Next Article in Journal
Leucoverdazyls as Novel Potent Inhibitors of Enterovirus Replication
Previous Article in Journal
Phenotypic and Genotypic Characterization of Staphylococcus aureus Isolated from Nasal Samples of Healthy Dairy Goats in Algeria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Enterococcus spp. in the Immunocompromised Host: A Comprehensive Review

by
Giuseppe Sangiorgio
1,*,
Maddalena Calvo
2,
Giuseppe Migliorisi
2,
Floriana Campanile
1 and
Stefania Stefani
1,2
1
Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
2
U.O.C. Laboratory Analysis Unit, University Hospital Policlinico-San Marco, Via Santa Sofia 78, 95123 Catania, Italy
*
Author to whom correspondence should be addressed.
Pathogens 2024, 13(5), 409; https://doi.org/10.3390/pathogens13050409
Submission received: 20 April 2024 / Revised: 5 May 2024 / Accepted: 14 May 2024 / Published: 15 May 2024
(This article belongs to the Section Bacterial Pathogens)

Abstract

:
The immunocompromised host is usually vulnerable to infectious diseases due to broad-spectrum treatments and immunological dysregulation. The Enterococcus genus consists of normal gut commensals, which acquire a leading role in infective processes among individuals with compromised immune systems. These microorganisms may express a potential virulence and resistance spectrum, enabling their function as severe pathogens. The Enterococcus spp. infections in immunocompromised hosts appear to be difficult to resolve due to the immunological response impairment and the possibility of facing antimicrobial-resistant strains. As regards the related risk factors, several data demonstrated that prior antibiotic exposure, medical device insertion, prolonged hospitalization and surgical interventions may lead to Enterococcus overgrowth, antibiotic resistance and spread among critical healthcare settings. Herein, we present a comprehensive review of Enterococcus spp. in the immunocompromised host, summarizing the available knowledge about virulence factors, antimicrobial-resistance mechanisms and host-pathogen interaction. The review ultimately yearns for more substantial support to further investigations about enterococcal infections and immunocompromised host response.

1. Introduction

Microorganisms belonging to the Enterococcus genus have long been recognized as important human gut commensals. Despite this assumption, their ability to persist in the environment and resist disinfection procedures has led to their widespread distribution in clinical settings [1]. Particularly, Enterococcus faecalis and Enterococcus faecium are notable for causing considerable management issues due to episodes of antimicrobial resistance. Their interactions with the host, including alterations in host cell signalling pathways and evasion of immune responses, further increase their pathogenicity and ability to cause long-lasting infections [2,3]. It is crucial to understand the characteristics, patterns of antibiotic resistance, and host interactions of E. faecalis and E. faecium for several reasons. Firstly, these bacteria are responsible for a wide range of infections, including urinary tract infections, bacteraemia, endocarditis, intra-abdominal infections, and surgical site infections. Furthermore, the clinical significance of Enterococcus species is heightened by their ability to cause both community-acquired and healthcare-associated illnesses [4,5,6]. Secondly, the emergence and spread of antibiotic-resistant strains, particularly those resistant to vancomycin, pose a significant threat to public health. This resistance profile complicates therapeutic approaches, increases the risk of treatment failure, and underscores the urgent need for effective antimicrobial stewardship procedures [7,8,9]. Thirdly, the development and persistence of infections are significantly influenced by the relationship between Enterococcus species and the host.
Enterococcus species is a leading cause of nosocomial infections, especially in immunocompromised patients. The rise in vancomycin-resistant Enterococcus (VRE) cases across Europe is particularly concerning, with resistance even emerging against last-resort antibiotics, severely limiting treatment options [10]. Infections caused by multidrug-resistant (MDR) enterococcal strains are associated with substantial economic burdens and higher rates of morbidity and mortality compared to those caused by susceptible strains [11,12]. Additionally, enterococci are highly resilient, withstanding harsh conditions, resisting biocides, forming biofilms, and possessing high genetic adaptability, all of which pose challenges to decontamination efforts. Consequently, enterococci serve as significant sources of nosocomial outbreaks and the dissemination of resistance genes. Diagnosis of antimicrobial tolerance in persistent infections in immunocompromised hosts, or in body sites with restricted immune access, is of particular concern as strong, effective bactericidal antimicrobials are necessary in these circumstances [13]. Moreover, enterococci play a significant role in maintaining intestinal homeostasis, contributing to continuous immune system stimulation [14]. Understanding these interactions between the host and these microorganisms can help identify new therapeutic targets and preventative measures for enterococcal infections [15,16].
In this review, we aim to provide a comprehensive insight into Enterococcus species as human pathogen and how they interact with immunocompromised patients, focusing on its pathogenicity, antibiotic resistance, and interaction mechanisms with the host.

2. Epidemiological Patterns of Enterococcal Infections

According to the World Health Organization (WHO) recent data, Enterococcus species mortality rate waves between 14.3% and 32.3% [17]. In 2020, vancomycin resistance in E. faecium showed significant variation across European countries and regions, with seven (18%) reporting percentages below 1%, including Finland, France, Iceland, the Netherlands, Norway, Sweden, and Ukraine. Conversely, 13 countries/areas (34%) reported antimicrobial resistance (AMR) percentages equal to or above 25%, with four (11% of 38 countries/areas) reporting percentages equal to or above 50%, including Bosnia and Herzegovina, Lithuania, North Macedonia, and Serbia [10]. Globally, the incidence of E. faecalis infections has remained relatively stable over the past two decades, although literary data report some localized outbreaks [18,19,20,21,22]. In Europe, according to the European Center for Disease Prevention and Control (ECDC) Surveillance Atlas of Infectious Diseases [23], E. faecium displays distinctive characteristics and epidemiology compared to E. faecalis, along with varying trends across different European countries and regions, showing an upward trajectory over the past two decades. For instance, in the Netherlands, the average number of invasive ampicillin-resistant enterococcal infections per hospital increased fivefold from 1999 to 2005 [24]. By 2007, enterococcal isolates exhibited significant variation, ranging from over 30% in countries such as Greece and Ireland to less than 1% in Scandinavian countries [25]. Meanwhile, a concerning report from Sweden during the period 2007–2009 highlighted an approximately fourfold increase [26].
In Italy, Enterococcus faecium generally cause healthcare-associated infections. Moreover, it is more commonly related with vancomycin resistance, and exhibits a higher propensity for multidrug resistance than Enterococcus faecalis (Figure 1). Enterococcus infections pose substantial challenges in clinical management due to their antibiotic resistance and ability to cause severe infections, especially in vulnerable patient populations. Among these populations, we performed a screening in the Southern Italy area using “Sicilian antibiotic resistance dashboard—Rete MIC” [27] (Figure 2). The SARS-Cov-2 pandemia reported a drastic increase in antimicrobial-resistant enterococci, probably due to insufficient treatment guidelines and antibiotic overprescription. According to literary data, several regions documented high resistance rates among Enterococcus spp., especially related to E. faecium [28]. Enterococcus faecalis and Enterococcus faecium represent the most diffused enterococcal species. However, less common enterococci such as Enterococcus avium, Enterococcus gallinarum, Enterococcus durans, Enterococcus hirae, and Enterococcus raffinosus can cause more than 24% of enterococcal infections [29].

2.1. Nosocomial Transmission

An initial crucial factor leading to nosocomial enterococcal infection appears to be the heightened density of colonization within the gastrointestinal tract [30]. Enterococci residing in the gastrointestinal tract can breach the intestinal barrier and traverse the liver, gaining access to the bloodstream. Once in circulation, these pathogens can travel to the heart, potentially leading to infective endocarditis. Environmental contamination from fecal sources, serving as a reservoir for colonization in other patients, along with contamination of the patient’s skin, primarily contributes to urinary tract infections and intravenous catheter-related infections [31]. Comprehending the transmission patterns of enterococci within hospital settings is crucial for infection management. The administration of antimicrobials to hospitalized patients frequently establishes VRE dominance in the gastrointestinal tract, thereby promoting the dissemination of these pathogens. Hence normally, intestinal epithelial cells and Paneth cells produce REGIIIγ, a C-type lectin that combats Gram-positive bacteria [32]. REGIIIγ production is initiated by the presence of Gram-negative bacteria and their microorganism-associated molecular patterns (MAMPs), such as lipopolysaccharide and flagellin [33]. However, antibiotic treatment reduces the population of Gram-negative bacteria, resulting in decreased REGIIIγ production. This decline in REGIIIγ secretion creates a favourable environment for enterococci to multiply and become the primary constituents of the gut microbiota [32,33]. In addition, the factors contributing to the risk of colonization and subsequent infection with VRE include close physical proximity to infected or colonized patients, prolonged hospitalization, stays in long-term care facilities, surgical or intensive care units, urinary catheterization, and repeated antibiotic treatments [34]. Antibiotic administration often increases the density of VRE in the gastrointestinal tract, facilitating their dissemination through faecal contamination of the hospital environment, including surfaces and the hands of healthcare workers and visitors. Moreover, enterococci exhibit remarkable resilience on environmental surfaces such as medical equipment, toilets, bed rails, and doorknobs, and demonstrate tolerance to heat, chlorine, and certain alcohol preparations, enhancing their survival and transmission potential [35].

2.2. Molecular Epidemiology in Clinical Setting

Population genetics studies have shed light on the evolutionary dynamics of enterococci [36,37]. Analyses employing multilocus sequence typing (MLST), which evaluates allelic variations in seven housekeeping genes, identified distinct clonal complexes among isolates recovered from hospitalized patients, distinguishing them from those of commensal or animal-derived isolates [38]. Notably, hospital-derived E. faecalis isolates predominantly cluster in clonal complexes CC2 and CC9 [39], while the increasing prevalence of E. faecium is attributed to a polyclonal subpopulation, particularly MLST sequence types 17, 18, 78, and 192, formerly known as clonal complex CC17 [40]. While E. faecalis is responsible for the majority of infections, the hospital-adapted genotype of E. faecium exhibits a higher propensity for MDR [29]. The global phylogeny of E. faecium reveals the prevalence of two discernible phylogenetic clades, labeled A and B. Clade A can be subdivided into two distinct subclades: A1, predominantly composed of clinical strains, and A2, comprising strains primarily isolated from animals but also some non-hospitalized individuals. Clade B encompasses isolates from community settings [41]. The plasticity of enterococcal genomes presents a challenge, with acquired elements potentially accounting for up to a quarter of the genome [42,43]. Conjugation events between enterococci can lead to the generation of hybrid strains, resulting in alterations in MLST patterns and genomic composition [44]. Notably, clustered regularly interspaced short palindromic repeats (CRISPR) loci and other genetic markers may contribute to provide insights into the phenotypic traits and genetic makeup of enterococci, as well as distinguishing between low-risk and high-risk strains [45].

2.3. Clinical Impact

Enterococci are one of the most common sources of healthcare-associated infections in developed countries, causing a range of infections such as bacteraemia, infective endocarditis, intra-abdominal and pelvic infections, skin infections, and central nervous infections [46,47]. Intra-abdominal, pelvic, and post-surgery wounds infections are the second most frequent enterococcal infection type. Additionally, enterococci rank the third most common causative agent in bloodstream infections and infective endocarditis, leading to significant morbidity and mortality rates [48]. Notably, enterococcal bloodstream infections account for around 10% of all cases of bacteraemia. While extensive studies reveal that enterococci are responsible for approximately 30% of hospital-associated endocarditis cases, following Staphylococcus spp., E. faecalis is the most frequent cause of both bloodstream and urinary tract infections, followed by E. faecium [49]. The spread of MDR strains limits treatment options, and enterococci have also been isolated from skin infections and, occasionally, reported to cause osteomyelitis, septic arthritis, and central nervous system infections like meningitis. Enterococcal infections, particularly those caused by VRE, are associated with high mortality rates, ranging from 25% to 50%, with a more significant impact on immunocompromised patients [50]. Although initially recognized more as intestinal colonization bacteria than virulent agents, they are increasingly considered causative agents of severe systemic infections, particularly in immunocompromised individuals, including haematological cancer patients. Haematological conditions, such as acute myeloid leukaemia and Hodgkin’s and non-Hodgkin’s lymphoma, that affect the blood and blood-forming organs appear to be related to an increased risk of enterococcal infections. Neutropenic individuals, especially, have a weakened immune response, facing high susceptibility to bacterial infections, including those caused by Enterococcus [51,52]. Patients undergoing haematopoietic stem cell transplantation, such as bone marrow or stem cell transplantation, often experience prolonged periods of immunosuppression, carrying vulnerability to various infections, including Enterococcus ones [53,54,55,56].

3. Antimicrobial Resistance

The use of glycopeptides has been extensively intensified because of the insufficient antimicrobial power of other therapeutical choices. This intensification in glycopeptides administration, along with an avoparcin use increase among farm animals, have boosted resistance diffusion [57]. Since 1995, the European Union (EU) banned the avoparcin use among farms, leading to a diminution in glycopeptide-resistant enterococci from human faecal samples. However, data demonstrated how the resistant enterococcal infection prevalence in humans remained elevated [57]. VRE have been a consistent infectious challenge in human medicine since 1980, when the first resistance isolates appeared [58,59]. Furthermore, literary evidence shows the high VRE incidence rates among immunocompromised patients [60]. The glycopeptides resistance is related to the acquisition of van genes operons, whose expression allows the production of alternative aminoacidic residuals in the peptidoglycan structure. Specifically, the alternative combinations D-alanin-D-lactate or D-alanin-D-serine substitute the wild-type D-alanin-D-alanin terminus. The substitution preserves the solidity of the enterococcal cell wall, but compromises the target recognition by the glycopeptide drugs. Depending on the involved van gene, different resistance phenotypes occur. VanA and VanB are the most common, followed by less diffused VanD and VanC types. The different Van phenotypes lead to several combinations of antibiotic resistance, alternatively involving vancomycin teicoplanin or both glycopeptide molecules [59,61]. Treating VRE poses a significant challenge due to the limited number of antibiotics available for effective treatment, with linezolid and daptomycin being the primary choices [62]. Enterococci are usually broadly susceptible to linezolid, but prior studies have shown that both linezolid- and daptomycin-resistant enterococci can emerge after these drug’s exposure or in the absence of their use [63,64,65]. The resistance mechanisms to linezolid are related to rRNA genes, whose number is variable based on the evaluated species (6 for E. faecium and 4 for E. faecalis). Therefore, the resistance expression level depends on the number of the involved genes. Moreover, enterococci can also develop linezolid resistance due to the acquisition of cfr genes, which are located in plasmids and contribute to methyltransferase modification [29]. Resistance to daptomycin is often related to phospholipid alterations in enterococcal cell membranes in E. faecalis and E. faecium. Also, membrane depolarization and cell wall fitness reduction may contribute to the same resistance phenomenon [66].
Enterococci also express resistance against aminoglycosides, β-lactams, and lincosamides [67]. As regards β-lactams resistance, E. faecalis often produces β-lactamases, while E. faecium expresses high-level penicillin resistance due to the expression of penicillin-binding protein 5 (PBP5). Epidemiological studies suggest how penicillin-resistant E. faecium can spread among hospital settings, accounting for more than 70% of incidence. E. faecalis strains can be susceptible to carbapenems, which are not a priority therapeutical choice. Some literature evidence highlights the E. faecalis capability to develop ceftobiprole resistance through pbp4 gene sequence alterations, which compromise ceftobiprole binding to its target [68]. The production of aminoglycoside-modifying enzymes and ribosomal mutations leads to intrinsic resistance to aminoglycosides. Enterococcus species exhibit mutations in topoisomerase gyrA and parC genes, leading to an acquired resistance to fluoroquinolones, whose use against enterococcal infections is limited to selected urinary tract infections. While all enterococcal isolates are typically susceptible to erythromycin and tetracyclines, they may occasionally acquire resistance to these antimicrobial agents. For instance, transposons carrying tet(M) protection gene mediate tetracycline resistance [29]. Enterococci may express macrolides, lincosamides and type b streptogramins (MLSb) resistance due to the acquisition of the erm gene, whose expression causes a reduced target affinity for all the MLSb drugs. For this reason, the MLSb phenotype identifies this type of extended antimicrobial resistance [61]. Figure 3 summarizes the principal mechanism of enterococcal antibiotic resistance.

4. Virulence Factors

Enterococcus spp. exhibit resilience in harsh environments and diverse ecological niches, transitioning from commensal to pathogen with mechanisms still not fully understood. Intrinsic traits of E. faecalis, such as stress responses and antibiotic resistance, likely aid in infection progression [69,70,71]. Bacterial adaptation to environmental changes often involves two-component signal transduction systems (TCS), where a histidine kinase detects signals and transfers a phosphoryl group to a response regulator, regulating gene expression [72]. Among the 17 TCS identified in the genome of E. faecalis [73], the Fsr system has been extensively studied and seems crucially connected to enterococcal virulence [74,75,76]. The fsr locus encompasses four genes: fsrA, fsrB, fsrC, and fsrD, forming a system responsive to extracellular gelatinase biosynthesis-activating pheromone (GBAP) accumulation [77]. FsrB functions as a cysteine protease-like processing enzyme for FsrD peptide [78]. Extracellular FsrD accumulation is sensed by FsrC, activating the response regulator and transcription factor FsrA. Stimulation of FsrC by chemically synthesized GBAP peptide has been demonstrated both in vivo and in vitro [79]. FsrABDC proteins play a pivotal role in activating two E. faecalis virulence-associated proteases, gelatinase (GelE) and serine protease (SprE), at a promoter upstream of the gelE gene [78,80]. Several studies on animal models have demonstrated the crucial role of these key factors in enterococcal virulence [81,82,83]. Additionally, gelatinase is essential for facilitating efficient biofilm formation and plays a significant role in the pathogenesis of enterococcal endocarditis [84,85]. The FsrA response regulator, similar to AgrA of Staphylococcus aureus, belongs to a protein family characterized by a LytTR DNA-binding domain, which is unique compared to the typical DNA-binding domains of response regulators and is frequently associated with the regulation of virulence factors such as toxins, bacteriocins, and extracellular polysaccharides [86]. These virulence factors can facilitate tissue invasion, promote immune evasion, and contribute to the overall virulence of the bacterium [87,88]. In E. faecium, extensively studied virulence genes involve surface proteins (Fms) crucial for adhesion, biofilm formation, and pili assembly. Specifically, these proteins include Esp (enterococcal surface protein), Acm (adhesin of collagen from Efm), Scm (second collagen adhesin from Efm), SgrA (serine-glutamate-repeat-containing-protein A), and EcbA (Efm-collagen-binding-protein A) [89,90,91]. Enterococcal cell membrane glycolipids and lipoteichoic acid play a significant role in pathogenesis [92]. A mutant E. faecalis strain lacking the glycolipid α-diglycosyl diacylglycerol (DGlcDAG) showed reduced adherence to enterocytes and biofilm formation [93]. Another virulence factor, SagA, identified as a major antigen in E. faecium, comprises three domains with distinct sequence and/or structural characteristics. SagA plays a crucial role in bacterial growth, potentially influencing cell wall metabolism, and can form oligomers when overexpressed, while also binding to fibrinogen and various ECM proteins, indicating broad adhesion capabilities and emphasizing its significance in bacterial virulence. [94]. The SagA gene, situated in a gene cluster involved in cell wall metabolism alongside MreCD proteins, encodes a protein sharing sequence homology with cell wall metabolism-related proteins from other bacteria, suggesting its potential involvement in cell wall metabolism. However, its hydrolase activity and structural characteristics differ from those of related proteins [94,95]. Some strains of E. faecalis produce a post-translationally modified antimicrobial peptide called cytolysin, capable of lysing both bacterial and eukaryotic cells, thereby contributing to pathogenesis [96]. Xiong et al. discovered two previously uncharacterized small β-barrel Pore Forming Toxins (PFTs), resembling members of the haemolysin family found in human samples, and identified them as part of a broader family of β-barrel PFTs in E. faecalis, E. faecium, and E. hirae. The most potent among these toxins exhibit binding affinity to HLA-I in humans and MHC-I in other animals, establishing them as a significant family of canonical protein toxins in enterococci [97]. Enterococcus virulence factors, prevalent in clinical isolates, include the carbohydrate phosphotransferase system (PTS), enabling glucose and alternative carbon nutrient uptake [98]. Regulatory proteins termed PTS-regulatory-domain-containing virulence regulators (PCVRs), phosphorylated via the PTS, are vital for bacterial adaptation to hostile and nutrient-limiting host environments [99]. Studies on PCVRs, such as MafR from E. faecalis, reveal that loss of MafR significantly diminishes virulence in a murine peritonitis model, suggesting a link between MafR and E. faecalis virulence [100]. Additionally, the biofilm and endocarditis-associated permease A (BepA) gene, part of PTS and highly expressed in E. faecium hospital-associated isolates [101] has been associated with endocarditis in a rat model. It has been demonstrated that BepA contributes to in vitro biofilm formation of E. faecium in the presence of human serum and to the metabolism of β-methyl-D-glucoside [102]. Figure 4 illustrates the main E. faecium and E. faecalis virulence factors. Additionally, Table 1 summarizes their biological characteristics and actions.

5. Interactions in Immunocompromised Host

The immune system is a complex network of molecules, cells, tissues, and organs that plays a crucial role in recognizing and eliminating foreign agents while maintaining a stable internal environment [121,122,123]. In individuals with intact immune systems, the detection of Enterococcus typically activates the host’s immune response, leading to the recruitment of immune cells like neutrophils and macrophages at the infection site for bacterial elimination, accompanied by the release of cytokines, chemokines, and antimicrobial peptides to combat the infection [121]. However, individuals with compromised immune systems, such as those undergoing immunosuppressive therapy or with certain underlying conditions, are more susceptible to Enterococcus faecium and Enterococcus faecalis infections [124,125,126,127]. Immunocompromised individuals are at an elevated risk of developing drug-resistant enterococcal infections, primarily because they frequently receive broad-spectrum antibiotics in hospital settings where resistant strains are prevalent [31,128]. Patients undergoing treatments like chemotherapy and hematopoietic stem cell transplantation face a heightened susceptibility due to their compromised immune systems, which include reduced innate immunity and weakened mucosal barriers [129]. These factors make it easier for multidrug-resistant organisms like VRE to colonize the skin and intestinal tract, eventually leading to infections [130]. The use of central venous catheters in hospitals provides an additional pathway for nosocomial pathogens to enter the bloodstream during hospitalization. When bacteria enter the bloodstream, they encounter different conditions compared to those in the skin or intestines. These conditions include altered nutrient availability, varying host immune responses, and fluctuating antibiotic concentrations [131]. Such factors create selective pressures that favour the growth of bacterial variants better suited for survival and proliferation in the bloodstream [132]. Previous studies involving immunocompromised patients revealed that VRE adapted to the bloodstream environment by activating a bacterial stringent response. This adaptation rendered antibiotics less effective in treating the infection, and resolution only occurred after immune function was restored through a granulocyte infusion [13,120].

5.1. Clinical Risk-Factors and Predictors of Mortality in Host

The correlation between VRE colonization or infection and specific pre-existing medical conditions such as diabetes mellitus, chronic renal failure, cancer, and transplantation is widely recognized [133,134,135,136,137,138,139]. Hospitalization in wards predominantly treating patients with hematological diseases is also linked to VRE colonization. This connection is compounded by the extensive use of broad-spectrum antibiotics in these wards, resulting in heightened colonization pressure and increased VRE transmission [133,140]. The presence of an invasive device has previously been identified as a significant clinical risk factor for VRE invasive infections [134]. Invasive procedures such as catheter insertion, ventilation, and surgery increase the risk by providing entry routes for Enterococcus, which can form biofilms on medical devices, or merely serve as indicators of debilitation, prolonged hospital stays, and severe comorbidities [141,142,143,144]. Patients with hematological conditions and invasive devices, undergoing prolonged antibiotic therapy in the same ward, may have acted as VRE reservoirs. Previous studies emphasize the significance of close proximity to culture-positive patients for VRE spread, such as Byers et al., that highlighted the proximity to non-isolated, colonized patients as a key risk factor for VRE acquisition [145]. Moreover, they suggested that while antibiotic exposure is crucial, it may not be adequate alone to yield a positive VRE culture in patients without prior VRE exposure.
Numerous studies have explored whether prior antimicrobial therapy serves as a risk factor for nosocomial VRE, with divergent results [146]. The main agents reported include vancomycin, cephalosporins, and antimicrobials targeting anaerobic organisms [141,147,148,149,150,151,152]. Continuous vancomycin administration has been proposed to create an intestinal niche conducive to VRE proliferation, potentially heightening the risk of VRE bloodstream invasion [149]. Glycopeptide usage might facilitate VRE carriers in transitioning to transmitters rather than heightening the likelihood of non-carriers becoming colonized [146]. Sakka et al. outline link between exposure to antimicrobial agents with anti-anaerobic properties and VRE colonization [140]. These medications potentially enhance VRE colonization in the lower gastrointestinal tract by suppressing anaerobic flora in the gut [136,147]. Consequently, individuals exposed to such agents are at an increased risk of VRE bloodstream infections. Therefore, it is suggested that restricting the use of antimicrobial agents with anti-anaerobic activity could aid in reducing the transmission and spread of VRE [21]. Hospitalization in a medical ward or admission to the ICU within the past 6 months are additional risk factors associated with VRE colonization in high-risk environments [134,153]. Research indicates that VRE colonization is often found in patients who later develop bacteremia. Moreover, in studies with immunocompromised patients, VRE colonization demonstrated a high negative predictive value (99.9%) and a positive predictive value of 29.3% for predicting the development of bacteremia [135]. VRE bacteremia can arise from prolonged gastrointestinal colonization, often occurring as an isolated breakthrough event amidst intense antimicrobial therapy. Invasive VRE infections are linked to significant morbidity and mortality rates [12,154]. Studies have previously linked the elevated crude mortality rate seen in VRE-infected patients to the severity of underlying illness [155].

5.2. Enterococcus Pathogenicity in Host Cells

The discovery of a widely distributed clonal lineage of the virulent E. faecium strain (CC-17), adapted to hospital environments, has revealed the swift dissemination of resistant strains within healthcare settings. The acquisition of ampicillin resistance and the putative esp pathogenicity island by E. faecium have bolstered its capacity to thrive in hospitals, enabling transmission and contributing to nosocomial outbreaks [40]. Comparative and functional genomics may detect indicators of VRE adaptation during colonization of the gastrointestinal tract and bloodstream infection in immunocompromised individuals [156]. Recent research on VRE adaptation to the human intestinal tract has also detected changes in carbohydrate metabolism during extended colonization periods [157]. Chilambi et al. report that a usual adjustment in carbohydrate utilization was related to the independent occurrence of a Y585C mutation in the sorbitol operon transcriptional regulator gutR across various STs [120]. The gut operon is more prevalent among hospital-adapted E. faecium isolates than commensal ones [156]. Therefore, sorbitol exposure, due to its use as a dietary sugar substitute and laxative in chemotherapy patients, might have contributed to the emergence and prevalence of the gutR Y585C mutation in VRE strains, a phenomenon observed in immunocompromised settings.
Typically, researchers identify common trends in how VRE adapts across various genetic backgrounds and establish connections between these genetic alterations and clinically differences in traits [40,62]. In particular, the discovery of significant genetic rearrangements, such as chromosomal inversions exceeding 1 Mb in size and similar genomic alterations, have been observed previously in the genomes of E. faecium associated with hospital environments [158,159]. However, the exact role and mechanisms through which these rearrangements may impact the bacterium’s ability to survive in a hospital setting still require further investigation.
Several studies demonstrated the ability of Enterococcus to influence multiple signaling pathways within host cells. Boonanantanasarn et al. found that E. faecalis induces EGFR activation in oral cancer cells through hydrogen peroxide (H2O2) production, either directly by the bacteria or via metalloproteinase-dependent EGF-like signals [160]. EGFR activation can significantly contribute to MEK/ERK activation, promoting cell proliferation [161,162,163]. The ability of gefitinib and catalase to attenuate E. faecalis-induced EGFR activation and cell proliferation, along with the inhibition of TGF-α activation by these inhibitors, suggests potential clinical applications for catalase and EGFR inhibitors in treating E. faecalis-induced oral carcinogenesis [160]. In experiments with HCT-116, an aggressive colorectal cancer cell line, E. faecalis was found to downregulate the expression of the FIAF gene (angiopoietin-like protein 4), which is typically associated with the development of certain cancer types [164,165]. In a mouse model of ulcerative colitis, an increase in E. faecalis colonization following vinegar treatment was associated with inflammation inhibition by suppressing T helper (Th)-1 and Th17 responses [166]. Additionally, in studies using human peripheral blood mononuclear cells, the heat-killed E. faecalis YM-73 strain exhibited immunomodulatory effects, increasing Th1-associated cytokines while reducing Th2-associated cytokines [167]. Wang et al. showed that E. faecalis Lipoteichoic Acid (LTA) induces the expression of the pro-inflammatory cytokine TNF-α by activating the p38 MAPK and NF-κB signaling pathways in differentiated THP-1 macrophages [168]. In addition, some studies have explored the potential involvement of miRNAs in Enterococcus infections. For instance, Li et al. investigated the role of miR-200a-3p in BMSC migration stimulated by E. faecalis supernatants, along with its downstream target FOXJ1. They also identified activation of the NFκB pathway, which promoted migration by upregulating MMP-3 and MMP-13 expressions. These findings offer a new perspective on the potential role of miRNAs in Enterococcus-host cell interactions [169]. Nevertheless, further research is needed to uncover the specific mechanisms and implications of miRNA regulation in Enterococcus infections, as well as to elucidate the broader role of miRNAs in the interplay between bacteria and host cells. In conclusion, all these pathways play critical roles in diverse cellular processes such as cell proliferation, survival, and inflammation. However, it’s likely that enterococci exert influence over additional signalling pathways that have yet to be elucidated, highlighting the complexity of their pathogenicity.

5.3. Biofilm Formation and Adhesion to Immunocompromised Host Tissues

Enterococcus faecalis is well-known for its ability to form biofilms [170,171]. Biofilms are structured communities of bacteria enclosed in a self-produced matrix. Biofilm formation involves the production of extracellular polymeric substances (EPS), which provide structural support and protect bacteria from host immune defenses and antimicrobial agents [172]. Several studies examined alterations in genes known to play a role in biofilm formation in E. faecium, such as ebpABC (biofilm-associated pilus), esp (enterococcal surface protein), asrR (antibiotic stress and response regulator), acm (collagen adhesion protein), and atlA (autolysin) [173]. Within biofilms, Enterococcus faecalis can attach to surfaces, such as medical devices or damaged tissues, shielding host immune responses and antibiotics. This capability makes biofilm-related infections challenging to eradicate in several clinical settings [174,175]. An interesting study by Fiorotto et al. highlighted how Enterococcus can interact with components of the host’s extracellular matrix, such as collagen and fibronectin, in an organoid model [176]. Therefore the interactions promote bacterial adhesion to host tissues and may contribute to the formation of biofilms [87,177,178]. The understanding of capsular polysaccharide (cps) in E. faecium is presently limited, with minimal knowledge extending beyond a potential capsule biosynthetic locus, which exhibits variability among different sequence types [37]. The literature reported that mutations in the cps locus may result in production changes of cell surface-associated polysaccharides. These changes could lead to increased biofilm formation, enhancing the colonization or persistence of VRE isolates carrying these mutations. Biofilm formation, in turn, amplifies bacterial infectivity and disease-causing potential by enhancing adhesion to host tissues and medical devices like catheters, increasing resistance to phagocytosis, and bolstering resilience against antibiotics [179,180]. Furthermore, the composition of enterococcal cell surface-associated polysaccharides has been demonstrated to influence bacterial sensitivity to lysozyme [181]. Lysozyme is a crucial component of the innate immune response. Lysozyme acts as a hydrolase, degrading the glycosidic bonds between N-acetylmuramic acid (NAM) and N-acetylglucosamide (NAG) in peptidoglycan in the bacterial cell wall, resulting in increased cell wall permeability and ultimately cell death [182]. The deletion of RasP (CdRasP) and RseP (EfsRseP), the site-2-metalloproteases of respectively E. faecalis and C. difficile, results in a decreased activity of σV, which is the extracytoplasmic function sigma factor which mediates the lysozyme resistance. This activity reduction consequently leads to a decreased lysozyme resistance [183,184,185]. Similar observations have been made in Enterococcus faecium, where mutations within RseP results in a 6–8-fold increase in lysozyme susceptibility as reductions in desiccation tolerance [183,186]. Therefore, the ability to tolerate constitutive innate immune system defenses such as lysozyme could have also played a role in selecting about capsule-lacking, biofilm-forming bacteria [183,187].

6. Therapeutical Approaches

6.1. Treatment for Susceptible Enterococci

Susceptible enterococcal strains commonly respond to β-lactam and aminoglycoside combinations. In order to reduce aminoglycoside side effects such as nephrotoxicity and ototoxicity, clinical guidelines also propose a double β-lactams therapy [188]. The synergic effect between amoxicillin and cefotaxime has been extensively demonstrated, along with the minimum inhibitory concentration (MIC) decrease in the case of simultaneous amoxicillin and ceftriaxone usage. Although Enterococcus intrinsic cephalosporins resistance, the association with ampicillin leads to penicillin-binding proteins (PBPs) inactivation, justifying the success of the combination. Specifically, several endocarditis episodes showed a reduced vegetation after this therapeutic regimen [189].

6.2. Challenges in Treatment of VRE Infection

VRE present challenges in treatment, particularly as resistance often emerges in strains of E. faecium already MDR [190]. These strains typically exhibit resistance to multiple antimicrobial drugs, including erythromycin, tetracycline, fluoroquinolones, and rifampicin, and do not respond synergistically to aminoglycosides [191]. Determining optimal therapy for infections caused by such strains remains uncertain. Physicians may resort to administering drugs or combinations with proven in vitro activity. However, discerning whether specific therapy is necessary for enterococcal isolates or assessing the infection’s role in the patient’s outcome relative to underlying disease can be challenging. Evaluating antimicrobial therapy efficacy for VRE infection is complicated by underlying disease severity and concurrent interventions. Nevertheless, literature suggests several antimicrobial drugs may offer benefits to patients infected with VRE, based on in vitro studies, animal research, anecdotal case reports, and small, uncontrolled series over the past decades [192,193,194,195,196]. The resistance mechanisms require new therapeutical strategies, which could be limited and related to possible complications [197,198]. VRE infections result in increased morbidity, mortality, and medical costs, necessitating a comprehensive treatment strategy involving all infection control professionals [199]. Certainly, the advent of VRE strains highlights the pressing need for brand-new antimicrobial drugs [200,201]. Treatment options for VRE infections mainly include linezolid as the only FDA-approved therapeutical choice. Off-label therapeutical strategies may involve daptomycin, often combined with fosfomycin or β-lactams, oritavancin, tigecycline, and tedizolid. All these agents are not approved yet in the case of VRE isolations due to the lack of extended clinical studies about their effectiveness. According to literature data, high daptomycin dosage may be bactericidal against VRE. However, some E. faecium isolates tend to survive those regimens, requiring a daptomycin combination with β-lactam antibiotics [202]. The daptomycin bactericidal activity is enhanced by the addition of carbapenems such as ertapenem or cephalosporins such as cefepime, ceftriaxone, and cefazolin. Moreover, the association of ceftaroline and daptomycin demonstrated high success percentage in E. faecium and E. faecalis infection treatment [203]. Previous studies have considered the role of tigecycline in VRE infection treatment, documenting an activity in the case of soft tissue and intra-abdominal infection episodes. However, further clinical trials will be essential to optimize the tigecycline regimen for those complicated Enterococcus spp. Infections [204].
Quinupristin/dalfopristin may represent a valid alternative against E. faecium due to E. faecalis intrinsic resistance to this double streptogramins combination [205]. Regarding new cephalosporins, ceftaroline and ceftobiprole demonstrated significant in vitro against the most common agents of infective endocarditis, including Enterococcus faecalis [206].

6.3. Prevention and Infection Control Strategies

Enterococcal antimicrobial resistance and virulence underscore the importance of controlling these microorganisms’ diffusion, especially among the hospital recovered patients. Reccomandation for controlling VRE involve isolating individuals with current or past VRE colonization, using protective gown and gloves, hand hygiene with antiseptic post glove removal, and allocating dedicated equipment like stethoscopes for VRE-colonized patient rooms [207]. Additionally, precautions should be taken to avoid touching environmental surfaces (e.g., doorknobs) after glove removal. Enhanced environmental cleaning methods may be recommended due to documented instances of persistent VRE presence in hospital rooms despite standard cleaning procedures [207,208]. Recent studies have demonstrated that decolonization through the use of chlorhexidine gluconate bathing for patients in Intensive Care Units (ICU) can prevent hospital-acquired infections and reduce the acquisition rate of MDR organisms including VRE [209]; this could be a potentially interesting path to follow for wards characterized by the presence of immunocompromised patients. Published data highlight the possibility of using capsular and cell wall polysaccharides as targets for immunotherapeutic choices. Specifically, these carbohydrates could be used as targets for antibodies, contributing to E. faecalis and E. faecium killing. Furthermore, the isolation of polysaccharides from enterococcal-grown colonies has been experienced in vaccine development and production [210]. The European Antimicrobial Resistance Surveillance Network (EARS-Net) currently include E. faecalis and E. faecium within its alert pathogens list. This inclusion in the network encourages the microbiology laboratories in furnishing identification and precise susceptibility profiles [211]. VRE poses a significant threat in healthcare settings, warranting the implementation of antimicrobial stewardship programs. Enhanced surveillance and screening have been effective in identifying colonized individuals and implementing necessary preventive measures [212,213,214]. Several studies underscore the importance of active rectal swab screening for detecting VRE strains. In ICU settings, surveillance swabs aid in patient monitoring, reducing VRE transmission to sterile sites. A positive VRE result might forecast glycopeptide resistance in subsequent infections [215,216,217]. We advocate for stringent surveillance, particularly in wards with immunocompromised patients.

7. Conclusions

Enterococci are significant human pathogens with diverse characteristics and clinical implications. Understanding the intricate interactions between Enterococcus and immunocompromised individuals is crucial for effective infection management. These patients, highly susceptible to Enterococcus infections, including drug-resistant strains like VRE, require enhanced surveillance and strict infection control measures, particularly in high-risk settings such as haematology, ICUs, and other wards with immunocompromised patients. Identifying clinical risk factors and predictors of mortality associated with VRE is vital for early intervention and optimal patient outcomes. Although therapeutic approaches for VRE infections present challenges, ongoing research aims to develop new strategies and treatments. Prevention and control strategies, including antimicrobial stewardship programs and environmental cleaning, are essential to curb VRE spread and reduce infection burden in healthcare settings. Despite continuous research efforts, it is necessary to address the complex challenges posed by Enterococcus infections. Our hope is to utilize these findings to develop more efficient approaches for managing and containing enterococcal infections in susceptible patient populations.

Author Contributions

Conceptualization: G.S., M.C. and G.M.; Methodology: G.S., M.C. and G.M.; Writing—original draft preparation: G.S., M.C. and G.M.; Writing—review and editing: G.S., M.C., G.M. and S.S.; Supervision: S.S. and F.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was partially supported by EU funding with the MUR PNRR Extended Partnership Initiative of Emerging Infection Disease Project n. PE 00000007 INF-ACT.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lebreton, F.; Manson, A.L.; Saavedra, J.T.; Straub, T.J.; Earl, A.M.; Gilmore, M.S. Tracing the Enterococci from Paleozoic Origins to the Hospital. Cell 2017, 169, 849–861.e13. [Google Scholar] [CrossRef] [PubMed]
  2. Lupia, T.; Roberto, G.; Scaglione, L.; Shbaklo, N.; De Benedetto, I.; Scabini, S.; Mornese Pinna, S.; Curtoni, A.; Cavallo, R.; De Rosa, F.G.; et al. Clinical and microbiological characteristics of bloodstream infections caused by Enterococcus spp. within internal medicine wards: A two-year single-centre experience. Intern. Emerg. Med. 2022, 17, 1129–1137. [Google Scholar] [CrossRef] [PubMed]
  3. Ha, S.; Huh, K.; Chung, D.R.; Ko, J.-H.; Cho, S.Y.; Huh, H.J.; Lee, N.Y.; Kang, C.-I.; Peck, K.R.; Song, J.-H.; et al. Efficacy of teicoplanin in bloodstream infections caused by Enterococcus faecium: Posthoc analysis of a nationwide surveillance. Int. J. Infect. Dis. 2022, 122, 506–513. [Google Scholar] [CrossRef] [PubMed]
  4. Berge, A.; Morenius, C.; Petropoulos, A.; Nilson, B.; Rasmussen, M. Epidemiology, bacteriology, and clinical characteristics of HACEK bacteremia and endocarditis: A population-based retrospective study. Eur. J. Clin. Microbiol. Infect. Dis. 2021, 40, 525–534. [Google Scholar] [CrossRef]
  5. Shrestha, S.; Kharel, S.; Homagain, S.; Aryal, R.; Mishra, S.K. Prevalence of vancomycin-resistant enterococci in Asia–A systematic review and meta-analysis. J. Clin. Pharm. Ther. 2021, 46, 1226–1237. [Google Scholar] [CrossRef]
  6. Ruksakiet, K.; Hanák, L.; Farkas, N.; Hegyi, P.; Sadaeng, W.; Czumbel, L.M.; Sang-Ngoen, T.; Garami, A.; Mikó, A.; Varga, G.; et al. Antimicrobial Efficacy of Chlorhexidine and Sodium Hypochlorite in Root Canal Disinfection: A Systematic Review and Meta-analysis of Randomized Controlled Trials. J. Endod. 2020, 46, 1032–1041.e7. [Google Scholar] [CrossRef] [PubMed]
  7. Rathod, S.N.; Bardowski, L.; Tse, I.; Churyla, A.; Fiehler, M.; Malczynski, M.; Qi, C.; Tanna, S.D.; Bulger, C.; Al-Qamari, A.; et al. Vancomycin-resistant Enterococcus outbreak in a pre- and post-cardiothoracic transplant population: Impact of discontinuing multidrug-resistant organism surveillance during the coronavirus disease 2019 pandemic. Transpl. Infect. Dis. 2022, 24, e13972. [Google Scholar] [CrossRef] [PubMed]
  8. Landete, J.M.; Peirotén, Á.; Medina, M.; Arqués, J.L.; Rodríguez-Mínguez, E. Virulence and Antibiotic Resistance of Enterococci Isolated from Healthy Breastfed Infants. Microb. Drug Resist. 2018, 24, 63–69. [Google Scholar] [CrossRef] [PubMed]
  9. Torres, C.; Alonso, C.A.; Ruiz-Ripa, L.; León-Sampedro, R.; Del Campo, R.; Coque, T.M. Antimicrobial Resistance in Enterococcus spp. of animal origin. Microbiol. Spectr. 2018, 6, 185–227. [Google Scholar] [CrossRef]
  10. World Health Organization. Antimicrobial Resistance Surveillance in Europe 2022–2020 Data; World Health Organization, Regional Office for Europe: Copenhagen, Denmark, 2022; ISBN 92-890-5668-1. [Google Scholar]
  11. Poudel, A.N.; Zhu, S.; Cooper, N.; Little, P.; Tarrant, C.; Hickman, M.; Yao, G. The economic burden of antibiotic resistance: A systematic review and meta-analysis. PLoS ONE 2023, 18, e0285170. [Google Scholar] [CrossRef]
  12. DiazGranados, C.A.; Zimmer, S.M.; Klein, M.; Jernigan, J.A. Comparison of mortality associated with vancomycin-resistant and vancomycin-susceptible enterococcal bloodstream infections: A meta-analysis. Clin. Infect. Dis. 2005, 41, 327–333. [Google Scholar] [CrossRef]
  13. Honsa, E.S.; Cooper, V.S.; Mhaissen, M.N.; Frank, M.; Shaker, J.; Iverson, A.; Rubnitz, J.; Hayden, R.T.; Lee, R.E.; Rock, C.O.; et al. RelA Mutant Enterococcus faecium with Multiantibiotic Tolerance Arising in an Immunocompromised Host. mBio 2017, 8, e02124-16. [Google Scholar] [CrossRef]
  14. Krawczyk, B.; Wityk, P.; Gałęcka, M.; Michalik, M. The Many Faces of Enterococcus spp.—Commensal, Probiotic and Opportunistic Pathogen. Microorganisms 2021, 9, 1900. [Google Scholar] [CrossRef]
  15. Ghazisaeedi, F.; Meens, J.; Hansche, B.; Maurischat, S.; Schwerk, P.; Goethe, R.; Wieler, L.H.; Fulde, M.; Tedin, K. A virulence factor as a therapeutic: The probiotic Enterococcus faecium SF68 arginine deiminase inhibits innate immune signaling pathways. Gut Microbes 2022, 14, 2106105. [Google Scholar] [CrossRef]
  16. Keogh, D.; Lam, L.N.; Doyle, L.E.; Matysik, A.; Pavagadhi, S.; Umashankar, S.; Low, P.M.; Dale, J.L.; Song, Y.; Ng, S.P.; et al. Extracellular Electron Transfer Powers Enterococcus faecalis Biofilm Metabolism. mBio 2018, 9, e00626-17. [Google Scholar] [CrossRef]
  17. Brinkwirth, S.; Ayobami, O.; Eckmanns, T.; Markwart, R. Hospital-acquired infections caused by enterococci: A systematic review and meta-analysis, WHO European Region, 1 January 2010 to 4 February 2020. Eurosurveillance 2021, 26, 2001628. [Google Scholar] [CrossRef]
  18. Simar, S.R.; Tran, T.T.; Rydell, K.B.; Panesso, D.; Contreras, G.A.; Munita, J.M.; Cifuentes, R.O.; Abbo, L.M.; Sahasrabhojane, P.; Dinh, A.Q.; et al. Multisite Detection of Tn1549-Mediated vanB Vancomycin Resistance in Multidrug-Resistant Enterococcus faecalis ST6 in Texas and Florida. Antimicrob. Agents Chemother. 2023, 67, e0128422. [Google Scholar] [CrossRef]
  19. Yuan, T.-Y.; Liang, B.; Jiang, B.-W.; Sun, S.-W.; Zhou, Y.-F.; Zhu, L.-W.; Liu, J.; Guo, X.-J.; Ji, X.; Sun, Y. Virulence genes and antimicrobial resistance in Enterococcus strains isolated from dogs and cats in Northeast China. J. Vet. Med. Sci. 2023, 85, 371–378. [Google Scholar] [CrossRef]
  20. Lee, Y.J.; Kim, K.; Lee, Y.J. Dissemination and characteristics of high-level erythromycin-resistant Enterococcus faecalis from bulk tank milk of dairy companies in Korea. Can. J. Vet. Res. Rev. 2023, 87, 51–58. [Google Scholar]
  21. Nüesch-Inderbinen, M.; Heyvaert, L.; Treier, A.; Zurfluh, K.; Cernela, N.; Biggel, M.; Stephan, R. High occurrence of Enterococcus faecalis, Enterococcus faecium, and Vagococcus lutrae harbouring oxazolidinone resistance genes in raw meat-based diets for companion animals—A public health issue, Switzerland, September 2018 to May 2020. Eurosurveillance 2023, 28, 2200496. [Google Scholar] [CrossRef] [PubMed]
  22. Mwikuma, G.; Kainga, H.; Kallu, S.A.; Nakajima, C.; Suzuki, Y.; Hang’ombe, B.M. Determination of the Prevalence and Antimicrobial Resistance of Enterococcus faecalis and Enterococcus faecium Associated with Poultry in Four Districts in Zambia. Antibiotics 2023, 12, 657. [Google Scholar] [CrossRef] [PubMed]
  23. Surveillance Atlas of Infectious Diseases. Available online: http://atlas.ecdc.europa.eu/public/index.aspx (accessed on 11 June 2023).
  24. Top, J.; Willems, R.; van der Velden, S.; Asbroek, M.; Bonten, M. Emergence of clonal complex 17 Enterococcus faecium in The Netherlands. J. Clin. Microbiol. 2008, 46, 214–219. [Google Scholar] [CrossRef]
  25. Werner, G.; Coque, T.M.; Hammerum, A.M.; Hope, R.; Hryniewicz, W.; Johnson, A.; Klare, I.; Kristinsson, K.G.; Leclercq, R.; Lester, C.H.; et al. Emergence and spread of vancomycin resistance among enterococci in Europe. Eurosurveillance 2008, 13, 19046. [Google Scholar] [CrossRef]
  26. Soderblom, T.; Aspevall, O.; Erntell, M.; Hedin, G.; Heimer, D.; Hokeberg, I.; Kidd-Ljunggren, K.; Melhus, A.; Olsson-Liljequist, B.; Sjogren, I.; et al. Alarming spread of vancomycin resistant enterococci in Sweden since 2007. Eurosurveillance 2010, 15, 19620. [Google Scholar] [CrossRef]
  27. Rete dei Laboratori di Microbiologia (Rete MIC) per la Sorveglianza Delle Resistenze Batteriche. Qualità Sicilia SSR. Available online: https://www.qualitasiciliassr.it/?q=rete-laboratori (accessed on 11 June 2023).
  28. Toc, D.A.; Botan, A.; Botescu, A.M.C.; Brata, V.D.; Colosi, I.A.; Costache, C.; Junie, L.M. A Tale of Two Pandemics: Antimicrobial Resistance Patterns of Enterococcus spp. in COVID-19 Era. Antibiotics 2023, 12, 312. [Google Scholar] [CrossRef] [PubMed]
  29. García-Solache, M.; Rice, L.B. The Enterococcus: A Model of Adaptability to Its Environment. Clin. Microbiol. Rev. 2019, 32, e00058-18. [Google Scholar] [CrossRef] [PubMed]
  30. Donskey, C.J.; Chowdhry, T.K.; Hecker, M.T.; Hoyen, C.K.; Hanrahan, J.A.; Hujer, A.M.; Hutton-Thomas, R.A.; Whalen, C.C.; Bonomo, R.A.; Rice, L.B. Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. N. Engl. J. Med. 2000, 343, 1925–1932. [Google Scholar] [CrossRef]
  31. Ubeda, C.; Taur, Y.; Jenq, R.R.; Equinda, M.J.; Son, T.; Samstein, M.; Viale, A.; Socci, N.D.; van den Brink, M.R.M.; Kamboj, M.; et al. Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J. Clin. Investig. 2010, 120, 4332–4341. [Google Scholar] [CrossRef]
  32. Brandl, K.; Plitas, G.; Mihu, C.N.; Ubeda, C.; Jia, T.; Fleisher, M.; Schnabl, B.; DeMatteo, R.P.; Pamer, E.G. Vancomycin-resistant enterococci exploit antibiotic-induced innate immune deficits. Nature 2008, 455, 804–807. [Google Scholar] [CrossRef] [PubMed]
  33. Kinnebrew, M.A.; Ubeda, C.; Zenewicz, L.A.; Smith, N.; Flavell, R.A.; Pamer, E.G. Bacterial flagellin stimulates Toll-like receptor 5-dependent defense against vancomycin-resistant Enterococcus infection. J. Infect. Dis. 2010, 201, 534–543. [Google Scholar] [CrossRef]
  34. Sydnor, E.R.M.; Perl, T.M. Hospital epidemiology and infection control in acute-care settings. Clin. Microbiol. Rev. 2011, 24, 141–173. [Google Scholar] [CrossRef]
  35. Bradley, C.R.; Fraise, A.P. Heat and chemical resistance of enterococci. J. Hosp. Infect. 1996, 34, 191–196. [Google Scholar] [CrossRef]
  36. Gilmore, M.S.; Lebreton, F.; van Schaik, W. Genomic transition of enterococci from gut commensals to leading causes of multidrug-resistant hospital infection in the antibiotic era. Curr. Opin. Microbiol. 2013, 16, 10–16. [Google Scholar] [CrossRef]
  37. Palmer, K.L.; Godfrey, P.; Griggs, A.; Kos, V.N.; Zucker, J.; Desjardins, C.; Cerqueira, G.; Gevers, D.; Walker, S.; Wortman, J.; et al. Comparative genomics of enterococci: Variation in Enterococcus faecalis, clade structure in E. faecium, and defining characteristics of E. gallinarum and E. casseliflavus. mBio 2012, 3, e00318-11. [Google Scholar] [CrossRef]
  38. Homan, W.L.; Tribe, D.; Poznanski, S.; Li, M.; Hogg, G.; Spalburg, E.; Van Embden, J.D.A.; Willems, R.J.L. Multilocus sequence typing scheme for Enterococcus faecium. J. Clin. Microbiol. 2002, 40, 1963–1971. [Google Scholar] [CrossRef]
  39. Ruiz-Garbajosa, P.; Bonten, M.J.M.; Robinson, D.A.; Top, J.; Nallapareddy, S.R.; Torres, C.; Coque, T.M.; Cantón, R.; Baquero, F.; Murray, B.E.; et al. Multilocus sequence typing scheme for Enterococcus faecalis reveals hospital-adapted genetic complexes in a background of high rates of recombination. J. Clin. Microbiol. 2006, 44, 2220–2228. [Google Scholar] [CrossRef]
  40. Willems, R.J.L.; Top, J.; van Santen, M.; Robinson, D.A.; Coque, T.M.; Baquero, F.; Grundmann, H.; Bonten, M.J.M. Global spread of vancomycin-resistant Enterococcus faecium from distinct nosocomial genetic complex. Emerg. Infect. Dis. 2005, 11, 821–828. [Google Scholar] [CrossRef]
  41. van Hal, S.J.; Willems, R.J.L.; Gouliouris, T.; Ballard, S.A.; Coque, T.M.; Hammerum, A.M.; Hegstad, K.; Pinholt, M.; Howden, B.P.; Malhotra-Kumar, S.; et al. The interplay between community and hospital Enterococcus faecium clones within health-care settings: A genomic analysis. Lancet Microbe 2022, 3, e133–e141. [Google Scholar] [CrossRef]
  42. Al Rubaye, M.; Janice, J.; Bjørnholt, J.V.; Kacelnik, O.; Haldorsen, B.C.; Nygaard, R.M.; Hegstad, J.; Sundsfjord, A.; Hegstad, K.; The Norwegian Vre Study Group. The population structure of vancomycin-resistant and -susceptible Enterococcus faecium in a low-prevalence antimicrobial resistance setting is highly influenced by circulating global hospital-associated clones. Microb. Genom. 2023, 9, 001160. [Google Scholar] [CrossRef]
  43. Laverde Gomez, J.A.; van Schaik, W.; Freitas, A.R.; Coque, T.M.; Weaver, K.E.; Francia, M.V.; Witte, W.; Werner, G. A multiresistance megaplasmid pLG1 bearing a hylEfm genomic island in hospital Enterococcus faecium isolates. Int. J. Med. Microbiol. 2011, 301, 165–175. [Google Scholar] [CrossRef]
  44. Manson, J.M.; Hancock, L.E.; Gilmore, M.S. Mechanism of chromosomal transfer of Enterococcus faecalis pathogenicity island, capsule, antimicrobial resistance, and other traits. Proc. Natl. Acad. Sci. USA 2010, 107, 12269–12274. [Google Scholar] [CrossRef] [PubMed]
  45. Gholizadeh, P.; Aghazadeh, M.; Ghotaslou, R.; Rezaee, M.A.; Pirzadeh, T.; Cui, L.; Watanabe, S.; Feizi, H.; Kadkhoda, H.; Kafil, H.S. Role of CRISPR-Cas system on antibiotic resistance patterns of Enterococcus faecalis. Ann. Clin. Microbiol. Antimicrob. 2021, 20, 49. [Google Scholar] [CrossRef] [PubMed]
  46. Cabiltes, I.; Coghill, S.; Bowe, S.J.; Athan, E. Enterococcal bacteraemia “silent but deadly”: A population-based cohort study. Intern. Med. J. 2020, 50, 434–440. [Google Scholar] [CrossRef] [PubMed]
  47. Giacobbe, D.R.; Labate, L.; Tutino, S.; Baldi, F.; Russo, C.; Robba, C.; Ball, L.; Dettori, S.; Marchese, A.; Dentone, C.; et al. Enterococcal bloodstream infections in critically ill patients with COVID-19: A case series. Ann. Med. 2021, 53, 1779–1786. [Google Scholar] [CrossRef] [PubMed]
  48. Zasowski, E.J.; Claeys, K.C.; Lagnf, A.M.; Davis, S.L.; Rybak, M.J. Time Is of the Essence: The Impact of Delayed Antibiotic Therapy on Patient Outcomes in Hospital-Onset Enterococcal Bloodstream Infections. Clin. Infect. Dis. 2016, 62, 1242–1250. [Google Scholar] [CrossRef]
  49. Lee, R.A.; Vo, D.T.; Zurko, J.C.; Griffin, R.L.; Rodriguez, J.M.; Camins, B.C. Infectious Diseases Consultation Is Associated with Decreased Mortality in Enterococcal Bloodstream Infections. Open Forum Infect. Dis. 2020, 7, ofaa064. [Google Scholar] [CrossRef] [PubMed]
  50. Cattaneo, C.; Rieg, S.; Schwarzer, G.; Müller, M.C.; Blümel, B.; Kern, W.V. Enterococcus faecalis bloodstream infection: Does infectious disease specialist consultation make a difference? Infection 2021, 49, 1289–1297. [Google Scholar] [CrossRef] [PubMed]
  51. Carvalho, A.S.; Lagana, D.; Catford, J.; Shaw, D.; Bak, N. Bloodstream infections in neutropenic patients with haematological malignancies. Infect. Dis. Health 2020, 25, 22–29. [Google Scholar] [CrossRef] [PubMed]
  52. Bussini, L.; Rosselli Del Turco, E.; Pasquini, Z.; Scolz, K.; Amedeo, A.; Beci, G.; Giglia, M.; Tedeschi, S.; Pascale, R.; Ambretti, S.; et al. Risk factors for persistent enterococcal bacteraemia: A multicentre retrospective study. J. Glob. Antimicrob. Resist. 2022, 29, 386–389. [Google Scholar] [CrossRef]
  53. Heidrich, V.; Bruno, J.S.; Knebel, F.H.; de Molla, V.C.; Miranda-Silva, W.; Asprino, P.F.; Tucunduva, L.; Rocha, V.; Novis, Y.; Arrais-Rodrigues, C.; et al. Dental Biofilm Microbiota Dysbiosis Is Associated with the Risk of Acute Graft-Versus-Host Disease after Allogeneic Hematopoietic Stem Cell Transplantation. Front. Immunol. 2021, 12, 692225. [Google Scholar] [CrossRef]
  54. Feng, D.; Zhang, Y.-T.; Qiu, T.-T.; Xu, K.-L. Clinical Features of Infection within 100 days after Hematopoietic Stem Cell Transplantation in 313 patients with Hematologic Diseases. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2020, 28, 602–608. [Google Scholar] [CrossRef]
  55. Yan, C.-H.; Wang, Y.; Mo, X.-D.; Sun, Y.-Q.; Wang, F.-R.; Fu, H.-X.; Chen, Y.; Han, T.-T.; Kong, J.; Cheng, Y.-F.; et al. Incidence, Risk Factors, Microbiology and Outcomes of Pre-engraftment Bloodstream Infection After Haploidentical Hematopoietic Stem Cell Transplantation and Comparison with HLA-identical Sibling Transplantation. Clin. Infect. Dis. 2018, 67, S162–S173. [Google Scholar] [CrossRef]
  56. Friedman, G.; Stepensky, P.; Abu Ahmad, W.; Masarwa, R.; Temper, V.; Oster, Y.; Amit, S.; Averbuch, D. Enterococcal Bacteremia in Children with Malignancies and Following Hematopoietic Stem Cell Transplantation: A 15-Year Single-Center Experience. Pediatr. Infect. Dis. J. 2020, 39, 318–324. [Google Scholar] [CrossRef] [PubMed]
  57. Casewell, M.; Friis, C.; Marco, E.; McMullin, P.; Phillips, I. The European ban on growth-promoting antibiotics and emerging consequences for human and animal health. J. Antimicrob. Chemother. 2003, 52, 159–161. [Google Scholar] [CrossRef] [PubMed]
  58. Aun, E.; Kisand, V.; Laht, M.; Telling, K.; Kalmus, P.; Väli, Ü.; Brauer, A.; Remm, M.; Tenson, T. Molecular Characterization of Enterococcus Isolates from Different Sources in Estonia Reveals Potential Transmission of Resistance Genes Among Different Reservoirs. Front. Microbiol. 2021, 12, 601490. [Google Scholar] [CrossRef] [PubMed]
  59. Levitus, M.; Rewane, A.; Perera, T.B. Vancomycin-Resistant Enterococci. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  60. Xie, O.; Slavin, M.A.; Teh, B.W.; Bajel, A.; Douglas, A.P.; Worth, L.J. Epidemiology, treatment and outcomes of bloodstream infection due to vancomycin-resistant enterococci in cancer patients in a vanB endemic setting. BMC Infect. Dis. 2020, 20, 228. [Google Scholar] [CrossRef]
  61. Raza, T.; Ullah, S.R.; Mehmood, K.; Andleeb, S. Vancomycin resistant Enterococci: A brief review. J. Pak. Med. Assoc. 2018, 68, 768–772. [Google Scholar]
  62. Lee, T.; Pang, S.; Abraham, S.; Coombs, G.W. Antimicrobial-resistant CC17 Enterococcus faecium: The past, the present and the future. J. Glob. Antimicrob. Resist. 2019, 16, 36–47. [Google Scholar] [CrossRef]
  63. Hasman, H.; Clausen, P.T.L.C.; Kaya, H.; Hansen, F.; Knudsen, J.D.; Wang, M.; Holzknecht, B.J.; Samulioniené, J.; Røder, B.L.; Frimodt-Møller, N.; et al. LRE-Finder, a Web tool for detection of the 23S rRNA mutations and the optrA, cfr, cfr(B) and poxtA genes encoding linezolid resistance in enterococci from whole-genome sequences. J. Antimicrob. Chemother. 2019, 74, 1473–1476. [Google Scholar] [CrossRef]
  64. Rahim, S.; Pillai, S.K.; Gold, H.S.; Venkataraman, L.; Inglima, K.; Press, R.A. Linezolid-resistant, vancomycin-resistant Enterococcus faecium infection in patients without prior exposure to linezolid. Clin. Infect. Dis. 2003, 36, E146–E148. [Google Scholar] [CrossRef]
  65. Arias, C.A.; Panesso, D.; McGrath, D.M.; Qin, X.; Mojica, M.F.; Miller, C.; Diaz, L.; Tran, T.T.; Rincon, S.; Barbu, E.M.; et al. Genetic basis for in vivo daptomycin resistance in enterococci. N. Engl. J. Med. 2011, 365, 892–900. [Google Scholar] [CrossRef] [PubMed]
  66. Zeng, W.; Feng, L.; Qian, C.; Chen, T.; Wang, S.; Zhang, Y.; Zheng, X.; Wang, L.; Liu, S.; Zhou, T.; et al. Acquisition of Daptomycin Resistance by Enterococcus faecium Confers Collateral Sensitivity to Glycopeptides. Front. Microbiol. 2022, 13, 815600. [Google Scholar] [CrossRef]
  67. Pfaller, M.A.; Cormican, M.; Flamm, R.K.; Mendes, R.E.; Jones, R.N. Temporal and Geographic Variation in Antimicrobial Susceptibility and Resistance Patterns of Enterococci: Results From the SENTRY Antimicrobial Surveillance Program, 1997–2016. Open Forum Infect. Dis. 2019, 6, S54–S62. [Google Scholar] [CrossRef]
  68. Lazzaro, L.M.; Cassisi, M.; Stefani, S.; Campanile, F. Impact of PBP4 Alterations on β-Lactam Resistance and Ceftobiprole Non-Susceptibility Among Enterococcus faecalis Clinical Isolates. Front. Cell. Infect. Microbiol. 2021, 11, 816657. [Google Scholar] [CrossRef] [PubMed]
  69. Maitreya, A.; Qureshi, A. Genomic and phenotypic characterisation of Enterococcus mundtii AM_AQ_BC8 for its anti-biofilm, antimicrobial and probiotic potential. Arch. Microbiol. 2024, 206, 84. [Google Scholar] [CrossRef]
  70. Chavers, L.S.; Moser, S.A.; Benjamin, W.H.; Banks, S.E.; Steinhauer, J.R.; Smith, A.M.; Johnson, C.N.; Funkhouser, E.; Chavers, L.P.; Stamm, A.M.; et al. Vancomycin-resistant enterococci: 15 years and counting. J. Hosp. Infect. 2003, 53, 159–171. [Google Scholar] [CrossRef]
  71. Rince, A.; Flahaut, S.; Auffray, Y. Identification of general stress genes in Enterococcus faecalis. Int. J. Food Microbiol. 2000, 55, 87–91. [Google Scholar] [CrossRef]
  72. Dougherty, T.J.; Pucci, M.J.; Zawadzke, L.; Barrett, J.F. American Society of Microbiology-100th General Meeting. IDrugs Investig. Drugs J. 2000, 3, 857–860. [Google Scholar]
  73. Hancock, L.; Perego, M. Two-component signal transduction in Enterococcus faecalis. J. Bacteriol. 2002, 184, 5819–5825. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, J.; Fang, R.; Peng, Q.; Wu, S.; Lei, L. The Regulations of Essential WalRK Two-Component System on Enterococcus faecalis. J. Clin. Med. 2023, 12, 767. [Google Scholar] [CrossRef]
  75. Hancock, L.E.; Perego, M. The Enterococcus faecalis fsr two-component system controls biofilm development through production of gelatinase. J. Bacteriol. 2004, 186, 5629–5639. [Google Scholar] [CrossRef] [PubMed]
  76. Ali, L.; Goraya, M.U.; Arafat, Y.; Ajmal, M.; Chen, J.-L.; Yu, D. Molecular Mechanism of Quorum-Sensing in Enterococcus faecalis: Its Role in Virulence and Therapeutic Approaches. Int. J. Mol. Sci. 2017, 18, 960. [Google Scholar] [CrossRef] [PubMed]
  77. Nakayama, J.; Cao, Y.; Horii, T.; Sakuda, S.; Akkermans, A.D.; de Vos, W.M.; Nagasawa, H. Gelatinase biosynthesis-activating pheromone: A peptide lactone that mediates a quorum sensing in Enterococcus faecalis. Mol. Microbiol. 2001, 41, 145–154. [Google Scholar] [CrossRef]
  78. Qin, X.; Singh, K.V.; Weinstock, G.M.; Murray, B.E. Characterization of fsr, a regulator controlling expression of gelatinase and serine protease in Enterococcus faecalis OG1RF. J. Bacteriol. 2001, 183, 3372–3382. [Google Scholar] [CrossRef] [PubMed]
  79. Ma, P.; Yuille, H.M.; Blessie, V.; Göhring, N.; Iglói, Z.; Nishiguchi, K.; Nakayama, J.; Henderson, P.J.F.; Phillips-Jones, M.K. Expression, purification and activities of the entire family of intact membrane sensor kinases from Enterococcus faecalis. Mol. Membr. Biol. 2008, 25, 449–473. [Google Scholar] [CrossRef]
  80. Willett, J.L.E.; Robertson, E.B.; Dunny, G.M. The Phosphatase Bph and Peptidyl-Prolyl Isomerase PrsA Are Required for Gelatinase Expression and Activity in Enterococcus faecalis. J. Bacteriol. 2022, 204, e0012922. [Google Scholar] [CrossRef] [PubMed]
  81. Qin, X.; Singh, K.V.; Weinstock, G.M.; Murray, B.E. Effects of Enterococcus faecalis fsr genes on production of gelatinase and a serine protease and virulence. Infect. Immun. 2000, 68, 2579–2586. [Google Scholar] [CrossRef]
  82. Mylonakis, E.; Engelbert, M.; Qin, X.; Sifri, C.D.; Murray, B.E.; Ausubel, F.M.; Gilmore, M.S.; Calderwood, S.B. The Enterococcus faecalis fsrB gene, a key component of the fsr quorum-sensing system, is associated with virulence in the rabbit endophthalmitis model. Infect. Immun. 2002, 70, 4678–4681. [Google Scholar] [CrossRef] [PubMed]
  83. Garsin, D.A.; Sifri, C.D.; Mylonakis, E.; Qin, X.; Singh, K.V.; Murray, B.E.; Calderwood, S.B.; Ausubel, F.M. A simple model host for identifying Gram-positive virulence factors. Proc. Natl. Acad. Sci. USA 2001, 98, 10892–10897. [Google Scholar] [CrossRef] [PubMed]
  84. Perez, M.; Calles-Enríquez, M.; del Rio, B.; Ladero, V.; Martín, M.C.; Fernández, M.; Alvarez, M.A. IS256 abolishes gelatinase activity and biofilm formation in a mutant of the nosocomial pathogen Enterococcus faecalis V583. Can. J. Microbiol. 2015, 61, 517–519. [Google Scholar] [CrossRef]
  85. Thurlow, L.R.; Thomas, V.C.; Narayanan, S.; Olson, S.; Fleming, S.D.; Hancock, L.E. Gelatinase contributes to the pathogenesis of endocarditis caused by Enterococcus faecalis. Infect. Immun. 2010, 78, 4936–4943. [Google Scholar] [CrossRef]
  86. Nikolskaya, A.N.; Galperin, M.Y. A novel type of conserved DNA-binding domain in the transcriptional regulators of the AlgR/AgrA/LytR family. Nucleic Acids Res. 2002, 30, 2453–2459. [Google Scholar] [CrossRef] [PubMed]
  87. Hashem, Y.A.; Abdelrahman, K.A.; Aziz, R.K. Phenotype-Genotype Correlations and Distribution of Key Virulence Factors in Enterococcus faecalis Isolated from Patients with Urinary Tract Infections. Infect. Drug Resist. 2021, 14, 1713–1723. [Google Scholar] [CrossRef] [PubMed]
  88. Francisco, P.A.; Fagundes, P.I.d.G.; Lemes-Junior, J.C.; Lima, A.R.; Passini, M.R.Z.; Gomes, B.P.F.A. Pathogenic potential of Enterococcus faecalis strains isolated from root canals after unsuccessful endodontic treatment. Clin. Oral Investig. 2021, 25, 5171–5179. [Google Scholar] [CrossRef] [PubMed]
  89. Sillanpää, J.; Nallapareddy, S.R.; Singh, K.V.; Prakash, V.P.; Fothergill, T.; Ton-That, H.; Murray, B.E. Characterization of the ebpfm pilus-encoding operon of Enterococcus faecium and its role in biofilm formation and virulence in a murine model of urinary tract infection. Virulence 2010, 1, 236–246. [Google Scholar] [CrossRef] [PubMed]
  90. Hendrickx, A.P.A.; van Wamel, W.J.B.; Posthuma, G.; Bonten, M.J.M.; Willems, R.J.L. Five genes encoding surface-exposed LPXTG proteins are enriched in hospital-adapted Enterococcus faecium clonal complex 17 isolates. J. Bacteriol. 2007, 189, 8321–8332. [Google Scholar] [CrossRef] [PubMed]
  91. Sillanpää, J.; Prakash, V.P.; Nallapareddy, S.R.; Murray, B.E. Distribution of genes encoding MSCRAMMs and Pili in clinical and natural populations of Enterococcus faecium. J. Clin. Microbiol. 2009, 47, 896–901. [Google Scholar] [CrossRef] [PubMed]
  92. Hendrickx, A.P.A.; van Schaik, W.; Willems, R.J.L. The cell wall architecture of Enterococcus faecium: From resistance to pathogenesis. Future Microbiol. 2013, 8, 993–1010. [Google Scholar] [CrossRef] [PubMed]
  93. Theilacker, C.; Sanchez-Carballo, P.; Toma, I.; Fabretti, F.; Sava, I.; Kropec, A.; Holst, O.; Huebner, J. Glycolipids are involved in biofilm accumulation and prolonged bacteraemia in Enterococcus faecalis. Mol. Microbiol. 2009, 71, 1055–1069. [Google Scholar] [CrossRef]
  94. Teng, F.; Kawalec, M.; Weinstock, G.M.; Hryniewicz, W.; Murray, B.E. An Enterococcus faecium secreted antigen, SagA, exhibits broad-spectrum binding to extracellular matrix proteins and appears essential for E. faecium growth. Infect. Immun. 2003, 71, 5033–5041. [Google Scholar] [CrossRef]
  95. Bubert, A.; Kuhn, M.; Goebel, W.; Köhler, S. Structural and functional properties of the p60 proteins from different Listeria species. J. Bacteriol. 1992, 174, 8166–8171. [Google Scholar] [CrossRef] [PubMed]
  96. Van Tyne, D.; Martin, M.J.; Gilmore, M.S. Structure, function, and biology of the Enterococcus faecalis cytolysin. Toxins 2013, 5, 895–911. [Google Scholar] [CrossRef]
  97. Xiong, X.; Tian, S.; Yang, P.; Lebreton, F.; Bao, H.; Sheng, K.; Yin, L.; Chen, P.; Zhang, J.; Qi, W.; et al. Emerging enterococcus pore-forming toxins with MHC/HLA-I as receptors. Cell 2022, 185, 1157–1171.e22. [Google Scholar] [CrossRef] [PubMed]
  98. Deutscher, J.; Aké, F.M.D.; Derkaoui, M.; Zébré, A.C.; Cao, T.N.; Bouraoui, H.; Kentache, T.; Mokhtari, A.; Milohanic, E.; Joyet, P. The bacterial phosphoenolpyruvate:carbohydrate phosphotransferase system: Regulation by protein phosphorylation and phosphorylation-dependent protein-protein interactions. Microbiol. Mol. Biol. Rev. 2014, 78, 231–256. [Google Scholar] [CrossRef] [PubMed]
  99. Rom, J.S.; Hart, M.T.; McIver, K.S. PRD-Containing Virulence Regulators (PCVRs) in Pathogenic Bacteria. Front. Cell. Infect. Microbiol. 2021, 11, 772874. [Google Scholar] [CrossRef]
  100. Ruiz-Cruz, S.; Espinosa, M.; Goldmann, O.; Bravo, A. Global Regulation of Gene Expression by the MafR Protein of Enterococcus faecalis. Front. Microbiol. 2015, 6, 1521. [Google Scholar] [CrossRef]
  101. Zhang, X.; Top, J.; de Been, M.; Bierschenk, D.; Rogers, M.; Leendertse, M.; Bonten, M.J.M.; van der Poll, T.; Willems, R.J.L.; van Schaik, W. Identification of a genetic determinant in clinical Enterococcus faecium strains that contributes to intestinal colonization during antibiotic treatment. J. Infect. Dis. 2013, 207, 1780–1786. [Google Scholar] [CrossRef]
  102. Paganelli, F.L.; Huebner, J.; Singh, K.V.; Zhang, X.; van Schaik, W.; Wobser, D.; Braat, J.C.; Murray, B.E.; Bonten, M.J.M.; Willems, R.J.L.; et al. Genome-wide Screening Identifies Phosphotransferase System Permease BepA to Be Involved in Enterococcus faecium Endocarditis and Biofilm Formation. J. Infect. Dis. 2016, 214, 189–195. [Google Scholar] [CrossRef]
  103. Eaton, T.J.; Gasson, M.J. Molecular Screening of Enterococcus Virulence Determinants and Potential for Genetic Exchange between Food and Medical Isolates. Appl. Environ. Microbiol. 2001, 67, 1628–1635. [Google Scholar] [CrossRef]
  104. Semedo, T.; Almeida Santos, M.; Martins, P.; Silva Lopes, M.F.; Figueiredo Marques, J.J.; Tenreiro, R.; Barreto Crespo, M.T. Comparative study using type strains and clinical and food isolates to examine hemolytic activity and occurrence of the cyl operon in enterococci. J. Clin. Microbiol. 2003, 41, 2569–2576. [Google Scholar] [CrossRef]
  105. Stępień-Pyśniak, D.; Hauschild, T.; Kosikowska, U.; Dec, M.; Urban-Chmiel, R. Biofilm formation capacity and presence of virulence factors among commensal Enterococcus spp. from wild birds. Sci. Rep. 2019, 9, 11204. [Google Scholar] [CrossRef]
  106. Ike, Y.; Hashimoto, H.; Clewell, D.B. Hemolysin of Streptococcus faecalis subspecies zymogenes contributes to virulence in mice. Infect. Immun. 1984, 45, 528–530. [Google Scholar] [CrossRef] [PubMed]
  107. Kim, B.; Wang, Y.-C.; Hespen, C.W.; Espinosa, J.; Salje, J.; Rangan, K.J.; Oren, D.A.; Kang, J.Y.; Pedicord, V.A.; Hang, H.C. Enterococcus faecium secreted antigen A generates muropeptides to enhance host immunity and limit bacterial pathogenesis. eLife 2019, 8, e45343. [Google Scholar] [CrossRef] [PubMed]
  108. Xu, W.; Flores-Mireles, A.L.; Cusumano, Z.T.; Takagi, E.; Hultgren, S.J.; Caparon, M.G. Host and bacterial proteases influence biofilm formation and virulence in a murine model of enterococcal catheter-associated urinary tract infection. NPJ Biofilms Microbiomes 2017, 3, 28. [Google Scholar] [CrossRef]
  109. Gök, Ş.M.; Türk Dağı, H.; Kara, F.; Arslan, U.; Fındık, D. Investigation of Antibiotic Resistance and Virulence Factors of Enterococcus faecium and Enterococcus faecalis Strains Isolated from Clinical Samples. Mikrobiyol. Bul. 2020, 54, 26–39. [Google Scholar] [CrossRef] [PubMed]
  110. Waters, C.M.; Hirt, H.; McCormick, J.K.; Schlievert, P.M.; Wells, C.L.; Dunny, G.M. An amino-terminal domain of Enterococcus faecalis aggregation substance is required for aggregation, bacterial internalization by epithelial cells and binding to lipoteichoic acid. Mol. Microbiol. 2004, 52, 1159–1171. [Google Scholar] [CrossRef]
  111. Schlievert, P.M.; Gahr, P.J.; Assimacopoulos, A.P.; Dinges, M.M.; Stoehr, J.A.; Harmala, J.W.; Hirt, H.; Dunny, G.M. Aggregation and binding substances enhance pathogenicity in rabbit models of Enterococcus faecalis endocarditis. Infect. Immun. 1998, 66, 218–223. [Google Scholar] [CrossRef]
  112. Yagi, Y.; Kessler, R.E.; Shaw, J.H.; Lopatin, D.E.; An, F.; Clewell, D.B. Plasmid content of Streptococcus faecalis strain 39-5 and identification of a pheromone (cPD1)-induced surface antigen. J. Gen. Microbiol. 1983, 129, 1207–1215. [Google Scholar] [CrossRef]
  113. Galli, D.; Lottspeich, F.; Wirth, R. Sequence analysis of Enterococcus faecalis aggregation substance encoded by the sex pheromone plasmid pAD1. Mol. Microbiol. 1990, 4, 895–904. [Google Scholar] [CrossRef]
  114. Kafil, H.S.; Mobarez, A.M.; Moghadam, M.F. Adhesion and virulence factor properties of Enterococci isolated from clinical samples in Iran. Indian J. Pathol. Microbiol. 2013, 56, 238–242. [Google Scholar] [CrossRef]
  115. Lowe, A.M.; Lambert, P.A.; Smith, A.W. Cloning of an Enterococcus faecalis endocarditis antigen: Homology with adhesins from some oral streptococci. Infect. Immun. 1995, 63, 703–706. [Google Scholar] [CrossRef]
  116. Strateva, T.; Atanasova, D.; Savov, E.; Petrova, G.; Mitov, I. Incidence of virulence determinants in clinical Enterococcus faecalis and Enterococcus faecium isolates collected in Bulgaria. Braz. J. Infect. Dis. 2016, 20, 127–133. [Google Scholar] [CrossRef]
  117. Hendrickx, A.P.A.; van Luit-Asbroek, M.; Schapendonk, C.M.E.; van Wamel, W.J.B.; Braat, J.C.; Wijnands, L.M.; Bonten, M.J.M.; Willems, R.J.L. SgrA, a nidogen-binding LPXTG surface adhesin implicated in biofilm formation, and EcbA, a collagen binding MSCRAMM, are two novel adhesins of hospital-acquired Enterococcus faecium. Infect. Immun. 2009, 77, 5097–5106. [Google Scholar] [CrossRef] [PubMed]
  118. Teng, F.; Singh, K.V.; Bourgogne, A.; Zeng, J.; Murray, B.E. Further characterization of the epa gene cluster and Epa polysaccharides of Enterococcus faecalis. Infect. Immun. 2009, 77, 3759–3767. [Google Scholar] [CrossRef]
  119. Nallapareddy, S.R.; Sillanpää, J.; Mitchell, J.; Singh, K.V.; Chowdhury, S.A.; Weinstock, G.M.; Sullam, P.M.; Murray, B.E. Conservation of Ebp-type pilus genes among Enterococci and demonstration of their role in adherence of Enterococcus faecalis to human platelets. Infect. Immun. 2011, 79, 2911–2920. [Google Scholar] [CrossRef] [PubMed]
  120. Chilambi, G.S.; Nordstrom, H.R.; Evans, D.R.; Ferrolino, J.A.; Hayden, R.T.; Marón, G.M.; Vo, A.N.; Gilmore, M.S.; Wolf, J.; Rosch, J.W.; et al. Evolution of vancomycin-resistant Enterococcus faecium during colonization and infection in immunocompromised pediatric patients. Proc. Natl. Acad. Sci. USA 2020, 117, 11703–11714. [Google Scholar] [CrossRef]
  121. Brodin, P.; Davis, M.M. Human immune system variation. Nat. Rev. Immunol. 2017, 17, 21–29. [Google Scholar] [CrossRef] [PubMed]
  122. Cohen, I.R.; Efroni, S. The Immune System Computes the State of the Body: Crowd Wisdom, Machine Learning, and Immune Cell Reference Repertoires Help Manage Inflammation. Front. Immunol. 2019, 10, 425417. [Google Scholar] [CrossRef]
  123. Fali, T.; Vallet, H.; Sauce, D. Impact of stress on aged immune system compartments: Overview from fundamental to clinical data. Exp. Gerontol. 2018, 105, 19–26. [Google Scholar] [CrossRef] [PubMed]
  124. Lee, R.A.; Goldman, J.; Haidar, G.; Lewis, J.; Arif, S.; Hand, J.; La Hoz, R.M.; Pouch, S.; Holaday, E.; Clauss, H.; et al. Daptomycin-Resistant Enterococcus Bacteremia Is Associated with Prior Daptomycin Use and Increased Mortality After Liver Transplantation. Open Forum Infect. Dis. 2022, 9, ofab659. [Google Scholar] [CrossRef]
  125. Wu, X.; Wu, L.; Wan, Q. Pathogen distribution and risk factors of bacterial and fungal infections after liver transplantation. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2022, 47, 1120–1128. [Google Scholar] [CrossRef] [PubMed]
  126. Tien, B.Y.Q.; Goh, H.M.S.; Chong, K.K.L.; Bhaduri-Tagore, S.; Holec, S.; Dress, R.; Ginhoux, F.; Ingersoll, M.A.; Williams, R.B.H.; Kline, K.A. Enterococcus faecalis Promotes Innate Immune Suppression and Polymicrobial Catheter-Associated Urinary Tract Infection. Infect. Immun. 2017, 85, e00378-17. [Google Scholar] [CrossRef] [PubMed]
  127. Oliveira, R.A.; Mancero, J.M.P.; Faria, D.F.; Poveda, V.d.B. A Retrospective Cohort Study of Risk Factors for Surgical Site Infection Following Liver Transplantation. Prog. Transplant. 2019, 29, 144–149. [Google Scholar] [CrossRef] [PubMed]
  128. Piewngam, P.; Chiou, J.; Ling, J.; Liu, R.; Pupa, P.; Zheng, Y.; Otto, M. Enterococcal bacteremia in mice is prevented by oral administration of probiotic Bacillus spores. Sci. Transl. Med. 2021, 13, eabf4692. [Google Scholar] [CrossRef] [PubMed]
  129. Souli, M.; Sakka, V.; Galani, I.; Antoniadou, A.; Galani, L.; Siafakas, N.; Zerva, L.; Fytrou, H.; Tsiodras, S.; Giamarellou, H. Colonisation with vancomycin- and linezolid-resistant Enterococcus faecium in a university hospital: Molecular epidemiology and risk factor analysis. Int. J. Antimicrob. Agents 2009, 33, 137–142. [Google Scholar] [CrossRef] [PubMed]
  130. Balian, C.; Garcia, M.; Ward, J. A Retrospective Analysis of Bloodstream Infections in Pediatric Allogeneic Stem Cell Transplant Recipients: The Role of Central Venous Catheters and Mucosal Barrier Injury. J. Pediatr. Oncol. Nurs. 2018, 35, 210–217. [Google Scholar] [CrossRef] [PubMed]
  131. Van Tyne, D.; Gilmore, M.S. Raising the Alarmone: Within-Host Evolution of Antibiotic-Tolerant Enterococcus faecium. mBio 2017, 8, e00066-17. [Google Scholar] [CrossRef] [PubMed]
  132. Van Tyne, D.; Manson, A.L.; Huycke, M.M.; Karanicolas, J.; Earl, A.M.; Gilmore, M.S. Impact of antibiotic treatment and host innate immune pressure on enterococcal adaptation in the human bloodstream. Sci. Transl. Med. 2019, 11, eaat8418. [Google Scholar] [CrossRef] [PubMed]
  133. Jafari, S.; Abdollahi, A.; Sabahi, M.; Salehi, M.; Asadollahi-Amin, A.; Hasannezhad, M.; Seifi, A. An Update to Enterococcal Bacteremia: Epidemiology, Resistance, and Outcome. Infect. Disord. Drug Targets 2022, 22, e170322187568. [Google Scholar] [CrossRef] [PubMed]
  134. Suntharam, N.; Lankford, M.G.; Trick, W.E.; Peterson, L.R.; Noskin, G.A. Risk factors for acquisition of vancomycin-resistant enterococci among hematology-oncology patients. Diagn. Microbiol. Infect. Dis. 2002, 43, 183–188. [Google Scholar] [CrossRef]
  135. Matar, M.J.; Tarrand, J.; Raad, I.; Rolston, K.V. Colonization and infection with vancomycin-resistant Enterococcus among patients with cancer. Am. J. Infect. Control 2006, 34, 534–536. [Google Scholar] [CrossRef]
  136. Edmond, M.B.; Ober, J.F.; Weinbaum, D.L.; Pfaller, M.A.; Hwang, T.; Sanford, M.D.; Wenzel, R.P. Vancomycin-resistant Enterococcus faecium bacteremia: Risk factors for infection. Clin. Infect. Dis. 1995, 20, 1126–1133. [Google Scholar] [CrossRef]
  137. Rafey, A.; Nizamuddin, S.; Qureshi, W.; Anjum, A.; Parveen, A. Trends of Vancomycin-Resistant Enterococcus Infections in Cancer Patients. Cureus 2022, 14, e31335. [Google Scholar] [CrossRef]
  138. Eryilmaz-Eren, E.; Izci, F.; Ture, Z.; Sagiroglu, P.; Kaynar, L.; Ulu-Kilic, A. Bacteremia in Hematopoietic Stem Cell Recipients Receiving Fluoroquinolone Prophylaxis: Incidence, Resistance, and Risk Factors. Infect. Chemother. 2022, 54, 446–455. [Google Scholar] [CrossRef]
  139. Bailly, B.; Lecheneaut, M.; Gbaguidi-Haore, H.; Chirouze, C.; Kleinclauss, F.; Bouiller, K. Epidemiology and risk factors for febrile ureteral stent-associated urinary tract infections: A prospective observational cohort study. J. Infect. 2023, 87, 12–17. [Google Scholar] [CrossRef]
  140. Sakka, V.; Tsiodras, S.; Galani, L.; Antoniadou, A.; Souli, M.; Galani, I.; Pantelaki, M.; Siafakas, N.; Zerva, L.; Giamarellou, H. Risk-factors and predictors of mortality in patients colonised with vancomycin-resistant enterococci. Clin. Microbiol. Infect. 2008, 14, 14–21. [Google Scholar] [CrossRef]
  141. Rodríguez-Noriega, E.; Hernández-Morfin, N.; Garza-Gonzalez, E.; Bocanegra-Ibarias, P.; Flores-Treviño, S.; Esparza-Ahumada, S.; González-Díaz, E.; Pérez-Gómez, H.R.; Mendoza-Mujica, C.; León-Garnica, G.; et al. Risk factors and outcome associated with the acquisition of linezolid-resistant Enterococcus faecalis. J. Glob. Antimicrob. Resist. 2020, 21, 405–409. [Google Scholar] [CrossRef]
  142. Meidrops, K.; Burkhardt, F.J.; Osipovs, J.D.; Petrosina, E.; Groma, V.; Stradins, P. Etiology, Risk Factors and Clinical Outcomes in Infective Endocarditis Patients Requiring Cardiac Surgery. J. Clin. Med. 2022, 11, 1957. [Google Scholar] [CrossRef]
  143. Danneels, P.; Hamel, J.-F.; Picard, L.; Rezig, S.; Martinet, P.; Lorleac’h, A.; Talarmin, J.-P.; Buzelé, R.; Guimard, T.; Le Moal, G.; et al. Impact of Enterococcus faecalis Endocarditis Treatment on Risk of Relapse. Clin. Infect. Dis. 2023, 76, 281–290. [Google Scholar] [CrossRef]
  144. Tsantes, A.G.; Altsitzioglou, P.; Papadopoulos, D.V.; Lorenzo, D.; Romanò, C.L.; Benzakour, T.; Tsukamoto, S.; Errani, C.; Angelini, A.; Mavrogenis, A.F. Infections of Tumor Prostheses: An Updated Review on Risk Factors, Microbiology, Diagnosis, and Treatment Strategies. Biology 2023, 12, 314. [Google Scholar] [CrossRef]
  145. Byers, K.E.; Anglim, A.M.; Anneski, C.J.; Germanson, T.P.; Gold, H.S.; Durbin, L.J.; Simonton, B.M.; Farr, B.M. A hospital epidemic of vancomycin-resistant Enterococcus risk factors and control. Infect. Control Hosp. Epidemiol. 2001, 22, 140–147. [Google Scholar] [CrossRef]
  146. Mascini, E.M.; Troelstra, A.; Beitsma, M.; Blok, H.E.M.; Jalink, K.P.; Hopmans, T.E.M.; Fluit, A.C.; Hene, R.J.; Willems, R.J.L.; Verhoef, J. Genotyping and preemptive isolation to control an outbreak of vancomycin-resistant Enterococcus faecium. Clin. Infect. Dis. 2006, 42, 739–746. [Google Scholar] [CrossRef]
  147. Carmeli, Y.; Eliopoulos, G.M.; Samore, M.H. Antecedent treatment with different antibiotic agents as a risk factor for vancomycin-resistant Enterococcus. Emerg. Infect. Dis. 2002, 8, 802. [Google Scholar] [CrossRef]
  148. Zhou, Q.; Moore, C.; Eden, S.; Tong, A.; McGeer, A. Mount Sinai Hospital Infection Control Team Factors associated with acquisition of vancomycin-resistant enterococci (VRE) in roommate contacts of patients colonized or infected with VRE in a tertiary care hospital. Infect. Control Hosp. Epidemiol. 2008, 29, 398–403. [Google Scholar] [CrossRef]
  149. Furtado, G.H.C.; Mendes, R.E.; Pignatari, A.C.C.; Wey, S.B.; Medeiros, E.A.S. Risk factors for vancomycin-resistant Enterococcus faecalis bacteremia in hospitalized patients: An analysis of two case-control studies. Am. J. Infect. Control 2006, 34, 447–451. [Google Scholar] [CrossRef]
  150. McNeil, S.A.; Malani, P.N.; Chenoweth, C.E.; Fontana, R.J.; Magee, J.C.; Punch, J.D.; Mackin, M.L.; Kauffman, C.A. Vancomycin-resistant enterococcal colonization and infection in liver transplant candidates and recipients: A prospective surveillance study. Clin. Infect. Dis. 2006, 42, 195–203. [Google Scholar] [CrossRef]
  151. Chacko, K.I.; Sullivan, M.J.; Beckford, C.; Altman, D.R.; Ciferri, B.; Pak, T.R.; Sebra, R.; Kasarskis, A.; Hamula, C.L.; Van Bakel, H. Genetic Basis of Emerging Vancomycin, Linezolid, and Daptomycin Heteroresistance in a Case of Persistent Enterococcus faecium Bacteremia. Antimicrob. Agents Chemother. 2018, 62, e02007-17. [Google Scholar] [CrossRef]
  152. Chen, M.; Pan, H.; Lou, Y.; Wu, Z.; Zhang, J.; Huang, Y.; Yu, W.; Qiu, Y. Epidemiological characteristics and genetic structure of linezolid-resistant Enterococcus faecalis. Infect. Drug Resist. 2018, 11, 2397–2409. [Google Scholar] [CrossRef]
  153. Orsi, G.B.; Ciorba, V. Vancomycin resistant enterococci healthcare associated infections. Ann Ig 2013, 25, 485–492. [Google Scholar] [CrossRef]
  154. Chou, Y.-Y.; Lin, T.-Y.; Lin, J.-C.; Wang, N.-C.; Peng, M.-Y.; Chang, F.-Y. Vancomycin-resistant enterococcal bacteremia: Comparison of clinical features and outcome between Enterococcus faecium and Enterococcus faecalis. J. Microbiol. Immunol. Infect. 2008, 41, 124–129. [Google Scholar]
  155. Garbutt, J.M.; Ventrapragada, M.; Littenberg, B.; Mundy, L.M. Association between resistance to vancomycin and death in cases of Enterococcus faecium bacteremia. Clin. Infect. Dis. 2000, 30, 466–472. [Google Scholar] [CrossRef]
  156. Lebreton, F.; van Schaik, W.; McGuire, A.M.; Godfrey, P.; Griggs, A.; Mazumdar, V.; Corander, J.; Cheng, L.; Saif, S.; Young, S.; et al. Emergence of epidemic multidrug-resistant Enterococcus faecium from animal and commensal strains. mBio 2013, 4, e00534-13. [Google Scholar] [CrossRef]
  157. Didelot, X.; Walker, A.S.; Peto, T.E.; Crook, D.W.; Wilson, D.J. Within-host evolution of bacterial pathogens. Nat. Rev. Microbiol. 2016, 14, 150–162. [Google Scholar] [CrossRef]
  158. Lam, M.M.C.; Seemann, T.; Bulach, D.M.; Gladman, S.L.; Chen, H.; Haring, V.; Moore, R.J.; Ballard, S.; Grayson, M.L.; Johnson, P.D.R.; et al. Comparative analysis of the first complete Enterococcus faecium genome. J. Bacteriol. 2012, 194, 2334–2341. [Google Scholar] [CrossRef]
  159. Arredondo-Alonso, S.; Top, J.; Schürch, A.C.; McNally, A.; Puranen, S.; Pesonen, M.; Pensar, J.; Marttinen, P.; Braat, J.C.; Rogers, M.R.; et al. Genomes of a major nosocomial pathogen Enterococcus faecium are shaped by adaptive evolution of the chromosome and plasmidome. bioRxiv 2019, 530725. [Google Scholar] [CrossRef]
  160. Boonanantanasarn, K.; Gill, A.L.; Yap, Y.; Jayaprakash, V.; Sullivan, M.A.; Gill, S.R. Enterococcus faecalis enhances cell proliferation through hydrogen peroxide-mediated epidermal growth factor receptor activation. Infect. Immun. 2012, 80, 3545–3558. [Google Scholar] [CrossRef]
  161. Maehata, Y.; Ozawa, S.; Kobayashi, K.; Kato, Y.; Yoshino, F.; Miyamoto, C.; Izukuri, K.; Kubota, E.; Hata, R.-I.; Lee, M.-C.-I. Reactive oxygen species (ROS) reduce the expression of BRAK/CXCL14 in human head and neck squamous cell carcinoma cells. Free Radic. Res. 2010, 44, 913–924. [Google Scholar] [CrossRef]
  162. Forsyth, C.B.; Banan, A.; Farhadi, A.; Fields, J.Z.; Tang, Y.; Shaikh, M.; Zhang, L.J.; Engen, P.A.; Keshavarzian, A. Regulation of oxidant-induced intestinal permeability by metalloprotease-dependent epidermal growth factor receptor signaling. J. Pharmacol. Exp. Ther. 2007, 321, 84–97. [Google Scholar] [CrossRef]
  163. Lee, E.; Yi, J.Y.; Chung, E.; Son, Y. Transforming growth factorbeta1 transactivates EGFR via an H2O2-dependent mechanism in squamous carcinoma cell line. Cancer Lett. 2010, 290, 43–48. [Google Scholar] [CrossRef]
  164. Grootaert, C.; Van de Wiele, T.; Van Roosbroeck, I.; Possemiers, S.; Vercoutter-Edouart, A.-S.; Verstraete, W.; Bracke, M.; Vanhoecke, B. Bacterial monocultures, propionate, butyrate and H2O2 modulate the expression, secretion and structure of the fasting-induced adipose factor in gut epithelial cell lines. Environ. Microbiol. 2011, 13, 1778–1789. [Google Scholar] [CrossRef]
  165. Wang, X.; Allen, T.D.; May, R.J.; Lightfoot, S.; Houchen, C.W.; Huycke, M.M. Enterococcus faecalis induces aneuploidy and tetraploidy in colonic epithelial cells through a bystander effect. Cancer Res. 2008, 68, 9909–9917. [Google Scholar] [CrossRef]
  166. Shen, F.; Feng, J.; Wang, X.; Qi, Z.; Shi, X.; An, Y.; Zhang, Q.; Wang, C.; Liu, M.; Liu, B.; et al. Vinegar Treatment Prevents the Development of Murine Experimental Colitis via Inhibition of Inflammation and Apoptosis. J. Agric. Food Chem. 2016, 64, 1111–1121. [Google Scholar] [CrossRef]
  167. Ou, C.-C.; Lin, S.-L.; Tsai, J.-J.; Lin, M.-Y. Heat-killed lactic acid bacteria enhance immunomodulatory potential by skewing the immune response toward Th1 polarization. J. Food Sci. 2011, 76, M260–M267. [Google Scholar] [CrossRef]
  168. Wang, S.; Liu, K.; Seneviratne, C.J.; Li, X.; Cheung, G.S.P.; Jin, L.; Chu, C.H.; Zhang, C. Lipoteichoic acid from an Enterococcus faecalis clinical strain promotes TNF-α expression through the NF-κB and p38 MAPK signaling pathways in differentiated THP-1 macrophages. Biomed. Rep. 2015, 3, 697–702. [Google Scholar] [CrossRef]
  169. Li, M.; Wei, L.; Zhou, W.; He, Z.; Ran, S.; Liang, J. miR-200a contributes to the migration of BMSCs induced by the secretions of E. faecalis via FOXJ1/NFκB/MMPs axis. Stem Cell Res. Ther. 2020, 11, 317. [Google Scholar] [CrossRef]
  170. You, R.; Kwon, O.Y.; Woo, H.J.; Lee, S.H. Hovenia Monofloral Honey can Attenuate Enterococcus faecalis Mediated Biofilm Formation and Inflammation. Food Sci. Anim. Resour. 2022, 42, 84–97. [Google Scholar] [CrossRef]
  171. Leong, J.Y.; Capella, C.E.; D’Amico, M.J.; Isguven, S.; Purtill, C.; Machado, P.; Delaney, L.J.; Henry, G.D.; Hickok, N.J.; Forsberg, F.; et al. A scoping review of penile implant biofilms-what do we know and what remains unknown? Transl. Androl. Urol. 2022, 11, 1210–1221. [Google Scholar] [CrossRef]
  172. Ali, I.A.A.; Lévesque, C.M.; Neelakantan, P. Fsr quorum sensing system modulates the temporal development of Enterococcus faecalis biofilm matrix. Mol. Oral Microbiol. 2022, 37, 22–30. [Google Scholar] [CrossRef]
  173. Ch’ng, J.-H.; Chong, K.K.L.; Lam, L.N.; Wong, J.J.; Kline, K.A. Biofilm-associated infection by enterococci. Nat. Rev. Microbiol. 2019, 17, 82–94. [Google Scholar] [CrossRef]
  174. Rezende-Pereira, G.; Albuquerque, J.P.; Souza, M.C.; Nogueira, B.A.; Silva, M.G.; Hirata, R.; Mattos-Guaraldi, A.L.; Duarte, R.S.; Neves, F.P.G. Biofilm Formation on Breast Implant Surfaces by Major Gram-Positive Bacterial Pathogens. Aesthet. Surg. J. 2021, 41, 1144–1151. [Google Scholar] [CrossRef]
  175. Kolodkin-Gal, I.; Cohen-Cymberknoh, M.; Zamir, G.; Tsesis, I.; Rosen, E. Targeting Persistent Biofilm Infections: Reconsidering the Topography of the Infection Site during Model Selection. Microorganisms 2022, 10, 1164. [Google Scholar] [CrossRef] [PubMed]
  176. Fiorotto, R.; Mariotti, V.; Taleb, S.A.; Zehra, S.A.; Nguyen, M.; Amenduni, M.; Strazzabosco, M. Cell-matrix interactions control biliary organoid polarity, architecture, and differentiation. Hepatol. Commun. 2023, 7, e0094. [Google Scholar] [CrossRef]
  177. Xiang, D.; Dong, P.-T.; Cen, L.; Bor, B.; Lux, R.; Shi, W.; Yu, Q.; He, X.; Wu, T. Antagonistic interaction between two key endodontic pathogens Enterococcus faecalis and Fusobacterium nucleatum. J. Oral Microbiol. 2023, 15, 2149448. [Google Scholar] [CrossRef] [PubMed]
  178. Parga, A.; Manoil, D.; Brundin, M.; Otero, A.; Belibasakis, G.N. Gram-negative quorum sensing signalling enhances biofilm formation and virulence traits in gram-positive pathogen Enterococcus faecalis. J. Oral Microbiol. 2023, 15, 2208901. [Google Scholar] [CrossRef]
  179. Hashem, Y.A.; Amin, H.M.; Essam, T.M.; Yassin, A.S.; Aziz, R.K. Biofilm formation in enterococci: Genotype-phenotype correlations and inhibition by vancomycin. Sci. Rep. 2017, 7, 5733. [Google Scholar] [CrossRef]
  180. Gilmore, M.S.; Clewell, D.B.; Ike, Y.; Shankar, N. Enterococci: From Commensals to Leading Causes of Drug Resistant Infection; Massachusetts Eye and Ear Infirmary: Boston, MA, USA, 2014. [Google Scholar]
  181. Smith, R.E.; Salamaga, B.; Szkuta, P.; Hajdamowicz, N.; Prajsnar, T.K.; Bulmer, G.S.; Fontaine, T.; Kołodziejczyk, J.; Herry, J.-M.; Hounslow, A.M.; et al. Decoration of the enterococcal polysaccharide antigen EPA is essential for virulence, cell surface charge and interaction with effectors of the innate immune system. PLoS Pathog. 2019, 15, e1007730. [Google Scholar] [CrossRef] [PubMed]
  182. Ragland, S.A.; Criss, A.K. From bacterial killing to immune modulation: Recent insights into the functions of lysozyme. PLoS Pathog. 2017, 13, e1006512. [Google Scholar] [CrossRef] [PubMed]
  183. Kristensen, S.S.; Diep, D.B.; Kjos, M.; Mathiesen, G. The role of site-2-proteases in bacteria: A review on physiology, virulence, and therapeutic potential. microLife 2023, 4, uqad025. [Google Scholar] [CrossRef]
  184. Rouchon, C.N.; Harris, J.; Zubair-Nizami, Z.; Weinstein, A.J.; Roky, M.; Frank, K.L. The Cationic Antimicrobial Peptide Activity of Lysozyme Reduces Viable Enterococcus faecalis Cells in Biofilms. Antimicrob. Agents Chemother. 2022, 66, e0233921. [Google Scholar] [CrossRef]
  185. Varahan, S.; Iyer, V.S.; Moore, W.T.; Hancock, L.E. Eep confers lysozyme resistance to Enterococcus faecalis via the activation of the extracytoplasmic function sigma factor SigV. J. Bacteriol. 2013, 195, 3125–3134. [Google Scholar] [CrossRef]
  186. Reinseth, I.; Tønnesen, H.H.; Carlsen, H.; Diep, D.B. Exploring the Therapeutic Potenital of the Leaderless Enterocins K1 and EJ97 in the Treatment of Vancomycin-Resistant Enterococcal Infection. Front. Microbiol. 2021, 12, 649339. [Google Scholar] [CrossRef] [PubMed]
  187. Hébert, L.; Courtin, P.; Torelli, R.; Sanguinetti, M.; Chapot-Chartier, M.-P.; Auffray, Y.; Benachour, A. Enterococcus faecalis constitutes an unusual bacterial model in lysozyme resistance. Infect. Immun. 2007, 75, 5390–5398. [Google Scholar] [CrossRef] [PubMed]
  188. Said, M.S.; Tirthani, E.; Lesho, E. Enterococcus Infections. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  189. Guerra, D.; Vidal, P.; Paccoud, O.; Maillard, A.; Cachera, L.; Junot, H.; Gauzit, R.; Zahar, J.R.; Abreu, M.A.; Bleibtreu, A. Dual beta-lactam treatment: Pros and cons. Porto Biomed. J. 2022, 7, e189. [Google Scholar] [CrossRef]
  190. Clark, N.C.; Cooksey, R.C.; Hill, B.C.; Swenson, J.M.; Tenover, F.C. Characterization of glycopeptide-resistant enterococci from U.S. hospitals. Antimicrob. Agents Chemother. 1993, 37, 2311–2317. [Google Scholar] [CrossRef] [PubMed]
  191. Wardal, E.; Żabicka, D.; Skalski, T.; Kubiak-Pulkowska, J.; Hryniewicz, W.; Sadowy, E. Characterization of a Tigecycline-, Linezolid- and Vancomycin-Resistant Clinical Enteroccoccus faecium Isolate, Carrying vanA and vanB Genes. Infect. Dis. Ther. 2023, 12, 2545–2565. [Google Scholar] [CrossRef] [PubMed]
  192. Cairns, K.A.; Udy, A.A.; Peel, T.N.; Abbott, I.J.; Dooley, M.J.; Peleg, A.Y. Therapeutics for Vancomycin-Resistant Enterococcal Bloodstream Infections. Clin. Microbiol. Rev. 2023, 36, e0005922. [Google Scholar] [CrossRef] [PubMed]
  193. Yim, J.; Smith, J.R.; Rybak, M.J. Role of Combination Antimicrobial Therapy for Vancomycin-Resistant Enterococcus faecium Infections: Review of the Current Evidence. Pharmacotherapy 2017, 37, 579–592. [Google Scholar] [CrossRef]
  194. Chong, Y.P.; Lee, S.-O.; Song, E.H.; Lee, E.J.; Jang, E.-Y.; Kim, S.-H.; Choi, S.-H.; Kim, M.-N.; Jeong, J.-Y.; Woo, J.H.; et al. Quinupristin-dalfopristin versus linezolid for the treatment of vancomycin-resistant Enterococcus faecium bacteraemia: Efficacy and development of resistance. Scand. J. Infect. Dis. 2010, 42, 491–499. [Google Scholar] [CrossRef] [PubMed]
  195. Linden, P.K. Treatment options for vancomycin-resistant enterococcal infections. Drugs 2002, 62, 425–441. [Google Scholar] [CrossRef]
  196. Noskin, G.A.; Siddiqui, F.; Stosor, V.; Kruzynski, J.; Peterson, L.R. Successful treatment of persistent vancomycin-resistant Enterococcus faecium bacteremia with linezolid and gentamicin. Clin. Infect. Dis. 1999, 28, 689–690. [Google Scholar] [CrossRef]
  197. Egan, S.A.; Kavanagh, N.L.; Shore, A.C.; Mollerup, S.; Samaniego Castruita, J.A.; O’Connell, B.; McManus, B.A.; Brennan, G.I.; Pinholt, M.; Westh, H.; et al. Genomic analysis of 600 vancomycin-resistant Enterococcus faecium reveals a high prevalence of ST80 and spread of similar vanA regions via IS1216E and plasmid transfer in diverse genetic lineages in Ireland. J. Antimicrob. Chemother. 2022, 77, 320–330. [Google Scholar] [CrossRef] [PubMed]
  198. Zerrouki, H.; Rebiahi, S.-A.; Hadjadj, L.; Ahlem, F.; Elhabiri, Y.; Sedrati, T.; Rolain, J.-M.; Diene, S.M. High frequency and diversity of Vancomycin-resistant Enterococci (VRE) in Algerian healthcare settings. Infect. Genet. Evol. 2021, 92, 104889. [Google Scholar] [CrossRef] [PubMed]
  199. Rosselli Del Turco, E.; Bartoletti, M.; Dahl, A.; Cervera, C.; Pericàs, J.M. How do I manage a patient with enterococcal bacteraemia? Clin. Microbiol. Infect. 2021, 27, 364–371. [Google Scholar] [CrossRef] [PubMed]
  200. Penven, M.; Zouari, A.; Nogues, S.; Collet, A.; Lecourt, M.; Birer, A.; Guerin, F.; Auger, G.; Cattoir, V. Web-based prediction of antimicrobial resistance in enterococcal clinical isolates by whole-genome sequencing. Eur. J. Clin. Microbiol. Infect. Dis. 2023, 42, 67–76. [Google Scholar] [CrossRef] [PubMed]
  201. Mollerup, S.; Elmeskov, C.; Pinholt, M.; Sejersen, T.S.; Pedersen, M.S.; Worning, P.; Frees, D.; Westh, H. Rapid in vivo development of resistance to daptomycin in vancomycin-resistant Enterococcus faecium due to genomic alterations. FEMS Microbiol. Lett. 2022, 369, fnac063. [Google Scholar] [CrossRef] [PubMed]
  202. Chuang, Y.-C.; Wang, J.-T.; Yang, J.-L.; Lin, C.-Y.; Huang, S.-H.; Chen, Y.-C.; Chang, S.-C. The combination of daptomycin with β-lactam antibiotics is more effective than daptomycin alone for vancomycin-resistant Enterococcus faecium bloodstream infection. J. Infect. Public Health 2022, 15, 1396–1402. [Google Scholar] [CrossRef]
  203. Smith, J.R.; Barber, K.E.; Raut, A.; Aboutaleb, M.; Sakoulas, G.; Rybak, M.J. β-Lactam combinations with daptomycin provide synergy against vancomycin-resistant Enterococcus faecalis and Enterococcus faecium. J. Antimicrob. Chemother. 2015, 70, 1738–1743. [Google Scholar] [CrossRef] [PubMed]
  204. Santimaleeworagun, W.; Hemapanpairoa, J.; Changpradub, D.; Thunyaharn, S. Optimizing the Dosing Regimens of Tigecycline against Vancomycin-Resistant Enterococci in the Treatment of Intra-abdominal and Skin and Soft Tissue Infections. Infect. Chemother. 2020, 52, 345–351. [Google Scholar] [CrossRef] [PubMed]
  205. Kristich, C.J.; Rice, L.B.; Arias, C.A. Enterococcal Infection—Treatment and Antibiotic Resistance. In Enterococci: From Commensals to Leading Causes of Drug Resistant Infection; Gilmore, M.S., Clewell, D.B., Ike, Y., Shankar, N., Eds.; Massachusetts Eye and Ear Infirmary: Boston, MA, USA, 2014. [Google Scholar]
  206. Rodríguez-García, R.; Rodríguez-Esteban, M.Á.; García-Carús, E.; Telenti, M.; Fernández, J. In vitro activity of ceftaroline and ceftobiprole against clinical isolates of Gram-positive bacteria from infective endocarditis: Are these drugs potential options for the initial management of this disease? Diagn. Microbiol. Infect. Dis. 2020, 98, 115153. [Google Scholar] [CrossRef]
  207. van der Starre, C.M.; Cremers-Pijpers, S.A.J.; van Rossum, C.; Bowles, E.C.; Tostmann, A. The in situ efficacy of whole room disinfection devices: A literature review with practical recommendations for implementation. Antimicrob. Resist. Infect. Control 2022, 11, 149. [Google Scholar] [CrossRef]
  208. Tan, C.; Linkenheld-Struk, A.; Williams, V.; Kozak, R.; Dhabaan, G.; Maze Dit Mieusement, L.; Salt, N.; Maguire, F.; Chan, A.K.; Leis, J.A. The role of the environment in transmission of vancomycin-resistant Enterococcus: A proof-of-concept study. Antimicrob. Steward. Healthc. Epidemiol. 2022, 2, e178. [Google Scholar] [CrossRef] [PubMed]
  209. Suh, J.W.; Kim, N.H.; Lee, M.J.; Lee, S.E.; Chun, B.C.; Lee, C.K.; Lee, J.; Kim, J.H.; Kim, S.B.; Yoon, Y.K.; et al. Real-world experience of how chlorhexidine bathing affects the acquisition and incidence of vancomycin-resistant enterococci (VRE) in a medical intensive care unit with VRE endemicity: A prospective interrupted time-series study. Antimicrob. Resist. Infect. Control 2021, 10, 160. [Google Scholar] [CrossRef]
  210. Kalfopoulou, E.; Huebner, J. Advances and Prospects in Vaccine Development against Enterococci. Cells 2020, 9, 2397. [Google Scholar] [CrossRef] [PubMed]
  211. EARS-Net EQA. Available online: https://antimicrobialresistance.dk/ears-net-eqa.aspx (accessed on 21 July 2023).
  212. Forde, B.M.; Bergh, H.; Cuddihy, T.; Hajkowicz, K.; Hurst, T.; Playford, E.G.; Henderson, B.C.; Runnegar, N.; Clark, J.; Jennison, A.V.; et al. Clinical Implementation of Routine Whole-genome Sequencing for Hospital Infection Control of Multi-drug Resistant Pathogens. Clin. Infect. Dis. 2023, 76, e1277–e1284. [Google Scholar] [CrossRef] [PubMed]
  213. Büchler, A.C.; Ragozzino, S.; Wicki, M.; Spaniol, V.; Jäger, S.; Seth-Smith, H.M.B.; Goldenberger, D.; Hinic, V.; Egli, A.; Frei, R.; et al. Patients exposed to vancomycin-resistant enterococci during in-hospital outbreaks in a low endemic setting: A proposal for risk-based screening. Antimicrob. Resist. Infect. Control 2022, 11, 60. [Google Scholar] [CrossRef] [PubMed]
  214. Abi Frem, J.; Ghanem, M.; Doumat, G.; Kanafani, Z.A. Clinical manifestations, characteristics, and outcome of infections caused by vancomycin-resistant enterococci at a tertiary care center in Lebanon: A case-case-control study. J. Infect. Public Health 2023, 16, 741–745. [Google Scholar] [CrossRef]
  215. Brasg, I.; Elligsen, M.; MacFadden, D.; Daneman, N. Predictive utility of swab screening for vancomycin-resistant Enterococcus in selection of empiric antibiotics for Enterococcus sterile-site infections: A retrospective cohort study. CMAJ Open 2017, 5, E632–E637. [Google Scholar] [CrossRef]
  216. Kram, S.J.; Kram, B.L.; Schultheis, J.M.; Kuhrt, M.M.; McRae, A.S.; Anderson, D.J. VanA rectal swab screening as a predictor of subsequent vancomycin-resistant enterococcal bloodstream infection in critically ill adults. Infect. Control Hosp. Epidemiol. 2021, 42, 411–416. [Google Scholar] [CrossRef]
  217. Gozaydin, A.; Kose, S.; Ece, G.; Ersan, G.; Gonullu, M. Detection of Vancomycin Resistant Enterococci from Rectal Swab Samples by Becton-Dickinson GeneOhm VanR assay and Culture at ICU of a Tertiary Care Center in Turkey. Pak. J. Med. Sci. 2013, 29, 682–686. [Google Scholar] [CrossRef]
Figure 1. Italian VRE isolate proportion, by year.
Figure 1. Italian VRE isolate proportion, by year.
Pathogens 13 00409 g001
Figure 2. (A) Sicilian Enterococcus Vancomycin-resistant isolate proportion per species (ENTFAE = Enterococcus faecalis and ENTFCM = Enterococcus faecium according to RETE MIC Dashboard acronyms). (B) Sicilian Enterococcus faecium and faecalis Vancomycin-resistant isolate proportion per year. Downloaded from https://qlik.qualitasiciliassr.it/anonimo/single/?appid=85ada16c-4b41-4bc6-9ca1-405b8243d0c2&sheet=6ad6f3ac3369-41c5-bd72-792243f9091b&opt=ctxmenu,currsel, accessed on 29 November 2023.
Figure 2. (A) Sicilian Enterococcus Vancomycin-resistant isolate proportion per species (ENTFAE = Enterococcus faecalis and ENTFCM = Enterococcus faecium according to RETE MIC Dashboard acronyms). (B) Sicilian Enterococcus faecium and faecalis Vancomycin-resistant isolate proportion per year. Downloaded from https://qlik.qualitasiciliassr.it/anonimo/single/?appid=85ada16c-4b41-4bc6-9ca1-405b8243d0c2&sheet=6ad6f3ac3369-41c5-bd72-792243f9091b&opt=ctxmenu,currsel, accessed on 29 November 2023.
Pathogens 13 00409 g002
Figure 3. Principal mechanisms of enterococcal antibiotic resistance. The acronyms “Vat” and “Vgb” indicates streptogramins resistance genes, while “MsrC” identifies a macrolides resistance marker. The acronyms “GdpD”, “LiaF” and “cls” indicate regulation system components whose mutations contribute to daptomycin resistance. Finally, the “PBP5” acronym identifies a penicillin-binding protein which express reduced β-lactams affinity. This figure was created by the authors using Biorender.com (app.biorender.com, accessed on 10 December 2023).
Figure 3. Principal mechanisms of enterococcal antibiotic resistance. The acronyms “Vat” and “Vgb” indicates streptogramins resistance genes, while “MsrC” identifies a macrolides resistance marker. The acronyms “GdpD”, “LiaF” and “cls” indicate regulation system components whose mutations contribute to daptomycin resistance. Finally, the “PBP5” acronym identifies a penicillin-binding protein which express reduced β-lactams affinity. This figure was created by the authors using Biorender.com (app.biorender.com, accessed on 10 December 2023).
Pathogens 13 00409 g003
Figure 4. Visual illustration of principal E. faecalis and E. faecium virulence factors. (this figure was created by the authors using Biorender.com, app.biorender.com, accessed on 10 December 2023).
Figure 4. Visual illustration of principal E. faecalis and E. faecium virulence factors. (this figure was created by the authors using Biorender.com, app.biorender.com, accessed on 10 December 2023).
Pathogens 13 00409 g004
Table 1. Summary of E. faecalis and E. faecium virulence factors in terms of biological characteristics and actions.
Table 1. Summary of E. faecalis and E. faecium virulence factors in terms of biological characteristics and actions.
Virulence FactorDescriptionReferences
Secreted factors
Cyl, haemolysin–cytolysin
  • Shows broad-spectrum binding to the extracellular matrix and is crucial for cell growth
  • Contributes to biofilm formation and lysis of red blood cells, macrophages and polymorphonuclear neutrophils
[103,104,105,106]
SagA, secreted antigen A
  • Shows broad-spectrum binding to the extracellular matrix and is crucial for cell growth
  • Contributes to biofilm formation
  • Highly immunogenic and a promising vaccine candidate
[94,107]
GelE, gelatinase
  • Plays a role in virulence in animal models of endocarditis, peritonitis, and endophthalmitis
  • Affects adherence, autolysin, and biofilm formation
  • Regulated by the Gsr quorum sensing system
  • Highly prevalent in the predominant CC17 clinical clones
[75,77,80,81,85]
sprE, serine protease
  • Serine protease that allow the pathogen to spread to cells and degrade connective components of tissue matrices.
  • These invasin can also be thought of as spreading factors that allow the bacteria to move throughout the host.
[108,109]
Cell surface determinants and their formation
Agg, aggregation substance
  • Aggregation protein involved in adherence to eukaryotic cells
  • Cell aggregation and conjugation
  • Transfer and survival of some plasmids in neutrophils
  • Role in E. faecalis endocarditis
[110,111,112,113]
efaA, Enterococcus faecium/faecalis antigen A
  • Cell wall adhesins expressed in serum by E. faecalis and E. faecium
[114,115]
Esp, Enterococcal surface protein
  • Contributes to forming biofilms and attaching to silicon-based surfaces
  • May be associated with cyl genes on a pathogenicity island
  • Acts as a virulence factor in UTI and endocarditis in animal models
  • Cell wall-associated protein involved in immune evasion
[89,90,91]
Ace and Acm, collagen-binding proteins
  • Contributes to the formation of biofilms
  • Acts as a virulence factor in UTI and endocarditis in animal models
  • Mediate adherence to collagen (Acm), and to collagen and laminin (Ace)
[89,90,91,116]
Scm, second collagen adhesin from E. fm
  • Binds to various ligands of the extracellular matrix
  • Highly prevalent in clinically related E. faecium
[89,90,91]
SgrA, EcbA
  • Surface-binding adhesins
  • Implicated in biofilm formation
  • Both serve as a markers for clinically associated E. faecium
[89,90,91,117]
epa, enterococcal polysaccharide antigen
  • Affect biofilm formation and translocation across intestinal epithelial cells
  • Role in experimental peritonitis and UTIs
  • Affect bacterial cell susceptibility to killing by polymorphonuclear neutrophils
[118]
Ebp, endocarditis- and biofilm-associated pili
  • Form pili
  • Mediate adherence to platelets, fibrinogen and collagen
  • Linked to infection
  • Contribute to adherence in biofilm formation and during UTIs
[89,114,115,119]
Other factors
Cps, capsular polysaccharides
  • Involved in adherence, virulence, and evasion of phagocytosis.
  • Crucial components for immune response
[120]
LTA, lipoteichoic acid
  • Component of the cell wall
  • Attached to lipid membrane
  • Contributes to resistance against microbes, biofilm formation, and virulence.
[92,93]
WTA, wall teichoic acid
  • Resists neutrophil killing mediated by complement
  • Involved in attaching to host cells
  • Contributes to beta-lactam resistance
[92,93]
Fsr regulator locus
  • The main regulatory system for quorum sensing in E. faecalis, it consists of the genes fsrA, fsrB, fsrD, and fsrC, which control the expression of gelatinase and serine protease.
  • Through this Fsr quorum-sensing system, biofilm formation is governed by the regulation of gelatinase production.
[74,75,76,77]
PTS, carbohydrate phosphotransferase system
  • Predominantly found in Clade A clinical isolates
  • Plays a role in intestinal colonization in a mouse model
  • BepA (PTS) contributes to in vivo biofilm formation and is associated with endocarditis in a mouse model
[99,100,101,102]
Ddl, D-Ala-D-Ala ligase
  • Non-vancomycin ligases that produce D-Ala-D-Ala, the standard cell wall precursor that increases susceptibility to glycopeptide antibiotics.
  • Changes in the ddl gene may lead to the production of ineffective or deactivated D-Ala-D-Ala ligases, potentially making bacteria reliant on glycopeptides based on the existence of vancomycin resistance clusters.
[45,87]
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

Sangiorgio, G.; Calvo, M.; Migliorisi, G.; Campanile, F.; Stefani, S. The Impact of Enterococcus spp. in the Immunocompromised Host: A Comprehensive Review. Pathogens 2024, 13, 409. https://doi.org/10.3390/pathogens13050409

AMA Style

Sangiorgio G, Calvo M, Migliorisi G, Campanile F, Stefani S. The Impact of Enterococcus spp. in the Immunocompromised Host: A Comprehensive Review. Pathogens. 2024; 13(5):409. https://doi.org/10.3390/pathogens13050409

Chicago/Turabian Style

Sangiorgio, Giuseppe, Maddalena Calvo, Giuseppe Migliorisi, Floriana Campanile, and Stefania Stefani. 2024. "The Impact of Enterococcus spp. in the Immunocompromised Host: A Comprehensive Review" Pathogens 13, no. 5: 409. https://doi.org/10.3390/pathogens13050409

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