Next Article in Journal
Biopolymer Green Lubricant for Sustainable Manufacturing
Next Article in Special Issue
Antimicrobial Approaches for Textiles: From Research to Market
Previous Article in Journal
Parametric Analysis to Study the Influence of Aerogel-Based Renders’ Components on Thermal and Mechanical Performance
Previous Article in Special Issue
Deposition of Antimicrobial Copper-Rich Coatings on Polymers by Atmospheric Pressure Jet Plasmas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Silver Nanocoating Technology in the Prevention of Prosthetic Joint Infection

1
Department of Orthopaedics, Faculty of Medicine and Dentistry, Palacký University Olomouc, I. P. Pavlova 6, Olomouc 779 00, Czech Republic
2
Regional Centre of Advanced Technologies and Materials, Palacký University Olomouc, Šlechtitelů 27, Olomouc 783 71, Czech Republic
3
Department of Immunology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, Olomouc 779 00, Czech Republic
*
Author to whom correspondence should be addressed.
Materials 2016, 9(5), 337; https://doi.org/10.3390/ma9050337
Submission received: 31 March 2016 / Revised: 26 April 2016 / Accepted: 27 April 2016 / Published: 5 May 2016
(This article belongs to the Special Issue Self-Cleaning and Antimicrobial Surfaces)

Abstract

:
Prosthetic joint infection (PJI) is a feared complication of total joint arthroplasty associated with increased morbidity and mortality. There is a growing body of evidence that bacterial colonization and biofilm formation are critical pathogenic events in PJI. Thus, the choice of biomaterials for implanted prostheses and their surface modifications may significantly influence the development of PJI. Currently, silver nanoparticle (AgNP) technology is receiving much interest in the field of orthopaedics for its antimicrobial properties and a strong anti-biofilm potential. The great advantage of AgNP surface modification is a minimal release of active substances into the surrounding tissue and a long period of effectiveness. As a result, a controlled release of AgNPs could ensure antibacterial protection throughout the life of the implant. Moreover, the antibacterial effect of AgNPs may be strengthened in combination with conventional antibiotics and other antimicrobial agents. Here, our main attention is devoted to general guidelines for the design of antibacterial biomaterials protected by AgNPs, its benefits, side effects and future perspectives in PJI prevention.

1. Introduction

Prosthetic joint infection (PJI) is a feared complication of modern orthopaedic surgery that substantially increases morbidity and even mortality following total joint arthroplasty (TJA) [1,2]. Generally, PJI leads to implant removal and long-term antibiotic therapy with a permanent, increased risk for PJI development in affected patients [3].
Current estimates suggest that up to 3% of primary hip and knee arthroplasties [4], up to 15.4% of revision hip and 25% of knee arthroplasties are complicated by PJI respectively [5]. According to some authors, these numbers are not only underestimated but they are also on the rise [6]. The annual cost of infected revisions in hospitals of the United States of America (USA) could increase from $566 million in 2009 to $1.62 billion by 2020 [7]. As a result, therapy of PJI continues to be associated with enormous costs.
The first postoperative months are the most typical period of PJI manifestation [8] with the incidence rate of late PJI in hip and knee arthroplasty at about 0.07% per prosthesis-year and a higher risk in knee arthroplasties when compared to hip [9].
The leading causes of PJIs are S. aureus and coagulase-negative staphylocci followed by streptococci and enterococci (all of these account for approximately 10% of PJI cases) [10,11]. Importantly, the prevalence of methicillin-resistant S. aureus (MRSA) in PJI is increasing, especially in the USA [12]. In addition, polymicrobial infections can occur in up to 15% of cases [13] despite the fact that some authors reported a substantial increase in the yearly occurrence of polymicrobial infections over the period of six years (2004 to 2010) with a greater increase in the proportion of gram-negative bacteria during the same period [14].

2. Pathogenesis of PJI

The distribution of PJI in time strongly points to the causative link towards surgery and the early postoperative period. A basic prerequisite for PJI development is the size of the bacterial load influencing the operating wound, immune response and the implant. The last two decades were under strong dominance of Gristina’s concept of “race for the surface” [15]. Accordingly, host and bacterial cells compete in determining the ultimate fate of the implant, when host cells colonize the implant surface first, the probability of attachment of bacterial cells is very low and vice versa. However, Gristina’s model is not able to predict PJI in “less clear” situations when the host cell coverage of an implant surface is incomplete and thus offering some places for bacteria adhesion. In addition, some prosthetic surfaces, either articulating or non-articulating, preclude host cell adhesion and development of a protective host film. This model can also be criticized for static conditions because fluid waves occurring many times per hour are typical for TJA. Finally, immune and host tissue responses contribute to the protection of an implant surface to a greater extent than only in terms of simple mechanistic competition for an implant surface. Despite the fact that not all the critical pathogen and host steps/factors have been elucidated to date [16], for instance an infection dose no doubt plays an important role. A higher bacterial load of S. aureus could alter the host immune response and accelerate biofilm formation [17] while a low level of “appropriate” bacterial contamination might even serve as a potent immunomodulatory factor preventing the development of PJI (“implant infection paradox”) [18]. Some evidence also suggests the role of genetic susceptibility [19,20]. Taken together, instead of Gristina’s metaphor, a specific local immunologic and tissue constellation type of pathogen as well as bacterial load interplay with each other and influence the implant-tissue interactions, either towards non-infective or infective statuses.
The most destabilizing factor is the basic yet highly successful survival strategy of bacteria in general: their ability to adhere and survive on virtually all natural and synthetic surfaces (Figure 1) [21,22]. The bacterial cell membrane contains various types of adhesins for a wide range of biomaterial surface receptor sites, members of the family of Microbial Surface Components Recognizing Adhesive Matrix Molecules (MSCRAMMs) [23]. Environmental and surface characteristics of a biomaterial such as surface roughness, hydrophobicity and electrostatic charge play only conditional roles [24]. A reservoir of receptors for bacterial adhesive ligands mediating adhesion of free-floating bacteria to the surface of the biomaterial, offers a conditional protein film covering the implant immediately after its placement into the host body [25]. The spectrum of binding molecules depends at least partly on the particular type of biomaterials attracting an exact set of host proteins and lipids [26,27,28].
Conceptually, the process of bacterial adhesion can be divided into two basic phases: reversible, and irreversible [29,30]. The former is mechanically and biologically less stable than the latter. The explanation lies partly in the origin of nonspecific interactions between the implant surface characteristics and bacterial surface adhesins, followed by molecular and cellular interactions closely associated with expression of biofilm specific gene clusters in reversibly attached bacteria [31]. At least four distinct classes of surface proteins have been identified to participate on firm adhesion of S. aureus micro-colonies to a biomaterial and to each other [32]. The adhesion phase is followed by gene expression for secretion of protective slime. This process makes bacteria extremely resistant to both the host immune system and antibiotic diffusion [30,33]. The transition between the reversible and irreversible phases of biofilm formation coupled with a phenotypical change, is the last window of opportunity for clinically reasonable preventative measures. Other parameters of biofilm formation are described in detail elsewhere [34], as well as the ability of bacteria to combine different pathogenic strategies [35].
In the host site, the details of tissue integration of a biomaterial are still poorly understood [36,37]. It is believed that immune as well as tissue resident cells recognize an implant surface and orchestrate the processes, leading to periprosthetic bone/soft-tissue regeneration and remodeling, preventing the development of biofilm in the majority of patients [38,39]. However, neither osseointegration nor fibrous tissue encapsulation of large non-fixation parts of an implant can eliminate long-term survivorship of bacterial micro-colonies. Moreover, the peri-implant fibrous barrier impedes contact between the host immunity sentinel cells and bacterial molecules. This interaction is critical for host immune responses dependent on recognition of bacterial pattern-recognition receptors (PRRs; also microbe associated molecular patterns = MAMPs). Importantly, it has been demonstrated that implantation of a medical device impairs the innate local host response and may facilitate the development of PJI [40,41].
As the majority of operating rooms are contaminated within the first few hours of service [42,43], most surgeries are not performed in a bacterial-free environment. All patients are exposed to the same environment within a particular operating room. The question therefore arises as to why some patients go on to have infections and others do not. There is a growing body of evidence that PJI results from a relatively unclear and perhaps unique combination of environmental and genetic factors. The environmental ones could be linked to immune and non-immune factors affecting host response to bacterial load (age, gender, malnutrition, weight, diabetes mellitus, smoking etc.); the factors related specifically to implant facilitating for instance, adhesion of bacteria and those related to the surgeon and surgery (operating skills, operating room parameters, surgical time etc.). The host genetics strongly influences an individual’s susceptibility to infectious diseases and there is some evidence available for genetic susceptibility to PJI [19].
As a result, there is a strong need for intrinsic implant surface antibacterial functionality that can protect the implant surface from a perioperative attack of pathogenic bacteria as well as help to overcome implant-induced defects in the local immune response.

3. Rationale and Basic Concepts of PJI Prevention

Strategies relying on a decreased bacterial load and creating a bacteria-free environment around an implant during the perioperative period are widely implemented in clinical practice [44,45]. There is sufficient evidence supporting systemic [46] and in some cases local antibiotic prophylaxis [47]. However, the optimal protocol for individual clinical situations is not known yet. At present, antibiotics are administered to all the patients undergoing TJA regardless of the individual risk for PJI development, at least in terms of the beginning, the type of antibiotic and the duration of antibiotic prophylaxis. With regard to the latter, the 24 h regime does not cover the time needed for early wound stabilization, or the period of time the suction drain is in contact with joint and deep tissues. In addition, the increasing occurrence of antibiotic resistance has been recognized to be a global problem. There is also some evidence for selecting antibiotic-resistant staphylococci in relation to wide-range antibiotic prophylaxis [48].
Attempts at formulating evidence-based standards for good clinical and logistic practice in orthopaedic operating rooms have been made [45,49]. There is a growing pressure on surgeons to improve their surgical skills in order to minimize the surgery-related factors. Educational programs aimed at educating/training orthopaedic surgeons (and all staff) in perioperative strategies of PJI prevention are under way [50].
Finally, strategies based on identification of risk patients and optimization of their conditions to decrease the probability of PJI development have been proposed. Even though modifiable PJI risk factors have been identified and well-described [50,51], it is often not possible to avoid operating “risk” patients who are not “optimized”. For instance, significant obesity precluded the indication for total hip or knee arthroplasty in some countries several years ago. However, it is unethical to reject surgery in these patients today, despite the fact that they have an increased risk for PJI [52]. Research testing is assessing whether the risk for PJI could be decreased after preoperative immunization of the patients at-risk, by a vaccine that targets either the most frequent pathogen as S. aureus, or the key molecules of bacterial adhesion and biofilm formation [53].
Taking into account the weaknesses associated with all the current preventative strategies, leaders in the field recommend a multistep preventative concept (Figure 2) covering simultaneously all the well-known targets, including the “anti-infective implant” [54,55,56].

4. Indications for Implants with Antibacterial Surface Treatment

In accordance with the evidence-based medical rules, it would be relevant to calculate the number of PJIs prevented, by usage of implants with an antibacterial surface. Theoretically, all the patients undergoing TJA are at risk for PJI. Revision cases carry an increased risk, partly due to the prolonged operating time during revision surgeries, in conjunction with a suboptimal local tissue environment [57]. Moreover, there is some evidence that the risk of PJI across the board in orthopaedic surgery, is on the rise [6]. As a result, one could argue that all patients should benefit from implants coated with a proven anti-infective surface. On the other hand, the risk for PJI is not homogenously distributed among arthroplasty patients [50]. Therefore, it might be convincing to implant “biofilm resistant” prostheses only in patients at an increased risk of PJI [51,58]. However, a validated tool for screening patients for an increased risk of PJI does not currently exist. Taken together, the preventative strategy involving all the patients undergoing primary and revision TJA seems to be more justifiable than a more restrictive approach targeting the high-risk patients. However, prior to implementation of such devices, it is necessary to demonstrate the significant reduction of PJI in a well-done, population-based, cost-benefit analysis [38]. An important consideration in designing implants with antibacterial coating relates to the characterization of reasonable and justifiable costs.

5. Recommendations for Construction of Implants with Anti-Infective Surfaces

A wide spectrum of substances and technological approaches has been proposed and tested for antibacterial features in orthopaedics (Table 1). In order to fully discuss and evaluate surface treatment technologies it is essential to review the strict criteria related generally to the process of innovation in this field. The requested parameters are as follows:
  • biocompatibility (the ability of a material to work efficiently with an appropriate host response in specific applications) [59];
  • strong evidence of anti-infective efficiency (the anti-bacterial efficiency should be demonstrated in vitro, in vivo and also in an appropriate model of PJI) [60,61,62];
  • fixation properties cannot be compromised (the antibacterial coating must not compromise long-term stable implant osseointegration or cement fixation);
  • durability of the anti-infective effect (while clear recommendations are lacking the epidemiological viewpoint suggests that at least two years would be appreciated) [63,64];
  • mechanical characteristics of the antibacterial coating (resistance to mechanical stresses and strains either during surgery or postoperatively) [65].
Recently, a new classification of the implant-related antibacterial strategies has been proposed distinguishing:
  • passive surface finishing/modification (PSM);
  • active surface finishing/modification (ASM);
  • perioperative antibacterial local carriers or coatings (LCCs) [56].
If the active substance is released from the surface of the implant, over time it may lead to its exhaustion and thereby, a loss of efficiency. It is therefore extremely relevant to design surface modifications with minimal but effective release of active substances into the surrounding tissue, thereby achieving a long, or even indefinite period of effectiveness. This approach may ensure antibacterial protection throughout the life of the implant. A specific set of problems are related to fluid dynamics and adhesion of host proteins, lipids, cells to “active” implant surfaces, limiting their antibacterial efficacy.

6. General Remarks on Prosthetic Implant Surface Modifications

Polyethylene, a modern generation of zirconia treated ceramic, stainless steel, cobalt-chrome and titanium alloys are the most commonly used materials in TJA implants. In TJA, each material/surface modification has its specific role (e.g., an articulating or a fixation surface) and can occupy a different place in the bulk implant (Figure 3). These parameters together, define the requirements for particular surface modifications in specific implant sites.
A number of principles from basic research have been proposed for translation into technologies potentially suitable for antibacterial treatment of orthopaedic implants [149]. It is easy to distinguish between technologies offering anti-adhesive properties, those working as antimicrobial agents and those combining the above-mentioned approaches. Anti-infective surfaces can be classified as contact killing” and antimicrobial agent eluting respectively [150].
Antibacterial surface technologies can employ metals (silver, zinc, copper, zirconium etc.), non-metal elements (e.g., selenium), organic substances (antibiotics, anti-infective peptides, chitosan, other substances) and their combinations. Antibacterial activity of the majority of metal coatings is closely linked to the ionic or nano-form, rather than to the bulk material [151]. Nanostructured surfaces and coatings (either of inorganic or organic origin) are therefore of great interest. Consequently, the nanoscale surface patterning methods have been applied to fabricate different nanopatterns (e.g., ordered stripes, pits, pillars or squares). Several studies have demonstrated that nanopatterning in conjunction with other surface treatment can inhibit bacterial adhesion [152,153].
In terms of functionality, one may divide surfaces as mono-functional and multi-functional. The latter are expected to target multiple biological tasks simultaneously (Figure 4), orchestrating early/long-term tissue adaptation to an implant, facilitating osseointegration and regulating the anti-infective immune response, all in addition to the “intrinsic” antibacterial surface effect [154]. Smart surface could be a completely different methodology designed to be a self-responsive multitask micro-machine that releases antimicrobial (and other) substances, after stimulation by microbial (or other) signals [155].

7. Why Silver Nanoparticle Technologies on the Implant Surfaces?

Currently, AgNP technology is receiving much interest in its use on implant surfaces, mainly for its antimicrobial properties and strong anti-biofilm potential together with relatively low cytotoxicity to mammalian cells. AgNPs effectively inhibit the growth of bacteria including highly resistant strains at very low concentrations in units of mg/L [92,156,157,158,159,160,161,162], whereas such concentrations do not exhibit an acute cytotoxic effect, which was proved at the concentrations higher than 20 mg/L [163,164,165].
Moreover, in the case of AgNPs, bacterial resistance has not been reported up to now, despite the fact that resistance to ionic silver has been observed. The multilevel antimicrobial (broad target attack) mode of AgNPs ensures that resistance cannot be easily acquired by single point mutations in contrast to antibiotics. Having a very low risk of development of bacterial resistance it is therefore relevant to know the antibacterial effects of AgNPs. This is an extremely valuable effect especially today, when we are facing growing antibacterial resistance observed in antibiotics and other antibacterial substances. Some experts even refer to the current state as to a “worldwide calamity” or “antibiotic resistance crisis”. Therefore, a joint multilevel and global interdisciplinary action including substituting antibiotics by non-antibiotic approaches could decrease the range and rate of bacterial resistance.
Moreover, AgNPs have a strong anti-biofilm potential [162,166,167,168,169,170,171,172,173,174,175,176,177,178,179]. Therefore, these are potentially very attractive for surface protection of orthopaedic implants since PJI is biofilm driven in the majority of clinical cases. As a result, silver is the most prevalent metal used in biomedical applications for antibacterial coating of prosthetic metal implants [180,181,182,183,184,185,186,187,188,189,190]. Both uncoated and coated AgNPs on various surfaces, such as titanium surfaces or catheter surfaces, thoroughly inhibit both planktonic and biofilm-forming bacteria [94,167,191,192]. Saleh et al. reported that biofilm and planktonic E. coli and P. aeruginosa cells showed very similar tolerance to AgNPs upon exposure [191]. Agarwala et al. reported high antimicrobial activity on catheters loaded with AgNPs towards planktonic as well as biofilm-forming cells [167]. Similarly, Zhong et al. and Harraser et al. reported that AgNP-loaded titanium can kill planktonic and adherent bacteria during 1, 4 and 12 days with similar effectiveness [94,192]. On the other side, several studies showed that biofilms decreased susceptibility to AgNPs compared to planktonic cells [193,194]. Choi et al. found that biofilms were four times less susceptible to AgNP exposure than planktonic cells were [194]. Starch-coated NPs reduced P. aeruginosa and S. aureus biofilm growth but completely inactivated planktonic cells at the same AgNP concentrations [195]. It is known that both planktonic and biofilm-forming bacteria produce extracellular polymeric substances (EPS), which has been proved to lower the diffusion rate of NPs [196]. EPS production is much greater in biofilms compared to planktonic bacteria and therefore may provide some protection to biofilm-forming cells from NPs.

8. Synthesis of Silver Nanoparticles on the Implant Surface

There are several approaches to synthesize the AgNPs on the implant surface. In the study [87], TiO2 (titanium dioxide) nanotubes (NT) on a titanium (Ti) surface were prepared by anodization of the Ti surface and consequently AgNTs were generated on the NT surface by ultraviolet reduction of silver ions. The TiO2-NTs loaded with Ag (silver) exhibited a strong antibacterial activity against methicillin-resistant S. aureus (MRSA, ATCC43300) in vitro for 30 days.
The cathodic arc silver plasma immersion ion implantation process can serve as another method of preparation and immobilization of AgNPs on a Ti surface. The immobilized AgNPs offered good defense against multiple cycles of bacteria (S. epidermidis) attacks in vitro and the mechanism was independent of silver release [197].
Also Pulse DC magnetron sputtering can be utilized for a generation of AgNPs on a Ti surface, where nanostructured Ti-Ag coatings with different Ag contents (1.2% to 21.6%) are able to kill S. aureus effectively during the first few days and remain moderately antibacterial after immersion for 75 days. Compared to pure Ti, the Ti-Ag coatings show good cytocompatibility as indicated by good osteoblast adhesion, proliferation, intracellular total protein synthesis and alkaline phosphatase activity [198].
AgNPs with the size of 50 nm can also be incorporated into a dopamine-modified alginate/chitosan (DAL/CHI) polyelectrolyte multilayer to modify titanium alloy surfaces. The polyelectrolyte multilayer coating enhanced wet ability of titanium alloy and promoted the fibroblast proliferation significantly, which could be attributed to the excellent biocompatibility of DAL/CHI [199]. Despite the slight fall of L929 cell activity after AgNP incorporation, AgNP-DAL/CHI multilayer inhibited the growth of both E. coli and S. aureus [199].
Hexagonal closed-packed TiO2 nanotubes with the diameter of 30–100 nm were prepared by anodization of a Ti foil, where the size of nanotubes was dependent on the parameters of anodization [200]. The size and shape of the generated AgNPs (12–40 nm) on TiO2 nanotubes by UV (ultraviolet) irradiation depends mainly on the size of TiO2 nanotubes and silver ion concentration. The highest antibacterial activity was obtained for TiO2 nanotubes with the opening diameter of about 100 nm and AgNPs with an average size of 20 nm, whereas good cell viability using osteoblast MG63 cells was remained [200]. The Ti/TiO2 nanotubes/AgNPs composites can also be prepared with the assistance of quaternary ammonium salt (QAS, 3-trimethoxysily-propyldimethyloctadecyl-ammonium chloride). The Ag nanoparticle loaded and QAS coated TiO2 nanotube substrates demonstrated long-term antibacterial effect and displayed good biocompatibility [201].

9. Antibacterial Effect of Silver Nanoparticles

The effects of silver, either as a metal (AgNPs) or in compounds is known to be non-specific, influencing many bacterial structures and metabolic processes at the same time (Figure 5). Among these are the following: inactivation of bacterial enzymes [202,203], disruption of bacterial metabolic processes [204,205,206] and the bacterial cell wall, accumulation in the cytoplasmic membrane and increase of its permeability [167,203,207], collapse the plasma membrane potential [206], interaction with DNA (deoxyribonucleic acid) [202] and generation of reactive oxygen species [208,209,210], which damage biomacromolecules [211]. Thanks to their multi-level mode of action, AgNPs destroy or inhibit the growth of pathogenic microorganisms including highly resistant bacterial strains at low concentrations from a few to several tens of mg/L [92,160,162,167,175,207]. Importantly, no relevant data describing bacterial resistance to AgNPs or inactivation of antibacterial action of AgNPs (nanoparticles) have been reported yet. Bacterial resistance to silver is driven only with the ionic form of silver and apart from others, was deeply researched by Silver et al. [212,213]. Bacterial resistance to ionic silver originated from clinical environments [214] and also from naturally occurring strains [215]. Besides reduction of Ag+ to a less toxic oxidation state, the probable Ag+ resistance mechanism involves an active efflux from the cell, by either P-type ATPases (adenosine triphosphatase) or chemiosmotic Ag+/H+ antiporters [216,217,218].
In recent years, a synergistic effect between AgNPs and various antibacterial agents has been investigated. Potara et al. studied antimicrobial activity of chitosan-coated AgNPs against two strains of S. aureus [219] and revealed that minimum inhibitory concentrations (MICs) of the composites were ten times lower than those of AgNPs and chitosan alone respectively. Another capping agent, myramistin increased activity of AgNPs against E. coli up to 20 times [220]. Combined treatments with a lactoferrin/xylitol hydrogel and silver-based wound dressings acted synergistically against the forming of biofilms of clinical wound isolates of methicillin-resistant S. aureus and P. aeruginosa [221]. Synergy of AgNPs and antimicrobial peptides polymyxin B and gramicidin S was reported also against different Gram-negative bacteria [222].
Recently several studies have indicated that AgNPs may strengthen the antibacterial effects of conventional antibiotics (beta-lactam antibiotics, macrolides, lincosamides, aminoglycosides) either additively or synergistically [223,224,225,226,227,228,229]. The synergistic effect of antibiotics and AgNPs was reported even at concentrations below their own effectiveness (i.e., below MICs) [225,230,231,232,233]. Brown et al. showed a synergistic effect of AgNPs functionalized with ampicillin, even against multiple-antibiotic-resistant isolates of P. aeruginosa, E. aerogenes and methicillin-resistant S. aureus [234]. Also Smekalova et al. and Panacek et al. proved enhancement of the antibacterial effect of antibiotics in combination with AgNPs against several animal and human pathogens and resistant bacterial strains [235,236,237]. These findings clearly showed that it is possible to find an effective combination of antibiotics and AgNPs or another antimicrobial with a multi-level mode of action, resulting in a synergistic antimicrobial effect allowing efficient inhibition of bacterial pathogens including highly resistant bacterial strains using significantly lower doses as compared to an antibiotic alone. Replacement of frequently used antibiotics by AgNPs or a combination of these antibiotics with AgNPs represent a promising tool on how to kill bacteria without the development of antibiotic resistance [238].

10. Potential Side Effects of Silver Nanoparticles

The main problem by using AgNPs on biomaterials is that they are considered toxic not only for bacteria, but also to human host cells. Toxicity of silver nanoparticles to mammalian cells is considerably lower in comparison with antibacterial effective concentrations also due to the fact that eukaryotic cells have an antioxidant cellular mechanism that protects them [239,240]. The extended use of AgNPs can lead to a number of health problems from argyria [241] to silver accumulation in human liver and kidney. Although silver and its derivatives are already in clinical use, evidence of serious health problems [242] and high toxicity are rare [243].
A number of in vitro studies have been performed exploring the effects of AgNPs on a variety of cell types [88,239,244,245,246,247,248,249,250,251,252,253]. The most common mechanisms of toxicity from nanosized silver particles, as well as silver ions released from them [245,254] are: oxidative stress [246,255], Trojan-horse mechanisms [256,257] and DNA damage [163].
The question arises as to what determines if silver nanoparticles are toxic or not. In general, toxicity is determined by many factors, either on the side of the nanoparticles or on the side of the body that they are in contact with. Regarding nanoparticles, these are mainly size, shape [258], charge, surface modification, tendency to release ions, dose and exposure time. The role of the particle size is more important than concentration or dose [259,260] because smaller nanoparticles have a higher surface/volume ratio leading to higher oxidation and dissolution, accompanied by higher silver ion release [261]. Therefore, smaller AgNPs may show higher toxicity due to their larger specific surface area and associated faster Ag+ release compared to larger AgNPs [260]. However, that does not necessarily mean higher toxicity in a particular material and situation. Silver ion release is controlled by surface modification/stabilization and can be further influenced by other compounds presented in biological environment. Moreover, Ag+ release is also dependent on the formation of a protective oxidized silver layer that prevents full oxidation and dissolution of AgNPs [262]. Likewise, it is known that spherical nanoparticles are less toxic than wires [263] and negatively charged NPs exhibit low toxicity [264,265].
On the host side, the potential toxicity of AgNPs is determined by patient health status, routes of exposure, gender and other factors. In some organs (liver, kidney), silver is accumulated soon after application, while in others (brain, lung) higher concentrations are detected after a prolonged period [266,267,268,269]. Pauksch et al. [270] investigated the effect of AgNPs on human osteoblasts and it turned out that AgNPs were toxic at concentrations higher than 10 μg/g. The authors suggested that there is a gap between the toxic and antibacterial doses of AgNPs. This statement was confirmed by Necula et al. [271] who tested the antibacterial efficacy and toxicity towards the human osteoblastic cell line. They demonstrated that the antibacterial dose is by an order of magnitude lower than that having a toxic effect on human cells. These observations support the promising usage of presence of a therapeutically useful window for the application of AgNPs in orthopaedics.

11. Protocol for Testing of Silver Nanoparticle Coating Technologies Intended for Usage in Orthopaedics

There is no doubt that nanotreatment of biomaterial surfaces offers new opportunities for PJI prevention. On the other hand, the main obstacles preventing broader usage of such technologies are cytotoxicity and resultant decreased biocompatibility. It should be cautioned that nanotechnologies can also induce unintended inflammatory responses related to activation of immune cells such as dendritic cells, macrophages and others. Concern also exists over the mechanical properties of implant nanocoatings since damage may occur during surgical implantation, especially in cementless implants inserted via press-fit methods. In addition, creating a coating-substrate interface robust enough to sustain the mechanical stresses involved in surgical implant insertion and ultimate loading once in vivo remains a challenge. Lastly, the risk of remote effects of absorbed nanosilver is still a potential problem.
Therefore, a set of in vitro tests (followed by in vivo experiments) is required to characterize in detail antibacterial efficacy, as well as biocompatibility and safety of such material modifications. The latter means to examine the cytotoxicity, cancerogenicity, interactions with osteoblasts and other cells and the potential of adverse stimulation of an immune response. As a result, specialists in nano-toxicology (esp. nano-genotoxicology, cytotoxicity, immunotoxicity), in vitro pharmacokinetics, pharmacodynamics and kinetics of particles are needed to collaborate in the development, preclinical testing and approval of any material modifications for clinical usage.

11.1. Demonstration of Antibacterial Efficacy

A critical step in progress lies in the demonstrating that newly developed biomaterials, or surface modifications possess antibacterial efficacy [272]. To date there is no widely accepted methodology available that could precisely and reproducibly demonstrate antibacterial behaviour of the proposed anti-infective technologies. Major criticisms are levelled at the static “closed” testing system, whereas in vivo, the implant has to face a dynamic, continuously changing, mechanically unstable and predominantly fluid environment [273]. As a result, the majority of studies to date have used inappropriate and insufficient protocols.
Controllable, standardized testing conditions that closely mimic the human in vivo environment are needed in order to overcome the aforementioned issues [273]. PJIs develop under low shear conditions and a multidirectional low-pressure fluid flow. A variety of testing tools have been proposed that attempt to simulate conditions of continuous or intermittent fluid-displacement in both the low and high shear conditions [274]. Protocols for cultivation of particular species (multispecies) biofilms under controllable, constant and reproducible conditions have been also described [275]. Finally, representative in vitro and in vivo models to test bacterial adhesion and biofilm formation on biomaterials for each particular clinical situation (i.e., total joint arthroplasty, internal, external fixation) should be further developed and appropriately validated. Given the large variability of antibacterial strategies, it is likely that testing methods must be better tailored to match the specific proposed strategy at hand [150].

11.2. Testing of Cytotoxicity

Although many studies presented new nanoparticle surface treatments proving in vitro safety [271], others demonstrated the potential danger of such materials [276]. Nanoparticles have different effects on human health depending on the bulk material from which they have been produced [277]. In addition to the elemental composition, factors like nanoparticle dose, size, shape, exposure time and surface chemistry can affect its biological behaviour. Regarding the shape, silver nanowires showed the strongest cytotoxicity and immunological responses, whereas spherical silver particles had negligible effects on cells when tested in human cells [278]. Liu et al. found that 5 nm AgNPs were more toxic than 20 and 50 nm AgNPs in four cell lines (A549, HepG2, MCF-7, SGC-7901), indicating a size-dependent effect on cell viability [253]. It should be noted that some cell lines (PC-12 and NIH-3T3) exhibit greater sensitivity to AgNPs than another mammalian cell lines [279]. The rate of ion release and its variation in different media should be taken in consideration as well. All this concludes that cytotoxicity testing should be always suited exactly for the proposed implant coating (i.e., exact nanoparticle size, concentration, shape, fixation method etc.) and its intended use in a specific tissue. In addition, a level of cytotoxicity can be dependent on the assay technique and a difference between extraction-based and direct contact assays has been found [280].
When testing new materials or surface modifications, the cytotoxicity testing is performed first and other tests (anti-bacterial, immunoreactivity etc.) are advanced only after the biomaterial is classified as biologically not harmful. Cytotoxicity testing is rapid, sensitive and inexpensive. Another big advantage of cytotoxicity testing is the standardization of the procedure, ISO 10993 and the FDA (Food and Drug Administration) blue book memorandum (#G95-1) and its suitability for the testing of biomaterial from any part of the medical device (i.e., TJA, internal, external fixation). This test is commonly performed using a mouse fibroblasts cell-line as target cells, following the exposure with the material (direct contact) as well as the extract of the material. Cells are very sensitive to biologically harmful extractables in certain quantities resulting in visible signs of toxicity, such as changes in cell morphology, vacuolization, or detachment. A different way of testing the nanotoxicity was described by Liu et al. via evaluation of induction of apoptosis [281].
Regarding novel anti-infective treatments with silver nanoparticles, a recent study reported that the BALB/c 3T3 cell line is 1000 times more sensitive for testing the toxicity of silver NPs than the in vivo animal models [282]. Although some studies showed a dose-dependent cytotoxic effect of nano-silver, new types of nano-silver were proven to be not cytotoxic [283], or it was shown that combination of a low amount of nano-silver with antibiotics provides an effective antibacterial action with negligible cytotoxic effect [236].

11.3. Testing of Immunoreactivity

Sensitization testing represents another part of the testing battery for new biomaterials, establishing the potential of a biomaterial to elicit immunogenic and allergenic responses (immunoreactivity). Currently, the most commonly used tests for novel materials and those medical devices that contact deep tissue, is the guinea pig maximization test (GPMT) where the extract of a biomaterial together with an adjuvant, is intradermally injected in model animals (Biological Evaluation of Medical Devices). Alternatively, a mouse local lymph node assay (LLNA) requiring less material than GPMT but needed to harvest the lymph nodes from sacrificed animals may be used with some precautions, such as a high number of false positives.
However, all animal tests are expensive and take days (weeks) to get results. It is therefore of great interest to replace or drastically reduce the utilization of tests based on experimental animals with suitable cell-based assays which exhibit required reliability, accuracy and importantly correlate to human reactivity. Several studies have shown great potential in the use of the MUTZ-3 human dendritic-cell cell-line for assessing in vitro sensitizing potency of chemicals and biomaterials, using a genomic biomarker signature [284,285,286]. Besides the MUTZ-3 assay, other tests are investigated for their potential to predict sensibilisation in humans as well [287]. However, it is likely that new testing methods must be validated and standardized to match the requirements for accuracy and ability, to be sensitive to the whole spectrum of molecules, with allergenic potential including nanosilver.

12. Time to Translation?

In the field of orthopaedics, there are no implants protected with silver nanotreatment available for clinical usage to date. At least two manufacturers already produce TJA treated by galvanic deposition of elementary silver on request (Implantcast GmbH–Medizintechnik, Buxtehude, Germany; Stanmore Implants, Borehamwood, UK). Initial clinical experiences with these “tailored” implants have been promising [288]. In addition, at least one study examined clinical usage of thermal-sprayed silver oxide in hydroxyapatite coating for total hip implants [84].
The situation is a little better in the field of indwelling medical devices protected by surface treatment with AgNPs. Again, in contrast to extensive experimental research, only several clinical studies have been conducted to demonstrate reduction of infections associated with the AgNP coating in these devices (left within a bodily organ for a limited time). For instance, the usage of external ventricular drainage catheters treated with AgNPs decreased the infection rate [289] while the venous catheters tailored with AgNPs failed to lower infection rates [290]. However, together these data preclude making any conclusions in support of their widespread clinical usage.
Examination of global grants and published studies of this topic suggests a striking discrepancy between proposed strategies of antibacterial surface treatment and ultimate completion of in vitro and in vivo experimentation. In fact, we believe that very little progress has actually been made in the translation of the aforementioned modalities into clinically useful technologies. Barriers to translational medicine in this area are not only related to economic, medicolegal and biotechnological issues but with major problems in the demonstration of the safety of clinical trials. Concerns about long-term durability of such new implants as compared to traditional implants are also realistic. Leaders in this field have recently proposed that in order for some of these obstacles to be overcome, we must improve efficiency and effectiveness amongst all the partners involved. Patients will benefit from these technologies only by improving collaborative efforts among governments, regulatory agencies, industry leaders and health care payers [291]. While pressures exist worldwide to diminish the incidence of PJIs, surprisingly there is not a single large clinical study examining the role of broad-range implementation of implants containing antibacterial surface treatments.

13. Future Developments

The ideal implant surface modification using whatever approach, should provide antibacterial protection throughout the life of the implant with minimal side effects. In relation to AgNPs and their usage in modification of implants, there are three crucial future developments. The first one is synthesis of AgNPs with defined optimal size, ensuring high antibacterial activity and concurrently low cytotoxicity to mammalian cells; that means good biocompatibility with tissues without acute or long-term adverse effects. The second is the development of a new coating technique, or improvement and optimization of a current one ensuring reliable formation of compact, continuous and durable layer of AgNPs. The third concerns the elimination of an inhibitory effect of human lipids and proteins preventing AgNPs from implementing their intended antibacterial effect. These substances cover surfaces of TJA immediately after an implant is placed into the human body. To meet this challenge, recent advances in the field of surface chemistry, fluid mechanics, fluid mechanobiology, bio-inspired materials and/or endogenous mechanisms of immune stimulation should be utilized.
Another important issue related to antibacterial efficiency of AgNPs is connected with the possible development of bacterial resistance to silver NPs. It can be expected that with increasing use of AgNPs in killing bacteria or in the prevention of bacterial colonization in clinical medicine, the bacterial resistance to AgNPs could develop. As a result, strategies combining AgNPs with other antibacterial substances/approaches (either composite or nanocomposite layers), in order to achieve additive/synergistic effects are highly reasonable and should be investigated.
Finally, further investigation should be carried out in the field of strategies combining AgNPs with approaches restoring/maintaining local tissue homeostasis and modulating the immunologic surveillance and patrolling. This concept might comply with a wide variety of clinical situations ranging from residual low dose bacterial load during the surgery, to late haematogenic spreading of infection.

14. Conclusions

There is no doubt that prevention is the best response to the growing problem of orthopaedic implant infections. Engineers believe they are able to develop reliable, durable, non-toxic and safe biomaterials preventing bacterial adhesion and formation of biofilm on surfaces. Strategies incorporating nanopatterning and other nanotechnologies show great promise. Research in the field of antibacterial surface treatment has demonstrated in vitro and in vivo effectiveness of the technologies based on AgNPs, combining a strong antibacterial effect with relative inertness to the inner environment of a patient. On the other hand, issues relating to the mechanical properties of these technologies and the potential for detrimental side effects, such as toxicity and interference with osseointegration require further investigation.

Acknowledgments

Funding was obtained from the Ministry of Health of the Czech Republic (AZV VES 15-27726A).

Author Contributions

J.G. gave the ideas, designed the review and wrote relevant parts of the manuscript. A.P., R.P., S.H. are responsible for the parts of the manuscript related to silver nanoparticles. E.K. contributed especially to the part on the methods appropriate to testing of silver nanocoating. M.Hob. read the literature and contributed to the orthopaedic parts of the manuscript. M.Hol. drew all the figures in the manuscript. All authors read and approved the final manuscript.

Conflicts of Interest

All the authors certify that they have no commercial associations that may pose a conflict of interest in connection with the submitted article.

Abbreviations

The following abbreviations are used in this manuscript:
Agsilver
AgNPsilver nanoparticle
ATPaseadenosine triphosphatase
DAL/CHIdopamine-modified alginate/chitosan
DNAdeoxyribonucleic acid
EPSextracellular polymeric substances
FDAFood and Drug Administration
GPMTguinea pig maximization test
LCClocal carriers or coating
LLNAlocal lymph node assay
MAMPmicrobe associated molecular pattern
MICminimum inhibitory concentration
MRSAmethicillin-resistant Staphylococcus aureus
MSCRAMMMicrobial Surface Components Recognizing Adhesive Matrix Molecule
NPnanoparticle
NTnanotube
PJIprosthetic joint infection
PRRpattern-recognition receptor
QAS3-trimethoxysily-propyldimethyloctadecyl-ammonium chloride
QASquaternary ammonium salt
Tititanium
TiO2titanium dioxide
TJAtotal joint arthroplasty
USAUnited States of America
UVultraviolet

References

  1. Berend, K.R.; Lombardi, A.V., Jr.; Morris, M.J.; Bergeson, A.G.; Adams, J.B.; Sneller, M.A. Two-stage treatment of hip periprosthetic joint infection is associated with a high rate of infection control but high mortality. Clin. Orthop. Relat. Res. 2013, 471, 510–518. [Google Scholar] [CrossRef] [PubMed]
  2. De Angelis, G.; Mutters, N.T.; Minkley, L.; Holderried, F.; Tacconelli, E. Prosthetic joint infections in the elderly. Infection 2015, 43, 629–637. [Google Scholar] [CrossRef] [PubMed]
  3. Bedair, H.; Goyal, N.; Dietz, M.J.; Urish, K.; Hansen, V.; Manrique, J.; Hamilton, W.; Deirmengian, G. A history of treated periprosthetic joint infection increases the risk of subsequent different site infection. Clin. Orthop. Relat. Res. 2015, 473, 2300–2304. [Google Scholar] [CrossRef] [PubMed]
  4. Voigt, J.; Mosier, M.; Darouiche, R. Systematic review and meta-analysis of randomized controlled trials of antibiotics and antiseptics for preventing infection in people receiving primary total hip and knee prostheses. Antimicrob. Agents Chemother. 2015, 59, 6696–6707. [Google Scholar] [CrossRef] [PubMed]
  5. Kamath, A.F.; Ong, K.L.; Lau, E.; Chan, V.; Vail, T.P.; Rubash, H.E.; Berry, D.J.; Bozic, K.J. Quantifying the burden of revision total joint arthroplasty for periprosthetic infection. J. Arthroplast. 2015, 30, 1492–1497. [Google Scholar] [CrossRef] [PubMed]
  6. Witso, E. The rate of prosthetic joint infection is underestimated in the arthroplasty registers. Acta Orthop. 2015, 277–278. [Google Scholar] [CrossRef] [PubMed]
  7. Kurtz, S.M.; Lau, E.; Watson, H.; Schmier, J.K.; Parvizi, J. Economic burden of periprosthetic joint infection in the united states. J. Arthroplast. 2012, 27, 61–65. [Google Scholar] [CrossRef] [PubMed]
  8. Lindgren, V.; Gordon, M.; Wretenberg, P.; Karrholm, J.; Garellick, G. Deep infection after total hip replacement: A method for national incidence surveillance. Infect. Control Hosp. Epidemiol. 2014, 35, 1491–1496. [Google Scholar] [CrossRef] [PubMed]
  9. Huotari, K.; Peltola, M.; Jamsen, E. The incidence of late prosthetic joint infections: A registry-based study of 112,708 primary hip and knee replacements. Acta Orthop. 2015, 86, 321–325. [Google Scholar] [CrossRef] [PubMed]
  10. Tande, A.J.; Patel, R. Prosthetic joint infection. Clin. Microbiol. Rev. 2014, 27, 302–345. [Google Scholar] [CrossRef] [PubMed]
  11. Gallo, J.; Kolar, M.; Dendis, M.; Loveckova, Y.; Sauer, P.; Zapletalova, J.; Koukalova, D. Culture and pcr analysis of joint fluid in the diagnosis of prosthetic joint infection. New Microbiol. 2008, 31, 97–104. [Google Scholar] [PubMed]
  12. Aggarwal, V.K.; Bakhshi, H.; Ecker, N.U.; Parvizi, J.; Gehrke, T.; Kendoff, D. Organism profile in periprosthetic joint infection: Pathogens differ at two arthroplasty infection referral centers in europe and in the united states. J. Knee Surg. 2014, 27, 399–406. [Google Scholar] [CrossRef] [PubMed]
  13. Bemer, P.; Plouzeau, C.; Tande, D.; Leger, J.; Giraudeau, B.; Valentin, A.S.; Jolivet-Gougeon, A.; Vincent, P.; Corvec, S.; Gibaud, S.; et al. Evaluation of 16s rRNA gene pcr sensitivity and specificity for diagnosis of prosthetic joint infection: A prospective multicenter cross-sectional study. J. Clin. Microbiol. 2014, 52, 3583–3589. [Google Scholar] [CrossRef] [PubMed]
  14. Benito, N.; Franco, M.; Coll, P.; Galvez, M.L.; Jordan, M.; Lopez-Contreras, J.; Pomar, V.; Monllau, J.C.; Mirelis, B.; Gurgui, M. Etiology of surgical site infections after primary total joint arthroplasties. J. Orthop. Res. 2014, 32, 633–637. [Google Scholar] [CrossRef] [PubMed]
  15. Gristina, A.G.; Naylor, P.; Myrvik, Q. Infections from biomaterials and implants: A race for the surface. Med. Prog. Technol. 1988, 14, 205–224. [Google Scholar] [PubMed]
  16. Nishitani, K.; Sutipornpalangkul, W.; de Mesy Bentley, K.L.; Varrone, J.J.; Bello-Irizarry, S.N.; Ito, H.; Matsuda, S.; Kates, S.L.; Daiss, J.L.; Schwarz, E.M. Quantifying the natural history of biofilm formation in vivo during the establishment of chronic implant-associated staphylococcus aureus osteomyelitis in mice to identify critical pathogen and host factors. J. Orthop. Res. 2015, 33, 1311–1319. [Google Scholar] [CrossRef] [PubMed]
  17. Vidlak, D.; Kielian, T. Infectious dose dictates the host response during S. Aureus orthopedic biofilm infection. Infect. Immun. 2016. [Google Scholar] [CrossRef] [PubMed]
  18. Yue, C.; Zhao, B.; Ren, Y.; Kuijer, R.; van der Mei, H.C.; Busscher, H.J.; Rochford, E.T. The implant infection paradox: Why do some succeed when others fail? Opinion and discussion paper. Eur. Cells Mater. 2015, 29, 303–310. [Google Scholar]
  19. Zhou, X.; Yishake, M.; Li, J.; Jiang, L.; Wu, L.; Liu, R.; Xu, N. Genetic susceptibility to prosthetic joint infection following total joint arthroplasty: A systematic review. Gene 2015, 563, 76–82. [Google Scholar] [CrossRef] [PubMed]
  20. Navratilova, Z.; Gallo, J.; Mrazek, F.; Lostak, J.; Petrek, M. Mbl2 gene variation affecting serum mbl is associated with prosthetic joint infection in czech patients after total joint arthroplasty. Tissue Antigens 2012, 80, 444–451. [Google Scholar] [CrossRef] [PubMed]
  21. Busscher, H.J.; van der Mei, H.C. How do bacteria know they are on a surface and regulate their response to an adhering state? PLoS Pathog. 2012, 8, e1002440. [Google Scholar] [CrossRef] [PubMed]
  22. Costerton, W.; Veeh, R.; Shirtliff, M.; Pasmore, M.; Post, C.; Ehrlich, G. The application of biofilm science to the study and control of chronic bacterial infections. J. Clin. Investig. 2003, 112, 1466–1477. [Google Scholar] [CrossRef] [PubMed]
  23. Eichenberger, E.M.; Thaden, J.T.; Sharma-Kuinkel, B.; Park, L.P.; Rude, T.H.; Ruffin, F.; Hos, N.J.; Seifert, H.; Rieg, S.; Kern, W.V.; et al. Polymorphisms in fibronectin binding proteins a and b among staphylococcus aureus bloodstream isolates are not associated with arthroplasty infection. PLoS ONE 2015, 10, e0141436. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, Y.; Busscher, H.J.; van der Mei, H.C.; Norde, W. Statistical analysis of long- and short-range forces involved in bacterial adhesion to substratum surfaces as measured using atomic force microscopy. Appl. Environ. Microbiol. 2011, 77, 5065–5070. [Google Scholar] [CrossRef] [PubMed]
  25. Chagnot, C.; Zorgani, M.A.; Astruc, T.; Desvaux, M. Proteinaceous determinants of surface colonization in bacteria: Bacterial adhesion and biofilm formation from a protein secretion perspective. Front. Microbiol. 2013, 4, 303. [Google Scholar] [CrossRef] [PubMed]
  26. Thevenot, P.; Hu, W.; Tang, L. Surface chemistry influences implant biocompatibility. Curr. Top. Med. Chem. 2008, 8, 270–280. [Google Scholar] [PubMed]
  27. Roach, P.; Eglin, D.; Rohde, K.; Perry, C.C. Modern biomaterials: A review—Bulk properties and implications of surface modifications. J. Mater. Sci. Mater. Med. 2007, 18, 1263–1277. [Google Scholar] [CrossRef] [PubMed]
  28. Wilson, C.J.; Clegg, R.E.; Leavesley, D.I.; Pearcy, M.J. Mediation of biomaterial-cell interactions by adsorbed proteins: A review. Tissue Eng. 2005, 11, 1–18. [Google Scholar] [CrossRef] [PubMed]
  29. Stoodley, P.; Ehrlich, G.D.; Sedghizadeh, P.P.; Hall-Stoodley, L.; Baratz, M.E.; Altman, D.T.; Sotereanos, N.G.; Costerton, J.W.; Demeo, P. Orthopaedic biofilm infections. Curr. Orthop. Pract. 2011, 22, 558–563. [Google Scholar] [CrossRef] [PubMed]
  30. 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] [PubMed]
  31. Laverty, G.; Gorman, S.P.; Gilmore, B.F. Biomolecular mechanisms of staphylococcal biofilm formation. Future Microbiol. 2013, 8, 509–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Foster, T.J.; Geoghegan, J.A.; Ganesh, V.K.; Hook, 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]
  33. Fux, C.A.; Costerton, J.W.; Stewart, P.S.; Stoodley, P. Survival strategies of infectious biofilms. Trends Microbiol. 2005, 13, 34–40. [Google Scholar] [CrossRef] [PubMed]
  34. Arciola, C.R.; Campoccia, D.; Ehrlich, G.D.; Montanaro, L. Biofilm-based implant infections in orthopaedics. Adv. Exp. Med. Biol. 2015, 830, 29–46. [Google Scholar] [PubMed]
  35. 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] [PubMed]
  36. Gardner, A.B.; Lee, S.K.; Woods, E.C.; Acharya, A.P. Biomaterials-based modulation of the immune system. BioMed Res. Int. 2013, 2013, 732182. [Google Scholar] [CrossRef] [PubMed]
  37. Pajarinen, J.; Lin, T.H.; Sato, T.; Yao, Z.; Goodman, S. Interaction of materials and biology in total joint replacement—Successes, challenges and future directions. J. Mater. Chem. B Mater. Biol. Med. 2014, 2, 7094–7108. [Google Scholar] [CrossRef] [PubMed]
  38. 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]
  39. Yue, C.; van der Mei, H.C.; Kuijer, R.; Busscher, H.J.; Rochford, E.T. Mechanism of cell integration on biomaterial implant surfaces in the presence of bacterial contamination. J. Biomed. Mater. Res. A 2015, 103, 3590–3598. [Google Scholar] [CrossRef] [PubMed]
  40. Higgins, D.M.; Basaraba, R.J.; Hohnbaum, A.C.; Lee, E.J.; Grainger, D.W.; Gonzalez-Juarrero, M. Localized immunosuppressive environment in the foreign body response to implanted biomaterials. Am. J. Pathol. 2009, 175, 161–170. [Google Scholar] [CrossRef] [PubMed]
  41. Zimmerli, W.; Sendi, P. Pathogenesis of implant-associated infection: The role of the host. Semin. Immunopathol. 2011, 33, 295–306. [Google Scholar] [CrossRef] [PubMed]
  42. An, Y.H.; Friedman, R.J. Prevention of sepsis in total joint arthroplasty. J. Hosp. Infect. 1996, 33, 93–108. [Google Scholar] [CrossRef]
  43. Humphreys, H. Surgical site infection, ultraclean ventilated operating theatres and prosthetic joint surgery: Where now? J. Hosp. Infect. 2012, 81, 71–72. [Google Scholar] [CrossRef] [PubMed]
  44. McHugh, S.M.; Hill, A.D.; Humphreys, H. Laminar airflow and the prevention of surgical site infection. More harm than good? Surgeon 2015, 13, 52–58. [Google Scholar] [CrossRef] [PubMed]
  45. Mejia, E.; Williams, A.; Long, M. Decreasing prosthetic joint surgical site infections: An interdisciplinary approach. AORN J. 2015, 101, 213–222. [Google Scholar] [CrossRef] [PubMed]
  46. Thornley, P.; Evaniew, N.; Riediger, M.; Winemaker, M.; Bhandari, M.; Ghert, M. Postoperative antibiotic prophylaxis in total hip and knee arthroplasty: A systematic review and meta-analysis of randomized controlled trials. CMAJ Open 2015, 3, E338–E343. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, J.; Zhu, C.; Cheng, T.; Peng, X.; Zhang, W.; Qin, H.; Zhang, X. A systematic review and meta-analysis of antibiotic-impregnated bone cement use in primary total hip or knee arthroplasty. PLoS ONE 2013, 8, e82745. [Google Scholar] [CrossRef] [PubMed]
  48. McMurray, C.L.; Hardy, K.J.; Verlander, N.Q.; Hawkey, P.M. Antibiotic surgical prophylaxis increases nasal carriage of antibiotic-resistant staphylococci. J. Med. Microbiol. 2015, 64, 1489–1495. [Google Scholar] [CrossRef] [PubMed]
  49. Illingworth, K.D.; Mihalko, W.M.; Parvizi, J.; Sculco, T.; McArthur, B.; el Bitar, Y.; Saleh, K.J. How to minimize infection and thereby maximize patient outcomes in total joint arthroplasty: A multicenter approach: Aaos exhibit selection. J. Bone Jt. Surg. Am. Vol. 2013, 95, e50. [Google Scholar] [CrossRef] [PubMed]
  50. Florschutz, A.V.; Fagan, R.P.; Matar, W.Y.; Sawyer, R.G.; Berrios-Torres, S.I. Surgical site infection risk factors and risk stratification. J. Am. Acad. Orthop. Surg. 2015, 23, S8–S11. [Google Scholar] [CrossRef] [PubMed]
  51. Maoz, G.; Phillips, M.; Bosco, J.; Slover, J.; Stachel, A.; Inneh, I.; Iorio, R. The otto aufranc award: Modifiable versus nonmodifiable risk factors for infection after hip arthroplasty. Clin. Orthop. Relat. Res. 2015, 473, 453–459. [Google Scholar] [CrossRef] [PubMed]
  52. Ma, Z.; Guo, F.; Qi, J.; Xiang, W.; Zhang, J. Meta-analysis shows that obesity may be a significant risk factor for prosthetic joint infections. Int. Orthop. 2015. [Google Scholar] [CrossRef] [PubMed]
  53. Alijanipour, P.; Heller, S.; Parvizi, J. Prevention of periprosthetic joint infection: What are the effective strategies? J. Knee Surg. 2014, 27, 251–258. [Google Scholar] [CrossRef] [PubMed]
  54. Rezapoor, M.; Parvizi, J. Prevention of periprosthetic joint infection. J. Arthroplast. 2015, 30, 902–907. [Google Scholar] [CrossRef] [PubMed]
  55. Getzlaf, M.A.; Lewallen, E.A.; Kremers, H.M.; Jones, D.L.; Bonin, C.A.; Dudakovic, A.; Thaler, R.; Cohen, R.C.; Lewallen, D.G.; van Wijnen, A.J. Multi-disciplinary antimicrobial strategies for improving orthopaedic implants to prevent prosthetic joint infections in hip and knee. J. Orthop. Res. 2016, 34, 177–186. [Google Scholar] [CrossRef] [PubMed]
  56. Romano, C.L.; Scarponi, S.; Gallazzi, E.; Romano, D.; Drago, L. Antibacterial coating of implants in orthopaedics and trauma: A classification proposal in an evolving panorama. J. Orthop. Surg. Res. 2015, 10, 157. [Google Scholar] [CrossRef] [PubMed]
  57. Tsaras, G.; Osmon, D.R.; Mabry, T.; Lahr, B.; St Sauveur, J.; Yawn, B.; Kurland, R.; Berbari, E.F. Incidence, secular trends, and outcomes of prosthetic joint infection: A population-based study, olmsted county, minnesota, 1969–2007. Infect. Control Hosp. Epidemiol. 2012, 33, 1207–1212. [Google Scholar] [CrossRef] [PubMed]
  58. Zhu, Y.; Zhang, F.; Chen, W.; Liu, S.; Zhang, Q.; Zhang, Y. Risk factors for periprosthetic joint infection after total joint arthroplasty: A systematic review and meta-analysis. J. Hosp. Infect. 2015, 89, 82–89. [Google Scholar] [CrossRef] [PubMed]
  59. Ratner, B.D.; Schoen, F.J. The concept and assessment of biocompatibility. In Biomaterials Science: An Introduction to Materials in Medicine, 3rd ed.; Ratner, B.D., Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.; Elsevier: Amsterdam, The Netherlands, 2013; Volume 1, pp. 588–592. [Google Scholar]
  60. Bernthal, N.M.; Stavrakis, A.I.; Billi, F.; Cho, J.S.; Kremen, T.J.; Simon, S.I.; Cheung, A.L.; Finerman, G.A.; Lieberman, J.R.; Adams, J.S.; et al. A mouse model of post-arthroplasty staphylococcus aureus joint infection to evaluate in vivo the efficacy of antimicrobial implant coatings. PLoS ONE 2010, 5, e12580. [Google Scholar] [CrossRef] [PubMed]
  61. Scherr, T.D.; Lindgren, K.E.; Schaeffer, C.R.; Hanke, M.L.; Hartman, C.W.; Kielian, T. Mouse model of post-arthroplasty staphylococcus epidermidis joint infection. Methods Mol. Biol. 2014, 1106, 173–181. [Google Scholar] [PubMed]
  62. Gatin, L.; Saleh-Mghir, A.; Massin, P.; Cremieux, A.C. Critical analysis of experimental models of periprosthetic joint infection. Orthop. Traumatol. Surg. Res. 2015, 101, 851–855. [Google Scholar] [CrossRef] [PubMed]
  63. Corvec, S.; Portillo, M.E.; Pasticci, B.M.; Borens, O.; Trampuz, A. Epidemiology and new developments in the diagnosis of prosthetic joint infection. Int. J. Artif. Organs 2012, 35, 923–934. [Google Scholar] [CrossRef] [PubMed]
  64. Kapadia, B.H.; Berg, R.A.; Daley, J.A.; Fritz, J.; Bhave, A.; Mont, M.A. Periprosthetic joint infection. Lancet 2016, 387, 386–394. [Google Scholar] [CrossRef]
  65. Ratner, B.D.; Hoffman, A.S. Physicochemical surface modifications of materials used in medicine. In Biomaterials Science: An Introduction to Materials in Medicine; Ratner, B.D., Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.; Academic Press (Elsevier): Waltham, MA, USA, 2013; Volume 1, pp. 259–276. [Google Scholar]
  66. Follmann, H.D.; Martins, A.F.; Gerola, A.P.; Burgo, T.A.; Nakamura, C.V.; Rubira, A.F.; Muniz, E.C. Antiadhesive and antibacterial multilayer films via layer-by-layer assembly of tmc/heparin complexes. Biomacromolecules 2012, 13, 3711–3722. [Google Scholar] [CrossRef] [PubMed]
  67. Neoh, K.G.; Kang, E.T. Combating bacterial colonization on metals via polymer coatings: Relevance to marine and medical applications. ACS Appl. Mater. Interfaces 2011, 3, 2808–2819. [Google Scholar] [CrossRef] [PubMed]
  68. Muszanska, A.K.; Rochford, E.T.; Gruszka, A.; Bastian, A.A.; Busscher, H.J.; Norde, W.; van der Mei, H.C.; Herrmann, A. Antiadhesive polymer brush coating functionalized with antimicrobial and rgd peptides to reduce biofilm formation and enhance tissue integration. Biomacromolecules 2014, 15, 2019–2026. [Google Scholar] [CrossRef] [PubMed]
  69. An, Y.H.; Stuart, G.W.; McDowell, S.J.; McDaniel, S.E.; Kang, Q.; Friedman, R.J. Prevention of bacterial adherence to implant surfaces with a crosslinked albumin coating in vitro. J. Orthop. Res. 1996, 14, 846–849. [Google Scholar] [CrossRef] [PubMed]
  70. Zhu, H.; Guo, Z.; Liu, W. Adhesion behaviors on superhydrophobic surfaces. Chem. Commun. 2014, 50, 3900–3913. [Google Scholar] [CrossRef] [PubMed]
  71. Stallard, C.P.; McDonnell, K.A.; Onayemi, O.D.; O’Gara, J.P.; Dowling, D.P. Evaluation of protein adsorption on atmospheric plasma deposited coatings exhibiting superhydrophilic to superhydrophobic properties. Biointerphases 2012, 7, 31. [Google Scholar] [CrossRef] [PubMed]
  72. Poncin-Epaillard, F.; Herry, J.M.; Marmey, P.; Legeay, G.; Debarnot, D.; Bellon-Fontaine, M.N. Elaboration of highly hydrophobic polymeric surface—A potential strategy to reduce the adhesion of pathogenic bacteria? Mater. Sci. Eng. C Mater. Biol. Appl. 2013, 33, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
  73. Shida, T.; Koseki, H.; Yoda, I.; Horiuchi, H.; Sakoda, H.; Osaki, M. Adherence ability of staphylococcus epidermidis on prosthetic biomaterials: An in vitro study. Int. J. Nanomed. 2013, 8, 3955–3961. [Google Scholar]
  74. Singh, A.V.; Vyas, V.; Patil, R.; Sharma, V.; Scopelliti, P.E.; Bongiorno, G.; Podesta, A.; Lenardi, C.; Gade, W.N.; Milani, P. Quantitative characterization of the influence of the nanoscale morphology of nanostructured surfaces on bacterial adhesion and biofilm formation. PLoS ONE 2011, 6, e25029. [Google Scholar] [CrossRef] [PubMed]
  75. Ivanova, E.P.; Truong, V.K.; Wang, J.Y.; Berndt, C.C.; Jones, R.T.; Yusuf, I.I.; Peake, I.; Schmidt, H.W.; Fluke, C.; Barnes, D.; et al. Impact of nanoscale roughness of titanium thin film surfaces on bacterial retention. Langmuir ACS J. Surf. Colloids 2010, 26, 1973–1982. [Google Scholar] [CrossRef] [PubMed]
  76. Truong, V.K.; Lapovok, R.; Estrin, Y.S.; Rundell, S.; Wang, J.Y.; Fluke, C.J.; Crawford, R.J.; Ivanova, E.P. The influence of nano-scale surface roughness on bacterial adhesion to ultrafine-grained titanium. Biomaterials 2010, 31, 3674–3683. [Google Scholar] [PubMed]
  77. Filova, E.; Fojt, J.; Kryslova, M.; Moravec, H.; Joska, L.; Bacakova, L. The diameter of nanotubes formed on ti-6al-4v alloy controls the adhesion and differentiation of saos-2 cells. Int. J. Nanomed. 2015, 10, 7145–7163. [Google Scholar]
  78. Pandit, V.; Zuidema, J.M.; Venuto, K.N.; Macione, J.; Dai, G.; Gilbert, R.J.; Kotha, S.P. Evaluation of multifunctional polysaccharide hydrogels with varying stiffness for bone tissue engineering. Tissue Eng. A 2013, 19, 2452–2463. [Google Scholar]
  79. Zhao, C.; Li, X.; Li, L.; Cheng, G.; Gong, X.; Zheng, J. Dual functionality of antimicrobial and antifouling of poly(n-hydroxyethylacrylamide)/salicylate hydrogels. Langmuir ACS J. Surf. Colloids 2013, 29, 1517–1524. [Google Scholar]
  80. Zan, X.; Kozlov, M.; McCarthy, T.J.; Su, Z. Covalently attached, silver-doped poly(vinyl alcohol) hydrogel films on poly(l-lactic acid). Biomacromolecules 2010, 11, 1082–1088. [Google Scholar]
  81. Drago, L.; Boot, W.; Dimas, K.; Malizos, K.; Hansch, G.M.; Stuyck, J.; Gawlitta, D.; Romano, C.L. Does implant coating with antibacterial-loaded hydrogel reduce bacterial colonization and biofilm formation in vitro? Clin. Orthop. Relat. Res. 2014, 472, 3311–3323. [Google Scholar]
  82. Bock, R.M.; McEntire, B.J.; Bal, B.S.; Rahaman, M.N.; Boffelli, M.; Pezzotti, G. Surface modulation of silicon nitride ceramics for orthopaedic applications. Acta Biomater. 2015, 26, 318–330. [Google Scholar]
  83. Webster, T.J.; Patel, A.A.; Rahaman, M.N.; Sonny Bal, B. Anti-infective and osteointegration properties of silicon nitride, poly(ether ether ketone), and titanium implants. Acta Biomater. 2012, 8, 4447–4454. [Google Scholar] [PubMed]
  84. Eto, S.; Kawano, S.; Someya, S.; Miyamoto, H.; Sonohata, M.; Mawatari, M. First clinical experience with thermal-sprayed silver oxide-containing hydroxyapatite coating implant. J. Arthroplast. 2015. [Google Scholar] [CrossRef] [PubMed]
  85. Guimond-Lischer, S.; Ren, Q.; Braissant, O.; Gruner, P.; Wampfler, B.; Maniura-Weber, K. Vacuum plasma sprayed coatings using ionic silver doped hydroxyapatite powder to prevent bacterial infection of bone implants. Biointerphases 2016, 11, 011012. [Google Scholar] [CrossRef] [PubMed]
  86. Unosson, E.; Rodriguez, D.; Welch, K.; Engqvist, H. Reactive combinatorial synthesis and characterization of a gradient ag-ti oxide thin film with antibacterial properties. Acta Biomater. 2015, 11, 503–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Cheng, H.; Li, Y.; Huo, K.; Gao, B.; Xiong, W. Long-lasting in vivo and in vitro antibacterial ability of nanostructured titania coating incorporated with silver nanoparticles. J. Biomed. Mater. Res. A 2014, 102, 3488–3499. [Google Scholar] [CrossRef] [PubMed]
  88. Gao, A.; Hang, R.Q.; Huang, X.B.; Zhao, L.Z.; Zhang, X.Y.; Wang, L.; Tang, B.; Ma, S.L.; Chu, P.K. The effects of titania nanotubes with embedded silver oxide nanoparticles on bacteria and osteoblasts. Biomaterials 2014, 35, 4223–4235. [Google Scholar] [CrossRef] [PubMed]
  89. Mei, S.; Wang, H.; Wang, W.; Tong, L.; Pan, H.; Ruan, C.; Ma, Q.; Liu, M.; Yang, H.; Zhang, L.; et al. Antibacterial effects and biocompatibility of titanium surfaces with graded silver incorporation in titania nanotubes. Biomaterials 2014, 35, 4255–4265. [Google Scholar] [CrossRef] [PubMed]
  90. Dong, W.; Zhu, Y.; Zhang, J.; Lu, L.; Zhao, C.; Qin, L.; Li, Y. Investigation on the antibacterial micro-porous titanium with silver nano-particles. J. Nanosci. Nanotechnol. 2013, 13, 6782–6786. [Google Scholar] [CrossRef] [PubMed]
  91. Panacek, A.; Balzerova, A.; Prucek, R.; Ranc, V.; Vecerova, R.; Husickova, V.; Pechousek, J.; Filip, J.; Zboril, R.; Kvitek, L. Preparation, characterization and antimicrobial efficiency of ag/pdda-diatomite nanocomposite. Colloids Surf. B Biointerfaces 2013, 110, 191–198. [Google Scholar] [CrossRef] [PubMed]
  92. Kvitek, L.; Panacek, A.; Soukupova, J.; Kolar, M.; Vecerova, R.; Prucek, R.; Holecova, M.; Zboril, R. Effect of surfactants and polymers on stability and antibacterial activity of silver nanoparticles (nps). J. Phys. Chem. C 2008, 112, 5825–5834. [Google Scholar] [CrossRef]
  93. Knetsch, M.L.W.; Koole, L.H. New strategies in the development of antimicrobial coatings: The example of increasing usage of silver and silver nanoparticles. Polymers 2011, 3, 340–366. [Google Scholar] [CrossRef]
  94. Harrasser, N.; Jussen, S.; Banke, I.J.; Kmeth, R.; von Eisenhart-Rothe, R.; Stritzker, B.; Gollwitzer, H.; Burgkart, R. Antibacterial efficacy of titanium-containing alloy with silver-nanoparticles enriched diamond-like carbon coatings. AMB Express 2015, 5, 77. [Google Scholar] [CrossRef] [PubMed]
  95. Bai, Y.; Bai, Y.; Wang, C.; Gao, J.; Ma, W. Fabrication and characterization of gold nanoparticle-loaded tio2 nanotube arrays for medical implants. J. Mater. Sci. Mater. Med. 2016, 27, 31. [Google Scholar] [CrossRef] [PubMed]
  96. Ahmed, R.A.; Fadl-allah, S.A.; El-Bagoury, N.; El-Rab, S.M.F.G. Improvement of corrosion resistance and antibacterial effect of niti orthopedic materials by chitosan and gold nanoparticles. Appl. Surf. Sci. 2014, 292, 390–399. [Google Scholar] [CrossRef]
  97. Koseki, H.; Asahara, T.; Shida, T.; Yoda, I.; Horiuchi, H.; Baba, K.; Osaki, M. Clinical and histomorphometrical study on titanium dioxide-coated external fixation pins. Int. J. Nanomed. 2013, 8, 593–599. [Google Scholar] [CrossRef] [PubMed]
  98. Haenle, M.; Fritsche, A.; Zietz, C.; Bader, R.; Heidenau, F.; Mittelmeier, W.; Gollwitzer, H. An extended spectrum bactericidal titanium dioxide (tio2) coating for metallic implants: In vitro effectiveness against mrsa and mechanical properties. J. Mater. Sci. Mater. Med. 2011, 22, 381–387. [Google Scholar] [CrossRef] [PubMed]
  99. Yue, C.; Kuijer, R.; Kaper, H.J.; van der Mei, H.C.; Busscher, H.J. Simultaneous interaction of bacteria and tissue cells with photocatalytically activated, anodized titanium surfaces. Biomaterials 2014, 35, 2580–2587. [Google Scholar] [CrossRef] [PubMed]
  100. Holinka, J.; Pilz, M.; Kubista, B.; Presterl, E.; Windhager, R. Effects of selenium coating of orthopaedic implant surfaces on bacterial adherence and osteoblastic cell growth. Bone Jt. J. 2013, 95-B, 678–682. [Google Scholar] [CrossRef] [PubMed]
  101. Tran, P.A.; Webster, T.J. Selenium nanoparticles inhibit staphylococcus aureus growth. Int. J. Nanomed. 2011, 6, 1553–1558. [Google Scholar]
  102. Rodriguez-Valencia, C.; Lopez-Alvarez, M.; Cochon-Cores, B.; Pereiro, I.; Serra, J.; Gonzalez, P. Novel selenium-doped hydroxyapatite coatings for biomedical applications. J. Biomed. Mater. Res. A 2013, 101, 853–861. [Google Scholar] [CrossRef] [PubMed]
  103. LewisOscar, F.; MubarakAli, D.; Nithya, C.; Priyanka, R.; Gopinath, V.; Alharbi, N.S.; Thajuddin, N. One pot synthesis and anti-biofilm potential of copper nanoparticles (cunps) against clinical strains of pseudomonas aeruginosa. Biofouling 2015, 31, 379–391. [Google Scholar] [CrossRef] [PubMed]
  104. Hoene, A.; Prinz, C.; Walschus, U.; Lucke, S.; Patrzyk, M.; Wilhelm, L.; Neumann, H.G.; Schlosser, M. In vivo evaluation of copper release and acute local tissue reactions after implantation of copper-coated titanium implants in rats. Biomed. Mater. 2013, 8. [Google Scholar] [CrossRef] [PubMed]
  105. Elizabeth, E.; Baranwal, G.; Krishnan, A.G.; Menon, D.; Nair, M. Zno nanoparticle incorporated nanostructured metallic titanium for increased mesenchymal stem cell response and antibacterial activity. Nanotechnology 2014, 25, 115101. [Google Scholar] [CrossRef] [PubMed]
  106. Hu, H.; Zhang, W.; Qiao, Y.; Jiang, X.; Liu, X.; Ding, C. Antibacterial activity and increased bone marrow stem cell functions of zn-incorporated tio2 coatings on titanium. Acta Biomater. 2012, 8, 904–915. [Google Scholar] [CrossRef] [PubMed]
  107. Tsuchiya, H.; Shirai, T.; Nishida, H.; Murakami, H.; Kabata, T.; Yamamoto, N.; Watanabe, K.; Nakase, J. Innovative antimicrobial coating of titanium implants with iodine. J. Orthop. Sci. 2012, 17, 595–604. [Google Scholar] [CrossRef] [PubMed]
  108. Bellucci, D.; Sola, A.; Cannillo, V. Hydroxyapatite and tricalcium phosphate composites with bioactive glass as second phase: State of the art and current applications. J. Biomed. Mater. Res. A 2016, 104, 1030–1056. [Google Scholar] [CrossRef] [PubMed]
  109. Durgalakshmi, D.; Balakumar, S.; Raja, C.A.; George, R.P.; Mudali, U.K. Structural, morphological and antibacterial investigation of ag-impregnated sol-gel-derived 45s5 nanobioglass systems. J. Nanosci. Nanotechnol. 2015, 15, 4285–4295. [Google Scholar] [CrossRef] [PubMed]
  110. Shi, Y.Y.; Li, M.; Liu, Q.; Jia, Z.J.; Xu, X.C.; Cheng, Y.; Zheng, Y.F. Electrophoretic deposition of graphene oxide reinforced chitosan-hydroxyapatite nanocomposite coatings on ti substrate. J. Mater. Sci. Mater. Med. 2016, 27, 48. [Google Scholar] [CrossRef] [PubMed]
  111. Richtera, L.; Chudobova, D.; Cihalova, K.; Kremplova, M.; Milosavljevic, V.; Kopel, P.; Blazkova, I.; Hynek, D.; Adam, V.; Kizek, R. The composites of graphene oxide with metal or semimetal nanoparticles and their effect on pathogenic microorganisms. Materials 2015, 8, 2994–3011. [Google Scholar] [CrossRef]
  112. Antoci, V., Jr.; Adams, C.S.; Parvizi, J.; Ducheyne, P.; Shapiro, I.M.; Hickok, N.J. Covalently attached vancomycin provides a nanoscale antibacterial surface. Clin. Orthop. Relat. Res. 2007, 461, 81–87. [Google Scholar] [CrossRef] [PubMed]
  113. Antoci, V., Jr.; King, S.B.; Jose, B.; Parvizi, J.; Zeiger, A.R.; Wickstrom, E.; Freeman, T.A.; Composto, R.J.; Ducheyne, P.; Shapiro, I.M.; et al. Vancomycin covalently bonded to titanium alloy prevents bacterial colonization. J. Orthop. Res. 2007, 25, 858–866. [Google Scholar] [CrossRef] [PubMed]
  114. Walter, M.S.; Frank, M.J.; Satue, M.; Monjo, M.; Ronold, H.J.; Lyngstadaas, S.P.; Haugen, H.J. Bioactive implant surface with electrochemically bound doxycycline promotes bone formation markers in vitro and in vivo. Dent. Mater. 2014, 30, 200–214. [Google Scholar] [CrossRef] [PubMed]
  115. Chennell, P.; Feschet-Chassot, E.; Devers, T.; Awitor, K.O.; Descamps, S.; Sautou, V. In vitro evaluation of tio2 nanotubes as cefuroxime carriers on orthopaedic implants for the prevention of periprosthetic joint infections. Int. J. Pharm. 2013, 455, 298–305. [Google Scholar] [CrossRef] [PubMed]
  116. Hickok, N.J.; Shapiro, I.M. Immobilized antibiotics to prevent orthopaedic implant infections. Adv. Drug Deliv. Rev. 2012, 64, 1165–1176. [Google Scholar] [CrossRef] [PubMed]
  117. Norowski, P.A.; Courtney, H.S.; Babu, J.; Haggard, W.O.; Bumgardner, J.D. Chitosan coatings deliver antimicrobials from titanium implants: A preliminary study. Implant Dent. 2011, 20, 56–67. [Google Scholar] [CrossRef] [PubMed]
  118. Chen, X.N.; Gu, Y.X.; Lee, J.H.; Lee, W.Y.; Wang, H.J. Multifunctional surfaces with biomimetic nanofibres and drug-eluting micro-patterns for infection control and bone tissue formation. Eur. Cells Mater. 2012, 24, 237–248. [Google Scholar]
  119. Renoud, P.; Toury, B.; Benayoun, S.; Attik, G.; Grosgogeat, B. Functionalization of titanium with chitosan via silanation: Evaluation of biological and mechanical performances. PLoS ONE 2012, 7, e39367. [Google Scholar] [CrossRef] [PubMed]
  120. Tan, H.; Ma, R.; Lin, C.; Liu, Z.; Tang, T. Quaternized chitosan as an antimicrobial agent: Antimicrobial activity, mechanism of action and biomedical applications in orthopedics. Int. J. Mol. Sci. 2013, 14, 1854–1869. [Google Scholar] [CrossRef] [PubMed]
  121. Yazici, H.; O’Neill, M.B.; Kacar, T.; Wilson, B.R.; Oren, E.E.; Sarikaya, M.; Tamerler, C. Engineered chimeric peptides as antimicrobial surface coating agents toward infection-free implants. ACS Appl. Mater. Interfaces 2016, 8, 5070–5081. [Google Scholar] [CrossRef] [PubMed]
  122. Rapsch, K.; Bier, F.F.; Tadros, M.; von Nickisch-Rosenegk, M. Identification of antimicrobial peptides and immobilization strategy suitable for a covalent surface coating with biocompatible properties. Bioconj. Chem. 2014, 25, 308–319. [Google Scholar] [CrossRef] [PubMed]
  123. Zheng, D.; Neoh, K.G.; Shi, Z.; Kang, E.T. Assessment of stability of surface anchors for antibacterial coatings and immobilized growth factors on titanium. J. Colloid Interface Sci. 2013, 406, 238–246. [Google Scholar] [CrossRef] [PubMed]
  124. Li, B.; McKeague, A.L. Emerging ideas: Interleukin-12 nanocoatings prevent open fracture-associated infections. Clin. Orthop. Relat. Res. 2011, 469, 3262–3265. [Google Scholar] [CrossRef] [PubMed]
  125. Thallinger, B.; Prasetyo, E.N.; Nyanhongo, G.S.; Guebitz, G.M. Antimicrobial enzymes: An emerging strategy to fight microbes and microbial biofilms. Biotechnol. J. 2013, 8, 97–109. [Google Scholar] [CrossRef] [PubMed]
  126. Chua, P.H.; Neoh, K.G.; Kang, E.T.; Wang, W. Surface functionalization of titanium with hyaluronic acid/chitosan polyelectrolyte multilayers and rgd for promoting osteoblast functions and inhibiting bacterial adhesion. Biomaterials 2008, 29, 1412–1421. [Google Scholar] [CrossRef] [PubMed]
  127. Glinel, K.; Thebault, P.; Humblot, V.; Pradier, C.M.; Jouenne, T. Antibacterial surfaces developed from bio-inspired approaches. Acta Biomater. 2012, 8, 1670–1684. [Google Scholar] [CrossRef] [PubMed]
  128. He, T.; Chan, V. Covalent layer-by-layer assembly of polyethyleneimine multilayer for antibacterial applications. J. Biomed. Mater. Res. A 2010, 95, 454–464. [Google Scholar] [CrossRef] [PubMed]
  129. Fu, J.; Ji, J.; Fan, D.; Shen, J. Construction of antibacterial multilayer films containing nanosilver via layer-by-layer assembly of heparin and chitosan-silver ions complex. J. Biomed. Mater. Res. A 2006, 79, 665–674. [Google Scholar] [CrossRef] [PubMed]
  130. Zhou, B.; Li, Y.; Deng, H.; Hu, Y.; Li, B. Antibacterial multilayer films fabricated by layer-by-layer immobilizing lysozyme and gold nanoparticles on nanofibers. Colloids Surf. B Biointerfaces 2014, 116, 432–438. [Google Scholar] [CrossRef] [PubMed]
  131. Huang, W.; Li, X.; Xue, Y.; Huang, R.; Deng, H.; Ma, Z. Antibacterial multilayer films fabricated by lbl immobilizing lysozyme and htcc on nanofibrous mats. Int. J. Biol. Macromol. 2013, 53, 26–31. [Google Scholar] [CrossRef] [PubMed]
  132. Min, J.; Braatz, R.D.; Hammond, P.T. Tunable staged release of therapeutics from layer-by-layer coatings with clay interlayer barrier. Biomaterials 2014, 35, 2507–2517. [Google Scholar] [CrossRef] [PubMed]
  133. Rizzello, L.; Pompa, P.P. Nanosilver-based antibacterial drugs and devices: Mechanisms, methodological drawbacks, and guidelines. Chem. Soc. Rev. 2013. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, M.; Zhao, Y.; Yan, L.; Peltier, R.; Hui, W.; Yao, X.; Cui, Y.; Chen, X.; Sun, H.; Wang, Z. Interfacial engineering of bimetallic ag/pt nanoparticles on reduced graphene oxide matrix for enhanced antimicrobial activity. ACS Appl. Mater. Interfaces 2016, 8, 8834–8840. [Google Scholar] [CrossRef] [PubMed]
  135. Funao, H.; Nagai, S.; Sasaki, A.; Hoshikawa, T.; Tsuji, T.; Okada, Y.; Koyasu, S.; Toyama, Y.; Nakamura, M.; Aizawa, M.; et al. A novel hydroxyapatite film coated with ionic silver via inositol hexaphosphate chelation prevents implant-associated infection. Sci. Rep. 2016, 6, 23238. [Google Scholar] [CrossRef] [PubMed]
  136. Gottenbos, B.; van der Mei, H.C.; Klatter, F.; Grijpma, D.W.; Feijen, J.; Nieuwenhuis, P.; Busscher, H.J. Positively charged biomaterials exert antimicrobial effects on gram-negative bacilli in rats. Biomaterials 2003, 24, 2707–2710. [Google Scholar] [CrossRef]
  137. Braem, A.; De Cremer, K.; Delattin, N.; De Brucker, K.; Neirinck, B.; Vandamme, K.; Martens, J.A.; Michiels, J.; Vleugels, J.; Cammue, B.P.; et al. Novel anti-infective implant substrates: Controlled release of antibiofilm compounds from mesoporous silica-containing macroporous titanium. Colloids Surf. B Biointerfaces 2015, 126, 481–488. [Google Scholar] [CrossRef] [PubMed]
  138. Yu, Q.; Cho, J.; Shivapooja, P.; Ista, L.K.; Lopez, G.P. Nanopatterned smart polymer surfaces for controlled attachment, killing, and release of bacteria. ACS Appl. Mater. Interfaces 2013, 5, 9295–9304. [Google Scholar] [CrossRef] [PubMed]
  139. Holzapfel, B.M.; Reichert, J.C.; Schantz, J.T.; Gbureck, U.; Rackwitz, L.; Noth, U.; Jakob, F.; Rudert, M.; Groll, J.; Hutmacher, D.W. How smart do biomaterials need to be? A translational science and clinical point of view. Adv. Drug Deliv. Rev. 2013, 65, 581–603. [Google Scholar] [CrossRef] [PubMed]
  140. Cipriano, A.F.; Miller, C.; Liu, H. Anodic growth and biomedical applications of tio2 nanotubes. J. Biomed. Nanotechnol. 2014, 10, 2977–3003. [Google Scholar] [CrossRef] [PubMed]
  141. Parvizi, J.; Antoci, V., Jr.; Hickok, N.J.; Shapiro, I.M. Selfprotective smart orthopedic implants. Expert Rev. Med. Devices 2007, 4, 55–64. [Google Scholar] [CrossRef] [PubMed]
  142. Mastronardi, E.; Foster, A.; Zhang, X.; Derosa, M.C. Smart materials based on DNA aptamers: Taking aptasensing to the next level. Sensors 2014, 14, 3156–3171. [Google Scholar] [CrossRef] [PubMed]
  143. Ehrlich, G.D.; Stoodley, P.; Kathju, S.; Zhao, Y.; McLeod, B.R.; Balaban, N.; Hu, F.Z.; Sotereanos, N.G.; Costerton, J.W.; Stewart, P.S.; et al. Engineering approaches for the detection and control of orthopaedic biofilm infections. Clin. Orthop. Relat.Res. 2005, 437, 59–66. [Google Scholar] [CrossRef] [PubMed]
  144. Shchukin, D.G.; Mohwald, H. Self-repairing coatings containing active nanoreservoirs. Small 2007, 3, 926–943. [Google Scholar] [CrossRef] [PubMed]
  145. Shchukin, D.; Mohwald, H. Materials science. A coat of many functions. Science 2013, 341, 1458–1459. [Google Scholar] [CrossRef] [PubMed]
  146. Keller, L.; Wagner, Q.; Offner, D.; Eap, S.; Musset, A.M.; Arruebo, M.; Kelm, J.M.; Schwinte, P.; Benkirane-Jessel, N. Integrating microtissues in nanofiber scaffolds for regenerative nanomedicine. Materials 2015, 8, 6863–6867. [Google Scholar] [CrossRef]
  147. Yilmaz, C.; Colak, M.; Yilmaz, B.C.; Ersoz, G.; Kutateladze, M.; Gozlugol, M. Bacteriophage therapy in implant-related infections: An experimental study. J. Bone Jt. Surg. Am. Vol. 2013, 95, 117–125. [Google Scholar] [CrossRef] [PubMed]
  148. Liu, Y.; Busscher, H.J.; Zhao, B.; Li, Y.; Zhang, Z.; van der Mei, H.C.; Ren, Y.; Shi, L. Surface-adaptive, antimicrobially loaded, micellar nanocarriers with enhanced penetration and killing efficiency in staphylococcal biofilms. ACS Nano 2016. [Google Scholar] [CrossRef] [PubMed]
  149. Yu, Q.; Wu, Z.; Chen, H. Dual-function antibacterial surfaces for biomedical applications. Acta Biomater. 2015, 16, 1–13. [Google Scholar] [CrossRef] [PubMed]
  150. Zaborowska, M.; Welch, K.; Branemark, R.; Khalilpour, P.; Engqvist, H.; Thomsen, P.; Trobos, M. Bacteria-material surface interactions: Methodological development for the assessment of implant surface induced antibacterial effects. J. Biomed. Mater. Res. B Appl. Biomater. 2014. [Google Scholar] [CrossRef] [PubMed]
  151. Lemire, J.A.; Harrison, J.J.; Turner, R.J. Antimicrobial activity of metals: Mechanisms, molecular targets and applications. Nat. Rev. Microbiol. 2013, 11, 371–384. [Google Scholar] [CrossRef] [PubMed]
  152. Ketabchi, A.; Komm, K.; Miles-Rossouw, M.; Cassani, D.A.; Variola, F. Nanoporous titanium surfaces for sustained elution of proteins and antibiotics. PLoS ONE 2014, 9, e92080. [Google Scholar] [CrossRef] [PubMed]
  153. Hizal, F.; Zhuk, I.; Sukhishvili, S.; Busscher, H.J.; van der Mei, H.C.; Choi, C.H. Impact of 3d hierarchical nanostructures on the antibacterial efficacy of a bacteria-triggered self-defensive antibiotic coating. ACS Appl. Mater. Interfaces 2015, 7, 20304–20313. [Google Scholar] [CrossRef] [PubMed]
  154. Tian, B.; Chen, W.; Yu, D.; Lei, Y.; Ke, Q.; Guo, Y.; Zhu, Z. Fabrication of silver nanoparticle-doped hydroxyapatite coatings with oriented block arrays for enhancing bactericidal effect and osteoinductivity. J. Mech. Behav. Biomed. Mater. 2016, 61, 345–359. [Google Scholar] [CrossRef] [PubMed]
  155. Gallo, J.; Holinka, M.; Moucha, C.S. Antibacterial surface treatment for orthopaedic implants. Int. J. Mol. Sci. 2014, 15, 13849–13880. [Google Scholar] [CrossRef] [PubMed]
  156. Baker, C.; Pradhan, A.; Pakstis, L.; Pochan, D.J.; Shah, S.I. Synthesis and antibacterial properties of silver nanoparticles. J. Nanosci. Nanotechnol. 2005, 5, 244–249. [Google Scholar] [CrossRef] [PubMed]
  157. Chen, C.Y.; Chiang, C.L. Preparation of cotton fibers with antibacterial silver nanoparticles. Mater. Lett. 2008, 62, 3607–3609. [Google Scholar] [CrossRef]
  158. Martinez-Castanon, G.A.; Nino-Martinez, N.; Martinez-Gutierrez, F.; Martinez-Mendoza, J.R.; Ruiz, F. Synthesis and antibacterial activity of silver nanoparticles with different sizes. J. Nanopart. Res. 2008, 10, 1343–1348. [Google Scholar] [CrossRef]
  159. Pal, S.; Tak, Y.K.; Song, J.M. Does the antibacterial activity of silver nanoparticles depend on the shape of the nanoparticle? A study of the gram-negative bacterium escherichia coli. Appl. Environ. Microbiol. 2007, 73, 1712–1720. [Google Scholar] [CrossRef] [PubMed]
  160. Panacek, A.; Kvitek, L.; Prucek, R.; Kolar, M.; Vecerova, R.; Pizurova, N.; Sharma, V.K.; Nevecna, T.; Zboril, R. Silver colloid nanoparticles: Synthesis, characterization, and their antibacterial activity. J. Phys. Chem. B 2006, 110, 16248–16253. [Google Scholar] [CrossRef] [PubMed]
  161. Sharma, V.K.; Yngard, R.A.; Lin, Y. Silver nanoparticles: Green synthesis and their antimicrobial activities. Adv. Colloid Interface Sci. 2009, 145, 83–96. [Google Scholar] [CrossRef] [PubMed]
  162. Ansari, M.A.; Khan, H.M.; Khan, A.A.; Cameotra, S.S.; Saquib, Q.; Musarrat, J. Gum arabic capped-silver nanoparticles inhibit biofilm formation by multi-drug resistant strains of pseudomonas aeruginosa. J. Basic Microbiol. 2014, 54, 688–699. [Google Scholar] [CrossRef] [PubMed]
  163. AshaRani, P.V.; Mun, G.L.K.; Hande, M.P.; Valiyaveettil, S. Cytotoxicity and genotoxicity of silver nanoparticles in human cells. Acs Nano 2009, 3, 279–290. [Google Scholar] [CrossRef] [PubMed]
  164. Carlson, C.; Hussain, S.M.; Schrand, A.M.; Braydich-Stolle, L.K.; Hess, K.L.; Jones, R.L.; Schlager, J.J. Unique cellular interaction of silver nanoparticles: Size-dependent generation of reactive oxygen species. J. Phys. Chem. B 2008, 112, 13608–13619. [Google Scholar] [CrossRef] [PubMed]
  165. Hussain, S.M.; Hess, K.L.; Gearhart, J.M.; Geiss, K.T.; Schlager, J.J. In vitro toxicity of nanoparticles in brl 3a rat liver cells. In Proceedings of the 13th International Workshop on in Vitro Toxicology, Zegrze, Poland, 8–11 September 2004; pp. 975–983.
  166. Afkhami, F.; Pourhashemi, S.J.; Sadegha, M.; Salehi, Y.; Fard, M.J.K. Antibiofilm efficacy of silver nanoparticles as a vehicle for calcium hydroxide medicament against enterococcus faecalis. J. Dent. 2015, 43, 1573–1579. [Google Scholar] [CrossRef] [PubMed]
  167. Agarwala, M.; Barman, T.; Gogoi, D.; Choudhury, B.; Pal, A.R.; Yadav, R.N.S. Highly effective antibiofilm coating of silver-polymer nanocomposite on polymeric medical devices deposited by one step plasma process. J. Biomed. Mater. Res. B Appl. Biomater. 2014, 102, 1223–1235. [Google Scholar] [CrossRef] [PubMed]
  168. Alberto Perez-Diaz, M.; Boegli, L.; James, G.; Velasquillo, C.; Sanchez-Sanchez, R.; Martinez-Martinez, R.-E.; Alejandro Martinez-Castanon, G.; Martinez-Gutierrez, F. Silver nanoparticles with antimicrobial activities against streptococcus mutans and their cytotoxic effect. Mater. Sci. Eng. C Mater. Biol. Appl. 2015, 55, 360–366. [Google Scholar] [CrossRef] [PubMed]
  169. Ali, K.; Ahmed, B.; Dwivedi, S.; Saquib, Q.; Al-Khedhairy, A.A.; Musarrat, J. Microwave accelerated green synthesis of stable silver nanoparticles with eucalyptus globulus leaf extract and their antibacterial and antibiofilm activity on clinical isolates. PLoS ONE 2015, 10, e0131178. [Google Scholar] [CrossRef] [PubMed]
  170. Besinis, A.; De Peralta, T.; Handy, R.D. Inhibition of biofilm formation and antibacterial properties of a silver nano-coating on human dentine. Nanotoxicology 2014, 8, 745–754. [Google Scholar] [CrossRef] [PubMed]
  171. Fufa, O.; Andronescu, E.; Grumezescu, V.; Holban, A.M.; Mogoanta, L.; Mogosanu, G.D.; Socol, G.; Iordache, F.; Chifiriuc, M.C.; Grumezescu, A.M. Silver nanostructurated surfaces prepared by maple for biofilm prevention. Biointerface Res. Appl. Chem. 2015, 5, 1011–1017. [Google Scholar]
  172. Ghosh, S.; Jagtap, S.; More, P.; Shete, U.J.; Maheshwari, N.O.; Rao, S.J.; Kitture, R.; Kale, S.; Bellare, J.; Patil, S.; et al. Dioscorea bulbifera mediated synthesis of novel aucoreagshell nanoparticles with potent antibiofilm and antileishmanial activity. J. Nanomater. 2015. [Google Scholar] [CrossRef]
  173. Jaiswal, S.; Bhattacharya, K.; McHale, P.; Duffy, B. Dual effects of beta-cyclodextrin-stabilised silver nanoparticles: Enhanced biofilm inhibition and reduced cytotoxicity. J. Mater. Sci. Mater. Med. 2015, 26, 5367. [Google Scholar] [CrossRef] [PubMed]
  174. Palanisamy, N.K.; Ferina, N.; Amirulhusni, A.N.; Mohd-Zain, Z.; Hussaini, J.; Ping, L.J.; Durairaj, R. Antibiofilm properties of chemically synthesized silver nanoparticles found against pseudomonas aeruginosa. J. Nanobiotechnol. 2014, 12, 2. [Google Scholar] [CrossRef] [PubMed]
  175. Rajiv, S.; Drilling, A.; Bassiouni, A.; James, C.; Vreugde, S.; Wormald, P.-J. Topical colloidal silver as an anti-biofilm agent in a staphylococcus aureus chronic rhinosinusitis sheep model. Int. Forum Allergy Rhinol. 2015, 5, 283–288. [Google Scholar] [CrossRef] [PubMed]
  176. Taglietti, A.; Arciola, C.R.; D’Agostino, A.; Dacarro, G.; Montanaro, L.; Campoccia, D.; Cucca, L.; Vercellino, M.; Poggi, A.; Pallavicini, P.; et al. Antibiofilm activity of a monolayer of silver nanoparticles anchored to an amino-silanized glass surface. Biomaterials 2014, 35, 1779–1788. [Google Scholar] [CrossRef] [PubMed]
  177. Taraszkiewicz, A.; Fila, G.; Grinholc, M.; Nakonieczna, J. Innovative strategies to overcome biofilm resistance. Biomed. Res. Int. 2013. [Google Scholar] [CrossRef] [PubMed]
  178. Thomas, R.; Soumya, K.R.; Mathew, J.; Radhakrishnan, E.K. Inhibitory effect of silver nanoparticle fabricated urinary catheter on colonization efficiency of coagulase negative staphylococci. J. Photochem. Photobiol. B Biol. 2015, 149, 68–77. [Google Scholar] [CrossRef] [PubMed]
  179. Wu, D.; Fan, W.; Kishen, A.; Gutmann, J.L.; Fan, B. Evaluation of the antibacterial efficacy of silver nanoparticles against enterococcus faecalis biofilm. J. Endod. 2014, 40, 285–290. [Google Scholar] [CrossRef] [PubMed]
  180. Xie, C.M.; Lu, X.; Wang, K.F.; Meng, F.Z.; Jiang, O.; Zhang, H.P.; Zhi, W.; Fang, L.M. Silver nanoparticles and growth factors incorporated hydroxyapatite coatings on metallic implant surfaces for enhancement of osteoinductivity and antibacterial properties. ACS Appl. Mater. Interfaces 2014, 6, 8580–8589. [Google Scholar] [CrossRef] [PubMed]
  181. Rodriguez-Cano, A.; Pacha-Olivenza, M.A.; Babiano, R.; Cintas, P.; Gonzalez-Martin, M.L. Non-covalent derivatization of aminosilanized titanium alloy implants silver-enhanced coating of antibacterial organics. Surf. Coat. Technol. 2014, 245, 66–73. [Google Scholar] [CrossRef]
  182. Zhao, C.J.; Feng, B.; Li, Y.T.; Tan, J.; Lu, X.; Weng, J. Preparation and antibacterial activity of titanium nanotubes loaded with ag nanoparticles in the dark and under the uv light. Appl. Surf. Sci. 2013, 280, 8–14. [Google Scholar] [CrossRef]
  183. Wang, Z.; Sun, Y.; Wang, D.; Liu, H.; Boughton, R.I. In situ fabrication of silver nanoparticle-filled hydrogen titanate nanotube layer on metallic titanium surface for bacteriostatic and biocompatible implantation. Int. J. Nanomed. 2013, 8, 2903–2916. [Google Scholar]
  184. Saidin, S.; Chevallier, P.; Kadir, M.R.A.; Hermawan, H.; Mantovani, D. Polydopamine as an intermediate layer for silver and hydroxyapatite immobilisation on metallic biomaterials surface. Mater. Sci. Eng. C Mater. Biol. Appl. 2013, 33, 4715–4724. [Google Scholar] [CrossRef] [PubMed]
  185. De Giglio, E.; Cafagna, D.; Cometa, S.; Allegretta, A.; Pedico, A.; Giannossa, L.C.; Sabbatini, L.; Mattioli-Belmonte, M.; Iatta, R. An innovative, easily fabricated, silver nanoparticle-based titanium implant coating: Development and analytical characterization. Anal. Bioanal. Chem. 2013, 405, 805–816. [Google Scholar] [CrossRef] [PubMed]
  186. Secinti, K.D.; Ozalp, H.; Attar, A.; Sargon, M.F. Nanoparticle silver ion coatings inhibit biofilm formation on titanium implants. J. Clin. Neurosci. 2011, 18, 391–395. [Google Scholar] [CrossRef] [PubMed]
  187. Cao, H.L.; Liu, X.Y.; Meng, F.H.; Chu, P.K. Biological actions of silver nanoparticles embedded in titanium controlled by micro-galvanic effects. Biomaterials 2011, 32, 693–705. [Google Scholar] [CrossRef] [PubMed]
  188. Della Valle, C.; Visai, L.; Santin, M.; Cigada, A.; Candiani, G.; Pezzoli, D.; Arciola, C.R.; Imbriani, M.; Chiesa, R. A novel antibacterial modification treatment of titanium capable to improve osseointegration. Int. J. Artif. Organs 2012, 35, 864–875. [Google Scholar] [CrossRef] [PubMed]
  189. Ionita, D.; Grecu, M.; Ungureanu, C.; Demetrescu, I. Antimicrobial activity of the surface coatings on tialzr implant biomaterial. J. Biosci. Bioeng. 2011, 112, 630–634. [Google Scholar] [CrossRef] [PubMed]
  190. Brennan, S.A.; Ni Fhoghlu, C.; Devitt, B.M.; O’Mahony, F.J.; Brabazon, D.; Walsh, A. Silver nanoparticles and their orthopaedic applications. Bone Jt. J. 2015, 97-B, 582–589. [Google Scholar] [CrossRef] [PubMed]
  191. Saleh, N.B.; Chambers, B.; Aich, N.; Plazas-Tuttle, J.; Phung-Ngoc, H.N.; Kirisits, M.J. Mechanistic lessons learned from studies of planktonic bacteria with metallic nanomaterials: Implications for interactions between nanomaterials and biofilm bacteria. Front. Microbiol. 2015, 6, 677. [Google Scholar] [CrossRef] [PubMed]
  192. Zhong, X.; Song, Y.J.; Yang, P.; Wang, Y.; Jiang, S.Y.; Zhang, X.; Li, C.Y. Titanium surface priming with phase-transited lysozyme to establish a silver nanoparticle-loaded chitosan/hyaluronic acid antibacterial multilayer via layer-by-layer self-assembly. PLoS ONE 2016, 11, e0146957. [Google Scholar] [CrossRef] [PubMed]
  193. Fabrega, J.; Renshaw, J.C.; Lead, J.R. Interactions of silver nanoparticles with pseudomonas putida biofilms. Environ. Sci. Technol. 2009, 43, 9004–9009. [Google Scholar] [CrossRef] [PubMed]
  194. Choi, O.Y.; Yu, C.P.; Fernandez, G.E.; Hu, Z.Q. Interactions of nanosilver with Escherichia coli cells in planktonic and biofilm cultures. Water Res. 2010, 44, 6095–6103. [Google Scholar] [CrossRef] [PubMed]
  195. Mohanty, S.; Mishra, S.; Jena, P.; Jacob, B.; Sarkar, B.; Sonawane, A. An investigation on the antibacterial, cytotoxic, and antibiofilm efficacy of starch-stabilized silver nanoparticles. Nanomed. Nanotechnol. 2012, 8, 916–924. [Google Scholar] [CrossRef] [PubMed]
  196. Peulen, T.O.; Wilkinson, K.J. Diffusion of nanoparticles in a biofilm. Environ. Sci. Technol. 2011, 45, 3367–3373. [Google Scholar] [CrossRef] [PubMed]
  197. Qin, H.; Cao, H.L.; Zhao, Y.C.; Zhu, C.; Cheng, T.; Wang, Q.J.; Peng, X.C.; Cheng, M.Q.; Wang, J.X.; Jin, G.D.; et al. In vitro and in vivo anti-biofilm effects of silver nanoparticles immobilized on titanium. Biomaterials 2014, 35, 9114–9125. [Google Scholar] [CrossRef] [PubMed]
  198. Bai, L.; Hang, R.Q.; Gao, A.; Zhang, X.Y.; Huang, X.B.; Wang, Y.Y.; Tang, B.; Zhao, L.Z.; Chu, P.K. Nanostructured titanium-silver coatings with good antibacterial activity and cytocompatibility fabricated by one-step magnetron sputtering. Appl. Surf. Sci. 2015, 355, 32–44. [Google Scholar] [CrossRef]
  199. Zhang, X.M.; Li, Z.Y.; Yuan, X.B.; Cui, Z.D.; Bao, H.J.; Li, X.; Liu, Y.D.; Yang, X.J. Cytotoxicity and antibacterial property of titanium alloy coated with silver nanoparticle-containing polyelectrolyte multilayer. Mater. Sci. Eng. C Mater. Biol. Appl. 2013, 33, 2816–2820. [Google Scholar] [CrossRef] [PubMed]
  200. Esfandiari, N.; Simchi, A.; Bagheri, R. Size tuning of ag-decorated TiO2 nanotube arrays for improved bactericidal capacity of orthopedic implants. J. Biomed. Mater. Res. A 2014, 102, 2625–2635. [Google Scholar] [CrossRef] [PubMed]
  201. Chen, X.Y.; Cai, K.Y.; Fang, J.J.; Lai, M.; Li, J.H.; Hou, Y.H.; Luo, Z.; Hu, Y.; Tang, L.L. Dual action antibacterial TiO2 nanotubes incorporated with silver nanoparticles and coated with a quaternary ammonium salt (QAS). Surf. Coat. Technol. 2013, 216, 158–165. [Google Scholar] [CrossRef]
  202. Li, W.R.; Xie, X.B.; Shi, Q.S.; Duan, S.S.; Ouyang, Y.S.; Chen, Y.B. Antibacterial effect of silver nanoparticles on staphylococcus aureus. Biometals 2011, 24, 135–141. [Google Scholar] [CrossRef] [PubMed]
  203. Li, W.R.; Xie, X.B.; Shi, Q.S.; Zeng, H.Y.; Ou-Yang, Y.S.; Chen, Y.B. Antibacterial activity and mechanism of silver nanoparticles on Escherichia coli. Appl. Microbiol. Biotechnol. 2010, 85, 1115–1122. [Google Scholar] [CrossRef] [PubMed]
  204. Cui, L.; Chen, P.Y.; Chen, S.D.; Yuan, Z.H.; Yu, C.P.; Ren, B.; Zhang, K.S. In situ study of the antibacterial activity and mechanism of action of silver nanoparticles by surface-enhanced raman spectroscopy. Anal. Chem. 2013, 85, 5436–5443. [Google Scholar] [CrossRef] [PubMed]
  205. Lara, H.H.; Ayala-Nunez, N.V.; Turrent, L.D.I.; Padilla, C.R. Bactericidal effect of silver nanoparticles against multidrug-resistant bacteria. World J. Microbiol. Biotechnol. 2010, 26, 615–621. [Google Scholar] [CrossRef]
  206. Lok, C.N.; Ho, C.M.; Chen, R.; He, Q.Y.; Yu, W.Y.; Sun, H.Z.; Tam, P.K.H.; Chiu, J.F.; Che, C.M. Proteomic analysis of the mode of antibacterial action of silver nanoparticles. J. Proteome Res. 2006, 5, 916–924. [Google Scholar] [CrossRef] [PubMed]
  207. Sondi, I.; Salopek-Sondi, B. Silver nanoparticles as antimicrobial agent: A case study on E-coli as a model for gram-negative bacteria. J. Colloid Interface Sci. 2004, 275, 177–182. [Google Scholar] [CrossRef] [PubMed]
  208. Choi, O.; Hu, Z.Q. Size dependent and reactive oxygen species related nanosilver toxicity to nitrifying bacteria. Environ. Sci. Technol. 2008, 42, 4583–4588. [Google Scholar] [CrossRef] [PubMed]
  209. Kim, J.S.; Kuk, E.; Yu, K.N.; Kim, J.H.; Park, S.J.; Lee, H.J.; Kim, S.H.; Park, Y.K.; Park, Y.H.; Hwang, C.Y.; et al. Antimicrobial effects of silver nanoparticles. Nanomed. Nanotechnol. 2007, 3, 95–101. [Google Scholar] [CrossRef] [PubMed]
  210. Xu, H.Y.; Qu, F.; Xu, H.; Lai, W.H.; Wang, Y.A.; Aguilar, Z.P.; Wei, H. Role of reactive oxygen species in the antibacterial mechanism of silver nanoparticles on Escherichia coli o157:H7. Biometals 2012, 25, 45–53. [Google Scholar] [CrossRef] [PubMed]
  211. Cabiscol, E.; Tamarit, J.; Ros, J. Oxidative stress in bacteria and protein damage by reactive oxygen species. Int. Microbiol. 2000, 3, 3–8. [Google Scholar] [PubMed]
  212. Silver, S. Bacterial silver resistance: Molecular biology and uses and misuses of silver compounds. FEMS Microbiol. Rev. 2003, 27, 341–353. [Google Scholar] [CrossRef]
  213. Silver, S.; Phung, L.T.; Silver, G. Silver as biocides in burn and wound dressings and bacterial resistance to silver compounds. J. Ind. Microbiol. Biotechnol. 2006, 33, 627–634. [Google Scholar] [CrossRef] [PubMed]
  214. Kremer, A.N.; Hoffmann, H. Subtractive hybridization yields a silver resistance determinant unique to nosocomial pathogens in the enterobacter cloacae complex. J. Clin. Microbiol. 2012, 50, 3249–3257. [Google Scholar] [CrossRef] [PubMed]
  215. Haefeli, C.; Franklin, C.; Hardy, K. Plasmid-determined silver resistance in pseudomonas-stutzeri isolated from a silver mine. J. Bacteriol. 1984, 158, 389–392. [Google Scholar] [PubMed]
  216. Gupta, A.; Matsui, K.; Lo, J.F.; Silver, S. Molecular basis for resistance to silver cations in salmonella. Nat. Med. 1999, 5, 183–188. [Google Scholar] [CrossRef] [PubMed]
  217. Li, X.Z.; Nikaido, H.; Williams, K.E. Silver-resistant mutants of Escherichia coli display active efflux of ag+ and are deficient in porins. J. Bacteriol. 1997, 179, 6127–6132. [Google Scholar] [PubMed]
  218. Nies, D.H. Efflux-mediated heavy metal resistance in prokaryotes. FEMS Microbiol. Rev. 2003, 27, 313–339. [Google Scholar] [CrossRef]
  219. Potara, M.; Jakab, E.; Damert, A.; Popescu, O.; Canpean, V.; Astilean, S. Synergistic antibacterial activity of chitosan-silver nanocomposites on staphylococcus aureus. Nanotechnology 2011, 22, 13. [Google Scholar] [CrossRef] [PubMed]
  220. Vertelov, G.K.; Krutyakov, Y.A.; Efremenkova, O.V.; Olenin, A.Y.; Lisichkin, G.V. A versatile synthesis of highly bactericidal myramistin (r) stabilized silver nanoparticles. Nanotechnology 2008, 19, 35. [Google Scholar] [CrossRef] [PubMed]
  221. Ammons, M.C.B.; Ward, L.S.; James, G.A. Anti-biofilm efficacy of a lactoferrin/xylitol wound hydrogel used in combination with silver wound dressings. Int. Wound J. 2011, 8, 268–273. [Google Scholar] [CrossRef] [PubMed]
  222. Ruden, S.; Hilpert, K.; Berditsch, M.; Wadhwani, P.; Ulrich, A.S. Synergistic interaction between silver nanoparticles and membrane-permeabilizing antimicrobial peptides. Antimicrob. Agents Chemother. 2009, 53, 3538–3540. [Google Scholar] [CrossRef] [PubMed]
  223. Birla, S.S.; Tiwari, V.V.; Gade, A.K.; Ingle, A.P.; Yadav, A.P.; Rai, M.K. Fabrication of silver nanoparticles by phoma glomerata and its combined effect against Escherichia coli, pseudomonas aeruginosa and staphylococcus aureus. Lett. Appl. Microbiol. 2009, 48, 173–179. [Google Scholar] [CrossRef] [PubMed]
  224. Fayaz, A.M.; Balaji, K.; Girilal, M.; Yadav, R.; Kalaichelvan, P.T.; Venketesan, R. Biogenic synthesis of silver nanoparticles and their synergistic effect with antibiotics: A study against gram-positive and gram-negative bacteria. Nanomed. Nanotechnol. 2010, 6, 103–109. [Google Scholar] [CrossRef] [PubMed]
  225. Ghosh, S.; Patil, S.; Ahire, M.; Kitture, R.; Kale, S.; Pardesi, K.; Cameotra, S.S.; Bellare, J.; Dhavale, D.D.; Jabgunde, A.; et al. Synthesis of silver nanoparticles using dioscorea bulbifera tuber extract and evaluation of its synergistic potential in combination with antimicrobial agents. Int. J. Nanomed. 2012, 7, 483–496. [Google Scholar]
  226. Muhsin, T.M.; Hachim, A.K. Mycosynthesis and characterization of silver nanoparticles and their activity against some human pathogenic bacteria. World J. Microbiol. Biotechnol. 2014, 30, 2081–2090. [Google Scholar] [CrossRef] [PubMed]
  227. Naqvi, S.Z.H.; Kiran, U.; Ali, M.I.; Jamal, A.; Hameed, A.; Ahmed, S.; Ali, N. Combined efficacy of biologically synthesized silver nanoparticles and different antibiotics against multidrug-resistant bacteria. Int. J. Nanomed. 2013, 8, 3187–3195. [Google Scholar] [CrossRef] [PubMed]
  228. Sathiyanarayanan, G.; Kiran, G.S.; Selvin, J. Synthesis of silver nanoparticles by polysaccharide bioflocculant produced from marine bacillus subtilis MSBN17. Colloid Surface B 2013, 102, 13–20. [Google Scholar] [CrossRef] [PubMed]
  229. Shahverdi, A.R.; Fakhimi, A.; Shahverdi, H.R.; Minaian, S. Synthesis and effect of silver nanoparticles on the antibacterial activity of different antibiotics against staphylococcus aureus and Escherichia coli. Nanomed. Nanotechnol. 2007, 3, 168–171. [Google Scholar] [CrossRef] [PubMed]
  230. Hwang, I.S.; Hwang, J.H.; Choi, H.; Kim, K.J.; Lee, D.G. Synergistic effects between silver nanoparticles and antibiotics and the mechanisms involved. J. Med. Microbiol. 2012, 61, 1719–1726. [Google Scholar] [CrossRef] [PubMed]
  231. Li, P.; Li, J.; Wu, C.Z.; Wu, Q.S.; Li, J. Synergistic antibacterial effects of beta-lactam antibiotic combined with silver nanoparticles. Nanotechnology 2005, 16, 1912–1917. [Google Scholar] [CrossRef]
  232. Markowska, K.; Grudniak, A.M.; Krawczyk, K.; Wrobel, I.; Wolska, K.I. Modulation of antibiotic resistance and induction of a stress response in pseudomonas aeruginosa by silver nanoparticles. J. Med. Microbiol. 2014, 63, 849–854. [Google Scholar] [CrossRef] [PubMed]
  233. Singh, R.; Wagh, P.; Wadhwani, S.; Gaidhani, S.; Kumbhar, A.; Bellare, J.; Chopade, B.A. Synthesis, optimization, and characterization of silver nanoparticles from acinetobacter calcoaceticus and their enhanced antibacterial activity when combined with antibiotics. Int. J. Nanomed. 2013, 8, 4277–4289. [Google Scholar]
  234. Brown, A.N.; Smith, K.; Samuels, T.A.; Lu, J.R.; Obare, S.O.; Scott, M.E. Nanoparticles functionalized with ampicillin destroy multiple-antibiotic-resistant isolates of pseudomonas aeruginosa and enterobacter aerogenes and methicillin-resistant staphylococcus aureus. Appl. Environ. Microbiol. 2012, 78, 2768–2774. [Google Scholar] [CrossRef] [PubMed]
  235. Smekalova, M.; Aragon, V.; Panacek, A.; Prucek, R.; Zboril, R.; Kvitek, L. Enhanced antibacterial effect of antibiotics in combination with silver nanoparticles against animal pathogens. Vet. J. 2016, 209, 174–179. [Google Scholar] [CrossRef] [PubMed]
  236. Panacek, A.; Smekalova, M.; Kilianova, M.; Prucek, R.; Bogdanova, K.; Vecerova, R.; Kolar, M.; Havrdova, M.; Plaza, G.A.; Chojniak, J.; et al. Strong and nonspecific synergistic antibacterial efficiency of antibiotics combined with silver nanoparticles at very low concentrations showing no cytotoxic effect. Molecules 2015, 21. [Google Scholar] [CrossRef] [PubMed]
  237. Panáček, A.; Smékalová, M.; Večeřová, R.; Bogdanová, K.; Röderová, M.; Kolář, M.; Kilianová, M.; Hradilová, Š.; Froning, J.P.; Havrdová, M.; et al. Silver nanoparticles strongly enhance and restore bactericidal activity of inactive antibiotics against multiresistant enterobacteriaceae. Colloids Surfaces B Biointerfaces 2016, 142, 392–399. [Google Scholar] [CrossRef] [PubMed]
  238. Franci, G.; Falanga, A.; Galdiero, S.; Palomba, L.; Rai, M.; Morelli, G.; Galdiero, M. Silver nanoparticles as potential antibacterial agents. Molecules 2015, 20, 8856–8874. [Google Scholar] [CrossRef] [PubMed]
  239. Arora, S.; Jain, J.; Rajwade, J.M.; Paknikar, K.M. Interactions of silver nanoparticles with primary mouse fibroblasts and liver cells. Toxicol. Appl. Pharmacol. 2009, 236, 310–318. [Google Scholar] [CrossRef] [PubMed]
  240. Scavone, M.; Armentano, I.; Fortunati, E.; Cristofaro, F.; Mattioli, S.; Torre, L.; Kenny, J.M.; Imbriani, M.; Arciola, C.R.; Visai, L. Antimicrobial properties and cytocompatibility of plga/ag nanocomposites. Materials 2016, 9. [Google Scholar] [CrossRef]
  241. Kwon, H.B.; Lee, J.H.; Lee, S.H.; Lee, A.Y.; Choi, J.S.; Ahn, Y.S. A case of argyria following colloidal silver ingestion. Ann. Dermatol. 2009, 21, 308–310. [Google Scholar] [CrossRef] [PubMed]
  242. Mayr, M.; Kim, M.J.; Wanner, D.; Helmut, H.; Schroeder, J.; Mihatsch, M.J. Argyria and decreased kidney function: Are silver compounds toxic to the kidney? Am. J. Kidney Dis. 2009, 53, 890–894. [Google Scholar] [CrossRef] [PubMed]
  243. Drake, P.L.; Hazelwood, K.J. Exposure-related health effects of silver and silver compounds: A review. Ann. Occup. Hyg. 2005, 49, 575–585. [Google Scholar] [CrossRef] [PubMed]
  244. Shahbazzadeh, D.; Ahari, H.; Motalebi, A.A.; Anvar, A.A.; Moaddab, S.; Asadi, T.; Shokrgozar, M.A.; Rahman-Nya, J. In vitro effect of nanosilver toxicity on fibroblast and mesenchymal stem cell lines. Iran. J. Fish. Sci. 2011, 10, 487–496. [Google Scholar]
  245. Mukherjee, S.G.; O’Claonadh, N.; Casey, A.; Chambers, G. Comparative in vitro cytotoxicity study of silver nanoparticle on two mammalian cell lines. Toxicol. In Vitro 2012, 26, 238–251. [Google Scholar] [CrossRef] [PubMed]
  246. Kim, S.; Choi, J.E.; Choi, J.; Chung, K.H.; Park, K.; Yi, J.; Ryu, D.Y. Oxidative stress-dependent toxicity of silver nanoparticles in human hepatoma cells. Toxicol. In Vitro 2009, 23, 1076–1084. [Google Scholar] [CrossRef] [PubMed]
  247. Kawata, K.; Osawa, M.; Okabe, S. In vitro toxicity of silver nanoparticles at noncytotoxic doses to HepG2 human hepatoma cells. Environ. Sci. Technol. 2009, 43, 6046–6051. [Google Scholar] [CrossRef] [PubMed]
  248. Avalos Funez, A.; Isabel Haza, A.; Mateo, D.; Morales, P. In vitro evaluation of silver nanoparticles on human tumoral and normal cells. Toxicol. Mech. Methods 2013, 23, 153–160. [Google Scholar] [CrossRef] [PubMed]
  249. Asare, N.; Instanes, C.; Sandberg, W.J.; Refsnes, M.; Schwarze, P.; Kruszewski, M.; Brunborg, G. Cytotoxic and genotoxic effects of silver nanoparticles in testicular cells. Toxicology 2012, 291, 65–72. [Google Scholar] [CrossRef] [PubMed]
  250. Albers, C.E.; Hofstetter, W.; Siebenrock, K.A.; Landmann, R.; Klenke, F.M. In vitro cytotoxicity of silver nanoparticles on osteoblasts and osteoclasts at antibacterial concentrations. Nanotoxicology 2013, 7, 30–36. [Google Scholar] [CrossRef] [PubMed]
  251. Frankova, J.; Pivodova, V.; Vagnerova, H.; Juranova, J.; Ulrichova, J. Effects of silver nanoparticles on primary cell cultures of fibroblasts and keratinocytes in a wound-healing model. J. Appl. Biomater. Funct. Mater. 2016. [Google Scholar] [CrossRef] [PubMed]
  252. Galandakova, A.; Frankova, J.; Ambrozova, N.; Habartova, K.; Pivodova, V.; Zalesak, B.; Safarova, K.; Smekalova, M.; Ulrichova, J. Effects of silver nanoparticles on human dermal fibroblasts and epidermal keratinocytes. Hum. Exp. Toxicol. 2015. [Google Scholar] [CrossRef] [PubMed]
  253. Liu, W.; Wu, Y.; Wang, C.; Li, H.C.; Wang, T.; Liao, C.Y.; Cui, L.; Zhou, Q.F.; Yan, B.; Jiang, G.B. Impact of silver nanoparticles on human cells: Effect of particle size. Nanotoxicology 2010, 4, 319–330. [Google Scholar] [CrossRef] [PubMed]
  254. De Matteis, V.; Malvindi, M.A.; Galeone, A.; Brunetti, V.; De Luca, E.; Kote, S.; Kshirsagar, P.; Sabella, S.; Bardi, G.; Pompa, P.P. Negligible particle-specific toxicity mechanism of silver nanoparticles: The role of ag+ ion release in the cytosol. Nanomed. Nanotechnol. 2015, 11, 731–739. [Google Scholar] [CrossRef] [PubMed]
  255. Chairuangkitti, P.; Lawanprasert, S.; Roytrakul, S.; Aueviriyavit, S.; Phummiratch, D.; Kulthong, K.; Chanvorachote, P.; Maniratanachote, R. Silver nanoparticles induce toxicity in a549 cells via ros-dependent and ros-independent pathways. Toxicol. In Vitro 2013, 27, 330–338. [Google Scholar] [CrossRef] [PubMed]
  256. Hsiao, I.L.; Hsieh, Y.K.; Wang, C.F.; Chen, I.C.; Huang, Y.J. Trojan-horse mechanism in the cellular uptake of silver nanoparticles verified by direct intra- and extracellular silver speciation analysis. Environ. Sci. Technol. 2015, 49, 3813–3821. [Google Scholar] [CrossRef] [PubMed]
  257. Park, E.J.; Yi, J.; Kim, Y.; Choi, K.; Park, K. Silver nanoparticles induce cytotoxicity by a trojan-horse type mechanism. Toxicol. In Vitro 2010, 24, 872–878. [Google Scholar] [CrossRef] [PubMed]
  258. Yang, H.; Liu, C.; Yang, D.F.; Zhang, H.S.; Xi, Z.G. Comparative study of cytotoxicity, oxidative stress and genotoxicity induced by four typical nanomaterials: The role of particle size, shape and composition. J. Appl. Toxicol. 2009, 29, 69–78. [Google Scholar] [CrossRef] [PubMed]
  259. Zhang, T.; Wang, L.; Chen, Q.; Chen, C. Cytotoxic potential of silver nanoparticles. Yonsei Med. J. 2014, 55, 283–291. [Google Scholar] [CrossRef] [PubMed]
  260. Park, M.; Neigh, A.M.; Vermeulen, J.P.; de la Fonteyne, L.J.J.; Verharen, H.W.; Briede, J.J.; van Loveren, H.; de Jong, W.H. The effect of particle size on the cytotoxicity, inflammation, developmental toxicity and genotoxicity of silver nanoparticles. Biomaterials 2011, 32, 9810–9817. [Google Scholar] [CrossRef] [PubMed]
  261. Liu, J.Y.; Sonshine, D.A.; Shervani, S.; Hurt, R.H. Controlled release of biologically active silver from nanosilver surfaces. ACS Nano 2010, 4, 6903–6913. [Google Scholar] [CrossRef] [PubMed]
  262. Molleman, B.; Hiemstra, T. Surface structure of silver nanoparticles as a model for understanding the oxidative dissolution of silver ions. Langmuir 2015, 31, 13361–13372. [Google Scholar] [CrossRef] [PubMed]
  263. Stoehr, L.C.; Gonzalez, E.; Stampfl, A.; Casals, E.; Duschl, A.; Puntes, V.; Oostingh, G.J. Shape matters: Effects of silver nanospheres and wires on human alveolar epithelial cells. Part. Fibre Toxicol. 2011, 8. [Google Scholar] [CrossRef] [PubMed]
  264. Kasemets, K.; Suppi, S.; Mantecca, P.; Kahru, A. Charge and size-dependent toxicity of silver nanoparticles to yeast cells. Toxicol. Lett. 2014, 229, S193–S194. [Google Scholar] [CrossRef]
  265. Schlinkert, P.; Casals, E.; Boyles, M.; Tischler, U.; Hornig, E.; Tran, N.; Zhao, J.; Himly, M.; Riediker, M.; Oostingh, G.J.; et al. The oxidative potential of differently charged silver and gold nanoparticles on three human lung epithelial cell types. J. Nanobiotechnol. 2015, 13. [Google Scholar] [CrossRef] [PubMed]
  266. Dziendzikowska, K.; Gromadzka-Ostrowska, J.; Lankoff, A.; Oczkowski, M.; Krawczynska, A.; Chwastowska, J.; Sadowska-Bratek, M.; Chajduk, E.; Wojewodzka, M.; Dusinska, M.; et al. Time-dependent biodistribution and excretion of silver nanoparticles in male wistar rats. J. Appl. Toxicol. 2012, 32, 920–928. [Google Scholar] [CrossRef] [PubMed]
  267. Loeschner, K.; Hadrup, N.; Qvortrup, K.; Larsen, A.; Gao, X.Y.; Vogel, U.; Mortensen, A.; Lam, H.R.; Larsen, E.H. Distribution of silver in rats following 28 days of repeated oral exposure to silver nanoparticles or silver acetate. Part. Fibre Toxicol. 2011, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Van der Zande, M.; Vandebriel, R.J.; Van Doren, E.; Kramer, E.; Rivera, Z.H.; Serrano-Rojero, C.S.; Gremmer, E.R.; Mast, J.; Peters, R.J.B.; Hollman, P.C.H.; et al. Distribution, elimination, and toxicity of silver nanoparticles and silver ions in rats after 28-day oral exposure. Acs Nano 2012, 6, 7427–7442. [Google Scholar] [CrossRef] [PubMed]
  269. Garza-Ocanas, L.; Ferrer, D.A.; Burt, J.; Diaz-Torres, L.A.; Ramirez Cabrera, M.; Tamez Rodriguez, V.; Lujan Rangel, R.; Romanovicz, D.; Jose-Yacaman, M. Biodistribution and long-term fate of silver nanoparticles functionalized with bovine serum albumin in rats. Metallomics 2010, 2, 204–210. [Google Scholar] [CrossRef] [PubMed]
  270. Pauksch, L.; Hartmann, S.; Rohnke, M.; Szalay, G.; Alt, V.; Schnettler, R.; Lips, K.S. Biocompatibility of silver nanoparticles and silver ions in primary human mesenchymal stem cells and osteoblasts. Acta Biomater. 2014, 10, 439–449. [Google Scholar] [CrossRef] [PubMed]
  271. Necula, B.S.; van Leeuwen, J.P.T.M.; Fratila-Apachitei, L.E.; Zaat, S.A.J.; Apachitei, I.; Duszczyk, J. In vitro cytotoxicity evaluation of porous TiO2-Ag antibacterial coatings for human fetal osteoblasts. Acta Biomater. 2012, 8, 4191–4197. [Google Scholar] [CrossRef] [PubMed]
  272. Campoccia, D.; Cangini, I.; Selan, L.; Vercellino, M.; Montanaro, L.; Visai, L.; Arciola, C.R. An overview of the methodological approach to the in vitro study of anti-infective biomaterials. Int. J. Artif. Organs 2012, 35, 800–816. [Google Scholar] [CrossRef] [PubMed]
  273. Stoodley, P.; Hall-Stoodley, L.; Costerton, B.; DeMeo, P.; Shirtliff, M.; Gawalt, E.; Kathju, S. Biofilms, biomaterials, and device-related infections. In Biomaterials Science: An Introduction to Materials in Medicine; Ratner, B.D., Hoffman, A.S., Schoen, F.J., Lemons, J.E., Eds.; Academic Press (Elsevier): Waltham, MA, USA, 2013; Volume 1, pp. 565–583. [Google Scholar]
  274. Barros, J.; Grenho, L.; Manuel, C.M.; Ferreira, C.; Melo, L.F.; Nunes, O.C.; Monteiro, F.J.; Ferraz, M.P. A modular reactor to simulate biofilm development in orthopedic materials. Int. Microbiol. 2013, 16, 191–198. [Google Scholar] [PubMed]
  275. Ludecke, C.; Jandt, K.D.; Siegismund, D.; Kujau, M.J.; Zang, E.; Rettenmayr, M.; Bossert, J.; Roth, M. Reproducible biofilm cultivation of chemostat-grown escherichia coli and investigation of bacterial adhesion on biomaterials using a non-constant-depth film fermenter. PLoS ONE 2014, 9, e84837. [Google Scholar]
  276. Rujanapun, N.; Aueviriyavit, S.; Boonrungsiman, S.; Rosena, A.; Phummiratch, D.; Riolueang, S.; Chalaow, N.; Viprakasit, V.; Maniratanachote, R. Human primary erythroid cells as a more sensitive alternative in vitro hematological model for nanotoxicity studies: Toxicological effects of silver nanoparticles. Toxicol. In Vitro 2015, 29, 1982–1992. [Google Scholar] [CrossRef] [PubMed]
  277. Albrecht, M.A.; Evans, C.W.; Raston, C.L. Green chemistry and the health implications of nanoparticles. Green Chem 2006, 8, 417–432. [Google Scholar] [CrossRef]
  278. Nunez-Anita, R.E.; Acosta-Torres, L.S.; Vilar-Pineda, J.; Martinez-Espinosa, J.C.; de la Fuente-Hernandez, J.; Castano, V.M. Toxicology of antimicrobial nanoparticles for prosthetic devices. Int. J. Nanomed. 2014, 9, 3999–4006. [Google Scholar]
  279. Sambale, F.; Wagner, S.; Stahl, F.; Khaydarov, R.R.; Scheper, T.; Bahnemann, D. Investigations of the toxic effect of silver nanoparticles on mammalian cell lines. J. Nanomater. 2015. [Google Scholar] [CrossRef]
  280. Sussman, E.M.; Casey, B.J.; Dutta, D.; Dair, B.J. Different cytotoxicity responses to antimicrobial nanosilver coatings when comparing extract-based and direct-contact assays. J. Appl. Toxicol. 2015, 35, 631–639. [Google Scholar] [CrossRef] [PubMed]
  281. Liu, Y.; Li, X.; Bao, S.; Lu, Z.; Li, Q.; Li, C.M. Plastic protein microarray to investigate the molecular pathways of magnetic nanoparticle-induced nanotoxicity. Nanotechnology 2013, 24, 175501. [Google Scholar] [CrossRef] [PubMed]
  282. Zou, J.; Feng, H.; Mannerstrom, M.; Heinonen, T.; Pyykko, I. Toxicity of silver nanoparticle in rat ear and BALB/c 3T3 cell line. J. Nanobiotechnol. 2014, 12, 52. [Google Scholar] [CrossRef] [PubMed]
  283. Chan, E.L.; Zhang, C.; Cheung, G.S. Cytotoxicity of a novel nano-silver particle endodontic irrigant. Clin. Cosmet. Investig. Dent. 2015, 7, 65–74. [Google Scholar] [CrossRef] [PubMed]
  284. Albrekt, A.S.; Johansson, H.; Borje, A.; Borrebaeck, C.; Lindstedt, M. Skin sensitizers differentially regulate signaling pathways in mutz-3 cells in relation to their individual potency. BMC Pharmacol. Toxicol. 2014, 15, 5. [Google Scholar] [CrossRef] [PubMed]
  285. Johansson, H.; Albrekt, A.S.; Borrebaeck, C.A.; Lindstedt, M. The gard assay for assessment of chemical skin sensitizers. Toxicol. In Vitro 2013, 27, 1163–1169. [Google Scholar] [CrossRef] [PubMed]
  286. Johansson, H.; Lindstedt, M.; Albrekt, A.S.; Borrebaeck, C.A. A genomic biomarker signature can predict skin sensitizers using a cell-based in vitro alternative to animal tests. BMC Genom. 2011, 12, 399. [Google Scholar] [CrossRef] [PubMed]
  287. Wong, C.L.; Ghassabian, S.; Smith, M.T.; Lam, A.L. In vitro methods for hazard assessment of industrial chemicals—Opportunities and challenges. Front. Pharmacol. 2015, 6, 94. [Google Scholar] [CrossRef] [PubMed]
  288. Wafa, H.; Grimer, R.J.; Reddy, K.; Jeys, L.; Abudu, A.; Carter, S.R.; Tillman, R.M. Retrospective evaluation of the incidence of early periprosthetic infection with silver-treated endoprostheses in high-risk patients: Case-control study. Bone Jt. J. 2015, 97, 252–257. [Google Scholar] [CrossRef] [PubMed]
  289. Lemcke, J.; Depner, F.; Meier, U. The impact of silver nanoparticle-coated and antibiotic-impregnated external ventricular drainage catheters on the risk of infections: A clinical comparison of 95 patients. Acta Neurochir. Suppl. 2012, 114, 347–350. [Google Scholar] [PubMed]
  290. Antonelli, M.; De Pascale, G.; Ranieri, V.M.; Pelaia, P.; Tufano, R.; Piazza, O.; Zangrillo, A.; Ferrario, A.; De Gaetano, A.; Guaglianone, E.; et al. Comparison of triple-lumen central venous catheters impregnated with silver nanoparticles (agtive(r)) vs. conventional catheters in intensive care unit patients. J. Hosp. Infect. 2012, 82, 101–107. [Google Scholar] [CrossRef] [PubMed]
  291. Grainger, D.W.; van der Mei, H.C.; Jutte, P.C.; van den Dungen, J.J.; Schultz, M.J.; van der Laan, B.F.; Zaat, S.A.; Busscher, H.J. Critical factors in the translation of improved antimicrobial strategies for medical implants and devices. Biomaterials 2013, 34, 9237–9243. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Free-floating bacteria (1) come close to the implant surface, here they interact via a set of chemical and physical mechanisms with a biomaterial surface covered by host cells/proteins. The majority of bacterial pathogens express specific surface adhesion molecules called adhesins (bacteria may have multiple adhesins for different surfaces); bacterial adhesion can be described as having an initial reversible, predominantly physically driven phase (2) and a time-dependent and irreversible molecular and cellular phase (4). The former is realized by Brownian motion, van der Waals attraction forces, gravitational forces, surface electrostatic charge and hydrophobic interactions (3); the latter employs a selective bridging function of bacterial surface polymeric structures, which include capsules, fimbriae or pili and slime; intermolecular interactions are facilitated by a protein film covering an implant immediately after its placement into the host body (5). Firm sticking of bacteria to the biomaterial surface allows them to create colonies (6) with biofilm formation, which is associated with a continuous release of free floating bacteria and signaling molecules (7).
Figure 1. Free-floating bacteria (1) come close to the implant surface, here they interact via a set of chemical and physical mechanisms with a biomaterial surface covered by host cells/proteins. The majority of bacterial pathogens express specific surface adhesion molecules called adhesins (bacteria may have multiple adhesins for different surfaces); bacterial adhesion can be described as having an initial reversible, predominantly physically driven phase (2) and a time-dependent and irreversible molecular and cellular phase (4). The former is realized by Brownian motion, van der Waals attraction forces, gravitational forces, surface electrostatic charge and hydrophobic interactions (3); the latter employs a selective bridging function of bacterial surface polymeric structures, which include capsules, fimbriae or pili and slime; intermolecular interactions are facilitated by a protein film covering an implant immediately after its placement into the host body (5). Firm sticking of bacteria to the biomaterial surface allows them to create colonies (6) with biofilm formation, which is associated with a continuous release of free floating bacteria and signaling molecules (7).
Materials 09 00337 g001
Figure 2. Prevention of PJI consists of a list of measurements optimizing host status/preparedness for surgery (identification of host risk factors, determination of host comorbidities; local antibacterial activities); reducing bacterial load during the surgery (intravenous antibiotic prophylaxis, operating room environment/traffic/management, surgical experience, measurements/tools preventing deliberation of bacteria from the surgeon/operating room personnel, protection of the implant from bacterial contamination/adhesion) and minimizing the chance for postoperative bacterial contamination (wound care strategy, rapid optimization of postoperative immune and metabolic conditions, early ambulance, experienced physiotherapy, eradication of local infections and haematogenous sources of bacteria).
Figure 2. Prevention of PJI consists of a list of measurements optimizing host status/preparedness for surgery (identification of host risk factors, determination of host comorbidities; local antibacterial activities); reducing bacterial load during the surgery (intravenous antibiotic prophylaxis, operating room environment/traffic/management, surgical experience, measurements/tools preventing deliberation of bacteria from the surgeon/operating room personnel, protection of the implant from bacterial contamination/adhesion) and minimizing the chance for postoperative bacterial contamination (wound care strategy, rapid optimization of postoperative immune and metabolic conditions, early ambulance, experienced physiotherapy, eradication of local infections and haematogenous sources of bacteria).
Materials 09 00337 g002
Figure 3. Total joint arthroplasty has several types of surfaces according to their locations and functions; ideally the whole implant should be covered via application of an antibacterial strategy; however, in practice the antibacterial strategy for a particular kind of surface has to respect its critical characteristics (for example the strategy for an articulating surface, let’s say a polyethylene one, has to be different from a non-articulating metallic one).
Figure 3. Total joint arthroplasty has several types of surfaces according to their locations and functions; ideally the whole implant should be covered via application of an antibacterial strategy; however, in practice the antibacterial strategy for a particular kind of surface has to respect its critical characteristics (for example the strategy for an articulating surface, let’s say a polyethylene one, has to be different from a non-articulating metallic one).
Materials 09 00337 g003
Figure 4. A particular implant surface has to address several implant-related tasks simultaneously and continuously (in the ideal case), therefore engineers have to solve the problem of how to bind (attract, fix) often contradictory functionalities via specific modifications/treatments in the particular surface location.
Figure 4. A particular implant surface has to address several implant-related tasks simultaneously and continuously (in the ideal case), therefore engineers have to solve the problem of how to bind (attract, fix) often contradictory functionalities via specific modifications/treatments in the particular surface location.
Materials 09 00337 g004
Figure 5. Silver nanoparticles simultaneously target bacteria via the destruction of their wall, inactivation of their enzymes, disrupting of critical metabolic pathways and interaction with bacterial DNA.
Figure 5. Silver nanoparticles simultaneously target bacteria via the destruction of their wall, inactivation of their enzymes, disrupting of critical metabolic pathways and interaction with bacterial DNA.
Materials 09 00337 g005
Table 1. Examples of anti-infective strategies proposed for treating of surfaces used in orthopaedic implants.
Table 1. Examples of anti-infective strategies proposed for treating of surfaces used in orthopaedic implants.
StrategyFeaturesExamplesReferences
Prevention in adhesion and adsorptionAnti-adhesive polymers[66,67,68]
Albumin[69]
Super-hydrophobic surfaces[70,71,72]
Nano-patterned surface[73,74,75,76,77]
Hydrogels[78,79,80,81]
Silicon nitride ceramics[82,83]
Methods to kill bacteriaInorganicSilver, silver-oxide[84,85,86]
Silver nanoparticles[87,88,89,90,91,92,93,94]
Gold nanoparticles[95,96]
Titanium dioxide[97,98,99]
Selenium ions[100,101,102]
Copper ions/nanoparticles[103,104]
Zinc ions[105,106]
Iodine coating[107]
Bioactive glass[108,109]
Graphene oxide[110,111]
OrganicCoated or covalently linked antibiotics[112,113,114,115,116]
Chitosan derivatives[117,118,119,120]
Signaling, inhibiting and antimicrobial peptides[121,122,123]
Cytokines[124]
Enzymes[125,126]
OtherNon-antibiotic bactericidal substances[127]
CombinedMultilayer coating[128,129,130,131,132]
Synergy material intensification[133,134,135]
Positively charged polymers[136]
Multi-functional and smart coatingPassiveNanostructured “smart” materials[68,137,138,139,140]
ActiveConcept: sensors conjoined to nanocontainers[141,142,143,144,145,146]
Alternative approach Lytic bacteriophages [147]
Surface-adaptive anti-biofilm nanocarriers[148]

Share and Cite

MDPI and ACS Style

Gallo, J.; Panacek, A.; Prucek, R.; Kriegova, E.; Hradilova, S.; Hobza, M.; Holinka, M. Silver Nanocoating Technology in the Prevention of Prosthetic Joint Infection. Materials 2016, 9, 337. https://doi.org/10.3390/ma9050337

AMA Style

Gallo J, Panacek A, Prucek R, Kriegova E, Hradilova S, Hobza M, Holinka M. Silver Nanocoating Technology in the Prevention of Prosthetic Joint Infection. Materials. 2016; 9(5):337. https://doi.org/10.3390/ma9050337

Chicago/Turabian Style

Gallo, Jiri, Ales Panacek, Robert Prucek, Eva Kriegova, Sarka Hradilova, Martin Hobza, and Martin Holinka. 2016. "Silver Nanocoating Technology in the Prevention of Prosthetic Joint Infection" Materials 9, no. 5: 337. https://doi.org/10.3390/ma9050337

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