Next Article in Journal
Dual Blockade of TNF and IL-17A Inhibits Inflammation and Structural Damage in a Rat Model of Spondyloarthritis
Next Article in Special Issue
Molecular Methodologies for Improved Polymicrobial Sepsis Diagnosis
Previous Article in Journal
Human Amnion-Derived Mesenchymal Stromal Cells: A New Potential Treatment for Carbapenem-Resistant Enterobacterales in Decompensated Cirrhosis
Previous Article in Special Issue
The Interplay between Host Defense, Infection, and Clinical Status in Septic Patients: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Look Who’s Talking: Host and Pathogen Drivers of Staphylococcus epidermidis Virulence in Neonatal Sepsis

1
Centre for Molecular Medicine & Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
2
Women’s and Infants Research Foundation Laboratory, King Edward Memorial Hospital, Subiaco, WA 6008, Australia
3
Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, WA 6009, Australia
4
Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD 20892, USA
5
Neonatal Directorate, Child and Adolescent Health Service, Nedlands, WA 6009, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(2), 860; https://doi.org/10.3390/ijms23020860
Submission received: 22 November 2021 / Revised: 10 January 2022 / Accepted: 10 January 2022 / Published: 13 January 2022

Abstract

:
Preterm infants are at increased risk for invasive neonatal bacterial infections. S. epidermidis, a ubiquitous skin commensal, is a major cause of late-onset neonatal sepsis, particularly in high-resource settings. The vulnerability of preterm infants to serious bacterial infections is commonly attributed to their distinct and developing immune system. While developmentally immature immune defences play a large role in facilitating bacterial invasion, this fails to explain why only a subset of infants develop infections with low-virulence organisms when exposed to similar risk factors in the neonatal ICU. Experimental research has explored potential virulence mechanisms contributing to the pathogenic shift of commensal S. epidermidis strains. Furthermore, comparative genomics studies have yielded insights into the emergence and spread of nosocomial S. epidermidis strains, and their genetic and functional characteristics implicated in invasive disease in neonates. These studies have highlighted the multifactorial nature of S. epidermidis traits relating to pathogenicity and commensalism. In this review, we discuss the known host and pathogen drivers of S. epidermidis virulence in neonatal sepsis and provide future perspectives to close the gap in our understanding of S. epidermidis as a cause of neonatal morbidity and mortality.

1. Introduction

Approximately 10% of infants are born preterm (defined as all live births <37 completed weeks of gestational age; GA) each year with numbers rising [1]. At the same time, advances in neonatal medicine have led to improved survival of preterm infants, including those born very and extremely preterm (<32 weeks GA and <28 weeks GA, respectively) [2]. However, prolonged hospitalization and invasive life support interventions are associated with an elevated risk of developing serious and life-threatening neonatal infections [3]. Bloodstream infections are the most common type of healthcare-associated infections, accounting for 11–19% of neonatal mortality worldwide [4]. Neonatal sepsis can be classified into early-onset sepsis (<72 h of life; EOS), which is usually caused by vertical transmission of maternal pathogens (i.e., Group B Streptococcus (GBS) and Escherichia coli (E. coli)) before or during birth [5], and late-onset sepsis (3–28 days of life; LOS) caused by nosocomial or community-acquired pathogens after birth [6]. LOS affects approximately in 1 of 10 very preterm infants (<32 weeks GA) with incidences reaching as high as 60% in the most extremely immature infants [7]. Gram-positive organisms are the most frequently isolated and in high-resource settings coagulase-negative staphylococci (CONS) are the predominant pathogens [8], accounting for up to 78% of LOS episodes [9]. Among CONS species, Staphylococcus epidermidis (S. epidermidis) is the most commonly isolated LOS pathogen in preterm infants [10]. Furthermore, S. epidermidis can initiate or aggravate other inflammation-associated morbidities in neonates, such as bronchopulmonary dysplasia, white matter injury, and retinopathy of prematurity [11]. In addition to maturity and birth weight, LOS risk factors include central venous catheters (CVC) intravenous lipids, mechanical ventilation, as well as prolonged use of antibiotics [6,12,13]. Early enteral feeding with unpasteurized breastmilk and supplementation with probiotics can reduce the risk of LOS in preterm infants [12,13]. While associated with lower mortality rates compared to sepsis caused by Gram-negative pathogens [14,15], short and long-term sequalae of S. epidermidis sepsis survivors include neurodevelopmental impairment and cerebral palsy [11]. The impact of S. epidermidis sepsis on long-term cognitive functions in preterm infants is still inconclusive [16,17]; however, persistent activation of neonatal inflammatory pathways, such as NF-κB, by S. epidermidis antigens may result in inflammatory disorders culminating in periventricular white matter injury [18].

Diagnosis, Prevention, and Treatment of LOS

Rapid and accurate diagnosis of neonatal sepsis remains challenging. Infants with LOS usually display non-specific symptoms and clinical signs alone are notoriously unreliable [19]. The current gold standard for neonatal sepsis diagnosis are microbial blood cultures, which have a long turn-around time (12–48 h) [20]. Moreover, while blood cultures are specific, their sensitivity is highly dependent on the inoculated blood volume, which is limited (often less than 1 mL) in preterm infants and LOS often has low and intermittent levels of bacteraemia (7–121 colony-forming units (CFU)/mL) [20,21]. Among S. epidermidis LOS cases, one third presented with <50 CFU/mL [22]. As a ubiquitous skin commensal, S. epidermidis is also a common contaminant in culture-based diagnostics, which poses an additional challenge in the diagnosis of S. epidermidis sepsis [23].
Because of diagnostic deficiencies, broad-spectrum antibiotics are often administered to preterm infants upon first signs of systemic infections. Flucloxacillin, a penicillin, or vancomycin are most commonly administered [24]. Due to the emergence and global spread of nosocomial S. epidermidis strains with reduced sensitivity towards antibiotics [25,26], these infections may result in persistent bacteraemia in preterm infants, despite aggressive antibiotic treatment [27]. Among US NICUs, 78.6% of very preterm infants and 87% of extremely preterm infants receive antibiotics within the first 3 days of life [28]. The SCOUT study found that among NICU-admitted infants (34–39 weeks GA), only a small fraction (5%) treated with antibiotics had a culture-proven infection [29]. This suggests that unnecessary antibiotics use may occur in a large number of preterm neonates, which is associated with potential adverse outcomes for the infant, including impaired gut microbiome development/gut dysbiosis as well increased risk for subsequent bacterial infections, particularly necrotizing enterocolitis (NEC) [30,31].
The most effective prevention strategies of neonatal CONS sepsis include meticulous hand hygiene and early enteral feeding with unpasteurized breast milk [32]. Furthermore, neonatal skin care with topical emollients, such as coconut oil, maintains skin condition in very preterm infants during the first weeks of life [33]. Except for the use of probiotics, no adjunct therapies for the treatment and prevention of neonatal infections have shown direct benefit [34].
Understanding the unique host–pathogen interactions between preterm infants and nosocomial CONS infections, particularly the dominant organism, S. epidermidis, is essential for the development of alternative and targeted preventative, diagnostic, as well as therapeutic tools to combat neonatal infections with this opportunistic pathogen.

2. S. epidermidis: Friend and Foe

2.1. S. epidermidis as an Early and Ubiquitous Colonizer of the Newborn

S. epidermidis is a commensal bacterium and can be found ubiquitously on human skin and mucous membranes of the respiratory tract and intestine [35,36,37]. The human fetus may encounter S. epidermidis already in the uterus, as recently proposed by Stinson et al., who found Staphylococcus spp. present in amniotic fluid using high resolution full-length 16S rRNA gene sequencing [38]. The initial colonization of preterm infant skin and intestine with S. epidermidis likely commences immediately after birth via contact with parents, hospital staff and equipment [39,40]. In the case of preterm infants, premature rupture of membranes (PROM) and intra-amniotic infection (chorioamnionitis) furthermore lead to microbial exposure before birth in 25–30% of infants [41]. The development of the early life neonatal skin and gut microbiome is GA-dependent and partly shaped by mode of delivery, feeding, and administration of antibiotics [42]. S. epidermidis is abundant in the colostrum and breast milk of healthy women [43,44]. Breast milk of preterm infant mothers contains higher counts of S. epidermidis which harbour virulence-associated genes, which may be due to prolonged hospital exposure [45]. S. epidermidis colonization of the preterm infant gut is promoted by enteral feeding with unpasteurized mothers’ milk and limits the intestinal spread of NICU-associated S. epidermidis strains [46]. Notably, feeding with bovine colostrum immediately after birth improved the clearance and clinical responses to S. epidermidis sepsis in a preterm piglet model [47].
The microbiome of preterm infants, which spend a prolonged time in the hospital, has been suggested to be additionally shaped by the “NICU microbiome”, consisting of a low number of mostly skin-associated bacterial taxa [48]. As such, preterm infants generally exhibit lower abundances of commensal, obligate anaerobic bacteria, but a higher prevalence of nosocomial isolates and multi-drug resistant strains, with enrichment of S. epidermidis, Enterococcus faecalis, and Klebsiella pneumoniae compared to term infants [49,50]. Colonization with S. epidermidis has been shown to be established by multiple founder lineages which form specialized communities at different skin sites (i.e., moist, oily, and dry) in a dynamic manner with shifts in intra-species abundances during the first month of life [51,52]. This diversity is maintained under high selective pressure throughout life with S. epidermidis strains continuously being transmitted between different skin sites [52].

2.2. Establishing Host Tolerance to Commensal S. epidermidis

The concept of disease tolerance as a defence strategy against infectious agents in animals was championed by Medzhitov et al. as an alternative to disease avoidance and resistance, which are associated with increased energetic and immune cost [53]. More recently, the concept was revisited by Harbeson et al. through the perspective of neonatal host defences in early life [54]. In brief, the authors suggest that the first postnatal weeks of life constitute a state of heightened immunogenic susceptibility to bacterial pathogens where the neonatal host must carefully balance between energy expended for immune defences (i.e., disease resistance) and the energy required for growth and development (i.e., resulting in increased disease tolerance). This is supported, for instance, by the observation that neonates exhibit a higher bacterial burden (CFU/g lung) and a delayed clearance of bacteria compared to adults in a murine model of methicillin-resistant Staphylococcus aureus (S. aureus) (MRSA)-pneumonia [55].
Additionally, the neonatal period is a time of active adaptive immunosuppression, with high abundances of regulatory T cells (Tregs) present in neonatal skin as well as peripheral tissue (Figure 1) [56]. A specialized subset of Tregs induces tolerance to S. epidermidis colonization during neonatal skin development, but not during adulthood [57]. Moreover, colonization with S. epidermidis, but not S. aureus, heightens Treg responses in neonatal mice [58]. Of note, Tregs in preterm infants have a distinct phenotype with heightened immunosuppressive capacity compared to term infants [56], and are implicated in the development of bronchopulmonary dysplasia and may play a role in EOS [56,59] by temporarily down-regulating host immune functions and facilitating increased bacterial establishment. Other subsets of neonatal cells contribute to inhibit hyperinflammation in response to rapid commensal colonization of the skin and mucosal membranes after birth, including CD71+ cells (immature erythrocytes) and intestinal epithelial cells (IECs) [60,61]. In their immunosuppressive role, CD71+ cells are also implicated in compromising innate and adaptive immunity against bacterial infections, for instance, with Listeria monocytogenes and Bordetella pertussis [62,63]. However, in a murine model of neonatal polymicrobial sepsis, ablation of CD71+ cells did not alter sepsis mortality [64]. Immunosuppressive molecules, such as S100-type alarmins (i.e., S100A8 and S100A9) also play a role in the postnatal gut microbial development by preventing excessive inflammation, with S100-knockout mice showing higher neonatal sepsis mortality [65,66,67]. Notably, preterm infants show reduced S100A8/S100A9 levels compared to term infants [67].
In conclusion, transient neonatal immunosuppression is crucial to establish tolerance against neonatal commensals, but simultaneously contributes to the elevated neonatal susceptibility to infectious disease in early life. However, the specific role of immunosuppressive cell subsets and molecules in regard to early life S. epidermidis colonization remains to be fully elucidated.

2.3. Contribution of S. epidermidis to Host Defence

Apart from direct competition for nutrients and host adhesion sites, commensal bacteria produce specific metabolites and antimicrobial peptides to inhibit pathogenic colonization [68]. In its commensal role, S. epidermidis modulates the host immune system and shapes the development of the skin and nasal microbiome [37,69] by preventing host colonization by more virulent bacterial and fungal organisms, as well as viruses (Figure 1) [70,71,72]. S. aureus nasal colonization, for instance, is inhibited by commensal S. epidermidis through the production of biofilm-disrupting compounds, such as serine protease Esp, which degrade S. aureus-specific host receptor proteins [73,74]. In hospitalized neonates, nasal S. epidermidis colonization has been shown to protect against S. aureus colonization by a Esp-independent mechanism [75].
Toll-like receptor (TLR) 2 stimulation by S. epidermidis induces the production of host antimicrobial peptides by keratinocytes and nasal epithelial cells to boost host immunity [39,74,76]. In a recent study, Pastar et al. reported the elevated ability of skin-resident γδ T cells in killing intracellular S. aureus following stimulation with S. epidermidis, via cell-specific upregulating of host perforin-2 (P-2) production [69]. Moreover, S. epidermidis induces epidermal IL-17-producing CD8+ T cells (Tc17) to upregulate S100-type alarmins, which limits pathogen invasion and promotes wound healing [70,77].
By potentially reducing the survival of opportunistic pathogens such as S. aureus and Streptococcus pyogenes through the secretion of antimicrobially active phenol-soluble modulins (PSMs) [76,78], S. epidermidis may protect from disease, such as atopic dermatitis (AD) [79]. However, while in vitro studies using high concentrations of synthetic PSM peptides reported antimicrobial activities, in another study, only proteolytically cleaved but not intact peptides exhibited activity against S. pyogenes [80], with their in vivo role and biological significance still unclear [81]. Furthermore, a more recent study by Cau et al. found that S. epidermidis colonization was associated with more severe AD disease [82], which may be due to S100A8/A9-dependent alterations of the skin barrier proteins [83].
Antiviral activities of S. epidermidis have also been documented. For instance, nasal commensal S. epidermidis can act as a frontline protection against influenza A virus through the upregulation of IFN-λ production in the nasal epithelium [71].
Lastly, a commensal S. epidermidis strain has recently been suggested to protect from UV-induced skin neoplasia by inhibiting DNA polymerase activity selectively in tumour cells through producing a nucleobase analogue [84].

2.4. S. epidermidis as a Human Pathogen

Compared to S. aureus, an important community pathogen causing skin and soft tissue infections [85], S. epidermidis is considered a low virulence organism and is typically non-haemolytic in human blood. It also does not express potent toxins or exoenzymes known to be determinants of S. aureus pathogenicity [86,87].
PSMsare a family of amphipathic peptides produced by S. epidermidis with multifactorial roles in its commensal as well as pathogenic niches [81]. PSMs differ in their length and net charge, with Staphylococci species each having a specific PSM profile [88]. Known PSMs produced by S. epidermidis include PSMα, PSMδ, PSMε, δ-toxin (PSMγ), PSM-mec, and two PSMβ peptides (PSMβ1, PSMβ2), which have been attributed a range of roles, including biofilm formation and dispersion, evasion of host immunity, and interspecies competition [81,89]. While some PSMs are highly cytolytic (i.e., α-type PSMs and PSMδ), in S. epidermidis, these are expressed in lower amounts and with reduced activity [81].
In accordance with its role as a ubiquitous commensal, S. epidermidis rarely causes invasive disease in healthy adults, children, or even term-born infants. However, opportunistic healthcare-associated infections with S. epidermidis are on the rise among adult patients with indwelling devices [90]. Biomedical device-related S. epidermidis infections are often of chronic nature and difficult to treat with antibiotics due to S. epidermidis strong abilities to produce biofilm on biotic and abiotic surfaces [91,92,93]. As a persistent inhabitant of skin and mucosal tissue, S. epidermidis has evolved diverse mechanisms for host attachment and accumulation, which contribute to both commensalism and pathogenicity [86]. This includes host protein-specific surface adhesion molecules, non-specific adhesion mechanisms (i.e., hydrophobic interactions), cell-wall-anchored proteins/adhesins (i.e., microbial surface components recognizing adhesive matrix molecules; MSCRAMMs), as well as peptidoglycan-bound wall teichoic acid (WTA) [94]. The production of extracellular polymeric substances (EPS) is considered a key virulence factor of S. epidermidis and has been shown to facilitate its attachment to medical devices during hospital-associated infections [91,93].
The ability to produce a mature biofilm is facilitated by complex multi-step mechanisms and is dependent on the presence of selected genes, particularly the intercellular adhesion (icaADBC) operon and insertion element IS256 [95,96], which are involved in the synthesis of the main biofilm exopolysaccharide polysaccharide intercellular adhesin (PIA) [97], also known as poly-N-acetylglucosmine (PNAG), as well as accumulation-associated proteins, such as extracellular matrix binding protein (Embp), biofilm associated-homologous protein(Bhp), and accumulation-associated protein (Aap) [98,99,100]. S. epidermidis secreted peptidoglycan hydrolases (i.e., autolysin AltE and autolysin/adhesin Aae), which play a role in the cellular processes involved in bacterial proliferation (e.g., cell separation), also promote primary attachment to medical devices [101,102]. Furthermore, alternative morphotypes, assembled from extracellular DNA (eDNA, released from dying cells) and (lipo)proteins have been described [103,104].
During changing environmental conditions, S. epidermidis uses the accessory gene regulator (agr) quorum-sensing system to detect increases in population density [105,106]. The agr system is encoded by the agrBDCA operon and multiple different agr types have been described in S. epidermidis [107]. S. epidermidis quorum-sensing facilitates the cross-inhibition of S. aureus [108]. Moreover, the colonization of a host niche by multiple S. epidermidis agr types has recently been suggested to suppress the expression of virulence factors to maintain homeostasis [52].
S. epidermidis PSMs, which are under strict control of the agr operon [109], play putative roles in biofilm maturation as well as in vivo biofilm dissemination [110,111]. Notably, S. epidermidis biofilm formation in response to harsh environmental condition, is accompanied by an extensive change in gene expression which facilitates the change from planktonic growth to quiescent mode, characterized by low expression of virulence factors, such as PSMs and agr [112].
A variety of mechanisms are employed by S. epidermidis to evade physical and chemical threats elicited by host innate and adaptive immunity [113,114]. A versatile range of antimicrobial peptides (AMPs) are produced by the host as a first line of antibacterial defence on human skin [115]. While lower in total host defence proteins, neonatal skin harbours higher concentrations of AMPs compared to adult skin [116,117]. S. epidermidis biofilm formation is a key mechanism to evade host immunity [118], including host AMPs [119], complement compound C3b and immunoglobulin G (IgG) [120]. S. epidermidis has also developed AMP sensor systems (aps) [121], efflux pumps for specific bacteriocins (vraFG ABC transporters) [122], and extracellular proteases (i.e., metalloprotease SepA) [114] to provide protection from host AMPs, complement components, and avoid neutrophil killing.

3. S. epidermidis: Inside, Outside—Everywhere?

As a human commensal species, S. epidermidis is ubiquitously present in the community as well as in the hospital environment, where healthcare workers serve as an important nosocomial reservoir for S. epidermidis, including methicillin-resistant S. epidermidis (MRSE) strains [123]. While invasive procedures are assumed to be a common route of entry for S. epidermidis in preterm infants, not all cases of S. epidermidis LOS are device-related and specific bacterial DNA signatures are often undetectable from removed material [124].
Emerging evidence suggests that the gut is an important alternative reservoir of LOS-causing S. epidermidis in preterm infants with immature intestinal mucosal barriers and bacterial dysbiosis (Figure 2) [125]. Disturbances in the development of mouth, gut, and lung microbiomes of preterm infants can be caused by common empiric antibiotic exposure, lack of enteral feeding, as well as use of proton pump inhibitors [126,127]. A disturbed intestinal microbiome can impair the development of colonic Treg cells, resulting in dysregulated T cell responses upon bacterial challenge [128]. Innate immune responses and IL-17 producing type 3 innate lymphoid cells (ILC3) in particular, play a critical role in immune homeostasis of the intestinal tissue during the perinatal period [129]. Even short-term antibiotic exposure can decrease ILC3s and IL-17 levels in neonatal mice resulting in increased bacterial translocation from the gut lumen and heightened susceptibility to LOS [130]. A reduced abundance of ILC3s may be due to decreased TLR signalling by gut commensals. In septic preterm infants the gut microbiome is less diverse compared to matched controls. Particularly increased carriage of Staphylococcus spp. and Enterobacteria and lower numbers of Bifidobacteria [131], may impair epithelial barrier function [132,133].
Direct evidence of intestinal S. epidermidis causing LOS is scarce; however, the presence of S. epidermidis has been detected in ~40% of preterm infant meconium samples and ~90% of faecal samples (collected 7 days after birth) [134]. Moreover, Staphylococcus was found more abundant in preterm infant faecal samples before and during sepsis [131]. However, a study by Stewart et al. found that while the dominant taxa in the gut microbiome of preterm infants with LOS was usually identical to the causative LOS pathogen identified through blood culture, when sepsis with S. epidermidis was diagnosed, Klebsiella and Escherichia dominated the intestine [135]. Most recently, Golinska et al. have found that 25% of CONS strains isolated from the bloodstream of VLBW/LWB neonates were identical to the strains isolated from faeces of the same neonates at the same time [125].

4. Genetic Determinants of Virulence in Commensal and Invasive S. epidermidis

S. epidermidis comprises a large number of clonal lineages with a pan-genome of about 2.5 Mb, consisting of approximately 80% core genome (containing genes present in all strains) and 20% accessory genome (genes present in only a subset of strains) [136,137]. The S. epidermidis pan-genome is open, indicating that the species undergoes frequent horizontal gene transfer (HGT) [138]. Bacteriophages are the major vehicles for HGT in staphylococci, governing the evolution of new clonal lineages [139,140]. Unlike other bacterial species, S. epidermidis isolates are genetically diverse with a large collection of genetic determinants for resistances against antibiotics and host immune defence (i.e., AMPs) [137]. S. epidermidis shares approximately half of its genome with its more virulent cousin S. aureus, and interspecies HGT of mobile genomic elements has been well documented between the species, including genes encoding for metal detoxification and enhanced pathogenicity [141]. S. epidermidis is an important reservoir of drug resistance genes, which they can transfer to S. aureus and Enterococcus spp., thereby driving the emergence of more virulent hospital-associated strains, particularly methicillin-resistant S. aureus (MRSA) [142,143]. The high genetic heterogeneity and rapid adaption through intra- and interspecies HGT underlies S. epidermidis ability to colonize and persist in various host niches and host populations [38,142,144].

4.1. Nosocomial S. epidermidis Strains Implicated in Neonatal Infection

S. epidermidis isolates can be grouped into two main phylogenetic genotype clusters (clonal complexes), lineage A/C (comprising most nosocomial isolates) and lineage B (mostly community isolates) [136,141]. Both clusters display a high genome plasticity with specific functions, suggested by the presence of a high number of genes exclusive to each cluster [145].
Multi-locus sequence typing (MLST) allows tracking of the evolution of S. epidermidis populations, and rise of new variants through mutations and HGT [146]. In hospital environments, two sequence types (STs), ST2 and ST23, account for most adult clinical disease (i.e., bacteremia, CVC infection, septic arthritis) [147]. Furthermore, having an ST2 or ST5 S. epidermidis bacteraemia has been found to be an independent predictor of complicated bloodstream infection [148]. Neonates in the NICU are most frequently colonized with S. epidermidis strains belonging to ST2, ST5, ST59, and ST81, and among LOS-causing isolates, ST2 and ST5 are most commonly identified [144].
Since the advent of next-generation sequencing, a vast collection of clinical and commensal S. epidermidis isolates have been sequenced [54,142,149]. By analysing the genetic determinants of isolates associated with asymptomatic carriage and disease, numerous studies have aimed to discover discriminating markers of S. epidermidis invasiveness among adults and neonates [138,149]. While there is some evidence for S. epidermidis adaptations within the host [150] and some differentially distributed virulence factors may contribute to invasiveness, previous studies were not able to conclusively pinpoint a genetic factor solely associated with virulence vs. commensalism in S. epidermidis [136,137,138,145,151]. Of note, a recent study by Du et al. has described a mutation in the tarIJLM gene cluster, which inhibited S. epidermidis capacity to colonize epithelial cells but promoted binding to endothelial cells, through changes in wall-teichoic acid structures expressed [140]. This mutation facilitated bloodstream invasion and increased sepsis mortality in a mouse model and was found only among a small number of infection-associated S. epidermidis isolates (6.9–13.9%), with increased prevalence in ST2, ST5, ST10, ST23, and ST87 strains.
In the next paragraphs, we will discuss some recent comparative genomics studies in adult and, where available, neonatal clinical and commensal S. epidermidis isolates, and put their findings into the context of previous key functional studies of S. epidermidis virulence.

4.2. The Role of S. epidermidis Biofilm in Neonatal Invasive Disease

S. epidermidis virulence in hospital-acquired infections is thought to be driven mainly by their ability to colonize the surfaces of medical devices and form persisting biofilm, thereby evading the host immune responses and antibiotic treatment [118]. Indeed, biofilm-associated genes, such as the icaADBC operon and IS256 are more prevalent in distinct clonal complexes and ST types (i.e., ST27), comprising mostly clinical S. epidermidis strains [152], and have been suggested as potential markers of invasive S. epidermidis [153,154]. Notably, ica-positive S. epidermidis strains are less adapted to human skin colonization [155] and the production of PIA and AltE is associated with an increased potential to cause bacteraemia in a rat model of central venous catheter (CVC)-associated infection [101]. In neonatal cord blood, S. epidermidis strains positive for icaADBC inhibit the release of IL-6, which was not observed in icaADBC-negative strains [156].
While the agr quorum-sensing system of S. epidermidis controls several putative determinants of acute virulence such as PSMs, proteases, and lipases [86,157], it reduces S. epidermidis primary host attachment and biofilm forming capabilities, but increases the direct virulence through the production of delta-toxins [158]. In line with this, agr non-functionality is more common among S. epidermidis strains isolated from joint protheses infections and S. epidermidis agr deletion mutants have increased ability to cause in vivo infection in a rabbit model of indwelling medical device-related infection [159]. By regulating the expression of pro-inflammatory PSMs, agr-positive S. epidermidis elicit elevated host immune responses and have been suggested to have increased invasive capacity [94,113,156,160].
Among S. epidermidis isolates from neonatal blood, the vast majority (85–90%) are capable of producing biofilm [161,162]. However, the rate of biofilm production and the presence of icaADB and altE in S. epidermidis isolates from neonatal bloodstream infection (BSI) was comparable to commensal S. epidermidis isolates from neonatal nares [149]. Furthermore, the rates of biofilm formation were comparable between persistent and nonpersistent neonatal S. epidermidis isolates upon growth in low-glucose media but higher in persistent isolates when grown in total parenteral nutrition (9.23% glucose) [163]. Similarly, icaA and other biofilm-associated genes, including atlE, embp, mecA, and IS256, were found equally prevalent in isolates of adult catheter-related BSI isolates compared to commensal isolates from healthy individuals [164], suggesting that biofilm-forming capabilities are not a distinguishing feature of S. epidermidis invasiveness among clinical and commensal isolates.
Investigating the role of biofilm formation during in vivo infection remains challenging, especially in the neonatal context, with only few established mouse and rabbit models. Moreover, the functional redundancy of S. epidermidis biofilm mechanisms complicate such studies [94]. The contribution of biofilm formation in acute neonatal sepsis and the neonatal immune response to S. epidermidis biofilm remains to be fully elucidated. However, biofilm-released S. epidermidis, such as from biomedical devices, have been established as an important source of bacteria seeding the bloodstream and causing acute sepsis [110,160]. Furthermore, the attenuation of host defences, particularly bacterial phagocytosis, by biofilm-producing S. epidermidis has been reported [122,165,166] and may inhibit the efficient clearing of bacteraemia in neonates.

4.3. The Role of S. epidermidis Host Adhesion in Neonatal Invasive Disease

A range of other putative S. epidermidis virulence determinants are likely involved in the onset and progression of LOS in the preterm infant (Figure 3). Indeed, the presence of genes encoding for specific surface adhesions have been suggested as discriminative markers for S. epidermidis invasiveness and virulence among neonates and adults, including Staphylococcus epidermidis surface protein I (sesI) [149,167]. Interestingly, sesI is mostly absent in S. epidermidis from healthy individuals [167] and is exclusively present in strains belonging to disease-associated S. epidermidis, such as LOS-causing ST2 clones [168]. sesI is also more prevalent in S. epidermidis strains harbouring other virulence-related genes, such as biofilm-associated genes aap and IS256 in adults and neonates [149,168]. Using in vitro adhesion and aggregation assays, sesI was found to promote the initial stages of bacterial agglomeration and biofilm-formation [168]. However, the correlation of sesI with bacterial pathogenicity remains unclear and further studies in the neonatal setting are needed to understand their contribution to S. epidermidis virulence in vivo [169,170].
The Ser-Asp-rich fibrinogen binding protein-encoding genes sdrF, sdrG (Fbe), and sdrH contribute to S. epidermidis adhesion to extracellular host matrix molecules [171] and sdrF has been found enriched among neonatal bloodstream isolates (65%) compared to nasal isolates (11%) [149]. Notably, SdrF plays a key role in initiating driveline-associated S. epidermidis infections in a murine model and may facilitate transcutaneous entry of commensal S. epidermidis [165]. In a rat model of intravascular-catheter-associated infection, sdrG-positive S. epidermidis were more likely to cause bacteraemia and metastatic disease than sdrG-deficient mutants [166].
The role of other adhesion-associated surface proteins and enzymes Embp, Aap, Bhp, small basic protein (Sbp), and glycerol ester hydrolase (Geh) in facilitating S. epidermidis attachment to biotic and abiotic surfaces has been investigated in vitro [172]; however, their role in vivo is less well defined [173]. In conclusion, while some adhesion factors seem to be associated with bloodstream infections, their contribution to neonatal sepsis pathogenesis remains incompletely understood.

4.4. The Role of S. epidermidis Toxins in Neonatal Invasive Disease

While S. epidermidis biofilm production is considered a key virulence factor contributing to the prevalence of biomedical device-associated infections, S. epidermidis sepsis is acute in nature and other virulence factors as well as host immunity (or lack thereof) likely drive infections in the neonatal host (Figure 3) [11].
S. epidermidis is a low virulence organism with limited toxin production apart from PSMs [81]. Qin et al. have linked MRSE PSM-mec to bacterial survival in human whole blood and resistance to neutrophil killing [174]. Moreover, the secretion of PSM-mec toxins increased cytokine expression (IL-1β, TNF-α and the mouse IL-8 homologue CXCL1), leading to elevated sepsis severity in a murine model of sepsis [174]. The secretion of another PSM, haemolytic δ-toxin (PSMγ), by S. epidermidis, has been associated with the development of neonatal NEC [175]. However, there is no evidence yet for any specific virulence factor or toxin contributing to S. epidermidis sepsis pathogenesis in neonatal animal models using isogenic deletion mutants.

4.5. S. epidermidis Antibiotic Resistance and Neonatal Invasive Disease

Nosocomial S. epidermidis strains have adapted under selective pressures in the hospital environment, resulting in multiple resistance determinants and resistance-conferring mutations. A large majority of S. epidermidis isolates from preterm neonatal infections are MRSE, carrying the mecA gene on the staphylococcal cassette chromosome mec (SCCmec) [176]. Furthermore, resistance against other aminoglycosides (e.g., gentamicin), and macrolides (e.g., erythromycin and clindamycin) was reported recently among S. epidermidis isolates from NICUs across Europe [138,168,177]. Linezolid and rifampicin resistance can be conferred through mutations in ribosomal gene rpoB (RNA polymerase) and the presence of plasmid-derived cfr (RNA methyl transferase), whose prevalence are rising among S. epidermidis ICU strains [27,28,151]. A potential relationship between rifampicin resistance and vancomyin/teicoplanin resistance in S. epidermidis strains with dual rpoB mutations (D471E and I527M; most commonly found in ST2 and ST23 lineages), has been suggested [147]. This may be driven, in part, by device impregnation with antibiotics such as rifampicin, as lower rates of resistance has been observed in countries where this is not common practice [147,178]. However, S. epidermidis isolates from neonates remain largely susceptible to vancomycin and linezolid [168,177,179].
Nosocomial S. epidermidis strains are more likely to cause invasive disease in both adults and neonates, and can disseminate between hospitals and even between countries [180,181]. In a recent study using comparative genomics, S. epidermidis isolates from adult prosthetic joint infections more frequently harboured resistance against antibiotics, including β-lactams and aminoglycosides, and chlorhexidine, compared to commensal nasal isolates from patients with planned surgery [178]. However, Morgenstern et al. found no difference in the treatment cure rate of device-related infection caused by methicillin/multi-drug resistant vs. susceptible S. epidermidis [182]. Moreover, in neonates, the presence of antibiotic resistance had no effect on neonatal C-reactive protein (CRP) levels [183], suggesting that antibiotic resistance does not influence the clinical course of neonatal LOS. Currently, the impact of different antibiotic resistances in S. epidermidis on neonatal sepsis onset, disease progression and outcome require further research and clarification.

5. Phenotypic Characteristics of S. epidermidis Clinical Isolates

Upon host invasion, S. epidermidis undergoes a rapid change of environment, including alterations in nutrient availability, variations in temperature, salt and pH, the presence of circulating host immune cells, pro-inflammatory molecules, and antibiotics. Invasive S. epidermidis isolates have demonstrated ability to transition from growth on skin to growth in blood or in the presence of blood components [184]. In line with their elevated pathogenic potential, S. epidermidis strains of the A/C lineage display increased ability to resist oxidative stress, evade host immunity and resist antibiotics in conditions mimicking blood infection, compared to less virulent B lineage strains [145]. Furthermore, growth rate in the presence of human plasma has been reported as a key predictor of whether an isolated S. epidermidis strain originated from true bacteriemia vs. contamination [184,185].
Méric et al. found that disease-associated S. epidermidis isolates induce higher levels of IL-8 production by blood cells (but not keratinocytes), compared to carriage-associated isolates [138]. S. epidermidis also induces higher IL-8 production by small airway epithelial cells in vitro compared to the more virulent S. aureus [177]. This may contribute to the development of bronchopulmonary dysplasia by promoting the persistent migration of inflammatory cells into the neonatal lungs [177].
A trend towards reduced toxicity with disease-associated S. epidermidis isolates was observed in vitro, as determined by an vesicle lysis test specific for staphylococcal PSM toxins [138]. This is in line with previous findings, which report a correlation between reduced bacterial toxicity and more severe disease in studies of MRSA bacteraemia and pneumonia [179,186], which has been suggested to be a trade-off for increased fitness in human serum in these strains [187]. Underlying the observed association between lower toxicity and increased invasiveness among clinical S. epidermidis, may be the predominance of agr-negative S. epidermidis strains among invasive isolates [159], which produce less PSMs and have increased biofilm-forming capacities [105,158] and hence, are more frequently implicated in biomedical device-associated infections.

6. S. epidermidis Gene Expression during Neonatal Host Invasion

Genotyping and phenotypically characterizing clinical S. epidermidis isolates is key in tracking the emergence of novel strains and lineages associated with nosocomial infections, as well as their antibiotic resistance, to guide appropriate treatment. Unfortunately, this is usually only done for genetic studies of nosocomial pathogens rather than as a part of clinical routine. Moreover, these analyses do not consider bacterial gene expression, posttranslational modifications, or novel gene mutations with unknown impact on the virulence phenotype. Ultimately, the virulence factors actively driving infection are likely dependent on the unique in vivo interplay between host and bacterium, which include not only direct bacterial virulence, but the host immunological and metabolic environment as well as the local host microbiota [188,189,190].
Invasion of host tissue constitutes a substantial change of environment for (commensal) pathogens. As such, bacterial exposure to blood and blood components is accompanied by a major ‘metabolic reset’, with the upregulation of bacterial genes for amino acid biosynthesis, transport, and metabolism [191]. Human plasma has been suggested as the main driver of S. epidermidis transcriptional responses as well as biofilm formation in a human blood challenge model [192]. Furthermore, the availability of exogeneous essential nutrients is crucial for S. epidermidis growth and biofilm formation [193,194]. The host strongly limits availability of nutrients, such as iron, that are essential for bacterial growth, a phenomenon that has been termed “host nutritional immunity” [195]. Hence, it is not surprising that genes encoding for iron sequestration have been found to be rapidly upregulated by S. epidermidis biofilm upon blood exposure [196]. Genes encoding for biotin metabolism, an essential cofactor in central pathways for bacterial survival, are also significantly upregulated in S. epidermidis in response to blood and plasma and may comprise a therapeutic target to inhibit S. epidermidis growth [192].
The only study, to our knowledge, investigating the transcriptional responses of S. epidermidis from infection isolates was conducted on adult patients with postoperative endophthalmitis [197]. This work identified iron and pyruvate metabolic genes and staphylococcal toxin SE1634 as potential contributors of S. epidermidis pathogenesis during endophthalmitis [197]. However, RNA-sequencing in this study was not performed on the bacteria directly isolated from the infection site, but after growth on blood agar plates, which does not completely reflect in vivo conditions.
Studies analysing the whole S. epidermidis transcriptome upon interaction with human blood remain scarce, focusing primarily on S. epidermidis biofilm virulence [198,199,200,201]. Furthermore, there are no studies to date using neonatal S. epidermidis challenge models to understand bacterial transcriptional responses to blood in this vulnerable host population.

7. The Neonatal Host as an Important Contributor to S. epidermidis Virulence

Neonatal blood cell immune responses elicited by clinical and reference S. epidermidis strains (live or heat-inactivated) have been investigated in a collection of in vitro (term and preterm neonatal blood samples) and in vivo (murine) studies, as previously reviewed by Dong et al. [11]. Briefly, the neonatal immune response against S. epidermidis is mediated by innate immune responses, characterized by high levels of pro-inflammatory cytokines (i.e., IL-6, IL-8, TNF-α), which are GA-dependent, and lack regulatory control. As recently reported by Hibbert et al., sepsis risk in neonates may be driven by a stark initial hyperinflammatory immune response combined with simultaneous immunosuppression caused, amongst other factors, by Tregs, MDSCs, and T cell inhibition [202]. While some aspects of preterm immunity may have a reduced functional capacity upon pathogenic stimulation (i.e., AMPs, reactive oxygen species, complement elements) compared to term infants and adults, S. epidermidis phagocytosis by preterm monocytes was not observed to be dysfunctional [11,203].
Of note, cytokine responses to S. epidermidis have recently been shown to be impaired in the whole blood of preterm infants who develop LOS, before clinical onset of their disease [198], suggesting that functional differences in immune responses play a role in the development of preterm S. epidermidis LOS.
S. epidermidis sepsis isolates may induce a greater oxidative burst compared to non-clinical reference strains; however, Dong and Speer et al. have stressed that the strain characteristics available are often unspecific [11]. When compared to in vitro challenge with other sepsis-causing pathogens such as E. coli or GBS, S. epidermidis induces similar or lower pro-inflammatory responses [199].
We are aware of four studies, to date, investigating the (preterm) neonatal host immune response and neurodevelopmental outcomes using a mouse model of S. epidermidis sepsis [200,204,205,206]. All studies demonstrated inoculum dose-dependent (TLR2-dependent and independent) inflammation with markedly increased pro-inflammatory cytokines and chemokines in blood and central nervous system (CNS; i.e., brain or cerebral spinal fluid; CSF), resulting in white and grey matter injury post challenge in one study. This heightened inflammatory state may explain the CNS injury associated with S. epidermidis infections in preterm infant [207]. Interestingly, S. epidermidis infection resulted in increased susceptibility to hypoxia-induced ischemia only in male but not female mice [200]. All studies used S. epidermidis 1457, an invasive clinical strain obtained from an adult catheter infection [201].
On a transcriptional level, LOS (regardless of causative pathogen) has been shown to induce a significant shift in gene expression, with elevated expression of IFN-α/β, IFN-γ, IL-1, and IL-6 pathways among very preterm infants [208]. Notably, among VLBW preterm infants, different gene expression clusters (‘endotypes’) corresponding to those detected in Gram-positive sepsis and Gram-negative sepsis were observed [209]. Cord blood monocytes from very preterm and term infants challenged with live S. epidermidis and E. coli shared a transcriptional response, driven mainly by TLR/NF-κB/TREM-1 signalling [210]. However, E. coli-stimulated, but not S. epidermidis-stimulated neonatal monocytes showed a GA-independent upregulation of IFN genes [210].
Collectively, these studies suggest that in addition to preterm infants exhibiting distinct immune capacities compared to term infant and adults [211,212], S. epidermidis elicits a potent and specific immune response in preterm neonates, compared to term infants. That alone, however, does not explain the unique susceptibility of preterm infants to S. epidermidis infections and their long-term adverse outcomes, as only a subset of the preterm infant population will develop serious infection in response to exposure to NICU S. epidermidis strains. Hence, the response of S. epidermidis to exposure and invasion of the preterm neonatal host may play a significant part in this interplay and understanding the bacterial drivers of preterm neonatal invasion in vivo will likely shed new insights into this tug-of-war.

8. Conclusions and Future Perspective

The adaptation of S. epidermidis is highly dynamic and directly shaped by the specific selection pressures encountered in the host environment. Notably, within-host evolution of S. epidermidis has been observed during the course of prolonged infection, which can impact virulence and antibiotic susceptibility [213]. Through their successful adaptation to harsh conditions, (nosocomial) S. epidermidis isolates may have a particularly diverse repertoire of commensal virulence factors readily employed upon host invasion. While there are some markers of virulence more frequently found in infection-associated vs. carriage-associated strains, the pathogenic potential of any given strain cannot be accurately predicted based on genetic analyses. The pathogenicity of S. epidermidis as an opportunistic pathogen is strongly influenced by the capacities of the (immunocompromised) host immune response. While preterm immunity has been shown to diverge from that in adults, and even the term infant counterpart, our understanding of the neonatal immune response to S. epidermidis infection remains incomplete. Of note, pathogen-specific neonatal (transcriptional) responses have been observed in several studies, indicating that virulence determinants beyond conserved bacterial surface structures modulate host immune responses. Considering that most preterm infants are faced with similar risks and exposure to S. epidermidis in the NICU, the host-inherent drivers of S. epidermidis sepsis development still need to be determined. What is needed is a comprehensive understanding of which S. epidermidis virulence determinants are contributing to invasive neonatal infections in vivo.
Using novel approaches, such as dual RNA-seq [214], host–pathogen interactions between the murine host and the opportunistic pathogens S. aureus and Streptococcus pneumoniae have recently been characterized [215,216,217,218] and provided new insights into the infection environment. Consequently, it has become evident, that in vitro gene expression studies are not representative of the complex in vivo host–pathogen interplay [217,218]. These studies showed that bacterial virulence gene expression is site-specific, with tissue-tropic virulence phenotypes in vivo [217,218] that are partly driven by the availability of different carbon sources [217]. Host (transcriptional) responses were shown to be isolate-specific and furthermore, host-intrinsic resistance directly related to pathogen gene expression during infection [218].
Blood remains a difficult biological site for analysing host–bacterial interactions during disease [217], given the low bacterial presence, even in culture-positive sepsis. Moreover, neonatal (sepsis) blood samples are particularly limited. However, with advances in the resolution of molecular techniques, such as next generation sequencing platforms, proteomics, and metabolomics, future research may be well suited to tackle these questions and decipher the unique interplay between the preterm neonatal host and the commensal pathogen S. epidermidis.

Author Contributions

Conceptualization, I.A.J.; Writing—Original Draft Preparation, I.A.J.; Writing—Review & Editing, M.O., A.J.C. and T.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by funding from the Wesfarmers Centre of Vaccines and Infectious Diseases (WCVID) and the Intramural Research Program of the National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH).

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. Chawanpaiboon, S.; Vogel, J.P.; Moller, A.-B.; Lumbiganon, P.; Petzold, M.; Hogan, D.; Landoulsi, S.; Jampathong, N.; Kongwattanakul, K.; Laopaiboon, M.; et al. Global, Regional, and National Estimates of Levels of Preterm Birth in 2014: A Systematic Review and Modelling Analysis. Lancet Glob. Health 2019, 7, e37–e46. [Google Scholar] [CrossRef] [Green Version]
  2. Horbar, J.D.; Edwards, E.M.; Greenberg, L.T.; Morrow, K.A.; Soll, R.F.; Buus-Frank, M.E.; Buzas, J.S. Variation in Performance of Neonatal Intensive Care Units in the United States. JAMA Pediatrics 2017, 171, e164396. [Google Scholar] [CrossRef] [PubMed]
  3. Zingg, W.; Hopkins, S.; Gayet-Ageron, A.; Holmes, A.; Sharland, M.; Suetens, C.; Almeida, M.; Asembergiene, J.; Borg, M.A.; Budimir, A.; et al. Health-Care-Associated Infections in Neonates, Children, and Adolescents: An Analysis of Paediatric Data from the European Centre for Disease Prevention and Control Point-Prevalence Survey. Lancet Infect. Dis. 2017, 17, 381–389. [Google Scholar] [CrossRef]
  4. Fleischmann-Struzek, C.; Goldfarb, D.M.; Schlattmann, P.; Schlapbach, L.J.; Reinhart, K.; Kissoon, N. The Global Burden of Paediatric and Neonatal Sepsis: A Systematic Review. Lancet Respir. Med. 2018, 6, 223–230. [Google Scholar] [CrossRef]
  5. Stoll, B.J.; Puopolo, K.M.; Hansen, N.I.; Sánchez, P.J.; Bell, E.F.; Carlo, W.A.; Cotten, C.M.; D’Angio, C.T.; Kazzi, S.N.J.; Poindexter, B.B.; et al. Early-Onset Neonatal Sepsis 2015 to 2017, the Rise of Escherichia Coli, and the Need for Novel Prevention Strategies. JAMA Pediatr. 2020, 174, e200593. [Google Scholar] [CrossRef] [PubMed]
  6. Dong, Y.; Speer, C.P. Late-Onset Neonatal Sepsis: Recent Developments. Arch. Dis. Child Fetal Neonatal Ed. 2015, 100, F257–F263. [Google Scholar] [CrossRef] [Green Version]
  7. Boghossian, N.S.; Page, G.P.; Bell, E.F.; Stoll, B.J.; Murray, J.C.; Cotten, C.M.; Shankaran, S.; Walsh, M.C.; Laptook, A.R.; Newman, N.S.; et al. Late-Onset Sepsis in Very Low Birth Weight Infants from Singleton and Multiple-Gestation Births. J. Pediatr. 2013, 162, 1120–1124. [Google Scholar] [CrossRef] [Green Version]
  8. Stoll, B.J.; Hansen, N.; Fanaroff, A.A.; Wright, L.L.; Carlo, W.A.; Ehrenkranz, R.A.; Lemons, J.A.; Donovan, E.F.; Stark, A.R.; Tyson, J.E.; et al. Late-Onset Sepsis in Very Low Birth Weight Neonates: The Experience of the NICHD Neonatal Research Network. Pediatrics 2002, 110, 285–291. [Google Scholar] [CrossRef]
  9. van den Hoogen, A.; Gerards, L.J.; Verboon-Maciolek, M.A.; Fleer, A.; Krediet, T.G. Long-Term Trends in the Epidemiology of Neonatal Sepsis and Antibiotic Susceptibility of Causative Agents. Neonatology 2010, 97, 22–28. [Google Scholar] [CrossRef] [PubMed]
  10. Marchant, E.A.; Boyce, G.K.; Sadarangani, M.; Lavoie, P.M. Neonatal Sepsis Due to Coagulase-Negative Staphylococci. Clin. Dev. Immunol. 2013, 2013, 586076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Dong, Y.; Speer, C.P.; Glaser, K. Beyond Sepsis: Staphylococcus Epidermidis Is an Underestimated but Significant Contributor to Neonatal Morbidity. Virulence 2018, 9, 621–633. [Google Scholar] [CrossRef] [Green Version]
  12. Patel, A.L.; Johnson, T.J.; Engstrom, J.L.; Fogg, L.F.; Jegier, B.J.; Bigger, H.R.; Meier, P.P. Impact of Early Human Milk on Sepsis and Health-Care Costs in Very Low Birth Weight Infants. J. Perinatol. 2013, 33, 514–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zhang, G.-Q.; Hu, H.-J.; Liu, C.-Y.; Shakya, S.; Li, Z.-Y. Probiotics for Preventing Late-Onset Sepsis in Preterm Neonates: A PRISMA-Compliant Systematic Review and Meta-Analysis of Randomized Controlled Trials. Medicine 2016, 95, e2581. [Google Scholar] [CrossRef]
  14. Fleischmann, C.; Reichert, F.; Cassini, A.; Horner, R.; Harder, T.; Markwart, R.; Tröndle, M.; Savova, Y.; Kissoon, N.; Schlattmann, P.; et al. Global Incidence and Mortality of Neonatal Sepsis: A Systematic Review and Meta-Analysis. Arch. Dis. Child. 2021, 106, 745–752. [Google Scholar] [CrossRef]
  15. Dong, Y.; Glaser, K.; Speer, C.P. Late-Onset Sepsis Caused by Gram-Negative Bacteria in Very Low Birth Weight Infants: A Systematic Review. Expert Rev. Anti Infect. Ther. 2019, 17, 177–188. [Google Scholar] [CrossRef]
  16. Alshaikh, B.; Yee, W.; Lodha, A.; Henderson, E.; Yusuf, K.; Sauve, R. Coagulase-Negative Staphylococcus Sepsis in Preterm Infants and Long-Term Neurodevelopmental Outcome. J. Perinatol. 2014, 34, 125–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Zonnenberg, I.A.; van Dijk, J.; van den Dungen, F.A.M.; Vermeulen, R.J.; van Weissenbruch, M.M. The Prognostic Value of NIRS in Preterm Infants with (Suspected) Late-Onset Sepsis in Relation to Long Term Outcome: A Pilot Study. PLoS ONE 2019, 14, e0220044. [Google Scholar] [CrossRef] [Green Version]
  18. Dong, Y.; Speer, C.P. The Role of Staphylococcus Epidermidis in Neonatal Sepsis: Guarding Angel or Pathogenic Devil? Int. J. Med Microbiol. 2014, 304, 513–520. [Google Scholar] [CrossRef] [PubMed]
  19. Healy, C.M.; Palazzi, D.L.; Edwards, M.S.; Campbell, J.R.; Baker, C.J. Features of Invasive Staphylococcal Disease in Neonates. Pediatrics 2004, 114, 953–961. [Google Scholar] [CrossRef]
  20. Iroh Tam, P.-Y.; Bendel, C.M. Diagnostics for Neonatal Sepsis: Current Approaches and Future Directions. Pediatr. Res. 2017, 82, 574–583. [Google Scholar] [CrossRef]
  21. Stranieri, I.; Kanunfre, K.A.; Rodrigues, J.C.; Yamamoto, L.; Nadaf, M.I.V.; Palmeira, P.; Okay, T.S. Assessment and Comparison of Bacterial Load Levels Determined by Quantitative Amplifications in Blood Culture-Positive and Negative Neonatal Sepsis. Rev. Inst. Med. Trop. Sao Paulo 2018, 60, e61. [Google Scholar] [CrossRef]
  22. Kellogg, J.A.; Manzella, J.P.; Bankert, D.A. Frequency of Low-Level Bacteremia in Children from Birth to Fifteen Years of Age. J. Clin. Microbiol. 2000, 38, 2181–2185. [Google Scholar] [CrossRef] [PubMed]
  23. Kleinschmidt, S.; Huygens, F.; Faoagali, J.; Rathnayake, I.U.; Hafner, L.M. Staphylococcus Epidermidis as a Cause of Bacteremia. Future Microbiol. 2015, 10, 1859–1879. [Google Scholar] [CrossRef] [Green Version]
  24. Garrido, F.; Allegaert, K.; Arribas, C.; Villamor, E.; Raffaeli, G.; Paniagua, M.; Cavallaro, G. Variations in Antibiotic Use and Sepsis Management in Neonatal Intensive Care Units: A European Survey. Antibiotics 2021, 10, 1046. [Google Scholar] [CrossRef] [PubMed]
  25. Li, X.; Arias, C.A.; Aitken, S.L.; Galloway Peña, J.; Panesso, D.; Chang, M.; Diaz, L.; Rios, R.; Numan, Y.; Ghaoui, S.; et al. Clonal Emergence of Invasive Multidrug-Resistant Staphylococcus Epidermidis Deconvoluted via a Combination of Whole-Genome Sequencing and Microbiome Analyses. Clin. Infect. Dis. 2018, 67, 398–406. [Google Scholar] [CrossRef] [PubMed]
  26. Huber, S.; Knoll, M.A.; Berktold, M.; Würzner, R.; Brindlmayer, A.; Weber, V.; Posch, A.E.; Mrazek, K.; Lepuschitz, S.; Ante, M.; et al. Genomic and Phenotypic Analysis of Linezolid-Resistant Staphylococcus Epidermidis in a Tertiary Hospital in Innsbruck, Austria. Microorganisms 2021, 9, 1023. [Google Scholar] [CrossRef]
  27. Linder, N.; Hernandez, A.; Amit, L.; Klinger, G.; Ashkenazi, S.; Levy, I. Persistent Coagulase-Negative Staphylococci Bacteremia in Very-Low-Birth-Weight Infants. Eur. J. Pediatr. 2011, 170, 989–995. [Google Scholar] [CrossRef]
  28. Flannery, D.D.; Ross, R.K.; Mukhopadhyay, S.; Tribble, A.C.; Puopolo, K.M.; Gerber, J.S. Temporal Trends and Center Variation in Early Antibiotic Use Among Premature Infants. JAMA Netw. Open 2018, 1, e180164. [Google Scholar] [CrossRef] [Green Version]
  29. Cantey, J.B.; Wozniak, P.S.; Sánchez, P.J. Prospective Surveillance of Antibiotic Use in the Neonatal Intensive Care Unit: Results from the SCOUT Study. Pediatr. Infect. Dis. J. 2015, 34, 267–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Russell, J.T.; Lauren Ruoss, J.; de la Cruz, D.; Li, N.; Bazacliu, C.; Patton, L.; McKinley, K.L.; Garrett, T.J.; Polin, R.A.; Triplett, E.W.; et al. Antibiotics and the Developing Intestinal Microbiome, Metabolome and Inflammatory Environment in a Randomized Trial of Preterm Infants. Sci. Rep. 2021, 11, 1943. [Google Scholar] [CrossRef]
  31. Ting, J.Y.; Roberts, A.; Sherlock, R.; Ojah, C.; Cieslak, Z.; Dunn, M.; Barrington, K.; Yoon, E.W.; Shah, P.S. Canadian Neonatal Network Investigators Duration of Initial Empirical Antibiotic Therapy and Outcomes in Very Low Birth Weight Infants. Pediatrics 2019, 143, e20182286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ramasethu, J. Prevention and Treatment of Neonatal Nosocomial Infections. Matern. Health Neonatol. Perinatol. 2017, 3, 5. [Google Scholar] [CrossRef] [Green Version]
  33. Strunk, T.; Pupala, S.; Hibbert, J.; Doherty, D.; Patole, S. Topical Coconut Oil in Very Preterm Infants: An Open-Label Randomised Controlled Trial. Neonatology 2018, 113, 146–151. [Google Scholar] [CrossRef]
  34. Schüller, S.S.; Kramer, B.W.; Villamor, E.; Spittler, A.; Berger, A.; Levy, O. Immunomodulation to Prevent or Treat Neonatal Sepsis: Past, Present, and Future. Front. Pediatr. 2018, 6, 199. [Google Scholar] [CrossRef]
  35. Garcia-Gutierrez, E.; Walsh, C.J.; Sayavedra, L.; Diaz-Calvo, T.; Thapa, D.; Ruas-Madiedo, P.; Mayer, M.J.; Cotter, P.D.; Narbad, A. Genotypic and Phenotypic Characterization of Fecal Staphylococcus Epidermidis Isolates Suggests Plasticity to Adapt to Different Human Body Sites. Front. Microbiol. 2020, 11, 688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The Human Skin Microbiome. Nat. Rev. Microbiol. 2018, 16, 143–155. [Google Scholar] [CrossRef] [PubMed]
  37. Liu, Q.; Liu, Q.; Meng, H.; Lv, H.; Liu, Y.; Liu, J.; Wang, H.; He, L.; Qin, J.; Wang, Y.; et al. Staphylococcus Epidermidis Contributes to Healthy Maturation of the Nasal Microbiome by Stimulating Antimicrobial Peptide Production. Cell Host Microbe 2020, 27, 68–78.e5. [Google Scholar] [CrossRef]
  38. Stinson, L.F.; Boyce, M.C.; Payne, M.S.; Keelan, J.A. The Not-so-Sterile Womb: Evidence That the Human Fetus Is Exposed to Bacteria Prior to Birth. Front. Microbiol. 2019, 10, 1124. [Google Scholar] [CrossRef]
  39. DiGiulio, D.B.; Romero, R.; Amogan, H.P.; Kusanovic, J.P.; Bik, E.M.; Gotsch, F.; Kim, C.J.; Erez, O.; Edwin, S.; Relman, D.A. Microbial Prevalence, Diversity and Abundance in Amniotic Fluid during Preterm Labor: A Molecular and Culture-Based Investigation. PLoS ONE 2008, 3, e3056. [Google Scholar] [CrossRef]
  40. Gómez, M.; Moles, L.; Melgar, A.; Ureta, N.; Bustos, G.; Fernández, L.; Rodríguez, J.M.; Jiménez, E. Early Gut Colonization of Preterm Infants: Effect of Enteral Feeding Tubes. J. Pediatr. Gastroenterol. Nutr. 2016, 62, 893–900. [Google Scholar] [CrossRef]
  41. Goldenberg, R.L.; Culhane, J.F.; Iams, J.D.; Romero, R. Epidemiology and Causes of Preterm Birth. Lancet 2008, 371, 75–84. [Google Scholar] [CrossRef]
  42. Yasmin, F.; Tun, H.M.; Konya, T.B.; Guttman, D.S.; Chari, R.S.; Field, C.J.; Becker, A.B.; Mandhane, P.J.; Turvey, S.E.; Subbarao, P.; et al. Cesarean Section, Formula Feeding, and Infant Antibiotic Exposure: Separate and Combined Impacts on Gut Microbial Changes in Later Infancy. Front. Pediatr. 2017, 5, 200. [Google Scholar] [CrossRef]
  43. Damaceno, Q.S.; Souza, J.P.; Nicoli, J.R.; Paula, R.L.; Assis, G.B.; Figueiredo, H.C.; Azevedo, V.; Martins, F.S. Evaluation of Potential Probiotics Isolated from Human Milk and Colostrum. Probiotics Antimicrob. Proteins 2017, 9, 371–379. [Google Scholar] [CrossRef] [PubMed]
  44. Lyons, K.E.; Ryan, C.A.; Dempsey, E.M.; Ross, R.P.; Stanton, C. Breast Milk, a Source of Beneficial Microbes and Associated Benefits for Infant Health. Nutrients 2020, 12, 1039. [Google Scholar] [CrossRef] [PubMed]
  45. Soeorg, H.; Metsvaht, T.; Eelmäe, I.; Metsvaht, H.K.; Treumuth, S.; Merila, M.; Ilmoja, M.-L.; Lutsar, I. Coagulase-Negative Staphylococci in Human Milk from Mothers of Preterm Compared with Term Neonates. J. Hum. Lact. 2017, 33, 329–340. [Google Scholar] [CrossRef]
  46. Soeorg, H.; Treumuth, S.; Metsvaht, H.K.; Eelmäe, I.; Merila, M.; Ilmoja, M.-L.; Lutsar, I.; Metsvaht, T. Higher Intake of Coagulase-Negative Staphylococci from Maternal Milk Promotes Gut Colonization with MecA-Negative Staphylococcus Epidermidis in Preterm Neonates. J. Perinatol. 2018, 38, 1344–1352. [Google Scholar] [CrossRef]
  47. Bæk, O.; Brunse, A.; Nguyen, D.N.; Moodley, A.; Thymann, T.; Sangild, P.T. Diet Modulates the High Sensitivity to Systemic Infection in Newborn Preterm Pigs. Front. Immunol. 2020, 11, 1019. [Google Scholar] [CrossRef]
  48. Brooks, B.; Olm, M.R.; Firek, B.A.; Baker, R.; Geller-McGrath, D.; Reimer, S.R.; Soenjoyo, K.R.; Yip, J.S.; Dahan, D.; Thomas, B.C.; et al. The Developing Premature Infant Gut Microbiome Is a Major Factor Shaping the Microbiome of Neonatal Intensive Care Unit Rooms. Microbiome 2018, 6, 112. [Google Scholar] [CrossRef] [Green Version]
  49. Gibson, M.K.; Wang, B.; Ahmadi, S.; Burnham, C.-A.D.; Tarr, P.I.; Warner, B.B.; Dantas, G. Developmental Dynamics of the Preterm Infant Gut Microbiota and Antibiotic Resistome. Nat. Microbiol. 2016, 1, 16024. [Google Scholar] [CrossRef]
  50. Gasparrini, A.J.; Wang, B.; Sun, X.; Kennedy, E.A.; Hernandez-Leyva, A.; Ndao, I.M.; Tarr, P.I.; Warner, B.B.; Dantas, G. Metagenomic Signatures of Early Life Hospitalization and Antibiotic Treatment in the Infant Gut Microbiota and Resistome Persist Long after Discharge. Nat. Microbiol. 2019, 4, 2285–2297. [Google Scholar] [CrossRef]
  51. Sharon, I.; Morowitz, M.J.; Thomas, B.C.; Costello, E.K.; Relman, D.A.; Banfield, J.F. Time Series Community Genomics Analysis Reveals Rapid Shifts in Bacterial Species, Strains, and Phage during Infant Gut Colonization. Genome Res. 2013, 23, 111–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zhou, W.; Spoto, M.; Hardy, R.; Guan, C.; Fleming, E.; Larson, P.J.; Brown, J.S.; Oh, J. Host-Specific Evolutionary and Transmission Dynamics Shape the Functional Diversification of Staphylococcus Epidermidis in Human Skin. Cell 2020, 180, 454–470.e18. [Google Scholar] [CrossRef] [PubMed]
  53. Medzhitov, R.; Schneider, D.S.; Soares, M.P. Disease Tolerance as a Defense Strategy. Science 2012, 335, 936–941. [Google Scholar] [CrossRef] [Green Version]
  54. Harbeson, D.; Ben-Othman, R.; Amenyogbe, N.; Kollmann, T.R. Outgrowing the Immaturity Myth: The Cost of Defending from Neonatal Infectious Disease. Front. Immunol. 2018, 9, 1077. [Google Scholar] [CrossRef] [Green Version]
  55. Fitzpatrick, E.A.; You, D.; Shrestha, B.; Siefker, D.; Patel, V.S.; Yadav, N.; Jaligama, S.; Cormier, S.A. A Neonatal Murine Model of MRSA Pneumonia. PLoS ONE 2017, 12, e0169273. [Google Scholar] [CrossRef]
  56. Pagel, J.; Twisselmann, N.; Rausch, T.K.; Waschina, S.; Hartz, A.; Steinbeis, M.; Olbertz, J.; Nagel, K.; Steinmetz, A.; Faust, K.; et al. Increased Regulatory T Cells Precede the Development of Bronchopulmonary Dysplasia in Preterm Infants. Front. Immunol. 2020, 11, 565257. [Google Scholar] [CrossRef] [PubMed]
  57. Scharschmidt, T.C.; Vasquez, K.S.; Truong, H.-A.; Gearty, S.V.; Pauli, M.L.; Nosbaum, A.; Gratz, I.K.; Otto, M.; Moon, J.J.; Liese, J.; et al. A Wave of Regulatory T Cells into Neonatal Skin Mediates Tolerance to Commensal Microbes. Immunity 2015, 43, 1011–1021. [Google Scholar] [CrossRef] [Green Version]
  58. Leech, J.M.; Dhariwala, M.O.; Lowe, M.M.; Chu, K.; Merana, G.R.; Cornuot, C.; Weckel, A.; Ma, J.M.; Leitner, E.G.; Gonzalez, J.R.; et al. Toxin-Triggered Interleukin-1 Receptor Signaling Enables Early-Life Discrimination of Pathogenic versus Commensal Skin Bacteria. Cell Host Microbe 2019, 26, 795–809.e5. [Google Scholar] [CrossRef]
  59. Pagel, J.; Hartz, A.; Figge, J.; Gille, C.; Eschweiler, S.; Petersen, K.; Schreiter, L.; Hammer, J.; Karsten, C.M.; Friedrich, D.; et al. Regulatory T Cell Frequencies Are Increased in Preterm Infants with Clinical Early-Onset Sepsis. Clin. Exp. Immunol. 2016, 185, 219–227. [Google Scholar] [CrossRef] [Green Version]
  60. He, Y.-M.; Li, X.; Perego, M.; Nefedova, Y.; Kossenkov, A.V.; Jensen, E.A.; Kagan, V.; Liu, Y.-F.; Fu, S.-Y.; Ye, Q.-J.; et al. Transitory Presence of Myeloid-Derived Suppressor Cells in Neonates Is Critical for Control of Inflammation. Nat. Med. 2018, 24, 224–231. [Google Scholar] [CrossRef]
  61. Grzywa, T.M.; Nowis, D.; Golab, J. The Role of CD71+ Erythroid Cells in the Regulation of the Immune Response. Pharmacol. Ther. 2021, 228, 107927. [Google Scholar] [CrossRef] [PubMed]
  62. Elahi, S.; Vega-López, M.A.; Herman-Miguel, V.; Ramírez-Estudillo, C.; Mancilla-Ramírez, J.; Motyka, B.; West, L.; Oyegbami, O. CD71+ Erythroid Cells in Human Neonates Exhibit Immunosuppressive Properties and Compromise Immune Response Against Systemic Infection in Neonatal Mice. Front Immunol. 2020, 11, 597433. [Google Scholar] [CrossRef] [PubMed]
  63. Namdar, A.; Koleva, P.; Shahbaz, S.; Strom, S.; Gerdts, V.; Elahi, S. CD71+ Erythroid Suppressor Cells Impair Adaptive Immunity against Bordetella Pertussis. Sci. Rep. 2017, 7, 7728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wynn, J.L.; Scumpia, P.O.; Stocks, B.T.; Romano-Keeler, J.; Alrifai, M.W.; Liu, J.-H.; Kim, A.S.; Alford, C.E.; Matta, P.; Weitkamp, J.-H.; et al. Neonatal CD71+ Erythroid Cells Do Not Modify Murine Sepsis Mortality. J. Immunol. 2015, 195, 1064–1070. [Google Scholar] [CrossRef] [Green Version]
  65. Pirr, S.; Richter, M.; Fehlhaber, B.; Pagel, J.; Härtel, C.; Roth, J.; Vogl, T.; Viemann, D. High Amounts of S100-Alarmins Confer Antimicrobial Activity on Human Breast Milk Targeting Pathogens Relevant in Neonatal Sepsis. Front Immunol. 2017, 8, 1822. [Google Scholar] [CrossRef] [Green Version]
  66. Ulas, T.; Pirr, S.; Fehlhaber, B.; Bickes, M.S.; Loof, T.G.; Vogl, T.; Mellinger, L.; Heinemann, A.S.; Burgmann, J.; Schöning, J.; et al. S100-Alarmin-Induced Innate Immune Programming Protects Newborn Infants from Sepsis. Nat. Immunol. 2017, 18, 622–632. [Google Scholar] [CrossRef]
  67. Willers, M.; Ulas, T.; Völlger, L.; Vogl, T.; Heinemann, A.S.; Pirr, S.; Pagel, J.; Fehlhaber, B.; Halle, O.; Schöning, J.; et al. S100A8 and S100A9 Are Important for Postnatal Development of Gut Microbiota and Immune System in Mice and Infants. Gastroenterology 2020, 159, 2130–2145.e5. [Google Scholar] [CrossRef]
  68. Dzidic, M.; Boix-Amorós, A.; Selma-Royo, M.; Mira, A.; Collado, M.C. Gut Microbiota and Mucosal Immunity in the Neonate. Med. Sci. 2018, 6, 56. [Google Scholar] [CrossRef] [Green Version]
  69. Pastar, I.; O’Neill, K.; Padula, L.; Head, C.R.; Burgess, J.L.; Chen, V.; Garcia, D.; Stojadinovic, O.; Hower, S.; Plano, G.V.; et al. Staphylococcus Epidermidis Boosts Innate Immune Response by Activation of Gamma Delta T Cells and Induction of Perforin-2 in Human Skin. Front. Immunol. 2020, 11, 2253. [Google Scholar] [CrossRef]
  70. Naik, S.; Bouladoux, N.; Linehan, J.L.; Han, S.-J.; Harrison, O.J.; Wilhelm, C.; Conlan, S.; Himmelfarb, S.; Byrd, A.L.; Deming, C.; et al. Commensal-Dendritic-Cell Interaction Specifies a Unique Protective Skin Immune Signature. Nature 2015, 520, 104–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Kim, H.J.; Jo, A.; Jeon, Y.J.; An, S.; Lee, K.-M.; Yoon, S.S.; Choi, J.Y. Nasal Commensal Staphylococcus Epidermidis Enhances Interferon-λ-Dependent Immunity against Influenza Virus. Microbiome 2019, 7, 80. [Google Scholar] [CrossRef] [Green Version]
  72. Nakatsuji, T.; Chen, T.H.; Narala, S.; Chun, K.A.; Two, A.M.; Yun, T.; Shafiq, F.; Kotol, P.F.; Bouslimani, A.; Melnik, A.V.; et al. Antimicrobials from Human Skin Commensal Bacteria Protect against Staphylococcus Aureus and Are Deficient in Atopic Dermatitis. Sci. Transl. Med. 2017, 9, eaah4680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Iwase, T.; Uehara, Y.; Shinji, H.; Tajima, A.; Seo, H.; Takada, K.; Agata, T.; Mizunoe, Y. Staphylococcus Epidermidis Esp Inhibits Staphylococcus Aureus Biofilm Formation and Nasal Colonization. Nature 2010, 465, 346–349. [Google Scholar] [CrossRef] [PubMed]
  74. Sugimoto, S.; Iwamoto, T.; Takada, K.; Okuda, K.-I.; Tajima, A.; Iwase, T.; Mizunoe, Y. Staphylococcus Epidermidis Esp Degrades Specific Proteins Associated with Staphylococcus Aureus Biofilm Formation and Host-Pathogen Interaction. J. Bacteriol. 2013, 195, 1645–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Lee, D.C.; Kananurak, A.; Tran, M.T.; Connolly, P.A.; Polage, C.R.; Iwase, T.; Bevins, C.L.; Underwood, M.A. Bacterial Colonization of the Hospitalized Newborn: Competition Between Staphylococcus Aureus and Staphylococcus Epidermidis. Pediatr. Infect. Dis. J. 2019, 38, 682–686. [Google Scholar] [CrossRef]
  76. Cogen, A.L.; Yamasaki, K.; Muto, J.; Sanchez, K.M.; Crotty Alexander, L.; Tanios, J.; Lai, Y.; Kim, J.E.; Nizet, V.; Gallo, R.L. Staphylococcus Epidermidis Antimicrobial Delta-Toxin (Phenol-Soluble Modulin-Gamma) Cooperates with Host Antimicrobial Peptides to Kill Group A Streptococcus. PLoS ONE 2010, 5, e8557. [Google Scholar] [CrossRef]
  77. Linehan, J.L.; Harrison, O.J.; Han, S.-J.; Byrd, A.L.; Vujkovic-Cvijin, I.; Villarino, A.V.; Sen, S.K.; Shaik, J.; Smelkinson, M.; Tamoutounour, S.; et al. Non-Classical Immunity Controls Microbiota Impact on Skin Immunity and Tissue Repair. Cell 2018, 172, 784–796.e18. [Google Scholar] [CrossRef] [Green Version]
  78. Cogen, A.L.; Yamasaki, K.; Sanchez, K.M.; Dorschner, R.A.; Lai, Y.; MacLeod, D.T.; Torpey, J.W.; Otto, M.; Nizet, V.; Kim, J.E.; et al. Selective Antimicrobial Action Is Provided by Phenol-Soluble Modulins Derived from Staphylococcus Epidermidis, a Normal Resident of the Skin. J. Invest. Dermatol. 2010, 130, 192–200. [Google Scholar] [CrossRef] [Green Version]
  79. Laborel-Préneron, E.; Bianchi, P.; Boralevi, F.; Lehours, P.; Fraysse, F.; Morice-Picard, F.; Sugai, M.; Sato’o, Y.; Badiou, C.; Lina, G.; et al. Effects of the Staphylococcus Aureus and Staphylococcus Epidermidis Secretomes Isolated from the Skin Microbiota of Atopic Children on CD4+ T Cell Activation. PLoS ONE 2015, 10, e0141067. [Google Scholar] [CrossRef] [Green Version]
  80. Joo, H.-S.; Cheung, G.Y.C.; Otto, M. Antimicrobial Activity of Community-Associated Methicillin-Resistant Staphylococcus Aureus Is Caused by Phenol-Soluble Modulin Derivatives. J. Biol. Chem. 2011, 286, 8933–8940. [Google Scholar] [CrossRef] [Green Version]
  81. Cheung, G.Y.C.; Joo, H.-S.; Chatterjee, S.S.; Otto, M. Phenol-Soluble Modulins--Critical Determinants of Staphylococcal Virulence. FEMS Microbiol. Rev. 2014, 38, 698–719. [Google Scholar] [CrossRef] [PubMed]
  82. Cau, L.; Williams, M.R.; Butcher, A.M.; Nakatsuji, T.; Kavanaugh, J.S.; Cheng, J.Y.; Shafiq, F.; Higbee, K.; Hata, T.R.; Horswill, A.R.; et al. Staphylococcus epidermidis Protease EcpA Can Be a Deleterious Component of the Skin Microbiome in Atopic Dermatitis. J. Allergy Clin. Immunol. 2021, 147, 955–966.e16. [Google Scholar] [CrossRef]
  83. Kim, M.J.; Im, M.A.; Lee, J.-S.; Mun, J.Y.; Kim, D.H.; Gu, A.; Kim, I.S. Effect of S100A8 and S100A9 on Expressions of Cytokine and Skin Barrier Protein in Human Keratinocytes. Mol. Med. Rep. 2019, 20, 2476–2483. [Google Scholar] [CrossRef]
  84. Nakatsuji, T.; Chen, T.H.; Butcher, A.M.; Trzoss, L.L.; Nam, S.-J.; Shirakawa, K.T.; Zhou, W.; Oh, J.; Otto, M.; Fenical, W.; et al. A Commensal Strain of Staphylococcus epidermidis Protects against Skin Neoplasia. Sci. Adv. 2018, 4, eaao4502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. DeLeo, F.R.; Otto, M.; Kreiswirth, B.N.; Chambers, H.F. Community-Associated Meticillin-Resistant Staphylococcus Aureus. Lancet 2010, 375, 1557–1568. [Google Scholar] [CrossRef] [Green Version]
  86. Otto, M. Staphylococcus epidermidis—the “accidental” Pathogen. Nat. Rev. Microbiol. 2009, 7, 555–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Otto, M. Staphylococcus Aureus Toxins. Curr. Opin. Microbiol. 2014, 17, 32–37. [Google Scholar] [CrossRef] [Green Version]
  88. Otto, M. Phenol-Soluble Modulins. Int. J. Med. Microbiol. 2014, 304, 164–169. [Google Scholar] [CrossRef] [Green Version]
  89. Le, K.Y.; Villaruz, A.E.; Zheng, Y.; He, L.; Fisher, E.L.; Nguyen, T.H.; Ho, T.V.; Yeh, A.J.; Joo, H.-S.; Cheung, G.Y.C.; et al. Role of Phenol-Soluble Modulins in Staphylococcus epidermidis Biofilm Formation and Infection of Indwelling Medical Devices. J. Mol. Biol. 2019, 431, 3015–3027. [Google Scholar] [CrossRef]
  90. Otto, M. Staphylococcus epidermidis: A Major Player in Bacterial Sepsis? Future Microbiol. 2017, 12, 1031–1033. [Google Scholar] [CrossRef] [PubMed]
  91. Otto, M. Molecular Basis of Staphylococcus epidermidis Infections. Semin. Immunopathol. 2012, 34, 201–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Zheng, Y.; He, L.; Asiamah, T.K.; Otto, M. Colonization of Medical Devices by Staphylococci. Environ. Microbiol. 2018, 20, 3141–3153. [Google Scholar] [CrossRef] [Green Version]
  93. Sabaté Brescó, M.; Harris, L.G.; Thompson, K.; Stanic, B.; Morgenstern, M.; O’Mahony, L.; Richards, R.G.; Moriarty, T.F. Pathogenic Mechanisms and Host Interactions in Staphylococcus epidermidis Device-Related Infection. Front. Microbiol. 2017, 8, 1401. [Google Scholar] [CrossRef] [Green Version]
  94. Otto, M. Staphylococcal Biofilms. Microbiol. Spectr. 2018, 6, 6.4.27. [Google Scholar] [CrossRef] [PubMed]
  95. Büttner, H.; Mack, D.; Rohde, H. Structural Basis of Staphylococcus Epidermidis Biofilm Formation: Mechanisms and Molecular Interactions. Front Cell Infect. Microbiol. 2015, 5, 14. [Google Scholar] [CrossRef] [Green Version]
  96. Heilmann, C.; Schweitzer, O.; Gerke, C.; Vanittanakom, N.; Mack, D.; Götz, F. Molecular Basis of Intercellular Adhesion in the Biofilm-Forming Staphylococcus Epidermidis. Mol. Microbiol. 1996, 20, 1083–1091. [Google Scholar] [CrossRef] [PubMed]
  97. Mack, D.; Fischer, W.; Krokotsch, A.; Leopold, K.; Hartmann, R.; Egge, H.; Laufs, R. The Intercellular Adhesin Involved in Biofilm Accumulation of Staphylococcus Epidermidis Is a Linear Beta-1,6-Linked Glucosaminoglycan: Purification and Structural Analysis. J. Bacteriol. 1996, 178, 175–183. [Google Scholar] [CrossRef] [Green Version]
  98. Christner, M.; Franke, G.C.; Schommer, N.N.; Wendt, U.; Wegert, K.; Pehle, P.; Kroll, G.; Schulze, C.; Buck, F.; Mack, D.; et al. The Giant Extracellular Matrix-Binding Protein of Staphylococcus Epidermidis Mediates Biofilm Accumulation and Attachment to Fibronectin. Mol. Microbiol. 2010, 75, 187–207. [Google Scholar] [CrossRef]
  99. Rohde, H.; Burdelski, C.; Bartscht, K.; Hussain, M.; Buck, F.; Horstkotte, M.A.; Knobloch, J.K.-M.; Heilmann, C.; Herrmann, M.; Mack, D. Induction of Staphylococcus Epidermidis Biofilm Formation via Proteolytic Processing of the Accumulation-Associated Protein by Staphylococcal and Host Proteases. Mol. Microbiol. 2005, 55, 1883–1895. [Google Scholar] [CrossRef]
  100. Tormo, M.Á.; Knecht, E.; Götz, F.; Lasa, I.; Penadés, J.R. Bap-Dependent Biofilm Formation by Pathogenic Species of Staphylococcus: Evidence of Horizontal Gene Transfer? Microbiology 2005, 151, 2465–2475. [Google Scholar] [CrossRef] [Green Version]
  101. Rupp, M.E.; Fey, P.D.; Heilmann, C.; Götz, F. Characterization of the Importance of Staphylococcus Epidermidis Autolysin and Polysaccharide Intercellular Adhesin in the Pathogenesis of Intravascular Catheter-Associated Infection in a Rat Model. J. Infect. Dis. 2001, 183, 1038–1042. [Google Scholar] [CrossRef] [Green Version]
  102. Heilmann, C.; Thumm, G.; Chhatwal, G.S.; Hartleib, J.; Uekötter, A.; Peters, G. Identification and Characterization of a Novel Autolysin (Aae) with Adhesive Properties from Staphylococcus Epidermidis. Microbiology 2003, 149, 2769–2778. [Google Scholar] [CrossRef] [Green Version]
  103. Campoccia, D.; Montanaro, L.; Arciola, C.R. Extracellular DNA (EDNA). A Major Ubiquitous Element of the Bacterial Biofilm Architecture. Int. J. Mol. Sci. 2021, 22, 9100. [Google Scholar] [CrossRef]
  104. Qin, Z.; Ou, Y.; Yang, L.; Zhu, Y.; Tolker-Nielsen, T.; Molin, S.; Qu, D. Role of Autolysin-Mediated DNA Release in Biofilm Formation of Staphylococcus Epidermidis. Microbiology 2007, 153, 2083–2092. [Google Scholar] [CrossRef] [Green Version]
  105. Olson, M.E.; Todd, D.A.; Schaeffer, C.R.; Paharik, A.E.; Van Dyke, M.J.; Büttner, H.; Dunman, P.M.; Rohde, H.; Cech, N.B.; Fey, P.D.; et al. Staphylococcus epidermidis agr Quorum-Sensing System: Signal Identification, Cross Talk, and Importance in Colonization. J. Bacteriol. 2014, 196, 3482–3493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Otto, M.; Süssmuth, R.; Jung, G.; Götz, F. Structure of the Pheromone Peptide of the Staphylococcus epidermidis agr System. FEBS Lett. 1998, 424, 89–94. [Google Scholar] [CrossRef] [Green Version]
  107. Dufour, P.; Jarraud, S.; Vandenesch, F.; Greenland, T.; Novick, R.P.; Bes, M.; Etienne, J.; Lina, G. High Genetic Variability of the Agr Locus in Staphylococcus Species. J. Bacteriol. 2002, 184, 1180–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Otto, M.; Echner, H.; Voelter, W.; Götz, F. Pheromone Cross-Inhibition between Staphylococcus aureus and Staphylococcus epidermidis. Infect. Immun. 2001, 69, 1957–1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Vuong, C.; Dürr, M.; Carmody, A.B.; Peschel, A.; Klebanoff, S.J.; Otto, M. Regulated Expression of Pathogen-Associated Molecular Pattern Molecules in Staphylococcus epidermidis: Quorum-Sensing Determines pro-Inflammatory Capacity and Production of Phenol-Soluble Modulins. Cell Microbiol. 2004, 6, 753–759. [Google Scholar] [CrossRef]
  110. Wang, R.; Khan, B.A.; Cheung, G.Y.C.; Bach, T.-H.L.; Jameson-Lee, M.; Kong, K.-F.; Queck, S.Y.; Otto, M. Staphylococcus epidermidis Surfactant Peptides Promote Biofilm Maturation and Dissemination of Biofilm-Associated Infection in Mice. J. Clin. Invest. 2011, 121, 238–248. [Google Scholar] [CrossRef]
  111. Periasamy, S.; Joo, H.-S.; Duong, A.C.; Bach, T.-H.L.; Tan, V.Y.; Chatterjee, S.S.; Cheung, G.Y.C.; Otto, M. How Staphylococcus aureus Biofilms Develop Their Characteristic Structure. Proc. Natl. Acad. Sci. USA 2012, 109, 1281–1286. [Google Scholar] [CrossRef] [Green Version]
  112. Yao, Y.; Sturdevant, D.E.; Otto, M. Genomewide Analysis of Gene Expression in Staphylococcus epidermidis Biofilms: Insights into the Pathophysiology of S. epidermidis Biofilms and the Role of Phenol-Soluble Modulins in Formation of Biofilms. J. Infect. Dis. 2005, 191, 289–298. [Google Scholar] [CrossRef] [Green Version]
  113. Vuong, C.; Voyich, J.M.; Fischer, E.R.; Braughton, K.R.; Whitney, A.R.; DeLeo, F.R.; Otto, M. Polysaccharide Intercellular Adhesin (PIA) Protects Staphylococcus epidermidis against Major Components of the Human Innate Immune System. Cell Microbiol. 2004, 6, 269–275. [Google Scholar] [CrossRef] [PubMed]
  114. Cheung, G.Y.C.; Rigby, K.; Wang, R.; Queck, S.Y.; Braughton, K.R.; Whitney, A.R.; Teintze, M.; DeLeo, F.R.; Otto, M. Staphylococcus Epidermidis Strategies to Avoid Killing by Human Neutrophils. PLoS Pathog. 2010, 6, e1001133. [Google Scholar] [CrossRef] [PubMed]
  115. Bardan, A.; Nizet, V.; Gallo, R.L. Antimicrobial Peptides and the Skin. Expert Opin. Biol. Ther. 2004, 4, 543–549. [Google Scholar] [CrossRef]
  116. Dorschner, R.A.; Lin, K.H.; Murakami, M.; Gallo, R.L. Neonatal Skin in Mice and Humans Expresses Increased Levels of Antimicrobial Peptides: Innate Immunity during Development of the Adaptive Response. Pediatr. Res. 2003, 53, 566–572. [Google Scholar] [CrossRef] [Green Version]
  117. Walker, V.P.; Akinbi, H.T.; Meinzen-Derr, J.; Narendran, V.; Visscher, M.; Hoath, S.B. Host Defense Proteins on the Surface of Neonatal Skin: Implications for Innate Immunity. J. Pediatr. 2008, 152, 777–781. [Google Scholar] [CrossRef]
  118. Le, K.Y.; Park, M.D.; Otto, M. Immune Evasion Mechanisms of Staphylococcus epidermidis Biofilm Infection. Front. Microbiol. 2018, 9, 359. [Google Scholar] [CrossRef] [PubMed]
  119. Otto, M. Bacterial Evasion of Antimicrobial Peptides by Biofilm Formation. Curr. Top. Microbiol. Immunol. 2006, 306, 251–258. [Google Scholar] [CrossRef]
  120. Kristian, S.A.; Birkenstock, T.A.; Sauder, U.; Mack, D.; Götz, F.; Landmann, R. Biofilm Formation Induces C3a Release and Protects Staphylococcus epidermidis from IgG and Complement Deposition and from Neutrophil-Dependent Killing. J. Infect. Dis. 2008, 197, 1028–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Li, M.; Lai, Y.; Villaruz, A.E.; Cha, D.J.; Sturdevant, D.E.; Otto, M. Gram-Positive Three-Component Antimicrobial Peptide-Sensing System. Proc. Natl Acad Sci USA 2007, 104, 9469–9474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Costa, S.K.; Cho, J.; Cheung, A.L. GraS Sensory Activity in Staphylococcus epidermidis Is Modulated by the “Guard Loop” of VraG and the ATPase Activity of VraF. J. Bacteriol. 2021, 203, e0017821. [Google Scholar] [CrossRef] [PubMed]
  123. Widerström, M.; Wiström, J.; Edebro, H.; Marklund, E.; Backman, M.; Lindqvist, P.; Monsen, T. Colonization of Patients, Healthcare Workers, and the Environment with Healthcare-Associated Staphylococcus epidermidis Genotypes in an Intensive Care Unit: A Prospective Observational Cohort Study. BMC Infect. Dis. 2016, 16, 743. [Google Scholar] [CrossRef] [Green Version]
  124. Lepainteur, M.; Desroches, M.; Bourrel, A.S.; Aberrane, S.; Fihman, V.; L’Hériteau, F.; Razafimahefa, H.; Derouin, V.; Doucet-Populaire, F.; Decousser, J.-W. Role of the Central Venous Catheter in Bloodstream Infections Caused by Coagulase-Negative Staphylococci in Very Preterm Neonates. Pediatric Infect. Dis. J. 2013, 32, 622–628. [Google Scholar] [CrossRef]
  125. Golińska, E.; Strus, M.; Tomusiak-Plebanek, A.; Więcek, G.; Kozień, Ł.; Lauterbach, R.; Pawlik, D.; Rzepecka-Węglarz, B.; Kędzierska, J.; Dorycka, M.; et al. Coagulase-Negative Staphylococci Contained in Gut Microbiota as a Primary Source of Sepsis in Low- and Very Low Birth Weight Neonates. J. Clin. Med. 2020, 9, 2517. [Google Scholar] [CrossRef]
  126. Gensollen, T.; Iyer, S.S.; Kasper, D.L.; Blumberg, R.S. How Colonization by Microbiota in Early Life Shapes the Immune System. Science 2016, 352, 539–544. [Google Scholar] [CrossRef] [Green Version]
  127. Levy, E.I.; Hoang, D.M.; Vandenplas, Y. The Effects of Proton Pump Inhibitors on the Microbiome in Young Children. Acta Paediatr. 2020, 109, 1531–1538. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, X.; Borbet, T.C.; Fallegger, A.; Wipperman, M.F.; Blaser, M.J.; Müller, A. An Antibiotic-Impacted Microbiota Compromises the Development of Colonic Regulatory T Cells and Predisposes to Dysregulated Immune Responses. mBio 2021, 12, e03335-20. [Google Scholar] [CrossRef]
  129. Mirpuri, J. The Emerging Role of Group 3 Innate Lymphoid Cells in the Neonate: Interaction with the Maternal and Neonatal Microbiome. Oxford Open Immunol. 2021, 2, iqab009. [Google Scholar] [CrossRef] [PubMed]
  130. Niu, X.; Daniel, S.; Kumar, D.; Ding, E.Y.; Savani, R.C.; Koh, A.Y.; Mirpuri, J. Transient Neonatal Antibiotic Exposure Increases Susceptibility to Late-Onset Sepsis Driven by Microbiota-Dependent Suppression of Type 3 Innate Lymphoid Cells. Sci. Rep. 2020, 10, 12974. [Google Scholar] [CrossRef]
  131. Korpela, K.; Blakstad, E.W.; Moltu, S.J.; Strømmen, K.; Nakstad, B.; Rønnestad, A.E.; Brække, K.; Iversen, P.O.; Drevon, C.A.; de Vos, W. Intestinal Microbiota Development and Gestational Age in Preterm Neonates. Sci. Rep. 2018, 8, 2453. [Google Scholar] [CrossRef] [Green Version]
  132. Al-Sadi, R.; Dharmaprakash, V.; Nighot, P.; Guo, S.; Nighot, M.; Do, T.; Ma, T.Y. Bifidobacterium Bifidum Enhances the Intestinal Epithelial Tight Junction Barrier and Protects against Intestinal Inflammation by Targeting the Toll-like Receptor-2 Pathway in an NF-ΚB-Independent Manner. Int. J. Mol. Sci. 2021, 22, 8070. [Google Scholar] [CrossRef]
  133. Ling, X.; Linglong, P.; Weixia, D.; Hong, W. Protective Effects of Bifidobacterium on Intestinal Barrier Function in LPS-Induced Enterocyte Barrier Injury of Caco-2 Monolayers and in a Rat NEC Model. PLoS ONE 2016, 11, e0161635. [Google Scholar] [CrossRef] [PubMed]
  134. Moles, L.; Gómez, M.; Moroder, E.; Bustos, G.; Melgar, A.; del Campo, R.; Rodríguez, J.M. Staphylococcus epidermidis in Feedings and Feces of Preterm Neonates. PLoS ONE 2020, 15, e0227823. [Google Scholar] [CrossRef] [Green Version]
  135. Stewart, C.J.; Embleton, N.D.; Marrs, E.C.L.; Smith, D.P.; Fofanova, T.; Nelson, A.; Skeath, T.; Perry, J.D.; Petrosino, J.F.; Berrington, J.E.; et al. Longitudinal Development of the Gut Microbiome and Metabolome in Preterm Neonates with Late Onset Sepsis and Healthy Controls. Microbiome 2017, 5, 75. [Google Scholar] [CrossRef] [Green Version]
  136. Conlan, S.; Mijares, L.A.; NISC Comparative Sequencing Program; Becker, J.; Blakesley, R.W.; Bouffard, G.G.; Brooks, S.; Coleman, H.; Gupta, J.; Gurson, N.; et al. Staphylococcus epidermidis Pan-Genome Sequence Analysis Reveals Diversity of Skin Commensal and Hospital Infection-Associated Isolates. Genome Biol. 2012, 13, R64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Su, F.; Tian, R.; Yang, Y.; Li, H.; Sun, G.; Li, Y.; Han, B.; Xu, X.; Chen, X.; Zhao, G.; et al. Comparative Genome Analysis Reveals the Molecular Basis of Niche Adaptation of Staphylococcus epidermidis Strains. Front. Genet. 2020, 11, 566080. [Google Scholar] [CrossRef] [PubMed]
  138. Méric, G.; Mageiros, L.; Pensar, J.; Laabei, M.; Yahara, K.; Pascoe, B.; Kittiwan, N.; Tadee, P.; Post, V.; Lamble, S.; et al. Disease-Associated Genotypes of the Commensal Skin Bacterium Staphylococcus epidermidis. Nat. Commun 2018, 9, 5034. [Google Scholar] [CrossRef] [Green Version]
  139. Chen, J.; Quiles-Puchalt, N.; Chiang, Y.N.; Bacigalupe, R.; Fillol-Salom, A.; Chee, M.S.J.; Fitzgerald, J.R.; Penadés, J.R. Genome Hypermobility by Lateral Transduction. Science 2018, 362, 207–212. [Google Scholar] [CrossRef] [Green Version]
  140. Du, X.; Larsen, J.; Li, M.; Walter, A.; Slavetinsky, C.; Both, A.; Sanchez Carballo, P.M.; Stegger, M.; Lehmann, E.; Liu, Y.; et al. Staphylococcus epidermidis Clones Express Staphylococcus aureus-Type Wall Teichoic Acid to Shift from a Commensal to Pathogen Lifestyle. Nat. Microbiol. 2021, 6, 757–768. [Google Scholar] [CrossRef] [PubMed]
  141. Méric, G.; Miragaia, M.; de Been, M.; Yahara, K.; Pascoe, B.; Mageiros, L.; Mikhail, J.; Harris, L.G.; Wilkinson, T.S.; Rolo, J.; et al. Ecological Overlap and Horizontal Gene Transfer in Staphylococcus aureus and Staphylococcus epidermidis. Genome Biol. Evol. 2015, 7, 1313–1328. [Google Scholar] [CrossRef] [Green Version]
  142. Cafini, F.; Nguyen, L.T.T.; Higashide, M.; Román, F.; Prieto, J.; Morikawa, K. Horizontal Gene Transmission of the Cfr Gene to MRSA and Enterococcus: Role of Staphylococcus epidermidis as a Reservoir and Alternative Pathway for the Spread of Linezolid Resistance. J. Antimicrob. Chemother. 2016, 71, 587–592. [Google Scholar] [CrossRef] [Green Version]
  143. Otto, M. Coagulase-Negative Staphylococci as Reservoirs of Genes Facilitating MRSA Infection: Staphylococcal Commensal Species Such as Staphylococcus epidermidis Are Being Recognized as Important Sources of Genes Promoting MRSA Colonization and Virulence. Bioessays 2013, 35, 4–11. [Google Scholar] [CrossRef] [Green Version]
  144. Soeorg, H.; Huik, K.; Parm, Ü.; Ilmoja, M.-L.; Metsvaht, T.; Lutsar, I. Molecular Epidemiology of Staphylococcus epidermidis in Neonatal Intensive Care Units. APMIS 2017, 125, 63–73. [Google Scholar] [CrossRef]
  145. Espadinha, D.; Sobral, R.G.; Mendes, C.I.; Méric, G.; Sheppard, S.K.; Carriço, J.A.; de Lencastre, H.; Miragaia, M. Distinct Phenotypic and Genomic Signatures Underlie Contrasting Pathogenic Potential of Staphylococcus epidermidis Clonal Lineages. Front. Microbiol. 2019, 10, 1971. [Google Scholar] [CrossRef] [PubMed]
  146. Sharma, P.; Satorius, A.E.; Raff, M.R.; Rivera, A.; Newton, D.W.; Younger, J.G. Multilocus Sequence Typing for Interpreting Blood Isolates of Staphylococcus Epidermidis. Interdiscip. Perspect. Infect. Dis. 2014, 2014, 787458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Lee, J.Y.H.; Monk, I.R.; Gonçalves da Silva, A.; Seemann, T.; Chua, K.Y.L.; Kearns, A.; Hill, R.; Woodford, N.; Bartels, M.D.; Strommenger, B.; et al. Global Spread of Three Multidrug-Resistant Lineages of Staphylococcus epidermidis. Nat. Microbiol. 2018, 3, 1175–1185. [Google Scholar] [CrossRef] [PubMed]
  148. Shelburne, S.A.; Dib, R.W.; Endres, B.T.; Reitzel, R.; Li, X.; Kalia, A.; Sahasrabhojane, P.; Chaftari, A.-M.; Hachem, R.; Vargas-Cruz, N.S.; et al. Whole-Genome Sequencing of Staphylococcus Epidermidis Bloodstream Isolates from a Prospective Clinical Trial Reveals That Complicated Bacteraemia Is Caused by a Limited Number of Closely Related Sequence Types. Clin. Microbiol. Infect. 2020, 26, 646.e1–646.e8. [Google Scholar] [CrossRef]
  149. Salgueiro, V.C.; Iorio, N.L.P.; Ferreira, M.C.; Chamon, R.C.; dos Santos, K.R.N. Methicillin Resistance and Virulence Genes in Invasive and Nasal Staphylococcus epidermidis Isolates from Neonates. BMC Microbiol. 2017, 17, 15. [Google Scholar] [CrossRef] [Green Version]
  150. Both, A.; Huang, J.; Qi, M.; Lausmann, C.; Weißelberg, S.; Büttner, H.; Lezius, S.; Failla, A.V.; Christner, M.; Stegger, M.; et al. Distinct Clonal Lineages and Within-Host Diversification Shape Invasive Staphylococcus epidermidis Populations. PLoS Pathog. 2021, 17, e1009304. [Google Scholar] [CrossRef]
  151. Yao, Y.; Vuong, C.; Kocianova, S.; Villaruz, A.E.; Lai, Y.; Sturdevant, D.E.; Otto, M. Characterization of the Staphylococcus epidermidis Accessory-Gene Regulator Response: Quorum-Sensing Regulation of Resistance to Human Innate Host Defense. J. Infect. Dis. 2006, 193, 841–848. [Google Scholar] [CrossRef] [Green Version]
  152. Kozitskaya, S.; Olson, M.E.; Fey, P.D.; Witte, W.; Ohlsen, K.; Ziebuhr, W. Clonal Analysis of Staphylococcus epidermidis Isolates Carrying or Lacking Biofilm-Mediating Genes by Multilocus Sequence Typing. J. Clin. Microbiol. 2005, 43, 4751–4757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gu, J.; Li, H.; Li, M.; Vuong, C.; Otto, M.; Wen, Y.; Gao, Q. Bacterial Insertion Sequence IS256 as a Potential Molecular Marker to Discriminate Invasive Strains from Commensal Strains of Staphylococcus epidermidis. J. Hosp. Infect. 2005, 61, 342–348. [Google Scholar] [CrossRef] [PubMed]
  154. Galdbart, J.O.; Allignet, J.; Tung, H.S.; Rydèn, C.; El Solh, N. Screening for Staphylococcus epidermidis Markers Discriminating between Skin-Flora Strains and Those Responsible for Infections of Joint Prostheses. J. Infect. Dis. 2000, 182, 351–355. [Google Scholar] [CrossRef] [Green Version]
  155. Rogers, K.L.; Rupp, M.E.; Fey, P.D. The Presence of IcaADBC Is Detrimental to the Colonization of Human Skin by Staphylococcus epidermidis. Appl. Environ. Microbiol. 2008, 74, 6155–6157. [Google Scholar] [CrossRef] [Green Version]
  156. Härtel, C.; Osthues, I.; Rupp, J.; Haase, B.; Röder, K.; Göpel, W.; Herting, E.; Schultz, C. Characterisation of the Host Inflammatory Response to Staphylococcus Epidermidis in Neonatal Whole Blood. Arch. Dis Child. Fetal Neonatal Ed. 2008, 93, F140–145. [Google Scholar] [CrossRef] [PubMed]
  157. Le, K.Y.; Otto, M. Quorum-Sensing Regulation in Staphylococci-an Overview. Front Microbiol 2015, 6, 1174. [Google Scholar] [CrossRef] [Green Version]
  158. Vuong, C.; Gerke, C.; Somerville, G.A.; Fischer, E.R.; Otto, M. Quorum-Sensing Control of Biofilm Factors in Staphylococcus epidermidis. J. Infect. Dis. 2003, 188, 706–718. [Google Scholar] [CrossRef] [Green Version]
  159. Vuong, C.; Kocianova, S.; Yao, Y.; Carmody, A.B.; Otto, M. Increased Colonization of Indwelling Medical Devices by Quorum-Sensing Mutants of Staphylococcus epidermidis in Vivo. J. Infect. Dis. 2004, 190, 1498–1505. [Google Scholar] [CrossRef] [Green Version]
  160. Nguyen, T.H.; Park, M.D.; Otto, M. Host Response to Staphylococcus epidermidis Colonization and Infections. Front. Cell Infect. Microbiol. 2017, 7, 90. [Google Scholar] [CrossRef] [Green Version]
  161. Al-Haqan, A.; Boswihi, S.S.; Pathan, S.; Udo, E.E. Antimicrobial Resistance and Virulence Determinants in Coagulase-Negative Staphylococci Isolated Mainly from Preterm Neonates. PLoS ONE 2020, 15, e0236713. [Google Scholar] [CrossRef] [PubMed]
  162. Skiba-Kurek, I.; Nowak, P.; Empel, J.; Tomczak, M.; Klepacka, J.; Sowa-Sierant, I.; Żak, I.; Pomierny, B.; Karczewska, E. Evaluation of Biofilm Formation and Prevalence of Multidrug-Resistant Strains of Staphylococcus epidermidis Isolated from Neonates with Sepsis in Southern Poland. Pathogens 2021, 10, 877. [Google Scholar] [CrossRef] [PubMed]
  163. Eftekhar, F.; Speert, D.P. Biofilm Formation by Persistent and Non-Persistent Isolates of Staphylococcus epidermidis from a Neonatal Intensive Care Unit. J. Hosp. Infect. 2009, 71, 112–116. [Google Scholar] [CrossRef] [PubMed]
  164. Rohde, H.; Kalitzky, M.; Kröger, N.; Scherpe, S.; Horstkotte, M.A.; Knobloch, J.K.-M.; Zander, A.R.; Mack, D. Detection of Virulence-Associated Genes Not Useful for Discriminating between Invasive and Commensal Staphylococcus epidermidis Strains from a Bone Marrow Transplant Unit. J. Clin. Microbiol. 2004, 42, 5614–5619. [Google Scholar] [CrossRef] [Green Version]
  165. Arrecubieta, C.; Toba, F.A.; von Bayern, M.; Akashi, H.; Deng, M.C.; Naka, Y.; Lowy, F.D. SdrF, a Staphylococcus epidermidis Surface Protein, Contributes to the Initiation of Ventricular Assist Device Driveline-Related Infections. PLoS Pathog. 2009, 5, e1000411. [Google Scholar] [CrossRef] [Green Version]
  166. Guo, B.; Zhao, X.; Shi, Y.; Zhu, D.; Zhang, Y. Pathogenic Implication of a Fibrinogen-Binding Protein of Staphylococcus epidermidis in a Rat Model of Intravascular-Catheter-Associated Infection. Infect. Immun. 2007, 75, 2991–2995. [Google Scholar] [CrossRef] [Green Version]
  167. Söderquist, B.; Andersson, M.; Nilsson, M.; Nilsdotter-Augustinsson, Å.; Persson, L.; Friberg, Ö.; Jacobsson, S. Staphylococcus epidermidis Surface Protein I (SesI): A Marker of the Invasive Capacity of S. Epidermidis? J. Med Microbiol. 2009, 58, 1395–1397. [Google Scholar] [CrossRef]
  168. Qi, X.; Jin, Y.; Duan, J.; Hao, Z.; Wang, S.; Guo, Y.; Lv, J.; Hu, L.; Wang, L.; Yu, F. SesI May Be Associated with the Invasiveness of Staphylococcus epidermidis. Front. Microbiol. 2017, 8, 2574. [Google Scholar] [CrossRef] [PubMed]
  169. Jamet, A.; Guglielmini, J.; Brancotte, B.; Coureuil, M.; Euphrasie, D.; Meyer, J.; Roux, J.; Barnier, J.-P.; Bille, E.; Ferroni, A.; et al. High-Resolution Typing of Staphylococcus epidermidis Based on Core Genome Multilocus Sequence Typing to Investigate the Hospital Spread of Multidrug-Resistant Clones. J. Clin. Microbiol. 2021, 59, e02454-20. [Google Scholar] [CrossRef]
  170. Bowden, M.G.; Chen, W.; Singvall, J.; Xu, Y.; Peacock, S.J.; Valtulina, V.; Speziale, P.; Höök, M. Identification and Preliminary Characterization of Cell-Wall-Anchored Proteins of Staphylococcus epidermidis. Microbiology 2005, 151, 1453–1464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Hartford, O.; O’Brien, L.; Schofield, K.; Wells, J.; Foster, T.J. The Fbe (SdrG) Protein of Staphylococcus epidermidis HB Promotes Bacterial Adherence to Fibrinogen. Microbiology 2001, 147, 2545–2552. [Google Scholar] [CrossRef] [PubMed]
  172. Schommer, N.N.; Christner, M.; Hentschke, M.; Ruckdeschel, K.; Aepfelbacher, M.; Rohde, H. Staphylococcus epidermidis Uses Distinct Mechanisms of Biofilm Formation to Interfere with Phagocytosis and Activation of Mouse Macrophage-like Cells 774A.1. Infect. Immun. 2011, 79, 2267–2276. [Google Scholar] [CrossRef] [Green Version]
  173. Foster, T.J. Surface Proteins of Staphylococcus epidermidis. Front. Microbiol. 2020, 11, 1829. [Google Scholar] [CrossRef] [PubMed]
  174. Qin, L.; Da, F.; Fisher, E.L.; Tan, D.C.S.; Nguyen, T.H.; Fu, C.-L.; Tan, V.Y.; McCausland, J.W.; Sturdevant, D.E.; Joo, H.-S.; et al. Toxin Mediates Sepsis Caused by Methicillin-Resistant Staphylococcus epidermidis. PLoS Pathog. 2017, 13, e1006153. [Google Scholar] [CrossRef]
  175. Scheifele, D.W.; Bjornson, G.L.; Dyer, R.A.; Dimmick, J.E. Delta-like Toxin Produced by Coagulase-Negative Staphylococci Is Associated with Neonatal Necrotizing Enterocolitis. Infect. Immun. 1987, 55, 2268–2273. [Google Scholar] [CrossRef] [Green Version]
  176. Svensson, K.; Hellmark, B.; Söderquist, B. Characterization of SCCmec Elements in Methicillin-Resistant Staphylococcus epidermidis Isolated from Blood Cultures from Neonates during Three Decades. APMIS 2011, 119, 885–893. [Google Scholar] [CrossRef]
  177. Ivarsson, M.; Schollin, J.; Björkqvist, M. Staphylococcus epidermidis and Staphylococcus aureus Trigger Different Interleukin-8 and Intercellular Adhesion Molecule-1 in Lung Cells: Implications for Inflammatory Complications Following Neonatal Sepsis. Acta Paediatr. 2013, 102, 1010–1016. [Google Scholar] [CrossRef]
  178. Månsson, E.; Bech Johannesen, T.; Nilsdotter-Augustinsson, Å.; Söderquist, B.; Stegger, M. Comparative Genomics of Staphylococcus epidermidis from Prosthetic-Joint Infections and Nares Highlights Genetic Traits Associated with Antimicrobial Resistance, Not Virulence. Microb. Genom. 2021, 7. [Google Scholar] [CrossRef]
  179. Rose, H.R.; Holzman, R.S.; Altman, D.R.; Smyth, D.S.; Wasserman, G.A.; Kafer, J.M.; Wible, M.; Mendes, R.E.; Torres, V.J.; Shopsin, B. Cytotoxic Virulence Predicts Mortality in Nosocomial Pneumonia Due to Methicillin-Resistant Staphylococcus aureus. J. Infect. Dis. 2015, 211, 1862–1874. [Google Scholar] [CrossRef] [Green Version]
  180. Gordon, R.J.; Miragaia, M.; Weinberg, A.D.; Lee, C.J.; Rolo, J.; Giacalone, J.C.; Slaughter, M.S.; Pappas, P.; Naka, Y.; Tector, A.J.; et al. Staphylococcus epidermidis Colonization Is Highly Clonal across US Cardiac Centers. J. Infect. Dis. 2012, 205, 1391–1398. [Google Scholar] [CrossRef] [Green Version]
  181. Widerström, M.; Monsen, T.; Karlsson, C.; Edebro, H.; Johansson, A.; Wiström, J. Clonality among Multidrug-Resistant Hospital-Associated Staphylococcus epidermidis in Northern Europe. Scand. J. Infect. Dis. 2009, 41, 642–649. [Google Scholar] [CrossRef]
  182. Morgenstern, M.; Post, V.; Erichsen, C.; Hungerer, S.; Bühren, V.; Militz, M.; Richards, R.G.; Moriarty, T.F. Biofilm Formation Increases Treatment Failure in Staphylococcus epidermidis Device-Related Osteomyelitis of the Lower Extremity in Human Patients. J. Orthop. Res. 2016, 34, 1905–1913. [Google Scholar] [CrossRef]
  183. Granslo, H.N.; Klingenberg, C.; Fredheim, E.G.A.; Rønnestad, A.; Mollnes, T.E.; Flægstad, T. Arginine Catabolic Mobile Element Is Associated with Low Antibiotic Resistance and Low Pathogenicity in Staphylococcus epidermidis From Neonates. Pediatr. Res. 2010, 68, 237–241. [Google Scholar] [CrossRef] [Green Version]
  184. VanAken, S.M.; Newton, D.; VanEpps, J.S. Improved Diagnostic Prediction of the Pathogenicity of Bloodstream Isolates of Staphylococcus epidermidis. PLoS ONE 2021, 16, e0241457. [Google Scholar] [CrossRef]
  185. Morioka, S.; Ichikawa, M.; Mori, K.; Kurai, H. Coagulase-Negative Staphylococcal Bacteraemia in Cancer Patients. Time to Positive Culture Can Distinguish Bacteraemia from Contamination. Infect. Dis. 2018, 50, 660–665. [Google Scholar] [CrossRef] [PubMed]
  186. Cheung, G.Y.C.; Kretschmer, D.; Duong, A.C.; Yeh, A.J.; Ho, T.V.; Chen, Y.; Joo, H.-S.; Kreiswirth, B.N.; Peschel, A.; Otto, M. Production of an Attenuated Phenol-Soluble Modulin Variant Unique to the MRSA Clonal Complex 30 Increases Severity of Bloodstream Infection. PLoS Pathog. 2014, 10, e1004298. [Google Scholar] [CrossRef] [PubMed]
  187. Laabei, M.; Uhlemann, A.-C.; Lowy, F.D.; Austin, E.D.; Yokoyama, M.; Ouadi, K.; Feil, E.; Thorpe, H.A.; Williams, B.; Perkins, M.; et al. Evolutionary Trade-Offs Underlie the Multi-Faceted Virulence of Staphylococcus aureus. PLoS Biol. 2015, 13, e1002229. [Google Scholar] [CrossRef] [Green Version]
  188. Pedroza-Dávila, U.; Uribe-Alvarez, C.; Morales-García, L.; Espinoza-Simón, E.; Méndez-Romero, O.; Muhlia-Almazán, A.; Chiquete-Félix, N.; Uribe-Carvajal, S. Metabolism, ATP Production and Biofilm Generation by Staphylococcus epidermidis in Either Respiratory or Fermentative Conditions. AMB Express 2020, 10, 31. [Google Scholar] [CrossRef] [Green Version]
  189. Stevens, E.J.; Bates, K.A.; King, K.C. Host Microbiota Can Facilitate Pathogen Infection. PLoS Pathog. 2021, 17, e1009514. [Google Scholar] [CrossRef]
  190. Uribe-Alvarez, C.; Chiquete-Félix, N.; Contreras-Zentella, M.; Guerrero-Castillo, S.; Peña, A.; Uribe-Carvajal, S. Staphylococcus epidermidis: Metabolic Adaptation and Biofilm Formation in Response to Different Oxygen Concentrations. Pathog. Dis. 2016, 74, ftv111. [Google Scholar] [CrossRef] [Green Version]
  191. França, A.; Pier, G.B.; Vilanova, M.; Cerca, N. Transcriptomic Analysis of Staphylococcus epidermidis Biofilm-Released Cells upon Interaction with Human Blood Circulating Immune Cells and Soluble Factors. Front. Microbiol. 2016, 7, 1143. [Google Scholar] [CrossRef]
  192. França, A.; Cerca, N. Plasma Is the Main Regulator of Staphylococcus epidermidis Biofilms Virulence Genes Transcription in Human Blood. Pathog. Dis. 2016, 74, ftv125. [Google Scholar] [CrossRef] [Green Version]
  193. Matinaho, S.; von Bonsdorff, L.; Rouhiainen, A.; Lönnroth, M.; Parkkinen, J. Dependence of Staphylococcus epidermidis on Non-Transferrin-Bound Iron for Growth. FEMS Microbiol. Lett. 2001, 196, 177–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Oliveira, F.; França, Â.; Cerca, N. Staphylococcus epidermidis Is Largely Dependent on Iron Availability to Form Biofilms. Int. J. Med. Microbiol. 2017, 307, 552–563. [Google Scholar] [CrossRef] [Green Version]
  195. Weinberg, E.D. Nutritional Immunity. Host’s Attempt to Withold Iron from Microbial Invaders. JAMA 1975, 231, 39–41. [Google Scholar] [CrossRef] [PubMed]
  196. França, A.; Carvalhais, V.; Maira-Litrán, T.; Vilanova, M.; Cerca, N.; Pier, G. Alterations in the Staphylococcus epidermidis Biofilm Transcriptome Following Interaction with Whole Human Blood. Pathog. Dis. 2014, 70, 444–448. [Google Scholar] [CrossRef] [Green Version]
  197. Liu, Q.; Chen, N.; Chen, H.; Huang, Y. RNA-Seq Analysis of Differentially Expressed Genes of Staphylococcus epidermidis Isolated from Postoperative Endophthalmitis and the Healthy Conjunctiva. Sci. Rep. 2020, 10, 14234. [Google Scholar] [CrossRef]
  198. Strunk, T.; Hibbert, J.; Doherty, D.; Nathan, E.; Simmer, K.; Richmond, P.; Currie, A.; Burgner, D. Impaired Cytokine Responses to Live Staphylococcus epidermidis in Preterm Infants Precede Gram-Positive, Late-Onset Sepsis. Clin. Infect. Dis. 2021, 72, 271–278. [Google Scholar] [CrossRef]
  199. Mohamed, M.A.; Cunningham-Rundles, S.; Dean, C.R.; Hammad, T.A.; Nesin, M. Levels of Pro-Inflammatory Cytokines Produced from Cord Blood in-Vitro Are Pathogen Dependent and Increased in Comparison to Adult Controls. Cytokine 2007, 39, 171–177. [Google Scholar] [CrossRef]
  200. Gravina, G.; Svedin, P.; Ardalan, M.; Levy, O.; Ek, C.J.; Mallard, C.; Lai, J.C.Y. Staphylococcus epidermidis Sensitizes Perinatal Hypoxic-Ischemic Brain Injury in Male but Not Female Mice. Front. Immunol. 2020, 11, 516. [Google Scholar] [CrossRef] [Green Version]
  201. Mack, D.; Siemssen, N.; Laufs, R. Parallel Induction by Glucose of Adherence and a Polysaccharide Antigen Specific for Plastic-Adherent Staphylococcus epidermidis: Evidence for Functional Relation to Intercellular Adhesion. Infect. Immun. 1992, 60, 2048–2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Hibbert, J.E.; Currie, A.; Strunk, T. Sepsis-Induced Immunosuppression in Neonates. Front. Pediatr. 2018, 6, 357. [Google Scholar] [CrossRef]
  203. Prosser, A.; Hibbert, J.; Strunk, T.; Kok, C.H.; Simmer, K.; Richmond, P.; Burgner, D.; Currie, A. Phagocytosis of Neonatal Pathogens by Peripheral Blood Neutrophils and Monocytes from Newborn Preterm and Term Infants. Pediatr. Res. 2013, 74, 503–510. [Google Scholar] [CrossRef] [PubMed]
  204. Kronforst, K.D.; Mancuso, C.J.; Pettengill, M.; Ninkovic, J.; Coombs, M.R.P.; Stevens, C.; Otto, M.; Mallard, C.; Wang, X.; Goldmann, D.; et al. A Neonatal Model of Intravenous Staphylococcus epidermidis Infection in Mice <24 h Old Enables Characterization of Early Innate Immune Responses. PLoS ONE 2012, 7, e43897. [Google Scholar] [CrossRef]
  205. Bi, D.; Qiao, L.; Bergelson, I.; Ek, C.J.; Duan, L.; Zhang, X.; Albertsson, A.-M.; Pettengill, M.; Kronforst, K.; Ninkovic, J.; et al. Staphylococcus epidermidis Bacteremia Induces Brain Injury in Neonatal Mice via Toll-like Receptor 2-Dependent and -Independent Pathways. J. Infect. Dis. 2015, 212, 1480–1490. [Google Scholar] [CrossRef]
  206. Lai, J.C.Y.; Svedin, P.; Ek, C.J.; Mottahedin, A.; Wang, X.; Levy, O.; Currie, A.; Strunk, T.; Mallard, C. Vancomycin Is Protective in a Neonatal Mouse Model of Staphylococcus epidermidis-Potentiated Hypoxic-Ischemic Brain Injury. Antimicrob. Agents Chemother. 2020, 64, e02003-19. [Google Scholar] [CrossRef] [PubMed]
  207. Strunk, T.; Inder, T.; Wang, X.; Burgner, D.; Mallard, C.; Levy, O. Infection-Induced Inflammation and Cerebral Injury in Preterm Infants. Lancet Infect. Dis. 2014, 14, 751–762. [Google Scholar] [CrossRef] [Green Version]
  208. Ng, S.; Strunk, T.; Lee, A.H.; Gill, E.E.; Falsafi, R.; Woodman, T.; Hibbert, J.; Hancock, R.E.W.; Currie, A. Whole Blood Transcriptional Responses of Very Preterm Infants during Late-Onset Sepsis. PLoS ONE 2020, 15, e0233841. [Google Scholar] [CrossRef]
  209. Cernada, M.; Pinilla-González, A.; Kuligowski, J.; Morales, J.M.; Lorente-Pozo, S.; Piñeiro-Ramos, J.D.; Parra-Llorca, A.; Lara-Cantón, I.; Vento, M.; Serna, E. Transcriptome Profiles Discriminate between Gram-Positive and Gram-Negative Sepsis in Preterm Neonates. Pediatr. Res. 2021. [Google Scholar] [CrossRef]
  210. de Jong, E.; Hancock, D.G.; Hibbert, J.; Wells, C.; Richmond, P.; Simmer, K.; Burgner, D.; Strunk, T.; Currie, A.J. Identification of Generic and Pathogen-Specific Cord Blood Monocyte Transcriptomes Reveals a Largely Conserved Response in Preterm and Term Newborn Infants. J. Mol. Med. 2018, 96, 147–157. [Google Scholar] [CrossRef]
  211. de Jong, E.; Strunk, T.; Burgner, D.; Lavoie, P.M.; Currie, A. The Phenotype and Function of Preterm Infant Monocytes: Implications for Susceptibility to Infection. J. Leukoc. Biol. 2017, 102, 645–656. [Google Scholar] [CrossRef] [Green Version]
  212. Strunk, T.; Currie, A.; Richmond, P.; Simmer, K.; Burgner, D. Innate Immunity in Human Newborn Infants: Prematurity Means More than Immaturity. J. Matern. Fetal Neonatal Med. 2011, 24, 25–31. [Google Scholar] [CrossRef]
  213. Westermann, A.J.; Förstner, K.U.; Amman, F.; Barquist, L.; Chao, Y.; Schulte, L.N.; Müller, L.; Reinhardt, R.; Stadler, P.F.; Vogel, J. Dual RNA-Seq Unveils Noncoding RNA Functions in Host-Pathogen Interactions. Nature 2016, 529, 496–501. [Google Scholar] [CrossRef] [PubMed]
  214. Minhas, V.; Aprianto, R.; McAllister, L.J.; Wang, H.; David, S.C.; McLean, K.T.; Comerford, I.; McColl, S.R.; Paton, J.C.; Veening, J.-W.; et al. In Vivo Dual RNA-Seq Analysis Reveals the Basis for Differential Tissue Tropism of Clinical Isolates of Streptococcus pneumoniae. bioRxiv 2019, 862755. [Google Scholar] [CrossRef]
  215. Minhas, V.; Aprianto, R.; McAllister, L.J.; Wang, H.; David, S.C.; McLean, K.T.; Comerford, I.; McColl, S.R.; Paton, J.C.; Veening, J.-W.; et al. In Vivo Dual RNA-Seq Reveals That Neutrophil Recruitment Underlies Differential Tissue Tropism of Streptococcus pneumoniae. Commun. Biol. 2020, 3, 293. [Google Scholar] [CrossRef] [PubMed]
  216. Shenoy, A.T.; Brissac, T.; Gilley, R.P.; Kumar, N.; Wang, Y.; Gonzalez-Juarbe, N.; Hinkle, W.S.; Daugherty, S.C.; Shetty, A.C.; Ott, S.; et al. Streptococcus pneumoniae in the Heart Subvert the Host Response through Biofilm-Mediated Resident Macrophage Killing. PLoS Pathog. 2017, 13, e1006582. [Google Scholar] [CrossRef] [Green Version]
  217. D’Mello, A.; Riegler, A.N.; Martínez, E.; Beno, S.M.; Ricketts, T.D.; Foxman, E.F.; Orihuela, C.J.; Tettelin, H. An in Vivo Atlas of Host–Pathogen Transcriptomes during Streptococcus pneumoniae Colonization and Disease. Proc. Natl. Acad. Sci. USA 2020, 117, 33507–33518. [Google Scholar] [CrossRef]
  218. Thänert, R.; Goldmann, O.; Beineke, A.; Medina, E. Host-Inherent Variability Influences the Transcriptional Response of Staphylococcus aureus during in Vivo Infection. Nat. Commun. 2017, 8, 14268. [Google Scholar] [CrossRef]
Figure 1. Summary of potential mechanisms underlying the establishment of neonatal tolerance to commensal S. epidermidis and its contribution to neonatal host defense. Antimicrobial peptides (AMPs), Phenol-soluble modulins (PSMs), Peforin-2 (P2), Toll-like receptor (TLR), Regulatory T cells (Tregs). Created with BioRender.com.
Figure 1. Summary of potential mechanisms underlying the establishment of neonatal tolerance to commensal S. epidermidis and its contribution to neonatal host defense. Antimicrobial peptides (AMPs), Phenol-soluble modulins (PSMs), Peforin-2 (P2), Toll-like receptor (TLR), Regulatory T cells (Tregs). Created with BioRender.com.
Ijms 23 00860 g001
Figure 2. Risk factors associated with skin-derived and gut-derived S. epidermidis late-onset sepsis (LOS) in the preterm neonate. Created with BioRender.com.
Figure 2. Risk factors associated with skin-derived and gut-derived S. epidermidis late-onset sepsis (LOS) in the preterm neonate. Created with BioRender.com.
Ijms 23 00860 g002
Figure 3. S. epidermidis functional and genetic determinants with potential role in late-onset sepsis (LOS pathogenicity). Created with BioRender.com.
Figure 3. S. epidermidis functional and genetic determinants with potential role in late-onset sepsis (LOS pathogenicity). Created with BioRender.com.
Ijms 23 00860 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Joubert, I.A.; Otto, M.; Strunk, T.; Currie, A.J. Look Who’s Talking: Host and Pathogen Drivers of Staphylococcus epidermidis Virulence in Neonatal Sepsis. Int. J. Mol. Sci. 2022, 23, 860. https://doi.org/10.3390/ijms23020860

AMA Style

Joubert IA, Otto M, Strunk T, Currie AJ. Look Who’s Talking: Host and Pathogen Drivers of Staphylococcus epidermidis Virulence in Neonatal Sepsis. International Journal of Molecular Sciences. 2022; 23(2):860. https://doi.org/10.3390/ijms23020860

Chicago/Turabian Style

Joubert, Isabella A., Michael Otto, Tobias Strunk, and Andrew J. Currie. 2022. "Look Who’s Talking: Host and Pathogen Drivers of Staphylococcus epidermidis Virulence in Neonatal Sepsis" International Journal of Molecular Sciences 23, no. 2: 860. https://doi.org/10.3390/ijms23020860

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