Next Article in Journal
Bacterial Infections among Patients with Chronic Diseases at a Tertiary Care Hospital in Saudi Arabia
Next Article in Special Issue
Postoperative Infection after Anterior Cruciate Ligament Reconstruction: Prevention and Management
Previous Article in Journal
Correction: McLennan et al. Assessing the Use of Molecular Barcoding and qPCR for Investigating the Ecology of Prorocentrum minimum (Dinophyceae), a Harmful Algal Species. Microorganisms 2021, 9, 510
Previous Article in Special Issue
Comparative Phenotypic and Genomic Features of Staphylococci from Sonication Fluid of Orthopedic Implant-Associated Infections with Poor Outcome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Staphylococcal Biofilm on the Surface of Implants in Orthopedic Infection

Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
*
Author to whom correspondence should be addressed.
Microorganisms 2022, 10(10), 1909; https://doi.org/10.3390/microorganisms10101909
Submission received: 17 August 2022 / Revised: 11 September 2022 / Accepted: 20 September 2022 / Published: 26 September 2022

Abstract

:
Despite advanced implant sterilization and aseptic surgical techniques, implant-associated infection remains a major challenge for orthopedic surgeries. The subject of bacterial biofilms is receiving increasing attention, probably as a result of the wide acknowledgement of the ubiquity of biofilms in the clinical environment, as well as the extreme difficulty in eradicating them. Biofilm can be defined as a structured microbial community of cells that are attached to a substratum and embedded in a matrix of extracellular polymeric substances (EPS) that they have produced. Biofilm development has been proposed as occurring in a multi-step process: (i) attachment and adherence, (ii) accumulation/maturation due to cellular aggregation and EPS production, and (iii) biofilm detachment (also called dispersal) of bacterial cells. In all these stages, characteristic proteinaceous and non-proteinaceous compounds are expressed, and their expression is strictly controlled. Bacterial biofilm formation around implants shelters the bacteria and encourages the persistence of infection, which could lead to implant failure and osteomyelitis. These complications need to be treated by major revision surgeries and extended antibiotic therapies, which could lead to high treatment costs and even increase mortality. Effective preventive and therapeutic measures to reduce risks for implant-associated infections are thus in urgent need.

1. Introduction

With the rapid progress of modern medical technology, medical devices such as prosthetic joints, pacemakers, and catheters, etc. are playing an increasingly important role in medical care, which has completely changed medicine [1,2,3]. This is especially true in the field of orthopedics, because orthopedic implants are widely used for fracture fixation, deformity correction, joint replacement, and soft tissue anchorage [4].
However, due to the lack of self-cleaning ability, the use of implants also increases the risk of infection, which is the main reason for the failure of implants [5]. The infection caused by opportunistic pathogens can be cleared by the host’s immune system, while planting plants will expose the body to the risk of permanent colonization and planting failure. For example, when there is a foreign body at the surgical site, infected rabbits need much fewer Staphylococcus aureus cells than when there is no implant [6].
Biofilm can be defined as a structured microbial community of cells that are attached to a substratum and embedded in a matrix of extracellular polymeric substances (EPS) that they have produced [7]. Foreign implants placed in the patient’s body provide a colonization surface for bacteria so that bacteria and plasma proteins can adhere to it, and further form bacterial biofilm. Studies have indicated that bacterial cells in biofilm show different phenotypes from planktonic bacteria in terms of growth rate and gene transcription. Biofilms enable the bacteria embedded in them more resistant to antibiotics and less sensitive to host immune defense biological attack and are a source of persistent bacteremia as well as localized tissue destruction [8,9].
Staphylococci, with the leading species Staphylococcus aureus and Staphylococcus epidermidis, are the most common causes of implant-associated infections [10]. Whereas S. aureus has many mechanisms and virulence factors to evade the immune system, S. epidermidis must rely primarily on the ability to form a biofilm to survive in the host [11].
Clinical practice and experimental studies indicated clearly that in most cases, antibiotic therapy alone is insufficient to eradicate biofilm infection, and implants infected by bacteria usually require surgical resection [12,13]. In addition, it must be mentioned that each surgery to replace failed implants has a higher risk of recurrence (up to 30%) than that of the initial operation [14]. To solve these problems, great research progress has been made in the therapeutic approaches of biofilm, such as the studies on surface modification of implants, phage therapy, vaccine technology, and nanomedicines, but these new treatment methods need further preclinical and clinical validation before they are widely promoted for clinical use [15,16].
In this review, we focused on implant-associated infections in the orthopedic field, but some of the principles discussed also apply to other implants. We briefly discussed the microbial epidemiology of infected orthopedic implants, the formation process, and the function of biofilm. We also summarized the current strategies for the prevention and treatment of implant-associated infections, and emphasized the need to further explore biofilm physiology and carry out innovative research on anti-biofilm methods.

2. Microbial Epidemiology of Infections in Orthopedic Implants

Infection represents a terrible complication after orthopedic surgery [17]. There are significant differences in the etiology of implant-associated infection, which mainly depends on the type and anatomical location of the implant as well as the postoperative time [18]. Although there are some subtle differences in the time division of different types of orthopedic implant infection, the infection can be divided into three stages: early infection, late-acute infection, and late infection according to the time interval between surgery and clinical feature. Early infections are mainly caused by toxic microorganisms such as Staphylococcus aureus. Late-acute infections are usually caused by bacteria with low toxicity, such as coagulase-negative Staphylococcus. Late infections can be symptomatic infection evolved from an initially asymptomatic surgical infection or new infections caused by hematogenous seeding [19,20].
So far, there have been many epidemiological studies on orthopedic implant-associated infections. The results derived from the new study matched rather well with, and confirmed those of, the old epidemiological investigation [21,22,23,24]. In general, Gram-positive bacteria are the most common causes of orthopedic implant-associated infection, such as Staphylococcus aureus and Staphylococcus epidermidis. Aerobic Gram-negative bacilli, including Enterobacteria and P. aeruginosa, are less frequent causes of infection, and Anaerobic bacteria are seen far less frequently [19,24,25].
The largest study (thousands of cases containing 2524 adult patients with a diagnosis of prosthetic joint infection (PJI)) aimed at exploring the etiology of PJI was completed by Benito et al. in 2016. The results showed that the majority of cases of PJIs were caused by Aerobic Gram-positive cocci (77.7%), followed by aerobic gram-negative bacilli (27.6%). In addition, the rate of multidrug-resistant infections by resistant Gram-negative bacilli increased from 2003 through 2012, suggesting that reassessing empiric and specific antimicrobial therapy of PJIs could be required [26]. Recently Valérie Zeller et al. took 926 patients with PJI as samples, analyzed the microorganisms causing joint prosthesis infection, and compared their distribution according to the classification of PJI. Considering the 997 PJIs, staphylococci were the most frequently isolated microorganism (50%), followed by 16% streptococci, 11% Gram-negative rods, 8% anaerobic bacteria (predominantly Cutibacteriumacnes), and 9% polymicrobial infections. Early postoperative PJIs are characterized by a high rate of staphylococcal infections (57%), especially S. aureus (25%) and S. epidermidis (25%). For late-chronic PJIs, the most frequent microbes were low virulent microbes, such as coagulase-negative staphylococci (61%), with a high rate of S. epidermidis infections (35%). It is worth mentioning that among all PJI categories, hematogenous PJIs accounted for the largest proportion (35%) [21]. This observation is in contrast to spinal implant-associated infections where about 98% of them originated from perioperative colonization [22]. Besides, Murdoch et al. observed that internal fixation devices have a lower risk of hematogenous infection than joint prostheses [27]. This phenomenon may be caused by the continuous production of synovial fluid, which may expose the joint prosthesis to microorganisms that filtered from the blood to the joint space.
A study of 242 orthopedic patients showed that S. aureus was the main pathogen among infections associated with internal and external fixation systems, while S. epidermidis was the most prevalent bacterial species among infections associated with knee and hip arthro-prostheses [24]. In another study of 250 patients diagnosed with spinal implant-associated infections, Gram-negative bacteria were found mainly distributed in the lumbosacral segment because its anatomy is close to the genitourinary system and intestinal system [22].

3. Staphylococcal Biofilm Development

In orthopedic infection, the bacteria that cause implant failure are often not dispersed, but form biofilms [28], which occurs through a series of physical, chemical, and biological processes. Biofilm is a microbial community of bacteria that irreversibly attaches to a biomaterial surface and embedded in an abundant matrix of extracellular polymeric substances (EPSs) that they have produced [29].
The classic model of Staphylococcal biofilm development, which applies to S. aureus and S. epidermidis, is a complex process that can be divided into several main stages: (i) attachment and adherence, (ii) accumulation/maturation due to cellular aggregation and EPS production, and (iii) biofilm detachment (also called dispersal) of bacterial cells [19,30].
The solid–liquid interface between the implant surface and liquid media (such as water and blood) provides an ideal environment for the attachment and growth of microorganisms. Bacteria must first attach and adhere to biological or abiotic implant surfaces, which is a prerequisite for plant-related infection [29].
Although scientists have carried out many studies to study the exact mechanism involved in the process of bacterial adhesion, we still do not fully understand it. According to Gristina [31], when a foreign body in the form of biomaterial is implanted into a mammalian host, “a race for the surface” begins. The concept of “race for the surface” is proposed to describe the competition between host tissue cell integration and bacterial colonization at implant surfaces. If the host tissue cells occupy the available plant surface earlier, then tissue integration can be successfully carried out later, so the risk of infection will be reduced. However, if bacterial cells adhere to the surface of biomaterials before the host tissue cells, the surface is colonized by bacteria, which inevitably leads to implant-associated infection [32].
The attachment of bacteria to abiotic surfaces is instantaneous and reversible [4]. The physical forces involved in this process are hydrophobic, electrostatic, and Van der Waals forces, with bacteria behaving like colloidal particles. Some cell-surface multi-subunit protein polymers, known as pili or fimbriae, have an important role in the attachment to abiotic surfaces by many pathogenic bacteria [33]. Others specifically increase the direct adhesion of bacteria to implants by binding to host cell surface molecules and/or ECM components, such as collagen and fibronectin [34]. The autolysins AtlA [35,36] from S. aureus and the homologous protein AtlE [37] from S. epidermidis are bi-functional cell wall-anchored proteins, mediating binding to abiotic surfaces such as naked polystyrene. Some components on the surface of Staphylococcus, such as teichoic acid, also contribute to the adhesion to the surface of abiotic materials [38,39].
Actually, in vivo bacteria mainly encounter biotic surfaces composed of host matrixes including fibrinogen, fibronectin and collagen, because implants are rapidly covered by extracellular matrix (ECM) proteins when submerged in physiological fluids [34,40]. The process of plasma proteins attaching to the biomaterial’s surface is governed by the physical and chemical properties of the material surface such as polarity and roughness of the material, as well as the characteristics of the plasma proteins such as size and charge [41]. After being attached to the surface of biomaterials, these proteins form an ideal scaffold which provides a foothold for bacterial adhesion and subsequent proliferation [14]. For this purpose, staphylococci have evolved a series of cell wall-anchored (CWA) proteins, of which the best-characterized group is called MSCRAMMs (microbial surface components recognizing adhesive matrix molecules) [42,43], each specifically binding to ECM proteins covering the devices. Some examples of staphylococcal MSCRAMMs have been studied to full advantage, including fibronectin-binding proteins (FnBPA and FnBPB) [44,45,46,47], the fibrinogen-binding clumping factor A (ClfA) and clumping factor B (ClfB) [48,49,50,51], the serine–aspartate repeat protein family (Sdr) [52,53,54,55,56] and the collagen-binding protein (Cna) [57,58,59]. All MSCRAMMs contain a conserved C-terminal cell wall-targeting Leu-Pro-X-Thr-Gly (LPXTG) motif that covalently anchors the MSCRAMMs to the peptidoglycan by the specified protein, sortases (a membrane protein of Staphylococcus aureus) [60]. The mechanism is mainly that sortase catalyzes the cleavage of polypeptides between threonine and glycine in the LGXTG motif, and then mediates the formation of an amide bond between the carboxyl group of threonine and the amino group of peptidoglycan cross-bridges [61,62,63]. Some MSCRAMMs, such as Staphylococcus epidermidis surface protein serine-aspartate repeat protein G (SdrG), have a specific domain (SdrGN2N3) composed of N2 and N3 at the N-terminus, which binds to a ligand by the “dock, lock and latch” (DLL) mechanism [64,65]. Staphylococcus aureus surface protein can also bind to keratin [50,66], complement system protein [67,68], and other chains of fibrinogen [69] and collagen [57] through a similar DLL mechanism. For a more detailed overview of MSCRAMMs of Gram-positive cocci, readers can refer to a review by Foster [42].
After initial adhesion to a surface, these bacteria start to proliferate, gradually forming a multi-layered microcolony. At the same time, the bacteria also start to produce EPS, which eventually forms the biofilm matrix. The biofilm matrix is mainly composed of polysaccharide, protein, teichoic acid, and extracellular DNA (eDNA) as well as other molecules on the whole constituting a physical barrier between the microbial community and the extracellular environment [70,71].
Staphylococcus epidermidis and Staphylococcus aureus mainly produce polysaccharide intercellular adhesin (PIA) as the main polysaccharide of the biofilm matrix [10,72]. Due to the enzyme-catalyzed removal of partial N-acetyl groups, PIA has a cationic character [73], which allows electrostatic interaction with other negatively charged biofilm components (such as teichoic acid). The production of PIA is dependent on the intracellular adhesion (ica) operon, which contains four genes in sequence: icaA, icaD, icaB, and icaC [72,74,75]. Under the promotion of IcaD, the enzyme IcaA (an N-acetylglucosaminytransferase) synthesizes PIA oligomers with UDP-N-acetylglucosamine (UDP-GlcNAc) as the precursor. IcaC is involved in the synthesis of long-chain PIA and allows the translocation of PIA across the cell membrane [76]. IcaB is a secreted N-deacetylase that is responsible for the above-mentioned partial deacetylation of PIA [77]. In addition, icaR and tcaR are two negative regulatory genes that regulate the transcription of icaADBC operon [78]. Studies have suggested TcaR functions as a repressor only in the absence of IcaR [79,80].
Some studies have found that although several strains of Staphylococcus epidermidis and Staphylococcus aureus do not produce PIA, they can also form a biofilm with protein as the main component both in vivo and in vitro [81,82,83]. These staphylococcal surface proteins include fibronectin-binding proteins (FnBPA and FnBPB), serine aspartate repeat protein (SdrC), accumulation-associated protein (Aap), staphylococcal surface protein G (SasG) and biofilm-associated protein (Bap). Some MSCRAMMs not only play a role in bacterial adhesion, but also promote cell–cell interaction, resulting in increased biofilm formation. For example, the A domain of FnBPs and SdrC participates in homologous interactions and promotes the cell accumulation of staphylococcal cells during biofilm formation [53,82,84,85]. In the PIA-independent staphylococcal biofilm, there are also some specific bacterial surface proteins, such as accumulation-associated protein (Aap) from S. epidermidis and Staphylococcus aureus Aap homologue SasG (Staphylococcus aureus surface protein G). Specifically, mature Aap or SasG protein can mediate intercellular adhesion through several repeated B domains [86,87,88,89,90]. In addition, some studies have shown that PIA-dependent biofilm has fibrous structures that are absent in protein-dependent biofilm, and has higher strength and ductility than protein-based biofilm [91,92,93].
Extracellular DNA (eDNA) was more recently discovered to take part in supramolecular structures such as stable filamentous networks which work as a net to hold biofilm cells together [94]. Having an extremely important role, eDNA has been found to mediate horizontal gene transfer [95], limit the spread of antibiotics [96], promote antibiotic resistance phenotypes [97], interact with cells of the host immune system and regulate innate and cell-mediated immune responses [98], guide biofilm diffusion [99], and serve as a nutrient source during periods of nutrient deficiency [100].
With the proliferation of bacteria and the increase of EPSs, mature biofilm is finally formed, which is characterized by the presence of nonuniform growth of microcolonies surrounded by water channels that help to distribute nutrients and signal molecules [101]. Extensive in vitro studies have shown that the formation of water channels in mature biofilm is mainly completed by a polypeptide family called phenol-soluble modulins (PSMs) [102].
PSMs are a family of small peptides (~21–45 amino acids long) and have amphipathic, α-helical secondary structures. The smaller, ~20 amino acid peptides are grouped into the α-class and the longer ~45 amino acid peptides into the β-class of PSMs. PSMs exist in a monomeric state, where they have surfactant-like properties, and in a polymeric state, where they fold into an amyloid structure which can stabilize the biofilm structure [103].
The surfactant and lytic properties are believed to allow PSMs to disrupt non-covalent (electrostatic or hydrophobic) interactions between biofilm matrix components, thereby producing the characteristic fluid channels that are embedded in the biofilm matrix.
To date, eight different PSMs have been characterized in S. aureus, including four PSMα peptides, two PSMβ, δ-toxin, and PSM-mec. Compared with wide-type strain, S. aureus psm-mec mutants only show slightly decreased capacity to form biofilms [104], while the other types of isogenic S. aureus psm mutants produced more compact and extended biofilms which demonstrated less channel formation [105]. S. epidermidis produces six PSM peptides, PSMα, PSMβ1, PSMβ2, PSMδ, PSMε, and δ-toxin, but only the β-type PSMs were well investigated for their biofilm effects. Studies show that different concentrations of PSMβ play disparate roles in different stages of S. epidermidis biofilm cycle. At medium concentrations, PSMβ promoted the formation of channels and the growth of biofilm, while at high concentrations, they destroyed biofilm, thereby resulting in biofilm dispersal [91].
Finally, the third stage of biofilm development is the dispersal of bacterial cells from biofilm, which is an active process of bacteria escaping from the biofilm [106]. Transcriptome analyses of drip reactor-grown P. aeruginosa biofilms show that the steepening of the chemical concentration gradient of nutrient resources, oxygen, and waste seems to drive the dispersion of bacterial biofilm, during the development of biofilm [107]. In order to survive when there is a limitation of nutrients or to establish infection at distant sites in the human body, some bacterial cells detach from the biofilm individually or in agglomerates, leaving behind eroded biofilms with central voids.
Compared with their biofilm and planktonic counterparts, dispersed bacteria express high levels of genes encoding virulence factors and genes important for motility and adhesion, which may lead to subsequent more serious infection [108].
Mechanical stress or shear force caused by blood flow and equipment flushing is considered to be the main mechanism limiting the overall biomass accumulation of the biofilm [109]. In addition, various enzymes and PSMs produced by bacteria that degrade the components of biofilm matrix all contribute to the stage of biofilm dispersal.
The important role of PSM in biofilm dispersal has been confirmed in vivo experiments. In a murine model of device-related infection caused by Staphylococcus epidermidis, bacteria harvested from body fluids and lymph nodes were overwhelmingly more of the WT than psmβ mutant strain, indicating PSMβ contributes to the dissemination of biofilm-associated infection [91].
With a deeper understanding of the matrix components of biofilm, the enzymes that degrade EPSs have also been widely studied. A variety of enzymes, including proteases, glycoside hydrolases, and nucleases, have been tested for their ability to degrade biofilm and promote biofilm dispersal [110]. The relative importance of each enzyme is decided by the strain-specific components of the biofilm matrix.
S. aureus produces and secretes four extracellular proteases of different protease families that may digest proteinaceous matrix components: metalloprotease aureolysin (Aur), serine protease SspA (also named V8 protease), and two cysteine proteases SspB and ScpA. Some of these proteases have been identified to degrade specific matrix proteins. For example, SspA digests FnBPs and Bap, whereas Aur degrades Bap as well as ClfB [110,111,112].
The biofilm matrix component eDNA is degraded by nuclease. To our knowledge, S. aureus nucleases Nuc1 and Nuc2 modulate biofilm formation [113,114].
A large number of studies have been carried out on the decomposing of extracellular polysaccharides by glycoside hydrolases as a means of dispersing biofilms. So far, an enzyme secreted by staphylococcus that can degrade PIA has never been found. Dispersin B, a glycoside hydrolase produced by Actinobacillus actinomycetemcomitans, has been shown to degrade PIA by hydrolyzing β-1,6-glycosidic linkages [115]. This enzyme is effective against biofilms made by a variety of bacteria, including S. aureus and S. epidermidis [116]. Cellulase is a glycoside hydrolase produced by multiple microbes that hydrolyzes the β-1,4-glycosidic linkage. It has been shown to cause the dispersal of S. aureus biofilms [117].
Dispersion is also associated with increased bacterial cell death in local hypoxic areas within the biofilm. Nitric oxide (NO) produced by bacteria through anaerobic denitrification react with superoxide to generate the cell-toxic radical ONOO. ONOO leads to cellular damage and cell lysis, which has a synergistic effect with enzyme-mediated destruction of biofilm matrix [118].

4. The Role of Staphylococcal Biofilm during Infection

The formation of biofilms creates significant challenges for treating implant-associated infections because bacteria in biofilms display unique characteristics and behavior compared with their planktonic counterparts, with the hallmark characteristics including an innate resistance to antimicrobials and host immune defenses.

5. Antibiotic Tolerance

It has been reported that the tolerance of bacteria in biofilm to antibiotics is 10- to 1000-fold higher for that of their planktonic counterparts [119]. Notably, bacteria with increased resistance in biofilm have the same minimum inhibitory concentration as the planktonic controls after dispersing from the biofilm. This indicates that the existence of biofilm plays a significant role in the enhancement of bacterial antibiotic tolerance [120]. The increase of antibiotic resistance of bacteria in biofilm is the comprehensive result of different mechanisms, including slow growth and reduced metabolic rates [121], the existence of persister cells [122], and the barrier effect of the biofilm matrix [123].
Compared with planktonic bacteria, bacteria growing at different locations in the biofilm community experience different concentration gradients of oxygen, nutrient resources, and metabolic wastes, as well as extracellular signal molecules [124], which will lead to changes in the metabolic rate of bacteria within the biofilm. In addition, the higher bacterial cell density in the biofilm community contributes to the transfer of tolerance genes between bacterial cells, which promotes the formation of subpopulations with different specialties in the community [125]. It is considered that an important reason why antibiotics cannot effectively remove biofilm bacteria is that biofilm communities can harbor tolerant and persister cells [126]. They contribute to the formation of chronic infections and complicate the treatment of implant infections. Common targets of antibiotics, such as RNA polymerase and cell-wall biosynthetic enzymes, represent low activity in metabolically inactive cells, and thus, slow-growing cells can survive transient or periodic antibiotic treatment and regrow when antibiotics are withdrawn. Unlike antibiotic resistance, antibiotic resistance can be caused by many, not a few, gene mutations, so tolerance mutations occur more frequently than resistance mutations. Persister cells are subpopulations of phenotypically dormant cells, originated from part of pre-existing non-growing cells or cells that are dormant at rest during stationary phase. Mutations in genes encoding tRNA synthetase, ribose-phosphate diphosphatekinase and toxin antitoxin (TA) systems are all tightly associated with induced cell dormancy, persister cell formation, and antibiotic tolerance [127].
Another factor promoting to development of antibiotic tolerance is the biofilm environment. Biofilm matrix containing numerous charged molecules (e.g., proteins, glycoproteins, polysaccharide, eDNA, etc.) can bind charged antimicrobial agents and provide a physical barrier that protects the microorganisms from immune components [128]. The specific interactions between the components of the biofilm matrix give rise to the necessarily global biofilm mechanical properties, which can protect the bacteria from external forces such as fluid shear and ensure the biofilm community remains attached to a surface. Moreover, the strength of the in vivo biofilm scaffold exceeds the maximum mechanical stress that neutrophils can exert, which makes it difficult for bacteria in the biofilm to be completely cleared by the immune system. Therefore, the biofilm matrix scaffold can still harbor tolerant or persister cells that can regrow and cause chronic infection even if the majority of actively growing pathogens in biofilm have been killed by antibiotics.

6. Immune Evasion

Bacterial invasion commonly stimulates a strong inflammatory response, which includes involves complement activation, recruitment of phagocytes (most importantly neutrophils), and subsequent killing of the pathogen. Neutrophils recruited to the infected site can kill bacteria by phagocytosis, degranulation of antimicrobial substances into the environment, or neutrophil extracellular trap (NET) formation [129]. In the past, it has been proposed that the resistance of bacteria in biofilm to immune cells is mainly due to the physical barrier effect of biofilm matrix [130]. However, in vitro studies have suggested that human leukocytes are able to penetrate biofilm matrix effectively and bacteria in biofilms are not inherently protected from phagocytic cells [131].
S. aureus relies on several immune-evasion proteins and cytolytic toxins that kill immune cells directly to evade the host immune response, and by skewing host immunity toward an anti-inflammatory, pro-fibrotic response instead of a pro-inflammatory, bactericidal response that favors staphylococcal persistence [132,133]. Fibronectin (Fn) binds to fibronectin-binding proteins (FnBPs) on the surface of Staphylococcus aureus and to α5β1 integrin on the surface of osteoblasts, thus forming a FnBP–Fn–α5β1 bridge that triggers the invasion of osteoblasts [134]. Inside the host cells, S. aureus is not vulnerable to extracellular antibiotics and immune cells. In addition, S. aureus secretes multiple proteins to prevent antibody opsonization and interfere with the complement system. For example, Staphylococcal protein A and Staphylococcal binder of immunoglobulin (Sbi) block Fc receptor-mediated phagocytosis by binding IgG in the wrong orientation [135]. Extracellular adherence protein (Eap) blocks the formation of lectin and classical pathway C3 convertases, thereby blocking C3b deposition and C5a production [136]. S. aureus also secretes toxins that induce immune cell lysis by disrupting the cell membrane [137]. Some toxins specifically target leukocytes via the interaction with receptors, including alpha-hemolysin (Hla) and bicomponent leukocidins Panton-Valentine leukocidin (PVL), gamma-hemolysin (HlgAB, HlgCB), leukocidinED (LukED), and leukocidinAB/GH (LukAB/GH).
Compared with S. aureus, S. epidermidis has fewer mechanisms to evade immune defenses, and its pathogenicity relies mainly on biofilm formation [138]. PIA by deacetylation of its poly-N-acetylglucosamine molecule forms a sort of positively charged “capsule” around S. epidermidis, which has been shown to protect against neutrophil phagocytosis and AMPs presumably by charge repulsion and physical barrier function [77,139]. Animal experiments and the observation of human Staphylococcus epidermidis biofilm infection showed that the immune response of wild-type Staphylococcus epidermidis strains to biofilm infection was generally weakened compared with the immune response from ica-negative isogenic mutants [140,141]. It is the low immunogenicity of S. epidermidis that makes it only diagnosed in a late stage of infection, when there is a mature biofilm.

7. Prevention and Therapy

Over the last decade, with the steady increase of the impact of multidrug-resistant bacteria in healthcare-associated infections, plant biofilm-associated infections are becoming a public health problem of growing importance worldwide. In the Unites States, the annual morbidity and mortality of biofilm-associated infection are very high, with over $18 billion in direct treatment costs spent on these infections [142]. Clinical management of biofilm-associated infections is a challenging and multifactorial problem. Although various strategies have been adopted, so far there is no method that can completely avoid biofilm-related infection. The risk of surgical site infections in orthopedic surgeries can be effectively reduced with preoperative interventions for patients [143].
A simple example of preoperative interventions is optimizing preoperative medical conditions. Systemic prophylaxis in a 24 h regimen, especially with high-risk patients, is also one of the strategies to prevent infection.
Anti-infective biomaterials currently represent a main preventive strategy [144,145]. The causes of implant-associated infection are multifactorial, and bacterial adhesion to the implant surface is the premise of infection. The ideal implant surface would be one that minimizes bacterial adhesion, confers effective sterilization, and has no interference with bone healing and osteointegration [20]. Great efforts have been made to deal with the adhesion of bacteria to implants by modifying its surface to lower adhesive features or by the design of antibacterial coatings [4,146].
Implant coatings can be divided into passive and active according to the mode of action. Passive coatings reduce bacterial adhesion by altering the implant surface chemistry and/or surface structure modification without local release of fungicides to surrounding tissues. The active coating releases pharmacologically active pre-incorporated bactericidal agents, such as antibiotics, preservatives, metal ions, non-metal ingredients (e.g., iodine, selenium) and functional peptides, thereby reducing infection [15]. Unfortunately, these approaches do not result in complete clinical success, possibly due to the fact that implants are prone to be covered by plasma proteins quickly so that the anti-biofilm compounds are masked and their efficacy diminished.
Although the purpose of the abovementioned strategies is to prevent the formation of biofilm, it is still necessary to deal with the biofilm that has already been formed. Currently, the only remedy to ensure complete resolution of the infection is surgical excision of the implant, thorough debridement of the peri-implant environment, and prolonged antimicrobial therapy [147], which not only causes extreme suffering for patients but also a huge economic burden on the healthcare system. Novel strategies have been researched to eradicate the biofilm in situ, but so far, no method of in-situ biofilm eradication that would provide a working alternative to the use of antibiotics has yet been taken to clinical use.
Rifampicin, an inhibitor of bacterial RNA polymerase, is an important antibiotic drug for the treatment of staphylococcal prosthetic joint infections, as it is able to penetrate staphylococcal biofilms [148]. Owing to the risk of the quick development of rifampicin-resistant isolates, rifampicin should be used with other antibiotics [149]. In addition, fluoroquinolones show the characteristics required to effectively eliminate gram-negative bacilli biofilm [150].
In the following, some novel strategies to eradicate biofilm in situ are presented, but so far, there has been limited translation into clinical practice.
Bacteriophages (phages), viruses that specifically infect and kill their bacterial hosts, have been investigated as an alternative to antibiotic treatment due to the growing degree of anti-microbial resistance, and they have been studied in some clinical trials demonstrating safety and possible efficacy [151]. The bactericidal effect of phage depends on phage-derived lysine, which can decompose peptidoglycan, the main component of the bacterial cell wall, thereby inducing cell lysis.
A recent study in a murine model of PJI suggested that the combination of phages with antibiotics may increase the removal of bacteria. Both the orthopedic wires coated with phage K or coated with linezolid alone demonstrate a modest decrease in the bacterial load, but the use of dual coated implants incorporating lytic phage as well as linezolid presents a stronger antibacterial effect [152]. Therefore, according to the different mechanisms of killing bacteria between antibiotics and phages, researchers speculated that the combination of phages and antibiotics may be more effective in controlling bacteria than one of them alone [153]. Moreover, Ferry et al. successfully treated multiple patients with recurrent S. aureus and P. aeruginosa PJI by combining antibiotic therapy with phage therapy, and preserved their prosthetic joints. This indicates that the combination of phage therapy and antibiotic therapy may become a salvage measure for preserving prosthetic joints of patients with recurrent PJI [154,155]. However, it seems that this approach is only experimental because antibiotics still have to be maintained during the phage therapy.
Another novel treatment strategy is the use of the vaccine, but all clinical trials of anti-staphylococcal vaccines have failed [156]. Nevertheless, several vaccine candidates have been investigated that specifically target which biofilm matrix components are present in the biofilm growth. For example, immunization with PIA reduced the consequences of osteomyelitis infection from PIA-producing intercellular adhesion in an S. aureus periprosthetic osteomyelitis rat model [157]. As for S. epidermidis, there appears to have been some preclinical success with anti-Ses antibodies or active vaccination using recombinant truncated Ses in subcutaneous catheter mouse models [158].
Materials of nano size with unique physical and chemical properties, strong bactericidal activity, and specific mechanisms have shown great potential for eradicating resident bacteria and pathogenic biofilm [159]. According to the elemental composition, these nanomaterials can be divided into organic (e.g., metal and metallic oxide nanoparticles) and inorganic (e.g., lipid nanoparticles and smart nanomaterials) platforms. In addition to their multiple mechanisms, such as physical damage, oxidative stress, thermal damage, and so on, nanomaterials’ high surface-to-volume ratios and multivalent interactions enable them act as carriers for antibiotics or drug combination loading that may produce synergistic anti-biofilm efficacy. Critically, nanomaterials can be designed to become activated in response to unique biofilm pathologic microenvironments (e.g., pH, light, temperature, hypoxia, and enzymatic activation) so as to accurately carry, retain, and release drugs when and where needed most. Moreover, these dispersed nanoparticles can be assembled on platforms suitable for therapeutic purposes as well as in the form of various devices (bandages, catheters, implant coatings, etc.) to prevent and control infections [160].
Although nanoparticles have shown good inhibitory effects on biofilm formation, studies on the inhibitory effect of nanoparticles on persister bacteria in biofilm are still rare. Different from traditional antibacterial drugs, smart nanoparticles with new anti-biofilm mechanisms (such as photothermal therapy (PTT) and photodynamic therapy (PDT)) might be a more promising method to kill the persister cells [161]. PTT relying on the conversion of near-infrared light (NIR) into local heat by photothermal agents destroys the integrity of bacteria or the structure of biofilm through local hyperthermia, and can promote the diffusion of photosensitizers to biofilm at the same time. The main disadvantage of this strategy is that bacteria can only be effectively removed at higher temperatures. However, when high temperature kills the formed biofilm, it will have a huge negative impact on the surrounding healthy tissues. Although healthy tissues can withstand temperatures above 50 °C for a long time, the antibacterial effect is limited at such temperatures. If the sensitivity of the biofilm to heat is improved by using a safe photosensitive heat (below 50 °C), the biofilm formed on the surface of the implant can be eradicated without damaging the surrounding tissue [162]. PDT is another promising way. Nano materials or photosensitizers stimulate reactive oxygen species (ROS) under near-infrared or visible light irradiation, which will destroy the integrity of bacterial cell membrane, and lead to the enhancement of bacterial sensitivity to heat and bacterial death. The ROS produced by nano materials or photosensitizers under near-infrared or visible light irradiation will destroy the integrity of bacterial cell membrane, and improve the thermal sensitivity of the biofilm [163]. Therefore, the combination of photothermal therapy and photodynamic therapy in the treatment of implant related infections is a hot research field today. Recently, a study used a red-phosphorus-IR780-arginine-glycine-aspartic-acid-cysteine coating on titanium bone implant with irradiation of NIR light (808 nm) to eradicate biofilms by combining PTT and PDT. Red phosphorus can be used as an efficient photothermal coating for its great biocompatibility. The temperature sensitivity of S. aureus biofilm is enhanced in the presence of ROS produced by IR780. At the same time, arginine-glycine-aspartic-acid-cysteine decorated on the surface of the implant can facilitate the integration of implant materials and bone tissue. Finally, the technique demonstrated a rapid bactericidal effect that was biosafe at a safe temperature (~50 °C) [164].
Driven by the motivation to write high impact articles, researchers have created a large number of anti-biofilm therapies based on nanotechnology, and have carried out in vitro experiments or animal experiments for verification [165]. Unfortunately, there are still many challenges that need to be solved before nanomaterials are applied to clinical practice. Off-target toxicity of nanomaterials should be paid attention to first [166]. Metal and inorganic nanomaterials can be absorbed within the gastrointestinal tract. Although the bioavailability is very low and nanomaterials may degrade metabolism, the chronic off-target effect caused by them may be long-lasting. For example, magnetite iron oxide nanoparticles increase endothelial permeability by producing robust radicals, which may lead to hepatotoxicity and nephrotoxicity. Therefore, it is necessary to use nanoparticle systems prepared from degradable biomaterials as much as possible. Next, due to the physiological differences between animals and humans, it is still open to question whether the nanomedicines verified as safe by animal experiments can be used in humans. In addition, the production cost of nanoparticles is high, so the economic benefits brought by using nanotechnology to solve clinical problems cannot be accurately predicted at present. Based on the abovementioned challenges, we believe that the future direction should be to effectively eradicate biofilms, minimize toxicity and drug resistance, and promote low-cost and high-efficiency clinical applications.

8. Conclusions

Despite the rapid development of modern medical treatment, implant related infection is still a serious clinical problem faced by surgeons. As mentioned above, the implants implanted in the body compromise host defenses and provide a site for bacteria to adhere to and colonize it. Staphylococcus is the most common bacterium causing implant related infection, and the main pathogenic microorganisms are Staphylococcus aureus and Staphylococcus epidermidis. These bacteria can adhere to the surface of implants and form biofilm through various mechanisms. Bacteria growing in biofilm have stronger antibiotic resistance than planktonic bacteria, which leads to the fact that antibiotic treatment alone is not enough to eradicate biofilm. Therefore, different treatment strategies need to be combined and integrated to fight implant-associated infections.
Currently, the only remedy to ensure complete resolution of the infection is surgical excision of the implant, thorough debridement of the peri-implant environment, and prolonged antimicrobial therapy, which causes heavy physical, emotional, and financial burdens.
Novel strategies have been researched to prevent and eradicate the biofilm in situ, but so far, no method of in-situ biofilm eradication that would provide a working alternative to the use of antibiotics has yet been taken to clinical use.
Given that biofilm-related infection of implants is a major public health problem that has not yet been solved, there is an urgent need to carry out more basic and clinical research on Staphylococcus and other related pathogens infecting implants. Importantly, the in vivo evaluation of the therapeutic effect of anti-biofilm deserves more attention.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work. W.-j.C. and Z.R.: Conducted the literature search and planned the review. Y.L. wrote the first draft of the manuscript and subsequently revised it. P.H. provided technical suggestions and reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was sponsored by the National Natural Science Foundation of China (81974325) and Program of Shanghai Committee of Science and Technology (22ZR1447500).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tjong, F.V.; Reddy, V.Y. Permanent Leadless Cardiac Pacemaker Therapy: A Comprehensive Review. Circulation 2017, 135, 1458–1470. [Google Scholar] [CrossRef] [PubMed]
  2. Trebse, R.; Roskar, S. Evaluation and interpretation of prosthetic joint infection diagnostic investigations. Int. Orthop. 2021, 45, 847–855. [Google Scholar] [CrossRef] [PubMed]
  3. Tripathi, S.; Kumar, S.; Kaushik, S. The Practice and Complications of Midline Catheters: A Systematic Review. Crit. Care Med. 2021, 49, e140–e150. [Google Scholar] [CrossRef] [PubMed]
  4. Filipović, U.; Dahmane, R.G.; Ghannouchi, S.; Zore, A.; Bohinc, K. Bacterial adhesion on orthopedic implants. Adv. Colloid Interface Sci. 2020, 283, 102228. [Google Scholar] [CrossRef] [PubMed]
  5. Rochford, E.T.; Richards, R.G.; Moriarty, T.F. Influence of material on the development of device-associated infections. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2012, 18, 1162–1167. [Google Scholar] [CrossRef] [PubMed]
  6. Southwood, R.T.; Rice, J.L.; McDonald, P.J.; Hakendorf, P.H.; Rozenbilds, M.A. Infection in experimental arthroplasties. Clin. Orthop. Relat. Res. 1987, 224, 33–36. [Google Scholar] [CrossRef]
  7. Donlan, R.M.; Costerton, J.W. Biofilms: Survival mechanisms of clinically relevant microorganisms. Clin. Microbiol. Rev. 2002, 15, 167–193. [Google Scholar] [CrossRef]
  8. Kot, B.; Sytykiewicz, H.; Sprawka, I. Expression of the Biofilm-Associated Genes in Methicillin-Resistant Staphylococcus aureus in Biofilm and Planktonic Conditions. Int. J. Mol. Sci. 2018, 19, 3487. [Google Scholar] [CrossRef]
  9. Freitas, A.I.; Lopes, N.; Oliveira, F.; Brás, S.; França, Â.; Vasconcelos, C.; Vilanova, M.; Cerca, N. Comparative analysis between biofilm formation and gene expression in Staphylococcus epidermidis isolates. Future Microbiol. 2018, 13, 415–427. [Google Scholar] [CrossRef]
  10. Otto, M. Staphylococcal Biofilms. Microbiol. Spectr. 2018, 6, 207–228. [Google Scholar] [CrossRef]
  11. Foster, T.J. Immune evasion by staphylococci. Nat. Rev. Microbiol. 2005, 3, 948–958. [Google Scholar] [CrossRef] [PubMed]
  12. Da Silva, R.A.G.; Afonina, I.; Kline, K.A. Eradicating biofilm infections: An update on current and prospective approaches. Curr. Opin. Microbiol. 2021, 63, 117–125. [Google Scholar] [CrossRef] [PubMed]
  13. Wu, H.; Moser, C.; Wang, H.Z.; Høiby, N.; Song, Z.J. Strategies for combating bacterial biofilm infections. Int. J. Oral Sci. 2015, 7, 1–7. [Google Scholar] [CrossRef] [PubMed]
  14. Pietrocola, G.; Campoccia, D.; Motta, C.; Montanaro, L.; Arciola, C.R.; Speziale, P. Colonization and Infection of Indwelling Medical Devices by Staphylococcus aureus with an Emphasis on Orthopedic Implants. Int. J. Mol. Sci. 2022, 23, 5958. [Google Scholar] [CrossRef] [PubMed]
  15. Rodríguez-Merchán, E.C.; Davidson, D.J.; Liddle, A.D. Recent Strategies to Combat Infections from Biofilm-Forming Bacteria on Orthopaedic Implants. Int. J. Mol. Sci. 2021, 22, 10243. [Google Scholar] [CrossRef] [PubMed]
  16. Sharahi, J.Y.; Azimi, T.; Shariati, A.; Safari, H.; Tehrani, M.K.; Hashemi, A. Advanced strategies for combating bacterial biofilms. J. Cell. Physiol. 2019, 234, 14689–14708. [Google Scholar] [CrossRef]
  17. Masters, E.A.; Trombetta, R.P.; de Mesy Bentley, K.L.; Boyce, B.F.; Gill, A.L.; Gill, S.R.; Nishitani, K.; Ishikawa, M.; Morita, Y.; Ito, H.; et al. Evolving concepts in bone infection: Redefining “biofilm”, “acute vs. chronic osteomyelitis”, “the immune proteome” and “local antibiotic therapy”. Bone Res. 2019, 7, 20. [Google Scholar] [CrossRef]
  18. Campoccia, D.; Montanaro, L.; Arciola, C.R. A review of the clinical implications of anti-infective biomaterials and infection-resistant surfaces. Biomaterials 2013, 34, 8018–8029. [Google Scholar] [CrossRef]
  19. Arciola, C.R.; Campoccia, D.; Montanaro, L. Implant infections: Adhesion, biofilm formation and immune evasion. Nat. Rev. Microbiol. 2018, 16, 397–409. [Google Scholar] [CrossRef]
  20. Zimmerli, W. Clinical presentation and treatment of orthopaedic implant-associated infection. J. Intern. Med. 2014, 276, 111–119. [Google Scholar] [CrossRef]
  21. Zeller, V.; Kerroumi, Y.; Meyssonnier, V.; Heym, B.; Metten, M.A.; Desplaces, N.; Marmor, S. Analysis of postoperative and hematogenous prosthetic joint-infection microbiological patterns in a large cohort. J. Infect. 2018, 76, 328–334. [Google Scholar] [CrossRef] [PubMed]
  22. Margaryan, D.; Renz, N.; Bervar, M.; Zahn, R.; Onken, J.; Putzier, M.; Vajkoczy, P.; Trampuz, A. Spinal implant-associated infections: A prospective multicentre cohort study. Int. J. Antimicrob. Agents 2020, 56, 106116. [Google Scholar] [CrossRef] [PubMed]
  23. Holleyman, R.J.; Baker, P.N.; Charlett, A.; Gould, K.; Deehan, D.J. Analysis of causative microorganism in 248 primary hip arthroplasties revised for infection: A study using the NJR dataset. Hip Int. J. Clin. Exp. Res. Hip Pathol. Ther. 2016, 26, 82–89. [Google Scholar] [CrossRef] [PubMed]
  24. Montanaro, L.; Speziale, P.; Campoccia, D.; Ravaioli, S.; Cangini, I.; Pietrocola, G.; Giannini, S.; Arciola, C.R. Scenery of Staphylococcus implant infections in orthopedics. Future Microbiol. 2011, 6, 1329–1349. [Google Scholar] [CrossRef] [PubMed]
  25. Oliveira, W.F.; Silva, P.M.S.; Silva, R.C.S.; Silva, G.M.M.; Machado, G.; Coelho, L.; Correia, M.T.S. Staphylococcus aureus and Staphylococcus epidermidis infections on implants. J. Hosp. Infect. 2018, 98, 111–117. [Google Scholar] [CrossRef] [PubMed]
  26. Benito, N.; Franco, M.; Ribera, A.; Soriano, A.; Rodriguez-Pardo, D.; Sorlí, L.; Fresco, G.; Fernández-Sampedro, M.; Dolores Del Toro, M.; Guío, L.; et al. Time trends in the aetiology of prosthetic joint infections: A multicentre cohort study. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2016, 22, 732.e1–732.e8. [Google Scholar]
  27. Murdoch, D.R.; Roberts, S.A.; Fowler, V.G., Jr.; Shah, M.A.; Taylor, S.L.; Morris, A.J.; Corey, G.R. Infection of orthopedic prostheses after Staphylococcus aureus bacteremia. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2001, 32, 647–649. [Google Scholar] [CrossRef]
  28. Neut, D.; van Horn, J.R.; van Kooten, T.G.; van der Mei, H.C.; Busscher, H.J. Detection of biomaterial-associated infections in orthopaedic joint implants. Clin. Orthop. Relat. Res. 2003, 413, 261–268. [Google Scholar] [CrossRef]
  29. Donlan, R.M. Biofilms: Microbial life on surfaces. Emerg. Infect. Dis. 2002, 8, 881–890. [Google Scholar] [CrossRef]
  30. Schilcher, K.; Horswill, A.R. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol. Mol. Biol. Rev. MMBR 2020, 84, e00026-19. [Google Scholar] [CrossRef]
  31. Gristina, A.G.; Naylor, P.; Myrvik, Q. Infections from biomaterials and implants: A race for the surface. Med. Prog. Through Technol. 1988, 14, 205–224. [Google Scholar]
  32. Busscher, H.J.; van der Mei, H.C.; Subbiahdoss, G.; Jutte, P.C.; van den Dungen, J.J.; Zaat, S.A.; Schultz, M.J.; Grainger, D.W. Biomaterial-associated infection: Locating the finish line in the race for the surface. Sci. Transl. Med. 2012, 4, 153rv10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Mandlik, A.; Swierczynski, A.; Das, A.; Ton-That, H. Pili in Gram-positive bacteria: Assembly, involvement in colonization and biofilm development. Trends Microbiol. 2008, 16, 33–40. [Google Scholar] [CrossRef] [PubMed]
  34. Kline, K.A.; Fälker, S.; Dahlberg, S.; Normark, S.; Henriques-Normark, B. Bacterial adhesins in host-microbe interactions. Cell Host Microbe 2009, 5, 580–592. [Google Scholar] [CrossRef] [PubMed]
  35. Houston, P.; Rowe, S.E.; Pozzi, C.; Waters, E.M.; O’Gara, J.P. Essential role for the major autolysin in the fibronectin-binding protein-mediated Staphylococcus aureus biofilm phenotype. Infect. Immun. 2011, 79, 1153–1165. [Google Scholar] [CrossRef] [PubMed]
  36. Bose, J.L.; Lehman, M.K.; Fey, P.D.; Bayles, K.W. Contribution of the Staphylococcus aureus Atl AM and GL murein hydrolase activities in cell division, autolysis, and biofilm formation. PLoS ONE 2012, 7, e42244. [Google Scholar] [CrossRef]
  37. Heilmann, C.; Hussain, M.; Peters, G.; Götz, F. Evidence for autolysin-mediated primary attachment of Staphylococcus epidermidis to a polystyrene surface. Mol. Microbiol. 1997, 24, 1013–1024. [Google Scholar] [CrossRef]
  38. Gross, M.; Cramton, S.E.; Götz, F.; Peschel, A. Key role of teichoic acid net charge in Staphylococcus aureus colonization of artificial surfaces. Infect. Immun. 2001, 69, 3423–3426. [Google Scholar] [CrossRef]
  39. Holland, L.M.; Conlon, B.; O’Gara, J.P. Mutation of tagO reveals an essential role for wall teichoic acids in Staphylococcus epidermidis biofilm development. Microbiology 2011, 157 Pt 2, 408–418. [Google Scholar] [CrossRef]
  40. Franz, S.; Rammelt, S.; Scharnweber, D.; Simon, J.C. Immune responses to implants—A review of the implications for the design of immunomodulatory biomaterials. Biomaterials 2011, 32, 6692–6709. [Google Scholar] [CrossRef]
  41. Bacakova, L.; Filova, E.; Parizek, M.; Ruml, T.; Svorcik, V. Modulation of cell adhesion, proliferation and differentiation on materials designed for body implants. Biotechnol. Adv. 2011, 29, 739–767. [Google Scholar] [CrossRef] [PubMed]
  42. Foster, T.J. The MSCRAMM Family of Cell-Wall-Anchored Surface Proteins of Gram-Positive Cocci. Trends Microbiol. 2019, 27, 927–941. [Google Scholar] [CrossRef] [PubMed]
  43. Gill, S.R.; Fouts, D.E.; Archer, G.L.; Mongodin, E.F.; Deboy, R.T.; Ravel, J.; Paulsen, I.T.; Kolonay, J.F.; Brinkac, L.; Beanan, M.; et al. Insights on evolution of virulence and resistance from the complete genome analysis of an early methicillin-resistant Staphylococcus aureus strain and a biofilm-producing methicillin-resistant Staphylococcus epidermidis strain. J. Bacteriol. 2005, 187, 2426–2438. [Google Scholar] [CrossRef] [PubMed]
  44. Burke, F.M.; Di Poto, A.; Speziale, P.; Foster, T.J. The A domain of fibronectin-binding protein B of Staphylococcus aureus contains a novel fibronectin binding site. FEBS J. 2011, 278, 2359–2371. [Google Scholar] [CrossRef] [PubMed]
  45. Geoghegan, J.A.; Monk, I.R.; O’Gara, J.P.; Foster, T.J. Subdomains N2N3 of fibronectin binding protein A mediate Staphylococcus aureus biofilm formation and adherence to fibrinogen using distinct mechanisms. J. Bacteriol. 2013, 195, 2675–2683. [Google Scholar] [CrossRef] [PubMed]
  46. Greene, C.; McDevitt, D.; Francois, P.; Vaudaux, P.E.; Lew, D.P.; Foster, T.J. Adhesion properties of mutants of Staphylococcus aureus defective in fibronectin-binding proteins and studies on the expression of fnb genes. Mol. Microbiol. 1995, 17, 1143–1152. [Google Scholar] [CrossRef]
  47. Speziale, P.; Pietrocola, G. The Multivalent Role of Fibronectin-Binding Proteins A and B (FnBPA and FnBPB) of Staphylococcus aureus in Host Infections. Front. Microbiol. 2020, 11, 2054. [Google Scholar] [CrossRef]
  48. Keane, F.M.; Loughman, A.; Valtulina, V.; Brennan, M.; Speziale, P.; Foster, T.J. Fibrinogen and elastin bind to the same region within the A domain of fibronectin binding protein A, an MSCRAMM of Staphylococcus aureus. Mol. Microbiol. 2007, 63, 711–723. [Google Scholar] [CrossRef]
  49. Ní Eidhin, D.; Perkins, S.; Francois, P.; Vaudaux, P.; Höök, M.; Foster, T.J. Clumping factor B (ClfB), a new surface-located fibrinogen-binding adhesin of Staphylococcus aureus. Mol. Microbiol. 1998, 30, 245–257. [Google Scholar] [CrossRef]
  50. Ganesh, V.K.; Barbu, E.M.; Deivanayagam, C.C.; Le, B.; Anderson, A.S.; Matsuka, Y.V.; Lin, S.L.; Foster, T.J.; Narayana, S.V.; Höök, M. Structural and biochemical characterization of Staphylococcus aureus clumping factor B/ligand interactions. J. Biol. Chem. 2011, 286, 25963–25972. [Google Scholar] [CrossRef]
  51. Xiang, H.; Feng, Y.; Wang, J.; Liu, B.; Chen, Y.; Liu, L.; Deng, X.; Yang, M. Crystal structures reveal the multi-ligand binding mechanism of Staphylococcus aureus ClfB. PLoS Pathog. 2012, 8, e1002751. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, H.; Lv, J.; Qi, X.; Ding, Y.; Li, D.; Hu, L.; Wang, L.; Yu, F. The carriage of the serine-aspartate repeat protein-encoding sdr genes among Staphylococcus aureus lineages. Braz. J. Infect. Dis. Off. Publ. Braz. Soc. Infect. Dis. 2015, 19, 498–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Feuillie, C.; Formosa-Dague, C.; Hays, L.M.; Vervaeck, O.; Derclaye, S.; Brennan, M.P.; Foster, T.J.; Geoghegan, J.A.; Dufrêne, Y.F. Molecular interactions and inhibition of the staphylococcal biofilm-forming protein SdrC. Proc. Natl. Acad. Sci. USA 2017, 114, 3738–3743. [Google Scholar] [CrossRef] [PubMed]
  54. Askarian, F.; Uchiyama, S.; Valderrama, J.A.; Ajayi, C.; Sollid, J.U.E.; van Sorge, N.M.; Nizet, V.; van Strijp, J.A.G.; Johannessen, M. Serine-Aspartate Repeat Protein D Increases Staphylococcus aureus Virulence and Survival in Blood. Infect. Immun. 2017, 85, e00559-16. [Google Scholar] [CrossRef] [PubMed]
  55. Trivedi, S.; Uhlemann, A.C.; Herman-Bausier, P.; Sullivan, S.B.; Sowash, M.G.; Flores, E.Y.; Khan, S.D.; Dufrêne, Y.F.; Lowy, F.D. The Surface Protein SdrF Mediates Staphylococcus epidermidis Adherence to Keratin. J. Infect. Dis. 2017, 215, 1846–1854. [Google Scholar] [CrossRef]
  56. Sellman, B.R.; Timofeyeva, Y.; Nanra, J.; Scott, A.; Fulginiti, J.P.; Matsuka, Y.V.; Baker, S.M. Expression of Staphylococcus epidermidis SdrG increases following exposure to an in vivo environment. Infect. Immun. 2008, 76, 2950–2957. [Google Scholar] [CrossRef]
  57. Zong, Y.; Xu, Y.; Liang, X.; Keene, D.R.; Höök, A.; Gurusiddappa, S.; Höök, M.; Narayana, S.V. A ‘Collagen Hug’ model for Staphylococcus aureus CNA binding to collagen. EMBO J. 2005, 24, 4224–4236. [Google Scholar] [CrossRef]
  58. Valotteau, C.; Prystopiuk, V.; Pietrocola, G.; Rindi, S.; Peterle, D.; De Filippis, V.; Foster, T.J.; Speziale, P.; Dufrêne, Y.F. Single-Cell and Single-Molecule Analysis Unravels the Multifunctionality of the Staphylococcus aureus Collagen-Binding Protein Cna. ACS Nano 2017, 11, 2160–2170. [Google Scholar] [CrossRef]
  59. Madani, A.; Garakani, K.; Mofrad, M.R.K. Molecular mechanics of Staphylococcus aureus adhesin, CNA, and the inhibition of bacterial adhesion by stretching collagen. PLoS ONE 2017, 12, e0179601. [Google Scholar] [CrossRef]
  60. Mazmanian, S.K.; Liu, G.; Ton-That, H.; Schneewind, O. Staphylococcus aureus sortase, an enzyme that anchors surface proteins to the cell wall. Science 1999, 285, 760–763. [Google Scholar] [CrossRef]
  61. Mazmanian, S.K.; Ton-That, H.; Schneewind, O. Sortase-catalysed anchoring of surface proteins to the cell wall of Staphylococcus aureus. Mol. Microbiol. 2001, 40, 1049–1057. [Google Scholar] [CrossRef] [PubMed]
  62. Schneewind, O.; Missiakas, D. Sec-secretion and sortase-mediated anchoring of proteins in Gram-positive bacteria. Biochim. Et Biophys. Acta 2014, 1843, 1687–1697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Siegel, S.D.; Reardon, M.E.; Ton-That, H. Anchoring of LPXTG-Like Proteins to the Gram-Positive Cell Wall Envelope. Curr. Top. Microbiol. Immunol. 2017, 404, 159–175. [Google Scholar] [PubMed]
  64. Bowden, M.G.; Heuck, A.P.; Ponnuraj, K.; Kolosova, E.; Choe, D.; Gurusiddappa, S.; Narayana, S.V.L.; Johnson, A.E.; Höök, M. Evidence for the “dock, lock, and latch” ligand binding mechanism of the staphylococcal microbial surface component recognizing adhesive matrix molecules (MSCRAMM) SdrG. J. Biol. Chem. 2008, 283, 638–647. [Google Scholar] [CrossRef] [PubMed]
  65. Milles, L.F.; Schulten, K.; Gaub, H.E.; Bernardi, R.C. Molecular mechanism of extreme mechanostability in a pathogen adhesin. Science 2018, 359, 1527–1533. [Google Scholar] [CrossRef]
  66. Walsh, E.J.; O’Brien, L.M.; Liang, X.; Hook, M.; Foster, T.J. Clumping factor B, a fibrinogen-binding MSCRAMM (microbial surface components recognizing adhesive matrix molecules) adhesin of Staphylococcus aureus, also binds to the tail region of type I cytokeratin 10. J. Biol. Chem. 2004, 279, 50691–50699. [Google Scholar] [CrossRef]
  67. Zhang, Y.; Wu, M.; Hang, T.; Wang, C.; Yang, Y.; Pan, W.; Zang, J.; Zhang, M.; Zhang, X. Staphylococcus aureus SdrE captures complement factor H’s C-terminus via a novel ‘close, dock, lock and latch’ mechanism for complement evasion. Biochem. J. 2017, 474, 1619–1631. [Google Scholar] [CrossRef]
  68. Sharp, J.A.; Echague, C.G.; Hair, P.S.; Ward, M.D.; Nyalwidhe, J.O.; Geoghegan, J.A.; Foster, T.J.; Cunnion, K.M. Staphylococcus aureus surface protein SdrE binds complement regulator factor H as an immune evasion tactic. PLoS ONE 2012, 7, e38407. [Google Scholar] [CrossRef]
  69. Da Costa, T.M.; Viljoen, A.; Towell, A.M.; Dufrêne, Y.F.; Geoghegan, J.A. Fibronectin binding protein B binds to loricrin and promotes corneocyte adhesion by Staphylococcus aureus. Nat. Commun. 2022, 13, 2517. [Google Scholar] [CrossRef]
  70. Flemming, H.C.; Wingender, J.; Szewzyk, U.; Steinberg, P.; Rice, S.A.; Kjelleberg, S. Biofilms: An emergent form of bacterial life. Nat. Rev. Microbiol. 2016, 14, 563–575. [Google Scholar]
  71. Ramírez-Larrota, J.S.; Eckhard, U. An Introduction to Bacterial Biofilms and Their Proteases, and Their Roles in Host Infection and Immune Evasion. Biomolecules 2022, 12, 306. [Google Scholar] [CrossRef] [PubMed]
  72. Arciola, C.R.; Campoccia, D.; Ravaioli, S.; Montanaro, L. Polysaccharide intercellular adhesin in biofilm: Structural and regulatory aspects. Front. Cell. Infect. Microbiol. 2015, 5, 7. [Google Scholar] [CrossRef] [PubMed]
  73. Mack, D.; Fischer, W.; Krokotsch, A.; Leopold, K.; Hartmann, R.; Egge, H.; Laufs, R. The intercellular adhesin involved in biofilm accumulation of Staphylococcus epidermidis is a linear beta-1,6-linked glucosaminoglycan: Purification and structural analysis. J. Bacteriol. 1996, 178, 175–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Heilmann, C.; Schweitzer, O.; Gerke, C.; Vanittanakom, N.; Mack, D.; Götz, F. Molecular basis of intercellular adhesion in the biofilm-forming Staphylococcus epidermidis. Mol. Microbiol. 1996, 20, 1083–1091. [Google Scholar] [CrossRef]
  75. Cramton, S.E.; Gerke, C.; Schnell, N.F.; Nichols, W.W.; Götz, F. The intercellular adhesion (ica) locus is present in Staphylococcus aureus and is required for biofilm formation. Infect. Immun. 1999, 67, 5427–5433. [Google Scholar] [CrossRef]
  76. Gerke, C.; Kraft, A.; Süssmuth, R.; Schweitzer, O.; Götz, F. Characterization of the N-acetylglucosaminyltransferase activity involved in the biosynthesis of the Staphylococcus epidermidis polysaccharide intercellular adhesin. J. Biol. Chem. 1998, 273, 18586–18593. [Google Scholar] [CrossRef]
  77. Vuong, C.; Kocianova, S.; Voyich, J.M.; Yao, Y.; Fischer, E.R.; DeLeo, F.R.; Otto, M. A crucial role for exopolysaccharide modification in bacterial biofilm formation, immune evasion, and virulence. J. Biol. Chem. 2004, 279, 54881–54886. [Google Scholar] [CrossRef]
  78. Conlon, K.M.; Humphreys, H.; O’Gara, J.P. icaR encodes a transcriptional repressor involved in environmental regulation of ica operon expression and biofilm formation in Staphylococcus epidermidis. J. Bacteriol. 2002, 184, 4400–4408. [Google Scholar] [CrossRef]
  79. Jefferson, K.K.; Pier, D.B.; Goldmann, D.A.; Pier, G.B. The teicoplanin-associated locus regulator (TcaR) and the intercellular adhesin locus regulator (IcaR) are transcriptional inhibitors of the ica locus in Staphylococcus aureus. J. Bacteriol. 2004, 186, 2449–2456. [Google Scholar] [CrossRef]
  80. Hoang, T.M.; Zhou, C.; Lindgren, J.K.; Galac, M.R.; Corey, B.; Endres, J.E.; Olson, M.E.; Fey, P.D. Transcriptional Regulation of icaADBC by both IcaR and TcaR in Staphylococcus epidermidis. J. Bacteriol. 2019, 201, e00524-18. [Google Scholar] [CrossRef]
  81. Rohde, H.; Burandt, E.C.; Siemssen, N.; Frommelt, L.; Burdelski, C.; Wurster, S.; Scherpe, S.; Davies, A.P.; Harris, L.G.; Horstkotte, M.A.; et al. Polysaccharide intercellular adhesin or protein factors in biofilm accumulation of Staphylococcus epidermidis and Staphylococcus aureus isolated from prosthetic hip and knee joint infections. Biomaterials 2007, 28, 1711–1720. [Google Scholar] [CrossRef] [PubMed]
  82. O’Neill, E.; Pozzi, C.; Houston, P.; Smyth, D.; Humphreys, H.; Robinson, D.A.; O’Gara, J.P. Association between methicillin susceptibility and biofilm regulation in Staphylococcus aureus isolates from device-related infections. J. Clin. Microbiol. 2007, 45, 1379–1388. [Google Scholar] [CrossRef] [PubMed]
  83. Fitzpatrick, F.; Humphreys, H.; O’Gara, J.P. Evidence for icaADBC-independent biofilm development mechanism in methicillin-resistant Staphylococcus aureus clinical isolates. J. Clin. Microbiol. 2005, 43, 1973–1976. [Google Scholar] [CrossRef] [Green Version]
  84. O’Neill, E.; Pozzi, C.; Houston, P.; Humphreys, H.; Robinson, D.A.; Loughman, A.; Foster, T.J.; O’Gara, J.P. A novel Staphylococcus aureus biofilm phenotype mediated by the fibronectin-binding proteins, FnBPA and FnBPB. J. Bacteriol. 2008, 190, 3835–3850. [Google Scholar] [CrossRef] [PubMed]
  85. Barbu, E.M.; Mackenzie, C.; Foster, T.J.; Höök, M. SdrC induces staphylococcal biofilm formation through a homophilic interaction. Mol. Microbiol. 2014, 94, 172–185. [Google Scholar] [CrossRef]
  86. Formosa-Dague, C.; Speziale, P.; Foster, T.J.; Geoghegan, J.A.; Dufrêne, Y.F. Zinc-dependent mechanical properties of Staphylococcus aureus biofilm-forming surface protein SasG. Proc. Natl. Acad. Sci. USA 2016, 113, 410–415. [Google Scholar] [CrossRef]
  87. Conrady, D.G.; Brescia, C.C.; Horii, K.; Weiss, A.A.; Hassett, D.J.; Herr, A.B. A zinc-dependent adhesion module is responsible for intercellular adhesion in staphylococcal biofilms. Proc. Natl. Acad. Sci. USA 2008, 105, 19456–19461. [Google Scholar] [CrossRef]
  88. Yarawsky, A.E.; Johns, S.L.; Schuck, P.; Herr, A.B. The biofilm adhesion protein Aap from Staphylococcus epidermidis forms zinc-dependent amyloid fibers. J. Biol. Chem. 2020, 295, 4411–4427. [Google Scholar] [CrossRef]
  89. Conrady, D.G.; Wilson, J.J.; Herr, A.B. Structural basis for Zn2+-dependent intercellular adhesion in staphylococcal biofilms. Proc. Natl. Acad. Sci. USA 2013, 110, E202–E211. [Google Scholar] [CrossRef]
  90. Shelton, C.L.; Conrady, D.G.; Herr, A.B. Functional consequences of B-repeat sequence variation in the staphylococcal biofilm protein Aap: Deciphering the assembly code. Biochem. J. 2017, 474, 427–443. [Google Scholar] [CrossRef]
  91. Wang, R.; Khan, B.A.; Cheung, G.Y.; Bach, T.H.; Jameson-Lee, M.; Kong, K.F.; Queck, S.Y.; Otto, M. Staphylococcus epidermidis surfactant peptides promote biofilm maturation and dissemination of biofilm-associated infection in mice. J. Clin. Investig. 2011, 121, 238–248. [Google Scholar] [CrossRef] [PubMed]
  92. Schaeffer, C.R.; Hoang, T.N.; Sudbeck, C.M.; Alawi, M.; Tolo, I.E.; Robinson, D.A.; Horswill, A.R.; Rohde, H.; Fey, P.D. Versatility of Biofilm Matrix Molecules in Staphylococcus epidermidis Clinical Isolates and Importance of Polysaccharide Intercellular Adhesin Expression during High Shear Stress. mSphere 2016, 1, e00165-16. [Google Scholar] [CrossRef] [PubMed]
  93. Hennig, S.; Nyunt Wai, S.; Ziebuhr, W. Spontaneous switch to PIA-independent biofilm formation in an ica-positive Staphylococcus epidermidis isolate. Int. J. Med. Microbiol. IJMM 2007, 297, 117–122. [Google Scholar] [CrossRef] [PubMed]
  94. Böckelmann, U.; Janke, A.; Kuhn, R.; Neu, T.R.; Wecke, J.; Lawrence, J.R.; Szewzyk, U. Bacterial extracellular DNA forming a defined network-like structure. FEMS Microbiol. Lett. 2006, 262, 31–38. [Google Scholar] [CrossRef] [PubMed]
  95. Hall, C.W.; Mah, T.F. Molecular mechanisms of biofilm-based antibiotic resistance and tolerance in pathogenic bacteria. FEMS Microbiol. Rev. 2017, 41, 276–301. [Google Scholar] [CrossRef]
  96. Doroshenko, N.; Tseng, B.S.; Howlin, R.P.; Deacon, J.; Wharton, J.A.; Thurner, P.J.; Gilmore, B.F.; Parsek, M.R.; Stoodley, P. Extracellular DNA impedes the transport of vancomycin in Staphylococcus epidermidis biofilms preexposed to subinhibitory concentrations of vancomycin. Antimicrob. Agents Chemother. 2014, 58, 7273–7282. [Google Scholar] [CrossRef]
  97. Lewenza, S. Extracellular DNA-induced antimicrobial peptide resistance mechanisms in Pseudomonas aeruginosa. Front. Microbiol. 2013, 4, 21. [Google Scholar] [CrossRef]
  98. Ciszek-Lenda, M.; Strus, M.; Walczewska, M.; Majka, G.; Machul-Żwirbla, A.; Mikołajczyk, D.; Górska, S.; Gamian, A.; Chain, B.; Marcinkiewicz, J. Pseudomonas aeruginosa biofilm is a potent inducer of phagocyte hyperinflammation. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2019, 68, 397–413. [Google Scholar] [CrossRef]
  99. Thammavongsa, V.; Missiakas, D.M.; Schneewind, O. Staphylococcus aureus degrades neutrophil extracellular traps to promote immune cell death. Science 2013, 342, 863–866. [Google Scholar] [CrossRef]
  100. Finkel, S.E.; Kolter, R. DNA as a nutrient: Novel role for bacterial competence gene homologs. J. Bacteriol. 2001, 183, 6288–6293. [Google Scholar] [CrossRef]
  101. Wilking, J.N.; Zaburdaev, V.; De Volder, M.; Losick, R.; Brenner, M.P.; Weitz, D.A. Liquid transport facilitated by channels in Bacillus subtilis biofilms. Proc. Natl. Acad. Sci. USA 2013, 110, 848–852. [Google Scholar] [CrossRef]
  102. Otto, M. Staphylococcal infections: Mechanisms of biofilm maturation and detachment as critical determinants of pathogenicity. Annu. Rev. Med. 2013, 64, 175–188. [Google Scholar] [CrossRef]
  103. Le, K.Y.; Dastgheyb, S.; Ho, T.V.; Otto, M. Molecular determinants of staphylococcal biofilm dispersal and structuring. Front. Cell. Infect. Microbiol. 2014, 4, 167. [Google Scholar] [CrossRef]
  104. Queck, S.Y.; Khan, B.A.; Wang, R.; Bach, T.H.; Kretschmer, D.; Chen, L.; Kreiswirth, B.N.; Peschel, A.; Deleo, F.R.; Otto, M. Mobile genetic element-encoded cytolysin connects virulence to methicillin resistance in MRSA. PLoS Pathog. 2009, 5, e1000533. [Google Scholar] [CrossRef] [PubMed]
  105. Periasamy, S.; Chatterjee, S.S.; Cheung, G.Y.; Otto, M. Phenol-soluble modulins in staphylococci: What are they originally for? Commun. Integr. Biol. 2012, 5, 275–277. [Google Scholar] [CrossRef]
  106. Rumbaugh, K.P.; Sauer, K. Biofilm dispersion. Nat. Rev. Microbiol. 2020, 18, 571–586. [Google Scholar] [CrossRef]
  107. Heacock-Kang, Y.; Sun, Z.; Zarzycki-Siek, J.; McMillan, I.A.; Norris, M.H.; Bluhm, A.P.; Cabanas, D.; Fogen, D.; Vo, H.; Donachie, S.P.; et al. Spatial transcriptomes within the Pseudomonas aeruginosa biofilm architecture. Mol. Microbiol. 2017, 106, 976–985. [Google Scholar] [CrossRef] [PubMed]
  108. Chua, S.L.; Liu, Y.; Yam, J.K.; Chen, Y.; Vejborg, R.M.; Tan, B.G.; Kjelleberg, S.; Tolker-Nielsen, T.; Givskov, M.; Yang, L. Dispersed cells represent a distinct stage in the transition from bacterial biofilm to planktonic lifestyles. Nat. Commun. 2014, 5, 4462. [Google Scholar] [CrossRef] [PubMed]
  109. Krsmanovic, M.; Biswas, D.; Ali, H.; Kumar, A.; Ghosh, R.; Dickerson, A.K. Hydrodynamics and surface properties influence biofilm proliferation. Adv. Colloid Interface Sci. 2021, 288, 102336. [Google Scholar] [CrossRef]
  110. Fleming, D.; Rumbaugh, K.P. Approaches to Dispersing Medical Biofilms. Microorganisms 2017, 5, 15. [Google Scholar] [CrossRef]
  111. Martí, M.; Trotonda, M.P.; Tormo-Más, M.A.; Vergara-Irigaray, M.; Cheung, A.L.; Lasa, I.; Penadés, J.R. Extracellular proteases inhibit protein-dependent biofilm formation in Staphylococcus aureus. Microbes Infect. 2010, 12, 55–64. [Google Scholar] [CrossRef] [PubMed]
  112. McGavin, M.J.; Zahradka, C.; Rice, K.; Scott, J.E. Modification of the Staphylococcus aureus fibronectin binding phenotype by V8 protease. Infect. Immun. 1997, 65, 2621–2628. [Google Scholar] [CrossRef]
  113. Tang, J.; Zhou, R.; Shi, X.; Kang, M.; Wang, H.; Chen, H. Two thermostable nucleases coexisted in Staphylococcus aureus: Evidence from mutagenesis and in vitro expression. FEMS Microbiol. Lett. 2008, 284, 176–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Hu, Y.; Xie, Y.; Tang, J.; Shi, X. Comparative expression analysis of two thermostable nuclease genes in Staphylococcus aureus. Foodborne Pathog. Dis. 2012, 9, 265–271. [Google Scholar] [CrossRef] [PubMed]
  115. Fazekas, E.; Kandra, L.; Gyémánt, G. Model for β-1,6-N-acetylglucosamine oligomer hydrolysis catalysed by DispersinB, a biofilm degrading enzyme. Carbohydr. Res. 2012, 363, 7–13. [Google Scholar] [CrossRef] [PubMed]
  116. Itoh, Y.; Wang, X.; Hinnebusch, B.J.; Preston, J.F., 3rd; Romeo, T. Depolymerization of beta-1,6-N-acetyl-D-glucosamine disrupts the integrity of diverse bacterial biofilms. J. Bacteriol. 2005, 187, 382–387. [Google Scholar] [CrossRef] [PubMed]
  117. Fleming, D.; Chahin, L.; Rumbaugh, K. Glycoside Hydrolases Degrade Polymicrobial Bacterial Biofilms in Wounds. Antimicrob. Agents Chemother. 2017, 61, e01998-16. [Google Scholar] [CrossRef] [PubMed]
  118. Thomas, V.C.; Hancock, L.E. Suicide and fratricide in bacterial biofilms. Int. J. Artif. Organs 2009, 32, 537–544. [Google Scholar] [CrossRef]
  119. Davies, D. Understanding biofilm resistance to antibacterial agents. Nat. Rev. Drug Discov. 2003, 2, 114–122. [Google Scholar] [CrossRef]
  120. Urish, K.L.; DeMuth, P.W.; Kwan, B.W.; Craft, D.W.; Ma, D.; Haider, H.; Tuan, R.S.; Wood, T.K.; Davis, C.M., 3rd. Antibiotic-tolerant Staphylococcus aureus Biofilm Persists on Arthroplasty Materials. Clin. Orthop. Relat. Res. 2016, 474, 1649–1656. [Google Scholar] [CrossRef]
  121. Nguyen, D.; Joshi-Datar, A.; Lepine, F.; Bauerle, E.; Olakanmi, O.; Beer, K.; McKay, G.; Siehnel, R.; Schafhauser, J.; Wang, Y.; et al. Active starvation responses mediate antibiotic tolerance in biofilms and nutrient-limited bacteria. Science 2011, 334, 982–986. [Google Scholar] [CrossRef] [PubMed]
  122. Fisher, R.A.; Gollan, B.; Helaine, S. Persistent bacterial infections and persister cells. Nat. Rev. Microbiol. 2017, 15, 453–464. [Google Scholar] [CrossRef] [PubMed]
  123. Singh, R.; Sahore, S.; Kaur, P.; Rani, A.; Ray, P. Penetration barrier contributes to bacterial biofilm-associated resistance against only select antibiotics, and exhibits genus-, strain- and antibiotic-specific differences. Pathog. Dis. 2016, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Høiby, N.; Bjarnsholt, T.; Givskov, M.; Molin, S.; Ciofu, O. Antibiotic resistance of bacterial biofilms. Int. J. Antimicrob. Agents 2010, 35, 322–332. [Google Scholar] [CrossRef] [PubMed]
  125. Savage, V.J.; Chopra, I.; O’Neill, A.J. Staphylococcus aureus biofilms promote horizontal transfer of antibiotic resistance. Antimicrob. Agents Chemother. 2013, 57, 1968–1970. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Yan, J.; Bassler, B.L. Surviving as a Community: Antibiotic Tolerance and Persistence in Bacterial Biofilms. Cell Host Microbe 2019, 26, 15–21. [Google Scholar] [CrossRef]
  128. Dragoš, A.; Kovács Á, T. The Peculiar Functions of the Bacterial Extracellular Matrix. Trends Microbiol. 2017, 25, 257–266. [Google Scholar] [CrossRef]
  129. Amulic, B.; Cazalet, C.; Hayes, G.L.; Metzler, K.D.; Zychlinsky, A. Neutrophil function: From mechanisms to disease. Annu. Rev. Immunol. 2012, 30, 459–489. [Google Scholar] [CrossRef]
  130. Høiby, N.; Krogh Johansen, H.; Moser, C.; Song, Z.; Ciofu, O.; Kharazmi, A. Pseudomonas aeruginosa and the in vitro and in vivo biofilm mode of growth. Microbes Infect. 2001, 3, 23–35. [Google Scholar] [CrossRef]
  131. Leid, J.G.; Shirtliff, M.E.; Costerton, J.W.; Stoodley, P. Human leukocytes adhere to, penetrate, and respond to Staphylococcus aureus biofilms. Infect. Immun. 2002, 70, 6339–6345. [Google Scholar] [CrossRef] [PubMed]
  132. Ricciardi, B.F.; Muthukrishnan, G.; Masters, E.; Ninomiya, M.; Lee, C.C.; Schwarz, E.M. Staphylococcus aureus Evasion of Host Immunity in the Setting of Prosthetic Joint Infection: Biofilm and Beyond. Curr. Rev. Musculoskelet. Med. 2018, 11, 389–400. [Google Scholar] [CrossRef] [PubMed]
  133. Hanke, M.L.; Angle, A.; Kielian, T. MyD88-dependent signaling influences fibrosis and alternative macrophage activation during Staphylococcus aureus biofilm infection. PLoS ONE 2012, 7, e42476. [Google Scholar]
  134. Foster, T.J. The remarkably multifunctional fibronectin binding proteins of Staphylococcus aureus. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2016, 35, 1923–1931. [Google Scholar] [CrossRef]
  135. Becker, S.; Frankel, M.B.; Schneewind, O.; Missiakas, D. Release of protein A from the cell wall of Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 2014, 111, 1574–1579. [Google Scholar] [CrossRef]
  136. Woehl, J.L.; Stapels, D.A.C.; Garcia, B.L.; Ramyar, K.X.; Keightley, A.; Ruyken, M.; Syriga, M.; Sfyroera, G.; Weber, A.B.; Zolkiewski, M.; et al. The extracellular adherence protein from Staphylococcus aureus inhibits the classical and lectin pathways of complement by blocking formation of the C3 proconvertase. J. Immunol. 2014, 193, 6161–6171. [Google Scholar] [CrossRef]
  137. Spaan, A.N.; Surewaard, B.G.; Nijland, R.; van Strijp, J.A. Neutrophils versus Staphylococcus aureus: A biological tug of war. Annu. Rev. Microbiol. 2013, 67, 629–650. [Google Scholar] [CrossRef]
  138. Le, K.Y.; Park, M.D.; Otto, M. Immune Evasion Mechanisms of Staphylococcus epidermidis Biofilm Infection. Front. Microbiol. 2018, 9, 359. [Google Scholar] [CrossRef]
  139. Vuong, C.; Voyich, J.M.; Fischer, E.R.; Braughton, K.R.; Whitney, A.R.; DeLeo, F.R.; Otto, M. Polysaccharide intercellular adhesin (PIA) protects Staphylococcus epidermidis against major components of the human innate immune system. Cell. Microbiol. 2004, 6, 269–275. [Google Scholar] [CrossRef]
  140. Kristian, S.A.; Birkenstock, T.A.; Sauder, U.; Mack, D.; Götz, F.; Landmann, R. Biofilm formation induces C3a release and protects Staphylococcus epidermidis from IgG and complement deposition and from neutrophil-dependent killing. J. Infect. Dis. 2008, 197, 1028–1035. [Google Scholar] [CrossRef]
  141. Rupp, M.E.; Ulphani, J.S.; Fey, P.D.; Bartscht, K.; Mack, D. Characterization of the importance of polysaccharide intercellular adhesin/hemagglutinin of Staphylococcus epidermidis in the pathogenesis of biomaterial-based infection in a mouse foreign body infection model. Infect. Immun. 1999, 67, 2627–2632. [Google Scholar] [CrossRef] [PubMed]
  142. Omar, A.; Wright, J.B.; Schultz, G.; Burrell, R.; Nadworny, P. Microbial Biofilms and Chronic Wounds. Microorganisms 2017, 5, 9. [Google Scholar] [CrossRef] [PubMed]
  143. Vicentini, C.; Bordino, V.; Cornio, A.R.; Canta, I.; Marengo, N.; Zotti, C.M. Surgical site infection prevention through bundled interventions in hip replacement surgery: A systematic review. Int. J. Surg. 2021, 95, 106149. [Google Scholar] [CrossRef] [PubMed]
  144. Campoccia, D.; Montanaro, L.; Arciola, C.R. A review of the biomaterials technologies for infection-resistant surfaces. Biomaterials 2013, 34, 8533–8554. [Google Scholar] [CrossRef] [PubMed]
  145. Alt, V. Antimicrobial coated implants in trauma and orthopaedics-a clinical review and risk-benefit analysis. Injury 2017, 48, 599–607. [Google Scholar] [CrossRef]
  146. Vallet-Regí, M.; Lozano, D.; González, B.; Izquierdo-Barba, I. Biomaterials against Bone Infection. Adv. Healthc. Mater. 2020, 9, e2000310. [Google Scholar] [CrossRef]
  147. Zimmerli, W.; Sendi, P. Orthopaedic biofilm infections. APMIS Acta Pathol. Microbiol. Et Immunol. Scand. 2017, 125, 353–364. [Google Scholar] [CrossRef]
  148. Coiffier, G.; Albert, J.D.; Arvieux, C.; Guggenbuhl, P. Optimizing combination rifampin therapy for staphylococcal osteoarticular infections. Jt. Bone Spine 2013, 80, 11–17. [Google Scholar] [CrossRef]
  149. Beldman, M.; Löwik, C.; Soriano, A.; Albiach, L.; Zijlstra, W.P.; Knobben, B.A.S.; Jutte, P.; Sousa, R.; Carvalho, A.; Goswami, K.; et al. If, When, and How to Use Rifampin in Acute Staphylococcal Periprosthetic Joint Infections, a Multicentre Observational Study. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2021, 73, 1634–1641. [Google Scholar] [CrossRef]
  150. Hsieh, P.H.; Lee, M.S.; Hsu, K.Y.; Chang, Y.H.; Shih, H.N.; Ueng, S.W. Gram-negative prosthetic joint infections: Risk factors and outcome of treatment. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2009, 49, 1036–1043. [Google Scholar] [CrossRef]
  151. Łusiak-Szelachowska, M.; Międzybrodzki, R.; Drulis-Kawa, Z.; Cater, K.; Knežević, P.; Winogradow, C.; Amaro, K.; Jończyk-Matysiak, E.; Weber-Dąbrowska, B.; Rękas, J.; et al. Bacteriophages and antibiotic interactions in clinical practice: What we have learned so far. J. Biomed. Sci. 2022, 29, 23. [Google Scholar] [CrossRef] [PubMed]
  152. Kaur, S.; Harjai, K.; Chhibber, S. In Vivo Assessment of Phage and Linezolid Based Implant Coatings for Treatment of Methicillin Resistant S. aureus (MRSA) Mediated Orthopaedic Device Related Infections. PLoS ONE 2016, 11, e0157626. [Google Scholar] [CrossRef] [PubMed]
  153. Torres-Barceló, C.; Hochberg, M.E. Evolutionary Rationale for Phages as Complements of Antibiotics. Trends Microbiol. 2016, 24, 249–256. [Google Scholar] [CrossRef] [PubMed]
  154. Ferry, T.; Kolenda, C.; Batailler, C.; Gustave, C.A.; Lustig, S.; Malatray, M.; Fevre, C.; Josse, J.; Petitjean, C.; Chidiac, C.; et al. Phage Therapy as Adjuvant to Conservative Surgery and Antibiotics to Salvage Patients With Relapsing S. aureus Prosthetic Knee Infection. Front. Med. 2020, 7, 570572. [Google Scholar] [CrossRef] [PubMed]
  155. Ferry, T.; Kolenda, C.; Batailler, C.; Gaillard, R.; Gustave, C.A.; Lustig, S.; Fevre, C.; Petitjean, C.; Leboucher, G.; Laurent, F. Case Report: Arthroscopic “Debridement Antibiotics and Implant Retention” with Local Injection of Personalized Phage Therapy to Salvage a Relapsing Pseudomonas aeruginosa Prosthetic Knee Infection. Front. Med. 2021, 8, 569159. [Google Scholar] [CrossRef] [PubMed]
  156. Wójcik-Bojek, U.; Różalska, B.; Sadowska, B. Staphylococcus aureus—A Known Opponent against Host Defense Mechanisms and Vaccine Development-Do We Still Have a Chance to Win? Int. J. Mol. Sci. 2022, 23, 948. [Google Scholar] [CrossRef]
  157. Søe, N.H.; Jensen, N.V.; Jensen, A.L.; Koch, J.; Poulsen, S.S.; Pier, G.B.; Johansen, H.K. Active and Passive Immunization Against Staphylococcus aureus Periprosthetic Osteomyelitis in Rats. Vivo 2017, 31, 45–50. [Google Scholar] [CrossRef]
  158. Hofmans, D.; Khodaparast, L.; Khodaparast, L.; Vanstreels, E.; Shahrooei, M.; Van Eldere, J.; Van Mellaert, L. Ses proteins as possible targets for vaccine development against Staphylococcus epidermidis infections. J. Infect. 2018, 77, 119–130. [Google Scholar] [CrossRef]
  159. Mubeen, B.; Ansar, A.N.; Rasool, R.; Ullah, I.; Imam, S.S.; Alshehri, S.; Ghoneim, M.M.; Alzarea, S.I.; Nadeem, M.S.; Kazmi, I. Nanotechnology as a Novel Approach in Combating Microbes Providing an Alternative to Antibiotics. Antibiotics 2021, 10, 1473. [Google Scholar] [CrossRef]
  160. Makabenta, J.M.V.; Nabawy, A.; Li, C.H.; Schmidt-Malan, S.; Patel, R.; Rotello, V.M. Nanomaterial-based therapeutics for antibiotic-resistant bacterial infections. Nat. Rev. Microbiol. 2021, 19, 23–36. [Google Scholar] [CrossRef]
  161. Huo, J.; Jia, Q.; Huang, H.; Zhang, J.; Li, P.; Dong, X.; Huang, W. Emerging photothermal-derived multimodal synergistic therapy in combating bacterial infections. Chem. Soc. Rev. 2021, 50, 8762–8789. [Google Scholar] [CrossRef] [PubMed]
  162. Xu, M.; Li, L.; Hu, Q. The recent progress in photothermal-triggered bacterial eradication. Biomater. Sci. 2021, 9, 1995–2008. [Google Scholar] [CrossRef] [PubMed]
  163. Warrier, A.; Mazumder, N.; Prabhu, S.; Satyamoorthy, K.; Murali, T.S. Photodynamic therapy to control microbial biofilms. Photodiagn. Photodyn. Ther. 2021, 33, 102090. [Google Scholar] [CrossRef] [PubMed]
  164. Tan, L.; Li, J.; Liu, X.; Cui, Z.; Yang, X.; Zhu, S.; Li, Z.; Yuan, X.; Zheng, Y.; Yeung, K.W.K.; et al. Rapid Biofilm Eradication on Bone Implants Using Red Phosphorus and Near-Infrared Light. Adv. Mater. 2018, 30, e1801808. [Google Scholar] [CrossRef] [PubMed]
  165. Liu, Y.; Shi, L.; Su, L.; van der Mei, H.C.; Jutte, P.C.; Ren, Y.; Busscher, H.J. Nanotechnology-based antimicrobials and delivery systems for biofilm-infection control. Chem. Soc. Rev. 2019, 48, 428–446. [Google Scholar] [CrossRef]
  166. Oberdörster, G. Safety assessment for nanotechnology and nanomedicine: Concepts of nanotoxicology. J. Intern. Med. 2010, 267, 89–105. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lu, Y.; Cai, W.-j.; Ren, Z.; Han, P. The Role of Staphylococcal Biofilm on the Surface of Implants in Orthopedic Infection. Microorganisms 2022, 10, 1909. https://doi.org/10.3390/microorganisms10101909

AMA Style

Lu Y, Cai W-j, Ren Z, Han P. The Role of Staphylococcal Biofilm on the Surface of Implants in Orthopedic Infection. Microorganisms. 2022; 10(10):1909. https://doi.org/10.3390/microorganisms10101909

Chicago/Turabian Style

Lu, Yu, Wei-jie Cai, Zun Ren, and Pei Han. 2022. "The Role of Staphylococcal Biofilm on the Surface of Implants in Orthopedic Infection" Microorganisms 10, no. 10: 1909. https://doi.org/10.3390/microorganisms10101909

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