytic toxins that damage cells and superantigens that activate T cells, which result in the production of proinflammatory cytokines and other inflammatory mediators (16-18).

Neutrophil responses are involved in host defense against S. aureus infections, including S. aureus skin infections. Indeed, the formation of a neutrophil abscess is a hallmark of S. aureus infections, and S. aureus is considered a classic pyogenic (pus-forming) infection. The important role of neutrophils is best exemplified by the finding that patients with congenital or acquired defects in neutrophil number or function are highly susceptible to S. aureus skin infections and other invasive infections (3). For neutrophils to function they must be recruited from the bloodstream to the site of the S. aureus infection where they promote their antimicrobial function via phagocytosis of the bacteria in phagosomes. Bacterial killing within the phagosomes is mediated by reactive oxygen species (ROS), antimicrobial peptides, enzymatic digestion and proteins that sequester essential nutrients, as well as via the formation of neutrophil extracellular traps (NETs) (3). Recently, there has been a focus on T cells and how they engage neutrophilic responses for enhanced clearance of S. aureus skin infections.

In an era of declining antibiotic development and rising drug resistance, a greater understanding of the immune responses that protect against S. aureus skin infections is needed to guide future host-directed therapies. This is especially relevant as all conventional antibody-based S. aureus vaccines have failed in clinical trials, including the recent clinical trial against bacteremia/deep sternal wound infections after cardiothoracic surgery in which the vaccinated patients who became infected were 5 times more likely to die (19-24). In addition, since S. aureus skin colonization is highly associated with flares of skin inflammation as in AD, a better understanding of how S. aureus promotes skin inflammation could reveal immune targets to reduce skin inflammation. Recently, there have been significant advances in the cutaneous innate and adaptive immune responses involved in host defense against S. aureus as well as advances in mechanisms by which S. aureus contributes to aberrant skin inflammation, which will be discussed in the review.

Antimicrobial peptides

Antimicrobial peptides (AMPs) are typically less than 50 amino acids in length and have amphipathic structures that bind selectively to negatively charged bacterial membranes to promote osmotic lysis and bacterial cell death. Several types of AMPs that that possess antimicrobial activity against S. aureus are produced by different cell types and play an important role in the innate immune response against S. aureus infections in the skin (Table I). These include β-defensins such as human β-defensin 1 (hBD1) as well as hBD2 and hBD3 (and their mouse orthologs mouse β-defensin 3 [mBD3] and mBD14), which are produced by keratinocytes and myeloid cells and have bacteriostatic and bactericidal activity against S. aureus (25-29). Notably, polymorphisms in hBD1 have been linked with increased S. aureus nasal colonization (30). β-defensins are induced by inflammatory signals such as Toll-like receptors (TLRs, see section below) (31-33), inflammatory cytokines such as IL-1β, IL-17 and IL-22 (34-36), and the activation of growth factor receptors on keratinocytes, as occurs during wound healing (37, 38). Cathelicidin (also called LL-37) is an AMP produced by keratinocytes and myeloid cells (including neutrophils) that has potent antimicrobial activity against S. aureus (27, 39, 40). In humans, cathelicidin production by keratinocytes and monocytes/macrophages is greatly induced by exposure to vitamin D as well as various inflammatory mediators (40-42), and vitamin D deficiency has been linked to an increase in S. aureus skin infections in children (43). Interestingly, a recent report revealed that if a S. aureus skin infection involved the deep dermis, adipocytes were activated to produce cathelicidin, which effectively mediated bacterial clearance (44, 45). Human keratinocytes also produce RNase7 (46, 47) and REG3A (the mouse ortholog Reg3γ), which are also important in providing antimicrobial activity against S. aureus (48, 49). In human sweat, the major AMP produced by eccrine glands is dermcidin, which also has potent activity against S. aureus (50, 51). Many of these AMPs, including hBD2, hBD3 and dermcidin, are decreased in the skin of AD patients, which provides an explanation for the increased S. aureus skin colonization and S. aureus skin infections (especially impetiginization) observed in this disease (27, 50, 52).

Table I.

Antimicrobial peptides with activity against Staphylococcus aureus.

Antimicrobial
peptides (AMPs)
Cell source Antimicrobial
activity
Other
properties
Regulation References
β-defensins (human β defensins [hBDs]) Keratinocytes Macrophages DCs hBDs 1 and 2 have weak bacteriostatic activity hBD3 has strong in vitro antimicrobial activity Chemotaxis of dendritic cells and T cells via CCR2 and CCR6 TLRs and proinflammatory cytokines hBD3: growth factor receptor activation (2529, 5658)
Cathelicidin (LL-37) Keratinocytes Macrophages Neutrophils Eccrine glands Mast cells Adipocytes Antimicrobial activity Chemotaxis of neutrophils, monocytes and T cells via formyl peptide receptors Angiogenesis Wound healing TLRs, inflammatory cytokines, Vitamin D (27, 39, 40, 5961)
RNase7 Keratinocytes Antimicrobial activity TLRs and proinflammatory cytokines (46, 47)
REG3A Keratinocytes Antimicrobial activity (48, 49)
Dermcidin Eccrine glands Antimicrobial activity Constitutive (50, 51)
α-defensin (human neutrophil peptides [HNPs]) Neutrophils Antimicrobial activity Chemotaxis of macrophages, dendritic cells and T cells Constitutive (53, 62)
Calprotectin (S100A8/S100A9) Keratinocytes Neutrophils Sequesters iron, manganese and zinc Constitutive and inducible (54, 55)

In addition to cells that reside in the skin, human neutrophils are rapidly recruited to the site of S. aureus skin infection, and they produce α-defensins (also known as human neutrophil peptides [HNPs]) that are secreted into the neutrophil phagosome to promote antimicrobial activity (53). If S. aureus bacteria enter the cytoplasm of neutrophils, about 50% of the protein fraction of human neutrophils contain calprotectin (S100A8/S100A9) which sequesters iron, manganese and zinc to inhibit bacterial growth (54, 55).

Many of these AMPs not only have antimicrobial activity but also promote chemotaxis of other immune cells. For example, HBDs have chemotactic activity for dendritic cells and T cells via interactions with chemokine receptors CCR2 and CCR6 (56-58); cathelicidin has chemotactic activity for neutrophils, monocytes and T cells via interaction with formyl peptide receptors (59-61); and HNPs have chemotactic activity for macrophages, dendritic cells and T cells (62). Therefore, multiple different AMPs are produced by keratinocytes and stromal and resident immune cells in the skin as well as recruited neutrophils and other immune cells, which all contribute to host defense against S. aureus skin infections.

Pattern recognition receptors

Pattern recognition receptors (PRRs) are host cell receptors that recognize pathogen associated molecular patterns (PAMPs) found on microorganisms, which enable PRRs to recognize foreign microbial components and initiate host defense responses. These PRRs activate different signaling pathways that ultimately result in the production of cytokines, chemokines, adhesion molecules and AMPs involved in host defense. Several PRRs have been implicated in the recognition S. aureus components, especially certain TLRs and nucleotide-binding oligomerization domain-containing protein 2 (NOD2) (Fig. 1). Of the 10 known human TLRs, TLR2 is found on the cell membrane. TLR2 heterodimerizes with TLR1 to recognize triacyl lipopeptides or with TLR6 to recognize diacyl lipopeptides, including S. aureus lipoteichoic acid (which is diacylated), and interacts with co-receptors CD14 and CD36 which impact ligand specificity (63-65). Gene polymorphisms in TLRs 1, 2 and 6 are associated with increased susceptibility to S. aureus skin infections (66), and TLR2-deficient mice have an increased susceptibility to S. aureus skin infections (65, 67, 68). TLRs 1, 2 and 6 are found in the cell membrane of a variety of cell types in the skin, including keratinocytes, Langerhans cells, monocytes/macrophages, dendritic cells, mast cells, endothelial cells, fibroblasts, and adipocytes. Thus, many cell types in the skin can recognize S. aureus lipopeptides and lipoteichoic acid and contribute to cutaneous host defense (69).

