Next Article in Journal
Device-Associated Infections in COVID-19 Patients: Frequency of Resistant Bacteria, Predictors and Mortality in Medellín, Colombia
Previous Article in Journal
Comparison of Rhizosphere Bacterial Communities of Pinus squamata, a Plant Species with Extremely Small Populations (PSESP) in Different Conservation Sites
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Biofilm Strategies: A Focused Review on Innovative Approaches

by
Antonella Iaconis
1,
Laura Maria De Plano
1,
Antonella Caccamo
1,
Domenico Franco
1,* and
Sabrina Conoci
1,2,3,*
1
Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
2
Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy
3
URT Lab Sens Beyond Nano—CNR-DSFTM, Department of Physical Sciences and Technologies of Matter, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy
*
Authors to whom correspondence should be addressed.
Microorganisms 2024, 12(4), 639; https://doi.org/10.3390/microorganisms12040639
Submission received: 4 March 2024 / Revised: 17 March 2024 / Accepted: 19 March 2024 / Published: 22 March 2024
(This article belongs to the Section Antimicrobial Agents and Resistance)

Abstract

:
Biofilm (BF) can give rise to systemic infections, prolonged hospitalization times, and, in the worst case, death. This review aims to provide an overview of recent strategies for the prevention and destruction of pathogenic BFs. First, the main phases of the life cycle of BF and maturation will be described to identify potential targets for anti-BF approaches. Then, an approach acting on bacterial adhesion, quorum sensing (QS), and the extracellular polymeric substance (EPS) matrix will be introduced and discussed. Finally, bacteriophage-mediated strategies will be presented as innovative approaches against BF inhibition/destruction.

1. Introduction

Biofilm (BF) production represents a strategy that bacteria use to survive in adverse conditions and to increase their survival success in the host [1]. Unfavorable conditions can induce bacteria to convert their physiological state from free-floating (planktonic) to sessile cells, acquiring the ability to adhere, grow, and form communities on biotic or abiotic surfaces [2,3]. This physio-metabolic change affects the entire bacterial community by a particular cell–cell communication mechanism, named quorum sensing (QS) [4]. Consequently, the bacterial population coordinates its metabolic activity towards the secretion of an extracellular polymeric substance (EPS), including lipids, polysaccharides, proteins, extracellular nucleic acids (eDNA), and ions [5]. Within this extracellular matrix, bacteria increase their resistance against drying, antimicrobial agents, and the action of the host’s immune system [6]. This finely controlled cooperation often involves different bacterial species, leading to polymicrobial BFs [7,8,9,10]. Bacteria in BFs obtain a common benefit, in terms of growth, virulence, persistence, and the acquisition of antimicrobial resistance (AMR) [11]. The BF extracellular matrix can be considered a hotspot for the diffusion of antibiotic resistance genes, due to the greater frequency and speed of horizontal gene transfer [12]. Therefore, BFs can act as a reservoir of multidrug-resistant (MDR) bacteria, often associated with serious illness and death [11].
The Centers for Disease Control and Prevention estimated that over 2 million infections and 23,000 deaths associated with MDR bacteria occur annually [13]. Among them, six highly virulent and antibiotic MDR bacteria have been included in the ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) groups [14]. Infections associated with ESKAPE bacteria are generally chronic in the presence of BF and require aggressive therapeutic treatments that subject the patient to serious complications [15]. BF infections can affect the lungs, mainly cystic fibrosis, wounds or medical implants, including orthopedic devices, and intravenous and urinary catheters [16,17]. Because BF infections are very difficult to eradicate, many lines of research are focused on altering the early stages of BF formation [18,19]. Among them, the alteration of QS is emerging as a promising and efficient prevention strategy [20,21,22]. On the other hand, since prevention approaches are not always applicable, several strategies have the EPS matrix as the main target to make pathogenic strains more susceptible to common therapeutic treatments [23,24,25]. Approaches based on bio- and nanotechnology have piqued the interest of numerous research groups due to the possibility of providing greater efficacy to actives and/or obsolete antibiotics [26,27,28,29,30].
This review aims to provide an overview of recent strategies for the prevention and destruction of pathogenic BFs. First, a schematic description of the main phases of the life cycle of BF and maturation will be described, focusing attention on the metabolic pathways involved and the structural components of BFs from Gram-positive and -negative bacteria. Then, recent anti-BF strategies acting on bacterial adhesion to surfaces, QS, and EPS will be introduced and discussed. Ultimately, recent approaches to BF inhibition/destruction by using bacterial viruses will be presented.

2. Life Cycle of Biofilm

The life cycle of BF is a complex phenomenon involving a large number of parameters [31,32]. The main phases of the life cycle of BF are the reversible (i) and irreversible (ii) surface adhesion, BF production (iii) and maturation (iv), and the dispersion (v) of planktonic cells or EPS-included cell aggregates (Figure 1) [33,34].
The life cycle of BF begins with the reversible surface adhesion of planktonic cells (phase 1). Adhesion to biotic or abiotic surfaces is mainly mediated by electrostatic intermolecular interactions, such as acid–base interactions and Van der Waals forces [35,36]. Conformational changes in bacterial surface proteins and an increase in acid–base and hydrophobic interactions progressively maximize the contact with the surface and the removal of interfacial water [37]. In some cases, surface exploration may take place by swarming processes, mediated by Type IV pili or flagella [38]. At this stage (phase 2), bacteria change their physiological state from planktonic to sessile cells by the loss of the superficial appendages and the activation of secondary metabolic pathways [39]. BF production (phase 3) begins with the replication of bacteria by forming mini aggregate microcolonies and the expression of genes related to EPS production and secretion [40,41]. EPS production involves the release of an adhesive matrix which makes cells adhere to each other and allows for the three-dimensional growth of the BF. During BF maturation (phase 4), some cells go towards death and can be used as scaffolds for BF growth. Meanwhile, viable cells import water, nutrients, and other metabolites from the external environment that will be necessary for their survival [42]. At this stage, BFs have the highest resistance against mechanical stresses and adverse environmental factors. The last stage is BF dispersion (phase 5), an active process due to the deterioration and detachment of BF portions with the release of planktonic cells or EPS-included cell aggregates [43]. This phase represents the starting point of a new life cycle of BF formation on other biotic or abiotic surfaces [44].

2.1. Reversible Adhesion

Reversible surface adhesion is a crucial step in the BF life cycle [42]. In this stage, bacteria are weakly bound to the surface, mainly due to the absence of EPS, and can explore neighbouring surfaces if deemed more favourable to BF production [36]. On the other hand, the instability of the bacteria makes their remotion easy and, consequently, the blocking of BF production. Motility systems, such as flagella, and the conditioning of the surface through the secretion of polysaccharides are involved in this step [45,46,47]. For example, S. aureus, devoid of any flagella, uses Brownian motions to approach surfaces, promoting initial adhesion by using polysaccharide intercellular adhesins (PIAs) and extracellular DNA (eDNA) [48]. Instead, P. aeruginosa regulates flagellar motility to promote both adhesion on surfaces and cell–cell adhesion. After reaching the appropriate surface, P. aeruginosa minimizes flagellar motility and uses the contraction motility of type IV pili to crawl on the surface and release an exopolysaccharide that promotes surface attachment [49]. Surface characteristics also influence the initial adhesion phase, since they can favour reversible bonds due to hydrophobic and electrostatic interactions [43]. Interactions of bacteria with positive surfaces are generally favoured due to the negative charge of the cell wall [50]. Adhesion strategies differ based on the peculiar characteristics of the cell wall [36]. In Gram-positive bacteria, adhesion is supported by adhesins, binding collagen and fibronectin proteins, and teichoic acids [51]. Bucher et al. identified specific targets, involved in biosynthesis pathways of cell wall components, for hampering BF formation and the anchoring of the extracellular matrix, without affecting planktonic growth [52]. In Gram-negative bacteria, adhesion is favoured by lipopolysaccharides (LPS) in the outer membrane, which makes the bacterial surface highly negative. Abdel-Rhman suggests that LPS in P. aeruginosa can stimulate and stabilize BFs [53]. In addition, LPS have a direct stimulatory effect on BFs from other bacterial strains, increasing their virulence [54]. Surfaces of biomedical devices, such as catheters, heart valves, and prostheses, favour the adhesion and subsequent proliferation of pathogenic bacteria, often resulting in chronic infection and health risks to patients [55,56,57].

2.2. Extracellular Polymeric Substances (EPSs)

Extracellular polymeric substances (EPSs) play a main role in bacteria survival since they protect bacteria from antibiotics and avoid drug penetration at bactericidal concentrations [58]. The extracellular matrix is an essential component for BF formation and maturation, both in Gram-positive and Gram-negative bacteria [59]. Its components are similar for both bacteria types and include proteins (expressed only during BF formation), polysaccharides, extracellular nucleic acids (eDNA), and some membrane vesicles [60]. BF-associated proteins (Bap), generally present in S. aureus [61], have in the central part multiple identical repeats that contain amyloid-like peptide sequences [62]. These proteins promote bacterial adhesion to an abiotic surface by self-assembling into amyloid-like aggregates in response to calcium concentration and low pH values. Gram-negatives also have surface proteins homologous to Bap [47]. Various bacterial species have different numbers of amyloid-like repeats that help in adhesion and immune evasion. Particularly, amyloid-like proteins are present in different species: PSM (phenol-soluble modulins) in S. aureus, TasA (translocation-dependent antimicrobial spore component) in B. subtilis, CsgA (major curli subunit) in E. coli, and Fap in Pseudomonas spp. [63,64]. These proteins also act as access channels for nutrients and exhibit surfactant properties that help with dispersal. Amyloid-like proteins are secreted on bacterial surfaces unprocessed, so they require polymerization and processing for final functional activity [47]. Several polysaccharides are also of fundamental importance for BFs. In S. aureus, adhesion is enhanced by the surface polysaccharide PNAG (poly-β(1,6)-N-acetyl-D-glucosamine) observed in the formation of immune evasion BF [65]. Meanwhile, in P. aeruginosa, exopolysaccharides are secreted including alginate, synthesis locus (Psl), and pellicle (Pel). Alginate may be the major exopolysaccharide responsible for P. aeruginosa mucoid formation during chronic infection, while Psl and Pel are more responsible for BF adhesion and maintenance [66]. In E. coli, cellulose plays a fundamental role in BF formation; it can also be regulatory and exert the feedback inhibition of BFs [67]. These polysaccharides, in addition to having a role in BF formation, are also important for its maintenance and could be used as targets for the inhibition of BF production [60]. Recently, antibodies capable of binding to the antigenic determinant (epitope) of BF-specific polysaccharides have been tested and developed: for example, human antibodies to P. aeruginosa polysaccharide Psl are available and enhance the immune response to BFs [68], while poly-N-acetyl beta-glucosamine has been tested as an antibody against staphylococci [69]. Extracellular DNA, both in Gram-positive and Gram-negative bacteria, confers antibiotic resistance to BFs. DeFrancesco et al. have shown that the use of DNase treatment reduces BFs [70], and therefore this component could be a potential target for future anti-BF therapies. Another important component in BF formation are vesicles which have a fundamental role as hydrophobic surface providers in the transport of EPS constituents, such as eDNA, adhesion-related proteins, and lipids. Membrane vesicles (MVs) are found in Gram-positive bacteria, while outer membrane vesicles (OMVs) in Gram-negative [47]. In S. aureus, MVs can carry active β-lactamase, which confers resistance to antibiotics [71]. Otherwise, OMVs produced by Gram-negative transport protein components of the matrix, endotoxins, DNA, and enzymes [72]. A proteomic study indicated that 20% of the protein components of the matrix derive from the OMVs [73]. Potentially, the vesicular components could also be a target for possible anti-BF therapies.

2.3. Quorum Sensing in Biofilm Production

Quorum sensing (QS) is a bacterial system for regulating gene expression in response to fluctuations in cell population density [4]. Bacteria can communicate and recognize the population density and control gene expression thanks to the release and accumulation of self-inducing extracellular signals, named autoinducers. As the bacterial population increases, autoinducers accumulate above the minimum threshold level: receptors that bind autoinducers trigger signal transduction cascades that lead to population-level changes in gene expression (Figure 2) [74,75]. There are many processes related to QS, including bioluminescence, the secretion of virulence factors, and the sporulation and production of antibiotics [76]. Gram-positive and Gram-negative bacteria have different types of autoinducers, although some systems are present in both types of bacteria [77].
In Gram-positive bacteria, intracellular communication is regulated by autoinducing peptides (AIPs), small post-translationally modified peptides. Several AIPs have been identified, including their structure–activity relationships and cognate receptors [78]. In S. aureus, the accessory gene regulator (Agr) is responsible for the regulation and secretion of AIPs (Figure 2A). Four Agr systems regulate the production of many AIPs and each specifically activates its cognate receptor [79]. The Agr system influences protease expression, which facilitates the collapse of mature stage BFs [80] by downregulating the formation of adhesion molecules such as autolysin E (AtIE) [5]. In S. aureus planktonic cells, the Agr system is highly active except in the BF state, indicating that the Agr system regulates the shedding of the BF [81]. The primary QS signalling molecule in Gram-negative is N-acylated homoserine-lactone (AHL) [82]. AHL regulates the so-called LuxI/LuxR system. AHL is released extracellularly, and when it accumulates, it enters the cell and binds to its receptor LuxR. Thanks to this binding, LuxR can lead the transcription of the gene responsible for the production of LuxI. LuxI, in turn, is liable for producing AHL itself. LuxI/LuxR regulates virulence factors and BF formation (Figure 2B) [83]. Gram-negative and -positive bacteria widely spread the system mediated by autoinducer-2 (AI-2) [84]. LuxS is a protease, encoded by the luxS gene, and regulates the synthesis of AI-2, which in turn can be transported out of the cells. During cell proliferation, the extracellular AI-2 concentration increases, and when reaching a threshold, bacteria sense a critical cell mass. This involves the regulation of QS gene expression, including the formation of BFs and the expression of virulence genes [85]. The two-component system (TCS) is ubiquitous and regulates several bacterial functions including growth, metabolism, pathogenicity, drug resistance, and host recognition [86]. The TCS is involved in planktonic cell morphology and consequently in BF formation [87]. The peculiar feature of this system is the presence of two components: the first component is a protein that acts as a sensor, in this case, histidine protein kinase (HPK), which is found in the inner membrane; the second component is a regulatory protein (RR) [5]. RR activation is responsible for the regulation of genes involved in morphogenesis, virulence factors, and QS [88].

3. Anti-BF Strategies Acting on Bacterial Adhesion to Surfaces

Surfaces in contact with biological fluids represent a good niche for bacteria adhesion. Surface adhesion is the crucial event for BF formation which can lead to complicated infections, even localized chronic infections, and serious limitations in the function of biomaterials [89]. Several surfaces favour bacterial adhesion, promoting the transition from planktonic to sessile cells and EPS-mediated cell anchoring [36,90]. Surface modulation represents one of the best approaches in preventing bacterial adhesion and an excellent prevention strategy against pathogenic BFs [91]. Surfaces can be directly altered or added with coatings able to make them inhospitable for bacteria [92]. Physical and chemical surface modulations can prevent bacterial adhesion and release lethal substances for microorganisms so that in addition to the anti-BF action, the surface would be able to prevent bacterial proliferation (Figure 3) [89].

3.1. Antiadhesive and Antibacterial Surface Modulation

Over the last 20 years, strategies have been developed to combat resistance associated with BFs but also to prevent microorganisms from being able to produce it [93]. Modulating a surface capable of preventing the first stages of BF formation seems to be a good anti-BF strategy. Zwitterionic materials have antifouling properties and have been widely used to construct antifouling surfaces for medical devices, biosensors, and marine coating applications [94]. Zwitterionic materials prevent bacterial cells from adhering thanks to their electric neutrality with equivalent positive- and negative-charged groups. They are also able to prevent protein attachment and bacterial colonization as their chemical properties allow them the binding of water molecules [95]. Silver-coated surfaces have been extensively studied due to their antimicrobial properties [96,97]. Lemire et al. have demonstrated that surfaces coated with silver oxynitrate have also successfully eliminated multispecies BFs [98]. Medical devices coated with aryl rhodanines specifically inhibit the early stages of BF development in Gram-positive strains, despite not having antibacterial activity [99]. The antiadhesive materials prevent the first surface adhesion, leading to the inhibition of the formation of BFs, in short-term installations as they cannot kill bacteria [100]. Xiang et al. used poly (carboxybetaine-co-dopamine methacrylamide) copolymer (PCBDA) to immobilize silver nanoparticles on cotton gauze resulting in an effective method not only to inhibit BF formation but also to accelerate healing processes [101]. Instead, a new strategy to inhibit the formation of BFs on surfaces could be represented by bacteriocin, a proteinaceous toxin produced by bacteria to inhibit the growth of similar bacterial strains [102]. Indeed, a bacteriocin derived from Lactobacillus sakei was able to destroy the BF of Listeria monocytogenes on stainless steel [103]. For example, bacteriocins bovicin HC5 and nisin can vary the microbial cell’s hydrophobicity and modulate the microbial cellular attachment, even at sub-inhibitory concentration [104].

3.2. Use of Nanoparticles (NPs) in Surface Modulation

In addition to the development of new materials that prevent microorganisms from adhering and starting all those mechanisms that lead to the formation of BF, nanoparticles (NPs) of various kinds have recently been used and deposited on surfaces, to prevent the formation of BFs [105]. NP-embedded materials have now been shown to inhibit BF formation in E. coli, P. aeruginosa, S. aureus, and S. epidermidis [106]. The use of NPs can be substantially divided into two strategies: the first involves the use of NPs, of a lipid or polymeric nature, as drug delivery carriers to activate antibiotics; the second involves the use of metallic NPs which act as an antimicrobial agent [107]. NPs can be used associated with enzymes that can degrade the BF’s adhesive structure [100]. For example, α-amylase, a glycoside hydrolase that catalyses the breaking of 1,4-glycosidic bonds, is also able to hydrolyze carbohydrates present in the BF matrix [108]. Silver NPs associated with amylases can eradicate and inhibit BF formation in different bacterial species [100,109]. Hybrids of α-amylase and zinc oxide in an NP form used to coat urinary catheters in a single-step sonochemical approach allowed for a drastic reduction in the incidence of bacteriuria in rabbit models [110]. Thanks to the combination of silver NPs with modified sulfobetaine in polyester membranes, the formation of BFs in S. aureus and E. coli was inhibited [111]. Furthermore, NPs could inhibit DNA replication and gene expression, thanks to the ability to attach themselves to the microbial surface allowing it to react with proteins and cellular DNA [112]. The nanoparticles most studied to implement anti-BF strategies are gold (Au), silver (Ag), and zinc (Zn) [113]. AuNPs have no antibacterial activity on their own, except when combined with antibiotics. Ampicillin bound to the surface of AuNPs (AuNPs-AMP) allows for the subversion of the drug resistance in different microorganisms such as E. coli, P. aeruginosa, and E. aerogenes [114]. Thanks to the presence of ampicillin, the AuNPs can enter the bacterial cells allowing for them to carry out their antimicrobial function [115,116]. AgNPs have antimicrobial activities and are also responsible for suppressing microbial resistance in terms of growth [117]. Silver ions (Ag+) have antimicrobial activity as they can bind to the negative part of cell membranes, damaging them and therefore allowing the cytoplasmic contents to escape [118]. Thanks to the presence of LPS, negatively charged, Gram-negative bacteria are more sensitive to the action of AgNPs, unlike Gram-positive bacteria which instead have peptoglycans, positively charged [119]. Regarding the use of ZnNPs, it is widely known that the antimicrobial activity of zinc oxide NPs (ZnONPs) is associated with the production of reactive oxygen species (ROS) that can affect cellular components such as lipids, proteins, and nucleic acids, with consequent cellular structural alterations [120]. The use of NPs, and more generally the modulation of surfaces, represents an excellent way forward in the prevention of bacterial adhesion, hindering the formation of BFs; however, there is a need to mainly improve the characterization of NPs on surfaces but also biocompatibility and the assessment of any toxic effect on humans, animals, and the environment [107]. Applying these strategies to inhibit BF formation appears to be the solution with the best perspective as this type of modulation would not lead to the development and worsening of antibiotic resistance [105].

4. Anti-BF Strategies Acting on Quorum Sensing

Quorum sensing (QS) is a mechanism for regulating gene expression based on microbial population density in response to environmental oscillations. QS offers better survival to microbial populations rather than individual cells [4]. In the human health field, because QS is correlated to BFs’ production, several authors are focusing their studies on cell-to-cell communication disruption as an innovative strategy against chronic infections [121]. In-depth studies on what the signals that regulate the formation of BFs are can represent the best strategy against bacterial resistance and immune evasion [5,122,123]. Different molecules, including enzymatic proteins, can degrade bacterial messengers or inactivate specific receptors related to bacterial QS, blocking the metabolic pathways associated with it [6]. Consequently, bacterial cells remain in a dispersed condition and are more susceptible to antimicrobial treatment than BF-residing cells [124]. This phenomenon of the interruption of QS signals is generally referred to as quorum quenching (QQ) (Figure 4) [125].

4.1. Quorum Sensing Inhibitors

Several molecules can inhibit or block cell-to-cell communication mediated by QS. Many of these molecules are naturally produced plant metabolites, such as cinnamaldehyde, one of the primary constituents of cinnamon, indicated for its therapeutic potential as an antimicrobial agent against pathogenic BFs [126]. Niu et al. showed that low concentrations of cinnamaldehyde negatively affect two types of QS related to acyl homoserine lactone (AHL) and autoinducer-2 [127]. In the same line of research, Topa et al. combined cinnamaldehyde with colistin to obtain synergistic activity in the inhibition and dispersion of preformed P. aeruginosa BF [128]. Again, Li et al. indicated a decrease in the virulence phenotypes of Aeromomonas hydrophila, due to QS inhibition and the downregulation of related genes following the addition of cinnamaldehyde [129]. Exploiting nanotechnology advancement, cinnamaldehyde has been loaded on chitosan NPs for anti-BF purposes [130]. Using a similar system, Subhaswaraj et al. obtained a significant anti-quorum sensing activity, mediated by the downregulation of virulence factors such as PAO1 related to QS in P. aeruginosa and associated with its BF formation [131]. Moreover, they indicated a significant alteration in the swimming and swarming motility of the same bacterial strain. In the end, Ramasamy et al. conjugated cinnamaldehyde to the surface of gold NPs to obtain a broad spectrum of anti-BF activity against Gram-negative (E. coli O157:H7 and P. aeruginosa) and -positive (methicillin-sensitive and -resistant S. aureus) bacteria [132]. Other compounds able to inhibit QS belong to natural flavonoids, such as baicalein and quercetin, for which a concentration-dependent decrease in violacein production in C. violaceum 12,472 and inhibition in pyocyanin production, proteolytic, and elastolytic activities, swarming motility, and BF formation in P. aeruginosa PAO1 have been demonstrated, due to quercetin and quercetin-3-O-arabinoside [133]. Paczkowski et al. showed inhibition via the antagonism of the autoinducer-binding receptors, LasR and RhlR, in P. aeruginosa. Specifically, a significant reduction in the ability of receptors to bind DNA-encoding QS-regulated promoters, mediated by two hydroxyl groups in the flavone A-ring, has been reported [134]. Similar results were obtained on A. hydrophila by citrus flavonoid hesperidin methylchalcone, with a consequent decrease in BF development and virulence factor production [135]. Finally, Pachaiappan et al. investigated the inhibition activity of N-acyl-homoserine lactone mediated by two flavonoids, namely apigenin and acacetin, and three isoflavonoids, namely genistein, daidzein, and biochanin, in P. aeruginosa [136]. Flavonoid derivatives have also been evaluated coupled with chitosan delivery systems to enhance the inhibitory activity of BFs from E. coli and P. aeruginosa [137,138]. Alternative approaches involved the use of silver and gold nanoparticles in flavonoid-based nanohybrids for multidrug-resistant bacteria [139,140]. Another plant metabolite able to prevent the production of QS-controlled virulence factors and related to BF formation is eugenol [141]. Fekrirad et al. indicated that this catechol was able to prevent the production of QS-controlled virulence factors, such as pigment prodigiosin, protease, and hemolysin in Serratia marcescens [142]. Specifically, they found that eugenol affected swarming motility, the formation of the microcolony, and extracellular polysaccharide via the downregulation of correlated genes. Also, in this case, approaches combined with other hydrophobic antimicrobial agents (triclosan) and nano- and micro-emulsions, reinforced with silver nanocomposites, have successfully been evaluated against BFs from Gram-positive and -negative bacteria [143,144,145,146]. The activity of QS inhibition has also been documented for some antibiotics, although their main activity is to inhibit growth or act as bactericidal [147,148,149,150,151]. Some authors showed that the macrolide antibiotic azithromycin was able to inhibit QS signal molecules in P. aeruginosa, attenuating its virulence [148,149]. Based on this finding, Zeng et al. have investigated the mechanism of action [150]. Since azithromycin acts on ribosomes, it has been evaluated if the transcriptional regulation of representative virulence genes could elicit alternative modes of gene expression mediated by the antibiotic. They suggest a relationship between lasI and rhlI, for which the first acts as a cell density sensor, while the second functions as a fine-tuning mechanism for the coordination of different QS systems. The ability to reduce the expression of virulence factors in bacterial populations is also documented for other antibiotics. For example, low concentrations of ceftazidime, cefepime, and imipenem caused the significant elimination of the QS signals in P. aeruginosa at a concentration 20 times lower than the MIC [151]. The activity is related to a decrease in elastase, protease, pyocyanin, and hemolysin, suggesting a potential use of β-lactam antibiotics as an effective approach for the prevention and treatment of BF infection [151]. To efficiently increase the penetration and retention of antibiotics in BFs, several approaches based on the loading of the antibiotic in small and differently charged NPs have been proposed [152,153]. It is known that a combination of oxazolidinone derivatives compound with β-lactam antibiotics (meropenem trihydrate) can reduce P. aeruginosa BFs by inhibiting the virulence factors such as elastase, pyocyanin, rhamnolipid, and protease and bacterial motility [154]. Anti-QS strategies often involve the use of such water-soluble cyclic oligosaccharides to enhance or synergistically act against BFs and other virulence factors [155,156,157].

4.2. Metal Nanoparticles as QS Inhibitors

The antibacterial activity associated with metal-based nanoparticles (MeNPs) includes the loss of cell wall and membrane integrity, as well as interference in many metabolic functions essential for bacterial cell viability [158,159]. The main mechanisms include physical damage due to chemical interactions, such as leaching and the dissolution of metal ions, and/or the production of reactive oxygen species (ROS) [160]. Recent evidence has demonstrated that metal NPs are also able to interfere with cell-to-cell communication, acting as QS inhibitors or inductors [161]. Srinivasan et al. have evaluated the anti-QS and anti-BF potential of Piper betle-based synthesized silver NPs against S. marcescens and P. mirabilis. Their results revealed the inhibition of QS-mediated virulence factors, such as prodigiosin, protease, and exopolysaccharides. Specifically, they indicated the downregulation of fimA, fimC, flhD, and bsmB genes in S. marcescens and flhB, flhD, and rsbA genes in P. mirabilis, respectively [162]. Similarly, Shah et al. showed anti-quorum sensing activity in nosocomial pathogen P. aeruginosa mediated by photosynthesized silver NPs. They deduced that eugenol-, a phenolic phytochemical, conjugated AgNPs exhibited a considerable binding interaction with QS-associated proteins, such as LasR, LasI, and MvfR [163]. Kumar et al. biosynthesized silver NPs from an aqueous leaf extract of Koelreuteria paniculata [164]. Their NPs resulted in the superior inhibition of QS-regulated virulence factors in P. aeruginosa PAO1 compared to chemically synthesized AgNPs. Moreover, no effects on cell viability were observed. On the other hand, Saeki et al. evaluated biogenic silver NPs acting on BF formation, the production of virulence factors, and the expression of QS-related genes PAO1 and PA14 in P. aeruginosa. [165]. However, their results indicated that exposure to low concentrations of bio-AgNPs could promote the expression of QS genes in P. aeruginosa, increasing the production of virulence factors such as elastase, pyocyanin, and BFs [165]. Anti-QS has also been obtained from other MeNPs. Elshaer and Shaaban microbially synthesized gold and selenium with anti-virulent activity against P. aeruginosa [166]. NPs inhibited QS-related virulence factors, such as pyocyanin, protease, and elastase, as well as significantly suppressed the expression of QS genes and toxins. Gold NPs from Capsicum annuum reduced P. aeruginosa and S. marcescens BFs, probably by inhibiting QS signals and blocking regulatory proteins [167]. Again, Gómez-Gómez et al. asserted the alteration of the QS signalling system mediated by selenium and tellurium NPs in P. aeruginosa [168]. Similar results were obtained by Maruthupandy et al. by using nickel oxide NPs [169]. Zinc and titanium oxide have been documented to have strong antibacterial activity against Gram-negative and -positive bacteria, affecting their adhesion on prosthetic scaffolds [170,171]. Khan et al. obtained two morphologically different sol–gel-fabricated ZnO nanospikes with inhibitory effects on quorum sensing and BF formation in P. aeruginosa [172]. Specifically, ZnO nanospikes obtained from 6-week separate incubation periods exhibited the highest effect on P. aeruginosa virulence factors, without affecting bacterial growth. Conversely, titanium dioxide NPs were observed to affect QS only when complexed with silver [173].

4.3. Quorum Quenching Enzymes

Another strategy to interrupt cell-to-cell communication is the removal of signalling molecules from the environment. In this context, several studies focused on the effect of several molecules on QS pathways and quorum quenching (QQ) possibilities in several bacterial model systems [174,175,176]. The discovery of AHL antagonists able to interfere with bacterial QS signalling and induce the accelerated degradation of the AHL-dependent transcription factors attracted many researchers [177]. Several bacterial isolates that can degrade AHL by hydrolyzing the lactone bond (acyl-homoserine lactonase) and the amide linkage (AHL-acylase) have been identified [178,179,180]. Most enzyme-based QS inhibition systems involve applications against BF-producing strains of P. aeruginosa [181]. Recently, Packiavathy et al. reported for the first time AHL-lactonase-mediated QQ activity from marine sediment bacteria Psychrobacter sp. [182]. To provide evidence of the specificity in QQ enzymes, Rémy et al. investigated the activity of two lactonases targeting the signal molecules N-(3-oxododecanoyl)-L-homoserine lactone and butyryl-homoserine lactone in P. aeruginosa PA14 [183]. They observed a similar decreasing effect of AHL concentrations and QS gene expression associated with them. On the other hand, only the lactonase with lower efficacy on butyryl-homoserine lactone was able to inhibit P. aeruginosa’s pathogenicity [183]. Khalid et al. identified several bacterial strains with QQ activity and subsequently tested them against an MDR P. aeruginosa [184]. Their findings suggest that QQ bacterial strains and their products could be a strategy to neutralize pathogenic BF formation. To significantly increase the lactonase activity for reducing EPS and BFs and altering cell surface hydrophobicity, biofunctionalization approaches using silver and gold have been evaluated against MDR K. pneumoniae and Proteus species [185,186]. Gupta et al. indicated good activity from the silver-coated lactonase without side effects on tissue cells, suggesting it is a suitable template for designing novel anti-BF drugs [185]. Similar results have been obtained by Vinoj et al. against BFs from Proteus species [186]. As for the other AHL-degrading enzyme, that is acylase, similar results were obtained [187,188,189]. As described above, nanohybrid strategies based on acylase enzymes and metal NPs, graphene, or antibiotics have also been suggested for obtaining a system with enhanced antibacterial and anti-BF activities. Ivanova et al. obtained silver NPs decorated by the layer-by-layer coating of amino-cellulose and acylase able to inhibit QS-regulated virulence factors from Chromobacterium violaceum and BF formation from P. aeruginosa [190]. The same research groups obtained an enhanced antibacterial effect of gentamicin with a synergistic effect on the BF due to the combination of the antibiotic with acylase [27]. Finally, other authors describe nanoparticle systems based on graphene oxide or polyurethane with acylase to obtain inhibitory action on BF formation and to mitigate the membrane’s biofouling [191,192]. Some antibodies can interfere in bacteria cell-to-cell signalling, in addition to being biocompatible and very efficient. Marin et al. firstly report antibody-based QS-inhibition, due to the hydrolysis of N-(3-oxo-acyl) homoserine lactone mediated by monoclonal antibodies, indicating XYD-11G2 as the most efficient for inhibiting QS in P. aeruginosa [193]. Similarly, Kaufmann et al. suggested an immuno-pharmacotherapeutic approach against P. aeruginosa infections by using the monoclonal antibody RS2-1G9 [194].

5. Anti-BF Strategies Acting on EPS

As previously described, the structure of the extracellular polymeric substance (EPS) allows for bacteria to protect themselves from dehydration, antibiotics, and drug penetration at bactericidal concentrations [24]. Strategies that aim to target the integrity and components of the EPS represent a promising anti-BF technique as affecting the integrity of the EPS matrix leads to the degradation of the BF [105]. The fundamental components of the matrix are proteins, polysaccharides, extracellular nucleic acids (eDNA), and some membrane vesicles. These components can act as targets to prevent the formation of the BF or for its destruction [24]. For example, anti-BF substances could prevent the polymerization and therefore the functionalization of proteins, preventing the formation of the BF itself [47]. Polysaccharides could also be used as targets for the inhibition of BF production [60]. For example, Psl and Pel in P. aeruginosa inhibit BF formation in S. aureus [31]. Conversely, in S. aureus protein-A inhibits Psl in P. aeruginosa and consequently the formation of the BF by the latter microorganism [195]. The main strategies that affect the integrity of the components of the extracellular polymeric substance will be described below (Figure 5).

5.1. Enzymes That Act on EPS Components

Different types of enzymes act on EPS components which lead to the disruption of the BF [105]. However, it is necessary to point out that various enzymes act as virulence factors, as they allow for the components of the EPS matrix to be degraded to promote bacterial dispersion (the last stage of the life cycle of BF formation) [196]. Dispersin B (DspB) is a protein responsible for the degradation of the BF of Actinobacillus pleuropneumoniea [197]. This allows for the dispersion of bacterial cells that can adhere to new nearby surfaces and therefore lead to an extension of the BF. Recombinant DspB is capable of destroying mature S. epidermidis BFs even at low concentrations. This characteristic is because DspB can specifically disrupt poly-N-acetylglucosamine (PNAG) which is one of the main polysaccharides of the BF of S. epidermidis [198,199]. Chen and Lee demonstrated how the combination of DspB with silver-binding peptide leads to the destruction of the matrix, and thanks to the production of AgNPs in situ, the dispersed cells are killed [200]. These authors demonstrate that although DspB is a virulence factor, with appropriate modifications, it can be used as an anti-BF strategy. Lefebvre et al. have demonstrated that proteases combined with ethylenediamine tetra-acetic acid (EDTA) can destabilize the BF and have been used for the eradication of S. aureus and P. aeruginosa BFs in patients with chronic wounds [201]. Nucleases can be considered an anti-BF [202,203]. Deoxyribonuclease I (DNase I) can degrade eDNA, causing a chain reaction that leads to a decrease in EPS matrix biomass, and as a result, makes the BF less resistant to any antibiotics [204]. Based on the same principle, Rubini et al. demonstrated that the combination of DNase with essential oils (EOs) leads to a reduction in the EPS by 85% [205]. Instead, according to Powell et al., the use of alginate oligosaccharide (OligoG) inhibits BF formation by causing an alteration in the EPS [206].

5.2. EPS Disruption Mediated by Nanoparticles

The use of enzymes as an anti-BF strategy is effective, but their use is limited by the high costs involved and the possible instability of the enzymes themselves [207]. In addition, mature BF makes it difficult to reach the deeper layers of the matrix. Consequently, systems capable of combining enzymes, antimicrobial agents, and nanoparticles have been designed to facilitate the dispersion of EPs and the destruction of cells in the deeper layers and to also prevent new colonization [196]. It is well known that nanoparticles can be used as anti-BF, mainly thanks to the electrostatic interactions between the NPs and the components of the EPS matrix [24]. NO-releasing silica NPs demonstrated the capacity to kill BF-based microbial cells, demonstrating how the use of nanoparticles for delivering is a promising strategy as an antimicrobial agent to microbial BFs [106]. When incorporated with silver NPs into alginate hydrogel, NO can be used for topical antibacterial applications with promising results for local applications in the combat of bacterial infections [208]. Different types of NPs have been combined with DNase and antimicrobial agents [24]. Tan et al. have effectively eradicated S. aureus mature BF (24 and 48 h-old) thanks to the use of positively charged chitosan NPs co-encapsulating oxacillin and DNase I. Furthermore, this system did not present cytotoxicity in the HaCat cell line (human immortalized keratinocytes) [209]. Several authors have used co-immobilized DNase I and cellobiose dehydrogenase in chitosan NPs in Candida albicans and S. aureus BFs obtaining excellent results in terms of BF destruction [210,211]. Meanwhile, Liu et al. designed MOF/Ce-based nanozymes with deoxyribonuclease (DNase) and peroxidase mimetic activities. This system can prevent bacteria from recolonizing thanks to the peroxidase-like activity of MOFs and the ability of cerium (IV) complexes to hydrolyze eDNA and disrupt established BFs [212].

5.3. Electrochemical Method to Deteriorate EPS

The “bioelectric effect” indicates the combination of low doses of antibiotic in a weak electric field, with the aim of disintegrating the mature biofilm [105]. It is possible to stimulate the detachment of the biofilm from a conductive surface through the application of a direct current [213]. The antibacterial activity of the electric current can be traced back to the production of toxic substances (for example, H2O2 and oxidizing radicals) following electrolysis but also to membrane damage with the consequent loss of cytoplasmic constituents [105,214]. These effects contribute to improving the minimum inhibitory concentration level leading to increased antibiotic sensitivity among BF and drug-resistant bacteria [215,216,217]. Antimicrobial agents under the influence of the electric field alter the permeability of the EPS matrix, causing the leakage of biocidal ions. The influx of those biocide ions destroys the bacterial cells through electrophoresis and electro-osmosis [218,219]. Blenkinsopp et al. have shown that this effect is not obtained with the sole application of electric current, in the absence of antimicrobial agents [220]. The antimicrobial effect related to electrical current also depends on the voltage during the electrical stimulation, as it affects the membrane potential and electrophysiology [221,222]. Alternating current (AC) or direct current (DC), or both, help implement the effect of antibiotics even at low doses [219,223,224]. Even the use of low temperature plasma, under low current, influences cell adhesion, as it decreases the EPS intensity surrounding the bacterial cells [225].

6. Anti-BF Strategies Mediated by Phage

Bacteriophages, or simply phages, are viruses that infect bacteria and are host-dependent during self-replication [226]. Each phage has a receptor-binding protein positioned on the tail fibre, which confers specificity for a selective bacteria host [227]. Recently, with the increase in AMR, the research focus has gradually made a comeback to develop phage-based treatments able to combat pathogenic bacteria infection and also BF formation [226]. In addition, since phages are natural killers of bacteria, they represent an excellent therapeutic agent not only in clinical applications but also in other areas, such as agriculture, food control, or industry, due to their specificity and ecological safety. The antibacterial activity of phages is carried out by depolymerase and lysins, responsible for degrading capsular polysaccharides and peptidoglycan in bacterial cells, respectively [228,229,230]. Phages can be applied to prevent BF formation or to destroy existing BFs. This last strategy can be classified into the following: (i) phage therapy, based on the intra- to extracellular degradation of the bacterial cell (using a single or cocktail of lytic phages); (ii) phage-derived enzyme based on the extra- to intracellular degradation of the bacterial matrix (using lysins and/or depolymerases); (iii) the combination of phages with other antimicrobial biotic or abiotic elements; (iv) the genetic modification of phage structure or genome. In this paragraph, we focus on the four ways of phage-mediated BF remotion (Figure 6).

6.1. Phage Therapy

During the intra- to extracellular degradation, a phage, in the first, uses the depolymerases, present on the viral tail structure, to penetrate the BF matrix. At this point, the phage interacts with bacteria hosts leading to viral infection by genome injection [231]. The formation of lytic progenies is accompanied by the activation of holins and endolysins, responsible for piercing the cytoplasmic membrane and the degradation of bacterial peptidoglycans [232]. The use of the entire phage structure, single or in a cocktail mix, is defined as phage therapy and has shown to be effective in eradicating bacterial BF exploiting the natural ability of the phage to kill bacteria [233]. Morris et al. demonstrated a 3.3-fold reduction in BF biomass caused by S. aureus on three-dimensional-printed titanium after 48 h of exposure to the StaPhage cocktail, based on the combination of five S. aureus-specific bacteriophages [234]. On the other hand, phages in a gel-like matrix have been coated on the catheter to reduce planktonic forms and BFs of 50 tested uropathogenic P. mirabilis strains found on the surface [235]. Moreover, phages PSTCR4 and PSTCR6, as part of 17 characterized novel phages, exhibited an efficient reduction in well-established P. stuartii BFs formed in catheter models [236]. Phages, isolated from human saliva samples, showed the effective prevention and reduction in the existing BF of S. mutans in cariogenic dentin models, such as a decrease of up to 97% in the expression of genes involved in BF production [237,238]. Manoharadas et al. used the combination of Φ44AHJD and ΦX174 phages to disrupt the hybrid BF of S. aureus and E. coli after 72 h of incubation [239]. They also demonstrated that the use of a single phage to the mixed E. coliS. aureus, instead, promoted the formation of the BF by the alternate strain that was not affected by the phage. A recent study showed that a phage cocktail based on four lytic phages inhibited the growth of MDR E. coli and caused a strong biomass reduction in the BF up to nearly 87% [240,241].

6.2. Phage-Derived Enzyme

As mentioned above, the enzymes depolymerases and lyases are used by phages to dissolve the BF matrix and to cleave bacterial cell walls causing the release of viral progenies, respectively. However, depolymerases are typically encoded as part of the phage structure and as such can be used as tail spike protein (TSP) or as free enzymes in the treatment of BF formation. Lysins are generally produced at the end of the phage lytic replication cycle, and in the context of BF degradation, can also be used as free enzymes [242]. For example, the depolymerase Dpo42, extracted from vB_EcoM_ECOO78 E. coli phage, showed the efficient degradation of the E. coli’s capsular polysaccharides (CPS) as well as the prevention of E. coli BF formation [243]. Gutiérrez et al. demonstrated the ability to inhibit and also disperse over 90% of BFs generated by different strains of S. epidermidis and S. aureus when using EPS depolymerase Dpo7, derived from bacteriophage vB_SepiS-phiIPLA7 [244]. In another study, recombinant TSP Dep42, from phage SH-KP152226, showed specific enzymatic activities against the K47 capsule of K. pneumoniae leading to the inhibition or degradation of its BFs. The study also showed that the combination of Dep42 with antibiotics could enhance polymyxin activity against K. pneumoniae BFs [245]. Recently, Shahed-Al-Mahmud et al. used φAB6 TSP to treat A. baumannii-adhered catheters and observed significantly fewer bacteria cells after 4 h of treatment. In an in vivo test, it was demonstrated that after 4 days, φAB6 TSP-treated zebrafish presented significantly higher survival rates compared to those without TSP treatment, suggesting the use of the treatment against MDR A. baumannii infections in the near future [246]. On the other hand, the use of lysin CF-301 as an anti-BF agent removed BFs from S. aureus or mixed-species on several surfaces, such as polystyrene, glass, surgical mesh, and catheters, with an improvement in anti-BF activity when combined with cell wall hydrolase lysostaphin [247]. Similarly, endolysin LysCSA13 showed high efficacy in removing about 80–90% of staphylococcal BF biomass on various surfaces [248]. Moreover, Yuan et al. showed the broad spectrum of antimicrobial activity of endolysin Abtn-4, isolated from A. baumannii phage D2, against MDR S. aureus, P. aeruginosa, K. pneumoniae, Enterococcus, and Salmonella, which in turn also resulted in being able to reduce formed BFs [249]. Chimeric lysin ClyH or ClyF has been found to reduce a large percentage of the BF mass of MRSA strains [250]. Recently, Vasina et al. found four endolysins LysAm24, LysAp22, LysECD7, and LysSi3 with high antibacterial activity against Gram-negative bacteria both in vitro and in vivo [251].

6.3. Phage Combination with Other Elements

Recent studies suggest that coupling phages with antibiotics or nanomaterials with antibacterial activity displays either the synergy or facilitation of BF treatment [29]. For example, the combination of phages with ciprofloxacin showed a synergistic effect, killing >6 log CFUs/g of fibrin clots within 6 h, in 64% of treated rats with experimental endocarditis caused by P. aeruginosa [252]. Similarly, a phage treatment before vancomycin or cefazolin exposure was more effective at eliminating S. aureus BF-associated cells. Probably, the high phage density led to the destruction of the BF matrix, then the antibiotic therapy was more efficient [253]. Recently, Cano et al. observed a biomass reduction in BF-associated prosthetic knee infection, after in vivo treatment with KpJH46Φ2 phage in combination with minocycline [254]. Stachler et al. demonstrated a potential synergistic effect between phages and chemical disinfection, such as sodium hypochlorite and benzalkonium chloride, in the remotion of BFs based on pathogen P. aeruginosa on the surfaces and to prevent the regeneration of dry BFs at the same time [255]. Recently, in some studies, phages were coupled with nanomaterials through physical adsorption to develop innovative alternatives for eradicating pathogenic BFs [256]. In another study, Podoviridae phages functionalized on magnetic nanoparticles removed about 95% of the multispecies BF (i.e., E. coli and P. aeruginosa) after 6 h of treatment [257].

6.4. Phage Engineering

Natural phage therapy is linked to the narrow host range and specificity [258]. However, phages can be modified by genetic engineering techniques to extend the host specificity and increase BF degradation for much broader applications [259]. Lu and Collins used engineered T7 phages to express dispersin B making it able to attack bacterial cells and facilitate the degradation of the EPS of the E. coli BF, resulting in a significant reduction of about 4.5 orders of magnitude of bacteria and 99% of BF mass [260]. A T7 bacteriophage was also engineered to encode a lactonase enzyme (AiiA), which has broad-range activity for the quenching of quorum sensing necessary for BF formation. The addition of this engineered T7 phage to mixed-species BFs based on P. aeruginosa and E. coli resulted in the inhibition of BF formation [261]. Moreover, Born et al. inserted the depolymerase dpoL1 gene into the genome of phage Y2, which led to enhanced bacterial killing and had a positive effect on the dispersion of the E. amylovora BF [262]. Phage efficacy has also been enhanced by genetic mutation leading to the conversion from the lysogenic to the lytic phage cycle. This change enabled the successful treatment of disseminated drug-resistant M. abscessus infection [263]. A recent study demonstrated that the recombinant receptor of the T4-like phage conferred to the engineered phage the ability to lyse four additional hosts compared to wild-type phages, allowing for a significant inhibitory effect on mixed E. coli [264]. Moreover, the phage display technique, developed in 1985 by George P. Smith [265], has been applied to find peptides with the ability to degrade BFs. Phage display is based on the expression of foreign peptides on phage capsid proteins. Starting from a pool of engineered phage particles, each one with a different random peptide exposed on its capsid, the selection of an engineered phage that selectively binds to a target is obtained by biopanning cycles. This process consists of the immobilization of the target on a surface to expose it to phage peptide libraries. Then, phages that did not bind efficiently to the target are washed, while strongly phage-binding targets are eluted by different methods. The phage display technique has been used to find peptides able to detect enzymes, whole eukaryotic and prokaryotic cells, including MDR strains, and assess if they presented microbicidal activities [266,267,268,269]. In addition, it has been observed that foreign peptides can be modified in their conformation on the phage surfaces and resistance to chemical–physical environment compared to the wild-type [270,271]. These findings expand the use of engineered phages as bio-probes for medical applications. In the future the ability to specifically recognize and interact with bacteria targets could be used to deliver any antibacterial and/or anti-BF agents.

7. Conclusions

Biofilm formation and the increase in bacterial antibiotic resistance are causing considerable concern in the scientific community. Although there are many anti-BF strategies, we believe that new alternatives are needed so that, especially when referring to human health, we can find effective solutions, which are easy to apply and easily replicable. There is the possibility of destroying mature BFs, but this strategy seems to be the least applicable, as it could lead to dispersion and therefore to an aggravation of the problem. The possibility of preventing BF formation could represent the best strategy to stop bacteria from creating an environment favourable to their proliferation, protected from external agents and capable of resisting antibiotics. New anti-BF strategies could overcome the now widespread and inexorable resistance to antibiotics. For example, in a hospital environment, materials capable of avoiding BF formation and bacterial proliferation can not only improve healing but also avoid potential chronic or even fatal infections, considerably reducing recovery times and the, in some ways excessive, use of antibiotics.

Author Contributions

A.I. and L.M.D.P., writing—original draft preparation; A.C., writing—review and editing; D.F. and S.C., conceptualization, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Koo, H.; Allan, R.N.; Howlin, R.P.; Stoodley, P.; Hall-Stoodley, L. Targeting Microbial Biofilms: Current and Prospective Therapeutic Strategies. Nat. Rev. Microbiol. 2017, 15, 740–755. [Google Scholar] [CrossRef] [PubMed]
  2. Li, J.; Liu, D.; Tian, X.; Koseki, S.; Chen, S.; Ye, X.; Ding, T. Novel Antibacterial Modalities against Methicillin Resistant Staphylococcus aureus Derived from Plants. Crit. Rev. Food Sci. Nutr. 2019, 59, S153–S161. [Google Scholar] [CrossRef] [PubMed]
  3. Guzmán-Soto, I.; McTiernan, C.; Gonzalez-Gomez, M.; Ross, A.; Gupta, K.; Suuronen, E.J.; Mah, T.-F.; Griffith, M.; Alarcon, E.I. Mimicking Biofilm Formation and Development: Recent Progress in In Vitro and In Vivo Biofilm Models. iScience 2021, 24, 102443. [Google Scholar] [CrossRef] [PubMed]
  4. Abisado, R.G.; Benomar, S.; Klaus, J.R.; Dandekar, A.A.; Chandler, J.R. Bacterial Quorum Sensing and Microbial Community Interactions. mBio 2018, 9, e02331-17. [Google Scholar] [CrossRef] [PubMed]
  5. Yi, L.; Li, J.; Liu, B.; Wang, Y. Advances in Research on Signal Molecules Regulating Biofilms. World J. Microbiol. Biotechnol. 2019, 35, 130. [Google Scholar] [CrossRef] [PubMed]
  6. Preda, V.G.; Săndulescu, O. Communication Is the Key: Biofilms, Quorum Sensing, Formation and Prevention. Discoveries 2019, 7, e100. [Google Scholar] [CrossRef]
  7. Fang, K.; Park, O.-J.; Hong, S.H. Controlling Biofilms Using Synthetic Biology Approaches. Biotechnol. Adv. 2020, 40, 107518. [Google Scholar] [CrossRef]
  8. Anju, V.T.; Busi, S.; Imchen, M.; Kumavath, R.; Mohan, M.S.; Salim, S.A.; Subhaswaraj, P.; Dyavaiah, M. Polymicrobial Infections and Biofilms: Clinical Significance and Eradication Strategies. Antibiotics 2022, 11, 1731. [Google Scholar] [CrossRef]
  9. Tian, F.; Li, J.; Nazir, A.; Tong, Y. Bacteriophage—A Promising Alternative Measure for Bacterial Biofilm Control. Infect. Drug Resist. 2021, 14, 205–217. [Google Scholar] [CrossRef]
  10. Wicaksono, W.A.; Erschen, S.; Krause, R.; Müller, H.; Cernava, T.; Berg, G. Enhanced Survival of Multi-Species Biofilms under Stress Is Promoted by Low-Abundant but Antimicrobial-Resistant Keystone Species. J. Hazard. Mater. 2022, 422, 126836. [Google Scholar] [CrossRef]
  11. Khan, A.A.; Manzoor, K.N.; Sultan, A.; Saeed, M.; Rafique, M.; Noushad, S.; Talib, A.; Rentschler, S.; Deigner, H.-P. Pulling the Brakes on Fast and Furious Multiple Drug-Resistant (MDR) Bacteria. Int. J. Mol. Sci. 2021, 22, 859. [Google Scholar] [CrossRef] [PubMed]
  12. Michaelis, C.; Grohmann, E. Horizontal Gene Transfer of Antibiotic Resistance Genes in Biofilms. Antibiotics 2023, 12, 328. [Google Scholar] [CrossRef] [PubMed]
  13. CDC. The Biggest Antibiotic-Resistant Threats in the U.S. Available online: https://www.cdc.gov/drugresistance/biggest-threats.html (accessed on 5 April 2022).
  14. De Oliveira, D.M.P.; Forde, B.M.; Kidd, T.J.; Harris, P.N.A.; Schembri, M.A.; Beatson, S.A.; Paterson, D.L.; Walker, M.J. Antimicrobial Resistance in ESKAPE Pathogens. Clin. Microbiol. Rev. 2020, 33, e00181-19. [Google Scholar] [CrossRef] [PubMed]
  15. Schulze, A.; Mitterer, F.; Pombo, J.P.; Schild, S. Biofilms by Bacterial Human Pathogens: Clinical Relevance—Development, Composition and Regulation—Therapeutical Strategies. Microb. Cell 2021, 8, 28. [Google Scholar] [CrossRef]
  16. Minkiewicz-Zochniak, A.; Jarzynka, S.; Iwańska, A.; Strom, K.; Iwańczyk, B.; Bartel, M.; Mazur, M.; Pietruczuk-Padzik, A.; Konieczna, M.; Augustynowicz-Kopeć, E.; et al. Biofilm Formation on Dental Implant Biomaterials by Staphylococcus aureus Strains Isolated from Patients with Cystic Fibrosis. Materials 2021, 14, 2030. [Google Scholar] [CrossRef] [PubMed]
  17. Su, Y.; Yrastorza, J.T.; Matis, M.; Cusick, J.; Zhao, S.; Wang, G.; Xie, J. Biofilms: Formation, Research Models, Potential Targets, and Methods for Prevention and Treatment. Adv. Sci. 2022, 9, 2203291. [Google Scholar] [CrossRef] [PubMed]
  18. Roy, R.; Tiwari, M.; Donelli, G.; Tiwari, V. Strategies for Combating Bacterial Biofilms: A Focus on Anti-Biofilm Agents and Their Mechanisms of Action. Virulence 2017, 9, 522–554. [Google Scholar] [CrossRef] [PubMed]
  19. Delik, E.; Eroğlu, B.; Çolak, Ç.Y.; Özçelik, A.T.; Tefon Öztürk, B.E. Alterations of Growth, Biofilm-Forming, and Gene Expression of Bordetella Pertussis by Antibiotics at Sub-Minimum Inhibitory Concentrations. Res. Microbiol. 2023, 174, 104058. [Google Scholar] [CrossRef]
  20. Rather, M.A.; Mandal, M. Attenuation of Biofilm and Quorum Sensing Regulated Virulence Factors of an Opportunistic Pathogen Pseudomonas aeruginosa by Phytofabricated Silver Nanoparticles. Microb. Pathog. 2023, 185, 106433. [Google Scholar] [CrossRef]
  21. Li, J.; Liu, H.; Zhao, C.; Zhang, J.; He, W. Autoinducer-2 Quorum Sensing Regulates Biofilm Formation and Chain Elongation Metabolic Pathways to Enhance Caproate Synthesis in Microbial Electrochemical System. Chemosphere 2023, 344, 140384. [Google Scholar] [CrossRef]
  22. Li, Y.; Wang, S.; Ding, H.; Xiao, K.; Huang, X. Quorum Sensing-Fe Metabolism Interplay Affects Biofouling on Reverse Osmosis Membrane: Evidences from Microbial Shift and Structure Alteration. Desalination 2023, 551, 116416. [Google Scholar] [CrossRef]
  23. Mirghani, R.; Saba, T.; Khaliq, H.; Mitchell, J.; Do, L.; Chambi, L.; Diaz, K.; Kennedy, T.; Alkassab, K.; Huynh, T.; et al. Biofilms: Formation, Drug Resistance and Alternatives to Conventional Approaches. AIMS Microbiol. 2022, 8, 239–277. [Google Scholar] [CrossRef]
  24. Pinto, R.M.; Soares, F.A.; Reis, S.; Nunes, C.; Van Dijck, P. Innovative Strategies toward the Disassembly of the EPS Matrix in Bacterial Biofilms. Front. Microbiol. 2020, 11, 952. [Google Scholar] [CrossRef]
  25. Ramakrishnan, R.; Singh, A.K.; Singh, S.; Chakravortty, D.; Das, D. Enzymatic Dispersion of Biofilms: An Emerging Biocatalytic Avenue to Combat Biofilm-Mediated Microbial Infections. J. Biol. Chem. 2022, 298, 102352. [Google Scholar] [CrossRef] [PubMed]
  26. Silpe, J.E.; Duddy, O.P.; Bassler, B.L. Natural and Synthetic Inhibitors of a Phage-Encoded Quorum-Sensing Receptor Affect Phage–Host Dynamics in Mixed Bacterial Communities. Proc. Natl. Acad. Sci. USA 2022, 119, e2217813119. [Google Scholar] [CrossRef] [PubMed]
  27. Ivanova, K.; Ivanova, A.; Hoyo, J.; Pérez-Rafael, S.; Tzanov, T. Nano-Formulation Endows Quorum Quenching Enzyme-Antibiotic Hybrids with Improved Antibacterial and Antibiofilm Activities against Pseudomonas aeruginosa. Int. J. Mol. Sci. 2022, 23, 7632. [Google Scholar] [CrossRef]
  28. Kotrange, H.; Najda, A.; Bains, A.; Gruszecki, R.; Chawla, P.; Tosif, M.M. Metal and Metal Oxide Nanoparticle as a Novel Antibiotic Carrier for the Direct Delivery of Antibiotics. Int. J. Mol. Sci. 2021, 22, 9596. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, S.; Lu, H.; Zhang, S.; Shi, Y.; Chen, Q. Phages against Pathogenic Bacterial Biofilms and Biofilm-Based Infections: A Review. Pharmaceutics 2022, 14, 427. [Google Scholar] [CrossRef]
  30. Sheridan, M.; Winters, C.; Zamboni, F.; Collins, M.N. Biomaterials: Antimicrobial Surfaces in Biomedical Engineering and Healthcare. Curr. Opin. Biomed. Eng. 2022, 22, 100373. [Google Scholar] [CrossRef]
  31. Beaudoin, C.M.; Cox, Z.; Dundore, T.; Thomas, T.; Kim, J.; Pillivant, D. Effect of Bench Press Load Knowledge on Repetitions, Rating of Perceived Exertion, and Attentional Focus. J. Strength Cond. Res. 2018, 32, 514. [Google Scholar] [CrossRef]
  32. Lopes, S.P.; Azevedo, N.F.; Pereira, M.O. Quantitative Assessment of Individual Populations within Polymicrobial Biofilms. Sci. Rep. 2018, 8, 9494. [Google Scholar] [CrossRef] [PubMed]
  33. Rumbaugh, K.P.; Sauer, K. Biofilm Dispersion. Nat. Rev. Microbiol. 2020, 18, 571–586. [Google Scholar] [CrossRef] [PubMed]
  34. Kragh, K.N.; Hutchison, J.B.; Melaugh, G.; Rodesney, C.; Roberts, A.E.L.; Irie, Y.; Jensen, P.Ø.; Diggle, S.P.; Allen, R.J.; Gordon, V.; et al. Role of Multicellular Aggregates in Biofilm Formation. mBio 2016, 7, e00237-16. [Google Scholar] [CrossRef] [PubMed]
  35. Carniello, V.; Peterson, B.W.; van der Mei, H.C.; Busscher, H.J. Physico-Chemistry from Initial Bacterial Adhesion to Surface-Programmed Biofilm Growth. Adv. Colloid Interface Sci. 2018, 261, 1–14. [Google Scholar] [CrossRef]
  36. Zheng, S.; Bawazir, M.; Dhall, A.; Kim, H.-E.; He, L.; Heo, J.; Hwang, G. Implication of Surface Properties, Bacterial Motility, and Hydrodynamic Conditions on Bacterial Surface Sensing and Their Initial Adhesion. Front. Bioeng. Biotechnol. 2021, 9, 643722. [Google Scholar] [CrossRef] [PubMed]
  37. Berne, C.; Ellison, C.K.; Ducret, A.; Brun, Y.V. Bacterial Adhesion at the Single-Cell Level. Nat. Rev. Microbiol. 2018, 16, 616–627. [Google Scholar] [CrossRef] [PubMed]
  38. Yang, A.; Tang, W.S.; Si, T.; Tang, J.X. Influence of Physical Effects on the Swarming Motility of Pseudomonas aeruginosa. Biophys. J. 2017, 112, 1462–1471. [Google Scholar] [CrossRef]
  39. Berne, C. Sticky Decisions: The Multilayered Regulation of Adhesin Production by Bacteria. PLoS Genet. 2023, 19, e1010648. [Google Scholar] [CrossRef]
  40. Singh, S.; Datta, S.; Narayanan, K.B.; Rajnish, K.N. Bacterial Exo-Polysaccharides in Biofilms: Role in Antimicrobial Resistance and Treatments. J. Genet. Eng. Biotechnol. 2021, 19, 140. [Google Scholar] [CrossRef]
  41. Schilcher, K.; Horswill, A.R. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol. Mol. Biol. Rev. 2020, 84, e00026-19. [Google Scholar] [CrossRef]
  42. Toyofuku, M.; Inaba, T.; Kiyokawa, T.; Obana, N.; Yawata, Y.; Nomura, N. Environmental Factors That Shape Biofilm Formation. Biosci. Biotechnol. Biochem. 2016, 80, 7–12. [Google Scholar] [CrossRef] [PubMed]
  43. Rather, M.A.; Gupta, K.; Mandal, M. Microbial Biofilm: Formation, Architecture, Antibiotic Resistance, and Control Strategies. Braz. J. Microbiol. 2021, 52, 1701–1718. [Google Scholar] [CrossRef] [PubMed]
  44. Ma, R.; Hu, X.; Zhang, X.; Wang, W.; Sun, J.; Su, Z.; Zhu, C. Strategies to Prevent, Curb and Eliminate Biofilm Formation Based on the Characteristics of Various Periods in One Biofilm Life Cycle. Front. Cell. Infect. Microbiol. 2022, 12, 1003033. [Google Scholar] [CrossRef] [PubMed]
  45. Valentin, J.D.P.; Straub, H.; Pietsch, F.; Lemare, M.; Ahrens, C.H.; Schreiber, F.; Webb, J.S.; van der Mei, H.C.; Ren, Q. Role of the Flagellar Hook in the Structural Development and Antibiotic Tolerance of Pseudomonas aeruginosa Biofilms. ISME J. 2022, 16, 1176–1186. [Google Scholar] [CrossRef] [PubMed]
  46. Zhai, Y.; Tian, W.; Chen, K.; Lan, L.; Kan, J.; Shi, H. Flagella-Mediated Adhesion of Escherichia coli O157:H7 to Surface of Stainless Steel, Glass and Fresh Produces during Sublethal Injury and Recovery. Food Microbiol. 2024, 117, 104383. [Google Scholar] [CrossRef]
  47. Ruhal, R.; Kataria, R. Biofilm Patterns in Gram-Positive and Gram-Negative Bacteria. Microbiol. Res. 2021, 251, 126829. [Google Scholar] [CrossRef]
  48. Foster, T.J.; Geoghegan, J.A.; Ganesh, V.K.; Höök, M. Adhesion, Invasion and Evasion: The Many Functions of the Surface Proteins of Staphylococcus aureus. Nat. Rev. Microbiol. 2014, 12, 49–62. [Google Scholar] [CrossRef] [PubMed]
  49. Schniederberend, M.; Williams, J.F.; Shine, E.; Shen, C.; Jain, R.; Emonet, T.; Kazmierczak, B.I. Modulation of Flagellar Rotation in Surface-Attached Bacteria: A Pathway for Rapid Surface-Sensing after Flagellar Attachment. PLoS Pathog. 2019, 15, e1008149. [Google Scholar] [CrossRef]
  50. Oh, J.K.; Yegin, Y.; Yang, F.; Zhang, M.; Li, J.; Huang, S.; Verkhoturov, S.V.; Schweikert, E.A.; Perez-Lewis, K.; Scholar, E.A.; et al. The Influence of Surface Chemistry on the Kinetics and Thermodynamics of Bacterial Adhesion. Sci. Rep. 2018, 8, 17247. [Google Scholar] [CrossRef]
  51. Dufrêne, Y.F.; Viljoen, A. Binding Strength of Gram-Positive Bacterial Adhesins. Front. Microbiol. 2020, 11, 1457. [Google Scholar] [CrossRef]
  52. Bucher, T.; Oppenheimer-Shaanan, Y.; Savidor, A.; Bloom-Ackermann, Z.; Kolodkin-Gal, I. Disturbance of the Bacterial Cell Wall Specifically Interferes with Biofilm Formation. Env. Microbiol. Rep. 2015, 7, 990–1004. [Google Scholar] [CrossRef] [PubMed]
  53. Abdel-Rhman, S.H. Role of Pseudomonas aeruginosa Lipopolysaccharides in Modulation of Biofilm and Virulence Factors of Enterobacteriaceae. Ann. Microbiol. 2019, 69, 299–305. [Google Scholar] [CrossRef]
  54. Huszczynski, S.M.; Lam, J.S.; Khursigara, C.M. The Role of Pseudomonas aeruginosa Lipopolysaccharide in Bacterial Pathogenesis and Physiology. Pathogens 2019, 9, 6. [Google Scholar] [CrossRef] [PubMed]
  55. Li, P.; Yin, R.; Cheng, J.; Lin, J. Bacterial Biofilm Formation on Biomaterials and Approaches to Its Treatment and Prevention. Int. J. Mol. Sci. 2023, 24, 11680. [Google Scholar] [CrossRef] [PubMed]
  56. Shree, P.; Singh, C.K.; Sodhi, K.K.; Surya, J.N.; Singh, D.K. Biofilms: Understanding the Structure and Contribution towards Bacterial Resistance in Antibiotics. Med. Microecol. 2023, 16, 100084. [Google Scholar] [CrossRef]
  57. Gondil, V.S.; Subhadra, B. Biofilms and Their Role on Diseases. BMC Microbiol. 2023, 23, 203. [Google Scholar] [CrossRef]
  58. Li, S.; Duan, G.; Xi, Y.; Chu, Y.; Li, F.; Ho, S.-H. Insights into the Role of Extracellular Polymeric Substances (EPS) in the Spread of Antibiotic Resistance Genes. Env. Pollut. 2023, 343, 123285. [Google Scholar] [CrossRef]
  59. Molina-Santiago, C.; de Vicente, A.; Romero, D. Bacterial Extracellular Matrix as a Natural Source of Biotechnologically Multivalent Materials. Comput. Struct. Biotechnol. J. 2021, 19, 2796–2805. [Google Scholar] [CrossRef]
  60. Cugini, C.; Shanmugam, M.; Landge, N.; Ramasubbu, N. The Role of Exopolysaccharides in Oral Biofilms. J. Dent. Res. 2019, 98, 739–745. [Google Scholar] [CrossRef]
  61. Latasa, C.; Solano, C.; Penadés, J.R.; Lasa, I. Biofilm-Associated Proteins. Comptes. Rendus. Biol. 2006, 329, 849–857. [Google Scholar] [CrossRef]
  62. Valle, J.; Fang, X.; Lasa, I. Revisiting Bap Multidomain Protein: More than Sticking Bacteria Together. Front. Microbiol. 2020, 11, 613581. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Y.; Jiang, J.; Gao, Y.; Sun, Y.; Dai, J.; Wu, Y.; Qu, D.; Ma, G.; Fang, X. Staphylococcus epidermidis Small Basic Protein (Sbp) Forms Amyloid Fibrils, Consistent with Its Function as a Scaffolding Protein in Biofilms. J. Biol. Chem. 2018, 293, 14296–14311. [Google Scholar] [CrossRef] [PubMed]
  64. Bleem, A.; Christiansen, G.; Madsen, D.J.; Maric, H.; Strømgaard, K.; Bryers, J.D.; Daggett, V.; Meyer, R.L.; Otzen, D.E. Protein Engineering Reveals Mechanisms of Functional Amyloid Formation in Pseudomonas aeruginosa Biofilms. J. Mol. Biol. 2018, 430, 3751–3763. [Google Scholar] [CrossRef]
  65. França, A.; Gaio, V.; Lopes, N.; Melo, L.D.R. Virulence Factors in Coagulase-Negative Staphylococci. Pathogens 2021, 10, 170. [Google Scholar] [CrossRef] [PubMed]
  66. Chung, J.; Eisha, S.; Park, S.; Morris, A.J.; Martin, I. How Three Self-Secreted Biofilm Exopolysaccharides of Pseudomonas aeruginosa, Psl, Pel, and Alginate, Can Each Be Exploited for Antibiotic Adjuvant Effects in Cystic Fibrosis Lung Infection. Int. J. Mol. Sci. 2023, 24, 8709. [Google Scholar] [CrossRef] [PubMed]
  67. Cordisco, E.; Zanor, M.I.; Moreno, D.M.; Serra, D.O. Selective Inhibition of the Amyloid Matrix of Escherichia coli Biofilms by a Bifunctional Microbial Metabolite. NPJ Biofilms Microbiomes 2023, 9, 81. [Google Scholar] [CrossRef]
  68. Ray, V.A.; Hill, P.J.; Stover, C.K.; Roy, S.; Sen, C.K.; Yu, L.; Wozniak, D.J.; DiGiandomenico, A. Anti-Psl Targeting of Pseudomonas aeruginosa Biofilms for Neutrophil-Mediated Disruption. Sci. Rep. 2017, 7, 16065. [Google Scholar] [CrossRef]
  69. Soliman, C.; Walduck, A.K.; Yuriev, E.; Richards, J.S.; Cywes-Bentley, C.; Pier, G.B.; Ramsland, P.A. Structural Basis for Antibody Targeting of the Broadly Expressed Microbial Polysaccharide Poly-N-Acetylglucosamine. J. Biol. Chem. 2018, 293, 5079–5089. [Google Scholar] [CrossRef]
  70. DeFrancesco, A.S.; Masloboeva, N.; Syed, A.K.; DeLoughery, A.; Bradshaw, N.; Li, G.-W.; Gilmore, M.S.; Walker, S.; Losick, R. Genome-Wide Screen for Genes Involved in eDNA Release during Biofilm Formation by Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 2017, 114, E5969–E5978. [Google Scholar] [CrossRef]
  71. Lee, A.R.; Park, S.B.; Kim, S.W.; Jung, J.W.; Chun, J.H.; Kim, J.; Kim, Y.R.; Lazarte, J.M.S.; Jang, H.B.; Thompson, K.D.; et al. Membrane Vesicles from Antibiotic-resistant Staphylococcus aureus Transfer Antibiotic-Resistance to Antibiotic-Susceptible Escherichia coli. J. Appl. Microbiol. 2022, 132, 2746–2759. [Google Scholar] [CrossRef]
  72. Wang, W.; Chanda, W.; Zhong, M. The Relationship between Biofilm and Outer Membrane Vesicles: A Novel Therapy Overview. FEMS Microbiol. Lett. 2015, 362, fnv117. [Google Scholar] [CrossRef] [PubMed]
  73. Juodeikis, R.; Carding, S.R. Outer Membrane Vesicles: Biogenesis, Functions, and Issues. Microbiol. Mol. Biol. Rev. 2022, 86, e00032-22. [Google Scholar] [CrossRef]
  74. Henke, J.M.; Bassler, B.L. Three Parallel Quorum-Sensing Systems Regulate Gene Expression in Vibrio Harveyi. J. Bacteriol. 2004, 186, 6902–6914. [Google Scholar] [CrossRef] [PubMed]
  75. Higgins, D.A.; Pomianek, M.E.; Kraml, C.M.; Taylor, R.K.; Semmelhack, M.F.; Bassler, B.L. The Major Vibrio Cholerae Autoinducer and Its Role in Virulence Factor Production. Nature 2007, 450, 883–886. [Google Scholar] [CrossRef] [PubMed]
  76. Wu, L.; Luo, Y. Bacterial Quorum-Sensing Systems and Their Role in Intestinal Bacteria-Host Crosstalk. Front. Microbiol. 2021, 12, 611413. [Google Scholar] [CrossRef] [PubMed]
  77. Mukherjee, S.; Bassler, B.L. Bacterial Quorum Sensing in Complex and Dynamically Changing Environments. Nat. Rev. Microbiol. 2019, 17, 371–382. [Google Scholar] [CrossRef] [PubMed]
  78. McBrayer, D.N.; Cameron, C.D.; Tal-Gan, Y. Development and Utilization of Peptide-Based Quorum Sensing Modulators in Gram-Positive Bacteria. Org. Biomol. Chem. 2020, 18, 7273–7290. [Google Scholar] [CrossRef]
  79. Papenfort, K.; Bassler, B.L. Quorum Sensing Signal–Response Systems in Gram-Negative Bacteria. Nat. Rev. Microbiol. 2016, 14, 576–588. [Google Scholar] [CrossRef]
  80. Loughran, A.J.; Atwood, D.N.; Anthony, A.C.; Harik, N.S.; Spencer, H.J.; Beenken, K.E.; Smeltzer, M.S. Impact of Individual Extracellular Proteases on Staphylococcus aureus Biofilm Formation in Diverse Clinical Isolates and Their Isogenic sarA Mutants. MicrobiologyOpen 2014, 3, 897–909. [Google Scholar] [CrossRef]
  81. Peng, Q.; Tang, X.; Dong, W.; Sun, N.; Yuan, W. A Review of Biofilm Formation of Staphylococcus aureus and Its Regulation Mechanism. Antibiotics 2022, 12, 12. [Google Scholar] [CrossRef]
  82. Coquant, G.; Grill, J.-P.; Seksik, P. Impact of N-Acyl-Homoserine Lactones, Quorum Sensing Molecules, on Gut Immunity. Front. Immunol. 2020, 11, 1827. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, D.J.; Jo, A.R.; Jang, M.C.; Nam, J.; Choi, H.J.; Choi, G.-W.; Sung, H.Y.; Bae, H.; Ku, Y.-G.; Chi, Y.-T. Analysis of Two Quorum Sensing-Deficient Isolates of Pseudomonas aeruginosa. Microb. Pathog. 2018, 119, 162–169. [Google Scholar] [CrossRef] [PubMed]
  84. Paul, E.; Mathur, P.; Sharma, C.; Chaturvedi, P. Chapter 9—Quorum Quenching Strategies of Endophytic Bacteria: Role in Plant Protection. In Plant-Microbe Interaction—Recent Advances in Molecular and Biochemical Approaches; Swapnil, P., Meena, M., Harish, Marwal, A., Vijayalakshmi, S., Zehra, A., Eds.; Academic Press: Cambridge, MA, USA, 2023; pp. 149–166. ISBN 978-0-323-91876-3. [Google Scholar]
  85. Wang, Y.; Wang, Y.; Sun, L.; Grenier, D.; Yi, L. The LuxS/AI-2 System of Streptococcus Suis. Appl. Microbiol. Biotechnol. 2018, 102, 7231–7238. [Google Scholar] [CrossRef] [PubMed]
  86. Guo, X.-P.; Sun, Y.-C. New Insights into the Non-Orthodox Two Component Rcs Phosphorelay System. Front. Microbiol. 2017, 8, 2014. [Google Scholar] [CrossRef] [PubMed]
  87. Qin, S.; Xiao, W.; Zhou, C.; Pu, Q.; Deng, X.; Lan, L.; Liang, H.; Song, X.; Wu, M. Pseudomonas aeruginosa: Pathogenesis, Virulence Factors, Antibiotic Resistance, Interaction with Host, Technology Advances and Emerging Therapeutics. Signal Transduct. Target. Ther. 2022, 7, 199. [Google Scholar] [CrossRef]
  88. The Two-Component Signal Transduction System and Its Regulation in Candida albicans. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8274445/ (accessed on 18 November 2023).
  89. Ghilini, F.; Pissinis, D.E.; Miñán, A.; Schilardi, P.L.; Diaz, C. How Functionalized Surfaces Can Inhibit Bacterial Adhesion and Viability. ACS Biomater. Sci. Eng. 2019, 5, 4920–4936. [Google Scholar] [CrossRef] [PubMed]
  90. Kreve, S.; Reis, A.C.D. Bacterial Adhesion to Biomaterials: What Regulates This Attachment? A Review. Jpn. Dent. Sci. Rev. 2021, 57, 85–96. [Google Scholar] [CrossRef]
  91. Pecoraro, C.; Carbone, D.; Parrino, B.; Cascioferro, S.; Diana, P. Recent Developments in the Inhibition of Bacterial Adhesion as Promising Anti-Virulence Strategy. Int. J. Mol. Sci. 2023, 24, 4872. [Google Scholar] [CrossRef]
  92. Chug, M.K.; Brisbois, E.J. Recent Developments in Multifunctional Antimicrobial Surfaces and Applications toward Advanced Nitric Oxide-Based Biomaterials. ACS Mater. Au 2022, 2, 525–551. [Google Scholar] [CrossRef]
  93. Alves, D.; Pereira, M.O. Bio-Inspired Coating Strategies for the Immobilization of Polymyxins to Generate Contact-Killing Surfaces. Macromol. Biosci. 2016, 16, 1450–1460. [Google Scholar] [CrossRef]
  94. He, M.; Gao, K.; Zhou, L.; Jiao, Z.; Wu, M.; Cao, J.; You, X.; Cai, Z.; Su, Y.; Jiang, Z. Zwitterionic Materials for Antifouling Membrane Surface Construction. Acta Biomaterialia 2016, 40, 142–152. [Google Scholar] [CrossRef]
  95. Colilla, M.; Izquierdo-Barba, I.; Vallet-Regí, M. The Role of Zwitterionic Materials in the Fight against Proteins and Bacteria. Medicines 2018, 5, 125. [Google Scholar] [CrossRef] [PubMed]
  96. Lemire, J.A.; Kalan, L.; Bradu, A.; Turner, R.J. Silver Oxynitrate, an Unexplored Silver Compound with Antimicrobial and Antibiofilm Activity. Antimicrob. Agents Chemother. 2015, 59, 4031–4039. [Google Scholar] [CrossRef] [PubMed]
  97. Kalan, L.R.; Pepin, D.M.; Ul-Haq, I.; Miller, S.B.; Hay, M.E.; Precht, R.J. Targeting Biofilms of Multidrug-Resistant Bacteria with Silver Oxynitrate. Int. J. Antimicrob. Agents 2017, 49, 719–726. [Google Scholar] [CrossRef] [PubMed]
  98. Lemire, J.A.; Kalan, L.; Gugala, N.; Bradu, A.; Turner, R.J. Silver Oxynitrate—An Efficacious Compound for the Prevention and Eradication of Dual-Species Biofilms. Biofouling 2017, 33, 460–469. [Google Scholar] [CrossRef] [PubMed]
  99. Chung, P.Y.; Toh, Y.S. Anti-Biofilm Agents: Recent Breakthrough against Multi-Drug Resistant Staphylococcus Aureus. Pathog. Dis. 2014, 70, 231–239. [Google Scholar] [CrossRef] [PubMed]
  100. Ferreres, G.; Ivanova, K.; Ivanov, I.; Tzanov, T. Nanomaterials and Coatings for Managing Antibiotic-Resistant Biofilms. Antibiotics 2023, 12, 310. [Google Scholar] [CrossRef]
  101. Xiang, J.; Zhu, R.; Lang, S.; Yan, H.; Liu, G.; Peng, B. Mussel-Inspired Immobilization of Zwitterionic Silver Nanoparticles toward Antibacterial Cotton Gauze for Promoting Wound Healing. Chem. Eng. J. 2021, 409, 128291. [Google Scholar] [CrossRef]
  102. Mgomi, F.C.; Yang, Y.; Cheng, G.; Yang, Z. Lactic acid bacteria biofilms and their antimicrobial potential against pathogenic microorganisms. Biofilm 2023, 5, 100118. [Google Scholar] [CrossRef]
  103. Winkelströter, L.K.; Gomes, B.C.; Thomaz, M.R.S.; Souza, V.M.; De Martinis, E.C.P. Lactobacillus sakei 1 and Its Bacteriocin Influence Adhesion of Listeria monocytogenes on Stainless Steel Surface. Food Control 2011, 22, 1404–1407. [Google Scholar] [CrossRef]
  104. Pimentel-Filho, N.D.J.; Martins, M.C.D.F.; Nogueira, G.B.; Mantovani, H.C.; Vanetti, M.C.D. Bovicin HC5 and Nisin Reduce Staphylococcus aureus Adhesion to Polystyrene and Change the Hydrophobicity Profile and Gibbs Free Energy of Adhesion. Int. J. Food Microbiol. 2014, 190, 1–8. [Google Scholar] [CrossRef] [PubMed]
  105. Srinivasan, R.; Santhakumari, S.; Poonguzhali, P.; Geetha, M.; Dyavaiah, M.; Xiangmin, L. Bacterial Biofilm Inhibition: A Focused Review on Recent Therapeutic Strategies for Combating the Biofilm Mediated Infections. Front. Microbiol. 2021, 12, 676458. [Google Scholar] [CrossRef] [PubMed]
  106. Hetrick, E.M.; Shin, J.H.; Paul, H.S.; Schoenfisch, M.H. Anti-Biofilm Efficacy of Nitric Oxide-Releasing Silica Nanoparticles. Biomaterials 2009, 30, 2782–2789. [Google Scholar] [CrossRef] [PubMed]
  107. Hemeg, H.A. Combatting Persisted and Biofilm Antimicrobial Resistant Bacterial by Using Nanoparticles. Z. Naturforsch. C J. Biosci. 2022, 77, 365–378. [Google Scholar] [CrossRef] [PubMed]
  108. Agirre, J.; Moroz, O.; Meier, S.; Brask, J.; Munch, A.; Hoff, T.; Andersen, C.; Wilson, K.S.; Davies, G.J. The Structure of the AliC GH13 α-Amylase from Alicyclobacillus sp. Reveals the Accommodation of Starch Branching Points in the α-Amylase Family. Acta Crystallogr. D Struct. Biol. 2019, 75, 1–7. [Google Scholar] [CrossRef] [PubMed]
  109. Abeleda, H.E.P.; Javier, A.P.; Murillo, A.Q.M.; Baculi, R.Q. Alpha-Amylase Conjugated Biogenic Silver Nanoparticles as Innovative Strategy against Biofilm-Forming Multidrug Resistant Bacteria. Biocatal. Agric. Biotechnol. 2020, 29, 101784. [Google Scholar] [CrossRef]
  110. Ivanova, A.; Ivanova, K.; Perelshtein, I.; Gedanken, A.; Todorova, K.; Milcheva, R.; Dimitrov, P.; Popova, T.; Tzanov, T. Sonochemically Engineered Nano-Enabled Zinc Oxide/Amylase Coatings Prevent the Occurrence of Catheter-Associated Urinary Tract Infections. Mater. Sci. Eng. C 2021, 131, 112518. [Google Scholar] [CrossRef] [PubMed]
  111. Xin, X.; Li, P.; Zhu, Y.; Shi, L.; Yuan, J.; Shen, J. Mussel-Inspired Surface Functionalization of PET with Zwitterions and Silver Nanoparticles for the Dual-Enhanced Antifouling and Antibacterial Properties. Langmuir 2019, 35, 1788–1797. [Google Scholar] [CrossRef]
  112. More, P.R.; Pandit, S.; Filippis, A.D.; Franci, G.; Mijakovic, I.; Galdiero, M. Silver Nanoparticles: Bactericidal and Mechanistic Approach against Drug Resistant Pathogens. Microorganisms 2023, 11, 369. [Google Scholar] [CrossRef]
  113. Islam, T.; Rahaman, M.M.; Mia, M.N.; Ara, I.; Islam, M.T.; Alam Riaz, T.; Araújo, A.C.J.; de Lima Silva, J.M.F.; de Lacerda, B.C.G.V.; de Andrade, E.M.; et al. Therapeutic Perspectives of Metal Nanoformulations. Drugs Drug Candidates 2023, 2, 232–278. [Google Scholar] [CrossRef]
  114. Chowdhury, N.; Kwan, B.W.; Wood, T.K. Persistence Increases in the Absence of the Alarmone Guanosine Tetraphosphate by Reducing Cell Growth. Sci. Rep. 2016, 6, 20519. [Google Scholar] [CrossRef] [PubMed]
  115. Torres, M.R.; Slate, A.J.; Ryder, S.F.; Akram, M.; Iruzubieta, C.J.C.; Whitehead, K.A. Ionic Gold Demonstrates Antimicrobial Activity against Pseudomonas aeruginosa Strains Due to Cellular Ultrastructure Damage. Arch. Microbiol. 2021, 203, 3015–3024. [Google Scholar] [CrossRef] [PubMed]
  116. Khan, F.; Kang, M.-G.; Jo, D.-M.; Chandika, P.; Jung, W.-K.; Kang, H.W.; Kim, Y.-M. Phloroglucinol-Gold and -Zinc Oxide Nanoparticles: Antibiofilm and Antivirulence Activities towards Pseudomonasaeruginosa PAO1. Mar. Drugs 2021, 19, 601. [Google Scholar] [CrossRef]
  117. Wang, Q.; Zhang, Y.; Li, Q.; Chen, L.; Liu, H.; Ding, M.; Dong, H.; Mou, Y. Therapeutic Applications of Antimicrobial Silver-Based Biomaterials in Dentistry. Int. J. Nanomed. 2022, 17, 443–462. [Google Scholar] [CrossRef] [PubMed]
  118. Ramalingam, B.; Parandhaman, T.; Das, S.K. Antibacterial Effects of Biosynthesized Silver Nanoparticles on Surface Ultrastructure and Nanomechanical Properties of Gram-Negative Bacteria Viz. Escherichia coli and Pseudomonas aeruginosa. ACS Appl. Mater. Interfaces 2016, 8, 4963–4976. [Google Scholar] [CrossRef] [PubMed]
  119. Song, Z.; Wu, Y.; Wang, H.; Han, H. Synergistic Antibacterial Effects of Curcumin Modified Silver Nanoparticles through ROS-Mediated Pathways. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 99, 255–263. [Google Scholar] [CrossRef]
  120. Kadiyala, U.; Turali-Emre, E.S.; Bahng, J.H.; Kotov, N.A.; VanEpps, J.S. Unexpected Insights into Antibacterial Activity of Zinc Oxide Nanoparticles against Methicillin Resistant Staphylococcus aureus (MRSA). Nanoscale 2018, 10, 4927–4939. [Google Scholar] [CrossRef]
  121. Zhang, H.; Zhang, Z.; Li, J.; Qin, G. New Strategies for Biocontrol of Bacterial Toxins and Virulence: Focusing on Quorum-Sensing Interference and Biofilm Inhibition. Toxins 2023, 15, 570. [Google Scholar] [CrossRef]
  122. Raju, D.V.; Nagarajan, A.; Pandit, S.; Nag, M.; Lahiri, D.; Upadhye, V. Effect of Bacterial Quorum Sensing and Mechanism of Antimicrobial Resistance. Biocatal. Agric. Biotechnol. 2022, 43, 102409. [Google Scholar] [CrossRef]
  123. Vashistha, A.; Sharma, N.; Nanaji, Y.; Kumar, D.; Singh, G.; Barnwal, R.P.; Yadav, A.K. Quorum Sensing Inhibitors as Therapeutics: Bacterial Biofilm Inhibition. Bioorganic Chem. 2023, 136, 106551. [Google Scholar] [CrossRef]
  124. Verderosa, A.D.; Totsika, M.; Fairfull-Smith, K.E. Bacterial Biofilm Eradication Agents: A Current Review. Front. Chem. 2019, 7, 824. [Google Scholar] [CrossRef] [PubMed]
  125. Grandclément, C.; Tannières, M.; Moréra, S.; Dessaux, Y.; Faure, D. Quorum Quenching: Role in Nature and Applied Developments. FEMS Microbiol. Rev. 2016, 40, 86–116. [Google Scholar] [CrossRef] [PubMed]
  126. Vasconcelos, N.G.; Croda, J.; Simionatto, S. Antibacterial Mechanisms of Cinnamon and Its Constituents: A Review. Microb. Pathog. 2018, 120, 198–203. [Google Scholar] [CrossRef]
  127. Niu, C.; Afre, S.; Gilbert, E.S. Subinhibitory Concentrations of Cinnamaldehyde Interfere with Quorum Sensing. Lett. Appl. Microbiol. 2006, 43, 489–494. [Google Scholar] [CrossRef]
  128. Topa, S.H.; Palombo, E.A.; Kingshott, P.; Blackall, L.L. Activity of Cinnamaldehyde on Quorum Sensing and Biofilm Susceptibility to Antibiotics in Pseudomonas aeruginosa. Microorganisms 2020, 8, 455. [Google Scholar] [CrossRef] [PubMed]
  129. Li, S.; Zhou, S.; Yang, Q.; Liu, Y.; Yang, Y.; Xu, N.; Ai, X.; Dong, J. Cinnamaldehyde Decreases the Pathogenesis of Aeromonas hydrophila by Inhibiting Quorum Sensing and Biofilm Formation. Fishes 2023, 8, 122. [Google Scholar] [CrossRef]
  130. Xu, J.; Lin, Q.; Sheng, M.; Ding, T.; Li, B.; Gao, Y.; Tan, Y. Antibiofilm Effect of Cinnamaldehyde-Chitosan Nanoparticles against the Biofilm of Staphylococcus aureus. Antibiotics 2022, 11, 1403. [Google Scholar] [CrossRef]
  131. Subhaswaraj, P.; Barik, S.; Macha, C.; Chiranjeevi, P.V.; Siddhardha, B. Anti Quorum Sensing and Anti Biofilm Efficacy of Cinnamaldehyde Encapsulated Chitosan Nanoparticles against Pseudomonas aeruginosa PAO1. LWT 2018, 97, 752–759. [Google Scholar] [CrossRef]
  132. Ramasamy, M.; Lee, J.-H.; Lee, J. Development of Gold Nanoparticles Coated with Silica Containing the Antibiofilm Drug Cinnamaldehyde and Their Effects on Pathogenic Bacteria. IJN 2017, 12, 2813–2828. [Google Scholar] [CrossRef]
  133. Vasavi, H.S.; Arun, A.B.; Rekha, P.-D. Anti-Quorum Sensing Activity of Psidium guajava L. Flavonoids against Chromobacterium violaceum and Pseudomonas aeruginosa PAO1. Microbiol. Immunol. 2014, 58, 286–293. [Google Scholar] [CrossRef]
  134. Paczkowski, J.E.; Mukherjee, S.; McCready, A.R.; Cong, J.-P.; Aquino, C.J.; Kim, H.; Henke, B.R.; Smith, C.D.; Bassler, B.L. Flavonoids Suppress Pseudomonas aeruginosa Virulence through Allosteric Inhibition of Quorum-Sensing Receptors. J. Biol. Chem. 2017, 292, 4064–4076. [Google Scholar] [CrossRef] [PubMed]
  135. Roshni, P.S.; Alexpandi, R.; Abirami, G.; Durgadevi, R.; Cai, Y.; Kumar, P.; Ravi, A.V. Hesperidin Methyl Chalcone, a Citrus Flavonoid, Inhibits Aeromonas hydrophila Infection Mediated by Quorum Sensing. Microb. Pathog. 2023, 177, 106029. [Google Scholar] [CrossRef] [PubMed]
  136. Pachaiappan, R.; Rajamuthu, T.P.; Sarkar, A.; Natrajan, P.; Krishnan, N.; Sakthivelu, M.; Velusamy, P.; Ramasamy, P.; Gopinath, S.C.B. N-Acyl-Homoserine Lactone Mediated Virulence Factor(s) of Pseudomonas aeruginosa Inhibited by Flavonoids and Isoflavonoids. Process. Biochem. 2022, 116, 84–93. [Google Scholar] [CrossRef]
  137. Omwenga, E.O.; Hensel, A.; Shitandi, A.; Goycoolea, F.M. Chitosan Nanoencapsulation of Flavonoids Enhances Their Quorum Sensing and Biofilm Formation Inhibitory Activities against an E. coli Top 10 Biosensor. Colloids Surf. B Biointerfaces 2018, 164, 125–133. [Google Scholar] [CrossRef] [PubMed]
  138. Tran, T.-T.; Hadinoto, K. A Potential Quorum-Sensing Inhibitor for Bronchiectasis Therapy: Quercetin-Chitosan Nanoparticle Complex Exhibiting Superior Inhibition of Biofilm Formation and Swimming Motility of Pseudomonas aeruginosa to the Native Quercetin. Int. J. Mol. Sci. 2021, 22, 1541. [Google Scholar] [CrossRef] [PubMed]
  139. Fernando, S.I.D.; Judan Cruz, K.G. Ethnobotanical Biosynthesis of Gold Nanoparticles and Its Downregulation of Quorum Sensing-Linked AhyR Gene in Aeromonas hydrophila. SN Appl. Sci. 2020, 2, 570. [Google Scholar] [CrossRef]
  140. Ilk, S.; Tan, G.; Emül, E.; Sağlam, N. Investigation the Potential Use of Silver Nanoparticles Synthesized by Propolis Extract as N-Acyl-Homoserine Lactone-Mediated Quorum Sensing Systems Inhibitor. Turk. J. Med. Sci. 2020, 50, 1147–1156. [Google Scholar] [CrossRef]
  141. Ashrafudoulla, M.; Mizan, M.F.R.; Ha, A.J.; Park, S.H.; Ha, S.-D. Antibacterial and Antibiofilm Mechanism of Eugenol against Antibiotic Resistance Vibrio Parahaemolyticus. Food Microbiol. 2020, 91, 103500. [Google Scholar] [CrossRef]
  142. Fekrirad, Z.; Gattali, B.; Kashef, N. Quorum Sensing-Regulated Functions of Serratia Marcescens Are Reduced by Eugenol. Iran. J. Microbiol. 2020, 12, 451–459. [Google Scholar] [CrossRef]
  143. Nabawy, A.; Makabenta, J.M.; Schmidt-Malan, S.; Park, J.; Li, C.-H.; Huang, R.; Fedeli, S.; Chattopadhyay, A.N.; Patel, R.; Rotello, V.M. Dual Antimicrobial-Loaded Biodegradable Nanoemulsions for Synergistic Treatment of Wound Biofilms. J. Control. Release 2022, 347, 379–388. [Google Scholar] [CrossRef]
  144. Sethuram, L.; Thomas, J.; Mukherjee, A.; Chandrasekaran, N. Eugenol Micro-Emulsion Reinforced with Silver Nanocomposite Electrospun Mats for Wound Dressing Strategies. Mater. Adv. 2021, 2, 2971–2988. [Google Scholar] [CrossRef]
  145. Lou, Z.; Letsididi, K.S.; Yu, F.; Pei, Z.; Wang, H.; Letsididi, R. Inhibitive Effect of Eugenol and Its Nanoemulsion on Quorum Sensing–Mediated Virulence Factors and Biofilm Formation by Pseudomonas aeruginosa. J. Food Prot. 2019, 82, 379–389. [Google Scholar] [CrossRef] [PubMed]
  146. Antunes, J.C.; Tavares, T.D.; Teixeira, M.A.; Teixeira, M.O.; Homem, N.C.; Amorim, M.T.P.; Felgueiras, H.P. Eugenol-Containing Essential Oils Loaded onto Chitosan/Polyvinyl Alcohol Blended Films and Their Ability to Eradicate Staphylococcus aureus or Pseudomonas aeruginosa from Infected Microenvironments. Pharmaceutics 2021, 13, 195. [Google Scholar] [CrossRef] [PubMed]
  147. Sofer, D.; Gilboa-Garber, N.; Belz, A.; Garber, N.C. “Subinhibitory” Erythromycin Represses Production of Pseudomonas aeruginosa Lectins, Autoinducer and Virulence Factors. Chemotherapy 1999, 45, 335–341. [Google Scholar] [CrossRef] [PubMed]
  148. Tateda, K.; Comte, R.; Pechere, J.-C.; Köhler, T.; Yamaguchi, K.; Van Delden, C. Azithromycin Inhibits Quorum Sensing in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2001, 45, 1930–1933. [Google Scholar] [CrossRef] [PubMed]
  149. Henkel, M.; Schmidberger, A.; Kühnert, C.; Beuker, J.; Bernard, T.; Schwartz, T.; Syldatk, C.; Hausmann, R. Kinetic Modeling of the Time Course of N-Butyryl-Homoserine Lactone Concentration during Batch Cultivations of Pseudomonas aeruginosa PAO1. Appl. Microbiol. Biotechnol. 2013, 97, 7607–7616. [Google Scholar] [CrossRef] [PubMed]
  150. Zeng, J.; Zhang, N.; Huang, B.; Cai, R.; Wu, B.; Shunmei, E.; Fang, C.; Chen, C. Mechanism of Azithromycin Inhibition of HSL Synthesis in Pseudomonas aeruginosa. Sci. Rep. 2016, 6, 24299. [Google Scholar] [CrossRef]
  151. El-Mowafy, S.A.; Abd El Galil, K.H.; Habib, E.-S.E.; Shaaban, M.I. Quorum Sensing Inhibitory Activity of Sub-Inhibitory Concentrations of β-Lactams. Afr. Health Sci. 2017, 17, 199–207. [Google Scholar] [CrossRef]
  152. Gao, Y.; Wang, J.; Chai, M.; Li, X.; Deng, Y.; Jin, Q.; Ji, J. Size and Charge Adaptive Clustered Nanoparticles Targeting the Biofilm Microenvironment for Chronic Lung Infection Management. ACS Nano 2020, 14, 5686–5699. [Google Scholar] [CrossRef]
  153. Kłodzińska, S.N.; Wan, F.; Jumaa, H.; Sternberg, C.; Rades, T.; Nielsen, H.M. Utilizing Nanoparticles for Improving Anti-Biofilm Effects of Azithromycin: A Head-to-Head Comparison of Modified Hyaluronic Acid Nanogels and Coated Poly (Lactic-Co-Glycolic Acid) Nanoparticles. J. Colloid Interface Sci. 2019, 555, 595–606. [Google Scholar] [CrossRef]
  154. Jiang, K.; Yan, X.; Yu, J.; Xiao, Z.; Wu, H.; Zhao, M.; Yue, Y.; Zhou, X.; Xiao, J.; Lin, F. Design, Synthesis, and Biological Evaluation of 3-Amino-2-Oxazolidinone Derivatives as Potent Quorum-Sensing Inhibitors of Pseudomonas aeruginosa PAO1. Eur. J. Med. Chem. 2020, 194, 112252. [Google Scholar] [CrossRef] [PubMed]
  155. Miller, K. Bacterial Communication and Its Role as a Target for Nanoparticle-Based Antimicrobial Therapy. Ph.D. Thesis, University of South Carolina, Columbia, SC, USA, 2015. [Google Scholar]
  156. Franco, D.; Zagami, R.; De Plano, L.M.; Burduja, N.; Guglielmino, S.P.P.; Scolaro, L.M.; Mazzaglia, A. Antimicrobial and Antibiofilm Photodynamic Action of Photosensitizing Nanoassemblies Based on Sulfobutylether-β-Cyclodextrin. Molecules 2023, 28, 2493. [Google Scholar] [CrossRef] [PubMed]
  157. Thanh Nguyen, H.; Goycoolea, F.M. Chitosan/Cyclodextrin/TPP Nanoparticles Loaded with Quercetin as Novel Bacterial Quorum Sensing Inhibitors. Molecules 2017, 22, 1975. [Google Scholar] [CrossRef] [PubMed]
  158. Slavin, Y.N.; Asnis, J.; Häfeli, U.O.; Bach, H. Metal Nanoparticles: Understanding the Mechanisms behind Antibacterial Activity. J. Nanobiotechnology 2017, 15, 65. [Google Scholar] [CrossRef] [PubMed]
  159. Godoy-Gallardo, M.; Eckhard, U.; Delgado, L.M.; de Roo Puente, Y.J.D.; Hoyos-Nogués, M.; Gil, F.J.; Perez, R.A. Antibacterial Approaches in Tissue Engineering Using Metal Ions and Nanoparticles: From Mechanisms to Applications. Bioact. Mater. 2021, 6, 4470–4490. [Google Scholar] [CrossRef] [PubMed]
  160. Franco, D.; Calabrese, G.; Guglielmino, S.P.P.; Conoci, S. Metal-Based Nanoparticles: Antibacterial Mechanisms and Biomedical Application. Microorganisms 2022, 10, 1778. [Google Scholar] [CrossRef]
  161. Gómez-Gómez, B.; Arregui, L.; Serrano, S.; Santos, A.; Pérez-Corona, T.; Madrid, Y. Unravelling Mechanisms of Bacterial Quorum Sensing Disruption by Metal-Based Nanoparticles. Sci. Total Environ. 2019, 696, 133869. [Google Scholar] [CrossRef] [PubMed]
  162. Srinivasan, R.; Vigneshwari, L.; Rajavel, T.; Durgadevi, R.; Kannappan, A.; Balamurugan, K.; Pandima Devi, K.; Veera Ravi, A. Biogenic Synthesis of Silver Nanoparticles Using Piper Betle Aqueous Extract and Evaluation of Its Anti-Quorum Sensing and Antibiofilm Potential against Uropathogens with Cytotoxic Effects: An in Vitro and in Vivo Approach. Env. Sci. Pollut. Res. 2018, 25, 10538–10554. [Google Scholar] [CrossRef]
  163. Shah, S.; Gaikwad, S.; Nagar, S.; Kulshrestha, S.; Vaidya, V.; Nawani, N.; Pawar, S. Biofilm Inhibition and Anti-Quorum Sensing Activity of Phytosynthesized Silver Nanoparticles against the Nosocomial Pathogen Pseudomonas aeruginosa. Biofouling 2019, 35, 34–49. [Google Scholar] [CrossRef]
  164. Kumar, S.; Paliya, B.S.; Singh, B.N. Superior Inhibition of Virulence and Biofilm Formation of Pseudomonas aeruginosa PAO1 by Phyto-Synthesized Silver Nanoparticles through Anti-Quorum Sensing Activity. Microb. Pathog. 2022, 170, 105678. [Google Scholar] [CrossRef]
  165. Saeki, E.K.; Martins, H.M.; de Camargo, L.C.; Anversa, L.; Tavares, E.R.; Yamada-Ogatta, S.F.; Lioni, L.M.Y.; Kobayashi, R.K.T.; Nakazato, G. Effect of Biogenic Silver Nanoparticles on the Quorum-Sensing System of Pseudomonas aeruginosa PAO1 and PA14. Microorganisms 2022, 10, 1755. [Google Scholar] [CrossRef] [PubMed]
  166. Elshaer, S.L.; Shaaban, M.I. Inhibition of Quorum Sensing and Virulence Factors of Pseudomonas aeruginosa by Biologically Synthesized Gold and Selenium Nanoparticles. Antibiotics 2021, 10, 1461. [Google Scholar] [CrossRef] [PubMed]
  167. Qais, F.A.; Ahmad, I.; Altaf, M.; Alotaibi, S.H. Biofabrication of Gold Nanoparticles Using Capsicum Annuum Extract and Its Antiquorum Sensing and Antibiofilm Activity against Bacterial Pathogens. ACS Omega 2021, 6, 16670–16682. [Google Scholar] [CrossRef] [PubMed]
  168. Gómez-Gómez, B.; Arregui, L.; Serrano, S.; Santos, A.; Pérez-Corona, T.; Madrid, Y. Selenium and Tellurium-Based Nanoparticles as Interfering Factors in Quorum Sensing-Regulated Processes: Violacein Production and Bacterial Biofilm Formation. Metallomics 2019, 11, 1104–1114. [Google Scholar] [CrossRef] [PubMed]
  169. Maruthupandy, M.; Rajivgandhi, G.N.; Quero, F.; Li, W.-J. Anti-Quorum Sensing and Anti-Biofilm Activity of Nickel Oxide Nanoparticles against Pseudomonas aeruginosa. J. Environ. Chem. Eng. 2020, 8, 104533. [Google Scholar] [CrossRef]
  170. Calabrese, G.; Franco, D.; Petralia, S.; Monforte, F.; Condorelli, G.G.; Squarzoni, S.; Traina, F.; Conoci, S. Dual-Functional Nano-Functionalized Titanium Scaffolds to Inhibit Bacterial Growth and Enhance Osteointegration. Nanomaterials 2021, 11, 2634. [Google Scholar] [CrossRef] [PubMed]
  171. Calabrese, G.; De Luca, G.; Franco, D.; Morganti, D.; Rizzo, M.G.; Bonavita, A.; Neri, G.; Fazio, E.; Neri, F.; Fazio, B.; et al. Structural and Antibacterial Studies of Novel ZnO and ZnxMn(1-x)O Nanostructured Titanium Scaffolds for Biomedical Applications. Biomater. Adv. 2023, 145, 213193. [Google Scholar] [CrossRef] [PubMed]
  172. Khan, M.F.; Husain, F.M.; Zia, Q.; Ahmad, E.; Jamal, A.; Alaidarous, M.; Banawas, S.; Alam, M.D.M.; Alshehri, B.A.; Jameel, M.; et al. Anti-Quorum Sensing and Anti-Biofilm Activity of Zinc Oxide Nanospikes. ACS Omega 2020, 5, 32203–32215. [Google Scholar] [CrossRef]
  173. Naik, K.; Kowshik, M. Anti-Quorum Sensing Activity of AgCl-TiO2 Nanoparticles with Potential Use as Active Food Packaging Material. J. Appl. Microbiol. 2014, 117, 972–983. [Google Scholar] [CrossRef]
  174. Barnaby, R.; Koeppen, K.; Stanton, B.A. Cyclodextrins Reduce the Ability of Pseudomonas aeruginosa Outer-Membrane Vesicles to Reduce CFTR Cl−Secretion. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L206–L215. [Google Scholar] [CrossRef]
  175. Ziegler, E.W.; Brown, A.B.; Nesnas, N.; Chouinard, C.D.; Mehta, A.K.; Palmer, A.G. β-Cyclodextrin Encapsulation of Synthetic AHLs: Drug Delivery Implications and Quorum-Quenching Exploits. ChemBioChem 2021, 22, 1292–1301. [Google Scholar] [CrossRef] [PubMed]
  176. Molnár, M.; Fenyvesi, É.; Berkl, Z.; Németh, I.; Fekete-Kertész, I.; Márton, R.; Vaszita, E.; Varga, E.; Ujj, D.; Szente, L. Cyclodextrin-Mediated Quorum Quenching in the Aliivibrio fischeri Bioluminescence Model System—Modulation of Bacterial Communication. Int. J. Pharm. 2021, 594, 120150. [Google Scholar] [CrossRef] [PubMed]
  177. Lu, L.; Li, M.; Yi, G.; Liao, L.; Cheng, Q.; Zhu, J.; Zhang, B.; Wang, Y.; Chen, Y.; Zeng, M. Screening Strategies for Quorum Sensing Inhibitors in Combating Bacterial Infections. J. Pharm. Anal. 2022, 12, 1–14. [Google Scholar] [CrossRef] [PubMed]
  178. Wang, L.-H.; Weng, L.-X.; Dong, Y.-H.; Zhang, L.-H. Specificity and Enzyme Kinetics of the Quorum-Quenching N-Acyl Homoserine Lactone Lactonase (AHL-Lactonase). J. Biol. Chem. 2004, 279, 13645–13651. [Google Scholar] [CrossRef] [PubMed]
  179. Kusada, H.; Zhang, Y.; Tamaki, H.; Kimura, N.; Kamagata, Y. Novel N-Acyl Homoserine Lactone-Degrading Bacteria Isolated from Penicillin-Contaminated Environments and Their Quorum-Quenching Activities. Front. Microbiol. 2019, 10, 455. [Google Scholar] [CrossRef] [PubMed]
  180. Torabi Delshad, S.; Soltanian, S.; Sharifiyazdi, H.; Haghkhah, M.; Bossier, P. Identification of N-Acyl Homoserine Lactone-Degrading Bacteria Isolated from Rainbow Trout (Oncorhynchus mykiss). J. Appl. Microbiol. 2018, 125, 356–369. [Google Scholar] [CrossRef] [PubMed]
  181. Rather, M.A.; Saha, D.; Bhuyan, S.; Jha, A.N.; Mandal, M. Quorum Quenching: A Drug Discovery Approach Against Pseudomonas aeruginosa. Microbiol. Res. 2022, 264, 127173. [Google Scholar] [CrossRef]
  182. Packiavathy, I.A.S.V.; Kannappan, A.; Thiyagarajan, S.; Srinivasan, R.; Jeyapragash, D.; Paul, J.B.J.; Velmurugan, P.; Ravi, A.V. AHL-Lactonase Producing Psychrobacter SP. From Palk Bay Sediment Mitigates Quorum Sensing-Mediated Virulence Production in Gram Negative Bacterial Pathogens. Front. Microbiol. 2021, 12, 634593. [Google Scholar] [CrossRef]
  183. Rémy, B.; Plener, L.; Decloquement, P.; Armstrong, N.; Elias, M.; Daudé, D.; Chabrière, É. Lactonase Specificity Is Key to Quorum Quenching in Pseudomonas aeruginosa. Front. Microbiol. 2020, 11, 762. [Google Scholar] [CrossRef]
  184. Khalid, S.J.; Ain, Q.; Khan, S.J.; Jalil, A.; Siddiqui, M.F.; Ahmad, T.; Badshah, M.; Adnan, F. Targeting Acyl Homoserine Lactones (AHLs) by the Quorum Quenching Bacterial Strains to Control Biofilm Formation in Pseudomonas aeruginosa. Saudi J. Biol. Sci. 2022, 29, 1673–1682. [Google Scholar] [CrossRef]
  185. Gupta, K.; Chhibber, S. Biofunctionalization of Silver Nanoparticles with Lactonase Leads to Altered Antimicrobial and Cytotoxic Properties. Front. Mol. Biosci. 2019, 6, 450649. [Google Scholar] [CrossRef] [PubMed]
  186. Vinoj, G.; Pati, R.; Sonawane, A.; Vaseeharan, B. In Vitro Cytotoxic Effects of Gold Nanoparticles Coated with Functional Acyl Homoserine Lactone Lactonase Protein from Bacillus licheniformis and Their Antibiofilm Activity against Proteus Species. Antimicrob. Agents Chemother. 2015, 59, 763–771. [Google Scholar] [CrossRef]
  187. Vogel, J.; Jansen, L.; Setroikromo, R.; Cavallo, F.M.; van Dijl, J.M.; Quax, W.J. Fighting Acinetobacter baumannii Infections with the Acylase PvdQ. Microbes Infect. 2022, 24, 104951. [Google Scholar] [CrossRef]
  188. De Celis, M.; Serrano-Aguirre, L.; Belda, I.; Liébana-García, R.; Arroyo, M.; Marquina, D.; de la Mata, I.; Santos, A. Acylase Enzymes Disrupting Quorum Sensing Alter the Transcriptome and Phenotype of Pseudomonas aeruginosa, and the Composition of Bacterial Biofilms from Wastewater Treatment Plants. Sci. Total Environ. 2021, 799, 149401. [Google Scholar] [CrossRef] [PubMed]
  189. Wang, H.; Lin, Q.; Dong, L.; Wu, W.; Liang, Z.; Dong, Z.; Ye, H.; Liao, L.; Zhang, L.-H. A Bacterial Isolate Capable of Quenching Both Diffusible Signal Factor- and N-Acylhomoserine Lactone-Family Quorum Sensing Signals Shows Much Enhanced Biocontrol Potencies. J. Agric. Food Chem. 2022, 70, 7716–7726. [Google Scholar] [CrossRef] [PubMed]
  190. Ivanova, A.; Ivanova, K.; Tied, A.; Heinze, T.; Tzanov, T. Layer-By-Layer Coating of Aminocellulose and Quorum Quenching Acylase on Silver Nanoparticles Synergistically Eradicate Bacteria and Their Biofilms. Adv. Funct. Mater. 2020, 30, 2001284. [Google Scholar] [CrossRef]
  191. Zhu, Z.; Wang, L.; Li, Q. A Bioactive Pol (Vinylidene Fluoride)/Graphene Oxide@acylase Nanohybrid Membrane: Enhanced Anti-Biofouling Based on Quorum Quenching. J. Membr. Sci. 2018, 547, 110–122. [Google Scholar] [CrossRef]
  192. Grover, N.; Plaks, J.G.; Summers, S.R.; Chado, G.R.; Schurr, M.J.; Kaar, J.L. Acylase-Containing Polyurethane Coatings with Anti-Biofilm Activity. Biotechnol. Bioeng. 2016, 113, 2535–2543. [Google Scholar] [CrossRef]
  193. De Lamo Marin, S.; Xu, Y.; Meijler, M.M.; Janda, K.D. Antibody Catalyzed Hydrolysis of a Quorum Sensing Signal Found in Gram-Negative Bacteria. Bioorganic Med. Chem. Lett. 2007, 17, 1549–1552. [Google Scholar] [CrossRef]
  194. Kaufmann, G.F.; Park, J.; Mee, J.M.; Ulevitch, R.J.; Janda, K.D. The Quorum Quenching Antibody RS2-1G9 Protects Macrophages from the Cytotoxic Effects of the Pseudomonas aeruginosa Quorum Sensing Signalling Molecule N-3-Oxo-Dodecanoyl-Homoserine Lactone. Mol. Immunol. 2008, 45, 2710–2714. [Google Scholar] [CrossRef]
  195. Armbruster, C.R.; Wolter, D.J.; Mishra, M.; Hayden, H.S.; Radey, M.C.; Merrihew, G.; MacCoss, M.J.; Burns, J.; Wozniak, D.J.; Parsek, M.R.; et al. Staphylococcus aureus Protein a Mediates Interspecies Interactions at the Cell Surface of Pseudomonas aeruginosa. mBio 2016, 7, e00538-16. [Google Scholar] [CrossRef] [PubMed]
  196. Flemming, H.-C.; Wingender, J.; Szewzyk, U.; Steinberg, P.; Rice, S.A.; Kjelleberg, S. Biofilms: An Emergent Form of Bacterial Life. Nat. Rev. Microbiol. 2016, 14, 563–575. [Google Scholar] [CrossRef] [PubMed]
  197. Kaplan, J.B.; Velliyagounder, K.; Ragunath, C.; Rohde, H.; Mack, D.; Knobloch, J.K.-M.; Ramasubbu, N. Genes Involved in the Synthesis and Degradation of Matrix Polysaccharide in Actinobacillus Actinomycetemcomitans and Actinobacillus Pleuropneumoniae Biofilms. J. Bacteriol. 2004, 186, 8213–8220. [Google Scholar] [CrossRef] [PubMed]
  198. Chaignon, P.; Sadovskaya, I.; Ragunah, C.; Ramasubbu, N.; Kaplan, J.B.; Jabbouri, S. Susceptibility of Staphylococcal Biofilms to Enzymatic Treatments Depends on Their Chemical Composition. Appl. Microbiol. Biotechnol. 2007, 75, 125–132. [Google Scholar] [CrossRef] [PubMed]
  199. Dobrynina, O.Y.; Bolshakova, T.N.; Umyarov, A.M.; Boksha, I.S.; Lavrova, N.V.; Grishin, A.V.; Lyashchuk, A.M.; Galushkina, Z.M.; Avetisian, L.R.; Chernukha, M.Y.; et al. Disruption of Bacterial Biofilms Using Recombinant Dispersin B. Microbiology 2015, 84, 498–501. [Google Scholar] [CrossRef]
  200. Chen, K.-J.; Lee, C.-K. Twofold Enhanced Dispersin B Activity by N-Terminal Fusion to Silver-Binding Peptide for Biofilm Eradication. Int. J. Biol. Macromol. 2018, 118, 419–426. [Google Scholar] [CrossRef] [PubMed]
  201. Lefebvre, E.; Vighetto, C.; Di Martino, P.; Larreta Garde, V.; Seyer, D. Synergistic Antibiofilm Efficacy of Various Commercial Antiseptics, Enzymes and EDTA: A Study of Pseudomonas aeruginosa and Staphylococcus aureus Biofilms. Int. J. Antimicrob. Agents 2016, 48, 181–188. [Google Scholar] [CrossRef]
  202. Torelli, R.; Cacaci, M.; Papi, M.; Paroni Sterbini, F.; Martini, C.; Posteraro, B.; Palmieri, V.; De Spirito, M.; Sanguinetti, M.; Bugli, F. Different Effects of Matrix Degrading Enzymes towards Biofilms Formed by E. faecalis and E. faecium Clinical Isolates. Colloids Surf. B Biointerfaces 2017, 158, 349–355. [Google Scholar] [CrossRef]
  203. Sugimoto, S.; Sato, F.; Miyakawa, R.; Chiba, A.; Onodera, S.; Hori, S.; Mizunoe, Y. Broad Impact of Extracellular DNA on Biofilm Formation by Clinically Isolated Methicillin-Resistant and -Sensitive Strains of Staphylococcus aureus. Sci. Rep. 2018, 8, 2254. [Google Scholar] [CrossRef]
  204. Sharma, P.; Garg, N.; Sharma, A.; Capalash, N.; Singh, R. Nucleases of Bacterial Pathogens as Virulence Factors, Therapeutic Targets and Diagnostic Markers. Int. J. Med. Microbiol. 2019, 309, 151354. [Google Scholar] [CrossRef]
  205. Rubini, D.; Banu, S.F.; Nisha, P.; Murugan, R.; Thamotharan, S.; Percino, M.J.; Subramani, P.; Nithyanand, P. Essential Oils from Unexplored Aromatic Plants Quench Biofilm Formation and Virulence of Methicillin Resistant Staphylococcus aureus. Microb. Pathog. 2018, 122, 162–173. [Google Scholar] [CrossRef] [PubMed]
  206. Powell, L.C.; Pritchard, M.F.; Ferguson, E.L.; Powell, K.A.; Patel, S.U.; Rye, P.D.; Sakellakou, S.-M.; Buurma, N.J.; Brilliant, C.D.; Copping, J.M.; et al. Targeted Disruption of the Extracellular Polymeric Network of Pseudomonas aeruginosa Biofilms by Alginate oligosaccharides. NPJ Biofilms Microbiomes 2018, 4, 13. [Google Scholar] [CrossRef] [PubMed]
  207. Wu, J.; Wang, X.; Wang, Q.; Lou, Z.; Li, S.; Zhu, Y.; Qin, L.; Wei, H. Nanomaterials with Enzyme-like Characteristics (Nanozymes): Next-Generation Artificial Enzymes (II). Chem. Soc. Rev. 2019, 48, 1004–1076. [Google Scholar] [CrossRef] [PubMed]
  208. Urzedo, A.L.; Gonçalves, M.C.; Nascimento, M.H.M.; Lombello, C.B.; Nakazato, G.; Seabra, A.B. Cytotoxicity and Antibacterial Activity of Alginate Hydrogel Containing Nitric Oxide Donor and Silver Nanoparticles for Topical Applications. ACS Biomater. Sci. Eng. 2020, 6, 2117–2134. [Google Scholar] [CrossRef] [PubMed]
  209. Tan, Y.; Ma, S.; Leonhard, M.; Moser, D.; Haselmann, G.M.; Wang, J.; Eder, D.; Schneider-Stickler, B. Enhancing Antibiofilm Activity with Functional Chitosan Nanoparticles Targeting Biofilm Cells and Biofilm Matrix. Carbohydr. Polym. 2018, 200, 35–42. [Google Scholar] [CrossRef] [PubMed]
  210. Gao, L.; Giglio, K.M.; Nelson, J.L.; Sondermann, H.; Travis, A.J. Ferromagnetic Nanoparticles with Peroxidase-like Activity Enhance the Cleavage of Biological Macromolecules for Biofilm Elimination. Nanoscale 2014, 6, 2588–2593. [Google Scholar] [CrossRef] [PubMed]
  211. Tan, Y.; Ma, S.; Leonhard, M.; Moser, D.; Ludwig, R.; Schneider-Stickler, B. Co-Immobilization of Cellobiose Dehydrogenase and Deoxyribonuclease I on Chitosan Nanoparticles against Fungal/Bacterial Polymicrobial Biofilms Targeting Both Biofilm Matrix and Microorganisms. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 108, 110499. [Google Scholar] [CrossRef] [PubMed]
  212. Liu, Z.; Wang, F.; Ren, J.; Qu, X. A Series of MOF/Ce-Based Nanozymes with Dual Enzyme-like Activity Disrupting Biofilms and Hindering Recolonization of Bacteria. Biomaterials 2019, 208, 21–31. [Google Scholar] [CrossRef]
  213. Van der Borden, A.J.; van der Werf, H.; van der Mei, H.C.; Busscher, H.J. Electric Current-Induced Detachment of Staphylococcus Epidermidis Biofilms from Surgical Stainless Steel. Appl. Environ. Microbiol. 2004, 70, 6871–6874. [Google Scholar] [CrossRef]
  214. Davis, C.P.; Shirtliff, M.E.; Trieff, N.M.; Hoskins, S.L.; Warren, M.M. Quantification, Qualification, and Microbial Killing Efficiencies of Antimicrobial Chlorine-Based Substances Produced by Iontophoresis. Antimicrob. Agents Chemother. 1994, 38, 2768–2774. [Google Scholar] [CrossRef]
  215. Borriello, G.; Werner, E.; Roe, F.; Kim, A.M.; Ehrlich, G.D.; Stewart, P.S. Oxygen Limitation Contributes to Antibiotic Tolerance of Pseudomonas Aeruginosa in Biofilms. Antimicrob. Agents Chemother. 2004, 48, 2659–2664. [Google Scholar] [CrossRef] [PubMed]
  216. Del Pozo, J.L.; Rouse, M.S.; Mandrekar, J.N.; Steckelberg, J.M.; Patel, R. The Electricidal Effect: Reduction of Staphylococcus and Pseudomonas Biofilms by Prolonged Exposure to Low-Intensity Electrical Current. Antimicrob. Agents Chemother. 2009, 53, 41–45. [Google Scholar] [CrossRef]
  217. Wolfmeier, H.; Pletzer, D.; Mansour, S.C.; Hancock, R.E.W. New Perspectives in Biofilm Eradication. ACS Infect. Dis. 2018, 4, 93–106. [Google Scholar] [CrossRef] [PubMed]
  218. Chang, Y.H.; Grodzinsky, A.J.; Wang, D.I. Augmentation of Mass Transfer through Electrical Means for Hydrogel-Entrapped Escherichia Coli Cultivation. Biotechnol. Bioeng. 1995, 48, 149–157. [Google Scholar] [CrossRef]
  219. Stewart, P.S.; Wattanakaroon, W.; Goodrum, L.; Fortun, S.M.; McLeod, B.R. Electrolytic Generation of Oxygen Partially Explains Electrical Enhancement of Tobramycin Efficacy against Pseudomonas Aeruginosa Biofilm. Antimicrob. Agents Chemother. 1999, 43, 292–296. [Google Scholar] [CrossRef]
  220. Blenkinsopp, S.A.; Khoury, A.E.; Costerton, J.W. Electrical Enhancement of Biocide Efficacy against Pseudomonas Aeruginosa Biofilms. Appl. Environ. Microbiol. 1992, 58, 3770–3773. [Google Scholar] [CrossRef] [PubMed]
  221. Sabelnikov, A.G.; Cymbalyuk, E.S.; Gongadze, G.; Borovyagin, V.L. Escherichia Coli Membranes during Electrotransformation: An Electron Microscopy Study. Biochim. Biophys. Acta 1991, 1066, 21–28. [Google Scholar] [CrossRef] [PubMed]
  222. Hancock, R.E.W.; Rozek, A. Role of Membranes in the Activities of Antimicrobial Cationic Peptides. FEMS Microbiol. Lett. 2002, 206, 143–149. [Google Scholar] [CrossRef]
  223. Del Pozo, J.L.; Rouse, M.S.; Mandrekar, J.N.; Sampedro, M.F.; Steckelberg, J.M.; Patel, R. Effect of Electrical Current on the Activities of Antimicrobial Agents against Pseudomonas aeruginosa, Staphylococcus aureus, and Staphylococcus epidermidis Biofilms. Antimicrob. Agents Chemother. 2009, 53, 35–40. [Google Scholar] [CrossRef]
  224. Kim, Y.W.; Subramanian, S.; Gerasopoulos, K.; Ben-Yoav, H.; Wu, H.-C.; Quan, D.; Carter, K.; Meyer, M.T.; Bentley, W.E.; Ghodssi, R. Effect of Electrical Energy on the Efficacy of Biofilm Treatment Using the Bioelectric Effect. Npj Biofilms Microbiomes 2015, 1, 15016. [Google Scholar] [CrossRef]
  225. Kovalova, Z.; Leroy, M.; Kirkpatrick, M.J.; Odic, E.; Machala, Z. Corona Discharges with Water Electrospray for Escherichia Coli Biofilm Eradication on a Surface. Bioelectrochemistry 2016, 112, 91–99. [Google Scholar] [CrossRef] [PubMed]
  226. Łusiak-Szelachowska, M.; Weber-Dąbrowska, B.; Górski, A. Bacteriophages and Lysins in Biofilm Control. Virol. Sin. 2020, 35, 125–133. [Google Scholar] [CrossRef] [PubMed]
  227. Dams, D.; Brøndsted, L.; Drulis-Kawa, Z.; Briers, Y. Engineering of Receptor-Binding Proteins in Bacteriophages and Phage Tail-like Bacteriocins. Biochem. Soc. Trans. 2019, 47, 449–460. [Google Scholar] [CrossRef] [PubMed]
  228. Chang, C.; Yu, X.; Guo, W.; Guo, C.; Guo, X.; Li, Q.; Zhu, Y. Bacteriophage-Mediated Control of Biofilm: A Promising New Dawn for the Future. Front. Microbiol. 2022, 13, 825828. [Google Scholar] [CrossRef] [PubMed]
  229. Schmelcher, M.; Donovan, D.M.; Loessner, M.J. Bacteriophage Endolysins as Novel Antimicrobials. Future Microbiol. 2012, 7, 1147–1171. [Google Scholar] [CrossRef] [PubMed]
  230. Yan, J.; Mao, J.; Xie, J. Bacteriophage Polysaccharide Depolymerases and Biomedical Applications. BioDrugs 2014, 28, 265–274. [Google Scholar] [CrossRef] [PubMed]
  231. Islam, M.Z.; Fokine, A.; Mahalingam, M.; Zhang, Z.; Garcia-Doval, C.; van Raaij, M.J.; Rossmann, M.G.; Rao, V.B. Molecular Anatomy of the Receptor Binding Module of a Bacteriophage Long Tail Fiber. PLoS Pathog. 2019, 15, e1008193. [Google Scholar] [CrossRef]
  232. Cisek, A.A.; Dąbrowska, I.; Gregorczyk, K.P.; Wyżewski, Z. Phage Therapy in Bacterial Infections Treatment: One Hundred Years After the Discovery of Bacteriophages. Curr. Microbiol. 2017, 74, 277–283. [Google Scholar] [CrossRef]
  233. Suh, G.A.; Lodise, T.P.; Tamma, P.D.; Knisely, J.M.; Alexander, J.; Aslam, S.; Barton, K.D.; Bizzell, E.; Totten, K.M.C.; Campbell, J.L.; et al. Considerations for the Use of Phage Therapy in Clinical Practice. Antimicrob. Agents Chemother. 2022, 66, e0207121. [Google Scholar] [CrossRef]
  234. Morris, J.; Kelly, N.; Elliott, L.; Grant, A.; Wilkinson, M.; Hazratwala, K.; McEwen, P. Evaluation of Bacteriophage Anti-Biofilm Activity for Potential Control of Orthopedic Implant-Related Infections Caused by Staphylococcus aureus. Surg. Infect. 2019, 20, 16–24. [Google Scholar] [CrossRef]
  235. Maszewska, A.; Zygmunt, M.; Grzejdziak, I.; Różalski, A. Use of Polyvalent Bacteriophages to Combat Biofilm of Proteus Mirabilis Causing Catheter-associated Urinary Tract Infections. J. Appl. Microbiol. 2018, 125, 1253–1265. [Google Scholar] [CrossRef] [PubMed]
  236. Rakov, C.; Ben Porat, S.; Alkalay-Oren, S.; Yerushalmy, O.; Abdalrhman, M.; Gronovich, N.; Huang, L.; Pride, D.; Coppenhagen-Glazer, S.; Nir-Paz, R.; et al. Targeting Biofilm of MDR Providencia Stuartii by Phages Using a Catheter Model. Antibiotics 2021, 10, 375. [Google Scholar] [CrossRef]
  237. Ben-Zaken, H.; Kraitman, R.; Coppenhagen-Glazer, S.; Khalifa, L.; Alkalay-Oren, S.; Gelman, D.; Ben-Gal, G.; Beyth, N.; Hazan, R. Isolation and Characterization of Streptococcus Mutans Phage as a Possible Treatment Agent for Caries. Viruses 2021, 13, 825. [Google Scholar] [CrossRef] [PubMed]
  238. Rajabi, Z.; Kermanshahi, R.; Dallal, M.M.S.; Erfani, Y.; Ranjbar, R. Isolation of the Bacteriophages Inhibiting the Expression of the Genes Involved in Biofilm Formation by Streptococcus mutans. Jundishapur. J. Microbiol. 2021, 14, e113206. [Google Scholar] [CrossRef]
  239. Manoharadas, S.; Altaf, M.; Alrefaei, A.F.; Hussain, S.A.; Devasia, R.M.; Badjah Hadj, A.Y.M.; Abuhasil, M.S.A. Microscopic Analysis of the Inhibition of Staphylococcal Biofilm Formation by Escherichia coli and the Disruption of Preformed Staphylococcal Biofilm by Bacteriophage. Microsc. Res. Technol. 2021, 84, 1513–1521. [Google Scholar] [CrossRef] [PubMed]
  240. Abou Zeid, A.A.; Swelim, M.A.; Reda, F.M.; Abd El Haveez, A.M.; Nasr-Eldin, M.A. Effectiveness of Four Lytic Phages against Biofilm-Producing and Multidrug-Resistant Escherichia coli. Benha J. Appl. Sci. 2021, 6, 53–65. [Google Scholar] [CrossRef]
  241. Amankwah, S.; Abdella, K.; Kassa, T. Bacterial Biofilm Destruction: A Focused Review on The Recent Use of Phage-Based Strategies with Other Antibiofilm Agents. Nanotechnol. Sci. Appl. 2021, 14, 161–177. [Google Scholar] [CrossRef] [PubMed]
  242. Maciejewska, B.; Olszak, T.; Drulis-Kawa, Z. Applications of Bacteriophages versus Phage Enzymes to Combat and Cure Bacterial Infections: An Ambitious and Also a Realistic Application? Appl. Microbiol. Biotechnol. 2018, 102, 2563–2581. [Google Scholar] [CrossRef]
  243. Guo, Z.; Huang, J.; Yan, G.; Lei, L.; Wang, S.; Yu, L.; Zhou, L.; Gao, A.; Feng, X.; Han, W.; et al. Identification and Characterization of Dpo42, a Novel Depolymerase Derived from the Escherichia coli Phage vB_EcoM_ECOO78. Front. Microbiol. 2017, 8, 273039. [Google Scholar]
  244. Gutiérrez, D.; Briers, Y.; Rodríguez-Rubio, L.; Martínez, B.; Rodríguez, A.; Lavigne, R.; García, P. Role of the Pre-Neck Appendage Protein (Dpo7) from Phage vB_SepiS-phiIPLA7 as an Anti-Biofilm Agent in Staphylococcal Species. Front. Microbiol. 2015, 6, 167086. [Google Scholar] [CrossRef]
  245. Wu, Y.; Wang, R.; Xu, M.; Liu, Y.; Zhu, X.; Qiu, J.; Liu, Q.; He, P.; Li, Q. A Novel Polysaccharide Depolymerase Encoded by the Phage SH-KP152226 Confers Specific Activity Against Multidrug-Resistant Klebsiella Pneumoniae via Biofilm Degradation. Front. Microbiol. 2019, 10, 2768. [Google Scholar] [CrossRef] [PubMed]
  246. Shahed-Al-Mahmud, M.; Roy, R.; Sugiokto, F.G.; Islam, M.N.; Lin, M.-D.; Lin, L.-C.; Lin, N.-T. Phage φAB6-Borne Depolymerase Combats Acinetobacter Baumannii Biofilm Formation and Infection. Antibiotics 2021, 10, 279. [Google Scholar] [CrossRef] [PubMed]
  247. Schuch, R.; Khan, B.K.; Raz, A.; Rotolo, J.A.; Wittekind, M. Bacteriophage Lysin CF-301, a Potent Antistaphylococcal Biofilm Agent. Antimicrob. Agents Chemother. 2017, 61, e02666-16. [Google Scholar] [CrossRef]
  248. Cha, Y.; Son, B.; Ryu, S. Effective Removal of Staphylococcal Biofilms on Various Food Contact Surfaces by Staphylococcus aureus Phage Endolysin LysCSA13. Food Microbiol. 2019, 84, 103245. [Google Scholar] [CrossRef] [PubMed]
  249. Yuan, Y.; Li, X.; Wang, L.; Li, G.; Cong, C.; Li, R.; Cui, H.; Murtaza, B.; Xu, Y. The Endolysin of the Acinetobacter Baumannii Phage vB_AbaP_D2 Shows Broad Antibacterial Activity. Microb. Biotechnol. 2021, 14, 403–418. [Google Scholar] [CrossRef]
  250. Yang, H.; Zhang, H.; Wang, J.; Yu, J.; Wei, H. A Novel Chimeric Lysin with Robust Antibacterial Activity against Planktonic and Biofilm Methicillin-Resistant Staphylococcus aureus. Sci. Rep. 2017, 7, 40182. [Google Scholar] [CrossRef]
  251. Vasina, D.V.; Antonova, N.P.; Grigoriev, I.V.; Yakimakha, V.S.; Lendel, A.M.; Nikiforova, M.A.; Pochtovyi, A.A.; Remizov, T.A.; Usachev, E.V.; Shevlyagina, N.V.; et al. Discovering the Potentials of Four Phage Endolysins to Combat Gram-Negative Infections. Front. Microbiol. 2021, 12, 748718. [Google Scholar] [CrossRef]
  252. Oechslin, F.; Piccardi, P.; Mancini, S.; Gabard, J.; Moreillon, P.; Entenza, J.M.; Resch, G.; Que, Y.-A. Synergistic Interaction Between Phage Therapy and Antibiotics Clears Pseudomonas aeruginosa Infection in Endocarditis and Reduces Virulence. J. Infect. Dis. 2017, 215, 703–712. [Google Scholar] [CrossRef] [PubMed]
  253. Kumaran, D.; Taha, M.; Yi, Q.; Ramirez-Arcos, S.; Diallo, J.-S.; Carli, A.; Abdelbary, H. Does Treatment Order Matter? Investigating the Ability of Bacteriophage to Augment Antibiotic Activity against Staphylococcus aureus Biofilms. Front. Microbiol. 2018, 9, 303285. [Google Scholar] [CrossRef] [PubMed]
  254. Cano, E.J.; Caflisch, K.M.; Bollyky, P.L.; Van Belleghem, J.D.; Patel, R.; Fackler, J.; Brownstein, M.J.; Horne, B.; Biswas, B.; Henry, M.; et al. Phage Therapy for Limb-Threatening Prosthetic Knee Klebsiella Pneumoniae Infection: Case Report and In Vitro Characterization of Anti-Biofilm Activity. Clin. Infect. Dis. 2021, 73, e144–e151. [Google Scholar] [CrossRef]
  255. Stachler, E.; Kull, A.; Julian, T.R. Bacteriophage Treatment before Chemical Disinfection Can Enhance Removal of Plastic-Surface-Associated Pseudomonas aeruginosa. Appl. Environ. Microbiol. 2021, 87, e00980-21. [Google Scholar] [CrossRef] [PubMed]
  256. Seethalakshmi, P.S.; Rajeev, R.; Kiran, G.S.; Selvin, J. Shrimp Disease Management for Sustainable Aquaculture: Innovations from Nanotechnology and Biotechnology. Aquacult. Int. 2021, 29, 1591–1620. [Google Scholar] [CrossRef]
  257. Yu, P.; Wang, Z.; Marcos-Hernandez, M.; Zuo, P.; Zhang, D.; Powell, C.; Pan, A.Y.; Villagrán, D.; Wong, M.S.; Alvarez, P.J.J. Bottom-up Biofilm Eradication Using Bacteriophage-Loaded Magnetic Nanocomposites: A Computational and Experimental Study. Environ. Sci. Nano 2019, 6, 3539–3550. [Google Scholar] [CrossRef]
  258. Koskella, B.; Meaden, S. Understanding Bacteriophage Specificity in Natural Microbial Communities. Viruses 2013, 5, 806–823. [Google Scholar] [CrossRef] [PubMed]
  259. Huss, P.; Raman, S. Engineered Bacteriophages as Programmable Biocontrol Agents. Curr. Opin. Biotechnol. 2020, 61, 116–121. [Google Scholar] [CrossRef] [PubMed]
  260. Lu, T.K.; Collins, J.J. Dispersing Biofilms with Engineered Enzymatic Bacteriophage. Proc. Natl. Acad. Sci. USA 2007, 104, 11197–11202. [Google Scholar] [CrossRef] [PubMed]
  261. Pei, R.; Lamas-Samanamud, G.R. Inhibition of Biofilm Formation by T7 Bacteriophages Producing Quorum-Quenching Enzymes. Appl. Environ. Microbiol. 2014, 80, 5340. [Google Scholar] [CrossRef] [PubMed]
  262. Born, Y.; Fieseler, L.; Thöny, V.; Leimer, N.; Duffy, B.; Loessner, M.J. Engineering of Bacteriophages Y2::dpoL1-C and Y2::luxAB for Efficient Control and Rapid Detection of the Fire Blight Pathogen, Erwinia Amylovora. Appl. Environ. Microbiol. 2017, 83, e00341-17. [Google Scholar] [CrossRef]
  263. Dedrick, R.M.; Guerrero-Bustamante, C.A.; Garlena, R.A.; Russell, D.A.; Ford, K.; Harris, K.; Gilmour, K.C.; Soothill, J.; Jacobs-Sera, D.; Schooley, R.T.; et al. Engineered Bacteriophages for Treatment of a Patient with a Disseminated Drug-Resistant Mycobacterium abscessus. Nat. Med. 2019, 25, 730–733. [Google Scholar] [CrossRef]
  264. Li, M.; Shi, D.; Li, Y.; Xiao, Y.; Chen, M.; Chen, L.; Du, H.; Zhang, W. Recombination of T4-like Phages and Its Activity against Pathogenic Escherichia coli in Planktonic and Biofilm Forms. Virol. Sin. 2020, 35, 651–661. [Google Scholar] [CrossRef]
  265. Smith, G.P. Filamentous Fusion Phage: Novel Expression Vectors That Display Cloned Antigens on the Virion Surface. Science 1985, 228, 1315–1317. [Google Scholar] [CrossRef] [PubMed]
  266. Muteeb, G.; Rehman, M.T.; Ali, S.Z.; Al-Shahrani, A.M.; Kamal, M.A.; Ashraf, G.M. Phage Display Technique: A Novel Medicinal Approach to Overcome an Tibiotic Resistance by Using Peptide-Based Inhibitors Against β-Lactamases. Curr. Drug Metab. 2017, 18, 90–95. [Google Scholar] [CrossRef] [PubMed]
  267. Flachbartova, Z.; Pulzova, L.; Bencurova, E.; Potocnakova, L.; Comor, L.; Bednarikova, Z.; Bhide, M. Inhibition of Multidrug Resistant Listeria Monocytogenes by Peptides Isolated from Combinatorial Phage Display Libraries. Microbiol. Res. 2016, 188–189, 34–41. [Google Scholar] [CrossRef] [PubMed]
  268. Gillespie, J.W.; Yang, L.; De Plano, L.M.; Stackhouse, M.A.; Petrenko, V.A. Evolution of a Landscape Phage Library in a Mouse Xenograft Model of Human Breast Cancer. Viruses 2019, 11, 988. [Google Scholar] [CrossRef] [PubMed]
  269. Petrenko, V.A.; Gillespie, J.W.; Xu, H.; O’Dell, T.; De Plano, L.M. Combinatorial Avidity Selection of Mosaic Landscape Phages Targeted at Breast Cancer Cells—An Alternative Mechanism of Directed Molecular Evolution. Viruses 2019, 11, 785. [Google Scholar] [CrossRef]
  270. Plano, L.M.D.; Franco, D.; Rizzo, M.G.; Zammuto, V.; Gugliandolo, C.; Silipigni, L.; Torrisi, L.; Guglielmino, S.P.P. Role of Phage Capsid in the Resistance to UV-C Radiations. Int. J. Mol. Sci. 2021, 22, 3408. [Google Scholar] [CrossRef]
  271. De Plano, L.M.; Silipigni, L.; Torrisi, L.; Torrisi, A.; Cutroneo, M.; Havranek, V.; Mackova, A.; Zammuto, V.; Gugliandolo, C.; Rizzo, M.G.; et al. Incidence of Phage Capsid Organization on the Resistance to High Energy Proton Beams. Appl. Sci. 2022, 12, 988. [Google Scholar] [CrossRef]
Figure 1. Representation of life cycle of BF formation, from reversible adhesion of bacteria to biofilm dispersion.
Figure 1. Representation of life cycle of BF formation, from reversible adhesion of bacteria to biofilm dispersion.
Microorganisms 12 00639 g001
Figure 2. Schematic representation of production and transduction of signals responsible for quorum sensing in Gram-positive (A) and Gram-negative (B) bacteria.
Figure 2. Schematic representation of production and transduction of signals responsible for quorum sensing in Gram-positive (A) and Gram-negative (B) bacteria.
Microorganisms 12 00639 g002
Figure 3. A representation of the possible surface modification strategies for inhibiting initial bacterial adhesion through the modification of the surface of the material used (A) or through the use of molecules or polymers that prevent bacteria from adhering to the surface (B). Antibacterial action may be facilitated by the presence of molecules capable of killing bacteria (C). Additionally, the combined approach of surface modifications with polymers (D) or the integration of nanoparticles (E) can lead to an antiadhesive surface with antibacterial properties.
Figure 3. A representation of the possible surface modification strategies for inhibiting initial bacterial adhesion through the modification of the surface of the material used (A) or through the use of molecules or polymers that prevent bacteria from adhering to the surface (B). Antibacterial action may be facilitated by the presence of molecules capable of killing bacteria (C). Additionally, the combined approach of surface modifications with polymers (D) or the integration of nanoparticles (E) can lead to an antiadhesive surface with antibacterial properties.
Microorganisms 12 00639 g003
Figure 4. An illustration of bacteria using receptors (purple) to sense the signals (red) produced by nearby bacteria, allowing them to communicate the density of their population. QS can be inhibited thanks to QS inhibitors (yellow) capable of binding receptors. Meanwhile, quorum quenching enzymes (green) can bind to autoinducer molecules.
Figure 4. An illustration of bacteria using receptors (purple) to sense the signals (red) produced by nearby bacteria, allowing them to communicate the density of their population. QS can be inhibited thanks to QS inhibitors (yellow) capable of binding receptors. Meanwhile, quorum quenching enzymes (green) can bind to autoinducer molecules.
Microorganisms 12 00639 g004
Figure 5. A representation of the main anti-BF strategies applied to the components of the EPS matrix. The lytic action of dispersin B on polysaccharides (A), the degradation of eDNA by DNase I (B), the use of nanoparticles functionalized with DNase and antimicrobial agents (C), and finally, the application of electric currents combined with molecules with antimicrobial action (D) are highlighted.
Figure 5. A representation of the main anti-BF strategies applied to the components of the EPS matrix. The lytic action of dispersin B on polysaccharides (A), the degradation of eDNA by DNase I (B), the use of nanoparticles functionalized with DNase and antimicrobial agents (C), and finally, the application of electric currents combined with molecules with antimicrobial action (D) are highlighted.
Microorganisms 12 00639 g005
Figure 6. A simplified image of phage anti-BF strategies: (1) Phage therapy: based on the use of a single or cocktail phage. (2) Phage-derived enzymes, such as depolymerases, lysins, or quorum quenching enzymes are used for the dispersion of the BF matrix. (3) Phage in combination therapy: using both phages and other antimicrobial compounds, such as antibiotics, nanoparticles, or chemical disinfectants. (4) Engineered phages: the genetical modification of phages to amplify the host–species interaction range or the phage efficiency.
Figure 6. A simplified image of phage anti-BF strategies: (1) Phage therapy: based on the use of a single or cocktail phage. (2) Phage-derived enzymes, such as depolymerases, lysins, or quorum quenching enzymes are used for the dispersion of the BF matrix. (3) Phage in combination therapy: using both phages and other antimicrobial compounds, such as antibiotics, nanoparticles, or chemical disinfectants. (4) Engineered phages: the genetical modification of phages to amplify the host–species interaction range or the phage efficiency.
Microorganisms 12 00639 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Iaconis, A.; De Plano, L.M.; Caccamo, A.; Franco, D.; Conoci, S. Anti-Biofilm Strategies: A Focused Review on Innovative Approaches. Microorganisms 2024, 12, 639. https://doi.org/10.3390/microorganisms12040639

AMA Style

Iaconis A, De Plano LM, Caccamo A, Franco D, Conoci S. Anti-Biofilm Strategies: A Focused Review on Innovative Approaches. Microorganisms. 2024; 12(4):639. https://doi.org/10.3390/microorganisms12040639

Chicago/Turabian Style

Iaconis, Antonella, Laura Maria De Plano, Antonella Caccamo, Domenico Franco, and Sabrina Conoci. 2024. "Anti-Biofilm Strategies: A Focused Review on Innovative Approaches" Microorganisms 12, no. 4: 639. https://doi.org/10.3390/microorganisms12040639

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