Next Article in Journal
The Role of Point-of-Care C-Reactive Protein Testing in Antibiotic Prescribing for Respiratory Tract Infections: A Survey among Swiss General Practitioners
Previous Article in Journal
Validation and Application of an HPLC-UV Method for Routine Therapeutic Drug Monitoring of Dalbavancin
Previous Article in Special Issue
Mechanisms of Resistance to Macrolide Antibiotics among Staphylococcus aureus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection

1
Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
2
Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
3
Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(5), 542; https://doi.org/10.3390/antibiotics11050542
Submission received: 24 March 2022 / Revised: 13 April 2022 / Accepted: 18 April 2022 / Published: 19 April 2022

Abstract

:
Staphylococcus aureus (S. aureus) causes a broad range of infections and is associated with significant morbidity and mortality. S. aureus produces a diverse range of cellular and extracellular factors responsible for its invasiveness and ability to resist immune attack. In recent years, increasing resistance to last-line anti-staphylococcal antibiotics daptomycin and vancomycin has been observed. Resistant strains of S. aureus are highly efficient in invading a variety of professional and nonprofessional phagocytes and are able to survive inside host cells. Eliciting immune protection against antibiotic-resistant S. aureus infection is a global challenge, requiring both innate and adaptive immune effector mechanisms. Dendritic cells (DC), which sit at the interface between innate and adaptive immune responses, are central to the induction of immune protection against S. aureus. However, it has been observed that S. aureus has the capacity to develop further antibiotic resistance and acquire increased resistance to immunological recognition by the innate immune system. In this article, we review the strategies utilised by S. aureus to circumvent antibiotic and innate immune responses, especially the interaction between S. aureus and DC, focusing on how this relationship is perturbed with the development of antibiotic resistance.

1. Introduction

S. aureus is mainly found living on the skin and mucus membranes of the anterior nares of most healthy human populations (20–80%) [1,2]. Under normal circumstances, S. aureus maintains a symbiotic relationship with the host but becomes virulent in deeper tissue sites accessed through trauma or parenteral route of infection [3]. Staphylococcal bacteraemia is the leading cause of human morbidity and mortality in hospital settings [4]. It is responsible for various diseases ranging from mild skin infections (impetigo, folliculitis) to more invasive infections (wound infections, endocarditis, osteomyelitis) and toxin-mediated diseases (toxic shock syndrome, scaled skin syndrome) [5]. Pathogenic strains of S. aureus are also associated with respiratory tract infections that range from asymptomatic colonisation to necrotizing pneumonia [6]. This necrotizing pneumonia is a progressive form of pneumonia causing respiratory distress, pleural effusion, haemoptysis, and leukopenia [7]. Disease progression in staphylococcal pneumonia is mainly mediated by the excessive and dysregulated host inflammatory response to infection causing lung injury [8,9].
S. aureus infection is of high socioeconomic burden in both developed and developing nations [10]. The emergence of ‘super bugs’ that have shown resistance to multiple antibiotics, leading to increased reliance on last-line antibiotics such as vancomycin and daptomycin, and that have shown an ability to simultaneously avoid host innate immunity [4,11] further complicates treatment strategies of S. aureus infections. Infections caused by daptomycin-resistant S. aureus are particularly more prolonged and difficult to treat [4,11,12]. Analyses of antibiotic-resistant S. aureus-host innate immune interactions may help to discover the fine details of pathogenesis of infections involving these bacterial strains and to develop effective therapeutic options.

2. Innate Sensing of S. aureus

The innate immune response is the first line of defence against invading pathogens. The innate immune system is comprised of diverse cells, proteins, and receptor systems that recognise pathogens and initiate a sequence of events that result in the production and secretion of inflammatory mediators [12]. The subsequent recruitment and activation of phagocytic cells to the infection site leads to a reduction in pathogen load and the downstream initiation of adaptive immunity [13,14]. Phagocytic cells of the innate immune system include neutrophils, macrophages, monocytes, and dendritic cells (DC). These cells are equipped to recognise evolutionarily conserved pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) through overlapping and cell-type-specific expression of pattern recognition receptors (PRRs) [15]. Major subtypes of PRR that have direct effects on initiating innate immunity are present on the cell surface, intracellular compartments, and in the cytosol. These include the toll-like receptors (TLRs), the retinoic acid inducible gene I-like receptors (RLRs), nucleotide-binding oligomerisation domain (NOD)-like receptors (NLR), C-type lectin receptors (CLR), and DNA sensing molecules such as cyclic GMP-AMP (cGAMP) synthase (cGAS) and stimulator of interferon genes (STING) (Table 1) [16,17].

2.1. TLRs

TLRs are the best-characterised pattern recognition receptors [16]. They have greatly advanced our understanding of how the body senses pathogen invasion, triggers innate immune responses, and primes antigen-specific adaptive immunity [15,18]. All TLRs except TLR3 utilise the MyD88-dependent signalling pathway that leads to nuclear factor-κB (NF-κB) activation and inflammatory responses. MyD88-deficient macrophages release much less cytokine than wild-type macrophages after staphylococcal infection [19]. Moreover, people with MyD88 deficiency have a high incidence of recurrent invasive and noninvasive S. aureus infections [41], highlighting the importance of TLR signalling for immunity to this bacterial strain. The roles of different TLRs in sensing PAMPS of S. aureus are outlined in this review (Table 1). TLR4, which classically recognises endotoxin, is not included in Table 1 since it is likely not involved in sensing of gram-positive bacteria such as S. aureus. However, recently, it has been demonstrated that S. aureus virulence factor Phenol-soluble modulin (PSM) α1–α3 inhibited HMGB1-induced NF-κB activation by binding with TLR4 in HEK-Blue hTLR4 and THP-1 cells [42]. Whether this interaction is definitively endotoxin-free remains to be elucidated.
TLR2 can recognise a wide range of PAMPs derived from bacteria, fungi, parasites, and viruses [20] and generally forms heterodimers with TLR1 or TLR6 [18]. TLR2 is considered critical for both systemic and localised S. aureus infection as it is highly expressed on resident macrophages and recruited neutrophils and monocytes [21]. Studies on TLR2−/− mice have shown that TLR2 is required for survival, abundant cytokine secretion, and effective clearance of S. aureus from the brain [22]. Even though TLR2 has not been reported to function as a phagocytic receptor, its absence is reported to cause a decrease in S. aureus phagocytosis [43,44,45]. Although others have reported that bacterial internalisation was not affected in the absence of the MyD88 adaptor in macrophages, suggesting that any role for TLR2 in facilitating bacterial uptake is independent of MyD88-dependent signalling [46].
TLR2/TLR6 heterodimers recognise diacylated lipopeptides and peptidoglycans from S. aureus [18]. Lipoteichoic acid (LTA) is a major outer cell wall component of S. aureus. LTA is thought to be recognised by TLR2 in cooperation with CD36 [47]. Indeed, loss of response to LTA via inborn defects in signalling downstream of TLR2 has been shown to lead to susceptibility to severe staphylococcal disease [48]. Of interest, antibodies to LTA can protect against severe staphylococcal disease in individuals with defects in the TLR2 signalling pathway [48].
Endosomal TLR7, 8 (human), 9, and 13 (murine only) sense the presence of single-stranded RNA (ssRNA), DNA, and ribosomal RNA (rRNA), respectively. Studies have shown that single deficiencies in TLR7, 9, or 13 have little or no effect in host defences in S. aureus infection models [23]. However, the combined deficiency of TLR7, 9, and 13 in macrophages resulted in the complete inhibition of interleukin (IL)-12 p70 and interferon (IFN)-β production, and increased susceptibility to both cutaneous and systemic S. aureus infection [23]. A strong TLR9-mediated Type I IFN response to DNA containing unmethylated CpG motifs of S. aureus was demonstrated in murine bone-marrow-derived DC [27]. In human monocytes, TLR7 and TLR8 can sense S. aureus tRNA and ssRNA degradation products, leading to IFN-β production [24,25,26]. Recently, it has been shown that TLRs 3, 7, and 9 all contribute to the sensing of S. aureus-released microvesicles by RAW 264.7 cells [24]. Overall, endosomal sensing of S. aureus nucleic acids does appear to play a role in innate sensing of S. aureus but the reliance on a single endosomal TLR is less clear, with likely differences between different cell types.

2.2. C-Type Lectin Receptor

C-type lectin receptors (CLRs) are a heterogenous group of transmembrane proteins, many of which are expressed on myeloid cells [49]. CLRs possess one or more carbohydrate recognition domains (CRDs) which mediate binding to its carbohydrate ligand. In addition, soluble/circulating CLRs exist that bind an array of carbohydrate patterns on pathogen surfaces [28].
The C-type lectin (CLEC) receptor langerin (CD207, also called CLEC4K) is expressed on Langerhans cells of the skin and subsets of conventional DC in mouse and human [50]. Human CD207 binds β-N-acetylglucosamine (GlcNAc) of teichoic acid of the S. aureus cell wall (WTA), inducing activation and innate cytokine responses by the DC subsets [51]. Certain strains of S. aureus express structurally different WTA, which is instead recognised by macrophage galactose-type lectin receptor (MGL; CD301), expressed by macrophages and DC, also inducing proinflammatory immune responses [52].
Mannose binding lectin (MBL) is a serum glycoprotein of humans and animals that is involved in opsonisation of complement on the surface of S. aureus through binding to carbohydrate linkages of WTA [53,54]. In humans, low levels of MBL and/or MBL2 polymorphism are responsible for increased susceptibility to S. aureus bacteraemia [55]. The MBL–WTA interaction is a particularly important innate defence mechanism against S. aureus infection in infants, which becomes redundant upon induction of antibodies against WTA [56]. Mice lacking MBL exhibit reduced proinflammatory cytokine production during S. aureus infection, indicating a role for MBL in regulating cytokine release [29]. MBL complexes with TLR2 within phagosomes and the cooperation amplifies the host response to S. aureus [30].
Many other CLRs have been implicated in uptake and recognition of bacteria [57]. It is likely that other CLRs interact with S. aureus, potentially dependent on host cell type and differences in lipid moieties present in different bacterial strains or cell wall changes induced upon acquisition of antibiotic resistance.

2.3. Cytosolic Sensors

2.3.1. NOD-like Receptors (NLR)

NLRs are located in the cytosol and respond to a range of both PAMPs and DAMPs [31]. They are multidomain proteins composed of a variable N-terminal effector region and C-terminal leucine-rich repeats (LRRs) that sense PAMPs [32]. N-terminal effector regions consist of caspase recruitment domain (CARD), pyrin domain (PYD), acidic domain, and a centrally located NOD domain. Based on their N-terminal domain, NLRs are divided into four subfamilies: NLRA, NLRB, NLRC, and NLRP [31]. The activated NLRs show various functions that can be divided into four broad categories: inflammasome formation, signalling transduction, transcription activation, and autophagy [58].
Mice deficient in the NLRC family member NOD2 have increased susceptibility to S. aureus infection and a significantly higher bacterial tissue burden. Their reduced viability is due in part to loss of NOD2 leading to defective neutrophil phagocytosis and reduced proinflammatory cytokines IL-1β and IL-6 and/or antimicrobial peptides, e.g., β-defensin production, against S. aureus infection [31,33].
S. aureus α, β-, and γ- hemolysins; Panton–Valentine leucocidin (PVL) [59]; and leucocidin A/B [60] can activate the NLRP3 inflammasome in monocytes and macrophages resulting in maturation and secretion of IL-1β and IL-18 [61,62,63]. In human macrophages, the NLRP7 inflammasome is induced in response to microbial acylated lipopeptides and promotes ASC-dependent caspase-1 activation, IL-1β and IL-18 maturation, and restriction of intracellular bacterial replication [64].

2.3.2. RIG-I-like Receptors Family

The retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) family consists of three DExD/H box RNA helicases located in the cytosol: retinoic acid-inducible gene (RIG-I), melanoma differentiation-associated gene 5 (MDA-5), and laboratory of genetics and physiology-2 (LGP-2) [65,66]. Both RIG-I and MDA-5 have tandem N-terminal caspase activation and recruitment domains (CARDs), a DExD/H box RNA helicase domain, and a C-terminal repressor domain (RD). Unlike RIG-I and MDA-5, LGP-2 contains only the RNA helicase domain and acts as a negative regulator of the other RLRs [67,68,69,70,71]. During infection, RIG-I dimerizes in an ATP-dependent manner [72] and the activated multimeric form of RIG-I or MDA5 then interacts with the downstream adaptor protein mitochondrial antiviral signalling protein (MAVS, also known as CARDIF and IPS-1) [36,68,73]. Ligation of RIG-I or MDA5 with MAVS activates the IKK-related kinase, TANK binding kinase 1 (TBK1), which further activates interferon regulatory factor (IRF)3/IRF7, resulting in the transcription of type I interferons [73,74]. MAVS also activates the NF-κB pathway through recruitment of TRADD, FADD, caspase-8, and caspase-10 [75,76].
RLRs are key sensors of virus infection that recognises intracellular RNA introduced to the cytosol [65,77]. Although RIG-I is known to recognise viral RNA, several studies have indicated the role of RIG-1 in the detection of bacterial RNA or DNA in a host cell type or pathogen specific manner [34,78,79,80,81,82]. Recently, it has been demonstrated that a human microglial cell line and primary astrocytes express RIG-I and stimulate IFN production via RIG-I-dependent signalling in the presence of S. aureus genomic DNA [34]. In another study, it was observed that the levels of MDA5 and RIG-1 mRNA were significantly increased in septicaemia patients infected with S. aureus when compared with healthy controls—whether there is a link to protective immunity during septicaemia is unclear [83].

2.3.3. cGAS–STING Pathway

Cytosolic DNA sensing by the cyclic GMP–AMP synthase (cGAS)–stimulator of interferon genes (STING) pathway is implicated in host immune defence mechanisms. cGAS senses viral, bacterial, or host dsDNA aberrantly localised in the cytosol [84] and promotes cGAS oligomerisation and activation [85,86]. Activated cGAS catalyses the formation of cyclic dinucleotides (CDN) from ATP and GTP to the CDN 2′3′-cGAMP [87]. CDNs such as cyclic di-AMP or cyclic di-GMP can also be produced from bacteria and activate STING directly [36]. CDNs bind to the endoplasmic reticulum (ER)-localised STING, which promotes STING dimerization and translocation from the ER to golgi compartments [36,37] where STING recruits and activates TBK1 and nuclear translocation of the transcription factor IRF3) and to a lesser extent, NF-κB) [38], leading to the production of type 1 IFNs and many other inflammatory cytokines. Type I IFN induction is a critical component of innate immune response to several bacterial pathogens including S. aureus [40].
In a murine pulmonary S. aureus infection model, STING deficiency resulted in increased mortality and a higher bacterial burden in the lungs and bronchoalveolar lavage (BAL) fluid, alongside a severe destruction of lung architecture [88]. However, STING deficiency did not worsen pulmonary inflammation during the early stage of infection [88]. Another study that implicated cGAS/STING signalling in S. aureus infection showed that S. aureus infection promoted IFN-β mRNA expression and TBK1/IRF3-dependent production of IFN-β in murine macrophages [40]. Production of IFN-β has been reported to be protective against S. aureus infection, as IFN-β is required to turn on downstream genes important for local inflammatory responses [89]. RNA-sequencing of bone-marrow-derived macrophages (BMDMs) treated with live S. aureus showed that approximately 95% of the induced genes within the first four hours were mostly regulated by TLR and STING signalling pathways [39]. This study has also implicated STING, but not TLR, in the type I IFN response with live, but not killed, S. aureus [39]. Overall, although the studies are limited outside of mouse, cGAS/STING signalling appears to be an important player in innate immune responses to S. aureus.