Figure 1. Pattern recognition receptors that recognize components of S. aureus.

Figure 1.

Different components of S. aureus are recognized by specific pattern recognition receptors (PRRs) that include the following: TLR1/2 heterodimer that recognizes triacyl lipopeptides and is found on the cell membrane; TLR2/6 heterodimer that recognizes diacyl lipopeptides as well as lipoteichoic acid (LTA) and is found on the cell membrane; NOD2 that recognizes the S. aureus peptidoglycan (PGN) breakdown product muramyl dipeptide (MDP) and is found in the cytoplasm; TLR9 that recognizes CpG motifs of S. aureus DNA and is found in endosomal membranes; and STING that recognizes DNA (cyclic guanosine-adenosine synthase [cGAS]) and is found in the cytoplasm. TLRs and NOD2 lead to inflammatory signaling pathways such as the activation of NF-κB (nuclear factor-κB) that leads to production of proinflammatory cytokines, chemokines, adhesion molecules and antimicrobial peptides involved in cutaneous host defense against S. aureus skin infections. STING has been shown to suppress protective TLR responses against S. aureus skin infections and thus serves as PRR that regulates cutaneous host defense mechanisms.

In addition to TLR2, TLR9 is found in endosomal membranes, recognizes double-stranded DNA, primarily induces a type I interferon response and has also been implicated in host defense against S. aureus infections. Human TLR9 recognizes CpG motifs of S. aureus double-stranded DNA (70). In addition, women with a particular polymorphism in the promoter region of TLR9 resulting in decreased TLR9 expression and persistent S. aureus nasal colonization (71). Recently, it was reported that stimulator of interferon genes (STING), a cytosolic DNA sensor (i.e., cyclic guanosine-adenosine synthase [cGAS]), suppressed TLR-induced genes in macrophages in vitro, and mice deficient in STING had enhanced clearance of an S. aureus skin infection compared with wt mice (72). These findings indicate that STING responds to S. aureus DNA in the cytoplasm and has an opposing role in host defense by downregulating TLR-mediated host defense mechanisms against S. aureus skin infections.

Another important PRR for S. aureus is NOD2 that recognizes the Gram-positive peptidoglycan breakdown product muramyl dipeptide (MDP), including MDP derived from S. aureus peptidoglycan (73). In cultured keratinocytes exposed to S. aureus, inhibition or overexpression of NOD2 resulted in more or less S. aureus survival respectively that correlated with IL-17C expression (74). Furthermore, mice deficient in NOD2 had an increased susceptibility to an S. aureus skin infection, which was mediated in part by reduced expression of IL-6 and IL-1β that impacted the neutrophilic response (67, 75). Taken together, several PRRs, including TLRs 1, 2, 6 in the cell membrane, TLR9 in endosomal membranes, and STING and NOD2 in the cytoplasm, promote and regulate cutaneous host defense against S. aureus skin infections. Importantly, the cellular compartment in which these different PRRs are located and the signaling pathways these PRRs initiate are crucial in how they ultimately contribute to host defense against S. aureus skin infections.

Inflammasomes

Our laboratory and others have found a key role for IL-1β in promoting neutrophil recruitment and host defense against S. aureus skin infections (67, 68, 76-78). Although transcription and translation of pro-IL-1β is induced by PRRs (such as TLR2 and NOD2, as described above), inflammatory cytokines, and other mediators (such as TNF), a second signal typically mediated by inflammasome complexes is required to process pro-IL-1β into active IL-1β (as well as pro-IL-18 to active IL-18) (79-81). These inflammasomes form complexes of proteins in the cytoplasm that undergo massive oligomerization, which ultimately results in release of mature and active IL-1β from the cell and subsequent triggering of IL-1R/MyD88-signaling (79-81). Several reports have found that the NLRP3 (nucleotide binding domain [NOD]-, leucine-rich repeat (LRR)- and pyrin domain-containing 3) inflammasome can be triggered by the creation pores in the cell membrane that lead to potassium (K+) efflux in response to the activity of S. aureus pore-forming toxins (α-, β-, and γ -toxins, and Panton-Valentin leukocidin [PVL]) and prurinergic receptors that recognize ATP produced by damaged cells during an infection (such as P2X7) as well as lysosomal rupture in macrophage cultures in vitro and in mouse models of S. aureus skin infections in vivo (Fig. 2) (77, 78, 82-86). The NLRP3 inflammasome has been shown to be critical for promoting IL-1β activity at the site of a S. aureus skin infection in mice by mediating neutrophil recruitment to the site of infection in the skin (67, 77). Interestingly, NLRP3 inflammasome activity is reduced in lesional AD skin and in keratinocytes in response to Th2 cytokines (i.e., IL-4, IL-5 and IL-13, which are increased in AD affected skin (87)), providing another mechanism for increased S. aureus colonization and S. aureus skin infections in AD.

Figure 2. Mechanisms of inflammasome-mediated production of IL-β production during S. aureus skin infections.

Figure 2.

The production and secretion of IL-1β during S. aureus skin infections requires two signals. Signal 1 involves the transcription of pro-IL-1β in response to PRRs (such as TLRs and NOD2) and proinflammatory cytokines (such as TNF). Signal 2 involves NLRP3 (nucleotide binding domain [NOD]-, leucine-rich repeat (LRR)-, and pyrin domain-containing 3) inflammasome activation, which results formation of these cytoplasmic complexes of proteins, including the inflammasome adaptor protein ASC (apoptosis-associated speck-like protein containing CARD [caspase activation and recruitment domain]) that undergo massive oligomerization ultimately leading to the processing of pro-caspase-1 to activate caspase-1 that cleaves pro-IL-1β into active and mature IL-1β, which then can be secreted from the cell to elicit IL-1R-dependent immune responses. It has been shown that NLRP3 inflammasome activation in response to S. aureus can be triggered by potassium (K+) efflux, which can be initiated by pore-forming toxins (such as α-, β- or γ-hemolysins and Panton-Valentine Leukocidin [PVL]), purinergic receptors (such as P2X7 activated by ATP released by damaged cells during infection) and lysosomal rupture during the intracellular digestion of S. aureus bacteria in phagocytes such as macrophages.

T cells in host defense against S. aureus skin infections

Although enhancing opsonophagocytosis (i.e., antibody-mediated phagocytosis) by inducing high titers of antibodies against bacterial surface components has been a successful vaccination approach for other bacterial pathogens, this has not been the case for S. aureus. Several vaccines targeting opsonophagocytosis by producing antibodies against S. aureus cell surface molecules, including capsular polysaccharides 5 and 8, clumping factor A (ClfA) and iron surface determinant B (IsdB), have all failed in human clinical trials (19-21). Although the reason for these failures is not entirely clear, S. aureus produces many virulence factors to evade antibody-mediated immune responses (88, 89). More recent vaccination attempts have alternatively targeted the production of antibodies that neutralize S. aureus toxins and virulence factors, and such attempts are currently in various stages of preclinical and clinical development and are reviewed elsewhere (90-93).

However, there has been a major focus on T cell responses, as they might provide an alternative strategy to targeting antibody responses for an effective adaptive immune response to protect against S. aureus skin infections and other invasive infections. Several findings in humans have suggested a role for T cells in immunity to S. aureus skin infections. These findings include the following: (i) patients with autosomal dominant hyper-IgE syndrome, who are highly susceptible to recurrent Candida albicans and S. aureus skin infections, were found to have a deficiency in IL-17A- and IL-17F-producing CD4+ T helper cells (i.e., Th17 cells) (94); (ii) rare humans identified with IL-17Ra-deficiency or IL-17F-deficiency who have an increased susceptibility to Candida albicans infections and a lessor susceptibility to S. aureus skin infections (95, 96); (iii) patients with atopic dermatitis who have increased S. aureus skin colonization and S. aureus impetiginization and skin infections have increased Th2 cells and cytokines in their affected skin (16-18); and (iv) patients with HIV disease who are highly susceptible to S. aureus skin infection that is associated with decreased CD4+ T helper cells in their blood (97, 98). Similarly, in mouse models of S. aureus skin infections, IL-17A and IL-17F (primarily produced by γδ T cells) promote neutrophil recruitment, AMP production and increased S. aureus bacterial clearance from the skin (99-103), including a specific role for IL-17A in host defense against repeated S. aureus skin infection exposures (104). Mice deficient in both IL-17A and IL-17F also develop spontaneous mucocutaneous S. aureus infections (105) and are more susceptible to S. aureus nasal colonization (106, 107). However, the findings in humans with primary deficiency disorders such as the aforementioned hyper-IgE syndrome, IL-17Ra-deficiency, IL-1F-deficiency and other disorders with defective IL-17 responses primarily suffer from mucocutaneous candidiasis rather than S. aureus skin infections (108), suggesting that mechanisms other than IL-17 responses are crucial for durable protection against S. aureus skin infections. Notably, protection against S. aureus skin infections in patients with HIV disease was recently found to be more dependent upon IFNγ rather than IL-17 (109). Similarly, mice deficient in both IL-17RA and IFNγR had more severe spontaneous mucocutaneous S. aureus infections than mice deficient in IL-17RA alone (110). Finally, we recently reported that following an initial S. aureus skin infection in mice, clonally expanded γδ T cells in skin draining lymph nodes were capable of mediating long-term memory protection lasting at least 140 days by trafficking from lymph nodes to the infected skin where they produced IFNγ and TNF to enhance the neutrophil response against a subsequent S. aureus skin challenge (111). Therefore, the role of T cells in immunity to S. aureus skin infections likely involves multiple T cell effector cytokines, including IL-17, IFNγ and TNF (Fig. 3), and these T cell responses could serve as more effective immune mechanisms to target in future vaccine efforts.

Figure 3. The role of T cells and effector cytokines in cutaneous host defense against S. aureus skin infections.

Figure 3.

Following the invasion of S. aureus into the skin, components of S. aureus are recognized by pattern recognition receptors (PRRs) expressed by keratinocytes and resident immune cells such as dendritic cells and macrophages as well as rapidly recruited immune cells such as neutrophils. These cells produce IL-1β and other proinflammatory mediators that promote T and B cell immune responses. IL-1β, in particular, has been shown to activate T cells (Th17 cells and γδ T cells) to produce IL-17 (i.e., IL-17A and IL-17F). The T cell effector cytokines IL-17A/F, IFNγ and TNF promote antimicrobial peptide production by keratinocytes as well as the production of chemokines, cytokines, adhesion molecules and granulopoiesis factors that promote neutrophil recruitment from the bloodstream to form an abscess to facilitate bacterial clearance. The B cell responses following a S. aureus skin infection include antibody production that promotes opsonophagocytosis (i.e., antibody-mediated phagocytosis) by monocytes/macrophages and neutrophils to help facilitate S. aureus skin infection clearance.

A novel mechanism by which S. aureus promotes skin inflammation

S. aureus colonization and superficial infection (impetiginization) of the skin is associated with skin inflammation during disease flares of AD, which was previously thought to be caused by to the action of S. aureus toxins and superantigens (16-18). In particular, superantigens of S. aureus have been shown to skew T cell responses towards Th2 cells that produce IL-4 (112). However, our laboratory and another group simultaneously reported that IL-36R (an IL-1R family member activated by IL-36α, IL-36β and IL-36γ that signals via the IL-1R/TLR signaling adapter molecule MyD88) was required to induce skin inflammation following S. aureus exposure to the surface of mouse skin (i.e., epicutaneous exposure) (Fig. 4) (113, 114). The mechanism by which IL-36R promoted skin inflammation involved increased expression of IL-36α in keratinocytes (in part by the activity of the S. aureus-derived toxin phenol-soluble modulin α [PSMα]) that subsequently induced IL-17-producing γδ and CD4+ T cells that were essential for mediating the skin inflammation. These results likely relate to human AD because IL-36α and IL-36γ transcripts were found to be increased in the affected human skin of AD patients (115). In addition, increased IL-17 cytokines and increased numbers of Th17 cells were reported in the skin and peripheral blood from humans with acute AD (116, 117). These findings extend the role of IL-36 cytokines in the skin as a prior study found that humans with loss-of-function mutations in IL-36RN, which encodes the IL-36 receptor antagonist (IL-36Ra) develop generalized pustular psoriasis (118), and IL-36 cytokines were subsequently found to be elevated in other forms of psoriasis (119). It should be mentioned that the anatomical site of S. aureus exposure of the skin was an important determinant in eliciting differential immune responses. As mentioned above, S. aureus epicutaneous exposure in mice resulted in IL-36-mediated skin inflammation (Fig 4) (113, 114). By contrast, exposure of S. aureus in an intradermal infection model in mice resulted in IL-1β-mediated neutrophil recruitment and abscess formation (67, 68, 76-78). Taken together, this newly reported role for IL-36 provides a potentially important mechanism by which S. aureus exposure to the skin surface induces skin inflammation by triggering T cell IL-17 responses (120). Future work will determine whether IL-36 could serve as a target for future biologic therapies to reduce skin inflammation in AD, psoriasis and perhaps other inflammatory skin diseases.

Figure 4. S. aureus induces skin inflammation versus abscess formation depending on the anatomical location of the bacterial exposure in the skin.

Figure 4.

Following S. aureus exposure to the skin surface (i.e., epicutaneous exposure), keratinocytes produce IL-36α (in a mechanism dependent upon S. aureus-derived phenol-soluble modulin α [PSMα]), which triggers γδ T cells and CD4+ T cells (i.e., Th17 cells) to produce IL-17 that subsequently induces skin inflammation. In contrast, following an S. aureus intradermal inoculation, IL-1β is produced by immune cells (especially neutrophils and monocytes/macrophages), which trigger γδ T cells and CD4+ T cells (i.e., Th17 cells) to produce IL-17 that subsequently induce neutrophil and monocyte recruitment to form an abscess to promote bacterial clearance.

