Next Article in Journal
Microbial Contamination and Antibiotic Resistance in Marketed Food in Bangladesh: Current Situation and Possible Improvements
Previous Article in Journal
Antimicrobial Peptides Designed against the Ω-Loop of Class A β-Lactamases to Potentiate the Efficacy of β-Lactam Antibiotics
Previous Article in Special Issue
Nanomaterials and Coatings for Managing Antibiotic-Resistant Biofilms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies

1
Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
2
LABBELS—Associate Laboratory in Biotechnology and Bioengineering and Microelectromechanical Systems, Braga and Guimarães, Portugal
Antibiotics 2023, 12(3), 554; https://doi.org/10.3390/antibiotics12030554
Submission received: 31 December 2022 / Revised: 24 February 2023 / Accepted: 6 March 2023 / Published: 10 March 2023
(This article belongs to the Special Issue Feature Reviews on Antibiofilm Strategies)

Abstract

:
Infections are one of the most significant complications of neonates, especially those born preterm, with sepsis as one of the principal causes of mortality. Coagulase-negative staphylococci (CoNS), a group of staphylococcal species that naturally inhabit healthy human skin and mucosa, are the most common cause of late-onset sepsis, especially in preterms. One of the risk factors for the development of CoNS infections is the presence of implanted biomedical devices, which are frequently used for medications and/or nutrient delivery, as they serve as a scaffold for biofilm formation. The major concerns related to CoNS infections have to do with the increasing resistance to multiple antibiotics observed among this bacterial group and biofilm cells’ increased tolerance to antibiotics. As such, the treatment of CoNS biofilm-associated infections with antibiotics is increasingly challenging and considering that antibiotics remain the primary form of treatment, this issue will likely persist in upcoming years. For that reason, the development of innovative and efficient therapeutic measures is of utmost importance. This narrative review assesses the current challenges and emerging diagnostic tools and therapies for the treatment of CoNS biofilm-associated infections, with a special focus on late-onset sepsis.

1. Introduction

According to the World Health Organization, 15 million babies are born preterm (before completing 37 weeks of gestation) every year [1], with preterm birth complications responsible for 1 million deaths annually [2]. One of the most significant complications in preterm infants is the development of infections, with neonatal sepsis (NS) as one of the principal causes of mortality and long-term neurological impairments [2,3,4]. The Global Burden of Disease estimated, in 2017, that there were 1.3 million annual incident cases of NS worldwide [5], resulting in 203,000 attributable deaths [6].
The definition of sepsis in adults was recently redefined, by the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) taskforce, as a life-threatening and dysregulated host response to an infection [7]. However, when it comes to sepsis in newborns, there is no international consensus, with criteria used to define NS varying among studies [8]. As such, an international consensus is necessary to standardize the criteria for NS definition, which is essential for accurate diagnoses and positive outcomes [8]. In general, NS is defined as a clinical syndrome detected during the first 28 days of life, which involves systemic signs of infection, circulatory shock and multisystem organ failure [9]. NS can be further classified as either early- or late-onset sepsis, depending on the presentation age and presumed infection mode [10] (Table 1). If signals of infection occur before 3 days of life, it is considered early-onset sepsis (EOS) and if it appears following that, and up to 28 days, it is considered late-onset sepsis (LOS) [11,12,13,14]. However, no time point clearly distinguishes EOS from LOS, with some clinicians defending different segregation time points [15].
While EOS is acquired vertically, with neonates becoming infected either in utero or during birth associated with passage through the vaginal canal, LOS is usually related to the postnatal nosocomial or community environments [21]. Although a reduction of EOS has been observed over the years, probably due to the use of prophylactic intrapartum antibiotics [22], LOS has shown stable or increased incidence, depending on the neonatal units evaluated [21,23]. As such, LOS is still one of the main causes of mortality among preterm infants and of several important morbidities with short- and long-term detrimental effects on neonatal outcomes [24]. In general, LOS affects 1 in 10 very preterm infants (28 to 32 weeks) with incidences reaching as high as 60% in extremely preterm infants (less than 28 weeks) [25]. Furthermore, due to extended hospital stays and additional diagnosis and treatments required, LOS constitutes an important economic burden, which is even higher when considering the costs related to the clinical support that patients may require due to the lifelong impact of the disease.
Due to its transmission venue, microorganisms causing EOS and LOS often differ. While EOS is mainly caused by bacteria present in the maternal vaginal canal, LOS is more likely caused by microorganisms present in neonatal rooms or community environments [13]. As such, considering that each hospital and community have a particular epidemiology profile, microorganisms causing LOS diverge depending on the hospital, country and also over time [21]. In high-income countries, the most frequent causative agents of LOS, especially in extremely and very low birth weight infants, are coagulase-negative staphylococci (CoNS) [21,26,27,28,29,30], accounting for 53 to 78% of the LOS cases [21]. In low- and middle-income countries, although CoNS are still responsible for about 35 to 47% of LOS, Staphylococcus aureus, Klebsiella pneumoniae and Escherichia coli show higher incidence [31,32,33]. However, the lack of studies in low- and middle-income countries makes accurate estimations difficult.
CoNS are a group of staphylococcal species that naturally inhabit healthy human skin and mucosa [34,35,36]. While the CoNS group is composed of more than 50 species [37], Staphylococcus epidermidis, Staphylococcus haemolyticus and Staphylococcus capitis are the most common etiological agents of LOS [38,39,40,41,42,43,44,45]. The straight relationship between CoNS and the emergence of LOS has to do, in part, with these bacteria’s ability to attach to the surface of medical devices and form biofilms, which is considered the major virulence factor of this group of bacteria [46,47,48,49]. One of the major concerns related to CoNS infections is the increasing resistance to antibiotics observed among this bacterial group, which is further aggravated not only by the phenomenon of multidrug resistance [50,51] but also because CoNS biofilm cells are inherently more tolerant to antibiotics than free-living cells [52,53]. Consequently, the treatment of CoNS infections is becoming increasingly challenging.
Given the significant social and economic implications of LOS, the development of both prophylactic and therapeutic measures has continually been explored; however, so far, antibiotics remain the only form of treatment. Considering that the number of preterm infants is increasing worldwide [1] and with the rapid spread of antimicrobial resistance [54], the management of CoNS infections is likely to deteriorate. Therefore, this narrative review evaluates the current challenges in diagnosis and emerging therapies for the treatment of LOS, in particular the ones caused by CoNS biofilms.

2. CoNS Role as Commensal in Newborns

CoNS are normal inhabitants of healthy human skin and mucosa of the respiratory tract and intestine, being one of the most abundant colonizers of all skin sites [34,55]. CoNS are involved in the maintenance of skin integrity and homeostasis, preventing colonization by opportunistic pathogens [56,57,58]. However, the immune system of newborns presents significant differences from that of adults [59,60], such as functional and quantitative deficiencies in antigen-presenting cells and phagocytes, lack of antigenic exposure, less production of pro-inflammatory cytokines and tissue-protective immunosuppressive mechanisms [61,62,63], and therefore the protective role of CoNS colonization is unclear. It could be expected that after contact with microorganisms, the neonates’ host immune system would mount an inflammatory response. However, although it is important to fight the infection, it is also essential to enable the establishment of a commensal bacterial community [64]. Hence, newborns present a particular immune response that relates to the unique environmental pressures and challenges of the first days of life [65,66]. This type of response is called disease tolerance, in which there is a reduction of the potential harm to the host without reducing the pathogen burden, a critical balance that enables fighting infections in early life but, at the same time, permits building a commensal community [64,67]. This is supported by the fact that regulatory T cells, which suppress other cells in the immune system to regulate the inflammatory response [68], induce tolerance to S. epidermidis colonization during the neonatal period, but not in adults [69]. Furthermore, the fact that neonates can survive significantly higher bacterial loads than adults during active infection [64] is another indicator of the disease tolerance process. Hence, the establishment of a healthy skin microbiome during this early period is essential to avoid colonization by virulent microbes and, thus, contributes to cutaneous homeostasis and the shaping of inflammatory responses [70].
It was shown that upon birth newborns are readily colonized [71], with staphylococcal species having a major representation [70]. However, the skin microbiome of newborns is very dynamic and the early colonization with staphylococci declines over time [70]. Indeed, the skin microbiome of neonates is not fully established, evolving during the first year of life or even beyond [70]. The colonization of neonates in the first hours/days is most likely the result of contact with medical staff, equipment and their parents [72]. The composition of the newborn microbiome is determined by the previous administration of antibiotics, by modes of feeding and delivery [71,73,74,75] and, very likely, depends on geographical and ethnic factors [63]. In addition, it has been suggested that the microbiome is also modulated by the microorganisms that colonize the neonatal intensive care unit (NICU) rooms if the infants are hospitalized for a long time [76,77]. In this context, it was noticed that these preterm infants harbor a higher prevalence of nosocomial isolates and multi-drug resistant strains compared to term infants [74,75].
Dominguez-Bello and collaborators showed that the skin microbiome of infants delivered by Caesarean section relates to that of the mother’s skin and the ones born through vaginal delivery relate to the mother’s vagina [71]. Furthermore, S. epidermidis was found to be the predominant bacterium in colostrum and milk from healthy women [78,79,80,81], enabling the passage of the bacterium through breastfeeding [82]. Hence, it was not surprising to find S. epidermidis in the first meconium obtained from both term and preterm breastfed neonates [83,84]. Interestingly, it was demonstrated that maternal milk mostly contains S. epidermidis isolates negative for the methicillin resistance gene mecA [81,85], which may limit the intestinal spread of mecA-positive S. epidermidis NICU-associated strains that are a major cause of LOS [86]. Hence, as in adults, CoNS seem to play an important role in the defense of neonates by competing with more virulent strains/species.

3. CoNS Role as a Pathogen in Newborns

Although LOS caused by CoNS seems to present less neonatal morbidity rates than LOS caused by other Gram-positive or Gram-negative bacteria [24], it is commonly associated with important morbidities such as bronchopulmonary dysplasia, white matter injury, necrotizing enterocolitis and retinopathy of prematurity [3,87,88,89,90,91,92]. In extremely low birth weight infants (<1000 g), an increased risk of cerebral palsy, mental and psychomotor developmental index and growth impairment was observed [24,87]. As such, even though CoNS can have a benign relationship with the host, the pathogenic lifestyle of CoNS needs to be further explored.

3.1. Risk Factors for LOS Caused by CoNS

Preterm infants are more likely to develop a generalized life-threatening infection due to their theoretically immunocompromised condition, in which several components of the immune system are not fully developed at birth [63]. However, the high frequency of LOS caused by CoNS in preterm infants is also related to the fact that they are more likely to require prolonged use of central catheters, parenteral nutrition and mechanical ventilation, which are increased risk factors for CoNS infection [14,25,93,94] (Figure 1). In that sense, it was found that the analyses of bodily fluids such as blood, which are often necessary for diagnosis and implicate the use of invasive procedures, can increase the risk for CoNS-related LOS independently [95]. Recently, it was demonstrated that umbilical catheters are another vector for skin microflora transmission of CoNS into the bloodstream, via biofilm formation [96].
While the use of indwelling medical devices generates an increased risk for CoNS-related LOS, one-third of LOS cases are not associated with medical devices [97], suggesting other risk factors/sources of infection. For instance, the use of formula feeding [95,98,99] and parenteral nutrition with glucose can increase the risk of preterm infants being infected with CoNS [100]. Gut colonization is another route of contamination in preterm infants with immature intestinal mucosal barriers and dysbiosis [101,102]. The gut of preterm neonates becomes colonized, within the first days, with S. epidermidis and S. haemolyticus carrying the mecA gene, which is associated with an increased risk of infection [103]. These strains spread in the NICU afterwards causing LOS [102,104,105]. Indeed, it was shown that S. epidermidis can translocate from the gastrointestinal tract into the bloodstream causing infection [101,102]. Furthermore, it was found that S. capitis also colonizes the gut of neonates. Butin et al. observed that among the neonates evaluated, 36% had at least one stool culture positive for S. capitis during hospitalization [106]. However, differently than S. epidermidis or S. haemolyticus, the presence of S. capitis in the gut microbiota was not a risk factor for LOS development. This suggests that the gut may serve as a reservoir of S. capitis but this is not enough to induce LOS [106]. The presence of such strains/species in the gut before the emergence of LOS [102,104,105,107] indicates that alterations in the gut microbiota could prevent the development of LOS [86]. As detailed in Section 2, this could be achieved by feeding neonates with maternal milk that is rich in CoNS that are mecA negative [80,85].
Finally, the colonisation rate among gynecological surgical staff with methicillin-resistant CoNS constitutes another important route of contamination of preterm babies. Indeed, it has been shown that the hands of healthcare personnel are the major sources of S. epidermidis transmission among neonates [97,102,108]. Thus, hygiene policies need to be revisited and strictly followed to decrease this venue of contamination.

3.2. CoNS Virulence Factors

The virulence potential of bacteria is normally defined by their capacity to produce toxins. In the case of CoNS species, although they produce and secrete toxins and exoenzymes, they normally rely on immune evasion mechanisms to cause infection [109]. One of the major factors contributing to CoNS pathogenesis is their ability to form biofilms [37,47,48], as biofilm-related infections are often chronic, persistent and, therefore, difficult to treat. As such, toxins, exoenzymes and all the molecules involved in biofilm formation are often considered good targets for the development of prophylactic and/or therapeutic strategies against CoNS infections.

3.2.1. Toxins and Exoenzymes

Toxin production in CoNS is mainly restricted to the production of phenol-soluble modulins (PSMs). PSM are peptides with surfactant properties that are produced by all Staphylococcal species; however, every species produces a specific repertoire, with some but not all sharing sequence similarities [110]. Studies in S. epidermidis and S. haemolyticus have shown that PSMs have multiple functions, including in pathogenesis by inducing neutrophils chemotaxis, cytokine release and leukocytes and erythrocytes lysis [111,112,113]. Importantly, Qin et al. have demonstrated, for the first time, that sepsis due to S. epidermidis resistant to methicillin is mediated, to a large extent, by the PSM-mec peptide toxin [114]. Of note, since PSM-mec is encoded by psm-mec gene locus located in the mobile genetic element staphylococcal chromosome cassette mec (SCCmec), which carries the mecA gene, the PSM-mec occurs in a series of methicillin-resistant CoNS [115], which are widespread in the clinical setting, specifically in the NICU [97,102,104,108].
Also contributing to CoNS pathogenicity is the ability to produce and secrete exoenzymes. In S. epidermidis the metalloprotease SepA plays an important role in the protection against the host immune system response, in particular against antimicrobial peptides (AMPs) and neutrophils [111,116]. Moreover, S. epidermidis produces an endopeptidase, Esp, that can degrade complement factors [117] and lipases (GehC and GehD) that facilitate bacteria survival in host fatty acid secretions [118,119,120]. In addition, it was shown that S. epidermidis produces a fatty acid-modifying enzyme that inactivates host fatty acids harmful to bacteria [121].

3.2.2. CoNS Biofilm Formation

Biofilms are recognized as the common form of microbial growth [122], including during infection (source of ~65% of medical device-related nosocomial infections [123]), conferring protection against the host immune system [124] and antibiotics challenges [125]. However, the role of biofilm formation by CoNS in the outcome of LOS is unclear, mainly because of the difficulty, both practical and ethical, of studying newborns, especially preterm. Yet, it is known that biofilms are formed right after the implantation of medical devices and that the persistence of biofilms may provide a basis for a continuous release of bacteria into the bloodstream [126,127]. Hence, it is likely that biofilms formed on medical devices are the source of persistent LOS [92].
Biofilms are defined as complex communities of bacteria surrounded by a protective matrix, attached or not to a surface [128]. Biofilms are formed following three main stages; (i) initial attachment, (ii) maturation and (iii) dispersal [129] (Figure 2). As can be expected, the first stage is only important in the case of biofilms attached to a surface, such as medical devices, which is the focus of this review. Although CoNS can be attached directly to the medical device surface, in vivo, medical devices are readily coated by host proteins. As such, CoNS normally form biofilms through direct interaction with host matrix proteins [130]. This process is mostly mediated by MSCRAMMs, i.e., microbial surface components recognizing adhesive matrix molecules [131,132], such as serine-aspartate repeat protein G (SdrG) and F (SdrF) [133]. In addition, Atl-type autolysins [134], extracellular matrix binding protein (Embp) [135] and wall teichoic acids [136] also aid the adhesion of S. epidermidis to host matrix proteins or tissue, respectively.
Having successfully attached to the surface, bacteria start to proliferate and secrete molecules that will give rise to the protective matrix, entering the second stage of biofilm formation, biofilm maturation [137]. The matrix is composed of several molecules including proteins, polysaccharides, teichoic acids, as well as DNA originating from lysed cells. Depending on the composition of the matrix, biofilms can be categorized as either polysaccharide- or protein-dependent biofilms. Importantly, the composition of the matrix can be modulated by environmental factors such as substrate availability and shear forces [137]. It was observed that S. epidermidis isolates recovered from environments with high shear forces are mainly polysaccharide-dependent in contrast with the ones isolated from low-shear environments [138]. In CoNS biofilms, notably in S. epidermidis, the polysaccharide poly-N-acetylglucosamine (PNAG), is one of the most prominent molecules [139] and up to 60% of the clinical isolates are known to produce PNAG [140,141,142,143,144]. PNAG, synthesized by the proteins encoded in the genes icaADBC [145], has a positive net charge, enabling the interaction with negatively charged molecules, thereby creating the distinguishing sticky characteristic of the matrix.
Although S. epidermidis biofilm formation mainly relies on PNAG, as referred to above, biofilms can also be formed through the mediation of proteins such as accumulation–associated protein (Aap) [146], biofilm homologue protein (Bhp) [147] and Embp [135]; however, these biofilms are less robust. Importantly, in addition to the adhesive molecules that maintain the biofilm structure, it is also necessary to generate channels for water and nutrients to reach the deeper layers of the biofilm, as well as to eliminate waste. These channels are shaped by the action of proteases (SepA, Esp, EcpA) [117,148,149,150], nucleases and molecules that disrupt non-covalent interactions, notably PSMs [127,151]. Depending on the extension of the action of these molecules, biofilm structuring can lead to the third stage of the biofilm lifecycle, biofilm dispersal. This last cycle is important to maintain the biofilm lifecycle, as the cells can colonize new locations and establish other foci of infection.

Biofilm Formation as an Antibiotic Tolerance/Resistance Mechanism

As recently defined by the consensus statement on the definitions and guidelines for research on antibiotic persistence, while antibiotic-resistant cells are defined as cells that can grow in the presence of antibiotics by carrying a resistance factor, antibiotic-tolerant cells are defined as cells that can survive antibiotic treatment (slower killing), without carrying a resistance factor, and can regrow after removal of the antibiotic; these tolerance factors can be either environmental or genetic [152]. At the beginning of biofilm studies, it was shown that biofilm cells were 10 to 1000 times more tolerant to antibiotics than their planktonic counterparts [153]. From then on, several studies were performed to identify the mechanisms allowing such a level of tolerance. Among them is the metabolic heterogeneity within biofilm cells [154,155]. Indeed, Rani and collaborators have characterized the spatial distribution of DNA replication, protein synthesis and oxygen concentration on staphylococcal biofilms and four subpopulations of cells were identified: (i) aerobically growing, (ii) fermentatively growing, (iii) dormant and (iv) dead cells, which accounted for ~10% of the total number of cells [156]. This heterogeneity is associated with the particular chemical and nutrient gradient within the several layers of the biofilm, resulting in different gene expression patterns and, thus, cells in different metabolic states [157]. Such a heterogeneous pattern of growth within a biofilm contributes to the alteration of the susceptibility profile to antibiotics. Indeed, this heterogeneity promotes the emergence of cell subpopulations with physiological characteristics which render them resilient to certain antibiotics [154,155]. As such, antibiotics whose targets are actively growing cells, i.e., actively dividing and synthesizing nucleic acids and proteins, will fail to act on slow-growing cells and/or cells with low metabolism [158].
Another important mechanism that hinders the activity/efficiency of antibiotics is the matrix. While it was demonstrated that many antibiotics such as vancomycin and cefotaxime have impaired capacity to penetrate the matrix of Staphylococcal biofilms [159,160], the matrix can also function as a decoy, trapping antibiotics [161,162]. In addition, due to the negative charge of polysaccharides and extracellular DNA, the matrix may interfere with the ability of positively charged antibiotics to act [163]. The matrix may also function as a reservoir of enzymes with activity against antibiotics, such as beta-lactamase [164] and aminoglycoside-modifying enzymes [162]. Interestingly, these enzymes are mainly accumulated in the outer layers of the biofilm, constituting the biofilm’s first line of defense [165]. Furthermore, PNAG-positive S. epidermidis strains showed increased resistance to several antibiotics and the cell wall degrading enzyme lysostaphin, as compared to PNAG-negative strains [166,167,168]. Noteworthy, sub-inhibitory concentrations of some antibiotics resulted in increased transcription of the ica locus presumably leading to higher contents of PNAG [169].
Concerning antibiotic resistance in biofilms, horizontal gene transfer (HGT) is one of the most important mechanisms of exogenous DNA acquisition in staphylococci and, thus, of obtaining antibiotic resistance genes [170]. Biofilms are major mediators of HGT due to the (i) high number and (ii) proximity of cells, (iii) high genetic competence and (iv) greater availability of exogenous DNA [171]. Indeed, it has been reported that the horizontal transfer of resistance-conferring genes is 700 times more efficient in biofilm than in free-living cells [172], further exacerbating the spread of antibiotic resistance [123]. Furthermore, mutations play an important role in the development of antibiotic resistance in staphylococcal species [173] and it was shown that biofilm cultures present increased mutation frequencies [174]. Ryder et al. have shown that S. epidermidis biofilm cells presented 4 times higher mutability than planktonic cells, increasing in this way their resistance to rifampicin [174].