3. Key Cells of the Innate Immune Response to S. aureus Infection

3.1. Keratinocytes (KCs)

KCs form the outermost barrier of the epidermis and are considered nonprofessional phagocytes, which are typically the first cells that encounter pathogens [21,90]. They recognise PAMPs via PRRs, scavenger receptors CD36, and MARCO, and subsequently signal to induce activation of transcription factors NF-κB, AP-1, and CREB, stimulating production of cytokines, chemokines, antimicrobial peptides, and inducible nitric oxide (NO) synthase [21,90,91,92]. KCs are important in immune responses to prevent S. aureus skin infections since they are able to phagocytose S. aureus and mediate caspase-1-dependent clearance through the activation of an inflammatory programmed cell death pathway, pyroptosis [93].

3.2. Macrophages

Macrophages are professional phagocytes and are critical for the clearance of invading pathogens from the site of infection. In macrophages, staphylococcal lipoproteins induce proinflammatory cytokines, NO, and reactive oxygen species (ROS) [94,95,96,97]. In a murine model of peritoneal infection, the particulate form of the staphylococcal cell envelope activated macrophages by inducing the production of chemotactic cytokines [98]. In S. aureus sepsis, increased microbial load and mortality was noticed in mice lacking macrophages [99]. Similar responses were also observed in murine airway infection models [100,101] and zebrafish [102], where loss of macrophages leads to increased S. aureus load and loss of normal alveolar structures. The antimicrobial peptide calprotectin present in monocytes and early macrophages successfully inhibits S. aureus growth in a mouse abscess model by reducing bioavailability of metal ions, starving the bacteria from essential nutrients [103,104,105].
In humans, Extracellular Vesicles (EVs) released by S. aureus are phagocytosed by macrophages and trigger NLRP3 inflammasome activation [106]. Another study showed that S. aureus infection induces a caspase-dependent pyroptotic cell death pathway in human macrophages by inhibiting mTORC1/STAT3 [107]. It is reported that upon phagocytosis by human macrophages, S. aureus persists in intracellular vacuoles for 3–4 days before escaping into the cytosol [108]. Treatment of human macrophages with interferon-γ at concentrations equivalent to human therapeutic doses at this stage is able to eliminate intracellular staphylococci [108].

3.3. Neutrophils

Neutrophils are capable of eliminating bacteria by producing nonspecific antimicrobial molecules. In a wide variety of tissues and organs, defects in neutrophil number or function result in an increased susceptibility to S. aureus infections [109,110,111,112]. Neutrophil priming is mediated by bacterial products including peptidoglycan and cytolytic toxins [113,114], inducing increased adhesion, phagocytosis, superoxide production, and degranulation cytotoxic granules [115]. For efficient killing and degradation, neutrophils utilise a combination of NADPH oxidase-derived reactive oxygen species (ROS), cytotoxic granule components, antimicrobial peptides, and neutrophil extracellular traps (NETs) [116,117]. In response to live S. aureus in humans, neutrophils can release NETs that trap the bacteria independent of ROS [118,119]. Human neutrophils are also efficient in phagocytizing biofilm-associated S. aureus in vitro [119,120]. However, the degree of clearance is dependent on the maturation state of the biofilm [120].

3.4. Dendritic Cells

DCs are the most potent antigen-presenting cell and are pivotal in the generation of adaptive immunity. Steinman and Cohn discovered the cell and applied the term “dendritic cells” based on their unique morphology [121]. Immature dendritic cells are present in all tissues including lymphoid and nonlymphoid organs [122]. Upon detecting ‘danger signals’, immature DCs are recruited to the sites of inflammation in peripheral tissues [123,124]. DCs subsequently take up antigen, migrate to lymphoid organs, and mature in the process [124]. Expression of chemokine receptors CCR6 and CCR7 also increase during maturation, which allows them to migrate to blood or lymph node to present the antigen [125,126] to prime naive T cells and initiate primary T-cell-mediated immune responses [127,128]. During maturation, DCs acquire an enhanced capacity to accumulate peptides, major histocompatibility complex (MHC) class I and II molecules, costimulatory molecules (such as CD40, CD80, and CD86), and soluble antigens (such as CD83 and DC-LAMP) [128,129].
DCs represent a heterogeneous population of cells and can be broadly divided into four categories based on surface marker expression and functional specialisation. The first two subsets, including the cross-presenting cDC1 and helper T-cell priming cDC2, comprise conventional DC (cDC) subsets [130,131] and are grouped into two categories based on their differential dependence on transcription factors for development [131]. The third DC subset includes the inflammatory monocyte-derived DCs (MoDCs), which are primarily involved in driving inflammation during infection and presenting antigen to T-cells [132]. Lastly, plasmacytoid DC (pDC) form the fourth functional group, which are involved in antiviral cytokine secretion and initiating innate immune responses [133].
While there is a body of literature focussing on the molecular mechanisms regulating innate recognition of S. aureus by DC, much of it is either assumed from DC receptor expression and their known ligands [134] or findings of S. aureus interactions with other phagocytes [119,135]. The latter tends to be obtained from either monocyte-inducible DCs or macrophage models that do not recapitulate primary DC subsets [136,137,138]. Especially lacking is literature pertaining to the role of primary cDC subsets in response to S. aureus, with few direct experiments investigating cDC subsets and their function either in vitro or ex vivo.
Study on BDCA1(+) myeloid DCs (mDCs) indicated that they can induce both innate and adaptive immune response against S. aureus [139]. In this study, BDCA1(+) mDCs phagocytosed S. aureus and upregulated expression of costimulatory markers MHC classes I and II, and increased production of proinflammatory cytokines and gamma interferon (IFN-γ) by CD4 and CD8 T cells [139]. Parcina and colleagues [140] examined the molecular mechanisms regulating the innate recognition of S. aureus by human pDC, demonstrating that activation is dependent on sensing of endosomal nucleic acids via TLR7 and TLR9 [140]. Therefore, it has been demonstrated that human pDCs can directly respond to S. aureus and produce tumour necrosis factor (TNF-α), IL-6, IFN-α, and upregulate CD86 expression [141]. Collectively, these observations have important implications for the immunoregulation and suppression of the host during clinical infection.
In murine models, it is clear that cDCs are important players in the recognition of S. aureus143. Indeed, cDC-deficient mice (CD11c-DTR transgenic) exhibit increased bacterial loads, accelerated mortality, and more severe pathology than controls [142]. Furthermore, S. aureus stimulation elicits cDC-dependent IL-12p70 [142], suggesting a possible role for cDC1 in Th1 induction. Nonetheless, these findings should be treated with caution due to the nonspecific ablation of other CD11c expressing cells, including monocytes, macrophages, NK, and B-cells in the CD11c-DTR model [143].
More recently, Richardson and colleagues [144] demonstrated a role for the S. aureus phenol soluble modulins in altering the Th balance in a murine infection model [144]. This study demonstrated the development of less stimulatory DCs, inducing the reduction in both Th1 and Th17 numbers, but increase in T-regs, in response to S. aureus lab strain USA300 expressing PSM, when compared with knockout strains [144].
In terms of DC pathogenesis, S. aureus is known to induce DC toxicity, with decreased numbers of murine splenic cDC1 and cDC2 during infection [144]. Recently, it has been shown that the toxicity of S. aureus leucocidins in monocyte-derived DCs are primarily induced through the action of pore forming leucocidin and leukocidin AB [145]. However, given the choice of monocyte-derived DCs for this research, these findings may not necessarily be reflective of primary DC subsets. Of note, S. aureus has also been shown to survive and replicate following internalisation by CD11c+ splenocytes in vitro [142], comprising a mixture of cDC, macrophages, and their precursors [131]. It is therefore clear that further work is required to understand the nuances of S. aureus immunopathogenesis, especially so with regard to distinct primary DC subsets.

4. Strategies of S. aureus to Evade Immune Defence Mechanisms

S. aureus has the capacity to survive even in the presence of an antigen-specific adaptive immune response [146]. It possesses several mechanisms to manipulate humoral or T-cell-mediated immune responses by secreting specific proteins or superantigens [146]. The remainder of this review will focus on the different immune evasion strategies used by S. aureus to modulate innate immune defences.

4.1. Inhibition of Pattern Recognition Pathways

Superantigen-like proteins (SSLs) are one of the major immune evasion molecules produced by S. aureus that interfere with a variety of innate immune defences [147,148]. The role of SSLs in downregulating early recognition signals through PRR and contributing to pathogenesis has been shown in a large number of studies [149,150,151,152,153]. SSL3 blocked binding of bacterial lipopeptides to the extracellular domain of TLR2 and prevented the formation of TLR2–TLR1 and TLR2–TLR6 heterodimers [153,154]. SSL3 has also been shown to block TLR2-mediated secretion of TNF-α in murine macrophages [153].
The TLRs and IL-1Rs superfamily have a conserved cytoplasmic region, known as the Toll/IL-1 receptor (TIR) domain [154,155,156]. The intracellular TIR domain can interact with a variety of TIR-containing adaptor proteins (MyD88, TIRAP, TRIF, TRAM, and TRAF6) and activate downstream signalling pathways [156,157]. The gene encoding a homologue of the human TIR domain TIR-containing protein (TirS) was identified in the mobile genetic element of ‘staphylococcal chromosomal cassettes’ (SCC) in S. aureus lab strain MSSA476 [156,158]. TirS in these S. aureus strains was localised within SCC, together with genes encoding fusidic acid resistance and genes providing resistance to methicillin. This protein exerted a specific inhibitory effect against TLR2-mediated NF-κB activation, JNK phosphorylation, and production of proinflammatory cytokines [156]. TirS was reported to be present in 12% of S. aureus (both Methicillin-sensitive and -resistant S. aureus) strains, and found to downregulate the NF-κB pathway through inhibition of not only TLR2, but also TLR4, TLR5, and TLR9 [159]. A comparison of pathogenicity of the MSSA476 wild-type strain vs. its isogenic tirS mutant in an intravenous mouse infection model revealed that presence of TirS increased the bacterial load in multiple organs [156].

4.2. Altering Secretion of Pro-Inflammatory Cytokines

Another staphylococcal virulence factor, adenosine synthase A (AdsA), catalyses the hydrolysis of adenosine monophosphate (AMP) to adenosine and increases the overall abundance of extracellular adenosine. Interaction of adenosine with its receptor triggers anti-inflammatory signalling cascades that cause the inhibition of platelet aggregation [160], cellular degranulation [161], IL-1α (IL-1) production, and increased production of the anti-inflammatory cytokine IL-10 [162]. Adenosine also impacts antigen presentation by decreasing the expression of MHC-II on macrophages [163] and DCs [164]. Further, adenosine reduces macrophage production of IL-12, which is a pivotal stimulus for Th1-type immune responses [165]. Thus, AdsA contributes to an overall downregulation of the innate immune response, whilst directly modulating the activation of downstream adaptive immunity.

4.3. Inhibition of Intracellular Degradation

S. aureus employs an array of mechanisms to interfere with normal host endosomal acidification, which is required for the clearance of the pathogen and the induction of an adaptive immune response. In immature bone-marrow-derived DCs, incomplete assembly of the S. aureus enzyme V-ATPase results in reduced acidity of host phagosomes [166]. Similarly, urease secreted by S. aureus increases endosomal pH by converting ammonia to urea [167]. Intracellular acidification has been shown to generate endogenous reactive oxygen species and mediate S. aureus cell death [168,169,170], whereas pH homeostasis mediated by urease protects cells from endogenous ROS in the presence of urea [167]. In the phagolysosomes, staphylococcal cascade proteins SodA, SodM, and KatA detoxify O2- into H2O2 and reduce the bacterial cell damage [171,172]. S. aureus also encodes accessory gene regulator (agr)-dependent factor(s) that induces autophagosome formation but prevents autophagolysosomal fusion events [173]. It has also been demonstrated that intracellular S. aureus within an autophagosome phosphorylates mitogen-activated protein kinase 14 (MAPK14) and autophagy-related 5 (ATG5) in murine fibroblasts, preventing autophagosome maturation and, therefore, bacterial degradation [174,175]. S. aureus can further escape into the cytosol from autophagosomes, where it proliferates and induces caspase-independent host cell death [173].
As elucidated for the gram-negative bacterium Klebsiella pneumoniae [176], S. aureus has also been shown to diminish macrophage efferocytosis and instead induce programmed necroptosis of neutrophils that have phagocytosed and/or been infected with S. aureus [177]. The secretory virulence factor alpha toxin of CA-MRSA has been implicated in impairing neutrophil efferocytosis by alveolar macrophages during lung infection [178]. This impaired clearance of neutrophils thereby delays clearance of the bacteria within the neutrophils, increases lung damage, and potentiates the capacity of antibiotic-resistant S. aureus to colonise the lung [178].
S. aureus produces a wide range of cellular and extracellular factors that helps them escape intracellular degradation and antigen presentation [179,180]. Resistance of S. aureus to host lysozymes is mediated by O-acetyltransferase A (OatA), which adds an acetyl group to N-acetylmuramic acid in the peptidoglycan of the cell wall [181]. The acetyl group causes steric hindrance, preventing the binding of lysozyme to peptidoglycan [182]. S. aureus produces PSM, which can form membrane pores in the phagosome facilitating escape from destruction and host antigen processing pathways [183,184,185]. PSMs can also activate the p38-CREB pathway in bone-marrow-derived DCs and reduces TLR2 signaling [186,187]. The ess-encoded virulence factor EsxA prevents host apoptosis, therefore impeding clearance, while EsxB reduces cytokine production by primary human DCs [188]. Host cell death is considered important to limit the dissemination of intracellular pathogens by revealing them to extracellular immune surveillance mechanisms [189,190]. Several studies showed that S. aureus can persist in human infection and serve as a reservoir for recurrent infection [191,192,193,194,195].

5. Development of Antibiotic Resistance and Evasion of Immune Recognition

The emergence of Penicillin-resistant S. aureus in the late 1940s resulted in the development of a semisynthetic penicillin (Methicillin) [196]. In the early 1960s, Methicillin-resistant S. aureus (MRSA) was reported and then spread worldwide. MRSA is now endemic in health care facilities in virtually all industrialised countries [197]. Community-associated MRSA (CA-MRSA) appeared in the 1990s and is currently a major problem in many countries worldwide [197,198]. Unlike healthcare-associated MRSA infections, CA-MRSA typically causes disease in healthy individuals. S. aureus has the capacity to acquire antibiotic resistance [199,200,201,202] by bacterial gene mutations and horizontal transfer of resistance genes from external sources [201,203,204]. Resistance to methicillin is mediated by the mecA gene carried on a distinct mobile genetic element, SCCmec [203]. mecA gene encodes a penicillin-binding protein, PBP2a, which is intrinsically resistant to inhibition by β-lactams [203,204,205]. Although SCCmec is central for antibiotic resistance, there is no direct evidence that SCCmec plays a clear role in MRSA virulence [206].
The acquisition of MRSA resistance to the last-line antibiotics vancomycin and daptomycin has been recorded over the last 15 years [207,208,209]. The mechanisms of daptomycin resistance (DAP-R) in S. aureus are generally thought to be associated with an overall change in the net charge of the bacterial membrane that results in the repulsion of the DAP antibiotic molecule from the cell surface [210,211]. The gene mprF is most consistently associated with the development of DAP-R in S. aureus both in vivo and in vitro [210,212,213,214]. Several studies have demonstrated that mutation in mprF is responsible for the cross-resistance of MRSA to DAP and Vancomycin (VCM) [215,216,217]. Other genes implicated in the development of DAP-R in S. aureus are those involved in phospholipid metabolism [210]. Importantly, S. aureus (and other Gram-positive organisms) harbor two (or more) cls genes (cls1 and cls2) and changes in this enzyme play a role in DAP-R by altering the pool of PG and Cardiolipin (CL) in the bacterial CM [218,219]. Another gene involved in phospholipid metabolism and DAP-R phenotype is pgsA, which encodes a CDP-diacylglycerol-glycerol-3-phosphate 3-phosphatidyltransferase [210]. Genomic analysis performed by Peleg and colleagues found that mutations in pgsA were often observed in DAP-R laboratory derivatives but not in DAP-R clinical isolates [211]. These mutations likely impair the enzymatic activity and thus decrease the overall pool of PG.
Interestingly, DAP-R clinical isolates of S. aureus have been shown to be significantly less virulent than susceptible parent strains following daptomycin therapy and clinical failure [220]. Cameron and colleagues [220] demonstrated that these resistant clinical isolates are not only less virulent, lending an increased rate of bacterial survival in murine challenge models, but that these strains also persist for longer in vivo than their DAP-susceptible parent strains [220].

6. Interplay between Antibiotic Resistance and Innate Immunity

With the mutations conferring antibiotic resistance being well-characterised, growing evidence is emerging regarding a duality in function, whereby antibiotic resistance mutations further modulate immune effector mechanisms (Figure 1).
Perhaps most well-characterised is that of the multipeptide resistance factor (MprF), which is an evolutionarily conserved protein expressed by various bacterial species including S. aureus [223,231,232]. The MprF protein attenuates the negative charge of anionic phospholipids in the bacterial cell membrane [233], serving to protect bacteria from the action of several host antimicrobial peptides including defensins, kinocidins, cathelicidins, and other cationic antimicrobial peptides [223,234,235]. Additionally, MprF is a flippase for Lys-PG and is required for translocation of lysinylated phospholipids to the outer leaflet of the membrane [236]. As previously discussed, gain of function mutations in MprF are associated with reduced daptomycin susceptibility. Importantly, DAP-R strains containing such point mutations have further demonstrated cross-resistance to three host cationic antimicrobial peptides [231]. While it remains unclear if these mutations are driven by host or antibiotic selective pressures (or some combination thereof), it is clear that antibiotic resistance mutations are able to simultaneously confer resistance to innate immune effectors.
Indeed, it has been demonstrated that the acquisition of specific point mutations in cls2, and, partly, mprF, correspond to a significant reduction in the secretion of proinflammatory cytokines including TNFα, interleukin-6 (IL-6), and macrophage inflammatory protein-1β (MIP-1β) as well as expression of CD80 by cDCs in response to daptomycin-resistant MRSA [225]. In vitro study of S. aureus also demonstrated that exposure to daptomycin alone or in combination with vancomycin or oxacillin (compared with vancomycin or oxacillin alone) led to a downregulation of TLR1, TLR2, TLR6, and TLR9 expression and dampened macrophage inflammatory response with diminished TNFα secretion and reduced synthesis of Nitric oxide (NO) [229,230]. In another in vivo study of Jiang et al., the cls2 point mutations allowed S. aureus to evade neutrophil chemotaxis and represented a metabolic strategy used by S. aureus to circumvent antibiotic and immune attack [11]. Mishra et al. found that development of DAP-R in rabbits with MRSA prosthetic joint infection is associated with resistance to host defence cationic peptides (HDP-R) [232].
Aside from directly interfering with innate effectors, S. aureus has the remarkable capacity to also accumulate antibiotic resistance mutations that simultaneously mask ligands of critical innate sensors. Increased biofilm formation capacity in hospitalised patients by MRSA compared with Methicillin Sensitive S. aureus (MSSA) strains is considered an important virulence factor influencing its persistence in both the environment and the host [234]. In addition, biofilms of MRSA attenuate the expression of IL-1β, TNF-α, CXCL2, and CCL2, circumventing TLR2 and TLR9 recognition pathways and macrophage invasion into the biofilm [235].
Indeed, it has been shown that specific mutations in the 23S rRNA of S. aureus confer resistance to macrolide, lincosamide, and streptogramin group antibiotics, whilst simultaneously preventing detection through TLR13 [224]. TLR13 is not present in humans, and whether these mutations are also able to evade human recognition of S. aureus 23S rRNA remain to be confirmed. The notion that bacteria can change throughout the course of infection to evade innate immune detection is indeed novel, and these findings set an important precedent for further investigation.

7. Concluding Remarks

The innate immune response is the key to the prevention of infection and generation of adaptive immunity. Initial sensing of bacteria through PRR initiates the inflammatory cascade to release cytokines, antimicrobial peptides, and recruit antigen presenting DCs to further activate adaptive immune responses. However, S. aureus possesses a large repertoire of virulence factors including antibiotic resistance genes that can lead to suboptimal innate immune responses, opening a window to establish and sustain infection. The precise mechanisms defining how antibiotic resistance mutations affect the bacterial isolate’s ability to evade innate immune activation is of clear importance. Understanding how resistant S. aureus modulates innate cells, including DCs, to escape recognition is critical to restore immunogenic potential and to allow recognition and clearance.

Author Contributions

N.J. and T.P. drafted manuscript. N.J. conceived Figure 1. M.O. reviewed and restructured the manuscript and all authors contributed to final editing. All authors have read and agreed to the published version of the manuscript.

Funding

M.O. received funding from Australian Research Council DP210103122, NHMRC: 1077633 and The CASS Foundation Ref 7319. N.J. was funded by a Monash University Faculty International Tuition Scholarship and co-funded Graduate Scholarship. T.P. was supported by an Australian Government Research Training Program (RTP) Stipend.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brown, A.F.; Leech, J.M.; Rogers, T.R.; McLoughlin, R.M. Staphylococcus aureus colonization: Modulation of host immune response and impact on human vaccine design. Front. Immunol. 2014, 4, 507. [Google Scholar] [CrossRef] [PubMed]
  2. Wertheim, H.F.; Melles, D.C.; Vos, M.C.; van Leeuwen, W.; van Belkum, A.; Verbrugh, H.A.; Nouwen, J.L. The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect. Dis. 2005, 5, 751–762. [Google Scholar] [CrossRef]
  3. Kluytmans, J.; van Belkum, A.; Verbrugh, H. Nasal carriage of Staphylococcus aureus: Epidemiology, underlying mechanisms, and associated risks. Clin. Microbiol. Rev. 1997, 10, 505–520. [Google Scholar] [CrossRef] [PubMed]
  4. Fowler, V.G., Jr.; Boucher, H.W.; Corey, G.R.; Abrutyn, E.; Karchmer, A.W.; Rupp, M.E.; Levine, D.P.; Chambers, H.F.; Tally, F.P.; Vigliani, G.A.; et al. Daptomycin versus standard therapy for bacteremia and endocarditis caused by Staphylococcus aureus. N. Engl. J. Med. 2006, 355, 653–665. [Google Scholar] [CrossRef] [Green Version]
  5. Howden, B.P.; Davies, J.K.; Johnson, P.D.; Stinear, T.P.; Grayson, M.L. Reduced vancomycin susceptibility in Staphylococcus aureus, including vancomycin-intermediate and heterogeneous vancomycin-intermediate strains: Resistance mechanisms, laboratory detection, and clinical implications. Clin. Microbiol. Rev. 2010, 23, 99–139. [Google Scholar] [CrossRef] [Green Version]
  6. Parker, D.; Prince, A. Immunopathogenesis of Staphylococcus aureus pulmonary infection. Semin. Immunopathol. 2012, 34, 281–297. [Google Scholar] [CrossRef] [Green Version]
  7. Gillet, Y.; Issartel, B.; Vanhems, P.; Fournet, J.-C.; Lina, G.; Bes, M.; Vandenesch, F.; Piémont, Y.; Brousse, N.; Floret, D.; et al. Association between Staphylococcus aureus strains carrying gene for Panton-Valentine leukocidin and highly lethal necrotising pneumonia in young immunocompetent patients. Lancet 2002, 359, 753–759. [Google Scholar] [CrossRef]
  8. Hayashida, A.; Bartlett, A.H.; Foster, T.J.; Park, P.W. Staphylococcus aureus beta-toxin induces lung injury through syndecan-1. Am. J. Pathol. 2009, 174, 509–518. [Google Scholar] [CrossRef] [Green Version]
  9. Rastogi, D.; Ratner, A.J.; Prince, A. Host–bacterial interactions in the initiation of inflammation. Paediatr. Respir. Rev. 2001, 2, 245–252. [Google Scholar] [CrossRef]
  10. Balasubramanian, D.; Harper, L.; Shopsin, B.; Torres, V.J. Staphylococcus aureus pathogenesis in diverse host environments. Pathog. Dis. 2017, 75, ftx005. [Google Scholar] [CrossRef] [Green Version]
  11. Jiang, J.H.; Bhuiyan, M.S.; Shen, H.H.; Cameron, D.R.; Rupasinghe, T.W.T.; Wu, C.M.; Le Brun, A.P.; Kostoulias, X.; Domene, C.; Fulcher, A.J.; et al. Antibiotic resistance and host immune evasion in Staphylococcus aureus mediated by a metabolic adaptation. Proc. Natl. Acad. Sci. USA 2019, 116, 3722–3727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chaplin, D.D. Overview of the immune response. J. Allergy Clin. Immunol. 2003, 111, S442–S459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Fournier, B.; Philpott, D.J. Recognition of Staphylococcus aureus by the innate immune system. Clin. Microbiol. Rev. 2005, 18, 521–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Steinman, R.M. The dendritic cell system and its role in immunogenicity. Annu. Rev. Immunol. 1991, 9, 271–296. [Google Scholar] [CrossRef] [PubMed]
  15. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Krishnaswamy, J.K.; Chu, T.; Eisenbarth, S.C. Beyond pattern recognition: NOD-like receptors in dendritic cells. Trends Immunol. 2013, 34, 224–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Palm, N.W.; Medzhitov, R. Pattern recognition receptors and control of adaptive immunity. Immunol. Rev. 2009, 227, 221–233. [Google Scholar] [CrossRef]
  18. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef]
  19. Takeuchi, O.; Hoshino, K.; Akira, S. Cutting edge: TLR2-deficient and MyD88-deficient mice are highly susceptible to Staphylococcus aureus infection. J. Immunol. 2000, 165, 5392–5396. [Google Scholar] [CrossRef] [Green Version]
  20. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [Green Version]
  21. Brandt, S.L.; Putnam, N.E.; Cassat, J.E.; Serezani, C.H. Innate immunity to Staphylococcus aureus: Evolving paradigms in soft tissue and invasive infections. J. Immunol. 2018, 200, 3871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Stenzel, W.; Soltek, S.; Sanchez-Ruiz, M.; Akira, S.; Miletic, H.; Schlüter, D.; Deckert, M. Both TLR2 and TLR4 are required for the effective immune response in Staphylococcus aureus-induced experimental murine brain abscess. Am. J. Pathol. 2008, 172, 132–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lentini, G.; Famà, A.; De Gaetano, G.V.; Galbo, R.; Coppolino, F.; Venza, M.; Teti, G.; Beninati, C. Role of endosomal TLRs in Staphylococcus aureus infection. J. Immunol. 2021, ji2100389. [Google Scholar] [CrossRef] [PubMed]
  24. Rodriguez, B.V.; Kuehn, M.J. Staphylococcus aureus secretes immunomodulatory RNA and DNA via membrane vesicles. Sci. Rep. 2020, 10, 18293. [Google Scholar] [CrossRef] [PubMed]
  25. Moen, S.H.; Ehrnström, B.; Kojen, J.F.; Yurchenko, M.; Beckwith, K.S.; Afset, J.E.; Damås, J.K.; Hu, Z.; Yin, H.; Espevik, T.; et al. Human Toll-like Receptor 8 (TLR8) is an important sensor of pyogenic bacteria, and is attenuated by cell surface TLR signaling. Front. Immunol. 2019, 10, 1209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Jöckel, S.; Nees, G.; Sommer, R.; Zhao, Y.; Cherkasov, D.; Hori, H.; Ehm, G.; Schnare, M.; Nain, M.; Kaufmann, A.; et al. The 2′-O-methylation status of a single guanosine controls transfer RNA-mediated Toll-like receptor 7 activation or inhibition. J. Exp. Med. 2012, 209, 235–241. [Google Scholar] [CrossRef]
  27. Parker, D.; Prince, A. Staphylococcus aureus induces type I IFN signaling in dendritic cells via TLR9. J. Immunol. 2012, 189, 4040–4046. [Google Scholar] [CrossRef] [Green Version]
  28. Zizzari, I.G.; Napoletano, C.; Battisti, F.; Rahimi, H.; Caponnetto, S.; Pierelli, L.; Nuti, M.; Rughetti, A. MGL receptor and immunity: When the ligand can make the difference. J. Immunol. Res. 2015, 2015, 450695. [Google Scholar] [CrossRef] [Green Version]
  29. Shi, L.; Takahashi, K.; Dundee, J.; Shahroor-Karni, S.; Thiel, S.; Jensenius, J.C.; Gad, F.; Hamblin, M.R.; Sastry, K.N.; Ezekowitz, R.A.B. Mannose-binding lectin-deficient mice are susceptible to infection with Staphylococcus aureus. J. Exp. Med. 2004, 199, 1379–1390. [Google Scholar] [CrossRef] [Green Version]
  30. Ip, W.K.E.; Takahashi, K.; Moore, K.J.; Stuart, L.M.; Ezekowitz, R.A.B. Mannose-binding lectin enhances Toll-like receptors 2 and 6 signaling from the phagosome. J. Exp. Med. 2008, 205, 169–181. [Google Scholar] [CrossRef] [Green Version]
  31. Kim, Y.K.; Shin, J.S.; Nahm, M.H. NOD-Like receptors in infection, immunity, and diseases. Yonsei Med. J. 2016, 57, 5–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Franchi, L.; Warner, N.; Viani, K.; Nuñez, G. Function of Nod-like receptors in microbial recognition and host defense. Immunol. Rev. 2009, 227, 106–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Deshmukh, H.S.; Hamburger, J.B.; Ahn, S.H.; McCafferty, D.G.; Yang, S.R.; Fowler, V.G., Jr. Critical role of NOD2 in regulating the immune response to Staphylococcus aureus. Infect. Immun. 2009, 77, 1376–1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Johnson, M.B.; Halman, J.R.; Burmeister, A.R.; Currin, S.; Khisamutdinov, E.F.; Afonin, K.A.; Marriott, I. Retinoic acid inducible gene-I mediated detection of bacterial nucleic acids in human microglial cells. J. Neuroinflamm. 2020, 17, 139. [Google Scholar] [CrossRef]
  35. Sun, Q.; Sun, L.; Liu, H.H.; Chen, X.; Seth, R.B.; Forman, J.; Chen, Z.J. The specific and essential role of MAVS in antiviral innate immune responses. Immunity 2006, 24, 633–642. [Google Scholar] [CrossRef] [Green Version]
  36. Marinho, F.V.; Benmerzoug, S.; Oliveira, S.C.; Ryffel, B.; Quesniaux, V.F.J. The emerging roles of STING in bacterial infections. Trends Microbiol. 2017, 25, 906–918. [Google Scholar] [CrossRef]
  37. Dobbs, N.; Burnaevskiy, N.; Chen, D.; Gonugunta, V.K.; Alto, N.M.; Yan, N. STING activation by translocation from the ER is associated with infection and autoinflammatory disease. Cell Host Microbe 2015, 18, 157–168. [Google Scholar] [CrossRef] [Green Version]
  38. Liu, S.; Cai, X.; Wu, J.; Cong, Q.; Chen, X.; Li, T.; Du, F.; Ren, J.; Wu, Y.T.; Grishin, N.V.; et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 2015, 347, aaa2630. [Google Scholar] [CrossRef] [Green Version]
  39. Scumpia, P.O.; Botten, G.A.; Norman, J.S.; Kelly-Scumpia, K.M.; Spreafico, R.; Ruccia, A.R.; Purbey, P.K.; Thomas, B.J.; Modlin, R.L.; Smale, S.T. Opposing roles of Toll-like receptor and cytosolic DNA-STING signaling pathways for Staphylococcus aureus cutaneous host defense. PLoS Pathog. 2017, 13, e1006496. [Google Scholar] [CrossRef]
  40. Zhou, Y.; Zhao, S.; Gao, X.; Jiang, S.; Ma, J.; Wang, R.; Li, Q.; Qin, L.; Tong, Z.; Wu, J.; et al. Staphylococcus aureus induces IFN-β production via a CARMA3-independent mechanism. Pathogens 2021, 10, 300. [Google Scholar] [CrossRef]
  41. Picard, C.; von Bernuth, H.; Ghandil, P.; Chrabieh, M.; Levy, O.; Arkwright, P.D.; McDonald, D.; Geha, R.S.; Takada, H.; Krause, J.C.; et al. Clinical features and outcome of patients with IRAK-4 and MyD88 deficiency. Medicine 2010, 89, 403–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Chu, M.; Zhou, M.; Jiang, C.; Chen, X.; Guo, L.; Zhang, M.; Chu, Z.; Wang, Y. Staphylococcus aureus Phenol-Soluble Modulins α1-α3 act as novel Toll-Like Receptor (TLR) 4 antagonists to inhibit HMGB1/TLR4/NF-κB signaling pathway. Front. Immunol. 2018, 9, 862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Mae, M.; Iyori, M.; Yasuda, M.; Shamsul, H.M.; Kataoka, H.; Kiura, K.; Hasebe, A.; Totsuka, Y.; Shibata, K. The diacylated lipopeptide FSL-1 enhances phagocytosis of bacteria by macrophages through a Toll-like receptor 2-mediated signalling pathway. FEMS Immunol. Med. Microbiol. 2007, 49, 398–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Jann, N.J.; Schmaler, M.; Ferracin, F.; Landmann, R. TLR2 enhances NADPH oxidase activity and killing of Staphylococcus aureus by PMN. Immunol. Lett. 2011, 135, 17–23. [Google Scholar] [CrossRef]
  45. Fang, L.; Wu, H.-M.; Ding, P.-S.; Liu, R.-Y. TLR2 mediates phagocytosis and autophagy through JNK signaling pathway in Staphylococcus aureus-stimulated RAW264.7 cells. Cell Signal. 2014, 26, 806–814. [Google Scholar] [CrossRef] [PubMed]
  46. Ip, W.K.E.; Sokolovska, A.; Charriere, G.M.; Boyer, L.; Dejardin, S.; Cappillino, M.P.; Yantosca, L.M.; Takahashi, K.; Moore, K.J.; Lacy-Hulbert, A.; et al. Phagocytosis and phagosome acidification are required for pathogen processing and MyD88-dependent responses to Staphylococcus aureus. J. Immunol. 2010, 184, 7071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Jimenez-Dalmaroni, M.J.; Xiao, N.; Corper, A.L.; Verdino, P.; Ainge, G.D.; Larsen, D.S.; Painter, G.F.; Rudd, P.M.; Dwek, R.A.; Hoebe, K.; et al. Soluble CD36 ectodomain binds negatively charged diacylglycerol ligands and acts as a co-receptor for TLR2. PLoS ONE 2009, 4, e7411. [Google Scholar] [CrossRef]
  48. Israel, L.; Wang, Y.; Bulek, K.; Della Mina, E.; Zhang, Z.; Pedergnana, V.; Chrabieh, M.; Lemmens, N.A.; Sancho-Shimizu, V.; Descatoire, M.; et al. Human Adaptive Immunity Rescues an Inborn Error of Innate Immunity. Cell 2017, 168, 789–800. [Google Scholar] [CrossRef] [Green Version]
  49. Kanazawa, N. C-Type Lectin Receptors. In Immunology of the Skin: Basic and Clinical Sciences in Skin Immune Responses; Kabashima, K., Ed.; Springer: Tokyo, Japan, 2016; pp. 255–274. [Google Scholar] [CrossRef]
  50. De Monte, A.; Olivieri, C.-V.; Vitale, S.; Bailleux, S.; Castillo, L.; Giordanengo, V.; Maryanski, J.L.; Segura, E.; Doglio, A. CD1c-related DCs that express CD207/Langerin, but are distinguishable from langerhans cells, are consistently present in human tonsils. Front. Immunol. 2016, 7, 197. [Google Scholar] [CrossRef] [Green Version]
  51. van Dalen, R.; De La Cruz Diaz Jacinto, S.; Rumpret, M.; Fuchsberger Felix, F.; van Teijlingen Nienke, H.; Hanske, J.; Rademacher, C.; Geijtenbeek Teunis, B.H.; van Strijp Jos, A.G.; Weidenmaier, C.; et al. Langerhans cells sense Staphylococcus aureus wall teichoic acid through langerin to induce inflammatory responses. mBio 2019, 10, e00330-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Mnich, M.E.; van Dalen, R.; Gerlach, D.; Hendriks, A.; Xia, G.; Peschel, A.; van Strijp, J.A.G.; van Sorge, N.M. The C-type lectin receptor MGL senses N-acetylgalactosamine on the unique Staphylococcus aureus ST395 wall teichoic acid. Cell. Microbiol. 2019, 21, e13072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kurokawa, K.; Jung, D.-J.; An, J.-H.; Fuchs, K.; Jeon, Y.-J.; Kim, N.-H.; Li, X.; Tateishi, K.; Park, J.A.; Xia, G.; et al. Glycoepitopes of Staphylococcal wall teichoic acid govern complement-mediated opsonophagocytosis via human serum antibody and mannose-binding lectin. J. Biol. Chem. 2013, 288, 30956–30968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. van Dalen, R.; Peschel, A.; van Sorge, N.M. Wall teichoic acid in Staphylococcus aureus host interaction. Trends Microbiol. 2020, 28, 985–998. [Google Scholar] [CrossRef] [PubMed]
  55. Chong, Y.P.; Park, K.H.; Kim, E.S.; Kim, M.N.; Kim, S.H.; Lee, S.O.; Choi, S.H.; Jeong, J.Y.; Woo, J.H.; Kim, Y.S. Association of mannose-binding lectin 2 gene polymorphisms with persistent Staphylococcus aureus bacteremia. PLoS ONE 2014, 9, e89139. [Google Scholar] [CrossRef] [PubMed]
  56. Park, K.-H.; Kurokawa, K.; Zheng, L.; Jung, D.-J.; Tateishi, K.; Jin, J.-O.; Ha, N.-C.; Kang, H.J.; Matsushita, M.; Kwak, J.-Y.; et al. Human serum mannose-binding lectin senses wall teichoic acid glycopolymer of Staphylococcus aureus, which is restricted in infancy. J. Biol. Chem. 2010, 285, 27167–27175. [Google Scholar] [CrossRef] [Green Version]
  57. Mnich, M.E.; van Dalen, R.; van Sorge, N.M. C-Type lectin receptors in host defense against bacterial pathogens. Front. Cell. Infect. Microbiol. 2020, 10, 309. [Google Scholar] [CrossRef]
  58. Motta, V.; Soares, F.; Sun, T.; Philpott, D.J. NOD-like receptors: Versatile cytosolic sentinels. Physiol. Rev. 2015, 95, 149–178. [Google Scholar] [CrossRef] [Green Version]
  59. Holzinger, D.; Gieldon, L.; Mysore, V.; Nippe, N.; Taxman, D.J.; Duncan, J.A.; Broglie, P.M.; Marketon, K.; Austermann, J.; Vogl, T.; et al. Staphylococcus aureus panton-valentine leukocidin induces an inflammatory response in human phagocytes via the NLRP3 inflammasome. J. Leukoc. Biol. 2012, 92, 1069–1081. [Google Scholar] [CrossRef] [Green Version]
  60. Melehani, J.H.; James, D.B.; DuMont, A.L.; Torres, V.J.; Duncan, J.A. Staphylococcus aureus Leukocidin A/B (LukAB) kills human monocytes via host NLRP3 and ASC when extracellular, but not intracellular. PLoS Pathog. 2015, 11, e1004970. [Google Scholar] [CrossRef]
  61. Shimada, T.; Park, B.G.; Wolf, A.J.; Brikos, C.; Goodridge, H.S.; Becker, C.A.; Reyes, C.N.; Miao, E.A.; Aderem, A.; Götz, F.; et al. Staphylococcus aureus evades lysozyme-based peptidoglycan digestion that links phagocytosis, inflammasome activation, and IL-1beta secretion. Cell Host Microbe 2010, 7, 38–49. [Google Scholar] [CrossRef] [Green Version]
  62. Muñoz-Planillo, R.; Franchi, L.; Miller, L.S.; Núñez, G. A critical role for hemolysins and bacterial lipoproteins in Staphylococcus aureus-induced activation of the Nlrp3 inflammasome. J. Immunol. 2009, 183, 3942–3948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Craven, R.R.; Gao, X.; Allen, I.C.; Gris, D.; Bubeck Wardenburg, J.; McElvania-Tekippe, E.; Ting, J.P.; Duncan, J.A. Staphylococcus aureus alpha-hemolysin activates the NLRP3-inflammasome in human and mouse monocytic cells. PLoS ONE 2009, 4, e7446. [Google Scholar] [CrossRef] [PubMed]
  64. Khare, S.; Dorfleutner, A.; Bryan, N.B.; Yun, C.; Radian, A.D.; de Almeida, L.; Rojanasakul, Y.; Stehlik, C. An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity 2012, 36, 464–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Thompson, M.R.; Kaminski, J.J.; Kurt-Jones, E.A.; Fitzgerald, K.A. Pattern recognition receptors and the innate immune response to viral infection. Viruses 2011, 3, 920. [Google Scholar] [CrossRef] [Green Version]
  66. Yoneyama, M.; Kikuchi, M.; Natsukawa, T.; Shinobu, N.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Akira, S.; Fujita, T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 2004, 5, 730–737. [Google Scholar] [CrossRef]
  67. Saito, T.; Hirai, R.; Loo, Y.M.; Owen, D.; Johnson, C.L.; Sinha, S.C.; Akira, S.; Fujita, T.; Gale, M., Jr. Regulation of innate antiviral defenses through a shared repressor domain in RIG-I and LGP2. Proc. Natl. Acad. Sci. USA 2007, 104, 582–587. [Google Scholar] [CrossRef] [Green Version]
  68. Yoneyama, M.; Kikuchi, M.; Matsumoto, K.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Foy, E.; Loo, Y.M.; Gale, M., Jr.; Akira, S.; et al. Shared and unique functions of the DExD/H-box helicases RIG-I, MDA5, and LGP2 in antiviral innate immunity. J. Immunol. 2005, 175, 2851–2858. [Google Scholar] [CrossRef] [Green Version]
  69. Komuro, A.; Horvath, C.M. RNA- and virus-independent inhibition of antiviral signaling by RNA helicase LGP2. J. Virol. 2006, 80, 12332–12342. [Google Scholar] [CrossRef] [Green Version]
  70. Venkataraman, T.; Valdes, M.; Elsby, R.; Kakuta, S.; Caceres, G.; Saijo, S.; Iwakura, Y.; Barber, G.N. Loss of DExD/H box RNA helicase LGP2 manifests disparate antiviral responses. J. Immunol. 2007, 178, 6444–6455. [Google Scholar] [CrossRef]
  71. Rothenfusser, S.; Goutagny, N.; DiPerna, G.; Gong, M.; Monks, B.G.; Schoenemeyer, A.; Yamamoto, M.; Akira, S.; Fitzgerald, K.A. The RNA helicase Lgp2 inhibits TLR-independent sensing of viral replication by retinoic acid-inducible gene-I. J. Immunol. 2005, 175, 5260–5268. [Google Scholar] [CrossRef] [Green Version]
  72. Takeuchi, O.; Akira, S. Innate immunity to virus infection. Immunol. Rev. 2009, 227, 75–86. [Google Scholar] [CrossRef] [PubMed]
  73. O’Neill, L.A.; Bowie, A.G. Sensing and signaling in antiviral innate immunity. Curr. Biol. 2010, 20, R328–R333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Schroder, M.; Baran, M.; Bowie, A.G. Viral targeting of DEAD box protein 3 reveals its role in TBK1/IKKepsilon-mediated IRF activation. EMBO J. 2008, 27, 2147–2157. [Google Scholar] [CrossRef] [PubMed]
  75. Michallet, M.C.; Meylan, E.; Ermolaeva, M.A.; Vazquez, J.; Rebsamen, M.; Curran, J.; Poeck, H.; Bscheider, M.; Hartmann, G.; Konig, M.; et al. TRADD protein is an essential component of the RIG-like helicase antiviral pathway. Immunity 2008, 28, 651–661. [Google Scholar] [CrossRef]
  76. Matsumiya, T.; Stafforini, D.M. Function and regulation of retinoic acid-inducible gene-I. Crit. Rev. Immunol. 2010, 30, 489–513. [Google Scholar] [CrossRef] [Green Version]
  77. Rehwinkel, J.; Gack, M.U. RIG-I-like receptors: Their regulation and roles in RNA sensing. Nat. Rev. Immunol. 2020, 20, 537–551. [Google Scholar] [CrossRef]
  78. Chiu, Y.-H.; MacMillan, J.B.; Chen, Z.J. RNA polymerase III detects cytosolic DNA and induces type I interferons through the RIG-I pathway. Cell 2009, 138, 576–591. [Google Scholar] [CrossRef] [Green Version]
  79. Monroe, K.M.; McWhirter, S.M.; Vance, R.E. Identification of host cytosolic sensors and bacterial factors regulating the Type I interferon response to Legionella pneumophila. PLoS Pathog. 2009, 5, e1000665. [Google Scholar] [CrossRef]
  80. Schmolke, M.; Patel, J.R.; Castro, E.d.; Sánchez-Aparicio, M.T.; Uccellini, M.B.; Miller, J.C.; Manicassamy, B.; Satoh, T.; Kawai, T.; Akira, S.; et al. RIG-I detects mRNA of intracellular Salmonella enterica serovar Typhimurium during bacterial infection. mBio 2014, 5, e01006-14. [Google Scholar] [CrossRef] [Green Version]
  81. Abdullah, Z.; Schlee, M.; Roth, S.; Mraheil, M.A.; Barchet, W.; Böttcher, J.; Hain, T.; Geiger, S.; Hayakawa, Y.; Fritz, J.H. RIG-I detects infection with live Listeria by sensing secreted bacterial nucleic acids. EMBO J. 2012, 31, 4153–4164. [Google Scholar] [CrossRef] [Green Version]
  82. Hagmann, C.A.; Herzner, A.M.; Abdullah, Z.; Zillinger, T.; Jakobs, C.; Schuberth, C.; Coch, C.; Higgins, P.G.; Wisplinghoff, H.; Barchet, W.; et al. RIG-I detects triphosphorylated RNA of Listeria monocytogenes during infection in non-immune cells. PLoS ONE 2013, 8, e62872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Asadpour-Behzadi, A.; Kariminik, A. RIG-1 and MDA5 are the important intracellular sensors against bacteria in septicemia suffering patients. J. Appl. Biomed. 2018, 16, 358–361. [Google Scholar] [CrossRef]
  84. Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Li, X.D.; Wu, J.; Gao, D.; Wang, H.; Sun, L.; Chen, Z.J. Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 2013, 341, 1390–1394. [Google Scholar] [CrossRef] [Green Version]
  86. Chen, Q.; Sun, L.; Chen, Z.J. Regulation and function of the cGAS–STING pathway of cytosolic DNA sensing. Nat. Immunol. 2016, 17, 1142. [Google Scholar] [CrossRef]
  87. Wu, J.; Sun, L.; Chen, X.; Du, F.; Shi, H.; Chen, C.; Chen, Z.J. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 2013, 339, 826–830. [Google Scholar] [CrossRef] [Green Version]
  88. Liu, Z.-Z.; Yang, Y.-J.; Zhou, C.-K.; Yan, S.-Q.; Ma, K.; Gao, Y.; Chen, W. STING contributes to host defense against Staphylococcus aureus pneumonia through suppressing necroptosis. Front. Immunol. 2021, 12, 2076. [Google Scholar] [CrossRef]
  89. Kaplan, A.; Ma, J.; Kyme, P.; Wolf, A.J.; Becker, C.A.; Tseng, C.W.; Liu, G.Y.; Underhill, D.M. Failure to induce IFN-β production during Staphylococcus aureus infection contributes to pathogenicity. J. Immunol. 2012, 189, 4537. [Google Scholar] [CrossRef] [Green Version]
  90. Mistry, R.D. Skin and soft tissue infections. Pediatr. Clin. N. Am. 2013, 60, 1063–1082. [Google Scholar] [CrossRef]
  91. Pasparakis, M.; Haase, I.; Nestle, F.O. Mechanisms regulating skin immunity and inflammation. Nat. Rev. Immunol. 2014, 14, 289–301. [Google Scholar] [CrossRef]
  92. Miller, L.S. Toll-like receptors in skin. Adv. Dermatol. 2008, 24, 71–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Soong, G.; Paulino, F.; Wachtel, S.; Parker, D.; Wickersham, M.; Zhang, D.; Brown, A.; Lauren, C.; Dowd, M.; West, E.; et al. Methicillin-resistant Staphylococcus aureus adaptation to human keratinocytes. mBio 2015, 6, e00289-15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Nguyen, M.T.; Kraft, B.; Yu, W.; Demicrioglu, D.D.; Hertlein, T.; Burian, M.; Schmaler, M.; Boller, K.; Bekeredjian-Ding, I.; Ohlsen, K.; et al. The νSaα specific Lipoprotein Like Cluster (lpl) of S. aureus USA300 contributes to immune stimulation and invasion in human ells. PLoS Pathog. 2015, 11, e1004984. [Google Scholar] [CrossRef] [Green Version]
  95. Kim, N.J.; Ahn, K.B.; Jeon, J.H.; Yun, C.-H.; Finlay, B.B.; Han, S.H. Lipoprotein in the cell wall of Staphylococcus aureus is a major inducer of nitric oxide production in murine macrophages. Mol. Immunol. 2015, 65, 17–24. [Google Scholar] [CrossRef] [PubMed]
  96. Nandi, A.; Dey, S.; Biswas, J.; Jaiswal, P.; Naaz, S.; Yasmin, T.; Bishayi, B. Differential induction of inflammatory cytokines and reactive oxygen species in murine peritoneal macrophages and resident fresh bone marrow cells by acute staphylococcus aureus infection: Contribution of toll-like receptor 2 (TLR2). Inflammation 2015, 38, 224–244. [Google Scholar] [CrossRef]
  97. Bekeredjian-Ding, I.; Stein, C.; Uebele, J. The innate immune response against Staphylococcus aureus. In Staphylococcus aureus: Microbiology, Pathology, Immunology, Therapy and Prophylaxis; Bagnoli, F., Rappuoli, R., Grandi, G., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 385–418. [Google Scholar] [CrossRef]
  98. Kim, B.; Jiang, T.; Bae, J.H.; Yun, H.S.; Jang, S.H.; Kim, J.H.; Kim, J.D.; Hur, J.H.; Shibata, K.; Kurokawa, K.; et al. In Staphylococcus aureus, the particulate state of the cell envelope is required for the efficient induction of host defense responses. Infect. Immun. 2019, 87. [Google Scholar] [CrossRef] [PubMed]
  99. Surewaard, B.G.; Deniset, J.F.; Zemp, F.J.; Amrein, M.; Otto, M.; Conly, J.; Omri, A.; Yates, R.M.; Kubes, P. Identification and treatment of the Staphylococcus aureus reservoir in vivo. J. Exp. Med. 2016, 213, 1141–1151. [Google Scholar] [CrossRef] [Green Version]
  100. Kitur, K.; Parker, D.; Nieto, P.; Ahn, D.S.; Cohen, T.S.; Chung, S.; Wachtel, S.; Bueno, S.; Prince, A. Toxin-induced necroptosis is a major mechanism of Staphylococcus aureus lung damage. PLoS Pathog. 2015, 11, e1004820. [Google Scholar] [CrossRef]
  101. Martin, F.J.; Parker, D.; Harfenist, B.S.; Soong, G.; Prince, A. Participation of CD11c(+) leukocytes in methicillin-resistant Staphylococcus aureus clearance from the lung. Infect. Immun. 2011, 79, 1898–1904. [Google Scholar] [CrossRef] [Green Version]
  102. Prajsnar, T.K.; Cunliffe, V.T.; Foster, S.J.; Renshaw, S.A. A novel vertebrate model of Staphylococcus aureus infection reveals phagocyte-dependent resistance of zebrafish to non-host specialized pathogens. Cell. Microbiol. 2008, 10, 2312–2325. [Google Scholar] [CrossRef]
  103. Nacken, W.; Roth, J.; Sorg, C.; Kerkhoff, C. S100A9/S100A8: Myeloid representatives of the S100 protein family as prominent players in innate immunity. Microsc. Res. Tech. 2003, 60, 569–580. [Google Scholar] [CrossRef] [PubMed]
  104. Corbin, B.D.; Seeley, E.H.; Raab, A.; Feldmann, J.; Miller, M.R.; Torres, V.J.; Anderson, K.L.; Dattilo, B.M.; Dunman, P.M.; Gerads, R.; et al. Metal chelation and inhibition of bacterial growth in tissue abscesses. Science 2008, 319, 962–965. [Google Scholar] [CrossRef] [PubMed]
  105. Damo, S.M.; Kehl-Fie, T.E.; Sugitani, N.; Holt, M.E.; Rathi, S.; Murphy, W.J.; Zhang, Y.; Betz, C.; Hench, L.; Fritz, G.; et al. Molecular basis for manganese sequestration by calprotectin and roles in the innate immune response to invading bacterial pathogens. Proc. Natl. Acad. Sci. USA 2013, 110, 3841–3846. [Google Scholar] [CrossRef] [Green Version]
  106. Wang, X.; Eagen, W.J.; Lee, J.C. Orchestration of human macrophage NLRP3 inflammasome activation by Staphylococcus aureus extracellular vesicles. Proc. Natl. Acad. Sci. USA 2020, 117, 3174. [Google Scholar] [CrossRef]
  107. Yao, R.; Chen, Y.; Hao, H.; Guo, Z.; Cheng, X.; Ma, Y.; Ji, Q.; Yang, X.; Wang, Y.; Li, X.; et al. Pathogenic effects of inhibition of mTORC1/STAT3 axis facilitates Staphylococcus aureus-induced pyroptosis in human macrophages. Cell Commun. Signal. 2020, 18, 187. [Google Scholar] [CrossRef]
  108. Kubica, M.; Guzik, K.; Koziel, J.; Zarebski, M.; Richter, W.; Gajkowska, B.; Golda, A.; Maciag-Gudowska, A.; Brix, K.; Shaw, L.; et al. A potential new pathway for Staphylococcus aureus dissemination: The silent survival of S. aureus phagocytosed by human monocyte-derived macrophages. PLoS ONE 2008, 3, e1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. González-Barca, E.; Carratalà, J.; Mykietiuk, A.; Fernández-Sevilla, A.; Gudiol, F. Predisposing factors and outcome of Staphylococcus aureus bacteremia in neutropenic patients with cancer. Eur. J. Clin. Microbiol. Infect. Dis 2001, 20, 117–119. [Google Scholar] [CrossRef]
  110. Bouma, G.; Ancliff, P.J.; Thrasher, A.J.; Burns, S.O. Recent advances in the understanding of genetic defects of neutrophil number and function. Br. J. Haematol. 2010, 151, 312–326. [Google Scholar] [CrossRef]
  111. Lakshman, R.; Finn, A. Neutrophil disorders and their management. J. Clin. Pathol. 2001, 54, 7–19. [Google Scholar] [CrossRef]
  112. Miller, L.S.; Cho, J.S. Immunity against Staphylococcus aureus cutaneous infections. Nat. Rev. Immunol. 2011, 11, 505–518. [Google Scholar] [CrossRef] [Green Version]
  113. El-Benna, J.; Dang, P.M.; Gougerot-Pocidalo, M.A. Priming of the neutrophil NADPH oxidase activation: Role of p47phox phosphorylation and NOX2 mobilization to the plasma membrane. Semin. Immunopathol. 2008, 30, 279–289. [Google Scholar] [CrossRef] [PubMed]
  114. Clarke, T.B.; Davis, K.M.; Lysenko, E.S.; Zhou, A.Y.; Yu, Y.; Weiser, J.N. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat. Med. 2010, 16, 228–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Ellis, T.N.; Beaman, B.L. Interferon-gamma activation of polymorphonuclear neutrophil function. Immunology 2004, 112, 2–12. [Google Scholar] [CrossRef] [PubMed]
  116. Teng, T.-S.; Ji, A.-l.; Ji, X.-Y.; Li, Y.-Z. Neutrophils and immunity: From bactericidal action to being conquered. J. Immunol. Res. 2017, 2017, 9671604. [Google Scholar] [CrossRef]
  117. Urban, C.F.; Lourido, S.; Zychlinsky, A. How do microbes evade neutrophil killing? Cell. Microbiol. 2006, 8, 1687–1696. [Google Scholar] [CrossRef]
  118. Pilsczek, F.H.; Salina, D.; Poon, K.K.H.; Fahey, C.; Yipp, B.G.; Sibley, C.D.; Robbins, S.M.; Green, F.H.Y.; Surette, M.G.; Sugai, M.; et al. A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J. Immunol. 2010, 185, 7413. [Google Scholar] [CrossRef] [Green Version]
  119. van Kessel, K.P.M.; Bestebroer, J.; van Strijp, J.A.G. Neutrophil-mediated phagocytosis of Staphylococcus aureus. Front. Immunol. 2014, 5, 467. [Google Scholar] [CrossRef] [Green Version]
  120. Günther, F.; Wabnitz, G.H.; Stroh, P.; Prior, B.; Obst, U.; Samstag, Y.; Wagner, C.; Hänsch, G.M. Host defence against Staphylococcus aureus biofilms infection: Phagocytosis of biofilms by polymorphonuclear neutrophils (PMN). Mol. Immunol. 2009, 46, 1805–1813. [Google Scholar] [CrossRef]
  121. Rowley, D.A.; Fitch, F.W. The road to the discovery of dendritic cells, a tribute to Ralph Steinman. Cell. Immunol. 2012, 273, 95–98. [Google Scholar] [CrossRef]
  122. Liu, K. Dendritic cells. Encyclopedia Cell Biol. 2016, 3, 741–749. [Google Scholar] [CrossRef]
  123. Liu, J.; Zhang, X.; Cheng, Y.; Cao, X. Dendritic cell migration in inflammation and immunity. Cell. Mol. Immunol. 2021, 18, 2461–2471. [Google Scholar] [CrossRef] [PubMed]
  124. Alvarez, D.; Vollmann, E.H.; von Andrian, U.H. Mechanisms and consequences of dendritic cell migration. Immunity 2008, 29, 325–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Solano-Gálvez, G.S.; Tovar-Torres, M.S.; Tron-Gómez, S.M.; Weiser-Smeke, E.A.; Álvarez-Hernández, A.D.; Franyuti-Kelly, A.G.; Tapia-Moreno, M.; Ibarra, A.; Gutiérrez-Kobeh, L.; Vázquez-López, R. Human dendritic cells: Ontogeny and their subsets in health and disease. Med. Sci. 2018, 6, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Chen, L.; Zhu, Y.; Zhang, G.; Gao, C.; Zhong, W.; Zhang, X. CD83-stimulated monocytes suppress T-cell immune responses through production of prostaglandin E2. Proc. Natl. Acad. Sci. USA 2011, 108, 18778–18783. [Google Scholar] [CrossRef] [Green Version]
  127. Banchereau, J.; Briere, F.; Caux, C.; Davoust, J.; Lebecque, S.; Liu, Y.J.; Pulendran, B.; Palucka, K. Immunobiology of dendritic cells. Annu. Rev. Immunol. 2000, 18, 767–811. [Google Scholar] [CrossRef] [PubMed]
  128. Kawamura, K.; Iyonaga, K.; Ichiyasu, H.; Nagano, J.; Suga, M.; Sasaki, Y. Differentiation, maturation, and survival of dendritic cells by osteopontin regulation. Clin. Diagn. Lab. Immunol. 2005, 12, 206–212. [Google Scholar] [CrossRef] [Green Version]
  129. de Saint-Vis, B.; Vincent, J.; Vandenabeele, S.; Vanbervliet, B.; Pin, J.J.; Aït-Yahia, S.; Patel, S.; Mattei, M.G.; Banchereau, J.; Zurawski, S.; et al. A novel lysosome-associated membrane glycoprotein, DC-LAMP, induced upon DC maturation, is transiently expressed in MHC Class II compartment. Immunity 1998, 9, 325–336. [Google Scholar] [CrossRef] [Green Version]
  130. Heath, W.R.; Carbone, F.R. Dendritic cell subsets in primary and secondary T cell responses at body surfaces. Nat. Immunol. 2009, 10, 1237–1244. [Google Scholar] [CrossRef]
  131. Macri, C.; Pang, E.S.; Patton, T.; O’Keeffe, M. Dendritic cell subsets. Semin. Cell Dev. Biol. 2018, 84, 11–21. [Google Scholar] [CrossRef]
  132. Hespel, C.; Moser, M. Role of inflammatory dendritic cells in innate and adaptive immunity. Eur. J. Immunol. 2012, 42, 2535–2543. [Google Scholar] [CrossRef]
  133. McKenna, K.; Beignon, A.-S.; Bhardwaj, N. Plasmacytoid dendritic cells: Linking innate and adaptive immunity. J. Virol. 2005, 79, 17–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Darisipudi, M.N.; Nordengrün, M.; Bröker, B.M.; Péton, V. Messing with the sentinels-The interaction of Staphylococcus aureus with dendritic Cells. Microorganisms 2018, 6, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Pidwill, G.R.; Gibson, J.F.; Cole, J.; Renshaw, S.A.; Foster, S.J. The Role of Macrophages in Staphylococcus aureus Infection. Front. Immunol. 2021, 11, 339. [Google Scholar] [CrossRef] [PubMed]
  136. Nishat, S.; Wuescher, L.M.; Worth, R.G. Platelets enhance dendritic cell responses against Staphylococcus aureus through CD40-CD40L. Infect. Immun. 2018, 86, e00186-18. [Google Scholar] [CrossRef] [Green Version]
  137. O’Keeffe, K.M.; Wilk, M.M.; Leech, J.M.; Murphy, A.G.; Laabei, M.; Monk, I.R.; Massey, R.C.; Lindsay, J.A.; Foster, T.J.; Geoghegan, J.A.; et al. Manipulation of autophagy in phagocytes facilitates Staphylococcus aureus bloodstream infection. Infect. Immun. 2015, 83, 3445–3457. [Google Scholar] [CrossRef] [Green Version]
  138. Uebele, J.; Stein, C.; Nguyen, M.-T.; Schneider, A.; Kleinert, F.; Tichá, O.; Bierbaum, G.; Götz, F.; Bekeredjian-Ding, I. Antigen delivery to dendritic cells shapes human CD4+ and CD8+ T cell memory responses to Staphylococcus aureus. PLoS Pathog. 2017, 13, e1006387. [Google Scholar] [CrossRef]
  139. Jin, J.O.; Zhang, W.; Du, J.Y.; Yu, Q. BDCA1-positive dendritic cells (DCs) represent a unique human myeloid DC subset that induces innate and adaptive immune responses to Staphylococcus aureus Infection. Infect. Immun. 2014, 82, 4466–4476. [Google Scholar] [CrossRef] [Green Version]
  140. Parcina, M.; Wendt, C.; Goetz, F.; Zawatzky, R.; Zähringer, U.; Heeg, K.; Bekeredjian-Ding, I. Staphylococcus aureus induced plasmacytoid dendritic cell activation is based on an IgG-mediated memory response. J. Immunol. 2008, 181, 3823. [Google Scholar] [CrossRef] [Green Version]
  141. Michea, P.; Vargas, P.; Donnadieu, M.-H.; Rosemblatt, M.; Bono, M.; Dumenil, G.; Soumelis, V. Epithelial control of the human pDC response to extracellular bacteria. Eur. J. Immunol. 2013, 43. [Google Scholar] [CrossRef]
  142. Schindler, D.; Gutierrez, M.G.; Beineke, A.; Rauter, Y.; Rohde, M.; Foster, S.; Goldmann, O.; Medina, E. Dendritic cells are central coordinators of the host immune response to Staphylococcus aureus bloodstream infection. Am. J. Pathol. 2012, 181, 1327–1337. [Google Scholar] [CrossRef]
  143. Compton, L.A.; Murphy, G.F.; Lian, C.G. Diagnostic immunohistochemistry in cutaneous neoplasia: An update. Dermatopathology 2015, 2, 15–42. [Google Scholar] [CrossRef] [PubMed]
  144. Richardson, J.R.; Armbruster, N.S.; Gunter, M.; Biljecki, M.; Klenk, J.; Heumos, S.; Autenrieth, S.E. PSM Peptides From Community-Associated Methicillin-Resistant Staphylococcus aureus Impair the Adaptive Immune Response via Modulation of Dendritic Cell Subsets in vivo. Front. Immunol. 2019, 10, 995. [Google Scholar] [CrossRef] [PubMed]
  145. Berends Evelien, T.M.; Zheng, X.; Zwack Erin, E.; Ménager Mickaël, M.; Cammer, M.; Shopsin, B.; Torres Victor, J.; Freitag Nancy, E. Staphylococcus aureus impairs the function of and kills human dendritic cells via the LukAB toxin. mBio 2019, 10, e01918-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Goldmann, O.; Medina, E. Staphylococcus aureus strategies to evade the host acquired immune response. Int. J. Med. Microbiol. 2018, 308, 625–630. [Google Scholar] [CrossRef]
  147. Zhao, Y.; van Kessel, K.P.M.; de Haas, C.J.C.; Rogers, M.R.C.; van Strijp, J.A.G.; Haas, P.-J.A. Staphylococcal superantigen-like protein 13 activates neutrophils via formyl peptide receptor 2. Cell microbiol 2018, 20, e12941. [Google Scholar] [CrossRef] [Green Version]
  148. Bestebroer, J.; Poppelier, M.J.; Ulfman, L.H.; Lenting, P.J.; Denis, C.V.; van Kessel, K.P.; van Strijp, J.A.; de Haas, C.J. Staphylococcal superantigen-like 5 binds PSGL-1 and inhibits P-selectin-mediated neutrophil rolling. Blood 2007, 109, 2936–2943. [Google Scholar] [CrossRef]
  149. Koymans, K.J.; Goldmann, O.; Karlsson, C.A.Q.; Sital, W.; Thanert, R.; Bisschop, A.; Vrieling, M.; Malmstrom, J.; van Kessel, K.P.M.; de Haas, C.J.C.; et al. The TLR2 Antagonist Staphylococcal Superantigen-Like Protein 3 Acts as a Virulence Factor to Promote Bacterial Pathogenicity in vivo. J. Innate Immun. 2017, 9, 561–573. [Google Scholar] [CrossRef]
  150. Walenkamp, A.M.; Boer, I.G.; Bestebroer, J.; Rozeveld, D.; Timmer-Bosscha, H.; Hemrika, W.; van Strijp, J.A.; de Haas, C.J. Staphylococcal superantigen-like 10 inhibits CXCL12-induced human tumor cell migration. Neoplasia 2009, 11, 333–344. [Google Scholar] [CrossRef] [Green Version]
  151. Bardoel, B.W.; Vos, R.; Bouman, T.; Aerts, P.C.; Bestebroer, J.; Huizinga, E.G.; Brondijk, T.H.; van Strijp, J.A.; de Haas, C.J. Evasion of Toll-like receptor 2 activation by staphylococcal superantigen-like protein 3. J. Mol. Med. 2012, 90, 1109–1120. [Google Scholar] [CrossRef]
  152. Koymans, K.J.; Feitsma, L.J.; Brondijk, T.H.; Aerts, P.C.; Lukkien, E.; Lossl, P.; van Kessel, K.P.; de Haas, C.J.; van Strijp, J.A.; Huizinga, E.G. Structural basis for inhibition of TLR2 by staphylococcal superantigen-like protein 3 (SSL3). Proc. Natl. Acad. Sci. USA 2015, 112, 11018–11023. [Google Scholar] [CrossRef] [Green Version]
  153. Yokoyama, R.; Itoh, S.; Kamoshida, G.; Takii, T.; Fujii, S.; Tsuji, T.; Onozaki, K. Staphylococcal superantigen-like protein 3 binds to the Toll-like receptor 2 extracellular domain and inhibits cytokine production induced by Staphylococcus aureus, cell wall component, or lipopeptides in murine macrophages. Infect. Immun. 2012, 80, 2816–2825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Takeda, K.; Akira, S. TLR signaling pathways. Semin. Immunol. 2004, 16, 3–9. [Google Scholar] [CrossRef]
  155. Xu, Y.; Tao, X.; Shen, B.; Horng, T.; Medzhitov, R.; Manley, J.L.; Tong, L. Structural basis for signal transduction by the Toll/interleukin-1 receptor domains. Nature 2000, 408, 111–115. [Google Scholar] [CrossRef] [PubMed]
  156. Askarian, F.; van Sorge, N.M.; Sangvik, M.; Beasley, F.C.; Henriksen, J.R.; Sollid, J.U.; van Strijp, J.A.; Nizet, V.; Johannessen, M. A Staphylococcus aureus TIR domain protein virulence factor blocks TLR2-mediated NF-κB signaling. J. Innate Immun. 2014, 6, 485–498. [Google Scholar] [CrossRef]
  157. Schiller, M.; Metze, D.; Luger, T.A.; Grabbe, S.; Gunzer, M. Immune response modifiers—Mode of action. Exp. Dermatol. 2006, 15, 331–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Askarian, F.; Wagner, T.; Johannessen, M.; Nizet, V. Staphylococcus aureus modulation of innate immune responses through Toll-like (TLR), (NOD)-like (NLR) and C-type lectin (CLR) receptors. FEMS Microbiol. Rev. 2018, 42, 656–671. [Google Scholar] [CrossRef] [PubMed]
  159. Patot, S.; Imbert, P.R.C.; Baude, J.; Martins Simões, P.; Campergue, J.-B.; Louche, A.; Nijland, R.; Bès, M.; Tristan, A.; Laurent, F.; et al. Correction: The TIR homologue lies near resistance genes in Staphylococcus aureus, coupling modulation of virulence and antimicrobial susceptibility. PLoS Pathog. 2017, 13, e1006291. [Google Scholar] [CrossRef]
  160. Kitakaze, M.; Hori, M.; Kamada, T. Role of adenosine and its interaction with alpha adrenoceptor activity in ischaemic and reperfusion injury of the myocardium. Cardiovasc. Res. 1993, 27, 18–27. [Google Scholar] [CrossRef]
  161. Kaufmann, I.; Hoelzl, A.; Schliephake, F.; Hummel, T.; Chouker, A.; Lysenko, L.; Peter, K.; Thiel, M. Effects of adenosine on functions of polymorphonuclear leukocytes from patients with septic shock. Shock 2007, 27, 25–31. [Google Scholar] [CrossRef]
  162. Csoka, B.; Nemeth, Z.H.; Virag, L.; Gergely, P.; Leibovich, S.J.; Pacher, P.; Sun, C.X.; Blackburn, M.R.; Vizi, E.S.; Deitch, E.A.; et al. A2A adenosine receptors and C/EBPbeta are crucially required for IL-10 production by macrophages exposed to Escherichia coli. Blood 2007, 110, 2685–2695. [Google Scholar] [CrossRef] [Green Version]
  163. Edwards, C.K., 3rd; Watts, L.M.; Parmely, M.J.; Linnik, M.D.; Long, R.E.; Borcherding, D.R. Effect of the carbocyclic nucleoside analogue MDL 201,112 on inhibition of interferon-gamma-induced priming of Lewis (LEW/N) rat macrophages for enhanced respiratory burst and MHC class II Ia+ antigen expression. J. Leukoc. Biol. 1994, 56, 133–144. [Google Scholar] [CrossRef] [PubMed]
  164. Panther, E.; Idzko, M.; Herouy, Y.; Rheinen, H.; Gebicke-Haerter, P.J.; Mrowietz, U.; Dichmann, S.; Norgauer, J. Expression and function of adenosine receptors in human dendritic cells. FASEB J. 2001, 15, 1963–1970. [Google Scholar] [CrossRef] [PubMed]
  165. HaskÓ, G.; Kuhel, D.G.; Chen, J.-F.; Schwarzschild, M.A.; Deitch, E.A.; Mabley, J.G.; Marton, A.; SzabÓ, C. Adenosine inhibits IL-12 and TNF-α production via adenosine A2a receptor-dependent and independent mechanisms. FASEB J. 2000, 14, 2065–2074. [Google Scholar] [CrossRef] [Green Version]
  166. Savina, A.; Amigorena, S. Phagocytosis and antigen presentation in dendritic cells. Immunol. Rev. 2007, 219, 143–156. [Google Scholar] [CrossRef] [PubMed]
  167. Zhou, C.; Bhinderwala, F.; Lehman, M.K.; Thomas, V.C.; Chaudhari, S.S.; Yamada, K.J.; Foster, K.W.; Powers, R.; Kielian, T.; Fey, P.D. Urease is an essential component of the acid response network of Staphylococcus aureus and is required for a persistent murine kidney infection. PLoS Pathog. 2019, 15, e1007538. [Google Scholar] [CrossRef] [PubMed]
  168. Collins, L.V.; Kristian, S.A.; Weidenmaier, C.; Faigle, M.; Van Kessel, K.P.; Van Strijp, J.A.; Götz, F.; Neumeister, B.; Peschel, A. Staphylococcus aureus strains lacking D-alanine modifications of teichoic acids are highly susceptible to human neutrophil killing and are virulence attenuated in mice. J. Infect. Dis. 2002, 186, 214–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Hampton, M.B.; Winterbourn, C.C. Modification of neutrophil oxidant production with diphenyleneiodonium and its effect on bacterial killing. Free Radic. Biol. Med. 1995, 18, 633–639. [Google Scholar] [CrossRef]
  170. Monteith, A.J.; Miller, J.M.; Maxwell, C.N.; Chazin, W.J.; Skaar, E.P. Neutrophil extracellular traps enhance macrophage killing of bacterial pathogens. Sci. Adv. 2021, 7, eabj2101. [Google Scholar] [CrossRef]
  171. Ballal, A.; Manna, A.C. Regulation of superoxide dismutase (sod) genes by SarA in Staphylococcus aureus. J. Bacteriol. 2009, 191, 3301–3310. [Google Scholar] [CrossRef] [Green Version]
  172. Cosgrove, K.; Coutts, G.; Jonsson, I.M.; Tarkowski, A.; Kokai-Kun, J.F.; Mond, J.J.; Foster, S.J. Catalase (KatA) and alkyl hydroperoxide reductase (AhpC) have compensatory roles in peroxide stress resistance and are required for survival, persistence, and nasal colonization in Staphylococcus aureus. J. Bacteriol. 2007, 189, 1025–1035. [Google Scholar] [CrossRef] [Green Version]
  173. Schnaith, A.; Kashkar, H.; Leggio, S.A.; Addicks, K.; Krönke, M.; Krut, O. Staphylococcus aureus subvert autophagy for induction of caspase-independent host cell death. J. Biol. Chem. 2007, 282, 2695–2706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Neumann, Y.; Bruns, S.A.; Rohde, M.; Prajsnar, T.K.; Foster, S.J.; Schmitz, I. Intracellular Staphylococcus aureus eludes selective autophagy by activating a host cell kinase. Autophagy 2016, 12, 2069–2084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Wang, M.; Fan, Z.; Han, H. Autophagy in Staphylococcus aureus infection. Front. Cell. Infect. Microbiol. 2021, 11, 222. [Google Scholar] [CrossRef] [PubMed]
  176. Jondle, C.N.; Gupta, K.; Mishra, B.B.; Sharma, J. Klebsiella pneumoniae infection of murine neutrophils impairs their efferocytic clearance by modulating cell death machinery. PLoS Pathog. 2018, 14, e1007338. [Google Scholar] [CrossRef] [Green Version]
  177. Greenlee-Wacker, M.C.; Rigby, K.M.; Kobayashi, S.D.; Porter, A.R.; DeLeo, F.R.; Nauseef, W.M. Phagocytosis of Staphylococcus aureus by human neutrophils prevents macrophage efferocytosis and induces programmed necrosis. J. Immunol. 2014, 192, 4709. [Google Scholar] [CrossRef] [Green Version]
  178. Cohen, T.S.; Jones-Nelson, O.; Hotz, M.; Cheng, L.; Miller, L.S.; Suzich, J.; Stover, C.K.; Sellman, B.R.S. aureus blocks efferocytosis of neutrophils by macrophages through the activity of its virulence factor alpha toxin. Sci. Rep. 2016, 6, 35466. [Google Scholar] [CrossRef]
  179. Huitema, L.; Phillips, T.; Alexeev, V.; Tomic-Canic, M.; Pastar, I.; Igoucheva, O. Intracellular escape strategies of Staphylococcus aureus in persistent cutaneous infections. Exp. Dermatol. 2021, 30, 1428–1439. [Google Scholar] [CrossRef]
  180. Nowicka, D.; Grywalska, E. Staphylococcus aureus and host immunity in recurrent furunculosis. Dermatology 2019, 235, 295–305. [Google Scholar] [CrossRef]
  181. Bera, A.; Herbert, S.; Jakob, A.; Vollmer, W.; Gotz, F. Why are pathogenic staphylococci so lysozyme resistant? The peptidoglycan O-acetyltransferase OatA is the major determinant for lysozyme resistance of Staphylococcus aureus. Mol. Microbiol. 2005, 55, 778–787. [Google Scholar] [CrossRef]
  182. Pushkaran, A.C.; Nataraj, N.; Nair, N.; Götz, F.; Biswas, R.; Mohan, C.G. Understanding the Structure–Function Relationship of Lysozyme Resistance in Staphylococcus aureus by Peptidoglycan O-Acetylation Using Molecular Docking, Dynamics, and Lysis Assay. J. Chem. Inf. Model. 2015, 55, 760–770. [Google Scholar] [CrossRef]
  183. Giese, B.; Glowinski, F.; Paprotka, K.; Dittmann, S.; Steiner, T.; Sinha, B.; Fraunholz, M.J. Expression of delta-toxin by Staphylococcus aureus mediates escape from phago-endosomes of human epithelial and endothelial cells in the presence of beta-toxin. Cell. Microbiol. 2011, 13, 316–329. [Google Scholar] [CrossRef] [PubMed]
  184. Grosz, M.; Kolter, J.; Paprotka, K.; Winkler, A.C.; Schafer, D.; Chatterjee, S.S.; Geiger, T.; Wolz, C.; Ohlsen, K.; Otto, M.; et al. Cytoplasmic replication of Staphylococcus aureus upon phagosomal escape triggered by phenol-soluble modulin alpha. Cell. Microbiol. 2014, 16, 451–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Surewaard, B.G.J.; de Haas, C.J.C.; Vervoort, F.; Rigby, K.M.; DeLeo, F.R.; Otto, M.; van Strijp, J.A.G.; Nijland, R. Staphylococcal alpha-phenol soluble modulins contribute to neutrophil lysis after phagocytosis. Cell. Microbiol. 2013, 15, 1427–1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Armbruster, N.S.; Richardson, J.R.; Schreiner, J.; Klenk, J.; Gunter, M.; Kretschmer, D.; Poschel, S.; Schenke-Layland, K.; Kalbacher, H.; Clark, K.; et al. PSM peptides of Staphylococcus aureus activate the p38-CREB pathway in dendritic Cells, thereby modulating cytokine production and T cell priming. J. Immunol. 2016, 196, 1284–1292. [Google Scholar] [CrossRef] [Green Version]
  187. Armbruster, N.S.; Richardson, J.R.; Schreiner, J.; Klenk, J.; Gunter, M.; Autenrieth, S.E. Staphylococcus aureus PSM peptides induce tolerogenic dendritic cells upon treatment with ligands of extracellular and intracellular TLRs. Int. J. Med. Microbiol. 2016, 306, 666–674. [Google Scholar] [CrossRef]
  188. Cruciani, M.; Etna, M.P.; Camilli, R.; Giacomini, E.; Percario, Z.A.; Severa, M.; Sandini, S.; Rizzo, F.; Brandi, V.; Balsamo, G.; et al. Staphylococcus aureus Esx factors control human dendritic cell functions conditioning Th1/Th17 response. Front. Cell Infect Microbiol. 2017, 7, 330. [Google Scholar] [CrossRef]
  189. Miao, E.A.; Leaf, I.A.; Treuting, P.M.; Mao, D.P.; Dors, M.; Sarkar, A.; Warren, S.E.; Wewers, M.D.; Aderem, A. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat. Immunol. 2010, 11, 1136–1142. [Google Scholar] [CrossRef]
  190. Soe, Y.M.; Bedoui, S.; Stinear, T.P.; Hachani, A. Intracellular Staphylococcus aureus and host cell death pathways. Cell. Microbiol. 2021, 23, e13317. [Google Scholar] [CrossRef]
  191. Proctor, R.A.; van Langevelde, P.; Kristjansson, M.; Maslow, J.N.; Arbeit, R.D. Persistent and relapsing infections associated with small-colony variants of Staphylococcus aureus. Clin. Infect. Dis. 1995, 20, 95–102. [Google Scholar] [CrossRef]
  192. Kipp, F.; Ziebuhr, W.; Becker, K.; Krimmer, V.; Höβ, N.; Peters, G.; von Eiff, C. Detection of Staphylococcus aureus by 16S rRNA directed in situ hybridisation in a patient with a brain abscess caused by small colony variants. J. Neurol. Neurosurg Psychiatry 2003, 74, 1000–1002. [Google Scholar] [CrossRef]
  193. Schröder, A.; Kland, R.; Peschel, A.; von Eiff, C.; Aepfelbacher, M. Live cell imaging of phagosome maturation in Staphylococcus aureus infected human endothelial cells: Small colony variants are able to survive in lysosomes. Med. Microbiol. Immunol. 2006, 195, 185–194. [Google Scholar] [CrossRef] [PubMed]
  194. Garzoni, C.; Kelley, W.L. Staphylococcus aureus: New evidence for intracellular persistence. Trends Microbiol. 2009, 17, 59–65. [Google Scholar] [CrossRef] [PubMed]
  195. Fraunholz, M.; Sinha, B. Intracellular Staphylococcus aureus: Live-in and let die. Front. Cell. Infect. Microbiol. 2012, 2, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Cohen, M.L. Epidemiology of drug resistance: Implications for a post-antimicrobial era. Science 1992, 257, 1050–1055. [Google Scholar] [CrossRef] [PubMed]
  197. Dantes, R.; Mu, Y.; Belflower, R.; Aragon, D.; Dumyati, G.; Harrison, L.H.; Lessa, F.C.; Lynfield, R.; Nadle, J.; Petit, S.; et al. National burden of invasive methicillin-resistant Staphylococcus aureus infections, United States, 2011. JAMA Intern. Med. 2013, 173, 1970–1978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. DeLeo, F.R.; Diep, B.A.; Otto, M. Host defense and pathogenesis in Staphylococcus aureus infections. Infect. Dis. Clin. N. Am. 2009, 23, 17–34. [Google Scholar] [CrossRef] [Green Version]
  199. Guillemot, D. Antibiotic use in humans and bacterial resistance. Curr. Opin. Microbiol. 1999, 2, 494–498. [Google Scholar] [CrossRef]
  200. Ito, T.; Okuma, K.; Ma, X.X.; Yuzawa, H.; Hiramatsu, K. Insights on antibiotic resistance of Staphylococcus aureus from its whole genome: Genomic island SCC. Drug Resist. Updat. 2003, 6, 41–52. [Google Scholar] [CrossRef]
  201. Chambers, H.F.; Deleo, F.R. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat. Rev. Microbiol. 2009, 7, 629–641. [Google Scholar] [CrossRef]
  202. Theuretzbacher, U. Resistance drives antibacterial drug development. Curr. Opin. Pharmacol. 2011, 11, 433–438. [Google Scholar] [CrossRef]
  203. Peacock, S.J.; Paterson, G.K. Mechanisms of Methicillin Resistance in Staphylococcus aureus. Annu. Rev. Biochem. 2015, 84, 577–601. [Google Scholar] [CrossRef] [PubMed]
  204. Fuda, C.C.; Fisher, J.F.; Mobashery, S. Beta-lactam resistance in Staphylococcus aureus: The adaptive resistance of a plastic genome. Cell. Mol. Life Sci. 2005, 62, 2617–2633. [Google Scholar] [CrossRef] [PubMed]
  205. Llarrull, L.I.; Fisher, J.F.; Mobashery, S. Molecular basis and phenotype of methicillin resistance in Staphylococcus aureus and insights into new beta-lactams that meet the challenge. Antimicrob. Agents Chemother. 2009, 53, 4051–4063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Stryjewski, M.E.; Corey, G.R. Methicillin-resistant Staphylococcus aureus: An evolving pathogen. Clin. Infect. Dis. 2014, 58, S10–S19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Mangili, A.; Bica, I.; Snydman, D.R.; Hamer, D.H. Daptomycin-resistant, methicillin-resistant Staphylococcus aureus bacteremia. Clin. Infect. Dis. 2005, 40, 1058–1060. [Google Scholar] [CrossRef] [Green Version]
  208. Vikram, H.R.; Havill, N.L.; Koeth, L.M.; Boyce, J.M. Clinical progression of methicillin-resistant Staphylococcus aureus vertebral osteomyelitis associated with reduced susceptibility to daptomycin. J. Clin. Microbiol. 2005, 43, 5384–5387. [Google Scholar] [CrossRef] [Green Version]
  209. Hayden, M.K.; Rezai, K.; Hayes, R.A.; Lolans, K.; Quinn, J.P.; Weinstein, R.A. Development of Daptomycin resistance in vivo in methicillin-resistant Staphylococcus aureus. J. Clin. Microbiol. 2005, 43, 5285–5287. [Google Scholar] [CrossRef] [Green Version]
  210. Tran, T.T.; Munita, J.M.; Arias, C.A. Mechanisms of drug resistance: Daptomycin resistance. Ann. N. Y. Acad. Sci. 2015, 1354, 32–53. [Google Scholar] [CrossRef] [Green Version]
  211. Peleg, A.Y.; Miyakis, S.; Ward, D.V.; Earl, A.M.; Rubio, A.; Cameron, D.R.; Pillai, S.; Moellering, R.C., Jr.; Eliopoulos, G.M. Whole genome characterization of the mechanisms of daptomycin resistance in clinical and laboratory derived isolates of Staphylococcus aureus. PLoS ONE 2012, 7, e28316. [Google Scholar] [CrossRef]
  212. Thitiananpakorn, K.; Aiba, Y.; Tan, X.-E.; Watanabe, S.; Kiga, K.; Sato’o, Y.; Boonsiri, T.; Li, F.-Y.; Sasahara, T.; Taki, Y.; et al. Association of mprF mutations with cross-resistance to daptomycin and vancomycin in methicillin-resistant Staphylococcus aureus (MRSA). Sci. Rep. 2020, 10. [Google Scholar] [CrossRef]
  213. Bhuiyan, M.S.; Jiang, J.H.; Kostoulias, X.; Theegala, R.; Lieschke, G.J.; Peleg, A.Y. The resistance to host antimicrobial peptides in infections caused by daptomycin-resistant Staphylococcus aureus. Antibiotics 2021, 10, 96. [Google Scholar] [CrossRef] [PubMed]