Conclusions

S. aureus is an important human skin bacterial pathogen, and CA-MRSA strains are spreading through the normal human population, becoming more resistant to antibiotics and creating a serious public health concern. Recent insights have revealed key roles for antimicrobial peptides, TLRs, NOD2, STING, the NLRP3 inflammasome and T cells (and their effector cytokines IL-17, IFNγ and TNF) in cutaneous host defense against S. aureus skin infections. In addition, S. aureus epicutaneous exposure to the skin resulted in IL-36-mediated, T cell-dependent IL-17 production that induced skin inflammation, providing a newly identified mechanism by which S. aureus induces skin inflammation in AD and potentially in other inflammatory skin diseases. These immune mechanisms might serve as more effective immune targets for the future development of vaccines and immunotherapies against S. aureus skin infections and S. aureusassociated inflammatory skin diseases.

Acknowledgements

This work was supported by grants R01AR073665 and R01AR069502 (to LSM) from the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the United States National Institutes of Health (NIH). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Footnotes

Conflict of Interest

L.S.M. has received grant support from MedImmune, Pfizer, Regeneron Pharmaceuticals, Moderna Therapeutics, and Boehringer Ingelheim, is a shareholder of Noveome Biotherapeutics, and is on the scientific advisory board for Integrated Biotherapeutics, which are all developing vaccines and therapeutics against S. aureus infections and inflammatory skin diseases.

Qi Liu and Momina Mazhar declare they have no conflicts of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Daum RS. Clinical practice. Skin and soft-tissue infections caused by methicillin-resistant Staphylococcus aureus. N Engl J Med 2007;357(4):380–90. 10.1056/NEJMcp070747. [DOI] [PubMed] [Google Scholar]
  • 2.Elston DM. Community-acquired methicillin-resistant Staphylococcus aureus. J Am Acad Dermatol 2007;56(1): 1–16; quiz 7-20. 10.1016/j.jaad.2006.04.018. [DOI] [PubMed] [Google Scholar]
  • 3.Miller LS, Cho JS. Immunity against Staphylococcus aureus cutaneous infections. Nat Rev Immunol 2011;11(8):505–18. 10.1038/nri3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Edelsberg J, Taneja C, Zervos M, Haque N, Moore C, Reyes K, et al. Trends in US hospital admissions for skin and soft tissue infections. Emerg Infect Dis 2009;15(9):1516–8. 10.3201/eid1509.081228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hersh AL, Chambers HF, Maselli JH, Gonzales R. National trends in ambulatory visits and antibiotic prescribing for skin and soft-tissue infections. Arch Intern Med 2008;168(14):1585–91. 10.1001/archinte.168.14.1585. [DOI] [PubMed] [Google Scholar]
  • 6.Suaya JA, Mera RM, Cassidy A, O’Hara P, Amrine-Madsen H, Burstin S, et al. Incidence and cost of hospitalizations associated with Staphylococcus aureus skin and soft tissue infections in the United States from 2001 through 2009. BMC Infect Dis 2014;14:296 10.1186/1471-2334-14-296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.DeLeo FR, Otto M, Kreiswirth BN, Chambers HF. Community-associated meticillin-resistant Staphylococcus aureus. Lancet. 2010;375(9725):1557–68. 10.1016/S0140-6736(09)61999-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Tong SY, Davis JS, Eichenberger E, Holland TL, Fowler VG Jr. Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management. Clin Microbiol Rev 2015;28(3):603–61. 10.1128/CMR.00134-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tong SY, Chen LF, Fowler VG Jr. Colonization, pathogenicity, host susceptibility, and therapeutics for Staphylococcus aureus: what is the clinical relevance? Semin Immunopathol 2012;34(2):185–200. 10.1007/s00281-011-0300-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Yang ES, Tan J, Eells S, Rieg G, Tagudar G, Miller LG. Body site colonization in patients with community-associated methicillin-resistant Staphylococcus aureus and other types of S. aureus skin infections. Clin Microbiol Infect 2010;16(5):425–31. 10.1111/j.1469-0691.2009.02836.x. [DOI] [PubMed] [Google Scholar]
  • 11.Byrd AL, Deming C, Cassidy SKB, Harrison OJ, Ng WI, Conlan S, et al. Staphylococcus aureus and Staphylococcus epidermidis strain diversity underlying pediatric atopic dermatitis. Sci Transl Med 2017;9(397). 10.1126/scitranslmed.aa14651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res 2012;22(5):850–9. 10.1101/gr.131029.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Drucker AM, Wang AR, Li WQ, Sevetson E, Block JK, Qureshi AA. The Burden of Atopic Dermatitis: Summary of a Report for the National Eczema Association. J Invest Dermatol 2017;137(1):26–30. 10.1016/j.jid.2016.07.012. [DOI] [PubMed] [Google Scholar]
  • 14.Weidinger S, Beck LA, Bieber T, Kabashima K, Irvine AD. Atopic dermatitis. Nat Rev Dis Primers. 2018;4(1):1 10.1038/s41572-018-0001-z. [DOI] [PubMed] [Google Scholar]
  • 15.Wesch D, Peters C, Oberg HH, Pietschmann K, Kabelitz D. Modulation of γδ T cell responses by TLR ligands. Cell Mol Life Sci 2011;68(14):2357–70. 10.1007/s00018-011-0699-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hepburn L, Hijnen DJ, Sellman BR, Mustelin T, Sleeman MA, May RD, et al. The complex biology and contribution of Staphylococcus aureus in atopic dermatitis, current and future therapies. Br J Dermatol 2017;177(1):63–71. 10.1111/bjd.15139. [DOI] [PubMed] [Google Scholar]
  • 17.Ong PY, Leung DY. Bacterial and Viral Infections in Atopic Dermatitis: a Comprehensive Review. Clin Rev Allergy Immunol 2016;51(3):329–37. 10.1007/s12016-016-8548-5. [DOI] [PubMed] [Google Scholar]
  • 18.Spaulding AR, Salgado-Pabon W, Kohler PL, Horswill AR, Leung DY, Schlievert PM. Staphylococcal and streptococcal superantigen exotoxins. Clin Microbiol Rev 2013;26(3):422–47. 10.1128/CMR.00104-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Daum RS, Spellberg B. Progress toward a Staphylococcus aureus vaccine. Clin Infect Dis 2012;54(4):560–7. 10.1093/cid/cir828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Fowler VG, Allen KB, Moreira ED, Moustafa M, Isgro F, Boucher HW, et al. Effect of an investigational vaccine for preventing Staphylococcus aureus infections after cardiothoracic surgery: a randomized trial. JAMA. 2013;309(13):1368–78. 10.1001/jama.2013.3010. [DOI] [PubMed] [Google Scholar]
  • 21.Fowler VG Jr., Proctor RA. Where does a Staphylococcus aureus vaccine stand? Clin Microbiol Infect 2014;20 Suppl 5:66–75. 10.1111/1469-0691.12570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.McNeely TB, Shah NA, Fridman A, Joshi A, Hartzel JS, Keshari RS, et al. Mortality among recipients of the Merck V710 Staphylococcus aureus vaccine after postoperative S. aureus infections: an analysis of possible contributing host factors. Hum Vaccin Immunother 2014;10(12):3513–6. 10.4161/hv.34407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Missiakas D, Schneewind O. Staphylococcus aureus vaccines: Deviating from the carol. J Exp Med 2016;213(9):1645–53. 10.1084/jem.20160569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rohrl J, Huber B, Koehl GE, Geissler EK, Hehlgans T. Mouse β-defensin 14 (Defbl4) promotes tumor growth by inducing angiogenesis in a CCR6-dependent manner. J Immunol 2012;188(10)4931–9. 10.4049/jimmunol.1102442. [DOI] [PubMed] [Google Scholar]
  • 25.Harder J, Bartels J, Christophers E, Schroder JM. Isolation and characterization of human β-defensin-3, a novel human inducible peptide antibiotic. J Biol Chem 2001;276(8):5707–13. 10.1074/jbc.M008557200. [DOI] [PubMed] [Google Scholar]
  • 26.Kisich KO, Howell MD, Boguniewicz M, Heizer HR, Watson NU, Leung DY. The constitutive capacity of human keratinocytes to kill Staphylococcus aureus is dependent on β-defensin 3. J Invest Dermatol 2007;127(10):2368–80. 10.1038/sj.jid.5700861. [DOI] [PubMed] [Google Scholar]
  • 27.Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T, et al. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N Engl J Med 2002;347(15):1151–60. 10.1056/NEJMoa021481. [DOI] [PubMed] [Google Scholar]
  • 28.Schroder JM, Harder J. Human β-defensin-2. Int J Biochem Cell Biol. 1999;31(6):645–51. [DOI] [PubMed] [Google Scholar]
  • 29.Zanger P, Holzer J, Schleucher R, Scherbaum H, Schittek B, Gabrysch S. Severity of Staphylococcus aureus infection of the skin is associated with inducibility of human β-defensin 3 but not human β-defensin 2. Infect Immun 2010;78(7):3112–7. 10.1128/IAI.00078-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Nurjadi D, Herrmann E, Hinderberger I, Zanger P. Impaired β-defensin expression in human skin links DEFB1 promoter polymorphisms with persistent Staphylococcus aureus nasal carriage. J Infect Dis 2013;207(4):666–74. 10.1093/infdis/jis735. [DOI] [PubMed] [Google Scholar]
  • 31.Menzies BE, Kenoyer A. Signal transduction and nuclear responses in Staphylococcus aureus-induced expression of human β-defensin 3 in skin keratinocytes. Infect Immun 2006;74(12):6847–54. 10.1128/IAI.00389-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ommori R, Ouji N, Mizuno F, Kita E, Ikada Y, Asada H. Selective induction of antimicrobial peptides from keratinocytes by staphylococcal bacteria. Microb Pathog 2013;56:35–9. https://doi.Org/10.1016/j.micpath.2012.11.005. [DOI] [PubMed] [Google Scholar]
  • 33.Sumikawa Y, Asada H, Hoshino K, Azukizawa H, Katayama I, Akira S, et al. Induction of β-defensin 3 in keratinocytes stimulated by bacterial lipopeptides through toll-like receptor 2. Microbes Infect 2006;8(6):1513–21. https://doi.Org/10.1016/j.micinf.2006.01.008. [DOI] [PubMed] [Google Scholar]