Biofilm Formation as an Immune System Resistance Mechanism

In addition to the protection against antibiotics, biofilm formation has also an important role in the defense of biofilm cells from the host immune system attack [175]. Biofilm cells are known to present a decreased ability to stimulate the innate immune system, resulting in a lower inflammatory response. This response is mediated by several mechanisms that include the presence of dormant cells [176,177], limited expression of toxins with proinflammatory effects (namely PSMs) [151,178] and the presence of PNAG [175]. In the case of S. epidermidis, it was shown that biofilms with higher proportions of dormant cells presented a decreased ability to induce the production of pro-inflammatory cytokines by in vitro cultured bone marrow-derived dendritic cells [176] and murine macrophages, either in vitro or in vivo [177]. Interestingly, it was found that S. epidermidis isolates obtained from device-related infections, mainly joint infections [178], often present a mutation in the quorum sensing Agr system that renders it dysfunctional. As such, since PSMs are under strict regulation of the Agr system [179], they are not produced; this has consequences not only for CoNS pathogenicity but also in biofilm structuring, presenting higher biofilm formation in vivo [178]. Hence, it seems that disabling the Agr system enhances the success of the bacterium in vivo. Furthermore, a recent study has followed the evolution of an S. epidermidis strain over the 16 weeks of pacemaker-associated endocarditis, and the capacity to form biofilm increased over time, as determined by in vitro assays [180]. Moreover, several mutations were detected, including in the Agr system [180]. Regarding PNAG, besides its important function in biofilm formation and maintenance, it also has a fundamental role in immune evasion. PNAG was shown to protect S. epidermidis biofilm cells, in in vitro assays, from neutrophil and AMPs killing [181], antibody-mediated phagocytosis [182] and reduced deposition of complement proteins and IgG on the bacterial surface [183]. In addition, PNAG was shown to present a protective role in vivo, using animal models [183,184,185,186]. Furthermore, ex vivo experiments using cord blood samples reported that the levels of interleukin-6 (IL-6) in blood cultures incubated with a PNAG-positive S. epidermidis strain were lower as compared to that of a PNAG-negative strain [187]. Similar results were observed in the blood of adults [188].

4. LOS Diagnosis and Treatment

All the mechanisms described above contribute to the development of CoNS biofilm-associated infections, with a chronic and persistent nature, which are very hard to cure with current antibiotic therapy. Furthermore, due to the commensal nature of CoNS [189] and the characteristic metabolic heterogeneity of the cells within biofilms [37,190], the infections caused by this group of bacteria are difficult to diagnose, increasing their economical and clinical burden. As such, it is important to comprehend the methods available for the diagnosis and treatment of CoNS biofilm-based infections, to be able to identify the limitations and, subsequently, develop efficient strategies to detect and treat CoNS-related infections, such as LOS.

4.1. Diagnosis of LOS

Despite the fair advances made in medicine over the years, LOS is still challenging to quickly and accurately diagnose, leading to delays in the application of an adequate treatment, which in turn may result in a higher risk of complications and, ultimately, a higher mortality rate [191]. This difficulty is attributed to several factors such as (i) variable or non-specific symptoms displayed by neonates [21,192], (ii) the maternal intrapartum antibiotic therapy that can mask the presence of bacteria [193] and (iii) the technical limitations of the diagnosis methods currently used [26,194]. Currently, the gold standard for the diagnosis of LOS is the identification of the causative agent through culture methods, namely from blood samples [195,196]. However, this method has time limitations resulting in a delayed diagnosis. As such, to try to speed up the diagnosis, other indicative analyses such as the use of molecular methods to help identify the causative agents, quantification of inflammatory molecules and the evaluation of clinical symptoms are often combined. Nevertheless, a timely and unambiguous diagnosis of LOS is still not available and, thus, there are still attempts to find new diagnostic strategies [12]. Figure 3 briefly summarizes the current, advanced and under-development diagnostic methods for the diagnosis of LOS.

4.1.1. Clinical Symptoms

The symptoms or clinical signs of NS, including LOS, are very comprehensive and can include symptoms that can naturally occur in preterm neonates [197]. In general, symptoms include, among others: (i) temperature instability, (ii) feeding difficulties/intolerance, (iii) apnoea and tachypnea, (iv) respiratory rate or desaturations, (v) heart rate variability, (vi) hypotension, (vii) irritability or seizures [15,20,192,198,199,200]. However, because of the variable or non-specific symptoms displayed by neonates [21,192], further analyses are necessary to be able to accurately diagnose LOS. However, the diagnosis of LOS can be made solely based on clinical symptoms, which is designated as clinical sepsis [195,196].

4.1.2. Biological Samples Culture

LOS is confirmed by isolating the causative agent from sterile body sites such as blood, urine and cerebrospinal, pleural, joint, and peritoneal fluids [15]. Blood cultures are the current gold standard for LOS diagnosis. However, the long turnaround time (up to 72 h or 48 h if automated systems are used), the small volume of blood and the reduced number of samples that can be drawn from preterm neonates, as well as the intermittent bacteremia, render this method often insufficient and insensitive [193]. Since most of the deaths occur within the first three days after blood cultures are obtained, faster and more sensitive identification of the causative pathogen is essential for early diagnosis and faster guidance of therapy [14]. In the case of CoNS biofilm-related infection, there are two additional limitations. On one hand, due to their commensal nature in human skin, CoNS-positive blood cultures are often interpreted as contamination during sample collection. To avoid misdiagnosis, specific criteria to define CoNS sepsis were delineated and although they may vary depending on the country and clinical staff, the most well-accepted criterion to consider LOS caused by CoNS is to obtain two positive blood cultures obtained within 2 days [14,201]. On the other hand, as described before staphylococcal biofilms are characterized by the presence of cells with different metabolic states. These include, among others, viable but non-culturable cells that, as the name indicates, are viable but cannot grow in culture media. Thus, despite being alive, these cells are not detected by culture-based methods hindering the interpretation of culture results [190]. Overall, these limitations have triggered the search for additional diagnosis methods, specifically culture-independent techniques.

4.1.3. Biomarkers—Inflammatory Molecules and Hematologic Indices

Considering that the host immune system reacts to the presence of microorganisms, it is reasonable to expect that the characterization of the molecules produced in the context of infection can support the diagnosis of NS. To be considered good biomarkers, among other characteristics, these molecules need to present high sensitivity (~100%), specificity (>85%) and high positive (>85%) and negative (~100%) predictive values of NS [202,203]. In that sense, over the last years, several studies have investigated the ability of inflammatory molecules to be used as biomarkers such as interleukins (IL-1β, IL-6, IL-8, TNF-α), cell adhesion molecules (CD11b, CD14, CD64), acute-phase reactants (procalcitonin, C-reactive protein, serum amyloid A), as well as hematologic indices such as white blood cells, neutrophils counts and platelets counts [192,202,204,205,206]. Despite the several developments regarding the identification of biomarkers for NS diagnosis, biomarkers specifically for LOS detection are poorly known.
LOS in preterm infants, in particular in extremely preterm infants, is mainly caused by CoNS, which seems to cause less inflammatory response compared with other bacteria [207,208,209]. Consequently, this less virulent profile can impair the application of inflammatory molecules as biomarkers in this case. Recently, Mwesigye and collaborators observed that there is growing evidence that C-reactive protein (CRP), an acute phase reactant synthesized by the liver in response to pro-inflammatory cytokines, is losing strength as a biomarker for LOS [210] despite being the most commonly used and widely implemented biomarker so far [192,204,206,211,212]. Notwithstanding the uncertainties regarding its predictive significance, CRP is still the object of analysis in several studies and it was shown that although CRP alone does not seem to be the best predictive biomarker, in combination with complete blood count, for instance, it has a good sensitivity [213]. However, other studies have shown that serum amyloid A, the expression of CD64 on neutrophils surface, presepsin and endocan have higher potential for a more accurate diagnosis of LOS [210,211,214]. Procalcitonin (PCT), another acute phase reactant, holds promising sensitivity for the prediction of LOS alone [215,216] and in combination with IL-6 [212]. Sherbiny and collaborators have also shown that hepcidin, which controls the levels of systemic iron [217], constitutes another possibility for rapid and accurate diagnostics of LOS [218,219]. Finally, it has been demonstrated that the levels of IL-6, PCT and CRP in CoNS-positive blood cultures were associated with increased mortality and, thus, a combination of these three molecules can establish a good biomarker [207]. Yet, these results were obtained with a small sample, requiring further investigation. Concerning hematologic indices, the immature-to-total and immature-to-mature neutrophil ratios and platelet counts were found to be highly predictive laboratory signs, but immature-to-total seems to be the most reliable [220,221,222]. It is important to stress that the selection of the best biomarker for NS diagnosis, namely LOS, depends on several aspects such as gestational age, time of infection and sampling [223]. Thus, the best biomarkers will depend on the particular clinical situation.
Although advantageous in some respects, the use of inflammatory biomarkers has a downside; the inflammatory molecules detected can be of non-infectious origin confusing the results. As such, these biochemical analyses do not have sufficient diagnostic accuracy to support a decision without considering the results of microbial cultures [224] but constitute a good “add-on” method.

4.1.4. Molecular Biology Methods—Nucleic Acids Analysis

To try to increase sensitivity and accelerate the diagnosis of LOS, molecular methods based on the detection of nucleic acids (DNA or RNA) are being increasingly used. These are either based on amplification (PCR, qPCR), probes hybridization (fluorescence in situ hybridization) or sequencing techniques (whole generation sequencing) [204]. The simplification of nucleic acids amplification procedures and instruments and the development of automated systems have fostered the use of amplification-based methods in the clinical setting. Therefore, it is not surprising that most of the methods used in neonatal studies are based on the amplification of nucleic acids, followed by hybridization- and sequencing-based methods, which are normally more time-consuming and expensive, respectively.
Several of the routine molecular methods currently used for LOS diagnosis include kits such as GeneXpert, FilmArray, Verigene, PNA-FISH and QuickFISH, which can yield results in as little as 3 h [225]. Although these kits enable the detection of CoNS, due to the relevance of CoNS in the development of LOS, there are kits specifically developed to detect Staphylococcus epidermidis, Staphylococcus hominis, Staphylococcus haemolyticus, Staphylococcus saprophyticus and Staphylococcus capitis, which are among the most frequently isolated in the skin microbiome and important in the context of LOS [34,48,49]. However, these methods rely on positive blood cultures and, thus, although fast, these methods do not constitute a major advantage as they do not preclude the need for previous cultures. In that sense, recent advances in molecular methods have enabled the amplification of nucleic acids directly from biological samples [192,225,226]. Two examples are the kits SeptiFast, from Roche, and SepsiTest, from Molzym, which require 1 to 1.5 mL whole blood to be able to detect, respectively, 25 to more than 300 pathogens at the same time, with a turnaround time of 6 h and 10 h [227]. In the case of SeptiFast, 100 µL of blood can be used but the sensitivity drops to 80% [228].
On the other hand, PCR/qPCR-based methods may result in false-positive results [229] and the reported divergences between blood culture and PCR/qPCR results [230,231], have raised questions about the efficiency of molecular methods. In addition, the sensitivity of these methods depends on the quality of the template (nucleic acids quantity, integrity and purity), which in turn depends on the principle of the methods used for nucleic acids isolation and the quality of the initial samples [229,232]. Furthermore, the quality of the molecular assay is also affected by contamination, either through other organisms or within the laboratory environment, and in situations of low-level bacteremia [233]. Thus, for now, molecular methods have promising potential as complementary tests but not alone [204,224].

4.1.5. Molecular Methods—Proteins Analysis

One of the ultimate techniques for bacterial pathogen identification is matrix-assisted laser desorption-ionization/time-of-flight (MALDI-TOF). Although in the past this method required isolated colonies [234], following several developments it can be performed directly from blood, urine, and cerebrospinal fluid samples, significantly improving the turnaround time [235,236,237,238]. Nevertheless, for the direct analysis of clinical samples, additional pre-treatment protocols are necessary to selectively recover bacterial cells [239] since multiple proteins from the host (hemoglobin in blood cultures, for instance) may be present in high quantities [238]. Hence, despite the clear advances, further studies are required to address the current limitations in the detection of microorganisms directly from biological samples. Another promising addition to this method is the ability to analyze the pathogens’ antimicrobial susceptibility, giving results at least 24 h earlier than when identification and/or resistance detection is performed using conventional methods [240]. This feature can greatly improve the treatment of LOS and the outcome of infection [241].
The downside of this method is the initial investment, but the cost of sample processing is cheaper than several of the methods used, mainly when considering the higher number of samples that need to be processed daily in the hospital environment. However, although this is true for high-income countries, it is difficult to implement in developing countries. Moreover, for now, it seems that MALDI-TOF has higher sensitivity to detect Gram-negative bacteria from culture bottles, particularly Enterobacteriaceae, than Gram-positive bacteria [242,243]. As such, improvements are still necessary to accurately detect LOS caused by CoNS and to fully implement this methodology in clinical laboratories.

4.1.6. Diagnosis Approaches under Development

Despite the several methods currently available, there is a continuous search to advance the methods available or for the development of new methods that provide prognostic information or definitive diagnostics promptly. Point-of-Care devices, which can be done at the bedside and can yield rapid results are the Holy Grail of NS diagnosis. The detection of volatile organic compounds in the breath of patients has been shown to have the capacity to differentiate sepsis cases from inflammation [244,245]. Although promising, validation in human studies is necessary [244,245]. Another solution that has been explored is the development of devices that can detect a variety of biomarkers and, this way, obtain a more accurate diagnosis of NS [246].
As in any other field of science, Omics technologies and machine learning may be very useful in the advance of the methods used to diagnose NS, including LOS. Transcriptomics [247,248,249,250], proteomics [251,252,253,254,255] and metabolomics [252,256,257,258] have been used to characterize the response of preterm neonates to NS [259,260] and, together with machine learning approaches, can help to identify promising biomarkers, to determine NS risk, treatment response and prognosis [261,262]. Some steps were already made in that direction and Pediatric Sepsis Biomarker Risk Model, PERSEVERE, was developed and validated as a prognostic enrichment tool for pediatric septic shock [260,263]. Ongoing research is evaluating the use of PERSEVERE in the prediction of NS [204].

4.2. LOS Treatment

So far, the only available treatment option for NS, including LOS, is the use of antibiotics. As referred to above, due to the lack of methods that can accurately and quickly diagnose LOS, in the case of suspicion of LOS, clinicians initiate empirical broad-spectrum antibiotic treatments [21]. Indeed, several studies covering different countries have reported that most of the treatments for NS do not have a confirmed diagnosis. This can increase antibiotic resistance and, even though it may save lives, it may also result in important sequels for newborns such as reduced gut microbial diversity [264], invasive fungal infections, necrotising enterocolitis [265], inflammatory diseases [266] and increased likelihood of early childhood obesity [267]. Hence, several other therapeutic options, mainly antibiotic-independent, but also including the improvement of the activity and delivery of current antibiotics, with fewer side effects, are being explored. Herein, some of the strategies that are being investigated in the context of CoNS biofilms are outlined (Figure 4), but a more exhaustive list of strategies can be seen in [158,268,269,270]. Of note, due to biofilm characteristic heterogeneity (cells with different metabolic states and under different pH and levels of oxygen and nutrients), the use of combined strategies may increase our likelihood of effectively targeting biofilm infections. As such, there are some developments in the area already, with the combination of antibiotics with other new methods being one of the most common strategies [271,272,273,274,275].

4.2.1. Currently Available Therapies—Antibiotics

As discussed above, in the presence of symptoms and signs suggestive of NS, empiric therapy is initiated [20]. Despite the serious consequences that this approach may hold, it has been proven to reduce both the mortality and morbidity of affected newborns [277]. The most commonly recommended treatment for NS is a β-lactam antibiotic, normally ampicillin, flucloxacillin or penicillin, combined with an aminoglycoside, most frequently gentamicin [20,21]. In the particular case of LOS, a cephalosporin, usually cefotaxime, or a glycopeptide, such as vancomycin, may be used instead due to the increased resistance demonstrated by CoNS to the other antibiotics frequently used to treat NS [21,278,279,280,281]. However, it is important to stress that depending on the local antibiotic resistance of the most common pathogens causing LOS, the antibiotic regimen may change [282]. Importantly, the empiric therapy should be altered as soon as diagnosis results are obtained, and an immediate cessation of antibiotics shall be performed if culture-negative results are obtained and the infant shows no subsequent clinical evidence of NS [283]. The increasing resistance to antibiotics, within the hospital and in the community, is seriously compromising our ability to treat LOS, with CoNS being increasingly resistant to a plethora of antibiotics, especially against beta-lactam antibiotics and, more importantly, frequently presenting multi-drug resistance phenotypes (resistant to 3 or more classes of antibiotics) [283,284,285]. Among S. epidermidis neonatal isolates, epidemiological studies have reported that ~52.7–85% present a multi-drug resistance profile, with ~92–99% resistant to penicillin and ~70–78% to gentamicin [43,286,287]. In the case of S. haemolyticus, the studies performed showed that ~82–87% of the isolates are multi-drug resistant, with 90% of the isolates being resistant to penicillin and ~61–79% to gentamycin [44,287,288]. Studies analyzing S. capitis neonatal isolates are mainly focused on the clone NRCS-A, since it has recently emerged as a major pathogen of LOS, having already been isolated in several countries all over the world [25,38,45]. This clone has shown high resistance to penicillin (99%), methicillin (95.6%) and gentamicin (95.1%) [38]. With regards to vancomycin, all the S. epidermidis and S. haemolyticus neonatal isolates studied so far have shown 100% susceptibility to vancomycin [43,286,289]. However, in the case of S. capitis neonatal isolates, in particular, the ones belonging to the NRCS-A clone showed resistance (37.5%) or heteroresistance (62.5%) to vancomycin [38]. As such, considering that vancomycin is one of the suggested antibiotics to treat LOS, it is not surprising that cases of prolonged S. capitis LOS have been reported [290,291]. Thus, alternatives to vancomycin have to be considered [292] and daptomycin and linezolid have been suggested [293,294,295,296]. Yet, Butin et al. reported that vancomycin resistance in S. capitis NRCS-A was associated with an increase in daptomycin and teicoplanin minimal inhibitory concentrations, but not in linezolid [297]. This indicates that daptomycin may not be the best approach, but highlights linezolid as a possible alternative for the treatment of LOS caused by S. capitis [298]. However, the use of linezolid needs to be conservative as resistance to this drug has already been observed in S. capitis isolates collected from non-neonate patients [292] and may not take long to upsurge in neonates.
When in biofilms, CoNS are more tolerant to antibiotics. Indeed, Qu et al. have shown that the resistance of CoNS to several antibiotics increases significantly as the biofilm develops [285]. Thus, although the strains may be susceptible in antimicrobials susceptibility tests, which are performed with planktonic cells, the antibiotic in question may not cure the infection caused by CoNS biofilms. One antibiotic that has performed well against biofilms is rifampicin [299]. However, rifampicin quickly develops resistance when used alone and, thus, it should be used as part of a combined therapy [300,301]. It was shown that when added to penicillin or vancomycin, it is effective in the treatment of refractory cases of bacteremia caused by CoNS in infants [302,303,304]. However, the combination of antibiotics needs to be tested since it may have synergistic but also antagonistic effects [305].
Notwithstanding their importance to treat LOS, the use of antibiotics always entails risks. Vancomycin, for instance, is known to cause fever and phlebitis and, in rare cases, nephrotoxicity and ototoxicity in adults [306]. In addition, an increased risk of nephrotoxicity occurs when amino-glycosides are combined with vancomycin [306,307,308]. Concerning the use of linezolid, care should be taken regarding the development of lactic acidosis and acidemia, which should be carefully monitored [309]. Concerning rifampicin, adverse effects such as hepatotoxicity, renal failure, rash, and hematological abnormalities such as thrombocytopenia have been reported in adults [310]. When it comes to infants, although some studies performed did not detect significant adverse effects of the use of rifampicin [311,312], it is suggested that the level of bilirubin should be monitored during therapy. Moreover, more studies are necessary to ensure proper doses and duration of the use of rifampicin in infants. In addition to the specific adverse effects of each antibiotic, extended use of any antibiotics can increase the risk of developing neonatal candidemia [313,314]. Noteworthy, except when detailed, these studies were performed in adults and, thus, the consequences of the use of these antibiotics in preterm neonates remain unclear.
Hence, due to (i) antibiotic resistance escalation in CoNS, (ii) biofilm inherent tolerance to antibiotics and a hot spot for HTG, (iii) the increased number of preterm infants and (iv) the fact that antibiotics are the only available treatment solution, CoNS biofilm-related infections are likely to upsurge and will be extremely difficult to eradicate. Therefore, new treatment strategies encompassing antibiotic-independent approaches, but also the alteration of already existing antibiotics or the design of new ones, are urgent.

Improved Antibiotic-Based Treatment Approaches

Although studies exploring new synthetic or altered antibiotics, used alone or in combination, for the treatment of CoNS biofilm-related infections are rare, encouraging results have been shown for S. aureus, indicating that the same approach can be applied to CoNS species. Antonoplis et al. have developed a synthetic vancomycin, vancomycin-d-octaarginine conjugate (V-r8), which was able to eradicate 97% of biofilm-associated methicillin-resistant S. aureus in a murine wound infection model [315]. Furthermore, another study has altered tobramycin in a way that it was able to kill S. aureus persister cells 4 to 6 logs more effectively than the original tobramycin [316]. With regard to the combination of classic antibiotics with new molecules, Kim et al. have shown that synthetic retinoid antibiotics CD437 and CD1530 were able to kill methicillin-resistant S. aureus cells, including persister cells, and their efficacy was enhanced in the presence of gentamicin [272]. Furthermore, the combination of bithionol and gentamicin reduced the load of S. aureus cells in a mouse model of chronic deep-seated methicillin-resistant S. aureus infections [271]. Similar results were obtained with 5-methylindole, which together with tobramycin was able to kill S. aureus persisters in a skin wound mouse model and showed good activity in vitro against S. epidermidis cells when in combination with gentamicin and kanamycin [273].

4.2.2. Antibiotic-Independent Treatment Approaches under Development

Although new treatment strategies are being explored, most are still under preclinical tests. Several recent reviews detail the most promising strategies being developed for the treatment of staphylococcal infection [128,137,158]. As such, herein, only a brief outline of the strategies that can be used in the future to treat CoNS biofilm-related infections is presented. Worth mentioning, the in vivo efficacy of these strategies still needs to be demonstrated.
Among the strategies being explored, there are some strategies that even though they present exciting results in several bacteria, in the case of Staphylococcal species, including CoNS, may not work that well. One of those examples is the use of AMPs. AMPs are very popular due to their strong antimicrobial activity, irrespective of cells’ metabolic state and resistance to antibiotics, and because AMPs are less prone to induce resistance [274]. However, when it comes to the treatment of staphylococcal infections, the use of AMPs may fail as Staphylococcal species have learned to sense AMPs and counteract them efficiently [317,318]. Yet, some studies are reporting the ability of AMPs to treat staphylococcal biofilms, namely S. aureus [319]. Hence further studies are necessary to accurately determine the ability of AMPs to treat CoNS-based infections. Another example is the use of molecules with the capacity to block quorum-sensing communications. These molecules may not be the best approach to deal with staphylococcal biofilms, since as was shown earlier the interference with the Agr system may increase biofilm formation [128,178].

Biofilm-Degrading Enzymes

Biofilm structure is mainly maintained by the matrix that protects and holds all the cells together. It is also the structure of the biofilm that promotes the different gradients of oxygen and nutrients, responsible for the heterogeneity of the cells within the biofilm. Hence, if this structure is destroyed, it is reasonable to assume that the particularities of the biofilm are lost and, thus, the free cells become more exposed and susceptible. Nevertheless, it is important to consider that the cells detached from the biofilm will likely lead to reinfection in the host [127]. Thus, the use of molecules that lead to biofilm disassembly needs to be used in combination with antibiotics to avoid subsequent regrowth [320,321]. Yet, some studies in S. epidermidis have revealed that the cells released from the biofilms are more tolerant to antibiotics and stimulate a more aggressive immune response than their planktonic or even biofilms counterparts [53,322]. Hence, the use of biofilm-degrading enzymes needs to be carefully evaluated. Examples of such biofilm degrading enzymes are dispersin B, produced by Actinobacillus actinomycetemcomitans, which degrades PNAG [321,323,324,325]; DNases I, that degrades extracellular DNA; protein K, that degrades proteins [137], and lysostaphin that degrades peptidoglycan from Staphylococcal species, including S. epidermidis [326]. In addition, it is important to consider the side effects of applying molecules with broad activity, such as proteases, as these may interfere with host proteins and tissues [327,328] and may have unexpected consequences.

Bacteriophages and Lysins

Bacteriophages (phages) are viruses that infect bacteria and their utilization for the treatment of infections, including biofilm-based, is on the rise due to their safe use in humans and for the environment and because they do not interfere with the healthy microbiota [158,329,330]. This has to do with the fact that bacteriophages (i) present narrow host specificity, preventing the killing of beneficial bacteria, (ii) affect both antibiotic-susceptible and resistant bacteria and (iii) are bactericidal (lytic phages), with their efficacy not relying on biofilm cells’ physiology [331]. Another important feature of phages is the ability to reach and infect the cells within the biofilm. Indeed, González and collaborators have shown that two staphylococcal phages (philPLA-RODI and philPLA-C1C) were able to move across the biofilm and propagate [332].
In S. epidermidis, the polyvalent Phage K has shown activity against biofilm cells [333] and a more recent study has demonstrated that a novel bacteriophage, DRA88, in combination with Phage K, strongly reduced the biomass of CoNS biofilms [334]. Furthermore, an S. epidermidis-specific phage, SEP1, was able to infect scraped biofilms, persister and biofilm-released cells. Interestingly, these findings suggest that its activity was affected by the biofilm matrix [335,336], which may be a downside of phage utilization. However, some staphylococcal phages harbor enzymes with the capacity to degrade polysaccharides, which can surpass the limitation posed by the biofilm matrix [337]. Despite the clear advantages of using phages, attention should be paid to the concentration of phages to be used, as it has been reported that the application of low doses resulted in Staphylococcus aureus biofilm development [338,339].
In addition to using phages, enzymes encoded by phages, namely lytic proteins such as endolysins and peptidoglycan hydrolases can also be used [158,340]. Similarly to phages, lytic proteins are specific to their target and were able to kill persister cells [341]. For instance, the endolysin LysGH15 was able to disrupt biofilms formed by some LOS-relevant CoNS, namely S. epidermidis, S. haemolyticus and S. hominis [342]. Noteworthy, the efficacy of LysGH15 was evaluated in vivo and a lower bacterial load was detected in both the blood and solid organs of endolysin-treated subjects [342]. CF-301, another endolysin, has also been shown to efficiently disturb biofilms formed by several CoNS species, including biofilms formed by different staphylococcal species [343]. Both phages and lytic proteins may be used alone or in combination with other management methods such as antibiotics. In fact, there are some promising studies in S. aureus showing the synergism between phages and antibiotics [275,344,345].
Although not directly related to the natural capabilities of phages to target CoNS biofilm-based infections, phages can be used as tools to deliver other mechanisms capable of interfering with biofilm viability. For instance, the delivery of CRISPR-Cas systems by modified bacteriophages was proposed as a potential strategy in the treatment of staphylococcal infections, including biofilm-based infections [346,347]. However, more studies are necessary to evaluate the CRISPR-Cas system’s impact on the treatment of staphylococcal biofilm infection.

5. Conclusions

Notwithstanding being the most frequently isolated Gram-positive bacteria in the context of LOS all over the globe, the infections caused by CoNS are normally perceived as an infection with relatively good outcomes, being often underestimated. However, previous and recent epidemiological studies, as well as the reported short-and long-term neurologic and developmental sequels, have raised awareness of the importance of these commensals as serious neonatal pathogens.
Although a lot of information is available regarding CoNS biofilms, most of these studies were performed in in vitro conditions, and it has become clear that in vitro observations cannot always be transferable to the in vivo level. Furthermore, information regarding CoNS biofilms’ role in NS is very limited. This is mainly related to the lack of in vivo models able to resemble preterm infants’ immune systems. As such, more in vivo studies are necessary to enable full comprehension of the role of CoNS biofilms in LOS outcomes. To surpass the limitations of working with reduced neonatal samples, omics and machine learning analysis can be of help.
Another complication associated with CoNS biofilm infections is the increased resistance to antibiotics that have been seen among these species, including multidrug resistance, together with the higher tolerance of biofilm cells. Despite the several advances made in the development of antibiotic-independent treatment strategies, most still need in vivo validation, followed by preclinical trials, anticipating that new treatment strategies will only be available in the market many years from now.
For all the aforementioned reasons, it is important to emphasize that despite all the studies that can be completed to comprehend the pathogenicity of CoNS biofilm infections and the response of preterm infants in the context of LOS, one of the most important aspects in the reduction of neonatal infection lies in preventive measures [24]. Therefore, the prevention of preterm births, prolonged use of antibiotics, and use of invasive procedures disrupting skin microbiome, as well as promotion of early feeding with breast milk and strict hand hygiene may constitute the best allies for the control of LOS caused by CoNS [24].

Funding

This work was supported by the Portuguese Foundation for Science and Technology (FCT) under the scope of the project with the reference EXPL/BIA-MIC/0032/2021.

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.

Abbreviations

AbbreviationDefinition
NSNeonatal sepsis
EOSEarly-onset sepsis
LOSLate-onset sepsis
CoNSCoagulase-negative staphylococci
GBSGroup B Streptococcus
NICUNeonatal intensive care unit
MSCRAMMsMicrobial surface components recognizing matrix molecules
AtlAutolysins
TATeichoic acids
eDNAExtracellular DNA
PNAGPoly-N-acetyl-glucosamine
EmbpExtracellular matrix binding protein
HGTHorizontal gene transfer
SrdSerine-aspartate repeat proteins
AapAccumulation-associated protein
BhpBiofilm-homologue protein
SepAS. epidermidis metalloprotease A
EspS. epidermidis serine protease
EcpAS. epidermidis protease A
PSMPhenol-soluble modulins
AMPsAntimicrobial peptides
CRPC-reactive protein
PCTProcalcitonin
PCRPolymerase chain reaction
qPCRQuantitative-polymerase chain reaction
FISHFluorescence in situ hybridization
PNA FISHPeptide nucleic acid fluorescence in situ hybridization
MALDI-TOFMatrix-assisted laser desorption-ionization/time-of-flight
CRISPR-CasClustered regularly interspaced short palindromic repeats

References

  1. Preterm Birth. Available online: https://www.who.int/news-room/fact-sheets/detail/preterm-birth (accessed on 1 November 2022).
  2. Liu, L.; Oza, S.; Hogan, D.; Chu, Y.; Perin, J.; Zhu, J.; Lawn, J.E.; Cousens, S.; Mathers, C.; Black, R.E. Global, Regional, and National Causes of under-5 Mortality in 2000–15: An Updated Systematic Analysis with Implications for the Sustainable Development Goals. Lancet 2016, 388, 3027–3035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Schlapbach, L.J.; Aebischer, M.; Adams, M.; Natalucci, G.; Bonhoeffer, J.; Latzin, P.; Nelle, M.; Bucher, H.U.; Latal, B.; Zeilinger, G.; et al. Impact of Sepsis on Neurodevelopmental Outcome in a Swiss National Cohort of Extremely Premature Infants. Pediatrics 2011, 128, e348–e357. [Google Scholar] [CrossRef] [PubMed]
  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. James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Incidence, Prevalence, and Years Lived with Disability for 354 Diseases and Injuries for 195 Countries and Territories, 1990-2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Roth, G.A.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Age-Sex-Specific Mortality for 282 Causes of Death in 195 Countries and Territories, 1980-2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1736–1788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Singer, M.; Deutschman, C.S.; Seymour, C.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801. [Google Scholar] [CrossRef]
  8. Molloy, E.J.; Wynn, J.L.; Bliss, J.; Koenig, J.M.; Keij, F.M.; McGovern, M.; Kuester, H.; Turner, M.A.; Giannoni, E.; Mazela, J.; et al. Neonatal Sepsis: Need for Consensus Definition, Collaboration and Core Outcomes. Pediatr. Res. 2020, 88, 2–4. [Google Scholar] [CrossRef] [Green Version]
  9. Ershad, M.; Mostafa, A.; Cruz, M.D.; Vearrier, D. Neonatal Sepsis Risk Factor American Academy of Pediatrics. Curr. Emerg. Hosp. Med. Rep. 2019, 7, 83–90. [Google Scholar] [CrossRef] [Green Version]
  10. Camacho-Gonzalez, A.; Spearman, P.W.; Stoll, B.J. Neonatal Infectious Diseases: Evaluation of Neonatal Sepsis. Pediatr. Clin. North Am. 2013, 60, 367–389. [Google Scholar] [CrossRef] [Green Version]
  11. Jyoti, A.; Kumar, S.; Kumar Srivastava, V.; Kaushik, S.; Govind Singh, S. Neonatal Sepsis at Point of Care. Clin. Chim. Acta 2021, 521, 45–58. [Google Scholar] [CrossRef]
  12. Singh, M.; Alsaleem, M.; Gray, C.P. Neonatal Sepsis. StatPearls 2022, 390, 1770–1780. [Google Scholar]
  13. Bizzarro, M.J.; Raskind, C.; Baltimore, R.S.; Gallagher, P.G. Seventy-Five Years of Neonatal Sepsis at Yale: 1928-2003. Pediatrics 2005, 116, 595–602. [Google Scholar] [CrossRef] [PubMed]
  14. 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]
  15. Shane, A.L.; Sánchez, P.J.; Stoll, B.J. Neonatal Sepsis. Lancet 2017, 390, 1770–1780. [Google Scholar] [CrossRef] [PubMed]
  16. Shah, B.A.; Padbury, J.F. Neonatal Sepsis: An Old Problem with New Insights. Virulence 2014, 5, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Simonsen, K.A.; Anderson-Berry, A.L.; Delair, S.F.; Dele Davies, H. Early-Onset Neonatal Sepsis. Clin. Microbiol. Rev. 2014, 27, 21–47. [Google Scholar] [CrossRef] [Green Version]
  18. Lawn, J.E.; Bianchi-Jassir, F.; Russell, N.J.; Kohli-Lynch, M.; Tann, C.J.; Hall, J.; Madrid, L.; Baker, C.J.; Bartlett, L.; Cutland, C.; et al. Group B Streptococcal Disease Worldwide for Pregnant Women, Stillbirths, and Children: Why, What, and How to Undertake Estimates? Clin. Infect. Dis. 2017, 65, S89–S99. [Google Scholar] [CrossRef] [Green Version]
  19. 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]
  20. Cortese, F.; Scicchitano, P.; Gesualdo, M.; Filaninno, A.; De Giorgi, E.; Schettini, F.; Laforgia, N.; Ciccone, M.M. Early and Late Infections in Newborns: Where Do We Stand? A Review. Pediatr. Neonatol. 2016, 57, 265–273. [Google Scholar] [CrossRef] [Green Version]
  21. 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]
  22. Shane, A.L.; Stoll, B.J. Neonatal Sepsis: Progress towards Improved Outcomes. J. Infect. 2014, 68 (Suppl 1), S24–S32. [Google Scholar] [CrossRef] [PubMed]
  23. Goh, G.L.; Lim, C.S.E.; Sultana, R.; De La Puerta, R.; Rajadurai, V.S.; Yeo, K.T. Risk Factors for Mortality From Late-Onset Sepsis Among Preterm Very-Low-Birthweight Infants: A Single-Center Cohort Study From Singapore. Front. Pediatr. 2022, 9, 801955. [Google Scholar] [CrossRef]
  24. 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] [PubMed] [Green Version]
  25. 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] [PubMed] [Green Version]
  26. 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]
  27. Berardi, A.; Sforza, F.; Baroni, L.; Spada, C.; Ambretti, S.; Biasucci, G.; Bolognesi, S.; Capretti, M.; Carretto, E.; Ciccia, M.; et al. Epidemiology and Complications of Late-Onset Sepsis: An Italian Area-Based Study. PLoS ONE 2019, 14, e0225407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Ferreira, A.; Sousa, E.; Freitas, J.; Viana, M.; Miranda, F.; Silva, F.P. da Positive Blood Culture and Neonatal Sepsis—A Five-Year Study. NASCER E CRESCER BIRTH GROWTH Med. J. 2022, 31, 106–114. [Google Scholar] [CrossRef]
  29. Størdal, E.H.; Solevåg, A.L.; Bjørnholt, J.V.; Rønnestad, A.; Stensvold, H.J. Sepsis Treatment Options Identified by 10-Year Study of Microbial Isolates and Antibiotic Susceptibility in a Level-Four Neonatal Intensive Care Unit. Acta Paediatr. Int. J. Paediatr. 2022, 111, 519–526. [Google Scholar] [CrossRef]
  30. Flannery, D.D.; Edwards, E.M.; Coggins, S.A.; Horbar, J.D.; Puopolo, K.M. Late-Onset Sepsis Among Very Preterm Infants. Pediatrics 2022, 150, e2022058813. [Google Scholar] [CrossRef]
  31. Sands, K.; Carvalho, M.J.; Spiller, O.B.; Portal, E.A.R.; Thomson, K.; Watkins, W.J.; Mathias, J.; Dyer, C.; Akpulu, C.; Andrews, R.; et al. Characterisation of Staphylococci Species from Neonatal Blood Cultures in Low- and Middle-Income Countries. BMC Infect. Dis. 2022, 22, 593. [Google Scholar] [CrossRef]
  32. Okomo, U.; Akpalu, E.N.K.; Le Doare, K.; Roca, A.; Cousens, S.; Jarde, A.; Sharland, M.; Kampmann, B.; Lawn, J.E. Aetiology of Invasive Bacterial Infection and Antimicrobial Resistance in Neonates in Sub-Saharan Africa: A Systematic Review and Meta-Analysis in Line with the STROBE-NI Reporting Guidelines. Lancet. Infect. Dis. 2019, 19, 1219–1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Medugu, N.; Iregbu, K.; Tam, P.Y.I.; Obaro, S. Aetiology of Neonatal Sepsis in Nigeria, and Relevance of Group b Streptococcus: A Systematic Review. PLoS ONE 2018, 13, e0200350. [Google Scholar] [CrossRef] [PubMed]
  34. Byrd, A.L.; Belkaid, Y.; Segre, J.A. The Human Skin Microbiome. Nat. Rev. Microbiol. 2018, 16. [Google Scholar] [CrossRef] [PubMed]
  35. Boxberger, M.; Cenizo, V.; Cassir, N.; La Scola, B. Challenges in Exploring and Manipulating the Human Skin Microbiome. Microbiome 2021, 9, 125. [Google Scholar] [CrossRef]
  36. Flowers, L.; Grice, E.A. The Skin Microbiota: Balancing Risk and Reward. Cell Host Microbe 2020, 28, 190–200. [Google Scholar] [CrossRef]
  37. França, A.; Gaio, V.; Lopes, N.; Melo, L.D.R.L.D.R. Virulence Factors in Coagulase-Negative Staphylococci. Pathogens 2021, 10, 170. [Google Scholar] [CrossRef]
  38. Rasigade, J.P.; Raulin, O.; Picaud, J.C.; Tellini, C.; Bes, M.; Grando, J.; Saïd, M.B.; Claris, O.; Etienne, J.; Tigaud, S.; et al. Methicillin-Resistant Staphylococcus Capitis with Reduced Vancomycin Susceptibility Causes Late-Onset Sepsis in Intensive Care Neonates. PLoS ONE 2012, 7, e31548. [Google Scholar] [CrossRef] [Green Version]
  39. Ben Said, M.; Hays, S.; Bonfils, M.; Jourdes, E.; Rasigade, J.P.; Laurent, F.; Picaud, J.C. Late-Onset Sepsis Due to Staphylococcus Capitis “neonatalis” in Low-Birthweight Infants: A New Entity? J. Hosp. Infect. 2016, 94, 95–98. [Google Scholar] [CrossRef]
  40. Björkqvist, M.; Söderquist, B.; Törnqvist, E.; Sjöberg, L.; Fredlund, H.; Kühn, I.; Colque-Navarro, P.; Schollin, J. Phenotypic and Genotypic Characterisation of Blood Isolates of Coagulase-Negative Staphylococci in the Newborn. APMIS 2002, 110, 332–339. [Google Scholar] [CrossRef]
  41. Foka, A.; Chini, V.; Petinaki, E.; Kolonitsiou, F.; Anastassiou, E.D.; Dimitracopoulos, G.; Spiliopoulou, I. Clonality of Slime-Producing Methicillin-Resistant Coagulase-Negative Staphylococci Disseminated in the Neonatal Intensive Care Unit of a University Hospital. Clin. Microbiol. Infect. 2006, 12, 1230–1233. [Google Scholar] [CrossRef] [Green Version]
  42. Neumeister, B.; Kastner, S.; Conrad, S.; Klotz, G.; Bartmann, P. Characterization of Coagulase-Negative Staphylococci Causing Nosocomial Infections in Preterm Infants. Eur. J. Clin. Microbiol. Infect. Dis. 1995, 14, 856–863. [Google Scholar] [CrossRef] [PubMed]
  43. 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]
  44. Pereira, P.M.A.; Binatti, V.B.; Sued, B.P.R.; Ramos, J.N.; Peixoto, R.S.; Simões, C.; de Castro, E.A.; Duarte, J.L.M.B.; Vieira, V.Ô.V.; Hirata, R.; et al. Staphylococcus Haemolyticus Disseminated among Neonates with Bacteremia in a Neonatal Intensive Care Unit in Rio de Janeiro, Brazil. Diagn. Microbiol. Infect. Dis. 2014, 78, 85–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Butin, M.; Rasigade, J.P.; Martins-Simões, P.; Meugnier, H.; Lemriss, H.; Goering, R.V.; Kearns, A.; Deighton, M.A.; Denis, O.; Ibrahimi, A.; et al. Wide Geographical Dissemination of the Multiresistant Staphylococcus Capitis NRCS-A Clone in Neonatal Intensive-Care Units. Clin. Microbiol. Infect. 2016, 22, 46–52. [Google Scholar] [CrossRef] [Green Version]
  46. Penesyan, A.; Paulsen, I.T.; Kjelleberg, S.; Gillings, M.R. Three Faces of Biofilms: A Microbial Lifestyle, a Nascent Multicellular Organism, and an Incubator for Diversity. npj Biofilms Microbiomes 2021, 7, 80. [Google Scholar] [CrossRef]
  47. Heilmann, C.; Ziebuhr, W.; Becker, K. Are Coagulase-Negative Staphylococci Virulent? Clin. Microbiol. Infect. 2019, 25, 1071–1080. [Google Scholar] [CrossRef]
  48. Becker, K.; Heilmann, C.; Peters, G. Coagulase-Negative Staphylococci. Clin. Microbiol. Rev. 2014, 27, 870–926. [Google Scholar] [CrossRef] [Green Version]
  49. Becker, K.; Both, A.; Weißelberg, S.; Heilmann, C.; Rohde, H. Emergence of Coagulase-Negative Staphylococci. Expert Rev. Anti. Infect. Ther. 2020, 18, 349–366. [Google Scholar] [CrossRef]
  50. Klingenberg, C.; Aarag, E.; Rønnestad, A.; Sollid, J.E.; Abrahamsen, T.G.; Kjeldsen, G.; Flœgstad, T. Coagulase-Negative Staphylococcal Sepsis in Neonates: Association between Antibiotic Resistance, Biofilm Formation and the Host Inflammatory Response. Pediatr. Infect. Dis. J. 2005, 24, 817–822. [Google Scholar] [CrossRef]
  51. Oliveira, F.; Cerca, N. Antibiotic Resistance and Biofilm Formation Ability among Coagulase-Negative Staphylococci in Healthy Individuals from Portugal. J. Antibiot. 2013, 66, 739–741. [Google Scholar] [CrossRef] [Green Version]
  52. Cerca, N.; Martins, S.; Cerca, F.; Jefferson, K.K.; Pier, G.B.; Oliveira, R.; Azeredo, J. Comparative Assessment of Antibiotic Susceptibility of Coagulase-Negative Staphylococci in Biofilm versus Planktonic Culture as Assessed by Bacterial Enumeration or Rapid XTT Colorimetry. J. Antimicrob. Chemother. 2005, 56, 331–336. [Google Scholar] [CrossRef] [PubMed]
  53. França, A.; Carvalhais, V.; Vilanova, M.; Pier, G.B.; Cerca, N. Characterization of an in Vitro Fed-Batch Model to Obtain Cells Released from S. Epidermidis Biofilms. AMB Express 2016, 6, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. O’ Neill, J. Antimicrobial Resistance: Tackling a Crisis for the Health and Wealth of Nations; HM Government: London, UK, 2014.
  55. Production, A.P.; Meng, H.; Dai, Y.; Otto, M.; Li, M.; Liu, Q.; Liu, Q.; Meng, H.; Lv, H.; Liu, Y.; et al. Staphylococcus Epidermidis Contributes to Healthy Maturation of the Nasal Microbiome by Stimulating Article 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]
  56. Lai, Y.; Di Nardo, A.; Nakatsuji, T.; Leichtle, A.; Yang, Y.; Cogen, A.L.; Wu, Z.; Hooper, L.V.; Von Aulock, S.; Radek, K.A.; et al. Commensal Bacteria Regulate TLR3-Dependent Inflammation Following Skin Injury. Nat. Med. 2009, 15, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
  57. 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. [Google Scholar] [CrossRef] [Green Version]
  58. Bitschar, K.; Staudenmaier, L.; Klink, L.; Focken, J.; Sauer, B.; Fehrenbacher, B.; Herster, F.; Bittner, Z.; Bleul, L.; Schaller, M.; et al. Staphylococcus Aureus Skin Colonization Is Enhanced by the Interaction of Neutrophil Extracellular Traps with Keratinocytes. J. Invest. Dermatol. 2020, 140, 1054–1065.e4. [Google Scholar] [CrossRef]
  59. Yoon, H.S. Neonatal Innate Immunity and Toll-like Receptor. Korean J. Pediatr. 2010, 53, 985–988. [Google Scholar] [CrossRef]
  60. Kai-Larsen, Y.; Gudmundsson, G.H.; Agerberth, B. A Review of the Innate Immune Defence of the Human Foetus and Newborn, with the Emphasis on Antimicrobial Peptides. Acta Paediatr. 2014, 103, 1000–1008. [Google Scholar] [CrossRef]
  61. Tsafaras, G.P.; Ntontsi, P.; Xanthou, G. Advantages and Limitations of the Neonatal Immune System. Front. Pediatr. 2020, 8, 1–10. [Google Scholar] [CrossRef] [Green Version]
  62. Kumar, S.K.M.; Bhat, B.V. Distinct Mechanisms of the Newborn Innate Immunity. Immunol. Lett. 2016, 173, 42–54. [Google Scholar] [CrossRef]
  63. Kan, B.; Razzaghian, H.R.; Lavoie, P.M. An Immunological Perspective on Neonatal Sepsis. Trends Mol. Med. 2016, 22, 290–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. 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] [PubMed] [Green Version]
  65. Kollmann, T.R.; Kampmann, B.; Mazmanian, S.K.; Marchant, A.; Levy, O. Protecting the Newborn and Young Infant from Infectious Diseases: Lessons from Immune Ontogeny. Immunity 2017, 46, 350–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Brook, B.; Harbeson, D.; Ben-Othman, R.; Viemann, D.; Kollmann, T.R. Newborn Susceptibility to Infection vs. Disease Depends on Complex in Vivo Interactions of Host and Pathogen. Semin. Immunopathol. 2017, 39, 615–625. [Google Scholar] [CrossRef] [PubMed]
  67. Medzhitov, R.; Schneider, D.S.; Soares, M.P. Disease Tolerance as a Defense Strategy. Science 2012, 335, 936–941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Rocamora-Reverte, L.; Melzer, F.L.; Würzner, R.; Weinberger, B. The Complex Role of Regulatory T Cells in Immunity and Aging. Front. Immunol. 2021, 11, 616949. [Google Scholar] [CrossRef]
  69. 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]
  70. Capone, K.A.; Dowd, S.E.; Stamatas, G.N.; Nikolovski, J. Diversity of the Human Skin Microbiome Early in Life. J. Invest. Dermatol. 2011, 131, 2026–2032. [Google Scholar] [CrossRef] [Green Version]
  71. Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery Mode Shapes the Acquisition and Structure of the Initial Microbiota across Multiple Body Habitats in Newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [Green Version]
  72. 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. [Google Scholar] [CrossRef]
  73. 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] [PubMed]
  74. Gasparrini, A.J.; Wang, B.; Sun, X.; Kennedy, E.A.; Hernandez-Leyva, A.; Ndao, I.M.; Tarr, P.I.; Warner, B.B.; Dantas, G. Persistent Metagenomic Signatures of Early-Life Hospitalization and Antibiotic Treatment in the Infant Gut Microbiota and Resistome. Nat. Microbiol. 2019, 4, 2285–2297. [Google Scholar] [CrossRef] [PubMed]
  75. 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] [PubMed] [Green Version]
  76. Brooks, B.; Olm, M.R.; Firek, B.A.; Baker, R.; Thomas, B.C.; Morowitz, M.J.; Banfield, J.F. Strain-Resolved Analysis of Hospital Rooms and Infants Reveals Overlap between the Human and Room Microbiome. Nat. Commun. 2017, 8, 1814. [Google Scholar] [CrossRef] [Green Version]
  77. 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]
  78. Altuntas, E.G. Isolation, Identification and Characterization of Staphylococcus Epidermidis in Human Milk. LWT Food Sci. Technol. 2015, 60, 36–41. [Google Scholar] [CrossRef]
  79. 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]
  80. Jiménez, E.; Delgado, S.; Fernández, L.; García, N.; Albújar, M.; Gómez, A.; Rodríguez, J.M. Assessment of the Bacterial Diversity of Human Colostrum and Screening of Staphylococcal and Enterococcal Populations for Potential Virulence Factors. Res. Microbiol. 2008, 159, 595–601. [Google Scholar] [CrossRef]
  81. Jiménez, E.; Delgado, S.; Maldonado, A.; Arroyo, R.; Albújar, M.; García, N.; Jariod, M.; Fernández, L.; Gómez, A.; Rodríguez, J.M. Staphylococcus Epidermidis: A Differential Trait of the Fecal Microbiota of Breast-Fed Infants. BMC Microbiol. 2008, 8, 143. [Google Scholar] [CrossRef] [Green Version]
  82. Martín, V.; Maldonado-Barragán, A.; Moles, L.; Rodriguez-Baños, M.; Del Campo, R.; Fernández, L.; Rodríguez, J.M.; Jiménez, E. Sharing of Bacterial Strains between Breast Milk and Infant Feces. J. Hum. Lact. 2012, 28, 36–44. [Google Scholar] [CrossRef]
  83. Jiménez, E.; Marín, M.L.; Martín, R.; Odriozola, J.M.; Olivares, M.; Xaus, J.; Fernández, L.; Rodríguez, J.M. Is Meconium from Healthy Newborns Actually Sterile? Res. Microbiol. 2008, 159, 187–193. [Google Scholar] [CrossRef] [PubMed]
  84. Moles, L.; Gómez, M.; Heilig, H.; Bustos, G.; Fuentes, S.; de Vos, W.; Fernández, L.; Rodríguez, J.M.; Jiménez, E. Bacterial Diversity in Meconium of Preterm Neonates and Evolution of Their Fecal Microbiota during the First Month of Life. PLoS ONE 2013, 8, e66986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. 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] [PubMed]
  86. 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] [PubMed]
  87. Stoll, B.J.; Hansen, N.I.; Adams-Chapman, I.; Fanaroff, A.A.; Hintz, S.R.; Vohr, B.; Higgins, R.D. Neurodevelopmental and Growth Impairment among Extremely Low-Birth-Weight Infants with Neonatal Infection. JAMA 2004, 292, 2357–2365. [Google Scholar] [CrossRef] [Green Version]
  88. 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] [Green Version]
  89. Alshaikh, B.; Yusuf, K.; Sauve, R. Neurodevelopmental Outcomes of Very Low Birth Weight Infants with Neonatal Sepsis: Systematic Review and Meta-Analysis. J. Perinatol. 2013, 33, 558–564. [Google Scholar] [CrossRef] [Green Version]
  90. Liljedahl, M.; Bodin, L.; Schollin, J. Coagulase-Negative Staphylococcal Sepsis as a Predictor of Bronchopulmonary Dysplasia. Acta Paediatr. 2004, 93, 211–215. [Google Scholar] [CrossRef]
  91. Shah, D.K.; Doyle, L.W.; Anderson, P.J.; Bear, M.; Daley, A.J.; Hunt, R.W.; Inder, T.E. Adverse Neurodevelopment in Preterm Infants with Postnatal Sepsis or Necrotizing Enterocolitis Is Mediated by White Matter Abnormalities on Magnetic Resonance Imaging at Term. J. Pediatr. 2008, 153, 170–175. [Google Scholar] [CrossRef]
  92. Anderson-Berry, A.; Brinton, B.; Lyden, E.; Faix, R.G. Risk Factors Associated with Development of Persistent Coagulase-Negative Staphylococci Bacteremia in the Neonate and Associated Short-Term and Discharge Morbidities. Neonatology 2011, 99, 23–31. [Google Scholar] [CrossRef]
  93. Jean-Baptiste, N.; Benjamin, D.K.; Cohen-Wolkowiez, M.; Fowler, V.G.; Laughon, M.; Clark, R.H.; Smith, P.B. Coagulase-Negative Staphylococcal Infections in the Neonatal Intensive Care Unit. Infect. Control Hosp. Epidemiol. 2011, 32, 679–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zea-Vera, A.; Ochoa, T.J. Challenges in the Diagnosis and Management of Neonatal Sepsis. J. Trop. Pediatr. 2015, 61, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. el Manouni el Hassani, S.; Berkhout, D.J.C.; Niemarkt, H.J.; Mann, S.; de Boode, W.P.; Cossey, V.; Hulzebos, C.V.; van Kaam, A.H.; Kramer, B.W.; van Lingen, R.A.; et al. Risk Factors for Late-Onset Sepsis in Preterm Infants: A Multicenter Case-Control Study. Neonatology 2019, 116, 42–51. [Google Scholar] [CrossRef] [PubMed]
  96. Sobczak, A.; Klepacka, J.; Amrom, D.; Żak, I.; Kruczek, P.; Kwinta, P. Umbilical Catheters as Vectors for Generalized Bacterial Infection in Premature Infants Regardless of Antibiotic Use. J. Med. Microbiol. 2019, 68, 1306–1313. [Google Scholar] [CrossRef] [PubMed]
  97. Hira, V.; Sluijter, M.; Estevão, S.; Horst-Kreft, D.; Ott, A.; De Groot, R.; Hermans, P.W.M.; Kornelisse, R.F. Clinical and Molecular Epidemiologic Characteristics of Coagulase-Negative Staphylococcal Bloodstream Infections in Intensive Care Neonates. Pediatr. Infect. Dis. J. 2007, 26, 607–612. [Google Scholar] [CrossRef] [PubMed]
  98. Cortez, J.; Makker, K.; Kraemer, D.F.; Neu, J.; Sharma, R.; Hudak, M.L. Maternal Milk Feedings Reduce Sepsis, Necrotizing Enterocolitis and Improve Outcomes of Premature Infants. J. Perinatol. 2018, 38, 71–74. [Google Scholar] [CrossRef] [PubMed]
  99. 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] [Green Version]
  100. Muk, T.; Brunse, A.; Henriksen, N.L.; Aasmul-Olsen, K.; Nguyen, D.N. Glucose Supply and Glycolysis Inhibition Shape the Clinical Fate of Staphylococcus Epidermidis-Infected Preterm Newborns. JCI Insight 2022, 7, e157234. [Google Scholar] [CrossRef]
  101. 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]
  102. 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]
  103. Tolo, I.; Thomas, J.C.; Fischer, R.S.B.; Brown, E.L.; Gray, B.M.; Robinson, D.A. Do Staphylococcus Epidermidis Genetic Clusters Predict Isolation Sources? J. Clin. Microbiol. 2016, 54, 1711–1719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Soeorg, H.; Metsvaht, H.K.; Keränen, E.E.; Eelmäe, I.; Merila, M.; Ilmoja, M.L.; Metsvaht, T.; Lutsar, I. Genetic Relatedness of Staphylococcus Haemolyticus in Gut and Skin of Preterm Neonates and Breast Milk of Their Mothers. Pediatr. Infect. Dis. J. 2019, 38, 308–313. [Google Scholar] [CrossRef]
  105. Soeorg, H.; Metsvaht, T.; Eelmäe, I.; Merila, M.; Treumuth, S.; Huik, K.; Jürna-Ellam, M.; Ilmoja, M.L.; Lutsar, I. The Role of Breast Milk in the Colonization of Neonatal Gut and Skin with Coagulase-Negative Staphylococci. Pediatr. Res. 2017, 82, 759–767. [Google Scholar] [CrossRef] [PubMed]
  106. Butin, M.; Rasigade, J.P.; Subtil, F.; Martins-Simões, P.; Pralong, C.; Freydière, A.M.; Vandenesch, F.; Tigaud, S.; Picaud, J.C.; Laurent, F. Vancomycin Treatment Is a Risk Factor for Vancomycin-Nonsusceptible Staphylococcus Capitis Sepsis in Preterm Neonates. Clin. Microbiol. Infect. 2017, 23, 839–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Soeorg, H.; Huik, K.; Parm, Ü.; Ilmoja, M.L.; Metelskaja, N.; Metsvaht, T.; Lutsar, I. Genetic Relatedness of Coagulase-Negative Staphylococci from Gastrointestinal Tract and Blood of Preterm Neonates with Late-Onset Sepsis. Pediatr. Infect. Dis. J. 2013, 32, 389–393. [Google Scholar] [CrossRef] [Green Version]
  108. Yu, J.; Liu, Y.; Xu, Z. Hand Carriage, Antimicrobial Resistance and Molecular Characterisation of Methicillin-Resistant Coagulase-Negative Staphylococci Isolated from Gynaecological Surgical Staff. J. Obstet. Gynaecol. 2022, 1–6. [Google Scholar] [CrossRef]
  109. Otto, M. Staphylococcus Epidermidis – the “Accidental” Pathogen. Nat. Rev. Microbiol. 2009, 7, 555–567. [Google Scholar] [CrossRef] [Green Version]
  110. 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]
  111. 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]
  112. Wang, R.; Braughton, K.R.; Kretschmer, D.; Bach, T.H.L.; Queck, S.Y.; Li, M.; Kennedy, A.D.; Dorward, D.W.; Klebanoff, S.J.; Peschel, A.; et al. Identification of Novel Cytolytic Peptides as Key Virulence Determinants for Community-Associated MRSA. Nat. Med. 2007, 13, 1510–1514. [Google Scholar] [CrossRef]
  113. Kretschmer, D.; Gleske, A.-K.; Rautenberg, M.; Wang, R.; Köberle, M.; Bohn, E.; Schöneberg, T.; Rabiet, M.-J.; Boulay, F.; Klebanoff, S.J.; et al. Human Formyl Peptide Receptor 2 Senses Highly Pathogenic Staphylococcus Aureus. Cell Host Microbe 2010, 7, 463–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. 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] [PubMed] [Green Version]
  115. Queck, S.Y.; Khan, B.A.; Wang, R.; Bach, T.H.L.; Kretschmer, D.; Chen, L.; Kreiswirth, B.N.; Peschel, A.; DeLeo, F.R.; Otto, M. Mobile Genetic Element-Encoded Cytolysin Connects Virulence to Methicillin Resistance in MRSA. PLoS Pathog. 2009, 5, e1000533. [Google Scholar] [CrossRef] [PubMed]
  116. Lai, Y.; Villaruz, A.E.; Li, M.; Cha, D.J.; Sturdevant, D.E.; Otto, M. The Human Anionic Antimicrobial Peptide Dermcidin Induces Proteolytic Defence Mechanisms in Staphylococci. Mol. Microbiol. 2007, 63, 497–506. [Google Scholar] [CrossRef]
  117. Dubin, G.; Chmiel, D.; Mak, P.; Rakwalska, M.; Rzychon, M.; Dubin, A. Molecular Cloning and Biochemical Characterization of Proteases from Staphylococcus Epidermidis. Biol. Chem. 2001, 382, 1575–1582. [Google Scholar] [CrossRef]
  118. Longshaw, C.M.; Farrell, A.M.; Wright, J.D.; Holland, K.T. Identification of a Second Lipase Gene, GehD, in Staphylococcus Epidermidis: Comparison of Sequence with Those of Other Staphylococcal Lipases. Microbiology 2000, 146 (Pt 6), 1419–1427. [Google Scholar] [CrossRef] [Green Version]
  119. Simons, J.W.F.A.; Van Kampen, M.D.; Riel, S.; Götz, F.; Egmond, M.R.; Verheij, H.M. Cloning, Purification and Characterisation of the Lipase from Staphylococcus Epidermidis--Comparison of the Substrate Selectivity with Those of Other Microbial Lipases. Eur. J. Biochem. 1998, 253, 675–683. [Google Scholar] [CrossRef]
  120. Gabriela Bowden, M.; Visai, L.; Longshaw, C.M.; Holland, K.T.; Speziale, P.; Höök, M. Is the GehD Lipase from Staphylococcus Epidermidis a Collagen Binding Adhesin? J. Biol. Chem. 2002, 277, 43017–43023. [Google Scholar] [CrossRef] [Green Version]
  121. Chamberlain, N.R.; Brueggemann, S.A. Characterisation and Expression of Fatty Acid Modifying Enzyme Produced by Staphylococcus Epidermidis. J. Med. Microbiol. 1997, 46, 693–697. [Google Scholar] [CrossRef] [Green Version]
  122. Flemming, H.C.; Wuertz, S. Bacteria and Archaea on Earth and Their Abundance in Biofilms. Nat. Rev. Microbiol. 2019, 17, 247–260. [Google Scholar] [CrossRef]
  123. Bowler, P.; Murphy, C.; Wolcott, R. Biofilm Exacerbates Antibiotic Resistance: Is This a Current Oversight in Antimicrobial Stewardship? Antimicrob. Resist. Infect. Control 2020, 9, 162. [Google Scholar] [CrossRef] [PubMed]
  124. Scherr, T.D.; Heim, C.E.; Morrison, J.M.; Kielian, T. Hiding in Plain Sight: Interplay between Staphylococcal Biofilms and Host Immunity. Front. Immunol. 2014, 5, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hogan, D.; Kolter, R. Why Are Bacteria Refractory to Antimicrobials? Curr. Opin. Microbiol. 2002, 5, 472–477. [Google Scholar] [CrossRef] [PubMed]
  126. Cheung, G.Y.; Otto, M. Understanding the Significance of Staphylococcus Epidermidis Bacteremia in Babies and Children. Curr. Opin. Infect. Dis. 2010, 23, 208–216. [Google Scholar] [CrossRef]
  127. 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] [PubMed]
  128. Otto, M. Staphylococcal Biofilms. Microbiol. Spectr. 2018, 6, 27. [Google Scholar] [CrossRef]
  129. Davey, M.E.; O’toole, G.A. Microbial Biofilms: From Ecology to Molecular Genetics. Microbiol. Mol. Biol. Rev. 2000, 64, 847–867. [Google Scholar] [CrossRef] [Green Version]
  130. Heilmann, C. Adhesion Mechanisms of Staphylococci. Adv. Exp. Med. Biol. 2011, 7, 105–123. [Google Scholar]
  131. Patti, J.M.; Allen, B.L.; McGavin, M.J.; Hook, M. MSCRAMM-Medicated Adherence of Microorganisms to Host Tissues. Annu. Rev. Microbiol. 1994, 48, 585–617. [Google Scholar] [CrossRef]
  132. Gill, S.R.; Fouts, D.E.; Archer, G.L.; Mongodin, E.F.; DeBoy, R.T.; Ravel, J.; Paulsen, I.T.; Kolonay, J.F.; Brinkac, L.; Beanan, M.; et al. Insights on Evolution of Virulence and Resistance from the Complete Genome Analysis of an Early Methicillin-Resistant Staphylococcus Aureus Strain and a Biofilm-Producing Methicillin-Resistant Staphylococcus Epidermidis Strain. J. Bacteriol. 2005, 187, 2426–2438. [Google Scholar] [CrossRef] [Green Version]
  133. McCrea, K.W.; Hartford, O.; Davis, S.; Eidhin, D.N.; Lina, G.; Speziale, P.; Foster, T.J.; Höök, M. The Serine-Aspartate Repeat (Sdr) Protein Family in Staphylococcus Epidermidis. Microbiology 2000, 146, 1535–1546. [Google Scholar] [CrossRef] [PubMed]
  134. Heilmann, C.; Hussain, M.; Peters, G.; Götz, F. Evidence for Autolysin-Mediated Primary Attachment of Staphylococcus Epidermidis to a Polystyrene Surface. Mol. Microbiol. 1997, 24, 1013–1024. [Google Scholar] [CrossRef] [PubMed]
  135. 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] [PubMed]
  136. Weidenmaier, C.; Kokai-Kun, J.F.; Kristian, S.A.; Chanturiya, T.; Kalbacher, H.; Gross, M.; Nicholson, G.; Neumeister, B.; Mond, J.J.; Peschel, A. Role of Teichoic Acids in Staphylococcus Aureus Nasal Colonization, a Major Risk Factor in Nosocomial Infections. Nat. Med. 2004, 10, 243–245. [Google Scholar] [CrossRef] [PubMed]
  137. Schilcher, K.; Horswill, A.R. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol. Mol. Biol. Rev. 2020, 84, e00026-19. [Google Scholar] [CrossRef]
  138. Schaeffer, C.R.; Hoang, T.-M.N.; Sudbeck, C.M.; Alawi, M.; Tolo, I.E.; Robinson, D.A.; Horswill, A.R.; Rohde, H.; Fey, P.D. Versatility of Biofilm Matrix Molecules in Staphylococcus Epidermidis Clinical Isolates and Importance of Polysaccharide Intercellular Adhesin Expression during High Shear Stress. mSphere 2016, 1, e00165-16. [Google Scholar] [CrossRef] [Green Version]
  139. 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 β-1,6-Linked Glucosaminoglycan: Purification and Structural Analysis. J. Bacteriol. 1996, 178, 175–183. [Google Scholar] [CrossRef] [Green Version]
  140. Cherifi, S.; Byl, B.; Deplano, A.; Nonhoff, C.; Denis, O.; Hallin, M. Comparative Epidemiology of Staphylococcus Epidermidis Isolates from Patients with Catheter-Related Bacteremia and from Healthy Volunteers. J. Clin. Microbiol. 2013, 51, 1541–1547. [Google Scholar] [CrossRef] [Green Version]
  141. Harris, L.G.; Murray, S.; Pascoe, B.; Bray, J.; Meric, G.; Magerios, L.; Wilkinson, T.S.; Jeeves, R.; Rohde, H.; Schwarz, S.; et al. Biofilm Morphotypes and Population Structure among Staphylococcus Epidermidis from Commensal and Clinical Samples. PLoS ONE 2016, 11, e0154510. [Google Scholar] [CrossRef]
  142. Ahmed, D.M.; Abel Messih, M.A.W.; Ibrahim, N.H.; Meabed, M.H.; Abdel-Salam, S.M. Frequency of IcaA and IcaD Determinants and Biofilm Formation among Coagulase-Negative Staphylococci Associated with Nasal Carriage in Neonatal Intensive Care Units. Germs 2019, 9, 61. [Google Scholar] [CrossRef]
  143. Sheikh, A.F.; Dezfuli, A.A.Z.; Navidifar, T.; Fard, S.S.; Dehdashtian, M. Association between Biofilm Formation, Structure and Antibiotic Resistance in Staphylococcus Epidermidis Isolated from Neonatal Septicemia in Southwest Iran. Infect. Drug Resist. 2019, 12, 1771–1782. [Google Scholar] [CrossRef] [Green Version]
  144. Martínez-Meléndez, A.; Morfín-Otero, R.; Villarreal-Treviño, L.; Camacho-Ortíz, A.; González-González, G.; Llaca-Díaz, J.; Rodríguez-Noriega, E.; Garza-González, E. Molecular Epidemiology of Coagulase-Negative Bloodstream Isolates: Detection of Staphylococcus Epidermidis ST2, ST7 and Linezolid-Resistant ST23. Brazilian J. Infect. Dis. 2016, 20, 419–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. 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]
  146. Hussain, M.; Herrmann, M.; Von Eiff, C.; Perdreau-Remington, F.; Peters, G. A 140-Kilodalton Extracellular Protein Is Essential for the Accumulation of Staphylococcus Epidermidis Strains on Surfaces. Infect. Immun. 1997, 65, 519–524. [Google Scholar] [CrossRef] [Green Version]
  147. 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]
  148. Paharik, A.E.; Kotasinska, M.; Both, A.; Hoang, T.M.N.; Büttner, H.; Roy, P.; Fey, P.D.; Horswill, A.R.; Rohde, H. The Metalloprotease SepA Governs Processing of Accumulation-Associated Protein and Shapes Intercellular Adhesive Surface Properties in Staphylococcus Epidermidis. Mol. Microbiol. 2017, 103, 860–874. [Google Scholar] [CrossRef] [Green Version]
  149. 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]
  150. Sugimoto, S.; Iwamoto, T.; Takada, K.; Okuda, K.I.; Tajima, A.; Iwase, T.; Mizunoe, Y. Staphylococcus Epidermidis Esp Degrades Specific Proteins Associatedwith Staphylococcus Aureus Biofilm Formation and Host-Pathogen Interaction. J. Bacteriol. 2013, 195, 1645–1655. [Google Scholar] [CrossRef] [Green Version]
  151. 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. 2012, 109, 1281–1286. [Google Scholar] [CrossRef] [Green Version]
  152. Balaban, N.Q.; Helaine, S.; Lewis, K.; Ackermann, M.; Aldridge, B.; Andersson, D.I.; Brynildsen, M.P.; Bumann, D.; Camilli, A.; Collins, J.J.; et al. Definitions and Guidelines for Research on Antibiotic Persistence. Nat. Rev. Microbiol. 2019, 17, 441–448. [Google Scholar] [CrossRef] [Green Version]
  153. Ceri, H.; Olson, M.E.; Stremick, C.; Read, R.R.; Morck, D.; Buret, A. The Calgary Biofilm Device: New Technology for Rapid Determination of Antibiotic Susceptibilities of Bacterial Biofilms. J. Clin. Microbiol. 1999, 37, 1771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Stewart, P.S. Mechanisms of Antibiotic Resistance in Bacterial Biofilms. Int. J. Med. Microbiol. 2002, 292, 107–113. [Google Scholar] [CrossRef] [PubMed]
  155. Stewart, P.S.; Franklin, M.J. Physiological Heterogeneity in Biofilms. Nat. Rev. Microbiol. 2008, 6, 199–210. [Google Scholar] [CrossRef] [PubMed]
  156. Rani, S.A.; Pitts, B.; Beyenal, H.; Veluchamy, R.A.; Lewandowski, Z.; Davison, W.M.; Buckingham-Meyer, K.; Stewart, P.S. Spatial Patterns of DNA Replication, Protein Synthesis, and Oxygen Concentration within Bacterial Biofilms Reveal Diverse Physiological States. J. Bacteriol. 2007, 189, 4223–4233. [Google Scholar] [CrossRef] [Green Version]
  157. García-Betancur, J.C.; Lopez, D. Cell Heterogeneity in Staphylococcal Communities. J. Mol. Biol. 2019, 431, 4699–4711. [Google Scholar] [CrossRef]
  158. Kranjec, C.; Angeles, D.M.; Mårli, M.T.; Fernández, L.; García, P.; Kjos, M.; Diep, D.B. Staphylococcal Biofilms: Challenges and Novel Therapeutic Perspectives. Antibiotics 2021, 10, 131. [Google Scholar] [CrossRef]
  159. Dunne, W.M.; Mason, E.O.; Kaplan, S.L. Diffusion of Rifampin and Vancomycin through a Staphylococcus Epidermidis Biofilm. Antimicrob. Agents Chemother. 1993, 37, 2522–2526. [Google Scholar] [CrossRef] [Green Version]
  160. Singh, R.; Ray, P.; Das, A.; Sharma, M. Penetration of Antibiotics through Staphylococcus Aureus and Staphylococcus Epidermidis Biofilms. J. Antimicrob. Chemother. 2010, 65, 1955–1958. [Google Scholar] [CrossRef] [Green Version]
  161. Gebreyohannes, G.; Nyerere, A.; Bii, C.; Sbhatu, D.B. Challenges of Intervention, Treatment, and Antibiotic Resistance of Biofilm-Forming Microorganisms. Heliyon 2019, 5, e02192. [Google Scholar] [CrossRef] [Green Version]
  162. Fluit, A.C.; Visser, M.R.; Schmitz, F. Molecular Detection of Antimicrobial Resistance. Clin. Microbiol. Rev. 2001, 14, 836–871. [Google Scholar] [CrossRef] [Green Version]
  163. Van Acker, H.; Van Dijck, P.; Coenye, T. Molecular Mechanisms of Antimicrobial Tolerance and Resistance in Bacterial and Fungal Biofilms. Trends Microbiol. 2014, 22, 326–333. [Google Scholar] [CrossRef] [PubMed]
  164. Olsen, J.E.; Christensen, H.; Aarestrup, F.M. Diversity and Evolution of BlaZ from Staphylococcus Aureus and Coagulase-Negative Staphylococci. J. Antimicrob. Chemother. 2006, 57, 450–460. [Google Scholar] [CrossRef]
  165. Vega, N.M.; Gore, J. Collective Antibiotic Resistance: Mechanisms and Implications. Curr. Opin. Microbiol. 2014, 21, 28–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Mahmoudi, H.; Pourhajibagher, M.; Chiniforush, N.; Soltanian, A.R.; Alikhani, M.Y.; Bahador, A. Biofilm Formation and Antibiotic Resistance in Meticillin-Resistant and Meticillin-Sensitive Staphylococcus Aureus Isolated from Burns. J. Wound Care 2019, 28, 66–73. [Google Scholar] [CrossRef]
  167. Qin, Z.; Yang, X.; Yang, L.; Jiang, J.; Ou, Y.; Molin, S.; Qu, D. Formation and Properties of in Vitro Biofilms of Ica-Negative Staphylococcus Epidermidis Clinical Isolates. J. Med. Microbiol. 2007, 56, 83–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Cafiso, V.; Bertuccio, T.; Santagati, M.; Campanile, F.; Amicosante, G.; Perilli, M.G.; Selan, L.; Artini, M.; Nicoletti, G.; Stefani, S. Presence of the Ica Operon in Clinical Isolates of Staphylococcus Epidermidis and Its Role in Biofilm Production. Clin. Microbiol. Infect. 2004, 10, 1081–1088. [Google Scholar] [CrossRef] [Green Version]
  169. Rachid, S.; Ohlsen, K.; Witte, W.; Hacker, J.; Ziebuhr, W. Effect of Subinhibitory Antibiotic Concentrations on Polysaccharide Intercellular Adhesin Expression in Biofilm-Forming Staphylococcus Epidermidis. Antimicrob. Agents Chemother. 2000, 44, 3357–3363. [Google Scholar] [CrossRef] [Green Version]
  170. Águila-Arcos, S.; Álvarez-Rodríguez, I.; Garaiyurrebaso, O.; Garbisu, C.; Grohmann, E.; Alkorta, I. Biofilm-Forming Clinical Staphylococcus Isolates Harbor Horizontal Transfer and Antibiotic Resistance Genes. Front. Microbiol. 2017, 8, 2018. [Google Scholar] [CrossRef]
  171. Lerminiaux, N.A.; Cameron, A.D.S. Horizontal Transfer of Antibiotic Resistance Genes in Clinical Environments. Can. J. Microbiol. 2019, 65, 34–44. [Google Scholar] [CrossRef]
  172. Flemming, H.-C.; Wingender, J.; Szewzyk, U.; Steinberg, P.; Rice, S.A.; Kjelleberg, S. Biofilms: An Emergent Form of Bacterial Life. Nat. Rev. Microbiol. 2016, 14, 563–575. [Google Scholar] [CrossRef]
  173. Woodford, N.; Ellington, M.J. The Emergence of Antibiotic Resistance by Mutation. Clin. Microbiol. Infect. 2007, 13, 5–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Ryder, V.J.; Chopra, I.; O’Neill, A.J. Increased Mutability of Staphylococci in Biofilms as a Consequence of Oxidative Stress. PLoS ONE 2012, 7, e47695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. 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] [Green Version]
  176. Cerca, F.; França, A.; Perez-Cabezas, B.; Carvalhais, V.; Ribeiro, A.; Azeredo, J.; Pier, G.; Cerca, N.; Vilanova, M. Dormant Bacteria within Staphylococcus Epidermidis Biofilms Have Low Inflammatory Properties and Maintain Tolerance to Vancomycin and Penicillin after Entering Planktonic Growth. J. Med. Microbiol. 2014, 63, 1274–1283. [Google Scholar] [CrossRef] [Green Version]
  177. Cerca, F.; Andrade, F.; Franca, A.; Andrade, E.B.; Ribeiro, A.; Almeida, A.A.; Cerca, N.; Pier, G.; Azeredo, J.; Vilanova, M. Staphylococcus Epidermidis Biofilms with Higher Proportions of Dormant Bacteria Induce a Lower Activation of Murine Macrophages. J. Med. Microbiol. 2011, 60, 1717–1724. [Google Scholar] [CrossRef] [Green Version]
  178. 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]
  179. Yao, Y.; Vuong, C.; Kocianova, S.; Villaruz, A.E.; Lai, Y.; Sturdavant, 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]
  180. Dengler Haunreiter, V.; Boumasmoud, M.; Häffner, N.; Wipfli, D.; Leimer, N.; Rachmühl, C.; Kühnert, D.; Achermann, Y.; Zbinden, R.; Benussi, S.; et al. In-Host Evolution of Staphylococcus Epidermidis in a Pacemaker-Associated Endocarditis Resulting in Increased Antibiotic Tolerance. Nat. Commun. 2019, 10, 1149. [Google Scholar] [CrossRef] [Green Version]
  181. Vuong, C.; Kocianova, S.; Voyich, J.M.; Yao, Y.; Fischer, E.R.; DeLeo, F.R.; Otto, M. A Crucial Role for Exopolysaccharide Modification in Bacterial Biofilm Formation, Immune Evasion, and Virulence. J. Biol. Chem. 2004, 279, 54881–54886. [Google Scholar] [CrossRef] [Green Version]
  182. Cerca, N.; Jefferson, K.K.; Oliveira, R.; Pier, G.B.; Azeredo, J. Comparative Antibody-Mediated Phagocytosis of Staphylococcus Epidermidis Cells Grown in a Biofilm or in the Planktonic State. Infect. Immun. 2006, 74, 4849–4855. [Google Scholar] [CrossRef] [Green Version]
  183. 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]
  184. Rupp, M.E.; Ulphani, J.S.; Fey, P.D.; Bartscht, K.; Mack, D. Characterization of the Importance of Polysaccharide Intercellular Adhesin/Hemagglutinin of Staphylococcus Epidermidis in the Pathogenesis of Biomaterial-Based Infection in a Mouse Foreign Body Infection Model. Infect. Immun. 1999, 67, 2627–2632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. 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]
  186. Rupp, M.E.; Ulphani, J.S.; Fey, P.D.; Mack, D. Characterization of Staphylococcus Epidermidis Polysaccharide Intercellular Adhesin/Hemagglutinin in the Pathogenesis of Intravascular Catheter-Associated Infection in a Rat Model. Infect. Immun. 1999, 67, 2656–2659. [Google Scholar] [CrossRef] [Green Version]
  187. 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–F145. [Google Scholar] [CrossRef] [PubMed]
  188. Fredheim, E.G.A.; Granslo, H.N.; Flægstad, T.; Figenschau, Y.; Rohde, H.; Sadovskaya, I.; Mollnes, T.E.; Klingenberg, C.; Aarag Fredheim, E.G.; Granslo, H.N.; et al. Staphylococcus Epidermidis Polysaccharide Intercellular Adhesin Activates Complement. FEMS Immunol. Med. Microbiol. 2011, 63, 269–280. [Google Scholar] [CrossRef]
  189. Hogan, S.; Stevens, N.T.; Humphreys, H.; O’Gara, J.P.; O’Neill, E. Current and Future Approaches to the Prevention and Treatment of Staphylococcal Medical Device-Related Infections. Curr. Pharm. Des. 2014, 21, 100–113. [Google Scholar] [CrossRef]
  190. Zandri, G.; Pasquaroli, S.; Vignaroli, C.; Talevi, S.; Manso, E.; Donelli, G.; Biavasco, F. Detection of Viable but Non-Culturable Staphylococci in Biofilms from Central Venous Catheters Negative on Standard Microbiological Assays. Clin. Microbiol. Infect. 2012, 18, E259–E261. [Google Scholar] [CrossRef] [Green Version]
  191. Ferrer, R.; Martin-Loeches, I.; Phillips, G.; Osborn, T.M.; Townsend, S.; Dellinger, R.P.; Artigas, A.; Schorr, C.; Levy, M.M. Empiric Antibiotic Treatment Reduces Mortality in Severe Sepsis and Septic Shock from the First Hour: Results from a Guideline-Based Performance Improvement Program. Crit. Care Med. 2014, 42, 1749–1755. [Google Scholar] [CrossRef] [Green Version]
  192. 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]
  193. Schelonka, R.L.; Chai, M.K.; Yoder, B.A.; Hensley, D.; Brockett, R.M.; Ascher, D.P. Volume of Blood Required to Detect Common Neonatal Pathogens. J. Pediatr. 1996, 129, 275–278. [Google Scholar] [CrossRef] [PubMed]
  194. Bury, G.; Leroux, S.; Borrego, C.L.; Leguen, C.G.; Mitanchez, D.; Gascoin, G.; Thollot, A.; Roué, J.M.; Carrault, G.; Pladys, P.; et al. Diagnosis of Neonatal Late-Onset Infection in Very Preterm Infant: Inter-Observer Agreement and International Classifications. Int. J. Environ. Res. Public Health 2021, 18, 882. [Google Scholar] [CrossRef] [PubMed]
  195. Wynn, J.L.; Wong, H.R.; Shanley, T.P.; Bizzarro, M.J.; Saiman, L.; Polin, R.A. Time for a Neonatal-Specific Consensus Definition for Sepsis. Pediatr. Crit. Care Med. 2014, 15, 523–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Lukacs, S.L.; Schrag, S.J. Clinical Sepsis in Neonates and Young Infants, United States, 1988-2006. J. Pediatr. 2012, 160. [Google Scholar] [CrossRef]
  197. Sofouli, G.A.; Kanellopoulou, A.; Vervenioti, A.; Dimitriou, G.; Gkentzi, D. Predictive Scores for Late-Onset Neonatal Sepsis as an Early Diagnostic and Antimicrobial Stewardship Tool: What Have We Done So Far? Antibiotics 2022, 11, 928. [Google Scholar] [CrossRef] [PubMed]
  198. Ohlin, A.; Björkqvist, M.; Montgomery, S.M.; Schollin, J. Clinical Signs and CRP Values Associated with Blood Culture Results in Neonates Evaluated for Suspected Sepsis. Acta Paediatr. Int. J. Paediatr. 2010, 99, 1635–1640. [Google Scholar] [CrossRef]
  199. Nelson, J.C.; Rizwan-uddin; Griffin, M.P.; Moorman, J.R. Probing the Order within Neonatal Heart Rate Variability. Pediatr. Res. 1998, 43, 823–831. [Google Scholar] [CrossRef] [Green Version]
  200. Griffin, M.P.; O’Shea, T.M.; Bissonette, E.A.; Harrell, F.E.; Lake, D.E.; Moorman, J.R. Abnormal Heart Rate Characteristics Preceding Neonatal Sepsis and Sepsis-like Illness. Pediatr. Res. 2003, 53, 920–926. [Google Scholar] [CrossRef]
  201. Healy, C.M.; Baker, C.J.; Palazzi, D.L.; Campbell, J.R.; Edwards, M.S. Distinguishing True Coagulase-Negative Staphylococcus Infections from Contaminants in the Neonatal Intensive Care Unit. J. Perinatol. 2013, 33, 52–58. [Google Scholar] [CrossRef]
  202. Sharma, D.; Farahbakhsh, N.; Shastri, S.; Sharma, P. Biomarkers for Diagnosis of Neonatal Sepsis: A Literature Review. J. Matern. Neonatal Med. 2018, 31, 1646–1659. [Google Scholar] [CrossRef]
  203. Ng, P.C.; Lam, H.S. Biomarkers for Late-Onset Neonatal Sepsis: Cytokines and Beyond. Clin. Perinatol. 2010, 37, 599–610. [Google Scholar] [CrossRef] [PubMed]
  204. Celik, I.H.; Hanna, M.; Canpolat, F.E. Mohan Pammi Diagnosis of Neonatal Sepsis: The Past, Present and Future. Pediatr. Res. 2021, 91, 337–350. [Google Scholar] [CrossRef]
  205. Gilfillan, M.; Bhandari, V. Biomarkers for the Diagnosis of Neonatal Sepsis and Necrotizing Enterocolitis: Clinical Practice Guidelines. Early Hum. Dev. 2017, 105, 25–33. [Google Scholar] [CrossRef] [PubMed]
  206. Chauhan, N.; Tiwari, S.; Jain, U. Potential Biomarkers for Effective Screening of Neonatal Sepsis Infections: An Overview. Microb. Pathog. 2017, 107, 234–242. [Google Scholar] [CrossRef]
  207. Rewa, O.; Muscedere, J.; Reynolds, S.; Jiang, X.; Heyland, D.K. Coagulase-Negative Staphylococcus, Catheter-Related, Bloodstream Infections and Their Association with Acute Phase Markers of Inflammation in the Intensive Care Unit: An Observational Study. Can. J. Infect. Dis. Med. Microbiol. 2012, 23, 204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Chiesa, C.; Natale, F.; Pascone, R.; Osborn, J.F.; Pacifico, L.; Bonci, E.; De Curtis, M. C Reactive Protein and Procalcitonin: Reference Intervals for Preterm and Term Newborns during the Early Neonatal Period. Clin. Chim. Acta 2011, 412, 1053–1059. [Google Scholar] [CrossRef]
  209. Dritsakou, K.; Liosis, G.; Gioni, M.; Glynou, E.; Avdeliodi, K.; Papagaroufalis, K. CRP Levels in Extremely Low Birth Weight (ELBW) Septic Infants. J. Matern. Fetal. Neonatal Med. 2015, 28, 237–239. [Google Scholar] [CrossRef]
  210. Mwesigye, P.; Rizwan, F.; Alassaf, N.; Khan, R. The Role and Validity of Diagnostic Biomarkers in Late-Onset Neonatal Sepsis. Cureus 2021, 13, e17065. [Google Scholar] [CrossRef]
  211. Brown, J.V.E.; Meader, N.; Wright, K.; Cleminson, J.; McGuire, W. Assessment of C-Reactive Protein Diagnostic Test Accuracy for Late-Onset Infection in Newborn Infants: A Systematic Review and Meta-Analysis. JAMA Pediatr. 2020, 174, 260–268. [Google Scholar] [CrossRef]
  212. Kurul, Ş.; Simons, S.H.P.; Ramakers, C.R.B.; De Rijke, Y.B.; Kornelisse, R.F.; Reiss, I.K.M.; Taal, H.R. Association of Inflammatory Biomarkers with Subsequent Clinical Course in Suspected Late Onset Sepsis in Preterm Neonates. Crit. Care 2021, 25, 12. [Google Scholar] [CrossRef]
  213. Beltempo, M.; Viel-Thériault, I.; Thibeault, R.; Julien, A.S.; Piedboeuf, B. C-Reactive Protein for Late-Onset Sepsis Diagnosis in Very Low Birth Weight Infants. BMC Pediatr. 2018, 18, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Hedegaard, S.S.; Wisborg, K.; Hvas, A.M. Diagnostic Utility of Biomarkers for Neonatal Sepsis--a Systematic Review. Infect. Dis. 2015, 47, 117–124. [Google Scholar] [CrossRef] [PubMed]
  215. Chiesa, C.; Panero, A.; Rossi, N.; Stegagno, M.; De Giusti, M.; Osborn, J.F.; Pacifico, L. Reliability of Procalcitonin Concentrations for the Diagnosis of Sepsis in Critically Ill Neonates. Clin. Infect. Dis. 1998, 26, 664–672. [Google Scholar] [CrossRef] [PubMed]
  216. Ruetsch, V.; Barreault, S.; Le Sache, N.; Tissères, P. Procalcitonin Is a Prognosis Biomarker in Very Preterm Neonates with Late Onset Sepsis: A Pilot Study. Eur. J. Pediatr. 2022, 181, 1459–1464. [Google Scholar] [CrossRef] [PubMed]
  217. Prentice, S.; Jallow, A.T.; Sinjanka, E.; Jallow, M.W.; Sise, E.A.; Kessler, N.J.; Wegmuller, R.; Cerami, C.; Prentice, A.M. Hepcidin Mediates Hypoferremia and Reduces the Growth Potential of Bacteria in the Immediate Post-Natal Period in Human Neonates. Sci. Rep. 2019, 9, 16596. [Google Scholar] [CrossRef] [Green Version]
  218. Sherbiny, H.S.; Mostafa, H.A.F.; Sherief, L.M.; Kamal, N.M.; El-Shal, A.S.; Halm, M.M.A.; Khan, H.Y.; Ali, A.S.A. Validity of Serum and Urinary Hepcidin as Biomarkers for Late-Onset in Premature Infants. Ther. Adv. Chronic Dis. 2022, 13, 204062232211225. [Google Scholar] [CrossRef]
  219. Wu, T.W.; Tabangin, M.; Kusano, R.; Ma, Y.; Ridsdale, R.; Akinbi, H. The Utility of Serum Hepcidin as a Biomarker for Late-Onset Neonatal Sepsis. J. Pediatr. 2013, 162, 67–71. [Google Scholar] [CrossRef]
  220. Griffin, M.P.; Lake, D.E.; O’Shea, T.M.; Moorman, J.R. Heart Rate Characteristics and Clinical Signs in Neonatal Sepsis. Pediatr. Res. 2007, 61, 222–227. [Google Scholar] [CrossRef] [Green Version]
  221. Makkar, M.; Pathak, R.; Garg, S.; Gupta, C.; Mahajan, N. Performance Evaluation of Hematologic Scoring System in Early Diagnosis of Neonatal Sepsis. J. Clin. Neonatol. 2013, 2, 25. [Google Scholar] [CrossRef] [Green Version]
  222. Narasimha, A.; Harendra Kumar, M.L. Significance of Hematological Scoring System (HSS) in Early Diagnosis of Neonatal Sepsis. Indian J. Hematol. Blood Transfus. 2011, 27, 14–17. [Google Scholar] [CrossRef] [Green Version]
  223. Eichberger, J.; Resch, E.; Resch, B. Diagnosis of Neonatal Sepsis: The Role of Inflammatory Markers. Front. Pediatr. 2022, 10, 281. [Google Scholar] [CrossRef] [PubMed]
  224. Pammi, M.; Flores, A.; Versalovic, J.; Leeflang, M.M.G. Molecular Assays for the Diagnosis of Sepsis in Neonates. Cochrane database Syst. Rev. 2017, 2, CD011926. [Google Scholar] [CrossRef] [PubMed]
  225. Kothari, A.; Morgan, M.; Haake, D.A. Emerging Technologies for Rapid Identification of Bloodstream Pathogens. Clin. Infect. Dis. 2014, 59, 272–278. [Google Scholar] [CrossRef] [PubMed]
  226. Van den Brand, M.; Peters, R.P.H.; Catsburg, A.; Rubenjan, A.; Broeke, F.J.; Van den Dungen, F.A.M.; Van Weissenbruch, M.M.; Van Furth, A.M.; Kõressaar, T.; Remm, M.; et al. Development of a Multiplex Real-Time PCR Assay for the Rapid Diagnosis of Neonatal Late Onset Sepsis. J. Microbiol. Methods 2014, 106, 8–15. [Google Scholar] [CrossRef] [PubMed]
  227. Sinha, M.; Jupe, J.; Mack, H.; Coleman, T.P.; Lawrence, S.M.; Fraley, S.I. Emerging Technologies for Molecular Diagnosis of Sepsis. Clin. Microbiol. Rev. 2018, 31, e00089-17. [Google Scholar] [CrossRef] [Green Version]
  228. Straub, J.; Paula, H.; Mayr, M.; Kasper, D.; Assadian, O.; Berger, A.; Rittenschober-Böhm, J. Diagnostic Accuracy of the ROCHE Septifast PCR System for the Rapid Detection of Blood Pathogens in Neonatal Sepsis—A Prospective Clinical Trial. PLoS ONE 2017, 12, e0187688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Tam, P.Y.I.; Hernandez-Alvarado, N.; Schleiss, M.R.; Hassan-Hanga, F.; Onuchukwu, C.; Umoru, D.; Obaro, S.K. Molecular Detection of Streptococcus Pneumoniae on Dried Blood Spots from Febrile Nigerian Children Compared to Culture. PLoS ONE 2016, 11, e0152253. [Google Scholar] [CrossRef] [Green Version]
  230. Liu, C.L.; Ai, H.W.; Wang, W.P.; Chen, L.; Hu, H.B.; Ye, T.; Zhu, X.H.; Wang, F.; Liao, Y.L.; Wang, Y.; et al. Comparison of 16S RRNA Gene PCR and Blood Culture for Diagnosis of Neonatal Sepsis. Arch. Pediatr. 2014, 21, 162–169. [Google Scholar] [CrossRef]
  231. Reier-Nilsen, T.; Farstad, T.; Nakstad, B.; Lauvrak, V.; Steinbakk, M. Comparison of Broad Range 16S RDNA PCR and Conventional Blood Culture for Diagnosis of Sepsis in the Newborn: A Case Control Study. BMC Pediatr. 2009, 9, 5. [Google Scholar] [CrossRef] [Green Version]
  232. Jordan, J.A.; Durso, M.B.; Butchko, A.R.; Jones, J.G.; Brozanski, B.S. Evaluating the Near-Term Infant for Early Onset Sepsis: Progress and Challenges to Consider with 16S RDNA Polymerase Chain Reaction Testing. J. Mol. Diagn. 2006, 8, 357–363. [Google Scholar] [CrossRef] [Green Version]
  233. Mancini, N.; Carletti, S.; Ghidoli, N.; Cichero, P.; Burioni, R.; Clementi, M. The Era of Molecular and Other Non-Culture-Based Methods in Diagnosis of Sepsis. Clin. Microbiol. Rev. 2010, 23, 235–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Luethy, P.M.; Johnson, J.K. The Use of Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry (MALDI-TOF MS) for the Identification of Pathogens Causing Sepsis. J. Appl. Lab. Med. 2019, 3, 675–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Schubert, S.; Kostrzewa, M. MALDI-TOF MS in the Microbiology Laboratory: Current Trends. Curr. Issues Mol. Biol. 2017, 23, 17–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Tschudin-Sutter, S.; Frei, R.; Dangel, M.; Jakob, M.; Balmelli, C.; Schaefer, D.J.; Weisser, M.; Elzi, L.; Battegay, M.; Widmer, A.F. Validation of a Treatment Algorithm for Orthopaedic Implant-Related Infections with Device-Retention-Results from a Prospective Observational Cohort Study. Clin. Microbiol. Infect. 2016, 22, 457.e1–457.e9. [Google Scholar] [CrossRef] [PubMed]
  237. Hou, T.Y.; Chiang-Ni, C.; Teng, S.H. Current Status of MALDI-TOF Mass Spectrometry in Clinical Microbiology. J. Food Drug Anal. 2019, 27, 404–414. [Google Scholar] [CrossRef] [PubMed]
  238. Ferreira, L.; Sánchez-Juanes, F.; Muñoz-Bellido, J.L.; González-Buitrago, J.M. Rapid Method for Direct Identification of Bacteria in Urine and Blood Culture Samples by Matrix-Assisted Laser Desorption Ionization Time-of-Flight Mass Spectrometry: Intact Cell vs. Extraction Method. Clin. Microbiol. Infect. 2011, 17, 1007–1012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Prod’Hom, G.; Bizzini, A.; Durussel, C.; Bille, J.; Greub, G. Matrix-Assisted Laser Desorption Ionization-Time of Flight Mass Spectrometry for Direct Bacterial Identification from Positive Blood Culture Pellets. J. Clin. Microbiol. 2010, 48, 1481–1483. [Google Scholar] [CrossRef] [Green Version]
  240. Verroken, A.; Defourny, L.; Le Polain De Waroux, O.; Belkhir, L.; Laterre, P.F.; Delmée, M.; Glupczynski, Y. Clinical Impact of MALDI-TOF MS Identification and Rapid Susceptibility Testing on Adequate Antimicrobial Treatment in Sepsis with Positive Blood Cultures. PLoS ONE 2016, 11, e0156299. [Google Scholar] [CrossRef] [Green Version]
  241. Hrabák, J.; Chudác ková, E.; Walková, R. Matrix-Assisted Laser Desorption Ionization-Time of Flight (Maldi-Tof) Mass Spectrometry for Detection of Antibiotic Resistance Mechanisms: From Research to Routine Diagnosis. Clin. Microbiol. Rev. 2013, 26, 103–114. [Google Scholar] [CrossRef] [Green Version]
  242. Ruiz-Aragón, J.; Ballestero-Téllez, M.; Gutiérrez-Gutiérrez, B.; de Cueto, M.; Rodríguez-Baño, J.; Pascual, Á. Direct Bacterial Identification from Positive Blood Cultures Using Matrix-Assisted Laser Desorption/Ionization Time-of-Flight (MALDI-TOF) Mass Spectrometry: A Systematic Review and Meta-Analysis. Enfermedades Infecc. y Microbiol. Clin. (Engl. ed.) 2018, 36, 484–492. [Google Scholar] [CrossRef]
  243. Hamilton, F.; Evans, R.; MacGowan, A. The Value of MALDI-TOF Failure to Provide an Identification of Staphylococcal Species Direct from Blood Cultures and Rule out Staphylococcus Aureus Bacteraemia: A Post-Hoc Analysis of the RAPIDO Trial. Access Microbiol. 2020, 3, 000192. [Google Scholar] [CrossRef] [PubMed]
  244. Fink, T.; Wolf, A.; Maurer, F.; Albrecht, F.W.; Heim, N.; Wolf, B.; Hauschild, A.C.; Bödeker, B.; Baumbach, J.I.; Volk, T.; et al. Volatile Organic Compounds during Inflammation and Sepsis in Rats: A Potential Breath Test Using Ion-Mobility Spectrometry. Anesthesiology 2015, 122, 117–126. [Google Scholar] [CrossRef] [PubMed]
  245. Guamán, A.V.; Carreras, A.; Calvo, D.; Agudo, I.; Navajas, D.; Pardo, A.; Marco, S.; Farré, R. Rapid Detection of Sepsis in Rats through Volatile Organic Compounds in Breath. J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci. 2012, 881–882, 76–82. [Google Scholar] [CrossRef] [PubMed]
  246. Oeschger, T.; McCloskey, D.; Kopparthy, V.; Singh, A.; Erickson, D. Point of Care Technologies for Sepsis Diagnosis and Treatment. Lab Chip 2019, 19, 728–737. [Google Scholar] [CrossRef] [PubMed]
  247. 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]
  248. 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. 2022, 91, 637–645. [Google Scholar] [CrossRef]
  249. Raymond, S.L.; López, M.C.; Baker, H.V.; Larson, S.D.; Efron, P.A.; Sweeney, T.E.; Khatri, P.; Moldawer, L.L.; Wynn, J.L. Unique Transcriptomic Response to Sepsis Is Observed among Patients of Different Age Groups. PLoS ONE 2017, 12, e0184159. [Google Scholar] [CrossRef] [Green Version]
  250. Bai, Y.; Zhao, N.; Zhang, Z.; Jia, Y.; Zhang, G.; Dong, G. Identification and Validation of a Novel Four-Gene Diagnostic Model for Neonatal Early-Onset Sepsis with Bacterial Infection. Eur. J. Pediatr. 2022. online ahead of print. [Google Scholar] [CrossRef]
  251. Miao, H.; Chen, S.; Ding, R. Evaluation of the Molecular Mechanisms of Sepsis Using Proteomics. Front. Immunol. 2021, 12, 733537. [Google Scholar] [CrossRef]
  252. Stewart, C.J.; Nelson, A.; Treumann, A.; Skeath, T.; Cummings, S.P.; Embleton, N.D.; Berrington, J.E. Metabolomic and Proteomic Analysis of Serum from Preterm Infants with Necrotising Entercolitis and Late-Onset Sepsis. Pediatr. Res. 2016, 79, 425–431. [Google Scholar] [CrossRef] [Green Version]
  253. Pilar-Orive, F.J.; Astigarraga, I.; Azkargorta, M.; Elortza, F.; Garcia-Obregon, S. A Three-Protein Panel to Support the Diagnosis of Sepsis in Children. J. Clin. Med. 2022, 11, 1563. [Google Scholar] [CrossRef] [PubMed]
  254. Garcia-Obregon, S.; Azkargorta, M.; Seijas, I.; Pilar-Orive, J.; Borrego, F.; Elortza, F.; Boyano, M.D.; Astigarraga, I. Identification of a Panel of Serum Protein Markers in Early Stage of Sepsis and Its Validation in a Cohort of Patients. J. Microbiol. Immunol. Infect. 2018, 51, 465–472. [Google Scholar] [CrossRef] [PubMed]
  255. Ludwig, K.R.; Hummon, A.B. Mass Spectrometry for the Discovery of Biomarkers of Sepsis. Mol. Biosyst. 2017, 13, 648–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Bjerkhaug, A.U.; Granslo, H.N.; Klingenberg, C. Metabolic Responses in Neonatal Sepsis-A Systematic Review of Human Metabolomic Studies. Acta Paediatr. 2021, 110, 2316–2325. [Google Scholar] [CrossRef]
  257. Tong, P.; Huang, F.R.; Xu, J.; Wu, Z.Q.; Hu, X.; Ling, M.; Wang, D.; Wu, B.F.; Yang, D.J.; Zhang, A.M. Metabolomic Changes of Neonatal Sepsis: An Exploratory Clinical Study. Zhongguo Dang Dai Er Ke Za Zhi 2022, 24, 675–680. [Google Scholar] [CrossRef] [PubMed]
  258. Mardegan, V.; Giordano, G.; Stocchero, M.; Pirillo, P.; Poloniato, G.; Donadel, E.; Salvadori, S.; Giaquinto, C.; Priante, E.; Baraldi, E. Untargeted and Targeted Metabolomic Profiling of Preterm Newborns with EarlyOnset Sepsis: A Case-Control Study. Metabolites 2021, 11, 115. [Google Scholar] [CrossRef] [PubMed]
  259. Abbas, M.; El-Manzalawy, Y. Machine Learning Based Refined Differential Gene Expression Analysis of Pediatric Sepsis. BMC Med. Genomics 2020, 13. [Google Scholar] [CrossRef]
  260. Wong, H.R.; Caldwell, J.T.; Cvijanovich, N.Z.; Weiss, S.L.; Fitzgerald, J.C.; Bigham, M.T.; Jain, P.N.; Schwarz, A.; Lutfi, R.; Nowak, J.; et al. Prospective Clinical Testing and Experimental Validation of the Pediatric Sepsis Biomarker Risk Model. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef]
  261. Mangioni, D.; Peri, A.M.; Rossolini, G.M.; Viaggi, B.; Perno, C.F.; Gori, A.; Bandera, A. Toward Rapid Sepsis Diagnosis and Patient Stratification: What’s New From Microbiology and Omics Science. J. Infect. Dis. 2020, 221, 1039–1047. [Google Scholar] [CrossRef]
  262. Lu, H.; Wen, D.; Wang, X.; Gan, L.; Du, J.; Sun, J.; Zeng, L.; Jiang, J.; Zhang, A. Host Genetic Variants in Sepsis Risk: A Field Synopsis and Meta-Analysis. Crit. Care 2019, 23, 26. [Google Scholar] [CrossRef] [Green Version]
  263. Wong, H.R.; Reeder, R.W.; Banks, R.; Berg, R.A.; Meert, K.L.; Hall, M.W.; McQuillen, P.S.; Mourani, P.M.; Chima, R.S.; Sorenson, S.; et al. Biomarkers for Estimating Risk of Hospital Mortality and Long-Term Quality-of-Life Morbidity After Surviving Pediatric Septic Shock: A Secondary Analysis of the Life After Pediatric Sepsis Evaluation Investigation. Pediatr. Crit. Care Med. 2021, 22, 8–15. [Google Scholar] [CrossRef] [PubMed]
  264. Fjalstad, J.W.; Esaiassen, E.; Juvet, L.K.; van den Anker, J.N.; Klingenberg, C. Antibiotic Therapy in Neonates and Impact on Gut Microbiota and Antibiotic Resistance Development: A Systematic Review. J. Antimicrob. Chemother. 2018, 73, 569–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Esaiassen, E.; Fjalstad, J.W.; Juvet, L.K.; van den Anker, J.N.; Klingenberg, C. Antibiotic Exposure in Neonates and Early Adverse Outcomes: A Systematic Review and Meta-Analysis. J. Antimicrob. Chemother. 2017, 72, 1858–1870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Kronman, M.P.; Zaoutis, T.E.; Haynes, K.; Feng, R.; Coffin, S.E. Antibiotic Exposure and IBD Development among Children: A Population-Based Cohort Study. Pediatrics 2012, 130. [Google Scholar] [CrossRef] [Green Version]
  267. Bailey, L.C.; Forrest, C.B.; Zhang, P.; Richards, T.M.; Livshits, A.; DeRusso, P.A. Association of Antibiotics in Infancy with Early Childhood Obesity. JAMA Pediatr. 2014, 168, 1063–1069. [Google Scholar] [CrossRef] [Green Version]
  268. Ferrer-Espada, R.; Liu, X.; Goh, X.S.; Dai, T. Antimicrobial Blue Light Inactivation of Polymicrobial Biofilms. Front. Microbiol. 2019, 10, 721. [Google Scholar] [CrossRef]
  269. Halstead, F.D.; Thwaite, J.E.; Burt, R.; Laws, T.R.; Raguse, M.; Moeller, R.; Webber, M.A.; Oppenheim, B.A. Antibacterial Activity of Blue Light against Nosocomial Wound Pathogens Growing Planktonically and as Mature Biofilms. Appl. Environ. Microbiol. 2016, 82, 4006–4016. [Google Scholar] [CrossRef] [Green Version]
  270. Hu, D.; Li, H.; Wang, B.; Ye, Z.; Lei, W.; Jia, F.; Jin, Q.; Ren, K.F.; Ji, J. Surface-Adaptive Gold Nanoparticles with Effective Adherence and Enhanced Photothermal Ablation of Methicillin-Resistant Staphylococcus Aureus Biofilm. ACS Nano 2017, 11, 9330–9339. [Google Scholar] [CrossRef]
  271. Kim, W.; Zou, G.; Hari, T.P.A.; Wilt, I.K.; Zhu, W.; Galle, N.; Faizi, H.A.; Hendricks, G.L.; Tori, K.; Pan, W.; et al. A Selective Membrane-Targeting Repurposed Antibiotic with Activity against Persistent Methicillin-Resistant Staphylococcus Aureus. Proc. Natl. Acad. Sci. USA 2019, 116, 16529–16534. [Google Scholar] [CrossRef] [Green Version]
  272. Kim, W.; Zhu, W.; Hendricks, G.L.; Van Tyne, D.; Steele, A.D.; Keohane, C.E.; Fricke, N.; Conery, A.L.; Shen, S.; Pan, W.; et al. A New Class of Synthetic Retinoid Antibiotics Effective against Bacterial Persisters. Nature 2018, 556, 103–107. [Google Scholar] [CrossRef]
  273. Sun, F.; Bian, M.; Li, Z.; Lv, B.; Gao, Y.; Wang, Y.; Fu, X. 5-Methylindole Potentiates Aminoglycoside Against Gram-Positive Bacteria Including Staphylococcus Aureus Persisters Under Hypoionic Conditions. Front. Cell. Infect. Microbiol. 2020, 10, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Grassi, L.; Maisetta, G.; Esin, S.; Batoni, G. Combination Strategies to Enhance the Efficacy of Antimicrobial Peptides against Bacterial Biofilms. Front. Microbiol. 2017, 8, 2409. [Google Scholar] [CrossRef] [PubMed]
  275. Akturk, E.; Oliveira, H.; Santos, S.B.; Costa, S.; Kuyumcu, S.; Melo, L.D.R.; Azeredo, J. Synergistic Action of Phage and Antibiotics: Parameters to Enhance the Killing Efficacy Against Mono and Dual-Species Biofilms. Antibiotics 2019, 8, 103. [Google Scholar] [CrossRef] [Green Version]
  276. Bi, Y.; Xia, G.; Shi, C.; Wan, J.; Liu, L.; Chen, Y.; Wu, Y.; Zhang, W.; Zhou, M.; He, H.; et al. Therapeutic Strategies against Bacterial Biofilms. Fundam. Res. 2021, 1, 193–212. [Google Scholar] [CrossRef]
  277. Bakhuizen, S.E.; De Haan, T.R.; Teune, M.J.; Van Wassenaer-Leemhuis, A.G.; Van Der Heyden, J.L.; Van Der Ham, D.P.; Mol, B.W.J. Meta-Analysis Shows That Infants Who Have Suffered Neonatal Sepsis Face an Increased Risk of Mortality and Severe Complications. Acta Paediatr. 2014, 103, 1211–1218. [Google Scholar] [CrossRef]
  278. Clark, R.H.; Bloom, B.T.; Spitzer, A.R.; Gerstmann, D.R. Empiric Use of Ampicillin and Cefotaxime, Compared with Ampicillin and Gentamicin, for Neonates at Risk for Sepsis Is Associated with an Increased Risk of Neonatal Death. Pediatrics 2006, 117, 67–74. [Google Scholar] [CrossRef]
  279. Tzialla, C.; Borghesi, A.; Serra, G.; Stronati, M.; Corsello, G. Antimicrobial Therapy in Neonatal Intensive Care Unit. Ital. J. Pediatr. 2015, 41, 27. [Google Scholar] [CrossRef] [Green Version]
  280. 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] [Green Version]
  281. Rubin, L.G.; Sánchez, P.J.; Siegel, J.; Levine, G.; Saiman, L.; Jarvis, W.R. Evaluation and Treatment of Neonates with Suspected Late-Onset Sepsis: A Survey of Neonatologists’ Practices. Pediatrics 2002, 110, e42. [Google Scholar] [CrossRef] [Green Version]
  282. Bizzarro, M.J.; Shabanova, V.; Baltimore, R.S.; Dembry, L.M.; Ehrenkranz, R.A.; Gallagher, P.G. Neonatal Sepsis 2004-2013: The Rise and Fall of Coagulase-Negative Staphylococci. J. Pediatr. 2015, 166, 1193–1199. [Google Scholar] [CrossRef] [Green Version]
  283. Korang, S.K.; Safi, S.; Nava, C.; Greisen, G.; Gupta, M.; Lausten-Thomsen, U.; Jakobsen, J.C. Antibiotic Regimens for Late-Onset Neonatal Sepsis. Cochrane database Syst. Rev. 2021, 5, CD013836. [Google Scholar] [CrossRef] [PubMed]
  284. Ma, X.X.; Wang, E.H.; Liu, Y.; Luo, E.J. Antibiotic Susceptibility of Coagulase-Negative Staphylococci (CoNS): Emergence of Teicoplanin-Non-Susceptible CoNS Strains with Inducible Resistance to Vancomycin. J. Med. Microbiol. 2011, 60, 1661–1668. [Google Scholar] [CrossRef] [PubMed]
  285. Qu, Y.; Daley, A.J.; Istivan, T.S.; Garland, S.M.; Deighton, M.A. Antibiotic Susceptibility of Coagulase-Negative Staphylococci Isolated from Very Low Birth Weight Babies: Comprehensive Comparisons of Bacteria at Different Stages of Biofilm Formation. Ann. Clin. Microbiol. Antimicrob. 2010, 9, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Lopes, N.; Freitas, A.I.; Ramos, H.; Vasconcelos, C.S. Epidermidis Isolates from a Tertiary Care Portuguese Hospital Show Very High Antibiotic Non-Susceptible Rates and Significant Ability to Form Biofilms. Appl. Microbiol. 2021, 1, 150–161. [Google Scholar] [CrossRef]
  287. Mehta, G.; Kumari, S. Multi-Resistant Staphylococcus Haemolyticus in a Neonatal Unit in New Delhi. Ann. Trop. Paediatr. 1997, 17, 15–20. [Google Scholar] [CrossRef] [PubMed]
  288. Westberg, R.; Stegger, M.; Söderquist, B. Molecular Epidemiology of Neonatal-Associated Staphylococcus Haemolyticus Reveals Endemic Outbreak. Microbiol. Spectr. 2022, 10, e0245222. [Google Scholar] [CrossRef]
  289. Klingenberg, C.; Rønnestad, A.; Anderson, A.S.; Abrahamsen, T.G.; Zorman, J.; Villaruz, A.; Flægstad, T.; Otto, M.; Sollid, E.J. Persistent Strains of Coagulase-Negative Staphylococci in a Neonatal Intensive Care Unit: Virulence Factors and Invasiveness. Clin. Microbiol. Infect. 2007, 13, 1100–1111. [Google Scholar] [CrossRef] [Green Version]
  290. Van Der Zwet, W.C.; Debets-Ossenkopp, Y.J.; Reinders, E.; Kapi, M.; Savelkoul, P.H.M.; Van Elburg, R.M.; Hiramatsu, K.; Vandenbroucke-Grauls, C.M.J.E. Nosocomial Spread of a Staphylococcus Capitis Strain with Heteroresistance to Vancomycin in a Neonatal Intensive Care Unit. J. Clin. Microbiol. 2002, 40, 2520–2525. [Google Scholar] [CrossRef] [Green Version]
  291. Ng, P.C.; Chow, V.C.Y.; Lee, C.H.; Ling, J.M.L.; Wong, H.L.; Chan, R.C.Y. Persistent Staphylococcus Capitis Septicemia in a Preterm Infant. Pediatr. Infect. Dis. J. 2006, 25, 652–654. [Google Scholar] [CrossRef]
  292. Laurent, F.; Butin, M. Staphylococcus Capitis and NRCS-A Clone: The Story of an Unrecognized Pathogen in Neonatal Intensive Care Units. Clin. Microbiol. Infect. 2019, 25, 1081–1085. [Google Scholar] [CrossRef]
  293. Asfour, S.S.; Asfour, R.S.; Khalil, T.M.; Al-Mouqdad, M.M. The Use of Daptomycin in the Treatment of Persistent Coagulase-Negative Staphylococcal Sepsis in Premature Infants: A Case Series. J. Pediatr. Pharmacol. Ther. JPPT 2021, 26, 92. [Google Scholar] [CrossRef]
  294. Principi, N.; Caironi, M.; Venturini, F.; Pani, L.; Esposito, S. Daptomycin in Paediatrics: Current Knowledge and the Need for Future Research. J. Antimicrob. Chemother. 2015, 70, 643–648. [Google Scholar] [CrossRef] [Green Version]
  295. Karageorgos, S.A.; Miligkos, M.; Dakoutrou, M.; Tsioutis, C. Clinical Effectiveness, Safety Profile, and Pharmacokinetics of Daptomycin in Pediatric Patients: A Systematic Review. J. Pediatric Infect. Dis. Soc. 2016, 5, 446–457. [Google Scholar] [CrossRef]
  296. Kocher, S.; Müller, W.; Resch, B. Linezolid Treatment of Nosocomial Bacterial Infection with Multiresistant Gram-Positive Pathogens in Preterm Infants: A Systematic Review. Int. J. Antimicrob. Agents 2010, 36, 106–110. [Google Scholar] [CrossRef] [Green Version]
  297. Butin, M.; Martins-Simões, P.; Picaud, J.C.; Kearns, A.; Claris, O.; Vandenesch, F.; Laurent, F.; Rasigade, J.P. Adaptation to Vancomycin Pressure of Multiresistant Staphylococcus Capitis NRCS-A Involved in Neonatal Sepsis. J. Antimicrob. Chemother. 2015, 70, 3027–3031. [Google Scholar] [CrossRef] [Green Version]
  298. Deville, J.G.; Adler, S.; Azimi, P.H.; Jantausch, B.A.; Morfin, M.R.; Beltran, S.; Edge-Padbury, B.; Naberhuis-Stehouwer, S.; Bruss, J.B. Linezolid versus Vancomycin in the Treatment of Known or Suspected Resistant Gram-Positive Infections in Neonates. Pediatr. Infect. Dis. J. 2003, 22, S158–S163. [Google Scholar] [CrossRef]
  299. Trampuz, A.; Zimmerli, W. Antimicrobial Agents in Orthopaedic Surgery: Prophylaxis and Treatment. Drugs 2006, 66, 1089–1106. [Google Scholar] [CrossRef]
  300. Monzón, M.; Oteiza, C.; Leiva, J.; Lamata, M.; Amorena, B. Biofilm Testing of Staphylococcus Epidermidis Clinical Isolates: Low Performance of Vancomycin in Relation to Other Antibiotics. Diagn. Microbiol. Infect. Dis. 2002, 44, 319–324. [Google Scholar] [CrossRef]
  301. 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]
  302. Tan, T.Q.; Mason, E.O.; Ou, C.N.; Kaplan, S.L. Use of Intravenous Rifampin in Neonates with Persistent Staphylococcal Bacteremia. Antimicrob. Agents Chemother. 1993, 37, 2401. [Google Scholar] [CrossRef] [Green Version]
  303. van der Lugt, N.M.; Steggerda, S.J.; Walther, F.J. Use of Rifampin in Persistent Coagulase Negative Staphylococcal Bacteremia in Neonates. BMC Pediatr. 2010, 10, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Shama, A.; Patole, S.K.; Whitehall, J.S. Intravenous Rifampicin in Neonates with Persistent Staphylococcal Bacteraemia. Acta Paediatr. 2002, 91, 670–673. [Google Scholar] [CrossRef] [PubMed]
  305. Dall, G.F.; Tsang, S.T.J.; Gwynne, P.J.; MacKenzie, S.P.; Simpson, A.H.R.W.; Breusch, S.J.; Gallagher, M.P. Unexpected Synergistic and Antagonistic Antibiotic Activity against Staphylococcus Biofilms. J. Antimicrob. Chemother. 2018, 73, 1830–1840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Rybak, M.J.; Lomaestro, B.M.; Rotschafer, J.C.; Moellering, R.C.; Craig, W.A.; Billeter, M.; Dalovisio, J.R.; Levine, D.P. Therapeutic Monitoring of Vancomycin in Adults Summary of Consensus Recommendations from the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists. Pharmacotherapy 2009, 29, 1275–1279. [Google Scholar] [CrossRef]
  307. Hailemeskel, B.; Namanny, M.; Wutoh, A. Frequency of Nephrotoxicity with Vancomycin and Aminoglycoside Antibiotic Therapy. Hosp. Pharm. 1999, 34, 1417–1420. [Google Scholar] [CrossRef]
  308. Sorrell, T.C.; Collignon, P.J. A Prospective Study of Adverse Reactions Associated with Vancomycin Therapy. J. Antimicrob. Chemother. 1985, 16, 235–241. [Google Scholar] [CrossRef]
  309. Ozkaya-Parlakay, A.; Kara, A.; Celik, M.; Ozsurekci, Y.; Oncel, E.K.; Ceyhan, M.; Cengiz, A.B. Early Lactic Acidosis Associated with Linezolid Therapy in Paediatric Patients. Int. J. Antimicrob. Agents 2014, 44, 334–336. [Google Scholar] [CrossRef]
  310. Alsayyed, B. Rifampin. Pediatr. Rev. 2004, 25, 216–217. [Google Scholar] [CrossRef]
  311. Arnold, C.J.; Ericson, J.; Kohman, J.; Corey, K.L.; Oh, M.; Onabanjo, J.; Hornik, C.P.; Clark, R.H.; Benjamin, D.K.; Smith, P.B.; et al. Rifampin Use and Safety in Hospitalized Infants. Am. J. Perinatol. 2015, 32, 565–570. [Google Scholar] [CrossRef] [Green Version]
  312. Brian Smith, P.; Michael Cotten, C.; Hudak, M.L.; Sullivan, J.E.; Poindexter, B.B.; Cohen-Wolkowiez, M.; Boakye-Agyeman, F.; Lewandowski, A.; Anand, R.; Benjamin, D.K.; et al. Rifampin Pharmacokinetics and Safety in Preterm and Term Infants. Antimicrob. Agents Chemother. 2019, 63, e00284-19. [Google Scholar] [CrossRef] [Green Version]
  313. Filioti, J.; Spiroglou, K.; Roilides, E. Invasive Candidiasis in Pediatric Intensive Care Patients: Epidemiology, Risk Factors, Management, and Outcome. Intensive Care Med. 2007, 33, 1272–1283. [Google Scholar] [CrossRef] [PubMed]
  314. Spiliopoulou, A.; Dimitriou, G.; Jelastopulu, E.; Giannakopoulos, I.; Anastassiou, E.D.; Christofidou, M. Neonatal Intensive Care Unit Candidemia: Epidemiology, Risk Factors, Outcome, and Critical Review of Published Case Series. Mycopathologia 2012, 173, 219–228. [Google Scholar] [CrossRef] [PubMed]
  315. Antonoplis, A.; Zang, X.; Huttner, M.A.; Chong, K.K.L.; Lee, Y.B.; Co, J.Y.; Amieva, M.R.; Kline, K.A.; Wender, P.A.; Cegelski, L. A Dual-Function Antibiotic-Transporter Conjugate Exhibits Superior Activity in Sterilizing MRSA Biofilms and Killing Persister Cells. J. Am. Chem. Soc. 2018, 140, 16140–16151. [Google Scholar] [CrossRef] [PubMed]
  316. Schmidt, N.W.; Deshayes, S.; Hawker, S.; Blacker, A.; Kasko, A.M.; Wong, G.C.L. Engineering Persister-Specific Antibiotics with Synergistic Antimicrobial Functions. Deshayes Contrib. Equal. 2014, 8, 8786–8793. [Google Scholar] [CrossRef] [Green Version]
  317. Joo, H.S.; Otto, M. Mechanisms of Resistance to Antimicrobial Peptides in Staphylococci. Biochim. Biophys. Acta 2015, 1848, 3055–3061. [Google Scholar] [CrossRef] [Green Version]
  318. Peschel, A.; Sahl, H.G. The Co-Evolution of Host Cationic Antimicrobial Peptides and Microbial Resistance. Nat. Rev. Microbiol. 2006, 4, 529–536. [Google Scholar] [CrossRef]
  319. Zhu, C.; Zhao, Y.; Zhao, X.; Liu, S.; Xia, X.; Zhang, S.; Wang, Y.; Zhang, H.; Xu, Y.; Chen, S.; et al. The Antimicrobial Peptide MPX Can Kill Staphylococcus Aureus, Reduce Biofilm Formation, and Effectively Treat Bacterial Skin Infections in Mice. Front. Vet. Sci. 2022, 9, 327. [Google Scholar] [CrossRef]
  320. Fleming, D.; Rumbaugh, K. The Consequences of Biofilm Dispersal on the Host. Sci. Rep. 2018, 8, 10738. [Google Scholar] [CrossRef] [Green Version]
  321. Donelli, G.; Francolini, I.; Romoli, D.; Guaglianone, E.; Piozzi, A.; Ragunath, C.; Kaplan, J.B. Synergistic Activity of Dispersin B and Cefamandole Nafate in Inhibition of Staphylococcal Biofilm Growth on Polyurethanes. Antimicrob. Agents Chemother. 2007, 51, 2733–2740. [Google Scholar] [CrossRef] [Green Version]
  322. França, A.; Pérez-Cabezas, B.; Correia, A.; Pier, G.B.; Cerca, N.; Vilanova, M. Staphylococcus Epidermidis Biofilm-Released Cells Induce a Prompt and More Marked in Vivo Inflammatory-Type Response than Planktonic or Biofilm Cells. Front. Microbiol. 2016, 7, 1530. [Google Scholar] [CrossRef] [Green Version]
  323. Izano, E.A.; Amarante, M.A.; Kher, W.B.; Kaplan, J.B. Differential Roles of Poly-N-Acetylglucosamine Surface Polysaccharide and Extracellular DNA in Staphylococcus Aureus and Staphylococcus Epidermidis Biofilms. Appl. Environ. Microbiol. 2008, 74, 470–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  324. Chaignon, P.; Sadovskaya, I.; Ragunah, C.; Ramasubbu, N.; Kaplan, J.B.; Jabbouri, S. Susceptibility of Staphylococcal Biofilms to Enzymatic Treatments Depends on Their Chemical Composition. Appl. Microbiol. Biotechnol. 2007, 75, 125–132. [Google Scholar] [CrossRef] [PubMed]
  325. Ramasubbu, N.; Thomas, L.M.; Ragunath, C.; Kaplan, J.B. Structural Analysis of Dispersin B, a Biofilm-Releasing Glycoside Hydrolase from the Periodontopathogen Actinobacillus Actinomycetemcomitans. J. Mol. Biol. 2005, 349, 475–486. [Google Scholar] [CrossRef] [PubMed]
  326. Wu, J.A.; Kusuma, C.; Mond, J.J.; Kokai-Kun, J.F. Lysostaphin Disrupts Staphylococcus Aureus and Staphylococcus Epidermidis Biofilms on Artificial Surfaces. Antimicrob. Agents Chemother. 2003, 47, 3407–3414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Sieprawska-Lupa, M.; Mydel, P.; Krawczyk, K.; Wójcik, K.; Puklo, M.; Lupa, B.; Suder, P.; Silberring, J.; Reed, M.; Pohl, J.; et al. Degradation of Human Antimicrobial Peptide LL-37 by Staphylococcus Aureus-Derived Proteinases. Antimicrob. Agents Chemother. 2004, 48, 4673–4679. [Google Scholar] [CrossRef] [Green Version]
  328. Prokešová, L.; Potuẑníková, B.; Potempa, J.; Zikán, J.; Radl, J.; Hachová, L.; Baran, K.; Porwit-Bbr, Z.; John, C. Cleavage of Human Immunoglobulins by Serine Proteinase from Staphylococcus Aureus. Immunol. Lett. 1992, 31, 259–265. [Google Scholar] [CrossRef]
  329. Pires, D.P.; Melo, L.D.R.; Vilas Boas, D.; Sillankorva, S.; Azeredo, J. Phage Therapy as an Alternative or Complementary Strategy to Prevent and Control Biofilm-Related Infections. Curr. Opin. Microbiol. 2017, 39, 48–56. [Google Scholar] [CrossRef] [Green Version]
  330. Melo, L.D.R.; Oliveira, H.; Pires, D.P.; Dabrowska, K.; Azeredo, J. Phage Therapy Efficacy: A Review of the Last 10 Years of Preclinical Studies. Crit. Rev. Microbiol. 2020, 46, 78–99. [Google Scholar] [CrossRef]
  331. Rohde, C.; Wittmann, J.; Kutter, E. Bacteriophages: A Therapy Concept against Multi-Drug-Resistant Bacteria. Surg. Infect. 2018, 19, 737–744. [Google Scholar] [CrossRef] [Green Version]
  332. González, S.; Fernández, L.; Campelo, A.B.; Gutiérrez, D.; Martínez, B.; Rodríguez, A.; García, P. The Behavior of Staphylococcus Aureus Dual-Species Biofilms Treated with Bacteriophage PhiIPLA-RODI Depends on the Accompanying Microorganism. Appl. Environ. Microbiol. 2017, 83, e02821-16. [Google Scholar] [CrossRef] [Green Version]
  333. Cerca, N.; Oliveira, R.; Azeredo, J. Susceptibility of Staphylococcus Epidermidis Planktonic Cells and Biofilms to the Lytic Action of Staphylococcus Bacteriophage K. Lett. Appl. Microbiol. 2007, 45, 313–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Alves, D.R.; Gaudion, A.; Bean, J.E.; Perez Esteban, P.; Arnot, T.C.; Harper, D.R.; Kot, W.; Hansen, L.H.; Enright, M.C.; Jenkins, A.T.A. Combined Use of Bacteriophage K and a Novel Bacteriophage to Reduce Staphylococcus Aureus Biofilm Formation. Appl. Environ. Microbiol. 2014, 80, 6694–6703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Melo, L.D.R.; Pinto, G.; Oliveira, F.; Vilas-Boas, D.; Almeida, C.; Sillankorva, S.; Cerca, N.; Azeredo, J. The Protective Effect of Staphylococcus Epidermidis Biofilm Matrix against Phage Predation. Viruses 2020, 12, 1076. [Google Scholar] [CrossRef]
  336. Melo, L.D.R.; França, A.; Brandão, A.; Sillankorva, S.; Cerca, N.; Azeredo, J. Assessment of Sep1virus Interaction with Stationary Cultures by Transcriptional and Flow Cytometry Studies. FEMS Microbiol. Ecol. 2018, 94, fiy143. [Google Scholar] [CrossRef] [PubMed]
  337. Gutiérrez, D.; Vandenheuvel, D.; Martínez, B.; Rodríguez, A.; Lavigne, R.; García, P. Two Phages, PhiIPLA-RODI and PhiIPLA-C1C, Lyse Mono- and Dual-Species Staphylococcal Biofilms. Appl. Environ. Microbiol. 2015, 81, 3336–3348. [Google Scholar] [CrossRef] [Green Version]
  338. Fernández, L.; González, S.; Campelo, A.B.; Martínez, B.; Rodríguez, A.; García, P. Low-Level Predation by Lytic Phage PhiIPLA-RODI Promotes Biofilm Formation and Triggers the Stringent Response in Staphylococcus Aureus. Sci. Rep. 2017, 7, 40965. [Google Scholar] [CrossRef] [Green Version]
  339. Hosseinidoust, Z.; Tufenkji, N.; van de Ven, T.G.M. Formation of Biofilms under Phage Predation: Considerations Concerning a Biofilm Increase. Biofouling 2013, 29, 457–468. [Google Scholar] [CrossRef] [Green Version]
  340. García, P.; Martínez, B.; Rodríguez, L.; Rodríguez, A. Synergy between the Phage Endolysin LysH5 and Nisin to Kill Staphylococcus Aureus in Pasteurized Milk. Int. J. Food Microbiol. 2010, 141, 151–155. [Google Scholar] [CrossRef] [Green Version]
  341. Gutiérrez, D.; Ruas-Madiedo, P.; Martínez, B.; Rodríguez, A.; García, P. Effective Removal of Staphylococcal Biofilms by the Endolysin LysH5. PLoS ONE 2014, 9, e107307. [Google Scholar] [CrossRef] [Green Version]
  342. Zhang, Y.; Cheng, M.; Zhang, H.; Dai, J.; Guo, Z.; Li, X.; Ji, Y.; Cai, R.; Xi, H.; Wang, X.; et al. Antibacterial Effects of Phage Lysin LysGH15 on Planktonic Cells and Biofilms of Diverse Staphylococci. Appl. Environ. Microbiol. 2018, 84, e00886-18. [Google Scholar] [CrossRef] [Green Version]
  343. Schuch, R.; Khan, B.K.; Raz, A.; Rotolo, J.A.; Wittekind, M. Bacteriophage Lysin CF-301, a Potent Antistaphylococcal Biofilm Agent. Antimicrob. Agents Chemother. 2017, 61, e02666-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Dickey, J.; Perrot, V. Adjunct Phage Treatment Enhances the Effectiveness of Low Antibiotic Concentration against Staphylococcus Aureus Biofilms in Vitro. PLoS ONE 2019, 14, e0209390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Tkhilaishvili, T.; Lombardi, L.; Klatt, A.B.; Trampuz, A.; Di Luca, M. Bacteriophage Sb-1 Enhances Antibiotic Activity against Biofilm, Degrades Exopolysaccharide Matrix and Targets Persisters of Staphylococcus Aureus. Int. J. Antimicrob. Agents 2018, 52, 842–853. [Google Scholar] [CrossRef] [PubMed]
  346. Cobb, L.H.; Park, J.Y.; Swanson, E.A.; Beard, M.C.; McCabe, E.M.; Rourke, A.S.; Seo, K.S.; Olivier, A.K.; Priddy, L.B. CRISPR-Cas9 Modified Bacteriophage for Treatment of Staphylococcus Aureus Induced Osteomyelitis and Soft Tissue Infection. PLoS ONE 2019, 14, e0220421. [Google Scholar] [CrossRef] [Green Version]
  347. Bikard, D.; Jiang, W.; Samai, P.; Hochschild, A.; Zhang, F.; Marraffini, L.A. Programmable Repression and Activation of Bacterial Gene Expression Using an Engineered CRISPR-Cas System. Nucleic Acids Res. 2013, 41, 7429–7437. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Summary of the several aspects contributing to the development of CoNS-caused infections in preterm infants. MSCRAMMS, microbial surface components recognizing matrix molecules; Atl, autolysins; TA, teichoic acids; eDNA, extracellular DNA; PNAG, poly-N-acetylglucosamine; HTG, horizontal gene transfer.
Figure 1. Summary of the several aspects contributing to the development of CoNS-caused infections in preterm infants. MSCRAMMS, microbial surface components recognizing matrix molecules; Atl, autolysins; TA, teichoic acids; eDNA, extracellular DNA; PNAG, poly-N-acetylglucosamine; HTG, horizontal gene transfer.
Antibiotics 12 00554 g001
Figure 2. Schematics representing CoNS biofilm formation main steps and molecules involved. HTG, horizontal gene transfer; AMPs, antimicrobial peptides; MSCRAMMS, microbial surface components recognizing matrix molecules; Atl, autolysins; Embp, extracellular matrix binding protein; TA, teichoic acids; PNAG, poly-N-acetylglucosamine; eDNA, extracellular DNA; PSMs, phenol-soluble modulins.
Figure 2. Schematics representing CoNS biofilm formation main steps and molecules involved. HTG, horizontal gene transfer; AMPs, antimicrobial peptides; MSCRAMMS, microbial surface components recognizing matrix molecules; Atl, autolysins; Embp, extracellular matrix binding protein; TA, teichoic acids; PNAG, poly-N-acetylglucosamine; eDNA, extracellular DNA; PSMs, phenol-soluble modulins.
Antibiotics 12 00554 g002
Figure 3. Summary of some of the currently used methods used for diagnosis of LOS, along with advanced and under-development methods. Of note, the list of techniques detailed under “current diagnostic methods” includes methods that are normally used alone, such as blood culture and those that are used as “add-on” tests, such as biomarkers.
Figure 3. Summary of some of the currently used methods used for diagnosis of LOS, along with advanced and under-development methods. Of note, the list of techniques detailed under “current diagnostic methods” includes methods that are normally used alone, such as blood culture and those that are used as “add-on” tests, such as biomarkers.
Antibiotics 12 00554 g003
Figure 4. Summary of the current and under-development therapeutic methods for CoNS-associated infections, including LOS. The use of nanoparticles, as well as photoinactivation, which involves the use of visible light, a photosensitizer and oxygen to produce reactive oxygen species and free radicals that create oxidative stress in bacteria [158], have shown promising results in staphylococci biofilms, including methicillin-resistant S. aureus [268,269,270,276], but more studies are necessary to investigate their consequences on host cells.
Figure 4. Summary of the current and under-development therapeutic methods for CoNS-associated infections, including LOS. The use of nanoparticles, as well as photoinactivation, which involves the use of visible light, a photosensitizer and oxygen to produce reactive oxygen species and free radicals that create oxidative stress in bacteria [158], have shown promising results in staphylococci biofilms, including methicillin-resistant S. aureus [268,269,270,276], but more studies are necessary to investigate their consequences on host cells.
Antibiotics 12 00554 g004
Table 1. Compilation of the differences that can be found between EOS and LOS [12,16,17,18,19,20].
Table 1. Compilation of the differences that can be found between EOS and LOS [12,16,17,18,19,20].
CharacteristicsEOSLOS
Onset0–3 days3–28 days
TransmissionVertical (female genitourinary system)Horizontal (neonatal/community environments)
Frequent causative agentsGroup B Streptococcus (GBS)CoNS
Escherichia coliStaphylococcus aureus
Haemophilus influenzaEscherichia coli
Listeria monocytogenes (dietary intake)Klebsiella pneumonia
Acinetobacter baumannii
Risk factors
-
Maternal GBS colonization
-
Prematurity
-
Chorioamnionitis
-
Low birth weight
-
Delivery before 37 weeks
-
Prolonged use of catheters/medical devices
-
Premature rupture of membranes
-
Invasive procedures
-
Prolonged rupture of membranes (>18 h)
-
Prolonged antibiotic therapy
-
Multiple gestations
-
Preterm birth (<37 weeks)
Mortality3–16%36–52%
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

França, A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics 2023, 12, 554. https://doi.org/10.3390/antibiotics12030554

AMA Style

França A. The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies. Antibiotics. 2023; 12(3):554. https://doi.org/10.3390/antibiotics12030554

Chicago/Turabian Style

França, Angela. 2023. "The Role of Coagulase-Negative Staphylococci Biofilms on Late-Onset Sepsis: Current Challenges and Emerging Diagnostics and Therapies" Antibiotics 12, no. 3: 554. https://doi.org/10.3390/antibiotics12030554

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