  214. Sabat, A.J.; Tinelli, M.; Grundmann, H.; Akkerboom, V.; Monaco, M.; Del Grosso, M.; Errico, G.; Pantosti, A.; Friedrich, A.W. Daptomycin resistant Staphylococcus aureus clinical strain with novel non-synonymous mutations in the mprF and vraS genes: A new insight into daptomycin resistance. Front. Microbiol. 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Mishra, N.N.; Yang, S.J.; Sawa, A.; Rubio, A.; Nast, C.C.; Yeaman, M.R.; Bayer, A.S. Analysis of cell membrane characteristics of in vitro-selected daptomycin-resistant strains of methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2009, 53, 2312–2318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Camargo, I.L.; Neoh, H.M.; Cui, L.; Hiramatsu, K. Serial daptomycin selection generates daptomycin-nonsusceptible Staphylococcus aureus strains with a heterogeneous vancomycin-intermediate phenotype. Antimicrob. Agents Chemother. 2008, 52, 4289–4299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Chen, F.-J.; Lauderdale, T.-L.; Lee, C.-H.; Hsu, Y.-C.; Huang, I.W.; Hsu, P.-C.; Yang, C.-S. Effect of a Point Mutation in mprF on Susceptibility to Daptomycin, Vancomycin, and Oxacillin in an MRSA Clinical Strain. Front. Microbiol. 2018, 9, 1086. [Google Scholar] [CrossRef] [PubMed]
  218. Ohniwa, R.L.; Kitabayashi, K.; Morikawa, K. Alternative cardiolipin synthase Cls1 compensates for stalled Cls2 function in Staphylococcus aureus under conditions of acute acid stress. FEMS Microbiol. Lett. 2013, 338, 141–146. [Google Scholar] [CrossRef] [Green Version]
  219. Koprivnjak, T.; Zhang, D.; Ernst, C.M.; Peschel, A.; Nauseef, W.M.; Weiss, J.P. Characterization of Staphylococcus aureus cardiolipin synthases 1 and 2 and their contribution to accumulation of cardiolipin in stationary phase and within phagocytes. J. Bacteriol. 2011, 193, 4134–4142. [Google Scholar] [CrossRef] [Green Version]
  220. Cameron, D.R.; Mortin, L.I.; Rubio, A.; Mylonakis, E.; Moellering, R.C., Jr.; Eliopoulos, G.M.; Peleg, A.Y. Impact of daptomycin resistance on Staphylococcus aureus virulence. Virulence 2015, 6, 127–131. [Google Scholar] [CrossRef] [Green Version]
  221. Ernst, C.M.; Staubitz, P.; Mishra, N.N.; Yang, S.J.; Hornig, G.; Kalbacher, H.; Bayer, A.S.; Kraus, D.; Peschel, A. The bacterial defensin resistance protein MprF consists of separable domains for lipid lysinylation and antimicrobial peptide repulsion. PLoS Pathog. 2009, 5, e1000660. [Google Scholar] [CrossRef] [Green Version]
  222. Wu, X.; Xu, F. Dendritic cells during Staphylococcus aureus infection: Subsets and roles. J. Transl. Med. 2014, 12, 358. [Google Scholar] [CrossRef] [Green Version]
  223. Balraadjsing, P.P.; de Jong, E.C.; van Wamel, W.J.B.; Zaat, S.A.J. Dendritic cells internalize Staphylococcus aureus more efficiently than Staphylococcus epidermidis, but do not differ in induction of antigen-specific T Cell proliferation. Microorganisms 2019, 8, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Oldenburg, M.; Krüger, A.; Ferstl, R.; Kaufmann, A.; Nees, G.; Sigmund, A.; Bathke, B.; Lauterbach, H.; Suter, M.; Dreher, S.; et al. TLR13 recognizes bacterial 23S rRNA devoid of erythromycin resistance-forming modification. Science 2012, 337, 1111–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Patton, T.; Jiang, J.-H.; Lundie, R.; Bafit, M.; Gao, W.; Peleg, A.; O’Keeffe, M. Daptomycin resistant Staphylococcus aureus clinical isolates are poorly sensed by dendritic cells. Immunol. Cell. Biol. 2019, 98. [Google Scholar] [CrossRef]
  226. Cruciani, M.; Sandini, S.; Etna, M.P.; Giacomini, E.; Camilli, R.; Severa, M.; Rizzo, F.; Bagnoli, F.; Hiscott, J.; Coccia, E.M. Differential responses of human dendritic cells to live or inactivated Staphylococcus aureus: Impact on cytokine production and T helper expansion. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef]
  227. Spaan, A.N.; Surewaard, B.G.; Nijland, R.; van Strijp, J.A. Neutrophils versus Staphylococcus aureus: A biological tug of war. Annu Rev. Microbiol. 2013, 67, 629–650. [Google Scholar] [CrossRef] [Green Version]
  228. Guerra, F.E.; Borgogna, T.R.; Patel, D.M.; Sward, E.W.; Voyich, J.M. Epic Immune Battles of History: Neutrophils vs. Staphylococcus aureus. Front. Cell. Infect. Microbiol. 2017, 7, 286. [Google Scholar] [CrossRef] [PubMed]
  229. English, B.K.; Maryniw, E.M.; Talati, A.J.; Meals, E.A. Diminished macrophage inflammatory response to Staphylococcus aureus isolates exposed to daptomycin versus vancomycin or oxacillin. Antimicrob. Agents Chemother. 2006, 50, 2225–2227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Kelesidis, T. The interplay between daptomycin and the immune system. Front. Immunol. 2014, 5, 52. [Google Scholar] [CrossRef] [Green Version]
  231. Mishra, N.N.; McKinnell, J.; Yeaman, M.R.; Rubio, A.; Nast, C.C.; Chen, L.; Kreiswirth, B.N.; Bayer, A.S. In vitro cross-resistance to daptomycin and host defense cationic antimicrobial peptides in clinical methicillin-resistant Staphylococcus aureus isolates. Antimicrob. Agents Chemother. 2011, 55, 4012–4018. [Google Scholar] [CrossRef] [Green Version]
  232. Mishra, N.N.; Yang, S.-J.; Chen, L.; Muller, C.; Saleh-Mghir, A.; Kuhn, S.; Peschel, A.; Yeaman, M.R.; Nast, C.C.; Kreiswirth, B.N.; et al. Emergence of daptomycin resistance in daptomycin-naïve babbits with methicillin-resistant Staphylococcus aureus prosthetic joint Infection is associated with resistance to host defense cationic peptides and mprF polymorphisms. PLoS ONE 2013, 8, e71151. [Google Scholar] [CrossRef]
  233. Zasloff, M. Antimicrobial peptides of multicellular organisms. Nature 2002, 415, 389–395. [Google Scholar] [CrossRef] [PubMed]
  234. Piechota, M.; Kot, B.; Frankowska-Maciejewska, A.; Grużewska, A.; Woźniak-Kosek, A. Biofilm formation by Methicillin-Resistant and Methicillin-Sensitive Staphylococcus aureus strains from hospitalized patients in Poland. BioMed Res. Int. 2018, 2018, 4657396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Thurlow, L.R.; Hanke, M.L.; Fritz, T.; Angle, A.; Aldrich, A.; Williams, S.H.; Engebretsen, I.L.; Bayles, K.W.; Horswill, A.R.; Kielian, T. Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo. J. Immunol. 2011, 186, 6585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Ernst, C.M.; Kuhn, S.; Slavetinsky, C.J.; Krismer, B.; Heilbronner, S.; Gekeler, C.; Kraus, D.; Wagner, S.; Peschel, A. The lipid-modifying multiple peptide resistance factor is an oligomer consisting of distinct interacting synthase and flippase subunits. mBio 2015, 6, e02340-14. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic presentation of the changes in the innate immune response coincident with the development of antibiotic resistance in S. aureus. (a) Changes in membrane phospholipid composition with the evolution of antibiotic resistance in S. aureus. Emergence of MRSA with reduced susceptibility to VCM and DAP has been associated with gain-of-function, in which lysinylation of PG is increased [217,221]. This positively charged L-PG increases net positive charge on the CM and reduced affinity for VCM and DAP [221]. Increased cell wall thickening is also suggested to cause ineffective binding of DAP/VCM to the CM [218,219]. (b) In the innate immune response, S. aureus is recognised by PRR of immune cells and phagocytosed, leading to the activation of intracellular PRRs and upregulation of costimulatory markers [222]. Cytokines produced by innate immune cells further mobilise recruitment of immune cells for pathogen clearance [222]. Among the innate immune cells, Antigen Presenting cells (APCs) induce the activation of adaptive immunity through antigen presentation [223]. However, antibiotic resistant S. aureus can evade innate immune detection, thus preventing activation. Notably, antibiotic-resistant S. aureus can modulate DC activation by avoiding recognition via TLR2 and 13 [159,224]. In response to DAP-R MRSA, DCs produced reduced proinflammatory cytokines including TNFα, IL-6, and MIP-1β as well as decreased CD80 expression compared with DAP-sensitive MRSA [225]. MRSA can also suppress DC activation by inducing the production of immunosuppressive cytokine IL-10 [226]. Neutrophils are one of the most fundamental host innate immune effectors against S. aureus [227]. During infection, neutrophils are initially recruited to the site of infection, a process termed chemotaxis, followed by adhesion and phagocytosis of S. aureus [228]. On the other hand, mutation in membrane phospholipid biosynthesis gene (cls2) conferring DAP resistance allowed S. aureus to evade neutrophil chemotaxis and overcome the immune attack [11]. Macrophages, which are also professional phagocytes, can also successfully control and degrade S. aureus using a range of mechanisms, including TLR activation, upregulation of proinflammatory cytokines, and generation of reactive ROS and NO [135]. However, the development of antibiotic resistance in S. aureus led to a downregulation of TLR1, TLR2, TLR6, and TLR9 expression in macrophages and dampened TNFα secretion and NO synthesis, allowing them to survive and escape macrophages [229,230].
Figure 1. Schematic presentation of the changes in the innate immune response coincident with the development of antibiotic resistance in S. aureus. (a) Changes in membrane phospholipid composition with the evolution of antibiotic resistance in S. aureus. Emergence of MRSA with reduced susceptibility to VCM and DAP has been associated with gain-of-function, in which lysinylation of PG is increased [217,221]. This positively charged L-PG increases net positive charge on the CM and reduced affinity for VCM and DAP [221]. Increased cell wall thickening is also suggested to cause ineffective binding of DAP/VCM to the CM [218,219]. (b) In the innate immune response, S. aureus is recognised by PRR of immune cells and phagocytosed, leading to the activation of intracellular PRRs and upregulation of costimulatory markers [222]. Cytokines produced by innate immune cells further mobilise recruitment of immune cells for pathogen clearance [222]. Among the innate immune cells, Antigen Presenting cells (APCs) induce the activation of adaptive immunity through antigen presentation [223]. However, antibiotic resistant S. aureus can evade innate immune detection, thus preventing activation. Notably, antibiotic-resistant S. aureus can modulate DC activation by avoiding recognition via TLR2 and 13 [159,224]. In response to DAP-R MRSA, DCs produced reduced proinflammatory cytokines including TNFα, IL-6, and MIP-1β as well as decreased CD80 expression compared with DAP-sensitive MRSA [225]. MRSA can also suppress DC activation by inducing the production of immunosuppressive cytokine IL-10 [226]. Neutrophils are one of the most fundamental host innate immune effectors against S. aureus [227]. During infection, neutrophils are initially recruited to the site of infection, a process termed chemotaxis, followed by adhesion and phagocytosis of S. aureus [228]. On the other hand, mutation in membrane phospholipid biosynthesis gene (cls2) conferring DAP resistance allowed S. aureus to evade neutrophil chemotaxis and overcome the immune attack [11]. Macrophages, which are also professional phagocytes, can also successfully control and degrade S. aureus using a range of mechanisms, including TLR activation, upregulation of proinflammatory cytokines, and generation of reactive ROS and NO [135]. However, the development of antibiotic resistance in S. aureus led to a downregulation of TLR1, TLR2, TLR6, and TLR9 expression in macrophages and dampened TNFα secretion and NO synthesis, allowing them to survive and escape macrophages [229,230].
Antibiotics 11 00542 g001
Table 1. PRRs involved in S. aureus recognition.
Table 1. PRRs involved in S. aureus recognition.
PRR FamilyReceptorSpeciesCellular
Location
PAMPSignal AdapterReferences
HumanMouse
TLRTLR1/TLR2 heterodimer++Plasma MembraneTriacyl lipoproteinsMyD88/TIRAP[18,19,20,21,22]
TLR2/TLR6 heterodimer++Plasma
Membrane
Diacyl lipoprotein,
Lipopeptide,
Peptidoglycan, Phenol soluble modulin (PSM)
MyD88/TIRAP[18,19,20]
TLR7++EndosometRNAMyD88[23,24]
TLR8+EndosomessRNAMyD88[25,26]
TLR9++EndosomeUnmethylated CpG DNAMyD88[23,27]
TLR13+Endosome23s RNAMyD88[23]
CLRMBL++Plasma
Membrane
Teichoic acid [28,29,30]
NLRNOD2++CytoplasmPeptidoglycan, α, β-, and γ- hemolysins; Panton–Valentine leukocidin (PVL); Leukocidin A/B; Acylated lipopeptidesRIP2, MAVS[31,32,33]
RLRsRIG-I++CytoplasmCytoplasmic RNAMAVS[34,35]
CDScGAS STING++CytoplasmDouble stranded DNA, Cyclic dinucleotideSTING[36,37,38,39,40]
CDS, Cytoplasmic DNA sensors; MyD88, Myeloid differentiation factor 88; TIRAP, Toll–interleukin 1 receptor (TIR) domain-containing adapter protein; CpG, cytosine-phosphate-guanosine; RIP2, Receptor-interacting-serine/threonine-protein kinase 2; MAVS, Mitochondrial antiviral-signalling protein.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jahan, N.; Patton, T.; O’Keeffe, M. The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection. Antibiotics 2022, 11, 542. https://doi.org/10.3390/antibiotics11050542

AMA Style

Jahan N, Patton T, O’Keeffe M. The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection. Antibiotics. 2022; 11(5):542. https://doi.org/10.3390/antibiotics11050542

Chicago/Turabian Style

Jahan, Nazneen, Timothy Patton, and Meredith O’Keeffe. 2022. "The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection" Antibiotics 11, no. 5: 542. https://doi.org/10.3390/antibiotics11050542

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop