Next Article in Journal
The Role of Copper Overload in Modulating Neuropsychiatric Symptoms
Previous Article in Journal
The Expression of Cell Cycle Cyclins in a Human Megakaryoblast Cell Line Exposed to Simulated Microgravity
Previous Article in Special Issue
Reduced Glycolysis and Cytotoxicity in Staphylococcus aureus Isolates from Chronic Rhinosinusitis as Strategies for Host Adaptation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Complex Intracellular Lifecycle of Staphylococcus aureus Contributes to Reduced Antibiotic Efficacy and Persistent Bacteremia

by
Cecilia F. Volk
1,
Richard A. Proctor
2,3,* and
Warren E. Rose
1,2,*
1
Pharmacy Practice and Translational Research Division, School of Pharmacy, Pharmacy University of Wisconsin-Madison, Madison, WI 53705, USA
2
Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
3
Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(12), 6486; https://doi.org/10.3390/ijms25126486
Submission received: 20 May 2024 / Revised: 3 June 2024 / Accepted: 7 June 2024 / Published: 12 June 2024
(This article belongs to the Special Issue Host-Pathogen Interactions during Persistent Bacterial Infections)

Abstract

:
Staphylococcus aureus bacteremia continues to be associated with significant morbidity and mortality, despite improvements in diagnostics and management. Persistent infections pose a major challenge to clinicians and have been consistently shown to increase the risk of mortality and other infectious complications. S. aureus, while typically not considered an intracellular pathogen, has been proven to utilize an intracellular niche, through several phenotypes including small colony variants, as a means for survival that has been linked to chronic, persistent, and recurrent infections. This intracellular persistence allows for protection from the host immune system and leads to reduced antibiotic efficacy through a variety of mechanisms. These include antimicrobial resistance, tolerance, and/or persistence in S. aureus that contribute to persistent bacteremia. This review will discuss the challenges associated with treating these complicated infections and the various methods that S. aureus uses to persist within the intracellular space.

1. Introduction

Antibiotic resistance in Staphylococcus aureus continues to pose significant risk to the healthcare landscape due to limited antimicrobial options [1]. However, persistent S. aureus infections present a major challenge to clinicians during the treatment of “susceptible infections” in patients. This is particularly true for persistent S. aureus bacteremia, characterized by repeatedly positive Staphylococcus aureus cultures in the bloodstream despite apparently appropriate antibiotic therapy [2]. This condition is associated with increased morbidity, mortality, and healthcare costs, making its management a priority in clinical practice [2,3]. From this perspective, we overview the problem of persistent bacteremia caused by S. aureus and highlight the intracellular proclivity of the organism as a mechanism for survival in the host and defense against antibiotic treatments.

2. The Clinical Challenges of Persistent Staphylococcus aureus Bacteremia (SAB)

Persistent SAB is defined by repeat positive blood cultures over multiple days; however, the number of days required for an infection to be considered persistent is widely debated, with more contemporary definitions shifting towards earlier cutoffs that support early switching to alternative regimens [2]. This has aligned with rapid advances in molecular organism identification (i.e., MALDI-TOF and others) and automated antimicrobial susceptibility testing [4,5]. However, because this definition is not standardized, it therefore allows for a range of in-practice interpretations of when to consider changes in antibiotic therapy [2]. Traditionally, and informed from historical clinical trials, the definition of persistent bacteremia derived from the average duration of bacteremia among clinical trials of antibiotic therapy—around 7 days [6,7]. Since then, several studies have suggested earlier definitions of persistent SAB (the 3-5-day duration of positive cultures) due to improved outcomes noted with shorter durations of bacteremia [3,8,9]. For example, we argued persistent or prolonged bacteremia be considered as the >4-day duration of positive cultures based upon the observed increase in mortality and cytokine markers associated with this threshold from regression analysis [8,10]. This time frame corresponds with the final identification and susceptibility profile of S. aureus obtained from blood cultures accounting for time to growth in blood culture vials, identification by molecular methods, and phenotypic susceptibility. However, recently, Holland et al. proposed considering persistent bacteremia earlier, after one calendar day, for the further diagnostic evaluation and consideration of antibiotic failure for implementing alternative treatment regimens [2]. This suggestion is supported by a study by Kuehl et al., which found that patients who had positive blood cultures beyond 24 h of antibiotic therapy were at a higher risk of mortality and metastatic infections [11]. The overwhelming clinical evidence implies an excessive morbidity and mortality risk of persistent bacteremia for patients and recommends a low threshold for early antibiotic changes for rapid bacteremia clearance.

3. Treatment Approaches for Persistent SAB

Several treatment options still exist for S. aureus; however, the treatment of complicated bacteremia is limited to only a handful of approved agents. This includes anti-staphylococcal β-lactams (nafcillin or oxacillin) and targeted cephalosporins (i.e., cefazolin) for MSSA, while vancomycin or daptomycin are typically reserved for MRSA. Recently, ceftobiprole received FDA approval for MSSA and MRSA bacteremia and endocarditis based on a randomized controlled trial demonstrating its non-inferiority to daptomycin [12], and ceftaroline has been used similarly without indication [13,14,15]. Following induction with these antibiotics, other agents are commonly used as step-down therapy, including linezolid (IV/PO), the lipoglycopeptides dalbavancin and oritavancin, and many oral options including β-lactams, trimethoprim–sulfamethoxazole, and clindamycin [16]. Clearly, there is a diverse range of options for the treatment of SAB. However, these options considerably narrow when confronted with MRSA or in patients with persistent bacteremia [3,16].
The successful management of persistent SAB requires a comprehensive approach that addresses both host and pathogen factors. Initial management involves timely and appropriate antibiotic therapy guided by antimicrobial susceptibility testing [17]. However, given the potential for antibiotic resistance and/or tolerance, combination therapy with agents targeting different bacterial pathways has been used successfully to improve bacterial clearance [3,16]. This is of highest concern in persistent MRSA bacteremia, where treatment options are extremely limited once the standard-of-care agents are deemed unsuccessful. Any randomized controlled trial of SAB implements a non-inferiority design [12,18,19,20,21]. No randomized controlled trial exists to understand the optimal treatment of persistent SAB; therefore, treatment approaches are guided by observational and retrospective case–control studies. From this evidence, treatment paradigms have been proposed to consider how to address persistent MRSA bacteremia and include combination therapy with priority towards daptomycin plus ceftaroline followed by daptomycin plus an antistaphylococcal beta-lactam, and then, finally, vancomycin plus a hydrophilic beta-lactam to reduce the risk of nephrotoxicity (e.g., cefazolin, ceftaroline) [16]. In addition to antibiotic therapy, the removal of potential sources of infection, such as infected intravascular devices or sites of metastatic infection, is essential. This may involve the removal or exchange of indwelling catheters, the debridement of infected tissues, or the surgical drainage of abscesses. Close monitoring for complications, such as infective endocarditis or metastatic infections, is also imperative during persistent bacteremia [16].
While antibiotic therapy is crucial for a successful outcome in SAB, host factors also play a significant role in the persistence of S. aureus and must be carefully evaluated and managed [22]. Underlying conditions that compromise host immune function, such as diabetes mellitus or immunosuppressive therapy, must be evaluated and managed when possible. Some host characteristics can be either modifiable or non-modifiable. For example, patients with diabetes can have blood glucose managed within the normal range; however, the underlying immunologic suppression and ischemia from this conditional is unchanged, which prevents the complete eradication of complicating and contributing factors in these patients. Optimal supportive care, including nutritional support and management of comorbidities, is also essential to enhance host defenses and improve outcomes. However, the host–pathogen dynamic in SAB remains poorly understood [23,24,25]. This is evident from the failed vaccination attempts in human trials aimed at preventing S. aureus invasive infections despite promising animal model data [23]. Only recently has the underlying host immunologic pathology occurring at the onset and during SAB begun to be understood. From these studies, it is noted that profound cytokine imbalances exist in patients with SAB [8,23,24,26,27]. The pro-inflammatory cytokine response appears to be important during early initial response and clearance bacteremia (e.g., IL-1β, IL-2, IL-6, TNF-α, and glutamine) [8,27]. In terms of survival outcome, high levels initially and throughout the duration of bacteremia appear to be detrimental either to excessive pro-inflammatory response (e.g., IL-6 and TNF) or host-immune paralysis (e.g., IL-10, IL-6, IL-17, CCR2, T4, adiponectin) [10,23,28,29]. Related to persistent bacteremia, a dampening of the pro-inflammatory cytokines IL-1β and TNF-α has been linked to the duration of bacteremia as a result of the pathogens’ response to antibiotic exposure and ability to evade host defenses through multiple mechanisms [8,10,27,28].
An additional factor that often complicates treatment is the diversity of toxins and virulence factors that S. aureus can produce. Multiple studies have linked differential toxin expression to patient outcomes in a variety of S. aureus infection types, including bacteremia [23,30,31,32]. Importantly, elevated innate levels of certain anti-toxin antibodies can be protective against severe disease [33,34,35]. These important observations have led to the evaluation of anti-toxin antibodies in early-stage clinical trials, including AR-301, an anti-α-toxin monoclonal antibody tested as an adjunctive therapy in patients with hospital-acquired bacterial pneumonia caused by S. aureus [36]. This trial only included 48 patients but found shorter durations of ventilation and faster microbiologic eradication in the patients who received the antibody. Antibiotics with anti-toxin effects are also being evaluated as adjunctive therapies for SAB. Specifically, clindamycin, a protein synthesis inhibitor that inhibits toxin production, was evaluated via an open-label randomized controlled trial [37]. This trial included 34 patients with severe S. aureus infections and found a lower mortality rate in patients receiving adjunctive clindamycin (0% vs. 24%). This treatment is currently being evaluated further in the S. aureus Network Adaptive Platform (SNAP) randomized controlled trial [38]. These preliminary studies highlight the likely importance of toxin inhibition in managing SAB.
In summary of its clinical importance and treatment challenges, persistent SAB poses a significant clinical problem, characterized by ongoing bloodstream infection despite appropriate therapy. Successful management requires a multifaceted approach that addresses both pathogen and host factors. Timely and appropriate antibiotic therapy, the removal of potential sources of infection, the optimization of host defenses, and efforts to prevent recurrent infections are key components of management. Ultimately, persistent SAB represents a complex clinical syndrome that requires a multidisciplinary approach involving infectious disease specialists, clinical microbiologists, pharmacists, nurses, and other healthcare professionals. Collaboration between specialties is essential to ensure timely diagnosis, appropriate treatment, and ongoing management of this challenging condition. However, the identification of why S. aureus persists from an organism, host, and mechanistic level remains key to solving this issue.

4. Small-Colony-Variant S. aureus Results in Reduced Antibiotic Efficacy and Infection Persistence

Although the reduction in the duration of SAB has been a noble goal over the last three decades, the mechanistic understanding of how and why S. aureus persists when susceptible to antibiotic therapy remains elusive. There are several proposed mechanisms contributing to this phenomenon. S. aureus has metabolic adaptations to survive and hide from the immune systems of humans. One of the most studied phenotypes contributing to persistence is the small colony variant (SCV) of S. aureus. This is a unique phenotype characterized by distinct morphological and growth characteristics [39]. SCVs contribute to the persistence of many invasive infections including bacteremia, pneumonia, and osteomyelitis, among others, and SCVs typically exhibit slow growth rates and form small, non-pigmented colonies on blood agar [39,40,41]. SCVs remain difficult to detect using standard laboratory methods for surveillance studies due to the lack of dedicated screening methods and noted instability of the phenotype, but they may contribute to up to 30% of invasive S. aureus infections [40].
Small colony variants were first described in 1911 as a slow-growing subpopulation [42] and later noted for their difference in “color, texture, and viscidity” [43]. Early research identified SCV occurrence correlated to harsh environmental conditions such as high salt content [44]. The clinical impact of SCVs arose in the early 1990s by identifying this phenotype as a cause of persistent and relapsing S. aureus infections [39]. Since this time, the understanding of the clinical relevance and emergence of SCVs has appeared through genetic mutations or adaptive responses to various environmental stresses during infection in patients, such as exposure to antibiotics or host immune defenses. These mutations often result in alterations in metabolic pathways, leading to changes in bacterial physiology and virulence [45]. Clinical manifestations of SCV infections vary depending on the site of infection (e.g., endocarditis vs. osteomyelitis) and underlying host factors [45,46].
A hallmark feature of SCVs is their reduced susceptibility to antibiotics, particularly aminoglycosides and cell wall-active agents like β-lactams [40,47]. This reduced susceptibility is attributed to decreased metabolic activity and alterations in membrane potential, which impair antibiotic uptake and efficacy through reductions in the production of active targets such as peptidoglycan and penicillin-binding proteins [45,48,49,50]. In addition to resistance to these key antibiotics, particularly β -lactams for MSSA, SCVs have reduced susceptibility to standard-of-care agents for MRSA—vancomycin—through reduced peptidoglycan formation and daptomycin since active membrane potential is required for activity [40,48,51]. The management of SCV infections is complex and often involves prolonged courses of antibiotic therapy tailored to the specific susceptibility profile of the isolate. Antimicrobials with high intracellular uptake, such as fluoroquinolones, may be preferred, when susceptible, to treat the intracellular reservoir [48]. Combination therapy may be necessary to enhance bacterial clearance and prevent the emergence of resistance, but the optimal combination for SCV treatment is not established [47,48]. The lipoglycopeptide oritavancin, which has the dual inhibition of peptidoglycan and cell membrane synthesis, may have specific activity against S. aureus SCVs [52,53]. Natural products also are being explored against SCVs, including tomatidine and its derivatives, which have been noted to block F0F1ATPase [54]. These molecules also synergize with aminoglycosides and prevent SCV formation [55].

5. Antimicrobial Resistance, Tolerance, and Persistence in S. aureus Contribute to Persistent Bacteremia

While major attention is rightfully paid toward antimicrobial resistance in S. aureus, the occurrence of antimicrobial tolerance and persistence also represents significant issues for antimicrobial therapy [56,57,58,59]. Observations of antimicrobial tolerance and persistence in S. aureus represent a growing area of interest and investigation [60,61]. Antimicrobial tolerance is distinct from antimicrobial resistance in that tolerant strains appear to be susceptible upon MIC testing in liquid culture. The nongrowing or slowly growing bacterial population remains viable and can “tolerate” or survive temporary antimicrobial exposure [62] (Figure 1). These tolerant populations may enter a dormancy state or reduce cellular processes to survive antimicrobial pressure through reductions in active targets (e.g., reduced cell-wall replication) [60]. Antimicrobial tolerance may develop through either a genetic mutation triggered by environmental conditions (e.g., reduced oxygen or nutrients leading to mutation(s) in TCA cycle genes) or antibiotics (single-point mutations) [58,60,63]. In contrast to tolerant cells, persisters are a subset of the bacterial population that can survive high concentrations of antimicrobial exposure. During treatment, the majority of the population may be killed by therapeutic concentrations, leaving the “persister cells” remaining to subvert antimicrobial treatment (Figure 1) [60]. Similar to tolerant cells, persisters may enter a state of dormancy and altered metabolism during this phase [64,65,66]. Once the antimicrobial exposure is removed, these cells may begin to replicate, leading to infection recurrence [64]. While SCVs described previously in detail can also be tolerant and persisters to antibiotics, these phenomena are not restricted to SCVs. Different mechanisms for SCVs and persisters exist, but a commonality between these two phenotypes is the decreased ability to make ATP [67].
An added layer to antimicrobial tolerance and persistence is the ability of S. aureus to produce robust biofilms [68,69]. These are complex communities of bacteria encased within a self-produced extracellular matrix consisting of polysaccharides, proteins, and extracellular DNA [70]. Biofilms provide protection against antibiotics and host immune responses, allowing bacteria within the biofilm to survive and persist despite antibiotic exposure [71,72]. Both tolerant bacterial populations and persister cells occur deep within the biofilm, and treatment is further compromised by a lack of antibiotic penetration through the extracellular matrix [73,74]. The low oxygen environment of biofilms combined with high bacterial burden may also reduce ATP and membrane potential [75]. In bacteremia, biofilms are known to play an important role in the pathology of the infection sources including endocarditis, prosthetic devices, and osteomyelitis [69].
Antimicrobial tolerant and persistent S. aureus pose a significant clinical challenge as they can lead to treatment failure, recurrent infections, and the spread of antibiotic resistance [60,76]. Arguably, antimicrobial tolerance is responsible for persistently positive SAB (consecutive days of bacteremia), while persister cells are responsible for bacteremia recurrence (positive cultures after day(s) of negative cultures) (Figure 1). The effective management of antimicrobial tolerant/persistent S. aureus infections often requires a multifaceted approach, including the use of combination antibiotic therapy, the removal of biofilm-associated infections, and the development of novel antimicrobial strategies to target tolerant/persistent bacterial populations [60]. However, the challenge of identifying these strains is a significant barrier for the early recognition and initiation of targeted therapy. Currently, the assays to test for tolerant/persistent S. aureus are experimental, and identifying these strains only occurs through the examination of an altered colony phenotype (e.g., SCV) or infection progression despite antibiotic therapy [56]. Understanding potential reservoirs for S. aureus tolerant populations and persistent cells is important for improving the response to antimicrobial therapy.

6. Proclivity of Staphylococcus aureus for Intracellular Growth and Persistence

Although S. aureus is traditionally considered an extracellular pathogen, there is now substantial evidence supporting the presence of an intracellular niche for this organism [77]. Using in vitro assays, S. aureus has been shown to survive within a wide variety of cell types, including epithelial cells, endothelial cells, fibroblasts, osteoblasts, osteocytes, keratinocytes, macrophages, and neutrophils [64,78,79,80,81,82,83]. These bacteria have often been shown to be in a semi-dormant, non-culturable, small-colony-variant state [83,84]. Evidence of intracellular S. aureus persistence as a cause of human disease has been harder to prove, although a few studies have successfully done so. The presence of intracellular S. aureus during human infections has now been demonstrated in patients with recurrent rhinosinusitis, tonsillitis, osteomyelitis, and bacteremia [83,85,86,87,88,89]. Importantly, these studies all examined patients with recurrent or chronic infections. This evidence highlights intracellular survival as an important part of the lifecycle of persistent S. aureus infections.
The evidence for intracellular S. aureus as a contributing mechanism of persistent S. aureus infections is evident by the research and understanding of SCVs in this environment. S. aureus SCVs have a noted preference for increased intracellular uptake because of bacterial surface proteins that bind host receptors, such as epithelial cells and macrophages [67]. This intracellular lifestyle enables SCVs to evade innate host defenses and antibiotics, leading to chronic or recurrent infections that are challenging to eradicate, contributing to persistent or relapsing infections. However, this proclivity for intracellular invasion is not restricted to SCVs, and S. aureus, regardless of colony phenotypes, becomes an intracellular pathogen.

7. Intracellular S. aureus Reduces Immune Recognition and Activation

S. aureus that persists intracellularly does so by avoiding the normal immune system activation during acute infection. Much of the typical immune activation is regulated by the accessory gene regulator (agr) system. Agr is a quorum-sensing system that regulates the expression of many toxins and other inflammatory factors [90] including α-toxin, which is known to induce inflammation and cell death when produced in an intracellular environment [91,92,93]. Several studies have tested clinical isolates from persistent S. aureus infections and found that these strains tend to have an agr deficiency [67,94,95,96]. Similarly, SCVs have been shown to have decreased agr expression [67]. In vitro studies have demonstrated that these agr-deficient strains have a higher rate of cellular uptake but lower induction of inflammatory responses [90,97]. SCVs have demonstrated lower production levels of IL-1β, IL-6, and IL-12 than wild-type S. aureus after the infection of epithelial cells [98]. In fact, a mouse model of chronic osteomyelitis infection has demonstrated that inflammatory markers return to normal levels during the infection, despite the persistent bacteria remaining intracellularly [99]. In addition, a study of patients with chronic rhinosinusitis with nasal polyps found no increase in the number of eosinophils, lymphocytes, and neutrophils in tissue samples, despite the presence of intracellular S. aureus [86]. Overall, this decreased immune activation allows for S. aureus to hide in the intracellular environment and largely avoid the host immune system.
There are multiple methods that S. aureus can utilize to survive in an intracellular environment. For S. aureus to enter host cells, the upregulation of alternative sigma factor B (SigB) is required. This regulatory system modulates the S. aureus stress response through the transcription of genes that control resistance to heat and oxidative and antibiotic stresses and contributes to the SCV phenotype [67]. After uptake into host cells, S. aureus is typically taken up into phagosomes. This is consistent for both phagocytic cells and non-professional phagocytes such as epithelial cells, osteoblasts, and fibroblasts [92,100,101,102]. The phagolysosome environment has many antibacterial properties, including an acidic pH of 4.5 [25]. S. aureus is capable of not only tolerating an acidic environment, but also deacidifying the environment through the production of ammonia [103]. Although the downregulation of agr is important for initial cell invasion, as noted above, the intracellular acidic environment then enhances the expression of the agr system, increasing the survival of S. aureus intracellularly [104,105,106]. This effect is likely strain specific [107,108]. S. aureus is also capable of protecting itself from defensins and other antimicrobial peptides within the phagosome environment. The GraRS system leads to the upregulation of multiple peptide resistance factor (MprF) which confers protection from host defense peptides by enhancing the lysinylation of phosphatidylglycerol to the outer portion of the cellular membrane [109]. Similarly, the dlt operon (dltABCD) and oatA lead to alterations in membrane teichoic acids and peptidoglycan, respectively, which lead to protection from the phagosome environment [109,110]. Finally, S. aureus may produce protective enzymes (catalase, superoxide dismutase, etc.) to protect themselves from reactive oxygen species produced within the phagosome [111,112]. An increase in SodM, an enzyme responsible for detoxifying reactive oxygen species, was discovered in S. aureus isolates from patients with persistent cystic fibrosis infections [113]. In vitro assays have determined that this increase is noted specifically after the internalization of S. aureus into airway epithelial cells [114].
Despite these adaptations to the harsh phagosome environment, many types of S. aureus will exit the phagosome as a method to increase survival. This phagosomal escape is an agr-mediated process, as agr mutant strains are not capable of escaping to the cytosol. An increase in agr expression has been measured just prior to phagosomal escape [115,116]. Phenol-soluble modulins (PSMs), agr-dependent cytotoxic peptides that are activated during the stringent response, have been proven to play an important role in escape into the cytosol [115,117,118,119,120]. The concentration of PSMs has been similarly shown to increase just prior to S. aureus phagosomal escape, which has proven to be a required component in this process [117,121]. Other S. aureus escape factors, including a non-ribosomal peptide synthetase and a Tet38 efflux pump, have been identified, but their mechanisms are less clear [122,123]. In addition to these mechanisms, S. aureus may escape phagosomal destruction during overwhelming infection. In these situations, host cells are not able to tackle the high bacterial burden and become exhausted, creating an intracellular niche for S. aureus replication [124,125,126]. After phagosomal escape, replication within the cytosol leads to host cell lysis and the release of S. aureus [108]. These released cells may be re-phagocytosed, leading to a cycle that maintains a portion of intracellular S. aureus [126,127]. This maintenance of intracellular S. aureus can also contribute to the spreading of infection to other host sites, when mobile phagocytes are infected [126,128].
The above survival mechanisms appear to contradict the fact that persistent infections are often caused by agr-deficient S. aureus strains, as these strains would be unable to escape the phagosome due to lack of PSMs [67,94,95,96]. Indeed, these agr-deficient strains employ different survival methods that do not require PSM-mediated phagosomal escape. Siegmund et al. determined that PSM-deficient S. aureus has a higher overall survival rate within endothelial cells [129]. The surviving bacteria were co-localized with LC3, a marker of autophagy, indicating that these bacteria can survive within vesicles, likely by interfering with lysosomal recruitment and the autophagy process. Unlike the process of phagosome escape and host cell lysis presented above, this PSM-independent process allows for a consistent niche of intracellular growth.

8. Anaerobic Metabolism of S. aureus Correlates to Intracellular Growth

There is a growing body of evidence to suggest that S. aureus utilizes anaerobic metabolism during persistent intracellular infections. Figure 2 presents a working model of key interactions of intracellular S. aureus metabolism and host factors leading to persistence. S. aureus SCVs have demonstrated a decrease in TCA cycle activity and a corresponding increase in glycolytic activity compared to wild-type strains, an effect that is consistent across all auxotrophic types of SCVs [50]. This effect, however, does not seem to be restricted to S. aureus with the SCV phenotype. One study conducted whole-genome sequencing of 206 MRSA isolates from patients with persistent bacteremia [130]. The authors found frequent mutations in genes involved in the TCA cycle (citZ and odhA). Mutations in these genes have been linked to antibiotic tolerance [65,66], which may help explain persistence despite adequate antimicrobial treatment. Similarly, S. aureus isolates from persistent cystic fibrosis infections have the downregulation of many proteins involved in amino acid metabolism, fatty acid metabolism, and energy metabolism [113,131]. These studies also found a reduction in the expression of proteins in the phosphoenolpyruvate carbohydrate phosphotransferase systems, which are responsible for the transport of glucose into the bacterial cell, a necessary precursor to glycolysis.
The redox regulator Rex inhibits important genes involved in anaerobic metabolism, including lactate dehydrogenase and alanine dehydrogenase. Rex plays an important role in activating anaerobic metabolism in response to intracellular uptake or in response to high concentrations of NADH or NO. RsaG is an sRNA that inhibits the expression of Rex [132,133]. RsaG is expressed in response to high levels of glucose-6-phosphate, as frequently found in the host cell cytosol or mucoid secretions. An increase in RsaG has been noted during in vitro internalization assays using both myoblasts and macrophages, as well as after exposure to mucus-secreting epithelial cells. This increase in RsaG therefore triggers a switch to anaerobic metabolism via the de-repression of Rex-regulated proteins. Similarly, the inhibition of Rex has been observed in response to high levels of NO, a signaling molecule utilized during wound repair that is present at high levels in lung epithelial cells in patients with inflammatory diseases such as asthma or COPD [134,135,136]. Rex is also the major regulator of staphylococcal respiratory response AB genes (srrAB), a two-component system that regulates virulence factors and genes involved in anaerobic metabolism [137,138,139]. Mutations in srrAB allow for S. aureus SCVs to grow rapidly yet retain aminoglycoside resistance that was acquired during the SCV phase [140]. S. aureus glycolysis is also necessary for the upregulation of itaconate production, as demonstrated in a mouse pulmonary infection model. High itaconate levels in turn lead to the production of extracellular polysaccharide and biofilm formation [136].
Like bacterial cells, host cell metabolism also plays an important role in immune response. Hypoxia-inducible factor 1α (hif1α) is an important gene that controls both immune response and cellular metabolism [141]. HIF1α activation stimulates the production of pro-IL-1β, an important part of the inflammasome response, and promotes glycolysis. Importantly, HIF1α expression is induced during S. aureus infection of human cells, and the metabolic state of the bacterial cell plays an important role in this expression [141,142]. S. aureus mutants deficient in cellular glycolysis are unable to stimulate host cell HIF1α expression and therefore host cell glycolysis [141]. Contrarily, S. aureus SCV infection stimulates more host glycolysis than wild-type S. aureus infection, an effect that has been demonstrated in multiple host cell types [142]. Bacterial glycolysis appears to also play an important role in establishing infection, an effect that has been demonstrated using an in vivo mouse cutaneous infection model. Mice that were infected with glycolysis-deficient S. aureus demonstrated a significantly lower bacterial burden than those infected with wild-type strains [141]. These findings highlight the complex and important interplay between bacterial and host metabolic processes.
Host cell anaerobic metabolism has an important secondary cellular consequence, the induction of necroptosis pathways [142]. Necroptosis is a type of host cell death that, unlike apoptosis, releases viable bacteria, thus promoting bacterial persistence [143]. Necroptosis is further upregulated by SCVs compared to wild-type S. aureus and is not activated when host cells are infected by glycolysis-deficient S. aureus. An in vivo mouse model demonstrated that mice unable to utilize necroptosis had greater S. aureus persistence compared to wild-type mice. This pathway provides an important mechanistic link between bacterial anaerobic metabolism and infection persistence.
Another notable effect of anaerobic metabolism is the inhibition of trained immunity. Trained immunity is the process wherein the innate immune system develops memory of an infection and is therefore strengthened against re-infection with the same organism [144]. Unlike the adaptive immune response, this memory is thought to be accomplished through a series of epigenetic changes and is highly regulated by the amount of intracellular fumarate [145,146]. Fumarate is a TCA cycle substrate that induces both the trained immunity response and glycolysis [147]. Infection with S. aureus SCVs leads to lower amounts of intracellular fumarate than infection with wild-type strains, an effect that is mediated by an increase in fumC activity, an enzyme that breaks down fumarate [142]. Therefore, SCVs do not induce trained immunity to the degree of wild-type S. aureus, as validated using an in vivo mouse model. Even in the presence of a mixed SCV and wild-type population, the excess fumC activity from the SCV isolates prevents the trained immunity response. Of note, non-SCV strains of S. aureus also can enhance the production of FumC [148,149]. This increases susceptibility to infection upon future S. aureus exposure.

9. Intracellular S. aureus Is More Resistant to the Effects of Antibiotics through Multifaceted Mechanisms

The ability for S. aureus to survive in the intracellular environment provides protection from many antibiotics. These mechanisms are outlined in Figure 3. First, most antibiotics are either unable to penetrate the intracellular space or lack sufficient penetration for microbial killing. For example, β-lactams, an important class in the clinical management of S. aureus infections, do not accumulate within phagocytes, despite their ability to diffuse through membranes [150]. This is thought to be due to their weak acidity, which leads to lower accumulation within the already acidic intracellular space [151]. Aminoglycosides are unable to cross the membrane due to their polarity but have been noted to enter the intracellular space by endocytosis [152]. This process requires a longer duration of drug exposure and ultimately leads to localization within lysosomes. However, optimizing aminoglycoside exposures for this effect is limited by their well-known toxicities, namely nephrotoxicity and ototoxicity [153]. Conversely, lincosamides (clindamycin), macrolides, and fluoroquinolones all demonstrate accumulation within the intracellular space [150].
Interestingly, the intracellular accumulation of antibiotics does not always correlate with the antibiotic’s activity within that space. One study evaluating the intracellular activity of a variety of antibiotics found that intracellular activity was consistently lower than extracellular, despite the high intracellular accumulation of some of the antibiotics used [154]. Overall, the extent of antibiotic activity was greatly dependent on both the concentration and time of antibiotic exposure for all agents. This disconnect between intracellular concentration and activity points to other important factors that affect antibiotic activity within the intracellular environment.
Intracellular bacteria undergo several changes that lead to increased antibiotic resistance. In SCVs, the pattern of antimicrobial resistance is dependent on the specific auxotrophic type. Hemin or menadione auxotrophic SCVs are typically resistant to aminoglycosides [40]. The inhibition of electron transport within these cells leads to a reduction in the electrochemical gradient across the membrane. This gradient is required for the uptake of aminoglycosides into S. aureus. The other type of SCV, thymidine auxotrophs, are resistant to sulfa antibiotics (trimethoprim–sulfamethoxazole) as a result of disruption of the tetrahydrofolic acid pathway [155]. Thymidine auxotrophy results in reduced ClpC activity, which is needed to activate aconitase, an enzyme involved in the intraconversion of the tricarboxylic acid citrate, cis-aconitate, and isocitrate in the TCA cycle [156]. Hence, the reduced TCA activity results in an SCV similar to menadione and hemin auxotrophs [155,157]. The slow-growing SCV phenotype also leads to decreased susceptibility to cell wall-active antibiotics, such as β-lactams and vancomycin [158].
Because all three SCV auxotrophic classes lead to disruption of the electron transport chain, SCVs are deficient in ATP. This ATP depletion has been linked to the formation of persister cells from exponential-phase normal-colony phenotype S. aureus [159]. An in vitro study found that persister cells arise due to a stochastic change from the exponential to stationary growth phase, a change that was accompanied by a large decrease in ATP levels. This change to the stationary phase was associated with a 100- to 1000-fold increase in survival after antibiotic challenge. Another study found that the inactivation of enzymes involved in the TCA cycle (sucA or sucB) increased the formation of persister cells in stationary-phase cultures [65]. Interestingly, this study did not find a consistent decrease in ATP levels amongst persister cells formed during the stationary phase, but it did note lower membrane potential. This effect was confirmed pharmacologically using a proton motive force inhibitor, which increased the formation of persister cells. As noted earlier, tomatidine and related derivatives inhibit bacterial ATP synthase (F0F1ATPase), which specifically kills S. aureus SCVs [54]. This selectivity in S. aureus, and not mammalian cells, points to the use of F0F1ATPase in auxotrophic SCVs to create a membrane gradient despite low ATP [160]. The inhibition of this enzyme is fatal for S. aureus as it collapses the membrane potential but leaves untargeted host cells unharmed [55].
Peyrusson et al. demonstrated that intracellular S. aureus persisters are produced because of antibiotic exposure. These bacteria exist in a non-dividing state and display activation of the stringent response and other stress responses [64]. The decreased metabolic state associated with intracellular persistence leads to the downregulation of many cellular processes that are the targets of bactericidal antibiotics, such as fluoroquinolones, aminoglycosides, and β-lactams, and causes widespread antibiotic tolerance. Collectively, the intracellular host environment along with the altered S. aureus metabolism (through multiple mechanisms) required to survive in this environment dramatically reduces the effect of common antibiotics used in bacteremia treatment, setting the stage for treatment failure.

10. Conclusions

Mortality from SAB has remained unacceptably high over the course of decades of improvements in diagnostics and therapeutics. In this review, we have summarized evidence on the role of tolerant and persistent populations of S. aureus compounded by their proclivity for anaerobic metabolism and intracellular invasion to escape host recognition and antibiotic treatment. This evidence suggests that intracellular S. aureus is a significant contributor to persistent bacteremia and treatment failure in patients. Current therapeutics either lack activity against tolerant and persistent populations due to their inability to kill quiescent cells and/or lack sufficient host cell penetration to eliminate intracellular S. aureus. Further mechanistic understanding of S. aureus intracellular invasion combined with targeting these bacterial populations for therapeutic treatment will be essential to reducing the prevalence of persistent SAB among patients.

Author Contributions

Conceptualization, R.A.P. and W.E.R.; writing—original draft preparation, C.F.V.; writing—review and editing, C.F.V., R.A.P. and W.E.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

W.E.R. has received consulting honoraria from Melinta and Basilea.

References

  1. Rodvold, K.A.; McConeghy, K.W. Methicillin-Resistant Staphylococcus aureus Therapy: Past, Present, and Future. Clin. Infect. Dis. 2014, 58, S20–S27. [Google Scholar] [CrossRef] [PubMed]
  2. Holland, T.L.; Bayer, A.S.; Fowler, V.G. Persistent Methicilin-Resistant Staphylococcus aureus Bacteremia: Resetting the Clock for Optimal Management. Clin. Infect. Dis. 2022, 75, 1668–1674. [Google Scholar] [CrossRef] [PubMed]
  3. Kullar, R.; Sakoulas, G.; Deresinski, S.; van Hal, S.J. When sepsis persists: A review of MRSA bacteraemia salvage therapy. J. Antimicrob. Chemother. 2015, 71, 576–586. [Google Scholar] [CrossRef] [PubMed]
  4. Bauer, K.A.; Perez, K.K.; Forrest, G.N.; Goff, D.A. Review of rapid diagnostic tests used by antimicrobial stewardship pro-grams. Clin. Infect. Dis. 2014, 59 (Suppl. 3), S134–S145. [Google Scholar] [CrossRef]
  5. Patel, R.; Fang, F.C. Diagnostic Stewardship: Opportunity for a Laboratory–Infectious Diseases Partnership. Clin. Infect. Dis. 2018, 67, 799–801. [Google Scholar] [CrossRef] [PubMed]
  6. Markowitz, N.; Quinn, E.L.; Saravolatz, L.D. Trimethoprim-Sulfamethoxazole Compared with Vancomycin for the Treatment of Staphylococcus aureus Infection. Ann. Intern. Med. 1992, 117, 390–398. [Google Scholar] [CrossRef] [PubMed]
  7. Levine, D.P.; Fromm, B.S.; Reddy, B.R. Slow Response to Vancomycin or Vancomycin plus Rifampin in Methicillin-resistant Staphylococcus aureus Endocarditis. Ann. Intern. Med. 1991, 115, 674–680. [Google Scholar] [CrossRef] [PubMed]
  8. Rose, W.E.; Eickhoff, J.C.; Shukla, S.K.; Pantrangi, M.; Rooijakkers, S.; Cosgrove, S.E.; Nizet, V.; Sakoulas, G. Elevated Serum Interleukin-10 at Time of Hospital Admission Is Predictive of Mortality in Patients with Staphylococcus aureus Bacteremia. J. Infect. Dis. 2012, 206, 1604–1611. [Google Scholar] [CrossRef] [PubMed]
  9. Kullar, R.; McKinnell, J.A.; Sakoulas, G. Avoiding the Perfect Storm: The Biologic and Clinical Case for Reevaluating the 7-Day Expectation for Methicillin-Resistant Staphylococcus aureus Bacteremia Before Switching Therapy. Clin. Infect. Dis. 2014, 59, 1455–1461. [Google Scholar] [CrossRef] [PubMed]
  10. Rose, W.E.; Shukla, S.K.; Berti, A.D.; Hayney, M.S.; Henriquez, K.M.; Ranzoni, A.; Cooper, M.A.; Proctor, R.A.; Nizet, V.; Sakoulas, G. Increased Endovascular Staphylococcus aureus Inoculum Is the Link Between Elevated Serum Interleukin 10 Concentrations and Mortality in Patients with Bacteremia. Clin. Infect. Dis. 2017, 64, 1406–1412. [Google Scholar] [CrossRef]
  11. Kuehl, R.; Morata, L.; Boeing, C.; Subirana, I.; Seifert, H.; Rieg, S.; Bin Kim, H.; Kim, E.S.; Liao, C.-H.; Tilley, R.; et al. Defining persistent Staphylococcus aureus bacteraemia: Secondary analysis of a prospective cohort study. Lancet Infect. Dis. 2020, 20, 1409–1417. [Google Scholar] [CrossRef]
  12. Holland, T.L.; Cosgrove, S.E.; Doernberg, S.B.; Jenkins, T.C.; Turner, N.A.; Boucher, H.W.; Pavlov, O.; Titov, I.; Kosulnykov, S.; Atanasov, B.; et al. Ceftobiprole for Treatment of Complicated Staphylococcus aureus Bacteremia. N. Engl. J. Med. 2023, 389, 1390–1401. [Google Scholar] [CrossRef] [PubMed]
  13. Sakoulas, G.; Moise, P.A.; Casapao, A.M.; Nonejuie, P.; Olson, J.; Okumura, C.Y.; Rybak, M.J.; Kullar, R.; Dhand, A.; Rose, W.E.; et al. Antimicrobial Salvage Therapy for Persistent Staphylococcal Bacteremia Using Daptomycin Plus Ceftaroline. Clin. Ther. 2014, 36, 1317–1333. [Google Scholar] [CrossRef]
  14. Rose, W.E.; Schulz, L.T.; Andes, D.; Striker, R.; Berti, A.D.; Hutson, P.R.; Shukla, S.K. Addition of Ceftaroline to Daptomycin after Emergence of Daptomycin-Nonsusceptible Staphylococcus aureus during Therapy Improves Antibacterial Activity. Antimicrob. Agents Chemother. 2012, 56, 5296–5302. [Google Scholar] [CrossRef]
  15. Lin, J.C.; Aung, G.; Thomas, A.; Jahng, M.; Johns, S.; Fierer, J. The use of ceftaroline fosamil in methicillin-resistant Staphy-lococcus aureus endocarditis and deep-seated MRSA infections: A retrospective case series of 10 patients. J. Infect. Chemother. 2013, 19, 42–49. [Google Scholar] [CrossRef] [PubMed]
  16. Rose, W.; Fantl, M.; Geriak, M.; Nizet, V.; Sakoulas, G. Current Paradigms of Combination Therapy in Methicillin-Resistant Staphylococcus aureus (MRSA) Bacteremia: Does it Work, which Combination, and for which Patients? Clin. Infect. Dis. 2021, 73, 2353–2360. [Google Scholar] [CrossRef]
  17. van Hal, S.J.; Jensen, S.O.; Vaska, V.L.; Espedido, B.A.; Paterson, D.L.; Gosbell, I.B. Predictors of Mortality in Staphylococcus aureus Bacteremia. Clin. Microbiol. Rev. 2012, 25, 362–386. [Google Scholar] [CrossRef] [PubMed]
  18. Fowler, V.G., Jr.; Boucher, H.W.; Corey, G.R.; Abrutyn, E.; Karchmer, A.W.; Rupp, M.E.; Levine, D.P.; Chambers, H.F.; Tally, F.P.; Vigliani, G.A.; et al. Daptomycin versus Standard Therapy for Bacteremia and Endocarditis Caused by Staphylococcus aureus. N. Engl. J. Med. 2006, 355, 653–665. [Google Scholar] [CrossRef] [PubMed]
  19. Pujol, M.; Miro, J.M.; Shaw, E.; Aguado, J.M.; San-Juan, R.; Puig-Asensio, M.; Pigrau, C.; Calbo, E.; Montejo, M.; Rodriguez-Alvarez, R.; et al. Daptomycin Plus Fosfomycin versus Daptomycin Alone for Methicillin-resistant Staphylococcus aureus Bacteremia and Endocarditis: A Randomized Clinical Trial. Clin. Infect. Dis. 2021, 72, 1517–1525. [Google Scholar] [CrossRef]
  20. Davis, J.S.; Sud, A.; O’Sullivan, M.V.; Robinson, J.O.; Ferguson, P.E.; Foo, H.; van Hal, S.J.; Ralph, A.P.; Howden, B.P.; Binks, P.M.; et al. Combination of Vancomycin and beta-Lactam Therapy for Methicillin-Resistant Staphylococcus aureus Bacteremia: A Pilot Multicenter Randomized Controlled Trial. Clin. Infect. Dis. 2016, 62, 173–180. [Google Scholar] [CrossRef]
  21. Tong, S.Y.C.; Lye, D.C.; Yahav, D.; Sud, A.; Robinson, J.O.; Nelson, J.; Archuleta, S.; Roberts, M.A.; Cass, A.; Paterson, D.L.; et al. Effect of Vancomycin or Daptomycin with vs without an Antistaphylococcal β-Lactam on Mortality, Bacteremia, Relapse, or Treatment Failure in Patients with MRSA Bacteremia: A Randomized Clinical Trial. JAMA 2020, 323, 527–537. [Google Scholar] [CrossRef] [PubMed]
  22. Fowler, V.G., Jr.; Miro, J.M.; Hoen, B.; Cabell, C.H.; Abrutyn, E.; Rubinstein, E.; Corey, G.R.; Spelman, D.; Bradley, S.F.; Barsic, B.; et al. Staphylococcus aureus endocarditis: A consequence of medical progress. JAMA 2005, 293, 3012–3021. [Google Scholar] [CrossRef] [PubMed]
  23. Miller, L.S.; Fowler, V.G.; Shukla, S.K.; Rose, W.E.; Proctor, R.A. Development of a vaccine against Staphylococcus aureus invasive infections: Evidence based on human immunity, genetics and bacterial evasion mechanisms. FEMS Microbiol. Rev. 2020, 44, 123–153. [Google Scholar] [CrossRef] [PubMed]
  24. Shukla, S.K.; Rose, W.; Schrodi, S.J. Complex host genetic susceptibility to Staphylococcus aureus infections. Trends Microbiol. 2015, 23, 529–536. [Google Scholar] [CrossRef] [PubMed]
  25. Horn, J.; Stelzner, K.; Rudel, T.; Fraunholz, M. Inside job: Staphylococcus aureus host-pathogen interactions. Int. J. Med. Microbiol. 2018, 308, 607–624. [Google Scholar] [CrossRef]
  26. Rooijakkers, S.H.; van Kessel, K.P.; van Strijp, J.A. Staphylococcal innate immune evasion. Trends Microbiol. 2005, 13, 596–601. [Google Scholar] [CrossRef] [PubMed]
  27. Minejima, E.; Bensman, J.; She, R.C.; Mack, W.J.; Tuan Tran, M.; Ny, P.; Lou, M.; Yamaki, J.; Nieberg, P.; Ho, J.; et al. A Dysregulated Balance of Proinflammatory and Anti-Inflammatory Host Cytokine Response Early during Therapy Predicts Persistence and Mortality in Staphylococcus aureus Bacteremia. Crit. Care Med. 2016, 44, 671–679. [Google Scholar] [CrossRef] [PubMed]
  28. Volk, C.F.; Burgdorf, S.; Edwardson, G.; Nizet, V.; Sakoulas, G.; Rose, W.E. Interleukin (IL)-1beta and IL-10 Host Responses in Patients with Staphylococcus aureus Bacteremia Determined by Antimicrobial Therapy. Clin. Infect. Dis. 2020, 70, 2634–2640. [Google Scholar] [CrossRef]
  29. Wozniak, J.M.; Mills, R.H.; Olson, J.; Caldera, J.; Sepich-Poore, G.D.; Carrillo-Terrazas, M.; Tsai, C.-M.; Vargas, F.; Knight, R.; Dorrestein, P.C.; et al. Mortality Risk Profiling of Staphylococcus aureus Bacteremia by Multiomic Serum Analysis Reveals Early Predictive and Pathogenic Signatures. Cell 2020, 182, 1311–1327. [Google Scholar] [CrossRef]
  30. Ferry, T.; Thomas, D.; Genestier, A.-L.; Bes, M.; Lina, G.; Vandenesch, F.; Etienne, J. Comparative Prevalence of Superantigen Genes in Staphylococcus aureus Isolates Causing Sepsis with and without Septic Shock. Clin. Infect. Dis. 2005, 41, 771–777. [Google Scholar] [CrossRef]
  31. Welsh, K.J.; Abbott, A.N.; Lewis, E.M.; Gardiner, J.M.; Kruzel, M.C.; Lewis, C.T.; Mohr, J.F.; Wanger, A.; Armitige, L.Y. Clinical characteristics, outcomes, and microbiologic features associated with methicillin-resistant Staphylococcus aureus bacteremia in pediatric patients treated with vancomycin. J. Clin. Microbiol. 2010, 48, 894–899. [Google Scholar] [CrossRef] [PubMed]
  32. Gillet, Y.; Issartel, B.; Vanhems, P.; Fournet, J.C.; Lina, G.; Bes, M.; Vandenesch, F.; Piemont, Y.; Brousse, N.; Floret, D.; et al. Association between Staphylococcus aureus strains carrying gene for Panton-Valentine leukocidin and highly lethal necrotising pneumonia in young immunocompetent patients. Lancet 2002, 359, 753–759. [Google Scholar] [CrossRef] [PubMed]
  33. Adhikari, R.P.; Ajao, A.O.; Aman, M.J.; Karauzum, H.; Sarwar, J.; Lydecker, A.D.; Johnson, J.K.; Nguyen, C.; Chen, W.H.; Roghmann, M.-C. Lower Antibody Levels to Staphylococcus aureus Exotoxins Are Associated with Sepsis in Hospitalized Adults with Invasive, S. aureus Infections. J. Infect. Dis. 2012, 206, 915–923. [Google Scholar] [CrossRef] [PubMed]
  34. Adhikari, R.P.; Kort, T.; Shulenin, S.; Kanipakala, T.; Ganjbaksh, N.; Roghmann, M.C.; Holtsberg, F.W.; Aman, M.J. Antibodies to S. aureus LukS-PV Attenuated Subunit Vaccine Neutralize a Broad Spectrum of Canonical and Non-Canonical Bi-component Leukotoxin Pairs. PLoS ONE 2015, 10, e0137874. [Google Scholar] [CrossRef]
  35. Fritz, S.A.; Tiemann, K.M.; Hogan, P.G.; Epplin, E.K.; Rodriguez, M.; Al-Zubeidi, D.N.; Wardenburg, J.B.; Hunstad, D.A. A Serologic Correlate of Protective Immunity Against Community-Onset Staphylococcus aureus Infection. Clin. Infect. Dis. 2013, 56, 1554–1561. [Google Scholar] [CrossRef] [PubMed]
  36. François, B.; Mercier, E.; Gonzalez, C.; Asehnoune, K.; Nseir, S.; Fiancette, M.; Desachy, A.; Plantefève, G.; Meziani, F.; de Lame, P.-A.; et al. Safety and tolerability of a single administration of AR-301, a human monoclonal antibody, in ICU patients with severe pneumonia caused by Staphylococcus aureus: First-in-human trial. Intensive Care Med. 2018, 44, 1787–1796. [Google Scholar] [CrossRef] [PubMed]
  37. Campbell, A.J.; Dotel, R.; Braddick, M.; Britton, P.N.; Eisen, D.P.; Francis, J.R.; Lynar, S.; McMullan, B.; Meagher, N.; Nelson, J.; et al. Clindamycin adjunctive therapy for severe Staphylococcus aureus treatment evaluation (CASSETTE)-an open-labelled pilot randomized controlled trial. JAC Antimicrob. Resist. 2022, 4, dlac014. [Google Scholar] [CrossRef] [PubMed]
  38. Anpalagan, K.; Dotel, R.; MacFadden, D.R.; Smith, S.; Voss, L.; Petersiel, N.; Marks, M.; Marsh, J.; Mahar, R.K.; McGlothlin, A.; et al. Does adjunctive clindamycin have a role in Staphylococcus aureus bacteremia? A protocol for the adjunctive treatment domain of the S. aureus Network Adaptive Platform (SNAP) randomized controlled trial. Clin. Infect. Dis. 2024, ciae28. [Google Scholar] [CrossRef]
  39. Proctor, R.A.; Balwit, J.M.; Vesga, O. Variant subpopulations of Staphylococcus aureus as cause of persistent and recurrent infections. Infect. Agents Dis. 1994, 3, 302–312. [Google Scholar]
  40. von Eiff, C. Staphylococcus aureus small colony variants: A challenge to microbiologists and clinicians. Int. J. Antimicrob. Agents 2008, 31, 507–510. [Google Scholar] [CrossRef]
  41. Kahl, B.C.; Belling, G.; Becker, P.; Chatterjee, I.; Wardecki, K.; Hilgert, K.; Cheung, A.L.; Peters, G.; Herrmann, M. Thymidine-dependent Staphylococcus aureus small-colony variants are associated with extensive alterations in regulator and virulence gene expression profiles. Infect. Immun. 2005, 73, 4119–4126. [Google Scholar] [CrossRef] [PubMed]
  42. Kolle, W.; Hetsch, H. Die Experimentelle Bakteriologie und die Infektionskrankheiten mit besonderer Berücksichtigung der Immunitätslehre; Urban und Schwarzenberg: Berlin, Germany, 1911; Volume 1. [Google Scholar]
  43. Bigger, J.W.; Boland, C.R.; O’meara, R.A.Q. Variant colonies of Staphylococcus aureus. J. Pathol. Bacteriol. 1927, 30, 261–269. [Google Scholar] [CrossRef]
  44. Youmans, G.P. Production of Small-Colony Variants of Staphylococcus aureus. Proc. Soc. Exp. Biol. Med. 1937, 36, 94–96. [Google Scholar] [CrossRef]
  45. Proctor, R.A.; von Eiff, C.; Kahl, B.C.; Becker, K.; McNamara, P.; Herrmann, M.; Peters, G. Small colony variants: A pathogenic form of bacteria that facilitates persistent and recurrent infections. Nat. Rev. Microbiol. 2006, 4, 295–305. [Google Scholar] [CrossRef] [PubMed]
  46. Kahl, B.C.; Becker, K.; Löffler, B. Clinical Significance and Pathogenesis of Staphylococcal Small Colony Variants in Persistent Infections. Clin. Microbiol. Rev. 2016, 29, 401–427. [Google Scholar] [CrossRef]
  47. Maduka-Ezeh, A.N.; Greenwood-Quaintance, K.E.; Karau, M.J.; Berbari, E.F.; Osmon, D.R.; Hanssen, A.D.; Steckelberg, J.M.; Patel, R. Antimicrobial susceptibility and biofilm formation of Staphylococcus epidermidis small colony variants associated with prosthetic joint infection. Diagn. Microbiol. Infect. Dis. 2012, 74, 224–229. [Google Scholar] [CrossRef] [PubMed]
  48. Garcia, L.G.; Lemaire, S.; Kahl, B.C.; Becker, K.; Proctor, R.A.; Denis, O.; Tulkens, P.M.; van Bambeke, F. Antibiotic activity against small-colony variants of Staphylococcus aureus: Review of in vitro, animal and clinical data. J. Antimicrob. Chemother. 2013, 68, 1455–1464. [Google Scholar] [CrossRef]
  49. Baltch, A.L.; Ritz, W.J.; Bopp, L.H.; Michelsen, P.; Smith, R.P. Activities of daptomycin and comparative antimicrobials, singly and in combination, against extracellular and intracellular Staphylococcus aureus and its stable small-colony variant in human monocyte-derived macrophages and in broth. Antimicrob. Agents Chemother. 2008, 52, 1829–1833. [Google Scholar] [CrossRef]
  50. Kriegeskorte, A.; Grubmuller, S.; Huber, C.; Kahl, B.C.; von Eiff, C.; Proctor, R.A.; Peters, G.; Eisenreich, W.; Becker, K. Staphylococcus aureus small colony variants show common metabolic features in central metabolism irrespective of the un-derlying auxotrophism. Front. Cell. Infect. Microbiol. 2014, 4, 141. [Google Scholar] [CrossRef]
  51. Mishra, N.N.; Bayer, A.S.; Weidenmaier, C.; Grau, T.; Wanner, S.; Stefani, S.; Cafiso, V.; Bertuccio, T.; Yeaman, M.R.; Nast, C.C.; et al. Phenotypic and Genotypic Characterization of Daptomycin-Resistant Methicillin-Resistant Staphylococcus aureus Strains: Relative Roles of mprF and dlt Operons. PLoS ONE 2014, 9, e107426. [Google Scholar] [CrossRef]
  52. Zhanel, G.G.; Schweizer, F.; Karlowsky, J.A. Oritavancin: Mechanism of Action. Clin. Infect. Dis. 2012, 54, S214–S219. [Google Scholar] [CrossRef] [PubMed]
  53. Bouza, E.; Burillo, A. Oritavancin: A novel lipoglycopeptide active against Gram-positive pathogens including multiresistant strains. Int. J. Antimicrob. Agents 2010, 36, 401–407. [Google Scholar] [CrossRef] [PubMed]
  54. Boulet, M.L.; Isabelle, C.; Guay, I.; Brouillette, E.; Langlois, J.-P.; Jacques, P.; Rodrigue, S.; Brzezinski, R.; Beauregard, P.B.; Bouarab, K.; et al. Tomatidine Is a Lead Antibiotic Molecule That Targets Staphylococcus aureus ATP Synthase Subunit C. Antimicrob. Agents Chemother. 2018, 62, e02197-17. [Google Scholar] [CrossRef] [PubMed]
  55. Langlois, J.-P.; Millette, G.; Guay, I.; Dubé-Duquette, A.; Chamberland, S.; Jacques, P.; Rodrigue, S.; Bouarab, K.; Marsault, É.; Malouin, F. Bactericidal Activity of the Bacterial ATP Synthase Inhibitor Tomatidine and the Combination of Tomatidine and Aminoglycoside against Persistent and Virulent Forms of Staphylococcus aureus. Front. Microbiol. 2020, 11, 805. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, J.; Gefen, O.; Ronin, I.; Bar-Meir, M.; Balaban, N.Q. Effect of tolerance on the evolution of antibiotic resistance under drug combinations. Science 2020, 367, 200–204. [Google Scholar] [CrossRef] [PubMed]
  57. Levin-Reisman, I.; Ronin, I.; Gefen, O.; Braniss, I.; Shoresh, N.; Balaban, N.Q. Antibiotic tolerance facilitates the evolution of resistance. Science 2017, 355, 826–830. [Google Scholar] [CrossRef] [PubMed]
  58. Miller, C.R.; Monk, J.M.; Szubin, R.; Berti, A.D. Rapid resistance development to three antistaphylococcal therapies in anti-biotic-tolerant Staphylococcus aureus bacteremia. PLoS ONE 2021, 16, e0258592. [Google Scholar] [CrossRef] [PubMed]
  59. Berti, A.D.; Shukla, N.; Rottier, A.D.; McCrone, J.S.; Turner, H.M.; Monk, I.R.; Baines, S.L.; Howden, B.P.; Proctor, R.A.; Rose, W.E. Daptomycin selects for genetic and phenotypic adaptations leading to antibiotic tolerance in MRSA. J. Antimicrob. Chemother. 2018, 73, 2030–2033. [Google Scholar] [CrossRef] [PubMed]
  60. Brauner, A.; Fridman, O.; Gefen, O.; Balaban, N.Q. Distinguishing between resistance, tolerance and persistence to antibiotic treatment. Nat. Rev. Microbiol. 2016, 14, 320–330. [Google Scholar] [CrossRef]
  61. Berti, A.D.; Hirsch, E.B. Tolerance to antibiotics affects response. Science 2020, 367, 141–142. [Google Scholar] [CrossRef]
  62. Windels, E.M.; Michiels, J.E.; van den Bergh, B.; Fauvart, M.; Michiels, J. Antibiotics: Combatting Tolerance to Stop Resistance. mBio 2019, 10, e02095-19. [Google Scholar] [CrossRef] [PubMed]
  63. Meredith, E.M.; Harven, L.T.; Berti, A.D. Antimicrobial Efficacy against Antibiotic-Tolerant Staphylococcus aureus Depends on the Mechanism of Antibiotic Tolerance. Antibiotics 2022, 11, 1810. [Google Scholar] [CrossRef] [PubMed]
  64. Peyrusson, F.; Varet, H.; Nguyen, T.K.; Legendre, R.; Sismeiro, O.; Coppee, J.Y.; Wolz, C.; Tenson, T.; van Bambeke, F. In-tracellular Staphylococcus aureus persisters upon antibiotic exposure. Nat. Commun. 2020, 11, 2200. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, Y.; Bojer, M.S.; George, S.E.; Wang, Z.; Jensen, P.R.; Wolz, C.; Ingmer, H. Inactivation of TCA cycle enhances Staph-ylococcus aureus persister cell formation in stationary phase. Sci. Rep. 2018, 8, 10849. [Google Scholar] [CrossRef]
  66. Zalis, E.A.; Nuxoll, A.S.; Manuse, S.; Clair, G.; Radlinski, L.C.; Conlon, B.P.; Adkins, J.; Lewis, K. Stochastic Variation in Expression of the Tricarboxylic Acid Cycle Produces Persister Cells. mBio 2019, 10. [Google Scholar] [CrossRef]
  67. Tuchscherr, L.; Loffler, B.; Proctor, R.A. Persistence of Staphylococcus aureus: Multiple Metabolic Pathways Impact the Ex-pression of Virulence Factors in Small-Colony Variants (SCVs). Front. Microbiol. 2020, 11, 1028. [Google Scholar] [CrossRef]
  68. Costerton, J.W.; Stewart, P.S.; Greenberg, E.P. Bacterial Biofilms: A Common Cause of Persistent Infections. Science 1999, 284, 1318–1322. [Google Scholar] [CrossRef]
  69. Donlan, R.M.; Costerton, J.W. Biofilms: Survival Mechanisms of Clinically Relevant Microorganisms. Clin. Microbiol. Rev. 2002, 15, 167–193. [Google Scholar] [CrossRef] [PubMed]
  70. Flemming, H.-C.; Wingender, J. The biofilm matrix. Nat. Rev. Microbiol. 2010, 8, 623–633. [Google Scholar] [CrossRef]
  71. Raad, I.; Hanna, H.; Jiang, Y.; Dvorak, T.; Reitzel, R.; Chaiban, G.; Sherertz, R.; Hachem, R. Comparative activities of dap-tomycin, linezolid, and tigecycline against catheter-related methicillin-resistant Staphylococcus bacteremic isolates embedded in biofilm. Antimicrob. Agents Chemother. 2007, 51, 1656–1660. [Google Scholar] [CrossRef]
  72. Foster, T.J.; Geoghegan, J.A.; Ganesh, V.K.; Höök, M. Adhesion, invasion and evasion: The many functions of the surface proteins of Staphylococcus aureus. Nat. Rev. Microbiol. 2014, 12, 49–62. [Google Scholar] [CrossRef] [PubMed]
  73. Aslam, S. Effect of antibacterials on biofilms. Am. J. Infect. Control 2008, 36, S175.e9–S175.e11. [Google Scholar] [CrossRef] [PubMed]
  74. Mah, T.-F.C.; O’Toole, G.A. Mechanisms of biofilm resistance to antimicrobial agents. Trends Microbiol. 2001, 9, 34–39. [Google Scholar] [CrossRef] [PubMed]
  75. Theis, T.J.; Daubert, T.A.; Kluthe, K.E.; Brodd, K.L.; Nuxoll, A.S. Staphylococcus aureus persisters are associated with reduced clearance in a catheter-associated biofilm infection. Front. Cell. Infect. Microbiol. 2023, 13, 1178526. [Google Scholar] [CrossRef] [PubMed]
  76. Safdar, A.; Rolston, K.V. Vancomycin tolerance, a potential mechanism for refractory gram-positive bacteremia observational study in patients with cancer. Cancer 2006, 106, 1815–1820. [Google Scholar] [CrossRef] [PubMed]
  77. Garzoni, C.; Kelley, W.L. Staphylococcus aureus: New evidence for intracellular persistence. Trends Microbiol. 2009, 17, 59–65. [Google Scholar] [CrossRef] [PubMed]
  78. Xu, D.; Hu, G.; Luo, J.; Cheng, J.; Wu, D.; Cheng, L.; Huang, X.; Fu, S.; Liu, J. Staphylococcus aureus induces mitophagy to promote its survival within bovine mammary epithelial cells. Vet. Microbiol. 2023, 280, 109697. [Google Scholar] [CrossRef] [PubMed]
  79. Rollin, G.; Tan, X.; Tros, F.; Dupuis, M.; Nassif, X.; Charbit, A.; Coureuil, M. Intracellular Survival of Staphylococcus aureus in Endothelial Cells: A Matter of Growth or Persistence. Front. Microbiol. 2017, 8, 1354. [Google Scholar] [CrossRef]
  80. Perez, K.; Patel, R. Survival of Staphylococcus epidermidis in Fibroblasts and Osteoblasts. Infect. Immun. 2018, 86, 10. [Google Scholar] [CrossRef]
  81. Abu-Humaidan, A.H.; Elvén, M.; Sonesson, A.; Garred, P.; Sørensen, O.E. Persistent Intracellular Staphylococcus aureus in Keratinocytes Lead to Activation of the Complement System with Subsequent Reduction in the Intracellular Bacterial Load. Front. Immunol. 2018, 9, 396. [Google Scholar] [CrossRef]
  82. Prajsnar, T.K.; Serba, J.J.; Dekker, B.M.; Gibson, J.F.; Masud, S.; Fleming, A.; Johnston, S.A.; Renshaw, S.A.; Meijer, A.H. The autophagic response to Staphylococcus aureus provides an intracellular niche in neutrophils. Autophagy 2020, 17, 888–902. [Google Scholar] [CrossRef]
  83. Yang, D.; Wijenayaka, A.R.; Solomon, L.B.; Pederson, S.M.; Findlay, D.M.; Kidd, S.P.; Atkins, G.J. Novel Insights into Staphylococcus aureus Deep Bone Infections: The Involvement of Osteocytes. mBio 2018, 9, e00415-18. [Google Scholar] [CrossRef]
  84. Zelmer, A.R.; Nelson, R.; Richter, K.; Atkins, G.J. Can intracellular Staphylococcus aureus in osteomyelitis be treated using current antibiotics? A systematic review and narrative synthesis. Bone Res. 2022, 10, 1–18. [Google Scholar] [CrossRef]
  85. Clement, S.; Vaudaux, P.; Francois, P.; Schrenzel, J.; Huggler, E.; Kampf, S.; Chaponnier, C.; Lew, D.; Lacroix, J. Evidence of an Intracellular Reservoir in the Nasal Mucosa of Patients with Recurrent Staphylococcus aureus Rhinosinusitis. J. Infect. Dis. 2005, 192, 1023–1028. [Google Scholar] [CrossRef]
  86. Ou, J.; Bassiouni, A.; Drilling, A.; Psaltis, A.; Vreugde, S.; Wormald, P. The persistence of intracellular Staphylococcus aureus in the sinuses: A longitudinal study. Rhinol. J. 2017, 55, 305–311. [Google Scholar] [CrossRef]
  87. Zautner, A.E.; Krause, M.; Stropahl, G.; Holtfreter, S.; Frickmann, H.; Maletzki, C.; Kreikemeyer, B.; Pau, H.W.; Podbielski, A. Intracellular Persisting Staphylococcus aureus Is the Major Pathogen in Recurrent Tonsillitis. PLoS ONE 2010, 5, e9452. [Google Scholar] [CrossRef]
  88. Ellington, J.K.; Harris, M.; Webb, L.; Smith, B.; Smith, T.; Tan, K.; Hudson, M. Intracellular Staphylococcus aureus. A mech-anism for the indolence of osteomyelitis. J. Bone Jt. Surg. Br. Vol. 2003, 85, 918–921. [Google Scholar] [CrossRef]
  89. Torlakovic, E.; Hibbs, J.R.; Miller, J.S.; Litz, C.E. Intracellular Bacteria in Blood Smears in Patients with Central Venous Catheters. Arch. Intern. Med. 1995, 155, 1547–1550. [Google Scholar] [CrossRef]
  90. Grundmeier, M.; Tuchscherr, L.; Brück, M.; Viemann, D.; Roth, J.; Willscher, E.; Becker, K.; Peters, G.; Löffler, B. Staphylococcal Strains Vary Greatly in Their Ability to Induce an Inflammatory Response in Endothelial Cells. J. Infect. Dis. 2010, 201, 871–880. [Google Scholar] [CrossRef]
  91. Menzies, B.E.; Kourteva, I. Staphylococcus aureus alpha-toxin induces apoptosis in endothelial cells. FEMS Immunol. Med. Microbiol. 2000, 29, 39–45. [Google Scholar] [CrossRef]
  92. Schnaith, A.; Kashkar, H.; Leggio, S.A.; Addicks, K.; Krönke, M.; Krut, O. Staphylococcus aureus Subvert Autophagy for Induction of Caspase-independent Host Cell Death. J. Biol. Chem. 2007, 282, 2695–2706. [Google Scholar] [CrossRef]
  93. Soong, G.; Chun, J.; Parker, D.; Prince, A. Staphylococcus aureus activation of caspase 1/calpain signaling mediates invasion through human keratinocytes. J. Infect. Dis. 2012, 205, 1571–1579. [Google Scholar] [CrossRef]
  94. Tuchscherr, L.; Medina, E.; Hussain, M.; Völker, W.; Heitmann, V.; Niemann, S.; Holzinger, D.; Roth, J.; Proctor, R.A.; Becker, K.; et al. Staphylococcus aureus phenotype switching: An effective bacterial strategy to escape host immune response and establish a chronic infection. EMBO Mol. Med. 2011, 3, 129–141. [Google Scholar] [CrossRef]
  95. Löffler, B.; Tuchscherr, L.; Niemann, S.; Peters, G. Staphylococcus aureus persistence in non-professional phagocytes. Int. J. Med. Microbiol. 2014, 304, 170–176. [Google Scholar] [CrossRef]
  96. Tuchscherr, L.; Löffler, B. Staphylococcus aureus dynamically adapts global regulators and virulence factor expression in the course from acute to chronic infection. Curr. Genet. 2015, 62, 15–17. [Google Scholar] [CrossRef]
  97. Haslinger-Loffler, B.; Kahl, B.C.; Grundmeier, M.; Strangfeld, K.; Wagner, B.; Fischer, U.; Cheung, A.L.; Peters, G.; Schulze-Osthoff, K.; Sinha, B. Multiple virulence factors are required for Staphylococcus aureus-induced apoptosis in endothelial cells. Cell. Microbiol. 2005, 7, 1087–1097. [Google Scholar] [CrossRef]
  98. Ou, J.J.; Drilling, A.J.; Cooksley, C.; Bassiouni, A.; Kidd, S.P.; Psaltis, A.J.; Wormald, P.J.; Vreugde, S. Reduced Innate Immune Response to a Staphylococcus aureus Small Colony Variant Compared to Its Wild-Type Parent Strain. Front. Cell. Infect. Microbiol. 2016, 6, 187. [Google Scholar] [CrossRef]
  99. Horst, S.A.; Hoerr, V.; Beineke, A.; Kreis, C.; Tuchscherr, L.; Kalinka, J.; Lehne, S.; Schleicher, I.; Kohler, G.; Fuchs, T.; et al. A novel mouse model of Staphylococcus aureus chronic osteo-myelitis that closely mimics the human infection: An integrated view of disease pathogenesis. Am. J. Pathol. 2012, 181, 1206–1214. [Google Scholar] [CrossRef]
  100. Seidl, K.; Solis, N.V.; Bayer, A.S.; Hady, W.A.; Ellison, S.; Klashman, M.C.; Xiong, Y.Q.; Filler, S.G. Divergent Responses of Different Endothelial Cell Types to Infection with Candida albicans and Staphylococcus aureus. PLoS ONE 2012, 7, e39633. [Google Scholar] [CrossRef]
  101. Jarry, T.M.; Cheung, A.L. Staphylococcus aureus Escapes More Efficiently from the Phagosome of a Cystic Fibrosis Bronchial Epithelial Cell Line than from Its Normal Counterpart. Infect. Immun. 2006, 74, 2568–2577. [Google Scholar] [CrossRef]
  102. Hudson, M.C.; Ramp, W.K.; Nicholson, N.C.; Williams, A.S.; Nousiainen, M.T. Internalization of Staphylococcus aureus by cultured osteoblasts. Microb. Pathog. 1995, 19, 409–419. [Google Scholar] [CrossRef]
  103. Weinrick, B.; Dunman, P.M.; McAleese, F.; Murphy, E.; Projan, S.J.; Fang, Y.; Novick, R.P. Effect of mild acid on gene expression in Staphylococcus aureus. J. Bacteriol. 2004, 186, 8407–8423. [Google Scholar] [CrossRef]
  104. Kubica, M.; Guzik, K.; Koziel, J.; Zarebski, M.; Richter, W.; Gajkowska, B.; Golda, A.; Maciag-Gudowska, A.; Brix, K.; Shaw, L.; et al. A Potential New Pathway for Staphylococcus aureus Dissemination: The Silent Survival of S. aureus Phagocytosed by Human Monocyte-Derived Macrophages. PLoS ONE 2008, 3, e1409. [Google Scholar] [CrossRef]
  105. Pang, Y.Y.; Schwartz, J.; Thoendel, M.; Ackermann, L.W.; Horswill, A.R.; Nauseef, W.M. agr-Dependent interactions of Staphylococcus aureus USA300 with human polymorphonuclear neutrophils. J. Innate Immun. 2010, 2, 546–559. [Google Scholar] [CrossRef]
  106. Tranchemontagne, Z.R.; Camire, R.B.; O’Donnell, V.J.; Baugh, J.; Burkholder, K.M. Staphylococcus aureus Strain USA300 Perturbs Acquisition of Lysosomal Enzymes and Requires Phagosomal Acidification for Survival inside Macrophages. Infect. Immun. 2016, 84, 241–253. [Google Scholar] [CrossRef]
  107. Watkins, K.E.; Unnikrishnan, M. Evasion of host defenses by intracellular Staphylococcus aureus. Adv. Appl. Microbiol. 2020, 112, 105–141. [Google Scholar] [CrossRef]
  108. Lacoma, A.; Cano, V.; Moranta, D.; Regueiro, V.; Dominguez-Villanueva, D.; Laabei, M.; Gonzalez-Nicolau, M.; Ausina, V.; Prat, C.; Bengoechea, J.A. Investigating intracellular persistence of Staphylococcus aureus within a murine alveolar macro-phage cell line. Virulence 2017, 8, 1761–1775. [Google Scholar] [CrossRef]
  109. Peschel, A.; Jack, R.W.; Otto, M.; Collins, L.V.; Staubitz, P.; Nicholson, G.; Kalbacher, H.; Nieuwenhuizen, W.F.; Jung, G.; Tarkowski, A.; et al. Staphylococcus aureus resistance to human defensins and evasion of neutrophil killing via the novel virulence factor MprF is based on modification of membrane lipids with l-lysine. J. Exp. Med. 2001, 193, 1067–1076. [Google Scholar] [CrossRef]
  110. Flannagan, R.S.; Kuiack, R.C.; McGavin, M.J.; Heinrichs, D.E. Staphylococcus aureus Uses the GraXRS Regulatory System to Sense and Adapt to the Acidified Phagolysosome in Macrophages. mBio 2018, 9, e01143-18. [Google Scholar] [CrossRef]
  111. Karavolos, M.H.; Horsburgh, M.J.; Ingham, E.; Foster, S.J. Role and regulation of the superoxide dismutases of Staphylococcus aureus. Microbiology 2003, 149, 2749–2758. [Google Scholar] [CrossRef]
  112. Cosgrove, K.; Coutts, G.; Jonsson, I.-M.; Tarkowski, A.; Kokai-Kun, J.F.; Mond, J.J.; Foster, S.J. Catalase (KatA) and Alkyl Hydroperoxide Reductase (AhpC) Have Compensatory Roles in Peroxide Stress Resistance and Are Required for Survival, Persistence, and Nasal Colonization in Staphylococcus aureus. J. Bacteriol. 2007, 189, 1025–1035. [Google Scholar] [CrossRef]
  113. Treffon, J.; Block, D.; Moche, M.; Reiss, S.; Fuchs, S.; Engelmann, S.; Becher, D.; Langhanki, L.; Mellmann, A.; Peters, G.; et al. Adaptation of Staphylococcus aureus to Airway Environments in Patients with Cystic Fibrosis by Upregulation of Super-oxide Dismutase M and Iron-Scavenging Proteins. J. Infect. Dis. 2018, 217, 1453–1461. [Google Scholar] [CrossRef]
  114. Treffon, J.; Chaves-Moreno, D.; Niemann, S.; Pieper, D.H.; Vogl, T.; Roth, J.; Kahl, B.C. Importance of superoxide dismutases A and M for protection of Staphylococcus aureus in the oxidative stressful environment of cystic fibrosis airways. Cell. Microbiol. 2020, 22, e13158. [Google Scholar] [CrossRef]
  115. Munzenmayer, L.; Geiger, T.; Daiber, E.; Schulte, B.; Autenrieth, S.E.; Fraunholz, M.; Wolz, C. Influence of Sae-regulated and Agr-regulated factors on the escape of Staphylococcus aureus from human macrophages. Cell. Microbiol. 2016, 18, 1172–1183. [Google Scholar] [CrossRef]
  116. Jarry, T.M.; Memmi, G.; Cheung, A.L. The expression of alpha-haemolysin is required for Staphylococcus aureus phagosomal escape after internalization in CFT-1 cells. Cell. Microbiol. 2008, 10, 1801–1814. [Google Scholar] [CrossRef]
  117. Grosz, M.; Kolter, J.; Paprotka, K.; Winkler, A.C.; Schafer, D.; Chatterjee, S.S.; Geiger, T.; Wolz, C.; Ohlsen, K.; Otto, M.; et al. Cytoplasmic replication of Staphylococcus aureus upon phagosomal escape triggered by phenolsoluble modulin alpha. Cell. Microbiol. 2014, 16, 451–465. [Google Scholar] [CrossRef]
  118. Wang, R.; Braughton, K.R.; Kretschmer, D.; Bach, T.H.; Queck, S.Y.; Li, M.; Kennedy, A.D.; Dorward, D.W.; Klebanoff, S.J.; Peschel, A.; et al. Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat. Med. 2007, 13, 1510–1514. [Google Scholar] [CrossRef]
  119. Queck, S.Y.; Jameson-Lee, M.; Villaruz, A.E.; Bach, T.-H.L.; Khan, B.A.; Sturdevant, D.E.; Ricklefs, S.M.; Li, M.; Otto, M. RNAIII-Independent Target Gene Control by the agr Quorum-Sensing System: Insight into the Evolution of Virulence Regulation in Staphylococcus aureus. Mol. Cell 2008, 32, 150–158. [Google Scholar] [CrossRef]
  120. Geiger, T.; Francois, P.; Liebeke, M.; Fraunholz, M.; Goerke, C.; Krismer, B.; Schrenzel, J.; Lalk, M.; Wolz, C. The Stringent Response of Staphylococcus aureus and Its Impact on Survival after Phagocytosis through the Induction of Intracellular PSMs Expression. PLoS Pathog. 2012, 8, e1003016. [Google Scholar] [CrossRef]
  121. Chatterjee, S.S.; Joo, H.-S.; Duong, A.C.; Dieringer, T.D.; Tan, V.Y.; Song, Y.; Fischer, E.R.; Cheung, G.Y.C.; Li, M.; Otto, M. Essential Staphylococcus aureus toxin export system. Nat. Med. 2013, 19, 364–367. [Google Scholar] [CrossRef]
  122. Blattner, S.; Das, S.; Paprotka, K.; Eilers, U.; Krischke, M.; Kretschmer, D.; Remmele, C.W.; Dittrich, M.; Muller, T.; Schuelein-Voelk, C.; et al. Staphylococcus aureus Exploits a Non-ribosomal Cyclic Dipeptide to Modulate Survival within Epithelial Cells and Phagocytes. PLoS Pathog. 2016, 12, e1005857. [Google Scholar] [CrossRef]
  123. Giese, B.; Glowinski, F.; Paprotka, K.; Dittmann, S.; Steiner, T.; Sinha, B.; Fraunholz, M.J. Expression of delta-toxin by Staph-ylococcus aureus mediates escape from phago-endosomes of human epithelial and endothelial cells in the presence of beta-toxin. Cell. Microbiol. 2011, 13, 316–329. [Google Scholar] [CrossRef]
  124. Clauditz, A.; Resch, A.; Wieland, K.-P.; Peschel, A.; GÖtz, F. Staphyloxanthin Plays a Role in the Fitness of Staphylococcus aureus and Its Ability to Cope with Oxidative Stress. Infect. Immun. 2006, 74, 4950–4953. [Google Scholar] [CrossRef]
  125. Flannagan, R.S.; Heit, B.; Heinrichs, D.E. Antimicrobial Mechanisms of Macrophages and the Immune Evasion Strategies of Staphylococcus aureus. Pathogens 2015, 4, 826–868. [Google Scholar] [CrossRef]
  126. Jubrail, J.; Morris, P.; Bewley, M.A.; Stoneham, S.; Johnston, S.A.; Foster, S.J.; Peden, A.A.; Read, R.C.; Marriott, H.M.; Dockrell, D.H. Inability to sustain intraphagolysosomal killing of Staphylococcus aureus predisposes to bacterial persistence in macrophages. Cell. Microbiol. 2015, 18, 80–96. [Google Scholar] [CrossRef]
  127. Flannagan, R.S.; Heit, B.; Heinrichs, D.E. Intracellular replication of Staphylococcus aureus in mature phagolysosomes in macrophages precedes host cell death, and bacterial escape and dissemination. Cell. Microbiol. 2016, 18, 514–535. [Google Scholar] [CrossRef]
  128. Lehar, S.M.; Pillow, T.; Xu, M.; Staben, L.; Kajihara, K.K.; Vandlen, R.; DePalatis, L.; Raab, H.; Hazenbos, W.L.; Morisaki, J.H.; et al. Novel antibody-antibiotic conjugate eliminates intracellular S. aureus. Nature 2015, 527, 323–328. [Google Scholar] [CrossRef]
  129. Siegmund, A.; Afzal, M.A.; Tetzlaff, F.; Keinhörster, D.; Gratani, F.; Paprotka, K.; Westermann, M.; Nietzsche, S.; Wolz, C.; Fraunholz, M.; et al. Intracellular persistence of Staphylococcus aureus in endothelial cells is promoted by the absence of phenol-soluble modulins. Virulence 2021, 12, 1186–1198. [Google Scholar] [CrossRef]
  130. Elgrail, M.M.; Chen, E.; Shaffer, M.G.; Srinivasa, V.; Griffith, M.P.; Mustapha, M.M.; Shields, R.K.; van Tyne, D.; Culyba, M.J. Convergent Evolution of Antibiotic Tolerance in Patients with Persistent Methicillin-Resistant Staphylococcus aureus Bacteremia. Infect. Immun. 2022, 90, e0000122. [Google Scholar] [CrossRef]
  131. Sriramulu, D.D.; Nimtz, M.; Romling, U. Proteome analysis reveals adaptation of Pseudomonas aeruginosa to the cystic fibrosis lung environment. Proteomics 2005, 5, 3712–3721. [Google Scholar] [CrossRef]
  132. Pagels, M.; Fuchs, S.; Pané-Farré, J.; Kohler, C.; Menschner, L.; Hecker, M.; McNamarra, P.J.; Bauer, M.C.; von Wachenfeldt, C.; Liebeke, M.; et al. Redox sensing by a Rex-family repressor is involved in the regulation of anaerobic gene expression in Staphylococcus aureus. Mol. Microbiol. 2010, 76, 1142–1161. [Google Scholar] [CrossRef] [PubMed]
  133. Desgranges, E.; Barrientos, L.; Herrgott, L.; Marzi, S.; Toledo-Arana, A.; Moreau, K.; Vandenesch, F.; Romby, P.; Caldelari, I. The 3’UTR-derived sRNA RsaG coordinates redox homeostasis and metabolism adaptation in response to glucose-6-phosphate uptake in Staphylococcus aureus. Mol. Microbiol. 2022, 117, 193–214. [Google Scholar] [CrossRef] [PubMed]
  134. Luo, J.-D.; Chen, A.F. Nitric oxide: A newly discovered function on wound healing. Acta Pharmacol. Sin. 2005, 26, 259–264. [Google Scholar] [CrossRef] [PubMed]
  135. Lane, C.; Knight, D.; Burgess, S.; Franklin, P.; Horak, F.; Legg, J.; Moeller, A.; Stick, S. Epithelial inducible nitric oxide synthase activity is the major determinant of nitric oxide concentration in exhaled breath. Thorax 2004, 59, 757–760. [Google Scholar] [CrossRef] [PubMed]
  136. Medina, L.M.P.; Becker, A.-K.; Michalik, S.; Surmann, K.; Hildebrandt, P.; Salazar, M.G.; Mekonnen, S.A.; Kaderali, L.; Völker, U.; van Dijl, J.M. Interaction of Staphylococcus aureus and Host Cells upon Infection of Bronchial Epithelium during Different Stages of Regeneration. ACS Infect. Dis. 2020, 6, 2279–2290. [Google Scholar] [CrossRef] [PubMed]
  137. Wilde, A.D.; Snyder, D.J.; Putnam, N.E.; Valentino, M.D.; Hammer, N.D.; Lonergan, Z.R.; Hinger, S.A.; Aysanoa, E.E.; Blanchard, C.; Dunman, P.M.; et al. Bacterial Hypoxic Responses Revealed as Critical Determinants of the Host-Pathogen Outcome by TnSeq Analysis of Staphylococcus aureus Invasive Infection. PLoS Pathog. 2015, 11, e1005341. [Google Scholar] [CrossRef] [PubMed]
  138. Throup, J.P.; Zappacosta, F.; Lunsford, R.D.; Annan, R.S.; Carr, S.A.; Lonsdale, J.T.; Bryant, A.P.; McDevitt, D.; Rosenberg, M.; Burnham, M.K.R. The srhSR Gene Pair from Staphylococcus aureus: Genomic and Proteomic Approaches to the Identification and Characterization of Gene Function. Biochemistry 2001, 40, 10392–10401. [Google Scholar] [CrossRef] [PubMed]
  139. Wu, Y.; Wu, Y.; Zhu, T.; Han, H.; Liu, H.; Xu, T.; Francois, P.; Fischer, A.; Bai, L.; Götz, F.; et al. Staphylococcus epidermidis SrrAB Regulates Bacterial Growth and Biofilm Formation Differently under Oxic and Microaerobic Conditions. J. Bacteriol. 2015, 197, 459–476. [Google Scholar] [CrossRef] [PubMed]
  140. Cao, S.; Huseby, D.L.; Brandis, G.; Hughes, D. Alternative Evolutionary Pathways for Drug-Resistant Small Colony Variant Mutants in Staphylococcus aureus. mBio 2017, 8, e00358-17. [Google Scholar] [CrossRef]
  141. Wickersham, M.; Wachtel, S.; Wong Fok Lung, T.; Soong, G.; Jacquet, R.; Richardson, A.; Parker, D.; Prince, A. Metabolic Stress Drives Keratinocyte Defenses against Staphylococcus aureus Infection. Cell Rep. 2017, 18, 2742–2751. [Google Scholar] [CrossRef]
  142. Lung, T.W.F.; Monk, I.R.; Acker, K.P.; Mu, A.; Wang, N.; Riquelme, S.A.; Pires, S.; Noguera, L.P.; Dach, F.; Gabryszewski, S.J.; et al. Staphylococcus aureus small colony variants impair host immunity by activating host cell glycolysis and inducing necroptosis. Nat. Microbiol. 2019, 5, 141–153. [Google Scholar] [CrossRef] [PubMed]
  143. Kitur, K.; Wachtel, S.; Brown, A.; Wickersham, M.; Paulino, F.; Peñaloza, H.F.; Soong, G.; Bueno, S.; Parker, D.; Prince, A. Necroptosis Promotes Staphylococcus aureus Clearance by Inhibiting Excessive Inflammatory Signaling. Cell Rep. 2016, 16, 2219–2230. [Google Scholar] [CrossRef] [PubMed]
  144. Kumar, R.; Kanev, L.; Woods, S.D.; Brenner, M.; Smith, B. Managing hyperkalemia in high-risk patients in long-term care. Am. J. Manag. Care 2017, 23, S27–S36. [Google Scholar] [PubMed]
  145. Saeed, S.; Quintin, J.; Kerstens, H.H.D.; Rao, N.A.; Aghajanirefah, A.; Matarese, F.; Cheng, S.-C.; Ratter, J.; Berentsen, K.; van der Ent, M.A.; et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science 2014, 345, 1578. [Google Scholar] [CrossRef] [PubMed]
  146. Arts, R.J.; Novakovic, B.; ter Horst, R.; Carvalho, A.; Bekkering, S.; Lachmandas, E.; Rodrigues, F.; Silvestre, R.; Cheng, S.-C.; Wang, S.-Y.; et al. Glutaminolysis and Fumarate Accumulation Integrate Immunometabolic and Epigenetic Programs in Trained Immunity. Cell Metab. 2016, 24, 807–819. [Google Scholar] [CrossRef] [PubMed]
  147. Kornberg, M.D.; Bhargava, P.; Kim, P.M.; Putluri, V.; Snowman, A.M.; Putluri, N.; Calabresi, P.A.; Snyder, S.H. Dimethyl fumarate targets GAPDH and aerobic glycolysis to modulate immunity. Science 2018, 360, 449–453. [Google Scholar] [CrossRef] [PubMed]
  148. Acker, K.P.; Lung, T.W.F.; West, E.; Craft, J.; Narechania, A.; Smith, H.; O’Brien, K.; Moustafa, A.M.; Lauren, C.; Planet, P.J.; et al. Strains of Staphylococcus aureus that Colonize and Infect Skin Harbor Mutations in Metabolic Genes. iScience 2019, 19, 281–290. [Google Scholar] [CrossRef]
  149. Gabryszewski, S.J.; Lung, T.W.F.; Annavajhala, M.K.; Tomlinson, K.L.; Riquelme, S.A.; Khan, I.N.; Noguera, L.P.; Wickersham, M.; Zhao, A.; Mulenos, A.M.; et al. Metabolic Adaptation in Methicillin-Resistant Staphylococcus aureus Pneumonia. Am. J. Respir. Cell Mol. Biol. 2019, 61, 185–197. [Google Scholar] [CrossRef] [PubMed]
  150. Tulkens, P.M. Intracellular distribution and activity of antibiotics. Eur. J. Clin. Microbiol. Infect. Dis. 1991, 10, 100–106. [Google Scholar] [CrossRef]
  151. Renard, C.; Vanderhaeghe, H.J.; Claes, P.J.; Zenebergh, A.; Tulkens, P.M. Influence of conversion of penicillin G into a basic derivative on its accumulation and subcellular localization in cultured macrophages. Antimicrob. Agents Chemother. 1987, 31, 410–416. [Google Scholar] [CrossRef]
  152. Auberttulkens, G.; Vanhoof, F.; Tulkens, P. Gentamicin-induced lysosomal phospholipidosis in cultured rat fibroblasts—Quantitative ultrastructural and biochemical-study. Lab. Investig. 1979, 40, 481–491. [Google Scholar] [PubMed]
  153. Cosgrove, S.E.; Vigliani, G.A.; Fowler, V.G., Jr.; Abrutyn, E.; Corey, G.R.; Levine, D.P.; Rupp, M.E.; Chambers, H.F.; Karchmer, A.W.; Boucher, H.W. Initial low-dose gentamicin for Staphylococcus aureus bacteremia and endocarditis is nephrotoxic. Clin. Infect. Dis. 2009, 48, 713–721. [Google Scholar] [CrossRef] [PubMed]
  154. Barcia-Macay, M.; Seral, C.; Mingeot-Leclercq, M.-P.; Tulkens, P.M.; van Bambeke, F. Pharmacodynamic Evaluation of the Intracellular Activities of Antibiotics against Staphylococcus aureus in a Model of THP-1 Macrophages. Antimicrob. Agents Chemother. 2006, 50, 841–851. [Google Scholar] [CrossRef] [PubMed]
  155. Proctor, R.A.; Kahl, B.; von Eiff, C.; Vaudaux, P.E.; Lew, D.P.; Peters, G. Staphylococcal Small Colony Variants Have Novel Mechanisms for Antibiotic Resistance. Clin. Infect. Dis. 1998, 27, S68–S74. [Google Scholar] [CrossRef] [PubMed]
  156. Somerville, G.A.; Chaussee, M.S.; Morgan, C.I.; Fitzgerald, J.R.; Dorward, D.W.; Reitzer, L.J.; Musser, J.M. Staphylococcus aureus Aconitase Inactivation Unexpectedly Inhibits Post-Exponential-Phase Growth and Enhances Stationary-Phase Survival. Infect. Immun. 2002, 70, 6373–6382. [Google Scholar] [CrossRef] [PubMed]
  157. Chatterjee, I.; Kriegeskorte, A.; Fischer, A.; Deiwick, S.; Theimann, N.; Proctor, R.A.; Peters, G.; Herrmann, M.; Kahl, B.C. In Vivo Mutations of Thymidylate Synthase (Encoded by thyA) Are Responsible for Thymidine Dependency in Clinical Small-Colony Variants of Staphylococcus aureus. J. Bacteriol. 2008, 190, 834–842. [Google Scholar] [CrossRef] [PubMed]
  158. Chambers, H.F.; Miller, M.H. Emergence of Resistance to Cephalothin and Gentamicin During Combination Therapy for Methicillin-Resistant Staphylococcus aureus Endocarditis in Rabbits. J. Infect. Dis. 1987, 155, 581–585. [Google Scholar] [CrossRef] [PubMed]
  159. Conlon, B.P.; Rowe, S.E.; Gandt, A.B.; Nuxoll, A.S.; Donegan, N.P.; Zalis, E.A.; Clair, G.; Adkins, J.N.; Cheung, A.L.; Lewis, K. Persister formation in Staphylococcus aureus is associated with ATP depletion. Nat. Microbiol. 2016, 1, 1–7. [Google Scholar] [CrossRef]
  160. Mitchell, G.; Gattuso, M.; Grondin, G.; Marsault, É.; Bouarab, K.; Malouin, F. Tomatidine Inhibits Replication of Staphylococcus aureus Small-Colony Variants in Cystic Fibrosis Airway Epithelial Cells. Antimicrob. Agents Chemother. 2011, 55, 1937–1945. [Google Scholar] [CrossRef]
Figure 1. Concept of antimicrobial resistance, tolerance, and persistence in S. aureus and potential impact on treatment outcomes. Different bacterial populations are represented by the color scheme (gold, susceptible; green, tolerant; reddish-brown, resistant).
Figure 1. Concept of antimicrobial resistance, tolerance, and persistence in S. aureus and potential impact on treatment outcomes. Different bacterial populations are represented by the color scheme (gold, susceptible; green, tolerant; reddish-brown, resistant).
Ijms 25 06486 g001
Figure 2. Model of intracellular lifecycle and anaerobic metabolism of S. aureus contributing to persistent infection.
Figure 2. Model of intracellular lifecycle and anaerobic metabolism of S. aureus contributing to persistent infection.
Ijms 25 06486 g002
Figure 3. Mechanisms of reduced antibiotic activity against intracellular S. aureus.
Figure 3. Mechanisms of reduced antibiotic activity against intracellular S. aureus.
Ijms 25 06486 g003
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

Volk, C.F.; Proctor, R.A.; Rose, W.E. The Complex Intracellular Lifecycle of Staphylococcus aureus Contributes to Reduced Antibiotic Efficacy and Persistent Bacteremia. Int. J. Mol. Sci. 2024, 25, 6486. https://doi.org/10.3390/ijms25126486

AMA Style

Volk CF, Proctor RA, Rose WE. The Complex Intracellular Lifecycle of Staphylococcus aureus Contributes to Reduced Antibiotic Efficacy and Persistent Bacteremia. International Journal of Molecular Sciences. 2024; 25(12):6486. https://doi.org/10.3390/ijms25126486

Chicago/Turabian Style

Volk, Cecilia F., Richard A. Proctor, and Warren E. Rose. 2024. "The Complex Intracellular Lifecycle of Staphylococcus aureus Contributes to Reduced Antibiotic Efficacy and Persistent Bacteremia" International Journal of Molecular Sciences 25, no. 12: 6486. https://doi.org/10.3390/ijms25126486

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