  • 34.Malhotra N, Yoon J, Leyva-Castillo JM, Galand C, Archer N, Miller LS, et al. IL-22 derived from γδ T cells restricts Staphylococcus aureus infection of mechanically injured skin. J Allergy Clin Immunol. 2016;138(4):1098–107 e3. 10.1016/j.jaci.2016.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Minegishi Y, Saito M, Nagasawa M, Takada H, Hara T, Tsuchiya S, et al. Molecular explanation for the contradiction between systemic Th17 defect and localized bacterial infection in hyper-IgE syndrome. J Exp Med 2009;206(6):1291–301. 10.1084/jem.20082767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wang R, Braughton KR, Kretschmer D, Bach TH, Queck SY, Li M, et al. Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat Med 2007;13(12):1510–4. 10.1038/nm1656. [DOI] [PubMed] [Google Scholar]
  • 37.Miller LS, Sorensen OE, Liu PT, Jalian HR, Eshtiaghpour D, Behmanesh BE, et al. TGF-α regulates TLR expression and function on epidermal keratinocytes. J Immunol 2005;174(10):6137–43. [DOI] [PubMed] [Google Scholar]
  • 38.Sorensen OE, Thapa DR, Roupe KM, Valore EV, Sjobring U, Roberts AA, et al. Injury-induced innate immune response in human skin mediated by transactivation of the epidermal growth factor receptor. J Clin Invest 2006;116(7):1878–85. 10.1172/JCI28422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Braff MH, Zaiou M, Fierer J, Nizet V, Gallo RL. Keratinocyte production of cathelicidin provides direct activity against bacterial skin pathogens. Infect Immun 2005;73(10):6771–81. 10.1128/IAI.73.10.6771-6781.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Peric M, Koglin S, Kim SM, Morizane S, Besch R, Prinz JC, et al. IL-17A enhances vitamin D3-induced expression of cathelicidin antimicrobial peptide in human keratinocytes. J Immunol 2008;181(12):8504–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311(5768):1770–3. 10.1126/science.1123933. [DOI] [PubMed] [Google Scholar]
  • 42.Schauber J, Dorschner RA, Coda AB, Buchau AS, Liu PT, Kiken D, et al. Injury enhances TLR2 function and antimicrobial peptide expression through a vitamin D-dependent mechanism. J Clin Invest 2007;117(3):803–11. 10.1172/JCI30142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang JW, Hogan PG, Hunstad DA, Fritz SA. Vitamin D sufficiency and Staphylococcus aureus infection in children. Pediatr Infect Dis J 2015;34(5):544–5. 10.1097/INF.0000000000000667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Miller LS. Adipocytes armed against Staphylococcus aureus. N Engl J Med 2015;372(14):1368–70. 10.1056/NEJMcibr1500271. [DOI] [PubMed] [Google Scholar]
  • 45.••.Zhang LJ, Guerrero-Juarez CF, Hata T, Bapat SP, Ramos R, Plikus MV, et al. Innate immunity. Dermal adipocytes protect against invasive Staphylococcus aureus skin infection. Science. 2015;347(6217):67–71. 10.1126/science.1260972. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This manuscript found that dermal adipocytes produce the antimicrobial peptide cathelicidin to promote clearance of an S. aureus skin infection in the deep dermis.
  • 46.Cho JS, Xuan C, Miller LS. Lucky number seven: RNase 7 can prevent Staphylococcus aureus skin colonization. J Invest Dermatol 2010;130(12):2703–6. 10.1038/jid.2010.294. [DOI] [PubMed] [Google Scholar]
  • 47.Simanski M, Dressel S, Glaser R, Harder J. RNase 7 protects healthy skin from Staphylococcus aureus colonization. J Invest Dermatol 2010;130(12):2836–8. 10.1038/jid.2010.217. [DOI] [PubMed] [Google Scholar]
  • 48.Choi SM, McAleer JP, Zheng M, Pociask DA, Kaplan MH, Qin S, et al. Innate Stat3-mediated induction of the antimicrobial protein Reg3γ is required for host defense against MRSA pneumonia. J Exp Med 2013;210(3):551–61. 10.1084/jem.20120260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lai Y, Li D, Li C, Muehleisen B, Radek KA, Park HJ, et al. The antimicrobial protein REG3A regulates keratinocyte proliferation and differentiation after skin injury. Immunity. 2012;37(1):74–84. https://doi.Org/10.1016/j.immuni.2012.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Rieg S, Steffen H, Seeber S, Humeny A, Kalbacher H, Dietz K, et al. Deficiency of dermcidin-derived antimicrobial peptides in sweat of patients with atopic dermatitis correlates with an impaired innate defense of human skin in vivo. J Immunol 2005;174(12):8003–10. [DOI] [PubMed] [Google Scholar]
  • 51.Steffen H, Rieg S, Wiedemann I, Kalbacher H, Deeg M, Sahl HG, et al. Naturally processed dermcidin-derived peptides do not permeabilize bacterial membranes and kill microorganisms irrespective of their charge. Antimicrob Agents Chemother 2006;50(8):2608–20. 10.1128/AAC.00181-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kisich KO, Carspecken CW, Fieve S, Boguniewicz M, Leung DY. Defective killing of Staphylococcus aureus in atopic dermatitis is associated with reduced mobilization of human β-defensin-3. J Allergy Clin Immunol 2008;122(1):62–8. 10.1016/j.jaci.2008.04.022. [DOI] [PubMed] [Google Scholar]
  • 53.Ericksen B, Wu Z, Lu W, Lehrer RI. Antibacterial activity and specificity of the six human α-defensins. Antimicrob Agents Chemother. 2005;49(1):269–75. https://doi.Org/10.1128/AAC.49.l.269-275.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nakashige TG, Zhang B, Krebs C, Nolan EM. Human calprotectin is an iron-sequestering host-defense protein. Nat Chem Biol 2015;11(10):765–71. 10.1038/nchembio.1891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kehl-Fie TE, Chitayat S, Hood MI, Damo S, Restrepo N, Garcia C, et al. Nutrient metal sequestration by calprotectin inhibits bacterial superoxide defense, enhancing neutrophil killing of Staphylococcus aureus. Cell Host Microbe. 2011; 10(2):158–64. https://doi.Org/10.1016/j.chom.2011.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Rohrl J, Yang D, Oppenheim JJ, Hehlgans T. Human β-defensin 2 and 3 and their mouse orthologs induce chemotaxis through interaction with CCR2. J Immunol 2010;184(12):6688–94. 10.4049/jimmunol.0903984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Niyonsaba F, Iwabuchi K, Matsuda H, Ogawa H, Nagaoka I. Epithelial cell-derived human β-defensin-2 acts as a chemotaxin for mast cells through a pertussis toxin-sensitive and phospholipase C-dependent pathway. Int Immunol 2002;14(4):421–6. [DOI] [PubMed] [Google Scholar]
  • 58.Yang D, Chertov O, Bykovskaia SN, Chen Q, Buffo MJ, Shogan J, et al. β-defensins: linking innate and adaptive immunity through dendritic and T cell CCR6. Science. 1999;286(5439):525–8. [DOI] [PubMed] [Google Scholar]
  • 59.Chen K, Xiang Y, Huang J, Gong W, Yoshimura T, Jiang Q, et al. The formylpeptide receptor 2 (Fpr2) and its endogenous ligand cathelin-related antimicrobial peptide (CRAMP) promote dendritic cell maturation. J Biol Chem 2014;289(25):17553–63. 10.1074/jbc.M113.535674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.De Y, Chen Q, Schmidt AP, Anderson GM, Wang JM, Wooters J, et al. LL-37, the neutrophil granule- and epithelial cell-derived cathelicidin, utilizes formyl peptide receptor-like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and T cells. J Exp Med 2000;192(7):1069–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Niyonsaba F, Iwabuchi K, Someya A, Hirata M, Matsuda H, Ogawa H, et al. A cathelicidin family of human antibacterial peptide LL-37 induces mast cell chemotaxis. Immunology. 2002;106(1):20–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Grigat J, Soruri A, Forssmann U, Riggert J, Zwirner J. Chemoattraction of macrophages, T lymphocytes, and mast cells is evolutionarily conserved within the human α-defensin family. J Immunol 2007;179(6):3958–65. [DOI] [PubMed] [Google Scholar]
  • 63.Nilsen NJ, Deininger S, Nonstad U, Skjeldal F, Husebye H, Rodionov D, et al. Cellular trafficking of lipoteichoic acid and Toll-like receptor 2 in relation to signaling: role of CD14 and CD36. J Leukoc Biol 2008;84(1):280–91. 10.1189/jlb.0907656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Triantafilou M, Gamper FG, Haston RM, Mouratis MA, Morath S, Hartung T, et al. Membrane sorting of toll-like receptor (TLR)-2/6 and TLR2/1 heterodimers at the cell surface determines heterotypic associations with CD36 and intracellular targeting. J Biol Chem 2006;281(41):31002–11. 10.1074/jbc.M602794200. [DOI] [PubMed] [Google Scholar]
  • 65.Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, et al. CD36 is a sensor of diacylglycerides. Nature. 2005;433(7025):523–7. 10.1038/nature03253. [DOI] [PubMed] [Google Scholar]
  • 66.Stappers MH, Thys Y, Oosting M, Plantinga TS, Ioana M, Reimnitz P, et al. TLR1, TLR2, and TLR6 gene polymorphisms are associated with increased susceptibility to complicated skin and skin structure infections. J Infect Dis 2014;210(2):311–8. 10.1093/infdis/jiu080. [DOI] [PubMed] [Google Scholar]
  • 67.Cho JS, Guo Y, Ramos RI, Hebroni F, Plaisier SB, Xuan C, et al. Neutrophil-derived IL-1β is sufficient for abscess formation in immunity against Staphylococcus aureus in mice. PLoS Pathog. 2012;8(11):el003047 10.1371/journal.ppat.1003047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Miller LS, O’Connell RM, Gutierrez MA, Pietras EM, Shahangian A, Gross CE, et al. MyD88 mediates neutrophil recruitment initiated by IL-1R but not TLR2 activation in immunity against Staphylococcus aureus. Immunity. 2006;24(1):79–91. https://doi.Org/10.1016/j.immuni.2005.11.011. [DOI] [PubMed] [Google Scholar]
  • 69.Miller LS. Toll-like receptors in skin. Adv Dermatol 2008;24:71–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Pohar J, Yamamoto C, Fukui R, Cajnko MM, Miyake K, Jerala R, et al. Selectivity of Human TLR9 for Double CpG Motifs and Implications for the Recognition of Genomic DNA. J Immunol 2017;198(5):2093–104. 10.4049/jimmunol.1600757. [DOI] [PubMed] [Google Scholar]
  • 71.Nurjadi D, Heeg K, Weber ANR, Zanger P. Toll-like receptor 9 (TLR-9) promotor polymorphisms and gene expression are associated with persistent Staphylococcus aureus nasal carriage. Clin Microbiol Infect 2018. https://doi.Org/10.1016/j.cmi.2018.02.014. [DOI] [PubMed] [Google Scholar]
  • 72. •.Scumpia PO, Botten GA, Norman JS, Kelly-Scumpia KM, Spreafico R, Ruccia AR, et al. Opposing roles of Toll-like receptor and cytosolic DNA-STING signaling pathways for Staphylococcus aureus cutaneous host defense. PLoS Pathog. 2017;13(7):e1006496 10.1371/journal.ppat.1006496. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This study demonstrated that the DNA sensor STING functions to suppress protective TLR-mediated immune responses against an S. aureus skin infection.
  • 73.Muller-Anstett MA, Muller P, Albrecht T, Nega M, Wagener J, Gao Q, et al. Staphylococcal peptidoglycan co-localizes with Nod2 and TLR2 and activates innate immune response via both receptors in primary murine keratinocytes. PLoS One. 2010;5(10):e13153 10.1371/journal.pone.0013153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Roth SA, Simanski M, Rademacher F, Schroder L, Harder J. The pattern recognition receptor NOD2 mediates Staphylococcus aureus-induced IL-17C expression in keratinocytes. J Invest Dermatol 2014;134(2):374–80. 10.1038/jid.2013.313. [DOI] [PubMed] [Google Scholar]
  • 75.Hruz P, Zinkernagel AS, Jenikova G, Botwin GJ, Hugot JP, Karin M, et al. NOD2 contributes to cutaneous defense against Staphylococcus aureus through α-toxin-dependent innate immune activation. Proc Natl Acad Sci U S A. 2009;106(31):12873–8. 10.1073/pnas.0904958106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cho JS, Zussman J, Donegan NP, Ramos RI, Garcia NC, Uslan DZ, et al. Noninvasive in vivo imaging to evaluate immune responses and antimicrobial therapy against Staphylococcus aureus and USA300 MRSA skin infections. J Invest Dermatol 2011;131(4):907–15. 10.1038/jid.2010.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Miller LS, Pietras EM, Uricchio LH, Hirano K, Rao S, Lin H, et al. Inflammasome-mediated production of IL-1β is required for neutrophil recruitment against Staphylococcus aureus in vivo. J Immunol 2007;179(10):6933–42. [DOI] [PubMed] [Google Scholar]
  • 78.Munoz-Planillo R, Franchi L, Miller LS, Nunez G. A critical role for hemolysins and bacterial lipoproteins in Staphylococcus aureus-induced activation of the Nlrp3 inflammasome. J Immunol 2009;183(6):3942–8. 10.4049/jimmunol.0900729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Franchi L, Munoz-Planillo R, Nunez G. Sensing and reacting to microbes through the inflammasomes. Nat Immunol 2012;13(4):325–32. 10.1038/ni.2231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Guo H, Callaway JP, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med 2015;21(7):677–87. 10.1038/nm.3893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–32. https://doi.Org/10.1016/j.cell.2010.01.040. [DOI] [PubMed] [Google Scholar]
  • 82.Craven RR, Gao X, Allen IC, Gris D, Bubeck Wardenburg J, McElvania-Tekippe E, et al. Staphylococcus aureus α-hemolysin activates the NLRP3-inflammasome in human and mouse monocytic cells. PLoS One. 2009;4(10):e7446 10.1371/journal.pone.0007446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Franchi L, Kanneganti TD, Dubyak GR, Nunez G. Differential requirement of P2X7 receptor and intracellular K+ for caspase-1 activation induced by intracellular and extracellular bacteria. J Biol Chem 2007;282(26): 18810–8. 10.1074/jbc.M610762200. [DOI] [PubMed] [Google Scholar]
  • 84.Holzinger D, Gieldon L, Mysore V, Nippe N, Taxman DJ, Duncan JA, et al. Staphylococcus aureus Panton-Valentine leukocidin induces an inflammatory response in human phagocytes via the NLRP3 inflammasome. J Leukoc Biol 2012;92(5):1069–81. https://doi.Org/10.1189/jlb.0112014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440(7081):228–32. 10.1038/nature04515. [DOI] [PubMed] [Google Scholar]
  • 86.Shimada T, Park BG, Wolf AJ, Brikos C, Goodridge HS, Becker CA, et al. Staphylococcus aureus evades lysozyme-based peptidoglycan digestion that links phagocytosis, inflammasome activation, and IL-1β secretion. Cell Host Microbe. 2010;7(1):38–49. https://doi.Org/10.1016/j.chom.2009.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Niebuhr M, Baumert K, Heratizadeh A, Satzger I, Werfel T. Impaired NLRP3 inflammasome expression and function in atopic dermatitis due to Th2 milieu. Allergy. 2014;69(8):1058–67. 10.1111/all.12428. [DOI] [PubMed] [Google Scholar]
  • 88.Spaan AN, Surewaard BG, Nijland R, van Strijp JA. Neutrophils versus Staphylococcus aureus: a biological tug of war. Annu Rev Microbiol 2013;67:629–50. https://doi.Org/10.1146/annurev-micro-092412-155746. [DOI] [PubMed] [Google Scholar]
  • 89.Thammavongsa V, Kim HK, Missiakas D, Schneewind O. Staphylococcal manipulation of host immune responses. Nat Rev Microbiol 2015; 13(9):529–43. 10.1038/nrmicro3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Aman MJ, Adhikari RP. Staphylococcal bicomponent pore-forming toxins: targets for prophylaxis and immunotherapy. Toxins (Basel). 2014;6(3):950–72. 10.3390/toxins6030950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Anderson AS, Miller AA, Donald RG, Scully IL, Nanra JS, Cooper D, et al. Development of a multicomponent Staphylococcus aureus vaccine designed to counter multiple bacterial virulence factors. Hum Vaccin Immunother 2012;8(11):1585–94. 10.4161/hv.21872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bagnoli F Staphylococcus aureus toxin antibodies: Good companions of antibiotics and vaccines. Virulence. 2017;8(7): 1037–42. 10.1080/21505594.2017.1295205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Rouha H, Badarau A, Visram ZC, Battles MB, Prinz B, Magyarics Z, et al. Five birds, one stone: neutralization of α-hemolysin and 4 bi-component leukocidins of Staphylococcus aureus with a single human monoclonal antibody. MAbs. 2015;7(1):243–54. 10.4161/19420862.2014.985132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Milner JD, Brenchley JM, Laurence A, Freeman AF, Hill BJ, Elias KM, et al. Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature. 2008;452(7188):773–6. 10.1038/nature06764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Levy R, Okada S, Beziat V, Moriya K, Liu C, Chai LY, et al. Genetic, immunological, and clinical features of patients with bacterial and fungal infections due to inherited IL-17RA deficiency. Proc Natl Acad Sci USA. 2016;113(51):E8277–E85. 10.1073/pnas.1618300114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Puel A, Cypowyj S, Bustamante J, Wright JF, Liu L, Lim HK, et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science. 2011;332(6025):65–8. 10.1126/science.1200439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Graber CJ, Jacobson MA, Perdreau-Remington F, Chambers HF, Diep BA. Recurrence of skin and soft tissue infection caused by methicillin-resistant Staphylococcus aureus in a HIV primary care clinic. J Acquir Immune Defic Syndr 2008;49(2):231–3. 10.1097/QAI.0b013e318183a947. [DOI] [PubMed] [Google Scholar]
  • 98.Vyas KJ, Shadyab AH, Lin CD, Crum-Cianflone NF. Trends and factors associated with initial and recurrent methicillin-resistant Staphylococcus aureus (MRSA) skin and soft-tissue infections among HIV-infected persons: an 18-year study. J Int Assoc Provid AIDS Care. 2014;13(3):206–13. 10.1177/2325957412473780. [DOI] [PubMed] [Google Scholar]
  • 99.Chan LC, Chaili S, Filler SG, Barr K, Wang H, Kupferwasser D, et al. Nonredundant Roles of Interleukin-17A (IL-17A) and IL-22 in Murine Host Defense against Cutaneous and Hematogenous Infection Due to Methicillin-Resistant Staphylococcus aureus. Infect Immun 2015;83(11):4427–37. 10.1128/IAI.01061-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cho JS, Pietras EM, Garcia NC, Ramos RI, Farzam DM, Monroe HR, et al. IL-17 is essential for host defense against cutaneous Staphylococcus aureus infection in mice. J Clin Invest 2010; 120(5):1762–73. 10.1172/JCI40891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Maher BM, Mulcahy ME, Murphy AG, Wilk M, O’Keeffe KM, Geoghegan JA, et al. Nlrp-3-driven interleukin 17 production by γδ T cells controls infection outcomes during Staphylococcus aureus surgical site infection. Infect Immun 2013;81(12):4478–89. 10.1128/IAI.01026-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Myles IA, Fontecilla NM, Valdez PA, Vithayathil PJ, Naik S, Belkaid Y, et al. Signaling via the IL-20 receptor inhibits cutaneous production of IL-1β and IL-17A to promote infection with methicillin-resistant Staphylococcus aureus. Nat Immunol 2013;14(8):804–11. 10.1038/ni.2637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Tkaczyk C, Hamilton MM, Datta V, Yang XP, Hilliard JJ, Stephens GL, et al. Staphylococcus aureus alpha toxin suppresses effective innate and adaptive immune responses in a murine dermonecrosis model. PLoS One. 2013;8(10):e75103 10.1371/journal.pone.0075103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Montgomery CP, Daniels M, Zhao F, Alegre ML, Chong AS, Daum RS. Protective immunity against recurrent Staphylococcus aureus skin infection requires antibody and interleukin-17A. Infect Immun 2014;82(5):2125–34. 10.1128/IAI.01491-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ishigame H, Kakuta S, Nagai T, Kadoki M, Nambu A, Komiyama Y, et al. Differential roles of interleukin-17A and -17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity. 2009;30(1):108–19. https://doi.Org/10.1016/j.immuni.2008.11.009. [DOI] [PubMed] [Google Scholar]
  • 106. •.Archer NK, Adappa ND, Palmer JN, Cohen NA, Harro JM, Lee SK, et al. Interleukin-17A (IL-17A) and IL-17F Are Critical for Antimicrobial Peptide Production and Clearance of Staphylococcus aureus Nasal Colonization. Infect Immun 2016;84(12):3575–83. 10.1128/IAI.00596-16. [DOI] [PMC free article] [PubMed] [Google Scholar]; • This paper demonstrates that IL-17 responses promote antimicrobial peptide production and neutrophil recruitment to promote clearance of S. aureus nasal colonization.
  • 107.Archer NK, Harro JM, Shirtliff ME. Clearance of Staphylococcus aureus nasal carriage is T cell dependent and mediated through interleukin-17A expression and neutrophil influx. Infect Immun 2013;81(6):2070–5. 10.1128/IAI.00084-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Patel DD, Kuchroo VK. Th17 Cell Pathway in Human Immunity: Lessons from Genetics and Therapeutic Interventions. Immunity. 2015;43(6):1040–51. https://doi.Org/10.1016/j.immuni.2015.12.003. [DOI] [PubMed] [Google Scholar]
  • 109. ••.Utay NS, Roque A, Timmer JK, Morcock DR, DeLeage C, Somasunderam A, et al. MRSA Infections in HIV-Infected People Are Associated with Decreased MRSA-Specific Th1 Immunity. PLoS Pathog 2016;12(4):el005580 10.1371/journal.ppat.1005580. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated that protection against S. aureus skin infections in patients with HIV disease were more dependent upon IFNγ than IL-17 responses, providing key evidence in humans that durable protection against S. aureus skin infections involves T cell cytokines other than IL-17.
  • 110.Barin JG, Talor MV, Schaub JA, Diny NL, Hou X, Hoyer M, et al. Collaborative Interferon-γ and Interleukin-17 Signaling Protects the Oral Mucosa from Staphylococcus aureus. Am J Pathol 2016;186(9):2337–52. https://doi.Org/10.1016/j.ajpath.2016.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. ••.Dillen CA, Pinsker BL, Marusina AI, Merleev AA, Farber ON, Liu H, et al. Clonally expanded γδ T cells protect against Staphylococcus aureus skin reinfection. J Clin Invest 2018;128(3):1026–42. 10.1172/JCI96481. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper was the first to demonstrate that a clonal population of γδ T cells expanded following an initial S. aureus skin infection and provided a long-term immunological memory response against a secondary S. aureus skin infection via production of IFNtγ and TNF that enhanced neutrophil recruitment to the skin.
  • 112.Laouini D, Kawamoto S, Yalcindag A, Bryce P, Mizoguchi E, Oettgen H, et al. Epicutaneous sensitization with superantigen induces allergic skin inflammation. J Allergy Clin Immunol 2003;112(5):981–7. 10.1016/j.jaci.2003.07.007. [DOI] [PubMed] [Google Scholar]
  • 113.••.Liu H, Archer NK, Dillen CA, Wang Y, Ashbaugh AG, Ortines RV, et al. Staphylococcus aureus Epicutaneous Exposure Drives Skin Inflammation via IL-36-Mediated T Cell Responses. Cell Host Microbe. 2017;22(5):653–66 e5. https://doi.Org/10.1016/j.chom.2017.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.••.Nakagawa S, Matsumoto M, Katayama Y, Oguma R, Wakabayashi S, Nygaard T, et al. Staphylococcus aureus Virulent PSMα Peptides Induce Keratinocyte Alarmin Release to Orchestrate IL-17-Dependent Skin Inflammation. Cell Host Microbe. 2017;22(5):667–77 e5. https://doi.Org/10.1016/j.chom.2017.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]; References 113 and 114 were the first to demonstrate that S. aureus epicutaneous exposure induced IL-36α production by keratinocytes that stimulated T cells to produce IL-17 to promote skin inflammation. This represents a previously unrecognized mechanism by which S. aureus promotes cutaneous inflammation that likely relates to disease flares of atopic dermatitis and potentially other inflammatory skin diseases.
  • 115. •.Suarez-Farinas M, Ungar B, Correa da Rosa J, Ewald DA, Rozenblit M, Gonzalez J, et al. RNA sequencing atopic dermatitis transcriptome profiling provides insights into novel disease mechanisms with potential therapeutic implications. J Allergy Clin Immunol 2015;135(5):1218–21. 10.1016/j.jaci.2015.03.003. [DOI] [PubMed] [Google Scholar]; This manuscript demonstrated by RNA-sequencing that transcripts for IL-36α and IL-36γ are increased in the affected skin from atopic dermatitis patients, providing evidence that IL-36 responses are involved in the pathogenesis of atopic dermatitis in humans.
  • 116.Koga C, Kabashima K, Shiraishi N, Kobayashi M, Tokura Y. Possible pathogenic role of Th17 cells for atopic dermatitis. J Invest Dermatol 2008;128(11):2625–30. 10.1038/jid.2008.111. [DOI] [PubMed] [Google Scholar]
  • 117.Toda M, Leung DY, Molet S, Boguniewicz M, Taha R, Christodoulopoulos P, et al. Polarized in vivo expression of IL-11 and IL-17 between acute and chronic skin lesions. J Allergy Clin Immunol 2003;111(4):875–81. [DOI] [PubMed] [Google Scholar]
  • 118.Marrakchi S, Guigue P, Renshaw BR, Puel A, Pei XY, Fraitag S, et al. Interleukin-36-receptor antagonist deficiency and generalized pustular psoriasis. N Engl J Med 2011;365(7):620–8. 10.1056/NEJMoa1013068. [DOI] [PubMed] [Google Scholar]
  • 119.Johnston A, Xing X, Wolterink L, Barnes DH, Yin Z, Reingold L, et al. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J Allergy Clin Immunol 2017;140(1):109–20. 10.1016/j.jaci.2016.08.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Williams MR, Nakatsuji T, Gallo RL. Staphylococcus aureus: Master Manipulator of the Skin. Cell Host Microbe. 2017;22(5):579–81. 10.1016/j.chom.2017.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES