Next Article in Journal
Hydrological Seasonality Drives DOM–Bacteria Interactions in the Rushan River Basin
Previous Article in Journal
Effect of Glutamate Concentration and Atmosphere of Incubation on the Production of ɣ-Aminobutyric Acid in Levilactobacillus brevis LB12
Previous Article in Special Issue
A Collection and Analysis of Simplified Data for a Better Understanding of the Complex Process of Biofilm Inactivation by Ultraviolet and Visible Irradiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Understanding Pseudomonas aeruginosa Biofilms: Quorum Sensing, c-di-GMP Signaling, and Emerging Antibiofilm Approaches

1
Department of Public Health, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia
2
Department of Physiology, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
3
Department of Public Health, College of Applied Medical Science, King Khalid University, Abha 61421, Saudi Arabia
4
Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Khamis Mishit 61421, Saudi Arabia
5
Family Medicine Department, King Fahad Armed Hospital, Jeddah 23311, Saudi Arabia
6
Home Health Care Department, King Fahad Armed Forces Hospital, Jeddah 23311, Saudi Arabia
7
Academic Affairs Department, Prince Sultan Military College for Health Sciences, Dhahran 34465, Saudi Arabia
8
Department of Pathology and Laboratory Diagnosis, College of Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
*
Author to whom correspondence should be addressed.
Microorganisms 2026, 14(1), 109; https://doi.org/10.3390/microorganisms14010109
Submission received: 30 November 2025 / Revised: 19 December 2025 / Accepted: 3 January 2026 / Published: 4 January 2026

Abstract

Pseudomonas aeruginosa (P. aeruginosa) forms biofilms that are difficult to eliminate. The matrix protects the cells, efflux pumps reduce intracellular drug levels, and dormant subpopulations survive treatment. Routine minimum inhibitory concentration (MIC) testing does not account for these features, which helps explain why infections often continue even when therapy appears appropriate. This review describes how quorum-sensing (QS) and cyclic di-guanosine monophosphate (c-di-GMP) regulate matrix production, efflux activity, and dormancy within P. aeruginosa biofilms. Important matrix components, including Psl, Pel, alginate, and extracellular DNA, slow the movement of antimicrobial agents. Regulatory proteins such as sagS and brlR increase the activity of the MexAB-OprM and MexEF-OprN efflux systems, further reducing intracellular drug concentrations. Oxygen and nutrient limitation promote persister cells and viable but nonculturable cells, with both having the ability to survive antibiotic levels that would normally be lethal. These defenses explain the gap between MIC values and biofilm-specific measurements, such as the minimum biofilm inhibitory concentration and the minimum biofilm eradication concentration. This review also summarizes emerging antibiofilm strategies. These include QS inhibitors, compounds that lower c-di-GMP, such as nitric oxide donors, nanoparticles, depolymerases, bacteriophages, and therapies that are directed at host targets. Modern diagnostic tools, such as confocal laser scanning microscopy, optical coherence tomography, and Raman spectroscopy, improve detection and guide treatment planning. A staged therapeutic approach is presented that begins with the dispersal or loosening of the matrix, continues with targeted antibiotics, and concludes with support for immune clearance. Viewing these strategies within a One Health framework highlights the role of biofilms in clinical disease and in environmental reservoirs and supports more effective surveillance and prevention.

1. Introduction

Biofilms represent a dynamic bacterial lifestyle rather than a fixed state. Bacterial cells coordinate behavior, communicate, and embed themselves in a self-produced extracellular polymeric substance (EPS) matrix that reshapes community function [1]. This collective mode of growth gives bacteria a high level of tolerance to antibiotics and host immunity, which explains the persistent nature of biofilm-related infections in clinical practice [2].
In patients with cystic fibrosis (CF), Pseudomonas aeruginosa (P. aeruginosa) establishes chronic airway infections that are dominated by biofilm growth. Mucoid conversion and increased alginate production alter the matrix and strengthen the chronic state of infection [3,4]. Experimental and clinical studies show that CF-adapted airway biofilms display elevated tolerance to several classes of antibiotics, which links biofilm behavior to progressive lung disease [5,6]. P. aeruginosa also forms invasive biofilms in burn wounds, and animal models reproduce the deep tissue architecture seen in human burns [7,8]. On urinary catheters, surface-attached biofilms lead to catheter-associated urinary tract infections (CAUTI) and often limit the effect of antimicrobial therapy [9]. In ventilator circuits, biofilms that form inside endotracheal tubes act as a source for ventilator-associated pneumonia (VAP) and allow bacterial migration into the lower airways [10,11].
P. aeruginosa serves as a model organism for understanding biofilm biology because reference strains PAO1 and PA14 are genetically tractable, well characterized, and show distinct virulence features [12]. The PAO1 genome, which measures about 6.3 megabases, revealed broad regulatory and metabolic capacity that supports environmental adaptability and intrinsic drug resistance [13]. This genomic foundation allows systems-based studies of biofilm control. Comparative analyses highlight the important differences between PAO1 and PA14 and help to distinguish conserved biofilm traits from strain-specific behavior [14]. Alongside genomic work, integrated in vitro, in vivo, and ex vivo model systems now allow a detailed investigation of P. aeruginosa biofilms under clinically relevant conditions [15].
A hierarchical quorum-sensing (QS) network that includes the Las, Rhl, and Pseudomonas quinolone signal (PQS) systems, with added inputs such as the integrated QS signal, is central to the resilience of P. aeruginosa biofilms [16,17]. These systems regulate many of the genes that are responsible for virulence, metabolism, and community behavior that support biofilm development and maintenance [16,17]. In parallel, the second messenger cyclic di-guanosine monophosphate (c-di-GMP) coordinates the transition from motile to sessile growth, guides surface attachment, and promotes matrix production. These activities are shaped by the opposing functions of diguanylate cyclases (DGCs) and phosphodiesterases (PDEs) [18,19]. Beyond early development, c-di-GMP sets the physiological state of biofilm communities and strongly influences metabolic activity in multiple experimental models [20]. Nitric oxide (NO)-induced dispersal depends on the activation of PDEs and a rapid decrease in intracellular c-di-GMP, which links environmental cues to biofilm destabilization [21,22].
Given the clinical threat of multidrug-resistant (MDR) P. aeruginosa and the complexity of its regulatory networks, it is essential to understand how QS and c-di-GMP intersect to control matrix production, persistence, and dispersal. This review maps the regulatory circuitry that supports biofilm resilience, explains how these signals shape virulence and antimicrobial tolerance, and evaluates translational strategies that target these systems. These strategies include QS inhibitors, c-di-GMP modulators, dispersal inducers, nanoparticle (NP)-based delivery systems, and bacteriophage-derived enzymes that can reprogram or disrupt biofilm behavior in chronic infections.

2. Architectural and Molecular Foundations of P. aeruginosa Biofilms

2.1. Staged Development: Attachment, Maturation, and Dispersal (pel, psl, algD, sadB, and bdlA)

P. aeruginosa follows a stepwise developmental program that includes reversible attachment, irreversible attachment, microcolony formation, three-dimensional maturation, and active dispersal to new sites of infection [23,24]. Irreversible attachment represents a true commitment checkpoint. Loss of sadB locks cells into a surface-defective, hyperswarming state and identifies sadB as a gatekeeper that also acts as a posttranslational modulator of AmrZ [25,26,27].
Dispersal is an actively controlled process that requires BdlA, a chemotaxis-like regulator whose phosphorylation state integrates environmental cues with c-di-GMP signaling [28,29,30,31]. Genetic and biochemical studies place BdlA together with partners such as DipA at the center of the dispersal switch [32,33,34,35]. Figure 1 summarizes this trajectory and highlights key genetic checkpoints (sadB, pel, psl, algD, and bdlA), EPS architecture components (Psl, Pel, alginate, CdrA, and eDNA), and oxygen gradients that support phenazine-driven redox cycling and the emergence of persister and viable but nonculturable (VBNC) subpopulations.

2.2. Matrix Composition and Mechanics: Polysaccharides, Proteins, eDNA, and Host Inputs (NETs, Mucins)

The EPS matrix functions as a composite material whose mechanical properties and signaling capacity arise from interactions among polysaccharides, proteins, and DNA. Psl provides architectural stability and binds the lectin LecB, which helps retain cells and EPS within the biofilm [29,36,37]. Pel is a cationic polymer with a wide genetic distribution and enhances both cohesion and tolerance to antimicrobial agents [29,36,37]. Alginate that is synthesized through algD reshapes CF biofilms and modifies drug responses, even though many nonmucoid P. aeruginosa biofilms form in its absence [31,38].
Matrix-associated proteins further strengthen and specialize the structure. The c-di-GMP-regulated adhesin CdrA crosslinks with Psl to promote aggregation and support robust biofilm formation [39,40]. eDNA acts as a load-bearing scaffold that interacts with Psl to create a skeletal lattice within the matrix. Treatment with DNase weakens this scaffold and improves antimicrobial penetration, particularly in early-stage biofilms [41,42]. Host-derived components are integral to matrix organization. Neutrophil extracellular traps (NETs) can surround P. aeruginosa communities in dense DNA-rich barriers, while airway mucins reshape the architecture of the biofilm. Depending on the local environment, mucins can stabilize tolerant aggregates or instead promote dispersal [43,44,45].

2.3. Spatial and Metabolic Heterogeneity: Oxygen and Nutrient Gradients, Persisters, and VBNC Cells

Steep gradients of oxygen and nutrients stratify P. aeruginosa biofilms and generate layers that differ in gene expression patterns, redox programs, and local growth rates [46,47]. In hypoxic or anoxic regions, endogenous phenazines shuttle electrons to sustain adenosine triphosphate (ATP) production. This redox cycling supports fitness under low oxygen conditions and contributes directly to antibiotic resistance [48,49,50]. This physical and chemical heterogeneity promotes phenotypic diversity within the biofilm. Drug-tolerant persister cells arise as dormant but reversible subpopulations and are repeatedly associated with treatment failure and infection relapse [51,52,53].
Other subpopulations can enter a VBNC state in response to disinfectants or antibiotic stress, as demonstrated in chlorine-exposed and catheter-associated biofilms [54,55,56]. Single-cell- and spectroscopy-based studies reveal that VBNC cells display distinct metabolic signatures when compared with neighboring culturable cells. Emerging single-cell transcriptomic maps further refine this view and show widespread transcriptional diversification within biofilms. These studies demonstrate a global metabolic downshift across biofilm cells when compared with planktonic populations [57].

3. Quorum Networks: The Social Intelligence of Biofilms

3.1. Hierarchical QS Systems Involving las, rhl, pqs, and iqs and Their Cross-Regulation

P. aeruginosa relies on two acyl homoserine lactone circuits, known as las and rhl, and on a quinolone-based circuit that is known as pqs. These systems also include an iqs module that links nutrient stress to QS activity. In the classical model, LasR activates both rhl and pqs, while PqsR controls the Pseudomonas quinolone signal and the related metabolite HHQ. These signals feed back into QS activity and guide biofilm programs [16].
The order of Las, Rhl, and Pqs activity is flexible. Many lasR variants maintain QS-controlled traits through alternative pathways [58,59]. Under phosphate limitation, PAO1 can shift to a state that does not depend on LasR. In this setting, RhlR dominates and represses pqsABCDE, which changes the hierarchy of the system [60]. Phosphate stress activates the iqs module. Under normal conditions, iqs is under Las control, but phosphate scarcity leads to PhoB-driven iqs upregulation and strengthens QS and virulence outputs [61]. Nutritional conditions also influence the relative activity of las, rhl, and pqs, which creates a context-dependent hierarchy [62].
Clinical isolates show similar plasticity. lasR variants are common in CF airways and can maintain or restructure QS-controlled pathways, which supports chronic infection [59,63]. A major mechanism behind this flexibility is the partnership between RhlR and PqsE. Structural and functional studies show that PqsE enhances RhlR-dependent transcription. This activity can reduce the need for the native Rhl autoinducer and preserve RhlR-driven behavior [64,65,66]. Recent work indicates that the PqsE and RhlR interactions fine-tune the RhlR output to match environmental cues [67]. The las, rhl, pqs, and iqs modules form a network that adapts to nutrient changes and stress. This network contains regulatory handoffs that can be targeted, including RhlR dominance under phosphate limitation and the RhlR and PqsE interaction [16,60,61,64].

3.2. Molecular Integration Between QS and c-di-GMP

QS operates within a broader group of second messenger and global control systems that include c-di-GMP signaling, the Gac and Rsm pathways, and the cyclic adenosine monophosphate (cAMP) Vfr system. These systems translate cell density information into motility changes, matrix production, and chronic phenotypes [68]. LasR and RhlR activate matrix-related loci. Evidence from PAO1 shows that psl is under QS control, and earlier studies link QS activity to pel. Together, these findings place Las and Rhl at the upper level of matrix regulation [69,70].
In parallel, c-di-GMP determines the shift from motile to sessile behavior. High c-di-GMP promotes irreversible attachment and EPS synthesis, while low c-di-GMP supports motility and dispersal. These shifts influence QS-controlled outputs such as rhamnolipid production [18,71,72]. At the core of the QS network, RhlR partners with PqsE to modify transcriptional programs. Structural work identifies a targetable interface between RhlR and PqsE [64,65,73].
Furthermore, c-di-GMP also maintains mucoid behavior. The binding of c-di-GMP to the PilZ domain protein Alg44 is essential for alginate polymerization. In addition, AlgU, which is released during MucA loss, drives algD expression and connects with QS to consolidate chronic traits [74,75,76,77]. Higher-level regulators, such as Gac and Rsm small RNAs and the cAMP Vfr axis, interact with DGCs and PDEs, including GcbA, to influence surface commitment and matrix output [78,79,80]. The overall conclusion is that QS and c-di-GMP form a coupled system that stabilizes matrix-rich biofilms, yet it preserves the capacity for motility and dispersal when conditions change [65,68,69,70,75].

3.3. QS-Controlled Virulence Portfolio

Across strains, QS controls a broad set of virulence traits that include secreted enzymes, surfactants, redox-active metabolites, and toxins. These traits strengthen biofilms and reduce the effectiveness of therapy. Las, Rhl, and Pqs regulate hundreds of genes, among them being LasB elastase, rhamnolipids that arise from rhlAB, several phenazines such as pyocyanin, hydrogen cyanide, and secretion systems that influence tissue injury, motility, and tolerance [81].
The pqs module shapes matrix behavior and dispersal. Under PqsR control, PQS and HHQ promote eDNA release through controlled lysis and prophage activation. Loss of pqsL increases HHQ levels and autolysis, which increases eDNA in early biofilms [82]. PQS also promotes the formation of outer membrane vesicles that carry eDNA and enzymes and assist in dispersion [83,84].
Phenazines, such as pyocyanin and HQNO, influence redox pathways and interactions between species. HQNO suppresses respiration in Staphylococcus aureus (S. aureus) and selects small-colony variants that rely on fermentative metabolism. These patterns involve sigma factor B and SrrAB and are common in CF airways [85,86,87,88,89]. Many QS-controlled traits are targetable. Natural compounds and synthetic inhibitors directed at RhlR or PqsR can reduce elastase, rhamnolipids, pyocyanin, and biofilm formation. Some of these inhibitors also modify c-di-GMP levels, which highlights cross-node intervention points [90,91,92].

3.4. Polymicrobial and Host Interactions

QS signals shape interactions within mixed communities that are often found in chronic airway and wound infections. HQNO limits respiration in S. aureus, reduces ATP levels, and selects fermentative small-colony variants. These responses appear in coculture systems, animal models, and clinical settings and involve SrrAB and SigB [86,93,94,95]. Outcomes depend on context. Oxygen availability affects HQNO production and activity, which determines whether the interaction results in competition or coexistence. Anaerobic conditions and media that mimic mucus can favor mixed microcolonies and isolate specific interactions [96,97].
Beyond quinolones, P. aeruginosa produces pyocyanin and the staphylolytic protease LasA. These factors impair S. aureus respiration or damage the cell wall, which shifts competitive balance within polymicrobial biofilms [98,99,100]. These exoproducts help explain why P. aeruginosa often suppresses or displaces S. aureus, but can also coexist with it under certain environmental constraints [101].
Table 1 summarizes the QS modules and their connections to global regulatory systems that include c-di-GMP, Gac and Rsm, and AlgU. The table lists the signals, the synthase and receptor pairs, the major regulatory interactions, the principal outputs, the environmental factors that influence each system, and the main targets described in the literature.

4. c-di-GMP Dynamics: The Molecular Switchboard of Biofilm Formation

4.1. Synthesis and Degradation Enzymes Involving DGCs and PDEs

In Pseudomonas aeruginosa, intracellular c-di-GMP is controlled by a large set of diguanylate cyclases (DGCs) that contain GGDEF domains and by phosphodiesterases (PDEs) that contain EAL or histidine aspartate glycine tyrosine proline domains. Genome-wide analyses of strain PAO1 showed that P. aeruginosa encodes approximately 41 proteins involved in c-di-GMP turnover. These include about 17 GGDEF-only proteins, 5 EAL-only proteins, and 16 proteins that carry both GGDEF and EAL domains, with additional HD-GYP proteins also annotated [18]. These opposing enzyme families shift physiology between motile states that require low c-di-GMP and matrix-producing states that require high c-di-GMP. This second messenger is central to biofilm initiation, maturation, and dispersal in PAO1 and PA14 [18,108,109].
The Wsp sensory system translates surface contact into increased c-di-GMP. Activation of WspA through WspE leads to phosphorylation of WspR. Phosphorylated WspR forms clusters that strengthen DGC activity and promote biofilm formation. Fluorescence and biochemical studies showed that WspR clustering occurs at defined cellular locations during surface growth, producing localized c-di-GMP signaling rather than a uniform cytoplasmic increase [110]. Evidence suggests that the Wsp system detects changes in the cell envelope during surface engagement and that flagellar regulators influence WspR positioning and signaling output [111,112,113].
Mechanosensory input flows directly into c-di-GMP synthesis through sadC. The flagellar stator MotC contacts the transmembrane region of sadC and stimulates its catalytic activity. This interaction links flagellar load to the switch from motile to sessile growth [114]. Division of labor among DGCs is clear. RoeA channels c-di-GMP toward Pel production, whereas sadC has a stronger influence on the control of flagellar motility [115].
On the degradation side, the EAL domain PDE encoded by bifA regulates biofilm formation and swarming. Loss of bifA increases c-di-GMP and produces a hyperbiofilm and swarming-deficient state that can be restored by complementation [116]. RbdA, a PAS GGDEF EAL membrane protein, promotes dispersal. Structural analysis of its cytoplasmic region reveals an activated PDE dimer and coordinated movements between linked domains, which provide a mechanistic basis for PDE-driven dispersal [117,118]. Genome-wide surveys confirm that P. aeruginosa encodes many DGC and PDE proteins.
A defining feature of this network is the high proportion of dual-domain GGDEF–EAL enzymes. In most of these proteins, only one domain is catalytically active under physiological conditions, while the second domain serves a regulatory or sensory role [108]. Several of these enzymes are membrane-associated or show polar or clustered localization, supporting the concept of local c-di-GMP pools that selectively regulate motility, attachment, matrix production, or dispersal rather than global signaling [108]. These findings show that c-di-GMP control is modular and locally organized [119].

4.2. Effectors and Downstream Targets

Effector binding of c-di-GMP shifts matrix synthesis from permissive to fully active. PelD, a degenerate GGDEF domain receptor, binds dimeric c-di-GMP through an RXXD motif. Structural studies of PelD in its apo and bound states show that nucleotide binding activates Pel machinery and commits cells to Pel-dependent biofilm formation [120]. A parallel checkpoint regulates alginate synthesis. The inner membrane PilZ domain protein Alg44 requires c-di-GMP binding for alginate polymerization and export. Genetic work established alg44 as essential for this activity, and structural studies confirm dimeric c-di-GMP as the activating ligand [74,107,121].
Furthermore, c-di-GMP also controls adhesin display through the LapD and LapG system. High c-di-GMP bound to LapD sequesters LapG and stabilizes surface adhesins. When c-di-GMP levels fall, LapG cleaves adhesins and reduces attachment. In P. aeruginosa, LapG cleaves the matrix adhesin CdrA, which fine-tunes aggregation and cohesion under c-di-GMP control [122,123,124]. Beyond matrix assembly and adhesion, c-di-GMP reshapes motility. High levels suppress flagellar motility and regulate type IV pili behavior during surface commitment. Mechanosensory coupling from the flagellar motor through MotC and sadC provides a direct path into c-di-GMP synthesis and consolidates the attached lifestyle [125,126].

4.3. Environmental and Host Signals Modulating c-di-GMP Turnover

Environmental and host-derived cues reset c-di-GMP levels and shift the balance among adhesion, matrix formation, motility, and dispersal. Low nonlethal NO activates PDEs, lowers c-di-GMP, and induces detachment. This mechanism is supported by biochemical studies, models that mimic the airway, and analyses of CF sputum. It is under clinical investigation as an adjunct to antibiotic therapy [72,127,128].
Temperature is another strong regulator. The thermosensory diguanylate cyclase A increases its catalytic rate more than one hundred-fold across a ten-degree change in temperature. This behavior tunes motility, biofilm development, and virulence. Similar thermosensory activity is reported in orthologs from other bacteria [129,130].
Cross-kingdom metabolites also shape lifestyle decisions. Ethanol produced by Candida albicans increases c-di-GMP in P. aeruginosa, inhibits swarming, strengthens Pel-dependent biofilms, and alters phenazine output. This response partly involves the Wsp system [131].
Oxygen limitation and related stresses feed into multisensor DGC and PDE proteins. PAS domain enzymes such as DipA and RbdA display higher PDE activity under nutrient or redox shifts. Increased PDE activity lowers c-di-GMP and triggers dispersal. ProE, a GGDEF–EAL hybrid protein, represses exopolysaccharide-related genes. These enzymes act as environmental sensors that couple oxygen and redox state to matrix-remodeling [117,132,133].
NO, temperature, ethanol, and hypoxia-related signals converge on c-di-GMP turnover and recalibrate the lifestyle of P. aeruginosa in clinical niches. These inputs either stabilize matrix-rich biofilms or prepare cells for dispersal [130,134].

4.4. Interplay with QS and Stress Pathways: Fine-Tuning Motility, EPS Synthesis, and Dispersal

c-di-GMP connects with QS and global regulators to coordinate the transition from acute to chronic states. LasR and RhlR promote matrix gene expression. For example, psl is under LasR and RhlR control. Higher c-di-GMP strengthens these QS-driven outputs and shifts cells toward matrix synthesis and away from motility [69]. This coordination sits within a larger network that links c-di-GMP, QS, cAMP Vfr, and Gac Rsm, aligning environmental information with virulence programs [68].
Posttranslational control further stabilizes outputs. Dimeric c-di-GMP binds Alg44 and activates alginate polymerization. AlgU drives algD expression and intersects with QS to promote mucoid behavior [74,135,136]. The Gac Rsm cascade shapes c-di-GMP flow and matrix-related genes. RsmA represses pslA and sadC, and the RetS and LadS pathways coordinate with DGCs such as sadC to bias populations toward either biofilm formation or motility [137,138]. The cAMP Vfr axis also interacts with c-di-GMP to reduce acute traits as chronic behavior rises [79].
This network remains capable of reversal. Low nonlethal NO activates PDEs, reduces c-di-GMP, and initiates detachment. This principle supports the investigation of NO-based approaches in airway infections [21,139]. Figure 2 and Table 2 summarize this landscape. The figure maps environmental inputs, enzymes, and effectors. The table lists representative DGC and PDE proteins, their stimuli, dominant phenotypes, and structural insights. These elements highlight c-di-GMP as a central switchboard that links environmental cues and quorum signals to chronic biofilm behavior.

5. Antimicrobial Tolerance and Persistence of the Biofilm Code

5.1. EPS Mediated Diffusion Restriction and Charge Buffering

The matrix of P. aeruginosa functions as an active filter rather than a passive outer layer. Psl and Pel form the structural scaffold and slow the movement of antibiotics, which allows interior cells to survive longer. In early biofilms, Psl provides a broad protective barrier that limits the penetration of chemically diverse drugs and even shields nearby cells that do not produce Psl but occupy Psl-rich clusters [140]. Pel adds mechanical strength and charge-based interactions. Because Pel is a cationic polysaccharide, it crosslinks with negatively charged eDNA, tightens the network, and restricts diffusion. Structural and imaging studies demonstrate this ionic Pel and eDNA binding, and Pel can partially compensate when Psl is reduced, explaining why Pel-dominated biofilms maintain resilience [141].
Electrostatic effects deepen these defenses. eDNA is abundant in P. aeruginosa biofilms and binds cationic antibiotics, including aminoglycosides, which lowers their activity. Removing eDNA increases aminoglycoside killing [142,143]. Divalent cations, such as calcium, bridge eDNA strands and compact the matrix, while alginate-derived guluronate oligomers disrupt these cation bridges and loosen the network. This disruption increases antibiotic access to P. aeruginosa biofilms [144,145]. In clinical contexts, these matrix-based effects matter. In mature biofilms, colistin tolerance often reflects penetration limits and altered physiology rather than classical target site changes. These findings illustrate how diffusion and charge buffering establish a strong baseline of protection in vivo [146]. In summary, Psl slows drug entry at the perimeter, Pel and eDNA interactions strengthen internal layers, and eDNA and cation chemistry restrict cationic agents. Together, they create a barrier that routine drug dosing rarely overcomes.

5.2. Efflux Pumps and Stress Regulons

Efflux contributes a second defensive layer that aligns with the biofilm lifestyle. In mature biofilms, the transcriptional regulator BrlR increases tolerance by promoting expression of the RND pumps mexAB-oprM and mexEF-oprN, which reduce intracellular antibiotic concentrations even when diffusion barriers are partially bypassed [147,148]. Efflux is primed early in development. The sensor and regulator SagS function as a surface commitment factor and increase BrlR activity, which links early adaptation to activation of efflux before the matrix is complete [149].
In clinical isolates, these systems operate in combination. Large surveys show context-dependent coordination between mexAB-oprM and mexEF-oprN, with common co-expression alongside other RND pumps such as mexXY and mexCD-oprJ under stress and host-like cues [150]. Reviews consistently identify these two pumps as major contributors to P. aeruginosa resistance [81,151]. This has therapeutic implications. In flow-cell biofilms, colistin tolerance depends on both the PmrAB two-component system and mexAB-oprM, linking environmental stress directly to active efflux and explaining reduced activity of cationic peptides in vivo [146,152]. Efflux also influences virulence and fitness. Inhibiting pumps can reduce pathogenicity, and variants in mexEF-oprN alter motility and secretion pathways, demonstrating combined effects on drug survival and disease progression [153,154]. In summary, biofilm stage regulators such as SagS and BrlR coordinate mexAB-oprM and mexEF-oprN within a flexible efflux repertoire that, together with the matrix, creates a two-tier defense that many standard regimens fail to overcome [147,149,150].

5.3. Dormant, Persister, and VBNC Populations

Steep oxygen and nutrient gradients create microenvironments of slow-growing or non-growing cells that tolerate antibiotics without heritable resistance. Low oxygen interiors reduce the activity of bactericidal drugs and activate stress responses, so neighboring cells with the same genotype may differ markedly in susceptibility. This phenomenon helps explain incomplete killing during otherwise appropriate therapy [2,155,156]. Persisters emerge within this setting. These rare, phenotypically tolerant cells survive lethal antibiotic exposure and repopulate the community once treatment pressure declines. Across P. aeruginosa models, persisters contribute to relapse and chronicity and are supported by nutrient limitation, redox stress, and matrix protection [2,51]. Clinically relevant stresses also generate VBNC states. Sub-inhibitory tobramycin can induce and maintain VBNC physiology in biofilms, and chlorine exposure produces VBNC populations with distinct metabolic signatures. These transitions arise more frequently in biofilms than in planktonic cultures [54,157,158,159]. Together, persisters and VBNC cells form a deep reservoir of tolerance that is strengthened by matrix barriers and efflux. Conventional MIC assays overlook these features. More suitable metrics include the minimum biofilm inhibitory concentration (MBIC) and the minimum biofilm eradication concentration (MBEC), which better reflect biofilm antibiotic responses [2,160].

5.4. Horizontal Gene Transfer and Plasmid Stabilization Inside the Matrix

Biofilms provide strong support for gene exchange. Surface-attached growth improves physical contact among cells and stabilizes interactions, which increases conjugation frequencies relative to planktonic states. At the same time, spatial structure can restrict exchange to outer layers and create organized gene exchange zones [161,162]. Biofilms can also serve as plasmid reservoirs. Even without antibiotics and despite the burden of plasmid carriage, communities maintain plasmids and rapidly expand plasmid-containing populations when selection returns. Evolution experiments and microfluidic studies identify biofilms as important refuges for multidrug-resistant (MDR) plasmids, which shape transfer routes and explain the persistent presence of resistance genes in chronic infection [163,164].
Plasmid-encoded stabilization modules strengthen this persistence. Toxin and antitoxin loci and partitioning systems reduce plasmid loss during cell division and influence host fitness. These factors increase the likelihood that resistance and tolerance genes remain in the population [165,166]. In P. aeruginosa, mobile DNA circulates through multiple routes. Integrative and conjugative elements, such as PAPI 1 and outer membrane vesicles, transfer genetic material, including resistance determinants, which broaden adaptive pathways inside biofilms [167,168]. Because plasmid fitness effects depend on the host background and the surrounding environment, persistence becomes highly probable in biofilm-dominated niches [169].
MIC assays do not capture these realities. MIC measurements reflect planktonic inhibition under well-mixed conditions and do not account for diffusion barriers, charge buffering, oxygen gradients, efflux, or dormancy. As a result, MBIC and MBEC values often exceed MIC values by large margins. Biofilm-specific antimicrobial susceptibility testing (AST) platforms offer more accurate guidance for infections that involve devices or tissue surfaces and are increasingly recommended for translational studies [170,171,172,173].
In summary, the matrix functions as both a chemical shield and a genetic exchange zone. Spatial structure increases encounters, stabilization systems secure inheritance, and multiple transfer pathways support the dissemination of adaptive traits. These factors preserve resistance even when MIC-based therapy appears suitable [162,167,168]. Figure 3 illustrates how QS hierarchies and the c-di-GMP network coordinate tolerance in P. aeruginosa biofilms by guiding EPS production, activating efflux, supporting metabolic dormancy, and stabilizing plasmids.

6. Cracking the Code: Emerging Strategies to Rewire or Disrupt the Network

6.1. Quorum-Sensing Inhibitors (QSIs) and Quorum-Quenching Enzymes

Reducing QS signaling can quiet community behavior without applying strong growth pressure. In P. aeruginosa, synthetic halogenated furanones inspired by metabolites from Delisea pulchra antagonize AHL-mediated signaling. These compounds suppress Las-regulated transcription and reduce biofilm-associated virulence while leaving growth largely unchanged [174,175]. Ajoene, derived from garlic, offers an additional route. It decreases QS-regulated traits and alters the Hfq and small RNA layer that coordinates QS outputs. These effects reduce pathogenic behavior in vitro and in infection models [176,177]. Natural product discovery continues to expand the field. Methyl gallate from Mangifera indica lowers QS-linked outputs that include pyocyanin, rhamnolipids, and motility at sub-MIC levels, and docking studies support LasR engagement together with broad antivirulence activity [178,179].
Enzymatic quorum quenching removes the signal directly. The periplasmic acylase PvdQ hydrolyzes long-chain AHLs involved in Las signaling. Structural work has mapped its active site, and administration of PvdQ in a murine lung model reduced inflammation and bacterial burden. Protein engineering now improves stability and catalytic performance [180,181,182]. Other AHL lactonases and acylases from diverse microbes also reduce QS-dependent virulence and biofilm formation and are being refined as adjuncts to antibiotics [183]. CRISPR interference provides another layer. Silencing of lasI, rhlI, and pqsR has been demonstrated in vitro and in vivo, and recent work connects QS, cAMP Vfr, and CRISPR systems, broadening antivirulence strategies [184,185,186,187]. By reducing Las, Rhl, and Pqs activity or degrading their autoinducers, QS inhibitors and quorum-quenching enzymes limit biofilm formation and virulence and often restore antibiotic susceptibility.

6.2. c-di-GMP Modulators and Dispersal Triggers

Reducing intracellular c-di-GMP can open established biofilms. Small molecules that activate native PDEs or inhibit DGCs shift cells from sessile to motile states and briefly increase antibiotic susceptibility. An example is the H6-335 chemotype. In P. aeruginosa, it reduced c-di-GMP, prevented biofilm formation, and dispersed established biofilms. An analog, H6-335-P1, activated the BifA PDE and, when combined with antibiotics, cleared implant-associated infections in mice [188,189].
NO remains the most validated physiological dispersal trigger. At low concentrations, it activates PDEs that target c-di-GMP, lowers intracellular c-di-GMP, and releases cells from the matrix. Dispersal depends on PDE activity in flow-cell models [21,190,191]. Host-derived cues can induce similar shifts. The mouse cathelicidin CRAMP triggers dispersal with associated changes in c-di-GMP regulators [192]. Combining a c-di-GMP-lowering compound with an appropriate antibiotic collapses biomass and increases killing. This approach is supported by mechanistic data on PDEs such as RbdA and by the in vivo results [117,118].

6.3. Nanoparticle-Based Synergistic Systems

NPs provide versatile platforms to disrupt P. aeruginosa biofilms by improving drug penetration, generating localized stress, and co-delivering agents that weaken tolerance networks. Surveys catalog metal and metal-oxide particles that include silver (Ag), zinc oxide (ZnO), and titanium dioxide (TiO2), as well as mesoporous silica frameworks, polymer and lipid carriers, and hybrid systems that restore antibiotic activity at lower doses [193,194].
Metal-based systems deliver broad stress. AgNPs damage membranes, respiratory enzymes, and nucleic acids and act synergistically with standard antibiotics against MDR P. aeruginosa. Recent reviews outline proteomic stress responses and dosing strategies that balance antimicrobial activity with host safety [195,196]. ZnONPs and TiO2NPs can generate reactive oxygen species under defined conditions and display antibiofilm and antibiotic potentiating activity. Very low zinc oxide can be hormetic, which underscores the need for controlled exposure [197,198]. Photocatalytic TiO2 coatings are being developed as self-cleaning and antibiofilm surfaces for devices and filters [199].
Mesoporous silica NPs (MSNPs) allow high drug loading and tunable release. Hybrid silica and gold nanomotors activated by near infrared (NIR) light penetrate biofilms and reduce biomass through photothermal and photocatalytic effects within short periods [200]. Silica-based NO-releasing particles kill biofilm cells and enhance antibiotic activity, confirming NO as a programmable dispersal and killing cue at the nanoscale [201]. Design studies describe mesoporous silica systems that couple antimicrobial drugs with stimuli-responsive release based on light, pH, or redox conditions [202].
In the airway, liposomal formulations improve residence time, reduce epithelial toxicity, and enhance penetration of mucus-rich and matrix-rich regions. In a chronic Pseudomonas rat model, liposomal bismuth ethanedithiol lowered bacterial counts. Reviews support inhaled, NP enabled antibiotic strategies for difficult lung infections [203,204,205]. Co-delivery of antibiotics with QS antagonists or c-di-GMP modulators aims to weaken multiple tolerance layers at once. Proof-of-concept work shows gold and selenium-based NPs reduce QS-controlled outputs such as pyocyanin and elastase, while design papers discuss gene targeting approaches and exposure controls to minimize environmental effects [206,207]. Dose, irradiation settings, and matrix interactions strongly shape outcomes. ZnO hormesis highlights the need for therapeutic windows and standardized biofilm testing that includes MBIC and MBEC alongside MIC [198]. By uniting targeted delivery with microenvironment-responsive release and combining antibiotics with QS or c-di-GMP inhibitors, NP systems penetrate EPS, promote dispersal, and increase susceptibility of entrenched biofilms [200,201,208].

6.4. Phage and Phage-Derived Enzymes

Bacteriophages offer precision tools for dismantling P. aeruginosa biofilms by damaging both matrix and cells. Native or engineered phages equipped with polysaccharide depolymerases degrade EPS and expose embedded cells to antibiotics and immune factors. Reviews document strong biofilm-degrading activity against Gram-negative biofilms, including P. aeruginosa [209,210].
Endolysins and next-generation Artilysins, which fuse lysins with peptides that permeabilize the outer membrane, can lyse P. aeruginosa despite the Gram-negative barrier. Rationally designed constructs, including LysPA26-based hybrids, show strong antibiofilm activity [211,212]. Phage antibiotic combinations often outperform either alone. In vitro and in vivo models that include zebrafish and murine airway and implant systems show accelerated clearance and improved survival, with reduced emergence of resistance [213,214,215].
Directed evolution to improve penetration and cocktail design to exploit resistance tradeoffs can strengthen performance against persistent biofilms [216]. The endogenous glycoside hydrolase (PslG) functions as a phage enzyme-like adjunct. Applied exogenously, it disperses Psl-rich biofilms and increases antibiotic susceptibility [217,218,219]. Phage, depolymerase, and engineered lysin strategies therefore provide targeted matrix digestion and bacterial killing that complement chemical inhibitors and antibiotics [220].

6.5. Host-Directed and Immunomodulatory Therapies

Because P. aeruginosa biofilms evade immune clearance, host-focused therapies are emerging alongside antibacterial agents. The bispecific monoclonal antibody MEDI3902, also known as gremubamab, targets Psl and the type three secretion protein PcrV. Phase one studies show favorable safety and pharmacokinetics (PKs) in healthy adults, and preclinical models show protection in bacteremia and pneumonia. Broad target conservation supports clinical applicability [221,222,223].
Vaccine development is active. Foundational work on alginate conjugates and the OprF and OprI formulation IC43 shaped the field. Newer strategies, such as NP presented antigens and multi-epitope constructs, aim to elicit strong antibody responses against adhesins and secretion components relevant to chronic lung disease [224,225,226]. Matrix targeting biologics complements these approaches. Enzymes directed at Psl, including variants of PslG, degrade EPS and increase antibiotic and neutrophil access, which improves host clearance [217,219]. These therapies are increasingly positioned as adjuncts combined with antibiotics, QS or c-di-GMP modulators, or phage to favor host control [224].
Host-directed therapies counter adhesins and secretion systems, generate protective antibodies, and weaken the matrix. These functions shift treatment from antibiotic killing alone to immune-enabled biofilm control, supported by clinical-stage antibodies and next-generation vaccine candidates [221,222].
Table 3 outlines the major therapeutic tracks that include QS disruption, quorum-quenching enzymes such as PvdQ, CRISPR interference of lasI, rhlI, and pqsR, c-di-GMP-reducing agents such as H6-335, NO donors, matrix-focused NP platforms, phage and depolymerase combinations, and host-directed biologics such as MEDI3902. The table links each approach with its molecular target, model system, antibiofilm readouts, and translational status. Figure 4 places these interventions within four major control hubs that include QS, c-di-GMP signaling, matrix integrity, and host immunity and shows how they converge on biofilm collapse and antibiotic resensitization. Together, these strategies provide a system-level view of biofilm dismantling that aims to weaken coordination, reduce tolerance, and restore therapeutic activity.

7. Diagnostic and Analytical Horizons: Reading the Biofilm Code

7.1. Advanced Imaging: CLSM, OCT, and Raman Mapping as a Spatial Code

Confocal laser scanning microscopy (CLSM) remains a central method for tracking P. aeruginosa biofilm development, stress responses, and regression. It provides three-dimensional information on volume, live and dead ratios, height, surface coverage, and roughness. Recent progress lies in standardized analysis. Harmonized workflows reduce operator bias, which is critical when evaluating QS inhibitors or c-di-GMP ligands [237,238]. Benchmark studies that compare dyes and software tools show that consistent image acquisition combined with COMSTAT-based quantification improves estimates of biovolume, secondary thickness, and viability in P. aeruginosa single species and mixed species communities [238,239].
Optical coherence tomography (OCT) covers a mesoscopic range. It is label-free, depth-resolved, and rapid. OCT supports the real-time monitoring of biomass, void fraction and porosity, roughness, and shear-induced detachment on medical materials and industrial lines. Bioluminescence platforms can be paired with OCT to obtain structure-linked viable load readouts or machine learning (ML) based texture scoring over time. Mid infrared (mid IR) and frequency domain OCT (FD OCT) provide better contrast in soft, water-rich materials [240,241,242]. From early capillary flow imaging to current OCT and bioluminescence combinations, studies show that OCT enables rapid and nondestructive measurements of thickness, roughness, void distribution, and detachment on device-scale materials in both laboratory and field settings, including irrigation tubing and medical polymers [240,243,244,245].
Raman microspectroscopy, including surface-enhanced Raman spectroscopy (SERS) and confocal Raman mapping, measures matrix chemistry in situ. In P. aeruginosa, SERS and confocal Raman imaging detect phenazines such as pyocyanin and alkyl quinolones through depth. These approaches visualize quorum signal gradients and redox changes as antibiotics diffuse [246,247,248]. A recent study generated three-dimensional chemical maps with better depth resolution and clearer metabolite-rich layers [249]. Method papers now recommend combining label-free Raman maps with mass spectrometry imaging (MSI), including matrix-assisted laser desorption ionization (MALDI) MSI, to co-register structure and metabolite patterns and reveal the small-molecule code that underlies biofilm behavior [246,250,251]. Practical SERS, with validation by electrochemical SERS, tracks evolving gradients of virulence factors over time [246,252]. MALDI guided MSI workflows identify phenazines and other metabolites across depth, allowing true overlays of chemotypes and structure [253,254,255]. In practice, CLSM provides biovolume, live and dead ratios, surface coverage, and roughness. OCT offers thickness, porosity, and void fraction, and detachment kinetics. Raman and SERS supply depth-resolved maps of pyocyanin, HHQ, PQS, redox state, and antibiotic penetration [238,243,246].

7.2. Omics Integration: Transcriptomic and Proteomic Mapping of QS and c-di-GMP Signatures

Time-resolved dual omics approaches now monitor P. aeruginosa interactions with airway epithelial cells in real time. Dual RNA sequencing (RNA-seq) in organoids and air–liquid interface (ALI) cultures measures both host and bacterial transcripts during biofilm development and remodeling [256,257,258]. These datasets show that the QS hierarchy is conditional. Las, Rhl, and Pqs can reorder under different stresses and media conditions, which agrees with recent reinterpretations of QS regulation [61]. Integrated work supports a simple principle. Environmental cues reshape local c-di-GMP signaling and shift cells toward motile or matrix-dwelling states [259]. Biochemical studies link selected PDEs, including rmcA and morA, to maintenance of mature architecture and tolerance. These findings connect second messenger balance directly to structure and resilience [260].
Genome-scale perturbation is refining the regulatory map. Multiplex CRISPR editing and CRISPR interference across GGDEF and EAL enzymes reveal how distributed enzyme activity sets c-di-GMP levels and defines distinct biofilm and virulence phenotypes [261,262]. These results support a model in which c-di-GMP acts through local hubs and protein interaction networks rather than a single global control [263,264]. Proteomic studies are also identifying actionable markers. For example, PA2146 marks biofilm formation on endoscope channel polymers and demonstrates MALDI mass spectrometry as a useful monitoring tool [265]. The most informative pipeline now integrates RNA-seq and proteomics with structured imaging such as CLSM and OCT, and chemical maps from Raman and MSI. This combination allows QS and c-di-GMP nodes to be aligned with phenazine and PQS gradients and with physical morphology [253,266].

7.3. Rapid Diagnostics: MALDI-TOF Biofilm Profiling, Impedance Biosensing, and Microfluidic Biofilm on Chip Models

Beyond species-level identification, emerging proteomic methods support a biofilm stage marker panel. A distinct approximately 5.45 kilodalton MALDI time of flight (MALDI-TOF) feature corresponding to PA2146 increases during P. aeruginosa biofilm growth on biopsy channel polymers. This marker is validated by liquid chromatography tandem mass spectrometry and is absent in strains lacking the gene, suggesting a practical monitoring signal for endoscope reprocessing [265]. Wider surveys propose that MALDI-TOF combined with MALDI MSI can generate small-molecule fingerprints that reflect quorum regulation and matrix development. These studies also note the current lack of a general biofilm producer database across ESKAPE pathogens [267].
Microfabricated electrochemical impedance spectroscopy (EIS) devices track early attachment and progression to mature biofilm in near real time, often within the first hour. They generate multiphase spectra that correlate with biomass and matrix formation and support closed-loop testing of dispersal triggers or QSIs [268]. Newer architectures enlarge the electroactive area and incorporate flow, which yields large and reproducible impedance changes during growth and after induced dispersal. These advances support compact and inexpensive sensors for monitoring hospital waterlines and device surfaces [269,270]. Reviews of electrochemical biosensors integrated into microfluidic systems highlight high sensitivity, nondestructive measurement, and true time series tracking of biofilm behavior [271].
Standardized biofilm on chip platforms now enable MBIC and MBEC testing under flow, straightforward polymicrobial coculture, and well-controlled gradients that better resemble clinical niches [272]. CF airway organ chips add mucus viscosity, shear, and epithelial barriers and provide a pharmacodynamic (PD) context for antibiotics, QSIs, and c-di-GMP modulators under patient-relevant conditions [273]. A new direction combines MALDI guided MSI with chip-grown P. aeruginosa to analyze quorum signals, phenazines, and structure in parallel [254,274]. This supports a same-day detect, stage, and treat concept. MALDI-TOF markers such as PA2146 report on biofilm stage, EIS defines kinetics and spread, and microfluidic chips provide rapid MBIC and MBEC data to guide therapy [265,268,272].

7.4. AI and Systems Biology: Predicting Biofilm Phenotypes and QSI Efficacy with ML

Explainable machine learning (ML) models now predict P. aeruginosa resistomes and biofilm vulnerabilities from genomic or integrated omics data. These tools include interfaces that validate phenotype predictions, such as responses to tobramycin in chip-grown biofilms [275,276]. Clinical evaluations favor interpretable approaches such as Bayesian and logistic models, while feature-weighted gradient boosting incorporates molecular diagnostic inputs [277]. For antivirulence discovery, quantitative structure-activity relationship (QSAR) models combined with ML help prioritize candidate LasR and RhlR ligands or peptides before testing in biofilm on chip systems [271].
Network-centered studies confirm that c-di-GMP operates through localized hubs. Recent mapping work defines protein neighborhoods that predict sessile versus motile outcomes. These patterns align with CRISPR-based perturbation screens across GGDEF and EAL enzymes and highlight targets for combined QSI and PDE activator strategies [261,263]. Clinically, NO-triggered dispersal remains the best validated PDE activation approach. It transiently lowers c-di-GMP and enhances antibiotic activity, and is supported by controlled CF trials and progress in donor chemistry and delivery [134,278].
A proposed pipeline links these tools into a single workflow. First, OCT and CLSM provide morphometric data. Second, Raman and SERS define metabolite layers. Third, depth fractionated RNA-seq and proteomics reveal transcriptional and protein-level signatures. Fourth, models such as XGBoost and long short-term memory (LSTM) networks predict MBIC, MBEC, and dispersal probability under QS inhibitors and c-di-GMP modulators. Fifth, prospective tests on microfluidic chips with EIS readouts validate these predictions [268,271,272]. This integration moves biofilm assessment toward predictive and mechanism-aware diagnostics.

8. Challenges, Translational Barriers, and One Health Perspectives

Moving antibiofilm strategies toward clinical use requires attention to safety, regulatory expectations, and ecological impact. For NPs and enzyme-based therapies, safety assessment begins with biocompatibility standards. ISO 10993 guides this process. Part 1 outlines test selection, Part 5 addresses in vitro cytotoxicity, and recent guidance from the United States Food and Drug Administration provides conditions under which chemical data or literature can replace animal testing [279,280,281]. Nanomaterials require additional scrutiny. ISO Technical Report 10993-22 calls for explicit characterization of particle size, shape, agglomeration, surface chemistry, and dissolution. It also notes that common cytotoxicity assays may behave differently at the nanoscale [282]. Interlaboratory comparisons reveal substantial variability in ISO 10993-5 cytotoxicity outcomes. These findings highlight the need to standardize extraction conditions, cell lines, and readouts when evaluating candidate coatings or nanoparticle payloads [283,284,285]. Best practice is clear: define the material, document exposure, match device relevant conditions, and pair biological tests with chemical characterization using ISO 10993-18, so that any safety signal can be interpreted accurately [286].
None of these approaches prevents evolution. P. aeruginosa can develop reduced susceptibility to QSIs. Although antivirulence pressure may select more slowly than antibiotic pressure, reviews argue for combination strategies to slow escape [287,288,289]. Phage-based therapies face similar constraints. Cocktails designed for biofilms that target different receptors and combine complementary activities reduce escape routes, and evolution in biofilm assays and phage antibiotic combinations improve durability. Even so, resistance can emerge when dosing or local ecology is not aligned with treatment design [216,290]. Clinically, NO-based dispersal remains a practical adjunct. It can resensitize biofilm aggregates, and early trials and translational reviews outline guidance on donor selection, delivery methods, and endpoints [134,291,292].
A One Health perspective shows why translation must extend beyond patient care. P. aeruginosa persists in premise plumbing. Sink drains, U bends, and faucet components act as long-term reservoirs that seed patient rooms. Whole genome studies repeatedly link isolates from patients to drains and identify high-risk clones such as ST111 and ST235, as well as carbapenemase-producing strains in hospital water systems [293,294,295]. Chlorine-tolerant P. aeruginosa and antimicrobial resistance (AMR) plasmids within plumbing biofilms help explain why routine disinfection may fail without engineering controls and targeted remediation [296,297]. Outside hospitals, drinking water plumbing is recognized as an AMR hot spot. Low flow, stagnation, and limited disinfectant residuals support opportunistic pathogens and increase the potential for resistance exchange [298,299,300]. Global reports emphasize the scale of the problem. Resistance in Gram-negative pathogens continues to rise in hospitals worldwide. These trends argue for diagnostics and interventions that are validated not only in laboratory and animal models, but also in the water systems where biofilms persist [301,302].

9. Conclusions

P. aeruginosa biofilms are dynamic and adaptive systems that are coordinated by QS and c-di-GMP. These networks build a matrix, limit antibiotic access, and support tolerant phenotypes that survive treatment. Understanding this biofilm code clarifies why standard MIC testing often fails in chronic lung disease, wounds, and device-related infections. Limited penetration, active efflux, and persister and VBNC populations act together to protect the community. A practical therapeutic sequence is to disperse, then attack, then clear. First, lowering c-di-GMP and damping quorum signals opens the matrix and releases cells. Next, targeted antimicrobials reach previously protected populations. Finally, host-directed therapies or matrix-degrading adjuncts remove residual biomass. The available toolkit includes c-di-GMP modulators, NO-based dispersal strategies, QS antagonists, signal-degrading enzymes, NPs, enzymatic depolymerases such as Psl targeting PslG, bacteriophage cocktails, and clinical-stage antibodies that recognize Psl or PcrV. Biofilm-aware susceptibility testing through MBIC and MBEC should guide treatment choices rather than planktonic MIC values. Successful translation requires safety by design that is informed by standards such as ISO 10993 for materials and nanosystems, together with standardized and clinically relevant diagnostics. A One Health perspective is also essential, with attention given to reservoirs in both patients and infrastructure. With mechanism-guided combinations, realistic biofilm testing, and upstream surveillance, dismantling the P. aeruginosa biofilm problem becomes an achievable goal.

Author Contributions

Conceptualization, A.E. and E.M.; Data curation, A.E., E.M., H.M.E., M.I., S.A. (Safiyah Alzahrani), S.A. (Sulaiman Anagreyyah), H.A., A.A. (Abdulaziz Alghamdi), A.A. (Ahmed Alzahrani), M.J., and A.A.-O.; Investigation, A.E., E.M., H.M.E., M.I., S.A. (Safiyah Alzahrani), S.A. (Sulaiman Anagreyyah), H.A., A.A. (Abdulaziz Alghamdi), A.A. (Ahmed Alzahrani), M.J., and A.A.-O.; Visualization, A.E., E.M., H.M.E., M.I., S.A. (Safiyah Alzahrani), S.A. (Sulaiman Anagreyyah), H.A., A.A. (Abdulaziz Alghamdi), A.A. (Ahmed Alzahrani), M.J., and A.A.-O.; Writing—original draft, A.E. and E.M.; Writing—review and editing, A.E., E.M., H.M.E., M.I., S.A. (Safiyah Alzahrani), S.A. (Sulaiman Anagreyyah), H.A., A.A. (Abdulaziz Alghamdi), A.A. (Ahmed Alzahrani), M.J., and A.A.-O.; Supervision, A.E.; Project administration, A.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The researchers would like to thank the Deanship of Graduate Studies and Scientific Research at Qassim University for financial support (QU-APC-2026).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. 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]
  2. Ciofu, O.; Tolker-Nielsen, T. Tolerance and resistance of Pseudomonas aeruginosa biofilms to antimicrobial agents—How P. aeruginosa can escape antibiotics. Front. Microbiol. 2019, 10, 913. [Google Scholar] [CrossRef] [PubMed]
  3. Maurice, N.M.; Bedi, B.; Sadikot, R.T. Pseudomonas aeruginosa biofilms: Host response and clinical implications in lung infections. Am. J. Respir. Cell Mol. Biol. 2018, 58, 428–439. [Google Scholar] [CrossRef] [PubMed]
  4. Hentzer, M.; Teitzel, G.M.; Balzer, G.J.; Heydorn, A.; Molin, S.; Givskov, M.; Parsek, M.R. Alginate overproduction affects Pseudomonas aeruginosa biofilm structure and function. J. Bacteriol. 2001, 183, 5395–5401. [Google Scholar] [CrossRef]
  5. Moreau-Marquis, S.; Stanton, B.A.; O’Toole, G.A. Pseudomonas aeruginosa biofilm formation in the cystic fibrosis airway. Pulm. Pharmacol. Ther. 2008, 21, 595–599. [Google Scholar] [CrossRef]
  6. Guillaume, O.; Butnarasu, C.; Visentin, S.; Reimhult, E. Interplay between biofilm microenvironment and pathogenicity of Pseudomonas aeruginosa in cystic fibrosis lung chronic infection. Biofilm 2022, 4, 100089. [Google Scholar] [CrossRef]
  7. Brandenburg, K.S.; Weaver, A.J., Jr.; Qian, L.; You, T.; Chen, P.; Karna, S.R.; Fourcaudot, A.B.; Sebastian, E.A.; Abercrombie, J.J.; Pineda, U. Development of Pseudomonas aeruginosa biofilms in partial-thickness burn wounds using a Sprague-Dawley rat model. J. Burn Care Res. 2019, 40, 44–57. [Google Scholar] [CrossRef]
  8. Brandenburg, K.S.; Weaver, A.J., Jr.; Karna, S.R.; You, T.; Chen, P.; Stryk, S.V.; Qian, L.; Pineda, U.; Abercrombie, J.J.; Leung, K.P. Formation of Pseudomonas aeruginosa biofilms in full-thickness scald burn wounds in rats. Sci. Rep. 2019, 9, 13627. [Google Scholar] [CrossRef]
  9. Cole, S.J.; Records, A.R.; Orr, M.W.; Linden, S.B.; Lee, V.T. Catheter-associated urinary tract infection by Pseudomonas aeruginosa is mediated by exopolysaccharide-independent biofilms. Infect. Immun. 2014, 82, 2048–2058. [Google Scholar] [CrossRef]
  10. Gil-Perotin, S.; Ramirez, P.; Marti, V.; Sahuquillo, J.M.; Gonzalez, E.; Calleja, I.; Menendez, R.; Bonastre, J. Implications of endotracheal tube biofilm in ventilator-associated pneumonia response: A state of concept. Crit. Care 2012, 16, R93. [Google Scholar] [CrossRef]
  11. Guillon, A.; Fouquenet, D.; Morello, E.; Henry, C.; Georgeault, S.; Si-Tahar, M.; Hervé, V. Treatment of Pseudomonas aeruginosa biofilm present in endotracheal tubes by poly-l-lysine. Antimicrob. Agents Chemother. 2018, 62, e00564-18. [Google Scholar] [CrossRef] [PubMed]
  12. Grace, A.; Sahu, R.; Owen, D.R.; Dennis, V.A. Pseudomonas aeruginosa reference strains PAO1 and PA14: A genomic, phenotypic, and therapeutic review. Front. Microbiol. 2022, 13, 1023523. [Google Scholar] [CrossRef] [PubMed]
  13. Stover, C.; Pham, X.; Erwin, A.; Mizoguchi, S.; Warrener, P.; Hickey, M.; Brinkman, F.; Hufnagle, W.; Kowalik, D.; Lagrou, M. Complete genome sequence of Pseudomonas aeruginosa PAO1, an opportunistic pathogen. Nature 2000, 406, 959–964. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, A.J.; Doing, G.; Neff, S.L.; Reiter, T.; Hogan, D.A.; Greene, C.S. Compendium-wide analysis of Pseudomonas aeruginosa core and accessory genes reveals transcriptional patterns across strains PAO1 and PA14. MSystems 2023, 8, e00342-00322. [Google Scholar] [CrossRef]
  15. Varadarajan, A.R.; Allan, R.N.; Valentin, J.D.; Castañeda Ocampo, O.E.; Somerville, V.; Pietsch, F.; Buhmann, M.T.; West, J.; Skipp, P.J.; van der Mei, H.C. An integrated model system to gain mechanistic insights into biofilm-associated antimicrobial resistance in Pseudomonas aeruginosa MPAO1. npj Biofilms Microbiomes 2020, 6, 46. [Google Scholar] [CrossRef]
  16. Lee, J.; Zhang, L. The hierarchy quorum sensing network in Pseudomonas aeruginosa. Protein Cell 2015, 6, 26–41. [Google Scholar] [CrossRef]
  17. Zhang, X.; Li, S.; Zhang, D.; Zheng, S.; Zhou, D.; Hou, Q.; Li, G.; Han, H. A comprehensive review of the pathogenic mechanisms of Pseudomonas aeruginosa: Synergistic effects of virulence factors, quorum sensing, and biofilm formation. Front. Microbiol. 2025, 16, 1619626. [Google Scholar] [CrossRef]
  18. Valentini, M.; Filloux, A. Biofilms and cyclic di-GMP (c-di-GMP) signaling: Lessons from Pseudomonas aeruginosa and other bacteria. J. Biol. Chem. 2016, 291, 12547–12555. [Google Scholar] [CrossRef]
  19. Kuchma, S.; Ballok, A.; Merritt, J.; Hammond, J.; Lu, W.; Rabinowitz, J.D.; O’Toole, G.A. Cyclic di-GMP-mediated repression of swarming motility by Pseudomonas aeruginosa PA14 requires the MotAB stator. J. Bacteriol. 2010, 197, 420–430. [Google Scholar] [CrossRef]
  20. Lichtenberg, M.; Kragh, K.N.; Fritz, B.; Kirkegaard, J.B.; Tolker-Nielsen, T.; Bjarnsholt, T. Cyclic-di-GMP signaling controls metabolic activity in Pseudomonas aeruginosa. Cell Rep. 2022, 41, 111515. [Google Scholar] [CrossRef]
  21. Barraud, N.; Schleheck, D.; Klebensberger, J.; Webb, J.S.; Hassett, D.J.; Rice, S.A.; Kjelleberg, S. Nitric oxide signaling in Pseudomonas aeruginosa biofilms mediates phosphodiesterase activity, decreased cyclic di-GMP levels, and enhanced dispersal. J. Bacteriol. 2009, 191, 7333–7342. [Google Scholar] [CrossRef]
  22. Cutruzzolà, F.; Frankenberg-Dinkel, N. Origin and impact of nitric oxide in Pseudomonas aeruginosa biofilms. J. Bacteriol. 2016, 198, 55–65. [Google Scholar] [CrossRef]
  23. Rasamiravaka, T.; Labtani, Q.; Duez, P.; El Jaziri, M. The formation of biofilms by Pseudomonas aeruginosa: A review of the natural and synthetic compounds interfering with control mechanisms. BioMed Res. Int. 2015, 2015, 759348. [Google Scholar] [CrossRef] [PubMed]
  24. Ma, L.; Conover, M.; Lu, H.; Parsek, M.R.; Bayles, K.; Wozniak, D.J. Assembly and development of the Pseudomonas aeruginosa biofilm matrix. PLoS Pathog. 2009, 5, e1000354. [Google Scholar] [CrossRef] [PubMed]
  25. Caiazza, N.C.; O’Toole, G.A. SadB is required for the transition from reversible to irreversible attachment during biofilm formation by Pseudomonas aeruginosa PA14. J. Bacteriol. 2004, 186, 4476–4485. [Google Scholar] [CrossRef]
  26. Ben-David, Y.; Sporny, M.; Brochin, Y.; Piscon, B.; Roth, S.; Zander, I.; Nisani, M.; Shoshani, S.; Yaron, O.; Karako-Lampert, S. SadB, a mediator of AmrZ proteolysis and biofilm development in Pseudomonas aeruginosa. npj Biofilms Microbiomes 2025, 11, 77. [Google Scholar] [CrossRef]
  27. Papangeli, M.; Luckett, J.; Heeb, S.; Alexander, M.R.; Williams, P.; Dubern, J.-F. SadB acts as a master regulator modulating Pseudomonas aeruginosa pathogenicity. bioRxiv 2025. [Google Scholar] [CrossRef]
  28. Ryder, C.; Byrd, M.; Wozniak, D.J. Role of polysaccharides in Pseudomonas aeruginosa biofilm development. Curr. Opin. Microbiol. 2007, 10, 644–648. [Google Scholar] [CrossRef]
  29. da Cruz Nizer, W.S.; Allison, K.N.; Adams, M.E.; Vargas, M.A.; Ahmed, D.; Beaulieu, C.; Raju, D.; Cassol, E.; Howell, P.L.; Overhage, J. The role of exopolysaccharides Psl and Pel in resistance of Pseudomonas aeruginosa to the oxidative stressors sodium hypochlorite and hydrogen peroxide. Microbiol. Spectr. 2024, 12, e00922–e00924. [Google Scholar] [CrossRef]
  30. Colvin, K.M.; Irie, Y.; Tart, C.S.; Urbano, R.; Whitney, J.C.; Ryder, C.; Howell, P.L.; Wozniak, D.J.; Parsek, M.R. The Pel and Psl polysaccharides provide Pseudomonas aeruginosa structural redundancy within the biofilm matrix. Environ. Microbiol. 2012, 14, 1913–1928. [Google Scholar] [CrossRef]
  31. Wozniak, D.J.; Wyckoff, T.J.; Starkey, M.; Keyser, R.; Azadi, P.; O’Toole, G.A.; Parsek, M.R. Alginate is not a significant component of the extracellular polysaccharide matrix of PA14 and PAO1 Pseudomonas aeruginosa biofilms. Proc. Natl. Acad. Sci. USA 2003, 100, 7907–7912. [Google Scholar] [CrossRef] [PubMed]
  32. Morgan, R.; Kohn, S.; Hwang, S.-H.; Hassett, D.J.; Sauer, K. BdlA, a chemotaxis regulator essential for biofilm dispersion in Pseudomonas aeruginosa. J. Bacteriol. 2006, 188, 7335–7343. [Google Scholar] [CrossRef] [PubMed]
  33. Petrova, O.E.; Sauer, K. Dispersion by Pseudomonas aeruginosa requires an unusual posttranslational modification of BdlA. Proc. Natl. Acad. Sci. USA 2012, 109, 16690–16695. [Google Scholar] [CrossRef]
  34. Petrova, O.E.; Cherny, K.E.; Sauer, K. The diguanylate cyclase GcbA facilitates Pseudomonas aeruginosa biofilm dispersion by activating BdlA. J. Bacteriol. 2015, 197, 174–187. [Google Scholar] [CrossRef] [PubMed]
  35. Li, Y.; Petrova, O.E.; Su, S.; Lau, G.W.; Panmanee, W.; Na, R.; Hassett, D.J.; Davies, D.G.; Sauer, K. BdlA, DipA and induced dispersion contribute to acute virulence and chronic persistence of Pseudomonas aeruginosa. PLoS Pathog. 2014, 10, e1004168. [Google Scholar] [CrossRef]
  36. Passos da Silva, D.; Matwichuk, M.L.; Townsend, D.O.; Reichhardt, C.; Lamba, D.; Wozniak, D.J.; Parsek, M.R. The Pseudomonas aeruginosa lectin LecB binds to the exopolysaccharide Psl and stabilizes the biofilm matrix. Nat. Commun. 2019, 10, 2183. [Google Scholar] [CrossRef]
  37. Le Mauff, F.; Razvi, E.; Reichhardt, C.; Sivarajah, P.; Parsek, M.R.; Howell, P.L.; Sheppard, D.C. The Pel polysaccharide is predominantly composed of a dimeric repeat of α-1, 4 linked galactosamine and N-acetylgalactosamine. Commun. Biol. 2022, 5, 502. [Google Scholar] [CrossRef]
  38. Qu, J.; Cai, Z.; Duan, X.; Zhang, H.; Cheng, H.; Han, S.; Yu, K.; Jiang, Z.; Zhang, Y.; Liu, Y. Pseudomonas aeruginosa modulates alginate biosynthesis and type VI secretion system in two critically ill COVID-19 patients. Cell Biosci. 2022, 12, 14. [Google Scholar] [CrossRef]
  39. Reichhardt, C. The Pseudomonas aeruginosa biofilm matrix protein CdrA has similarities to other fibrillar adhesin proteins. J. Bacteriol. 2023, 205, e00019–e00023. [Google Scholar] [CrossRef]
  40. Fong, J.N.; Yildiz, F.H. Biofilm matrix proteins. Microb. Biofilms 2015, 3, 201–222. [Google Scholar]
  41. Panlilio, H.; Rice, C.V. The role of extracellular DNA in the formation, architecture, stability, and treatment of bacterial biofilms. Biotechnol. Bioeng. 2021, 118, 2129–2141. [Google Scholar] [CrossRef] [PubMed]
  42. Deng, W.; Lei, Y.; Tang, X.; Li, D.; Liang, J.; Luo, J.; Liu, L.; Zhang, W.; Ye, L.; Kong, J. DNase inhibits early biofilm formation in Pseudomonas aeruginosa-or Staphylococcus aureus-induced empyema models. Front. Cell. Infect. Microbiol. 2022, 12, 917038. [Google Scholar] [CrossRef] [PubMed]
  43. Thanabalasuriar, A.; Scott, B.N.V.; Peiseler, M.; Willson, M.E.; Zeng, Z.; Warrener, P.; Keller, A.E.; Surewaard, B.G.J.; Dozier, E.A.; Korhonen, J.T. Neutrophil extracellular traps confine Pseudomonas aeruginosa ocular biofilms and restrict brain invasion. Cell Host Microbe 2019, 25, 526–536.e4. [Google Scholar] [CrossRef] [PubMed]
  44. Landry, R.M.; An, D.; Hupp, J.T.; Singh, P.K.; Parsek, M.R. Mucin–Pseudomonas aeruginosa interactions promote biofilm formation and antibiotic resistance. Mol. Microbiol. 2006, 59, 142–151. [Google Scholar] [CrossRef]
  45. Co, J.Y.; Cárcamo-Oyarce, G.; Billings, N.; Wheeler, K.M.; Grindy, S.C.; Holten-Andersen, N.; Ribbeck, K. Mucins trigger dispersal of Pseudomonas aeruginosa biofilms. NPJ Biofilms Microbiomes 2018, 4, 23. [Google Scholar] [CrossRef]
  46. Dietrich, L.E.; Okegbe, C.; Price-Whelan, A.; Sakhtah, H.; Hunter, R.C.; Newman, D.K. Bacterial community morphogenesis is intimately linked to the intracellular redox state. J. Bacteriol. 2013, 195, 1371–1380. [Google Scholar] [CrossRef]
  47. Williamson, K.S.; Richards, L.A.; Perez-Osorio, A.C.; Pitts, B.; McInnerney, K.; Stewart, P.S.; Franklin, M.J. Heterogeneity in Pseudomonas aeruginosa biofilms includes expression of ribosome hibernation factors in the antibiotic-tolerant subpopulation and hypoxia-induced stress response in the metabolically active population. J. Bacteriol. 2012, 194, 2062–2073. [Google Scholar] [CrossRef]
  48. Glasser, N.R.; Kern, S.E.; Newman, D.K. Phenazine redox cycling enhances anaerobic survival in Pseudomonas aeruginosa by facilitating generation of ATP and a proton-motive force. Mol. Microbiol. 2014, 92, 399–412. [Google Scholar] [CrossRef]
  49. Cornell, W.C.; Zhang, Y.; Bendebury, A.; Hartel, A.J.; Shepard, K.L.; Dietrich, L.E. Phenazine oxidation by a distal electrode modulates biofilm morphogenesis. Biofilm 2020, 2, 100025. [Google Scholar] [CrossRef]
  50. Schiessl, K.T.; Hu, F.; Jo, J.; Nazia, S.Z.; Wang, B.; Price-Whelan, A.; Min, W.; Dietrich, L.E. Phenazine production promotes antibiotic tolerance and metabolic heterogeneity in Pseudomonas aeruginosa biofilms. Nat. Commun. 2019, 10, 762. [Google Scholar] [CrossRef]
  51. Soares, A.; Alexandre, K.; Etienne, M. Tolerance and persistence of Pseudomonas aeruginosa in biofilms exposed to antibiotics: Molecular mechanisms, antibiotic strategies and therapeutic perspectives. Front. Microbiol. 2020, 11, 2057. [Google Scholar] [CrossRef]
  52. Žiemytė, M.; Carda-Diéguez, M.; Rodríguez-Díaz, J.C.; Ventero, M.P.; Mira, A.; Ferrer, M.D. Real-time monitoring of Pseudomonas aeruginosa biofilm growth dynamics and persister cells’ eradication. Emerg. Microbes Infect. 2021, 10, 2062–2075. [Google Scholar] [CrossRef]
  53. Kunnath, A.P.; Suodha Suoodh, M.; Chellappan, D.K.; Chellian, J.; Palaniveloo, K. Bacterial persister cells and development of antibiotic resistance in chronic infections: An update. Br. J. Biomed. Sci. 2024, 81, 12958. [Google Scholar] [CrossRef]
  54. Qi, Z.; Huang, Z.; Liu, C. Metabolism differences of biofilm and planktonic Pseudomonas aeruginosa in viable but nonculturable state induced by chlorine stress. Sci. Total Environ. 2022, 821, 153374. [Google Scholar] [CrossRef] [PubMed]
  55. Wilks, S.A.; Koerfer, V.V.; Prieto, J.A.; Fader, M.; Keevil, C.W. Biofilm development on urinary catheters promotes the appearance of viable but nonculturable bacteria. MBio 2021, 12, e03584-20. [Google Scholar] [CrossRef]
  56. Qi, Z.; Liu, C. Temporal metabolic dynamics and heterogeneity of viable but nonculturable Pseudomonas aeruginosa induced by chlorine disinfection at single-cell resolution. J. Environ. Chem. Eng. 2024, 12, 113514. [Google Scholar] [CrossRef]
  57. Korshoj, L.E.; Kielian, T. Bacterial single-cell RNA sequencing captures biofilm transcriptional heterogeneity and differential responses to immune pressure. Nat. Commun. 2024, 15, 10184. [Google Scholar] [CrossRef] [PubMed]
  58. Dekimpe, V.; Deziel, E. Revisiting the quorum-sensing hierarchy in Pseudomonas aeruginosa: The transcriptional regulator RhlR regulates LasR-specific factors. Microbiology 2009, 155, 712–723. [Google Scholar] [CrossRef]
  59. Feltner, J.B.; Wolter, D.J.; Pope, C.E.; Groleau, M.-C.; Smalley, N.E.; Greenberg, E.P.; Mayer-Hamblett, N.; Burns, J.; Déziel, E.; Hoffman, L.R. LasR variant cystic fibrosis isolates reveal an adaptable quorum-sensing hierarchy in Pseudomonas aeruginosa. MBio 2016, 7, e01513-16. [Google Scholar] [CrossRef]
  60. Soto-Aceves, M.P.; Cocotl-Yañez, M.; Servín-González, L.; Soberón-Chávez, G. The Rhl quorum-sensing system is at the top of the regulatory hierarchy under phosphate-limiting conditions in Pseudomonas aeruginosa PAO1. J. Bacteriol. 2021, 203, e00475-20. [Google Scholar] [CrossRef]
  61. Raya, J.; Montagut, E.-J.; Marco, M.-P. Correction: Analysing the integrated quorum sensing (iqs) system and its potential role in Pseudomonas aeruginosa pathogenesis. Front. Cell. Infect. Microbiol. 2025, 15, 1634997. [Google Scholar] [CrossRef]
  62. Welsh, M.A.; Blackwell, H.E. Chemical genetics reveals environment-specific roles for quorum sensing circuits in Pseudomonas aeruginosa. Cell Chem. Biol. 2016, 23, 361–369. [Google Scholar] [CrossRef]
  63. Hoffman, L.R.; Kulasekara, H.D.; Emerson, J.; Houston, L.S.; Burns, J.L.; Ramsey, B.W.; Miller, S.I. Pseudomonas aeruginosa lasR mutants are associated with cystic fibrosis lung disease progression. J. Cyst. Fibros. 2009, 8, 66–70. [Google Scholar] [CrossRef]
  64. Simanek, K.A.; Taylor, I.R.; Richael, E.K.; Lasek-Nesselquist, E.; Bassler, B.L.; Paczkowski, J.E. The PqsE-RhlR interaction regulates RhlR DNA binding to control virulence factor production in Pseudomonas aeruginosa. Microbiol. Spectr. 2022, 10, e02108–e02121. [Google Scholar] [CrossRef]
  65. Feathers, J.R.; Richael, E.K.; Simanek, K.A.; Fromme, J.C.; Paczkowski, J.E. Structure of the RhlR-PqsE complex from Pseudomonas aeruginosa reveals mechanistic insights into quorum-sensing gene regulation. Structure 2022, 30, 1626–1636.e4. [Google Scholar] [CrossRef] [PubMed]
  66. Groleau, M.-C.; de Oliveira Pereira, T.; Dekimpe, V.; Déziel, E. PqsE is essential for RhlR-dependent quorum sensing regulation in Pseudomonas aeruginosa. Msystems 2020, 5, e00194-20. [Google Scholar] [CrossRef] [PubMed]
  67. Tchadi, B.V.; Derringer, J.J.; Detweiler, A.K.; Taylor, I.R. PqsE adapts the activity of the Pseudomonas aeruginosa quorum-sensing transcription factor RhlR to both autoinducer concentration and promoter sequence identity. J. Bacteriol. 2025, 207, e00516-24. [Google Scholar] [CrossRef] [PubMed]
  68. Coggan, K.A.; Wolfgang, M.C. Global regulatory pathways and cross-talk control Pseudomonas aeruginosa environmental lifestyle and virulence phenotype. Curr. Issues Mol. Biol. 2012, 14, 1–14. [Google Scholar] [CrossRef]
  69. Carey, J.N.; Lamont, S.; Wozniak, D.J.; Dandekar, A.A.; Parsek, M.R. Quorum sensing regulation of Psl polysaccharide production by Pseudomonas aeruginosa. J. Bacteriol. 2024, 206, e00312–24. [Google Scholar] [CrossRef]
  70. Sakuragi, Y.; Kolter, R. Quorum-sensing regulation of the biofilm matrix genes (pel) of Pseudomonas aeruginosa. J. Bacteriol. 2007, 189, 5383–5386. [Google Scholar] [CrossRef]
  71. Ha, D.-G.; O’Toole, G.A. c-di-GMP and its effects on biofilm formation and dispersion: A Pseudomonas aeruginosa review. Microb. Biofilms 2015, 3, 301–317. [Google Scholar] [CrossRef]
  72. An, S.; Wu, J.; Zhang, L.-H. Modulation of Pseudomonas aeruginosa biofilm dispersal by a cyclic-Di-GMP phosphodiesterase with a putative hypoxia-sensing domain. Appl. Environ. Microbiol. 2010, 76, 8160–8173. [Google Scholar] [CrossRef]
  73. Borgert, S.R.; Henke, S.; Witzgall, F.; Schmelz, S.; Zur Lage, S.; Hotop, S.-K.; Stephen, S.; Lübken, D.; Krüger, J.; Gomez, N.O. Moonlighting chaperone activity of the enzyme PqsE contributes to RhlR-controlled virulence of Pseudomonas aeruginosa. Nat. Commun. 2022, 13, 7402. [Google Scholar] [CrossRef]
  74. Whitney, J.C.; Whitfield, G.B.; Marmont, L.S.; Yip, P.; Neculai, A.M.; Lobsanov, Y.D.; Robinson, H.; Ohman, D.E.; Howell, P.L. Dimeric c-di-GMP is required for post-translational regulation of alginate production in Pseudomonas aeruginosa. J. Biol. Chem. 2015, 290, 12451–12462. [Google Scholar] [CrossRef]
  75. Liang, Z.; Rybtke, M.; Kragh, K.N.; Johnson, O.; Schicketanz, M.; Zhang, Y.E.; Andersen, J.B.; Tolker-Nielsen, T. Transcription of the alginate operon in Pseudomonas aeruginosa is regulated by c-di-GMP. Microbiol. Spectr. 2022, 10, e00675-22. [Google Scholar] [CrossRef] [PubMed]
  76. Okkotsu, Y.; Little, A.S.; Schurr, M.J. The Pseudomonas aeruginosa AlgZR two-component system coordinates multiple phenotypes. Front. Cell. Infect. Microbiol. 2014, 4, 82. [Google Scholar] [CrossRef]
  77. Bazire, A.; Shioya, K.; Soum-Soutéra, E.; Bouffartigues, E.; Ryder, C.; Guentas-Dombrowsky, L.; Hémery, G.; Linossier, I.; Chevalier, S.; Wozniak, D.J. The sigma factor AlgU plays a key role in formation of robust biofilms by nonmucoid Pseudomonas aeruginosa. J. Bacteriol. 2010, 192, 3001–3010. [Google Scholar] [CrossRef] [PubMed]
  78. Petrova, O.E.; Sauer, K. SagS contributes to the motile-sessile switch and acts in concert with BfiSR to enable Pseudomonas aeruginosa biofilm formation. J. Bacteriol. 2011, 193, 6614–6628. [Google Scholar] [CrossRef] [PubMed]
  79. Almblad, H.; Harrison, J.J.; Rybtke, M.; Groizeleau, J.; Givskov, M.; Parsek, M.R.; Tolker-Nielsen, T. The cyclic AMP–Vfr signaling pathway in Pseudomonas aeruginosa is inhibited by cyclic di-GMP. J. Bacteriol. 2015, 197, 2190–2200. [Google Scholar] [CrossRef]
  80. Li, K.; Yang, G.; Debru, A.B.; Li, P.; Zong, L.; Li, P.; Xu, T.; Wu, W.; Jin, S.; Bao, Q. SuhB regulates the motile-sessile switch in Pseudomonas aeruginosa through the Gac/Rsm pathway and c-di-GMP signaling. Front. Microbiol. 2017, 8, 1045. [Google Scholar] [CrossRef]
  81. 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]
  82. Giallonardi, G.; Letizia, M.; Mellini, M.; Frangipani, E.; Halliday, N.; Heeb, S.; Cámara, M.; Visca, P.; Imperi, F.; Leoni, L. Alkyl-quinolone-dependent quorum sensing controls prophage-mediated autolysis in Pseudomonas aeruginosa colony biofilms. Front. Cell. Infect. Microbiol. 2023, 13, 1183681. [Google Scholar] [CrossRef]
  83. Cooke, A.C.; Florez, C.; Dunshee, E.B.; Lieber, A.D.; Terry, M.L.; Light, C.J.; Schertzer, J.W. Pseudomonas quinolone signal-induced outer membrane vesicles enhance biofilm dispersion in Pseudomonas aeruginosa. mSphere 2020, 5, e01109-20. [Google Scholar] [CrossRef]
  84. Florez, C.; Raab, J.E.; Cooke, A.C.; Schertzer, J.W. Membrane distribution of the Pseudomonas quinolone signal modulates outer membrane vesicle production in Pseudomonas aeruginosa. mBio 2017, 8, e01034-17. [Google Scholar] [CrossRef] [PubMed]
  85. Mitchell, G.; Séguin, D.L.; Asselin, A.-E.; Déziel, E.; Cantin, A.M.; Frost, E.H.; Michaud, S.; Malouin, F. Staphylococcus aureus sigma B-dependent emergence of small-colony variants and biofilm production following exposure to Pseudomonas aeruginosa 4-hydroxy-2-heptylquinoline-N-oxide. BMC Microbiol. 2010, 10, 33. [Google Scholar] [CrossRef]
  86. Proctor, R. Respiration and small-colony variants of Staphylococcus aureus. Microbiol. Spectr. 2019, 7, GPP3-0069-2019. [Google Scholar] [CrossRef] [PubMed]
  87. Roman-Rodriguez, F.; Kim, J.; Parker, D.; Boyd, J.M. An effective response to respiratory inhibition by a Pseudomonas aeruginosa-excreted quinoline promotes Staphylococcus aureus fitness and survival in co-culture. bioRxiv 2025. [Google Scholar] [CrossRef]
  88. Orazi, G.; O’Toole, G.A. Pseudomonas aeruginosa alters Staphylococcus aureus sensitivity to vancomycin in a biofilm model of cystic fibrosis infection. mBio 2017, 8, e00873-17. [Google Scholar] [CrossRef] [PubMed]
  89. Biswas, L.; Biswas, R.; Schlag, M.; Bertram, R.; Götz, F. Small-colony variant selection as a survival strategy for Staphylococcus aureus in the presence of Pseudomonas aeruginosa. Appl. Environ. Microbiol. 2009, 75, 6910–6912. [Google Scholar] [CrossRef]
  90. Ahmed, S.A.; Rudden, M.; Smyth, T.J.; Dooley, J.S.; Marchant, R.; Banat, I.M. Natural quorum sensing inhibitors effectively downregulate gene expression of Pseudomonas aeruginosa virulence factors. Appl. Microbiol. Biotechnol. 2019, 103, 3521–3535. [Google Scholar] [CrossRef]
  91. Lim, T.; Ham, S.-Y.; Nam, S.; Kim, M.; Lee, K.Y.; Park, H.-D.; Byun, Y. Recent advance in small molecules targeting RhlR of Pseudomonas aeruginosa. Antibiotics 2022, 11, 274. [Google Scholar] [CrossRef]
  92. Rodríguez-Urretavizcaya, B.; Vilaplana, L.; Marco, M.-P. Strategies for quorum sensing inhibition as a tool for controlling Pseudomonas aeruginosa infections. Int. J. Antimicrob. Agents 2024, 64, 107323. [Google Scholar] [CrossRef]
  93. Hoffman, L.R.; Déziel, E.; d’Argenio, D.A.; Lépine, F.; Emerson, J.; McNamara, S.; Gibson, R.L.; Ramsey, B.W.; Miller, S.I. Selection for Staphylococcus aureus small-colony variants due to growth in the presence of Pseudomonas aeruginosa. Proc. Natl. Acad. Sci. USA 2006, 103, 19890–19895. [Google Scholar] [CrossRef]
  94. Barraza, J.P.; Whiteley, M. A Pseudomonas aeruginosa antimicrobial affects the biogeography but not fitness of Staphylococcus aureus during coculture. mBio 2021, 12, e00047-21. [Google Scholar] [CrossRef]
  95. Nguyen, A.T.; Oglesby-Sherrouse, A.G. Interactions between Pseudomonas aeruginosa and Staphylococcus aureus during co-cultivations and polymicrobial infections. Appl. Microbiol. Biotechnol. 2016, 100, 6141–6148. [Google Scholar] [CrossRef] [PubMed]
  96. Pallett, R.; Leslie, L.J.; Lambert, P.A.; Milic, I.; Devitt, A.; Marshall, L.J. Anaerobiosis influences virulence properties of Pseudomonas aeruginosa cystic fibrosis isolates and the interaction with Staphylococcus aureus. Sci. Rep. 2019, 9, 6748. [Google Scholar] [CrossRef]
  97. Gomes-Fernandes, M.; Gomez, A.-C.; Bravo, M.; Huedo, P.; Coves, X.; Prat-Aymerich, C.; Gibert, I.; Lacoma, A.; Yero, D. Strain-specific interspecies interactions between co-isolated pairs of Staphylococcus aureus and Pseudomonas aeruginosa from patients with tracheobronchitis or bronchial colonization. Sci. Rep. 2022, 12, 3374. [Google Scholar] [CrossRef] [PubMed]
  98. Shah, R.; Narh, J.K.; Urlaub, M.; Jankiewicz, O.; Johnson, C.; Livingston, B.; Dahl, J.-U. Pseudomonas aeruginosa kills Staphylococcus aureus in a polyphosphate-dependent manner. mSphere 2024, 9, e00686-24. [Google Scholar] [CrossRef]
  99. Kessler, E.; Safrin, M.; Abrams, W.R.; Rosenbloom, J.; Ohman, D.E. Inhibitors and specificity of Pseudomonas aeruginosa LasA. J. Biol. Chem. 1997, 272, 9884–9889. [Google Scholar] [CrossRef] [PubMed]
  100. Keim, K.; Bhattacharya, M.; Crosby, H.A.; Jenul, C.; Mills, K.; Schurr, M.; Horswill, A. Polymicrobial interactions between Staphylococcus aureus and Pseudomonas aeruginosa promote biofilm formation and persistence in chronic wound infections. bioRxiv 2024. [Google Scholar] [CrossRef]
  101. Hotterbeekx, A.; Kumar-Singh, S.; Goossens, H.; Malhotra-Kumar, S. In vivo and In vitro Interactions between Pseudomonas aeruginosa and Staphylococcus spp. Front. Cell. Infect. Microbiol. 2017, 7, 106. [Google Scholar]
  102. Kostylev, M.; Kim, D.Y.; Smalley, N.E.; Salukhe, I.; Greenberg, E.P.; Dandekar, A.A. Evolution of the Pseudomonas aeruginosa quorum-sensing hierarchy. Proc. Natl. Acad. Sci. USA 2019, 116, 7027–7032. [Google Scholar] [CrossRef]
  103. Diggle, S.P.; Matthijs, S.; Wright, V.J.; Fletcher, M.P.; Chhabra, S.R.; Lamont, I.L.; Kong, X.; Hider, R.C.; Cornelis, P.; Cámara, M. The Pseudomonas aeruginosa 4-quinolone signal molecules HHQ and PQS play multifunctional roles in quorum sensing and iron entrapment. Chem. Biol. 2007, 14, 87–96. [Google Scholar] [CrossRef]
  104. Lin, J.; Cheng, J.; Wang, Y.; Shen, X. The Pseudomonas quinolone signal (PQS): Not just for quorum sensing anymore. Front. Cell. Infect. Microbiol. 2018, 8, 230. [Google Scholar] [CrossRef] [PubMed]
  105. Déziel, E.; Gopalan, S.; Tampakaki, A.P.; Lépine, F.; Padfield, K.E.; Saucier, M.; Xiao, G.; Rahme, L.G. The contribution of MvfR to Pseudomonas aeruginosa pathogenesis and quorum sensing circuitry regulation: Multiple quorum sensing-regulated genes are modulated without affecting lasRI, rhlRI or the production of N-acyl-L-homoserine lactones. Mol. Microbiol. 2005, 55, 998–1014. [Google Scholar] [CrossRef]
  106. Lee, J.; Wu, J.; Deng, Y.; Wang, J.; Wang, C.; Wang, J.; Chang, C.; Dong, Y.; Williams, P.; Zhang, L.-H. A cell–cell communication signal integrates quorum sensing and stress response. Nat. Chem. Biol. 2013, 9, 339–343. [Google Scholar] [CrossRef] [PubMed]
  107. Merighi, M.; Lee, V.T.; Hyodo, M.; Hayakawa, Y.; Lory, S. The second messenger bis-(3′-5′)-cyclic-GMP and its PilZ domain-containing receptor Alg44 are required for alginate biosynthesis in Pseudomonas aeruginosa. Mol. Microbiol. 2007, 65, 876–895. [Google Scholar] [CrossRef] [PubMed]
  108. Valentini, M.; Filloux, A. Multiple roles of c-di-GMP signaling in bacterial pathogenesis. Annu. Rev. Microbiol. 2019, 73, 387–406. [Google Scholar] [CrossRef]
  109. Park, S.; Sauer, K. Controlling biofilm development through cyclic di-GMP signaling. Adv. Exp. Med. Biol. 2022, 1386, 69–94. [Google Scholar]
  110. Güvener, Z.T.; Harwood, C.S. Subcellular location characteristics of the Pseudomonas aeruginosa GGDEF protein, WspR, indicate that it produces cyclic-di-GMP in response to growth on surfaces. Mol. Microbiol. 2007, 66, 1459–1473. [Google Scholar] [CrossRef]
  111. Huangyutitham, V.; Güvener, Z.T.; Harwood, C.S. Subcellular clustering of the phosphorylated WspR response regulator protein stimulates its diguanylate cyclase activity. mBio 2013, 4, e00242-13. [Google Scholar] [CrossRef]
  112. O’Neal, L.; Baraquet, C.; Suo, Z.; Dreifus, J.E.; Peng, Y.; Raivio, T.L.; Wozniak, D.J.; Harwood, C.S.; Parsek, M.R. The Wsp system of Pseudomonas aeruginosa links surface sensing and cell envelope stress. Proc. Natl. Acad. Sci. USA 2022, 119, e2117633119. [Google Scholar] [CrossRef] [PubMed]
  113. Guan, C.; Huang, Y.; Zhou, Y.; Han, Y.; Liu, S.; Liu, S.; Kong, W.; Wang, T.; Zhang, Y. FlhF affects the subcellular clustering of WspR through HsbR in Pseudomonas aeruginosa. Appl. Environ. Microbiol. 2024, 90, e01548-23. [Google Scholar] [CrossRef] [PubMed]
  114. Baker, A.E.; Webster, S.S.; Diepold, A.; Kuchma, S.L.; Bordeleau, E.; Armitage, J.P.; O’Toole, G.A. Flagellar stators stimulate c-di-GMP production by Pseudomonas aeruginosa. J. Bacteriol. 2019, 201, e00741-18. [Google Scholar] [CrossRef]
  115. Merritt, J.H.; Ha, D.-G.; Cowles, K.N.; Lu, W.; Morales, D.K.; Rabinowitz, J.; Gitai, Z.; O’Toole, G.A. Specific control of Pseudomonas aeruginosa surface-associated behaviors by two c-di-GMP diguanylate cyclases. mBio 2010, 1, e00183-10. [Google Scholar] [CrossRef]
  116. Kuchma, S.L.; Brothers, K.M.; Merritt, J.H.; Liberati, N.T.; Ausubel, F.M.; O’Toole, G.A. BifA, a cyclic-Di-GMP phosphodiesterase, inversely regulates biofilm formation and swarming motility by Pseudomonas aeruginosa PA14. J. Bacteriol. 2007, 189, 8165–8178. [Google Scholar] [CrossRef]
  117. Liu, C.; Liew, C.W.; Wong, Y.H.; Tan, S.T.; Poh, W.H.; Manimekalai, M.S.; Rajan, S.; Xin, L.; Liang, Z.-X.; Grüber, G. Insights into biofilm dispersal regulation from the crystal structure of the PAS-GGDEF-EAL region of RbdA from Pseudomonas aeruginosa. J. Bacteriol. 2018, 200, e00515-17. [Google Scholar] [CrossRef]
  118. Cordery, C.; Craddock, J.; Malý, M.; Basavaraja, K.; Webb, J.S.; Walsh, M.A.; Tews, I. Control of phosphodiesterase activity in the regulator of biofilm dispersal RbdA from Pseudomonas aeruginosa. RSC Chem. Biol. 2024, 5, 1052–1059. [Google Scholar] [CrossRef] [PubMed]
  119. Eilers, K.; Kuok Hoong Yam, J.; Morton, R.; Mei Hui Yong, A.; Brizuela, J.; Hadjicharalambous, C.; Liu, X.; Givskov, M.; Rice, S.A.; Filloux, A. Phenotypic and integrated analysis of a comprehensive Pseudomonas aeruginosa PAO1 library of mutants lacking cyclic-di-GMP-related genes. Front. Microbiol. 2022, 13, 949597. [Google Scholar] [CrossRef]
  120. Li, Z.; Chen, J.-H.; Hao, Y.; Nair, S.K. Structures of the PelD cyclic diguanylate effector involved in pellicle formation in Pseudomonas aeruginosa PAO1. J. Biol. Chem. 2012, 287, 30191–30204. [Google Scholar] [CrossRef]
  121. Remminghorst, U.; Rehm, B.H. Alg44, a unique protein required for alginate biosynthesis in Pseudomonas aeruginosa. FEBS Lett. 2006, 580, 3883–3888. [Google Scholar] [CrossRef]
  122. Newell, P.D.; Boyd, C.D.; Sondermann, H.; O’Toole, G.A. A c-di-GMP effector system controls cell adhesion by inside-out signaling and surface protein cleavage. PLoS Biol. 2011, 9, e1000587. [Google Scholar] [CrossRef]
  123. Cooley, R.B.; O’Donnell, J.P.; Sondermann, H. Coincidence detection and bi-directional transmembrane signaling control a bacterial second messenger receptor. Elife 2016, 5, e21848. [Google Scholar] [CrossRef]
  124. Rybtke, M.; Berthelsen, J.; Yang, L.; Høiby, N.; Givskov, M.; Tolker-Nielsen, T. The LapG protein plays a role in Pseudomonas aeruginosa biofilm formation by controlling the presence of the CdrA adhesin on the cell surface. Microbiologyopen 2015, 4, 917–930. [Google Scholar] [CrossRef]
  125. Ribbe, J.; Baker, A.E.; Euler, S.; O’Toole, G.A.; Maier, B. Role of cyclic di-GMP and exopolysaccharide in type IV pilus dynamics. J. Bacteriol. 2017, 199, e00859-16. [Google Scholar] [CrossRef] [PubMed]
  126. Webster, S.S.; Lee, C.K.; Schmidt, W.C.; Wong, G.C.; O’Toole, G.A. Interaction between the type 4 pili machinery and a diguanylate cyclase fine-tune c-di-GMP levels during early biofilm formation. Proc. Natl. Acad. Sci. USA 2021, 118, e2105566118. [Google Scholar] [CrossRef]
  127. Zemke, A.C.; D’Amico, E.J.; Snell, E.C.; Torres, A.M.; Kasturiarachi, N.; Bomberger, J.M. Dispersal of epithelium-associated Pseudomonas aeruginosa biofilms. mSphere 2020, 5, e00630-20. [Google Scholar] [CrossRef]
  128. Wille, J.; Coenye, T. Biofilm dispersion: The key to biofilm eradication or opening Pandora’s box? Biofilm 2020, 2, 100027. [Google Scholar] [CrossRef]
  129. Randall, T.E.; Eckartt, K.; Kakumanu, S.; Price-Whelan, A.; Dietrich, L.E.; Harrison, J.J. Sensory perception in bacterial cyclic diguanylate signal transduction. J. Bacteriol. 2022, 204, e00433-21. [Google Scholar] [CrossRef] [PubMed]
  130. Almblad, H.; Randall, T.E.; Liu, F.; Leblanc, K.; Groves, R.A.; Kittichotirat, W.; Winsor, G.L.; Fournier, N.; Au, E.; Groizeleau, J. Bacterial cyclic diguanylate signaling networks sense temperature. Nat. Commun. 2021, 12, 1986. [Google Scholar] [CrossRef] [PubMed]
  131. Chen, A.I.; Dolben, E.F.; Okegbe, C.; Harty, C.E.; Golub, Y.; Thao, S.; Ha, D.G.; Willger, S.D.; O’Toole, G.A.; Harwood, C.S. Candida albicans ethanol stimulates Pseudomonas aeruginosa WspR-controlled biofilm formation as part of a cyclic relationship involving phenazines. PLoS Pathog. 2014, 10, e1004480. [Google Scholar] [CrossRef] [PubMed]
  132. Roy, A.B.; Petrova, O.E.; Sauer, K. The phosphodiesterase DipA (PA5017) is essential for Pseudomonas aeruginosa biofilm dispersion. J. Bacteriol. 2012, 194, 2904–2915. [Google Scholar] [CrossRef]
  133. Feng, Q.; Ahator, S.D.; Zhou, T.; Liu, Z.; Lin, Q.; Liu, Y.; Huang, J.; Zhou, J.; Zhang, L.-H. Regulation of exopolysaccharide production by ProE, a cyclic-di-GMP phosphodiesterase in Pseudomonas aeruginosa PAO1. Front. Microbiol. 2020, 11, 1226. [Google Scholar] [CrossRef] [PubMed]
  134. Howlin, R.P.; Cathie, K.; Hall-Stoodley, L.; Cornelius, V.; Duignan, C.; Allan, R.N.; Fernandez, B.O.; Barraud, N.; Bruce, K.D.; Jefferies, J. Low-dose nitric oxide as targeted anti-biofilm adjunctive therapy to treat chronic Pseudomonas aeruginosa infection in cystic fibrosis. Mol. Ther. 2017, 25, 2104–2116. [Google Scholar] [CrossRef]
  135. Hay, I.D.; Wang, Y.; Moradali, M.F.; Rehman, Z.U.; Rehm, B.H. Genetics and regulation of bacterial alginate production. Environ. Microbiol. 2014, 16, 2997–3011. [Google Scholar] [CrossRef]
  136. Franklin, M.J.; Nivens, D.E.; Weadge, J.T.; Howell, P.L. Biosynthesis of the Pseudomonas aeruginosa extracellular polysaccharides, alginate, Pel, and Psl. Front. Microbiol. 2011, 2, 167. [Google Scholar]
  137. Shang, L.; Yan, Y.; Zhan, Y.; Ke, X.; Shao, Y.; Liu, Y.; Yang, H.; Wang, S.; Dai, S.; Lu, J. A regulatory network involving Rpo, Gac and Rsm for nitrogen-fixing biofilm formation by Pseudomonas stutzeri. npj Biofilms Microbiomes 2021, 7, 54. [Google Scholar] [CrossRef]
  138. Moscoso, J.A.; Jaeger, T.; Valentini, M.; Hui, K.; Jenal, U.; Filloux, A. The diguanylate cyclase SadC is a central player in Gac/Rsm-mediated biofilm formation in Pseudomonas aeruginosa. J. Bacteriol. 2014, 196, 4081–4088. [Google Scholar] [CrossRef]
  139. Kim, H.-S.; Ham, S.-Y.; Ryoo, H.-S.; Kim, D.-H.; Yun, E.-T.; Park, H.-D.; Park, J.-H. Inhibiting bacterial biofilm formation by stimulating c-di-GMP regulation using citrus peel extract from Jeju Island. Sci. Total Environ. 2023, 872, 162180. [Google Scholar] [CrossRef]
  140. Billings, N.; Ramirez Millan, M.; Caldara, M.; Rusconi, R.; Tarasova, Y.; Stocker, R.; Ribbeck, K. The extracellular matrix component Psl provides fast-acting antibiotic defense in Pseudomonas aeruginosa biofilms. PLoS Pathog. 2013, 9, e1003526. [Google Scholar] [CrossRef] [PubMed]
  141. Jennings, L.K.; Storek, K.M.; Ledvina, H.E.; Coulon, C.; Marmont, L.S.; Sadovskaya, I.; Secor, P.R.; Tseng, B.S.; Scian, M.; Filloux, A. Pel is a cationic exopolysaccharide that cross-links extracellular DNA in the Pseudomonas aeruginosa biofilm matrix. Proc. Natl. Acad. Sci. USA 2015, 112, 11353–11358. [Google Scholar] [CrossRef]
  142. Mulcahy, H.; Charron-Mazenod, L.; Lewenza, S. Extracellular DNA chelates cations and induces antibiotic resistance in Pseudomonas aeruginosa biofilms. PLoS Pathog. 2008, 4, e1000213. [Google Scholar] [CrossRef]
  143. Chiang, W.-C.; Nilsson, M.; Jensen, P.Ø.; Høiby, N.; Nielsen, T.E.; Givskov, M.; Tolker-Nielsen, T. Extracellular DNA shields against aminoglycosides in Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 2013, 57, 2352–2361. [Google Scholar] [CrossRef]
  144. Das, T.; Sehar, S.; Koop, L.; Wong, Y.K.; Ahmed, S.; Siddiqui, K.S.; Manefield, M. Influence of calcium in extracellular DNA mediated bacterial aggregation and biofilm formation. PLoS ONE 2014, 9, e91935. [Google Scholar] [CrossRef]
  145. 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. Targeted disruption of the extracellular polymeric network of Pseudomonas aeruginosa biofilms by alginate oligosaccharides. npj Biofilms Microbiomes 2018, 4, 13. [Google Scholar] [CrossRef]
  146. Pamp, S.J.; Gjermansen, M.; Johansen, H.K.; Tolker-Nielsen, T. Tolerance to the antimicrobial peptide colistin in Pseudomonas aeruginosa biofilms is linked to metabolically active cells, and depends on the pmr and mexAB-oprM genes. Mol. Microbiol. 2008, 68, 223–240. [Google Scholar] [CrossRef]
  147. Liao, J.; Schurr, M.J.; Sauer, K. The MerR-like regulator BrlR confers biofilm tolerance by activating multidrug efflux pumps in Pseudomonas aeruginosa biofilms. J. Bacteriol. 2013, 195, 3352–3363. [Google Scholar] [CrossRef] [PubMed]
  148. Chambers, J.R.; Sauer, K. The MerR-like regulator BrlR impairs Pseudomonas aeruginosa biofilm tolerance to colistin by repressing PhoPQ. J. Bacteriol. 2013, 195, 4678–4688. [Google Scholar] [CrossRef] [PubMed]
  149. Gupta, K.; Marques, C.N.; Petrova, O.E.; Sauer, K. Antimicrobial tolerance of Pseudomonas aeruginosa biofilms is activated during an early developmental stage and requires the two-component hybrid SagS. J. Bacteriol. 2013, 195, 4975–4987. [Google Scholar] [CrossRef] [PubMed]
  150. Horna, G.; López, M.; Guerra, H.; Saénz, Y.; Ruiz, J. Interplay between MexAB-OprM and MexEF-OprN in clinical isolates of Pseudomonas aeruginosa. Sci. Rep. 2018, 8, 16463. [Google Scholar] [CrossRef]
  151. Wu, W.; Huang, J.; Xu, Z. Antibiotic influx and efflux in Pseudomonas aeruginosa: Regulation and therapeutic implications. Microb. Biotechnol. 2024, 17, e14487. [Google Scholar] [CrossRef] [PubMed]
  152. Lin, J.; Xu, C.; Fang, R.; Cao, J.; Zhang, X.; Zhao, Y.; Dong, G.; Sun, Y.; Zhou, T. Resistance and heteroresistance to colistin in Pseudomonas aeruginosa isolates from Wenzhou, China. Antimicrob. Agents Chemother. 2019, 63, e00556-19. [Google Scholar] [CrossRef] [PubMed]
  153. Rampioni, G.; Pillai, C.R.; Longo, F.; Bondì, R.; Baldelli, V.; Messina, M.; Imperi, F.; Visca, P.; Leoni, L. Effect of efflux pump inhibition on Pseudomonas aeruginosa transcriptome and virulence. Sci. Rep. 2017, 7, 11392. [Google Scholar] [CrossRef]
  154. Vaillancourt, M.; Limsuwannarot, S.P.; Bresee, C.; Poopalarajah, R.; Jorth, P. Pseudomonas aeruginosa mexR and mexEF antibiotic efflux pump variants exhibit increased virulence. Antibiotics 2021, 10, 1164. [Google Scholar] [CrossRef] [PubMed]
  155. Jo, J.; Price-Whelan, A.; Dietrich, L.E. Gradients and consequences of heterogeneity in biofilms. Nat. Rev. Microbiol. 2022, 20, 593–607. [Google Scholar] [CrossRef]
  156. 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]
  157. Mangiaterra, G.; Cedraro, N.; Vaiasicca, S.; Citterio, B.; Galeazzi, R.; Laudadio, E.; Mobbili, G.; Minnelli, C.; Bizzaro, D.; Biavasco, F. Role of tobramycin in the induction and maintenance of viable but non-culturable Pseudomonas aeruginosa in an in vitro biofilm model. Antibiotics 2020, 9, 399. [Google Scholar] [CrossRef]
  158. Mangiaterra, G.; Cedraro, N.; Vaiasicca, S.; Citterio, B.; Frangipani, E.; Biavasco, F.; Vignaroli, C. Involvement of acquired tobramycin resistance in the shift to the viable but non-culturable state in Pseudomonas aeruginosa. Int. J. Mol. Sci. 2023, 24, 11618. [Google Scholar] [CrossRef]
  159. Mao, G.; Song, Y.; Bartlam, M.; Wang, Y. Long-term effects of residual chlorine on Pseudomonas aeruginosa in simulated drinking water fed with low AOC medium. Front. Microbiol. 2018, 9, 879. [Google Scholar] [CrossRef]
  160. Stewart, P.S.; Franklin, M.J.; Williamson, K.S.; Folsom, J.P.; Boegli, L.; James, G.A. Contribution of stress responses to antibiotic tolerance in Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 2015, 59, 3838–3847. [Google Scholar] [CrossRef]
  161. Molin, S.; Tolker-Nielsen, T. Gene transfer occurs with enhanced efficiency in biofilms and induces enhanced stabilisation of the biofilm structure. Curr. Opin. Biotechnol. 2003, 14, 255–261. [Google Scholar] [CrossRef]
  162. Stalder, T.; Top, E. Plasmid transfer in biofilms: A perspective on limitations and opportunities. NPJ Biofilms Microbiomes 2016, 2, 1–5. [Google Scholar] [CrossRef]
  163. Røder, H.L.; Trivedi, U.; Russel, J.; Kragh, K.N.; Herschend, J.; Thalsø-Madsen, I.; Tolker-Nielsen, T.; Bjarnsholt, T.; Burmølle, M.; Madsen, J.S. Biofilms can act as plasmid reserves in the absence of plasmid specific selection. npj Biofilms Microbiomes 2021, 7, 78. [Google Scholar] [CrossRef]
  164. Metzger, G.A.; Ridenhour, B.J.; France, M.; Gliniewicz, K.; Millstein, J.; Settles, M.L.; Forney, L.J.; Stalder, T.; Top, E.M. Biofilms preserve the transmissibility of a multi-drug resistance plasmid. npj Biofilms Microbiomes 2022, 8, 95. [Google Scholar] [CrossRef]
  165. Pizzolato-Cezar, L.R.; Spira, B.; Machini, M.T. Bacterial toxin-antitoxin systems: Novel insights on toxin activation across populations and experimental shortcomings. Curr. Res. Microb. Sci. 2023, 5, 100204. [Google Scholar] [CrossRef]
  166. Lin, J.; Guo, Y.; Yao, J.; Tang, K.; Wang, X. Applications of toxin-antitoxin systems in synthetic biology. Eng. Microbiol. 2023, 3, 100069. [Google Scholar] [CrossRef] [PubMed]
  167. Dangla-Pélissier, G.; Roux, N.; Schmidt, V.; Chambonnier, G.; Ba, M.; Sebban-Kreuzer, C.; de Bentzmann, S.; Giraud, C.; Bordi, C. The horizontal transfer of Pseudomonas aeruginosa PA14 ICE PAPI-1 is controlled by a transcriptional triad between TprA, NdpA2 and MvaT. Nucleic Acids Res. 2021, 49, 10956–10974. [Google Scholar] [CrossRef]
  168. Johnston, E.L.; Zavan, L.; Bitto, N.J.; Petrovski, S.; Hill, A.F.; Kaparakis-Liaskos, M. Planktonic and biofilm-derived Pseudomonas aeruginosa outer membrane vesicles facilitate horizontal gene transfer of plasmid DNA. Microbiol. Spectr. 2023, 11, e05179-22. [Google Scholar] [CrossRef] [PubMed]
  169. Alonso-del Valle, A.; León-Sampedro, R.; Rodríguez-Beltrán, J.; DelaFuente, J.; Hernández-García, M.; Ruiz-Garbajosa, P.; Cantón, R.; Peña-Miller, R.; San Millán, A. Variability of plasmid fitness effects contributes to plasmid persistence in bacterial communities. Nat. Commun. 2021, 12, 2653. [Google Scholar] [CrossRef]
  170. Thieme, L.; Hartung, A.; Tramm, K.; Klinger-Strobel, M.; Jandt, K.D.; Makarewicz, O.; Pletz, M.W. MBEC versus MBIC: The lack of differentiation between biofilm reducing and inhibitory effects as a current problem in biofilm methodology. Biol. Proced. Online 2019, 21, 18. [Google Scholar] [CrossRef] [PubMed]
  171. Coenye, T. Biofilm antimicrobial susceptibility testing: Where are we and where could we be going? Clin. Microbiol. Rev. 2023, 36, e00024-23. [Google Scholar] [CrossRef] [PubMed]
  172. Macia, M.; Rojo-Molinero, E.; Oliver, A. Antimicrobial susceptibility testing in biofilm-growing bacteria. Clin. Microbiol. Infect. 2014, 20, 981–990. [Google Scholar] [CrossRef]
  173. Okae, Y.; Nishitani, K.; Sakamoto, A.; Kawai, T.; Tomizawa, T.; Saito, M.; Kuroda, Y.; Matsuda, S. Estimation of minimum biofilm eradication concentration (MBEC) on in vivo biofilm on orthopedic implants in a rodent femoral infection model. Front. Cell. Infect. Microbiol. 2022, 12, 896978. [Google Scholar] [CrossRef]
  174. Hentzer, M.; Wu, H.; Andersen, J.B.; Riedel, K.; Rasmussen, T.B.; Bagge, N.; Kumar, N.; Schembri, M.A.; Song, Z.; Kristoffersen, P. Attenuation of Pseudomonas aeruginosa virulence by quorum sensing inhibitors. EMBO J. 2003, 22, 3803–3815. [Google Scholar] [CrossRef]
  175. Hentzer, M.; Riedel, K.; Rasmussen, T.B.; Heydorn, A.; Andersen, J.B.; Parsek, M.R.; Rice, S.A.; Eberl, L.; Molin, S.; Høiby, N. Inhibition of quorum sensing in Pseudomonas aeruginosa biofilm bacteria by a halogenated furanone compound. Microbiology 2002, 148, 87–102. [Google Scholar] [CrossRef]
  176. Jakobsen, T.H.; van Gennip, M.; Phipps, R.K.; Shanmugham, M.S.; Christensen, L.D.; Alhede, M.; Skindersoe, M.E.; Rasmussen, T.B.; Friedrich, K.; Uthe, F. Ajoene, a sulfur-rich molecule from garlic, inhibits genes controlled by quorum sensing. Antimicrob. Agents Chemother. 2012, 56, 2314–2325. [Google Scholar] [CrossRef]
  177. Jakobsen, T.H.; Warming, A.N.; Vejborg, R.M.; Moscoso, J.A.; Stegger, M.; Lorenzen, F.; Rybtke, M.; Andersen, J.B.; Petersen, R.; Andersen, P.S. A broad range quorum sensing inhibitor working through sRNA inhibition. Sci. Rep. 2017, 7, 9857. [Google Scholar] [CrossRef]
  178. Naga, N.G.; Zaki, A.A.; El-Badan, D.E.; Rateb, H.S.; Ghanem, K.M.; Shaaban, M.I. Inhibition of Pseudomonas aeruginosa quorum sensing by methyl gallate from Mangifera indica. Sci. Rep. 2023, 13, 17942. [Google Scholar] [CrossRef] [PubMed]
  179. Flores-Maldonado, O.; Lezcano-Domínguez, C.I.; Dávila-Aviña, J.; González, G.M.; Ríos-López, A.L. Methyl gallate attenuates virulence and decreases antibiotic resistance in extensively drug-resistant Pseudomonas aeruginosa. Microb. Pathog. 2024, 194, 106830. [Google Scholar] [CrossRef]
  180. Bokhove, M.; Jimenez, P.N.; Quax, W.J.; Dijkstra, B.W. The quorum-quenching N-acyl homoserine lactone acylase PvdQ is an Ntn-hydrolase with an unusual substrate-binding pocket. Proc. Natl. Acad. Sci. USA 2010, 107, 686–691. [Google Scholar] [CrossRef] [PubMed]
  181. Utari, P.D.; Setroikromo, R.; Melgert, B.N.; Quax, W.J. PvdQ quorum quenching acylase attenuates Pseudomonas aeruginosa virulence in a mouse model of pulmonary infection. Front. Cell. Infect. Microbiol. 2018, 8, 119. [Google Scholar] [CrossRef]
  182. Sompiyachoke, K.; Elias, M.H. Engineering quorum quenching acylases with improved kinetic and biochemical properties. Protein Sci. 2024, 33, e4954. [Google Scholar] [CrossRef] [PubMed]
  183. Murugayah, S.A.; Gerth, M.L. Engineering quorum quenching enzymes: Progress and perspectives. Biochem. Soc. Trans. 2019, 47, 793–800. [Google Scholar] [CrossRef] [PubMed]
  184. Tan, S.Z.; Reisch, C.R.; Prather, K.L.J. A robust CRISPR interference gene repression system in Pseudomonas. J. Bacteriol. 2018, 200, e00575-17. [Google Scholar] [CrossRef]
  185. Yu, M.A.; Banta, A.B.; Ward, R.D.; Prasad, N.K.; Kwon, M.S.; Rosenberg, O.S.; Peters, J.M. Investigating Pseudomonas aeruginosa gene function during pathogenesis using mobile-CRISPRi. In Pseudomonas aeruginosa: Methods and Protocols; Springer: New York, NY, USA, 2023; pp. 13–32. [Google Scholar]
  186. Høyland-Kroghsbo, N.M.; Paczkowski, J.; Mukherjee, S.; Broniewski, J.; Westra, E.; Bondy-Denomy, J.; Bassler, B.L. Quorum sensing controls the Pseudomonas aeruginosa CRISPR-Cas adaptive immune system. Proc. Natl. Acad. Sci. USA 2017, 114, 131–135. [Google Scholar] [CrossRef]
  187. Dela Ahator, S.; Liu, Y.; Wang, J.; Zhang, L.-H. The virulence factor regulator and quorum sensing regulate the type IF CRISPR-Cas mediated horizontal gene transfer in Pseudomonas aeruginosa. Front. Microbiol. 2022, 13, 987656. [Google Scholar] [CrossRef] [PubMed]
  188. Andersen, J.B.; Hultqvist, L.D.; Jansen, C.U.; Jakobsen, T.H.; Nilsson, M.; Rybtke, M.; Uhd, J.; Fritz, B.G.; Seifert, R.; Berthelsen, J. Identification of small molecules that interfere with c-di-GMP signaling and induce dispersal of Pseudomonas aeruginosa biofilms. NPJ Biofilms Microbiomes 2021, 7, 59. [Google Scholar] [CrossRef]
  189. Hultqvist, L.D.; Andersen, J.B.; Nilsson, C.M.; Jansen, C.U.; Rybtke, M.; Jakobsen, T.H.; Nielsen, T.E.; Qvortrup, K.; Moser, C.; Graz, M. High efficacy treatment of murine Pseudomonas aeruginosa catheter-associated urinary tract infections using the c-di-GMP modulating anti-biofilm compound Disperazol in combination with ciprofloxacin. Antimicrob. Agents Chemother. 2024, 68, e01481-23. [Google Scholar] [CrossRef]
  190. Li, Y.; Heine, S.; Entian, M.; Sauer, K.; Frankenberg-Dinkel, N. NO-induced biofilm dispersion in Pseudomonas aeruginosa is mediated by an MHYT domain-coupled phosphodiesterase. J. Bacteriol. 2013, 195, 3531–3542. [Google Scholar] [CrossRef]
  191. Zhu, X.; Oh, H.-S.; Ng, Y.C.B.; Tang, P.Y.P.; Barraud, N.; Rice, S.A. Nitric oxide-mediated induction of dispersal in Pseudomonas aeruginosa biofilms is inhibited by flavohemoglobin production and is enhanced by imidazole. Antimicrob. Agents Chemother. 2018, 62, e01832-17. [Google Scholar] [CrossRef]
  192. Zhang, Y.; Cheng, P.; Wang, S.; Li, X.; Peng, L.; Fang, R.; Xiong, J.; Li, H.; Mei, C.; Gao, J. Pseudomonas aeruginosa biofilm dispersion by the mouse antimicrobial peptide CRAMP. Vet. Res. 2022, 53, 80. [Google Scholar] [CrossRef]
  193. Omran, B.A.; Tseng, B.S.; Baek, K.-H. Nanocomposites against Pseudomonas aeruginosa biofilms: Recent advances, challenges, and future prospects. Microbiol. Res. 2024, 282, 127656. [Google Scholar] [CrossRef]
  194. Guo, Y.; Mao, Z.; Ran, F.; Sun, J.; Zhang, J.; Chai, G.; Wang, J. Nanotechnology-based drug delivery systems to control bacterial-biofilm-associated lung infections. Pharmaceutics 2023, 15, 2582. [Google Scholar] [CrossRef]
  195. Rodrigues, A.S.; Batista, J.G.; Rodrigues, M.Á.; Thipe, V.C.; Minarini, L.A.; Lopes, P.S.; Lugão, A.B. Advances in silver nanoparticles: A comprehensive review on their potential as antimicrobial agents and their mechanisms of action elucidated by proteomics. Front. Microbiol. 2024, 15, 1440065. [Google Scholar] [CrossRef]
  196. Kamer, A.M.A.; El Maghraby, G.M.; Shafik, M.M.; Al-Madboly, L.A. Silver nanoparticle with potential antimicrobial and antibiofilm efficiency against multiple drug resistant, extensive drug resistant Pseudomonas aeruginosa clinical isolates. BMC Microbiol. 2024, 24, 277. [Google Scholar] [CrossRef] [PubMed]
  197. Haji, S.H.; Ganjo, A.R.; Faraj, T.A.; Fatah, M.H.; Smail, S.B. The enhanced antibacterial and antibiofilm properties of titanium dioxide nanoparticles biosynthesized by multidrug-resistant Pseudomonas aeruginosa. BMC Microbiol. 2024, 24, 379. [Google Scholar] [CrossRef] [PubMed]
  198. Al-Momani, H.; Aolymat, I.; Ibrahim, L.; Albalawi, H.; Al Balawi, D.; Albiss, B.A.; Almasri, M.; Alghweiri, S. Low-dose zinc oxide nanoparticles trigger the growth and biofilm formation of Pseudomonas aeruginosa: A hormetic response. BMC Microbiol. 2024, 24, 290. [Google Scholar] [CrossRef] [PubMed]
  199. Jalvo, B.; Faraldos, M.; Bahamonde, A.; Rosal, R. Antimicrobial and antibiofilm efficacy of self-cleaning surfaces functionalized by TiO2 photocatalytic nanoparticles against Staphylococcus aureus and Pseudomonas putida. J. Hazard. Mater. 2017, 340, 160–170. [Google Scholar] [CrossRef]
  200. Maric, T.; Løvind, A.; Zhang, Z.; Geng, J.; Boisen, A. Near-infrared light-driven mesoporous SiO2/Au nanomotors for eradication of Pseudomonas aeruginosa biofilm. Adv. Healthc. Mater. 2023, 12, 2203018. [Google Scholar] [CrossRef]
  201. 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]
  202. Colilla, M.; Vallet-Regí, M. Organically modified mesoporous silica nanoparticles against bacterial resistance. Chem. Mater. 2023, 35, 8788–8805. [Google Scholar] [CrossRef]
  203. Alhariri, M.; Omri, A. Efficacy of liposomal bismuth-ethanedithiol-loaded tobramycin after intratracheal administration in rats with pulmonary Pseudomonas aeruginosa infection. Antimicrob. Agents Chemother. 2013, 57, 569–578. [Google Scholar] [CrossRef]
  204. Bassetti, M.; Vena, A.; Russo, A.; Peghin, M. Inhaled liposomal antimicrobial delivery in lung infections. Drugs 2020, 80, 1309–1318. [Google Scholar] [CrossRef] [PubMed]
  205. Dallal Bashi, Y.H.; Mairs, R.; Murtadha, R.; Kett, V. Pulmonary delivery of antibiotics to the lungs: Current state and future prospects. Pharmaceutics 2025, 17, 111. [Google Scholar] [CrossRef] [PubMed]
  206. 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]
  207. Afrasiabi, S.; Partoazar, A. Targeting bacterial biofilm-related genes with nanoparticle-based strategies. Front. Microbiol. 2024, 15, 1387114. [Google Scholar] [CrossRef]
  208. Panthi, V.K.; Fairfull-Smith, K.E.; Islam, N. Liposomal drug delivery strategies to eradicate bacterial biofilms: Challenges, recent advances, and future perspectives. Int. J. Pharm. 2024, 655, 124046. [Google Scholar] [CrossRef]
  209. Topka-Bielecka, G.; Dydecka, A.; Necel, A.; Bloch, S.; Nejman-Faleńczyk, B.; Węgrzyn, G.; Węgrzyn, A. Bacteriophage-derived depolymerases against bacterial biofilm. Antibiotics 2021, 10, 175. [Google Scholar] [CrossRef]
  210. Wang, H.; Liu, Y.; Bai, C.; Leung, S.S.Y. Translating bacteriophage-derived depolymerases into antibacterial therapeutics: Challenges and prospects. Acta Pharm. Sin. B 2024, 14, 155–169. [Google Scholar] [CrossRef]
  211. Sisson, H.M.; Jackson, S.A.; Fagerlund, R.D.; Warring, S.L.; Fineran, P.C. Gram-negative endolysins: Overcoming the outer membrane obstacle. Curr. Opin. Microbiol. 2024, 78, 102433. [Google Scholar] [CrossRef]
  212. Wang, T.; Zheng, Y.; Dai, J.; Zhou, J.; Yu, R.; Zhang, C. Design SMAP29-LysPA26 as a highly efficient artilysin against Pseudomonas aeruginosa with bactericidal and antibiofilm activity. Microbiol. Spectr. 2021, 9, e00546-21. [Google Scholar] [CrossRef]
  213. De Soir, S.; Parée, H.; Kamarudin, N.H.N.; Wagemans, J.; Lavigne, R.; Braem, A.; Merabishvili, M.; De Vos, D.; Pirnay, J.-P.; Van Bambeke, F. Exploiting phage-antibiotic synergies to disrupt Pseudomonas aeruginosa PAO1 biofilms in the context of orthopedic infections. Microbiol. Spectr. 2024, 12, e03219-23. [Google Scholar] [CrossRef]
  214. Manohar, P.; Loh, B.; Nachimuthu, R.; Leptihn, S. Phage-antibiotic combinations to control Pseudomonas aeruginosa–Candida two-species biofilms. Sci. Rep. 2024, 14, 9354. [Google Scholar] [CrossRef]
  215. Lin, L.-C.; Tsai, Y.-C.; Lin, N.-T. Phage–Antibiotic Synergy Enhances Biofilm Eradication and Survival in a Zebrafish Model of Pseudomonas aeruginosa Infection. Int. J. Mol. Sci. 2025, 26, 5337. [Google Scholar] [CrossRef] [PubMed]
  216. Kunisch, F.; Campobasso, C.; Wagemans, J.; Yildirim, S.; Chan, B.K.; Schaudinn, C.; Lavigne, R.; Turner, P.E.; Raschke, M.J.; Trampuz, A. Targeting Pseudomonas aeruginosa biofilm with an evolutionary trained bacteriophage cocktail exploiting phage resistance trade-offs. Nat. Commun. 2024, 15, 8572. [Google Scholar] [CrossRef] [PubMed]
  217. Yu, S.; Su, T.; Wu, H.; Liu, S.; Wang, D.; Zhao, T.; Jin, Z.; Du, W.; Zhu, M.-J.; Chua, S.L. PslG, a self-produced glycosyl hydrolase, triggers biofilm disassembly by disrupting exopolysaccharide matrix. Cell Res. 2015, 25, 1352–1367. [Google Scholar] [CrossRef]
  218. Baker, P.; Whitfield, G.B.; Hill, P.J.; Little, D.J.; Pestrak, M.J.; Robinson, H.; Wozniak, D.J.; Howell, P.L. Characterization of the Pseudomonas aeruginosa glycoside hydrolase PslG reveals that its levels are critical for Psl polysaccharide biosynthesis and biofilm formation. J. Biol. Chem. 2015, 290, 28374–28387. [Google Scholar] [CrossRef] [PubMed]
  219. Su, T.; He, J.; Li, N.; Liu, S.; Xu, S.; Gu, L. A rational designed PslG with normal biofilm hydrolysis and enhanced resistance to trypsin-like protease digestion. Front. Microbiol. 2020, 11, 760. [Google Scholar] [CrossRef]
  220. Chegini, Z.; Khoshbayan, A.; Taati Moghadam, M.; Farahani, I.; Jazireian, P.; Shariati, A. Bacteriophage therapy against Pseudomonas aeruginosa biofilms: A review. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 45. [Google Scholar] [CrossRef]
  221. Ali, S.O.; Yu, X.Q.; Robbie, G.J.; Wu, Y.; Shoemaker, K.; Yu, L.; DiGiandomenico, A.; Keller, A.E.; Anude, C.; Hernandez-Illas, M. Phase 1 study of MEDI3902, an investigational anti–Pseudomonas aeruginosa PcrV and Psl bispecific human monoclonal antibody, in healthy adults. Clin. Microbiol. Infect. 2019, 25, 629.e1–629.e6. [Google Scholar] [CrossRef]
  222. Chastre, J.; François, B.; Bourgeois, M.; Komnos, A.; Ferrer, R.; Rahav, G.; De Schryver, N.; Lepape, A.; Koksal, I.; Luyt, C.-E. Safety, efficacy, and pharmacokinetics of gremubamab (MEDI3902), an anti-Pseudomonas aeruginosa bispecific human monoclonal antibody, in P. aeruginosa-colonised, mechanically ventilated intensive care unit patients: A randomised controlled trial. Crit. Care 2022, 26, 355. [Google Scholar] [CrossRef] [PubMed]
  223. Long, M.B.; Gilmour, A.; Kehl, M.; Tabor, D.E.; Keller, A.E.; Warrener, P.; Gopalakrishnan, V.; Rosengren, S.; Crichton, M.L.; McIntosh, E. A bispecific monoclonal antibody targeting Psl and PcrV enhances neutrophil-mediated killing of Pseudomonas aeruginosa in patients with bronchiectasis. Am. J. Respir. Crit. Care Med. 2024, 210, 35–46. [Google Scholar] [CrossRef]
  224. Killough, M.; Rodgers, A.M.; Ingram, R.J. Pseudomonas aeruginosa: Recent advances in vaccine development. Vaccines 2022, 10, 1100. [Google Scholar] [CrossRef]
  225. Santamarina-Fernández, R.; Fuentes-Valverde, V.; Silva-Rodríguez, A.; García, P.; Moscoso, M.; Bou, G. Pseudomonas aeruginosa Vaccine Development: Lessons, Challenges, and Future Innovations. Int. J. Mol. Sci. 2025, 26, 2012. [Google Scholar] [CrossRef]
  226. Azimi, S.; Zanjani, L.S. Immunization against Pseudomonas aeruginosa using Alg-PLGA nano-vaccine. Iran. J. Basic Med. Sci. 2021, 24, 476. [Google Scholar]
  227. Hossain, M.A.; Lee, S.-J.; Park, N.-H.; Mechesso, A.F.; Birhanu, B.T.; Kang, J.; Reza, M.A.; Suh, J.-W.; Park, S.-C. Impact of phenolic compounds in the acyl homoserine lactone-mediated quorum sensing regulatory pathways. Sci. Rep. 2017, 7, 10618. [Google Scholar] [CrossRef]
  228. Sio, C.F.; Otten, L.G.; Cool, R.H.; Diggle, S.P.; Braun, P.G.; Bos, R.; Daykin, M.; Cámara, M.; Williams, P.; Quax, W.J. Quorum quenching by an N-acyl-homoserine lactone acylase from Pseudomonas aeruginosa PAO1. Infect. Immun. 2006, 74, 1673–1682. [Google Scholar] [CrossRef]
  229. Cho, S.W.; Kim, S.; Kim, J.M.; Kim, J.-S. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat. Biotechnol. 2013, 31, 230–232. [Google Scholar] [CrossRef]
  230. Barraud, N.; Hassett, D.J.; Hwang, S.-H.; Rice, S.A.; Kjelleberg, S.; Webb, J.S. Involvement of nitric oxide in biofilm dispersal of Pseudomonas aeruginosa. J. Bacteriol. 2006, 188, 7344–7353. [Google Scholar] [CrossRef] [PubMed]
  231. Dakal, T.C.; Kumar, A.; Majumdar, R.S.; Yadav, V. Mechanistic basis of antimicrobial actions of silver nanoparticles. Front. Microbiol. 2016, 7, 1831. [Google Scholar] [CrossRef] [PubMed]
  232. Raghunath, A.; Perumal, E. Metal oxide nanoparticles as antimicrobial agents: A promise for the future. Int. J. Antimicrob. Agents 2017, 49, 137–152. [Google Scholar] [CrossRef]
  233. Guo, Z.; Liu, M.; Zhang, D. Potential of phage depolymerase for the treatment of bacterial biofilms. Virulence 2023, 14, 2273567. [Google Scholar] [CrossRef]
  234. Manohar, P.; Loh, B.; Turner, D.; Tamizhselvi, R.; Mathankumar, M.; Elangovan, N.; Nachimuthu, R.; Leptihn, S. In vitro and in vivo evaluation of the biofilm-degrading Pseudomonas phage Motto, as a candidate for phage therapy. Front. Microbiol. 2024, 15, 1344962. [Google Scholar] [CrossRef]
  235. Urgeya, K.D.; Subedi, D.; Kumar, N.; Willcox, M. The Ability of Bacteriophages to Reduce Biofilms Produced by Pseudomonas aeruginosa Isolated from Corneal Infections. Antibiotics 2025, 14, 629. [Google Scholar] [CrossRef] [PubMed]
  236. Tabor, D.; Oganesyan, V.; Keller, A.; Yu, L.; McLaughlin, R.; Song, E.; Warrener, P.; Rosenthal, K.; Esser, M.; Qi, Y. Pseudomonas aeruginosa PcrV and Psl, the molecular targets of bispecific antibody MEDI3902, are conserved among diverse global clinical isolates. J. Infect. Dis. 2018, 218, 1983–1994. [Google Scholar] [CrossRef]
  237. Reichhardt, C.; Parsek, M.R. Confocal laser scanning microscopy for analysis of Pseudomonas aeruginosa biofilm architecture and matrix localization. Front. Microbiol. 2019, 10, 677. [Google Scholar] [CrossRef]
  238. Mhade, S.; Kaushik, K.S. Tools of the trade: Image analysis programs for confocal laser-scanning microscopy studies of biofilms and considerations for their use by experimental researchers. ACS Omega 2023, 8, 20163–20177. [Google Scholar] [CrossRef]
  239. Mountcastle, S.E.; Vyas, N.; Villapun, V.M.; Cox, S.C.; Jabbari, S.; Sammons, R.L.; Shelton, R.M.; Walmsley, A.D.; Kuehne, S.A. Biofilm viability checker: An open-source tool for automated biofilm viability analysis from confocal microscopy images. npj Biofilms Microbiomes 2021, 7, 44. [Google Scholar] [CrossRef] [PubMed]
  240. Demidov, V.V.; Jackson, O.P.; Demidova, N.; Gunn, J.R.; Gitajn, I.L.; Elliott, J.T. Integrating optical coherence tomography and bioluminescence with predictive modeling for quantitative assessment of methicillin-resistant S. aureus biofilms. J. Biomed. Opt. 2025, 30, S34111. [Google Scholar] [CrossRef] [PubMed]
  241. Devine, B.C.; Dogan, A.B.; Sobol, W.M. Recent Optical Coherence Tomography (OCT) Innovations for Increased Accessibility and Remote Surveillance. Bioengineering 2025, 12, 441. [Google Scholar] [CrossRef]
  242. Heise, B.; Zorin, I.; Duswald, K.; Karl, V.; Brouczek, D.; Eichelseder, J.; Schwentenwein, M. Mid-infrared optical coherence tomography and machine learning for inspection of 3D-printed ceramics at the micron scale. Front. Mater. 2024, 11, 1441812. [Google Scholar] [CrossRef]
  243. Xi, C.; Marks, D.; Schlachter, S.; Luo, W.; Boppart, S.A. High-resolution three-dimensional imaging of biofilm development using optical coherence tomography. J. Biomed. Opt. 2006, 11, 034001. [Google Scholar]
  244. Hou, J.; Wang, C.; Rozenbaum, R.T.; Gusnaniar, N.; de Jong, E.D.; Woudstra, W.; Geertsema-Doornbusch, G.I.; Atema-Smit, J.; Sjollema, J.; Ren, Y. Bacterial density and biofilm structure determined by optical coherence tomography. Sci. Rep. 2019, 9, 9794. [Google Scholar] [CrossRef] [PubMed]
  245. Haval, M.; Unakal, C.; Ghagane, S.C.; Pandit, B.R.; Daniel, E.; Siewdass, P.; Ekimeri, K.; Rajamanickam, V.; Justiz-Vaillant, A.; Lootawan, K.-A.A. Biofilms Exposed: Innovative Imaging and Therapeutic Platforms for Persistent Infections. Antibiotics 2025, 14, 865. [Google Scholar] [CrossRef] [PubMed]
  246. Polisetti, S.; Baig, N.F.; Morales-Soto, N.; Shrout, J.D.; Bohn, P.W. Spatial mapping of pyocyanin in Pseudomonas aeruginosa bacterial communities using surface enhanced Raman scattering. Appl. Spectrosc. 2017, 71, 215–223. [Google Scholar] [CrossRef]
  247. Bodelon, G.; Montes-García, V.; Perez-Juste, J.; Pastoriza-Santos, I. Surface-enhanced Raman scattering spectroscopy for label-free analysis of P. aeruginosa quorum sensing. Front. Cell. Infect. Microbiol. 2018, 8, 143. [Google Scholar]
  248. Jia, J. Spatiotemporal Mapping of Quinolone and Phenazine Secretion in Pseudomonas aeruginosa Biofilms by Confocal Raman Imaging. Ph.D. Thesis, University of Notre Dame, Notre Dame, IN, USA, 2021. [Google Scholar]
  249. Kotowska, A.M.; Zhang, J.; Carabelli, A.; Watts, J.; Aylott, J.W.; Gilmore, I.S.; Williams, P.; Scurr, D.J.; Alexander, M.R. Toward comprehensive analysis of the 3D chemistry of Pseudomonas aeruginosa biofilms. Anal. Chem. 2023, 95, 18287–18294. [Google Scholar] [CrossRef]
  250. Rodriguez, L.; Zhang, Z.; Wang, D. Recent advances of Raman spectroscopy for the analysis of bacteria. Anal. Sci. Adv. 2023, 4, 81–95. [Google Scholar] [CrossRef]
  251. Cao, T.; Weaver, A.A.; Baek, S.; Jia, J.; Shrout, J.D.; Bohn, P.W. Depth distributions of signaling molecules in Pseudomonas aeruginosa biofilms mapped by confocal Raman microscopy. J. Chem. Phys. 2021, 154, 204201. [Google Scholar] [CrossRef]
  252. Do, H.; Kwon, S.-R.; Fu, K.; Morales-Soto, N.; Shrout, J.D.; Bohn, P.W. Electrochemical surface-enhanced Raman spectroscopy of pyocyanin secreted by Pseudomonas aeruginosa communities. Langmuir 2019, 35, 7043–7049. [Google Scholar] [CrossRef]
  253. Masyuko, R.N.; Lanni, E.J.; Driscoll, C.M.; Shrout, J.D.; Sweedler, J.V.; Bohn, P.W. Spatial organization of Pseudomonas aeruginosa biofilms probed by combined matrix-assisted laser desorption ionization mass spectrometry and confocal Raman microscopy. Analyst 2014, 139, 5700–5708. [Google Scholar] [CrossRef]
  254. Lanni, E.J.; Masyuko, R.N.; Driscoll, C.M.; Aerts, J.T.; Shrout, J.D.; Bohn, P.W.; Sweedler, J.V. MALDI-guided SIMS: Multiscale imaging of metabolites in bacterial biofilms. Anal. Chem. 2014, 86, 9139–9145. [Google Scholar] [CrossRef]
  255. Shen, Y.; Wang, Y.; Wang, J.; Xie, P.; Xie, C.; Chen, Y.; Banaei, N.; Ren, K.; Cai, Z. High-resolution 3D spatial distribution of complex microbial colonies revealed by mass spectrometry imaging. J. Adv. Res. 2024, in press. [Google Scholar] [CrossRef]
  256. Pleguezuelos-Manzano, C.; Beenker, W.A.; van Son, G.J.; Begthel, H.; Amatngalim, G.D.; Beekman, J.M.; Clevers, H.; den Hertog, J. Dual RNA sequencing of a co-culture model of Pseudomonas aeruginosa and human 2D upper airway organoids. Sci. Rep. 2025, 15, 2222. [Google Scholar] [CrossRef] [PubMed]
  257. Pleguezuelos-Manzano, C.; Beenker, W.A.; van Son, G.J.; Begthel, H.; Amatngalim, G.D.; Beekman, J.M.; Clevers, H.; den Hertog, J. Establishment and characterization of a new Pseudomonas aeruginosa infection model using 2D airway organoids and dual RNA sequencing. bioRxiv 2023. [Google Scholar] [CrossRef]
  258. Asensio, N.C.; Rendón, J.M.; López, J.J.G.; Tizzano, S.G.; Gómez, M.T.M.; Burgas, M.T. Time-resolved dual transcriptomics of Pseudomonas aeruginosa biofilm formation in cystic fibrosis. Biofilm 2025, 10, 100301. [Google Scholar] [CrossRef]
  259. Hengge, R. High-specificity local and global c-di-GMP signaling. Trends Microbiol. 2021, 29, 993–1003. [Google Scholar] [CrossRef]
  260. Katharios-Lanwermeyer, S.; Whitfield, G.B.; Howell, P.L.; O’Toole, G.A. Pseudomonas aeruginosa uses c-di-GMP phosphodiesterases RmcA and MorA to regulate biofilm maintenance. mBio 2021, 12, e03384-20. [Google Scholar] [CrossRef] [PubMed]
  261. Martino, R.A.; Volke, D.C.; Tenaglia, A.H.; Tribelli, P.M.; Nikel, P.I.; Smania, A.M. Genetic Dissection of Cyclic di-GMP Signalling in Pseudomonas aeruginosa via Systematic Diguanylate Cyclase Disruption. Microb. Biotechnol. 2025, 18, e70137. [Google Scholar] [CrossRef]
  262. Noirot-Gros, M.-F.; Forrester, S.; Malato, G.; Larsen, P.E.; Noirot, P. CRISPR interference to interrogate genes that control biofilm formation in Pseudomonas fluorescens. Sci. Rep. 2019, 9, 15954. [Google Scholar] [CrossRef]
  263. Vasenina, A.; Fu, Y.; O’Toole, G.A.; Mucha, P.J. Local control: A hub-based model for the c-di-GMP network. Msphere 2024, 9, e00178-24. [Google Scholar] [CrossRef] [PubMed]
  264. Junkermeier, E.H.; Hengge, R. Local signaling enhances output specificity of bacterial c-di-GMP signaling networks. Microlife 2023, 4, uqad026. [Google Scholar] [CrossRef]
  265. van der Ploeg, K.; de Vogel, C.P.; Klaassen, C.; Luider, T.; Zeneyedpour, L.; Mason-Slingerland, B.C.; Vos, M.C.; Bruno, M.; Bexkens, M.L.; Severin, J.A. Proteomic identification of Pa2146 as a biofilm marker of Pseudomonas aeruginosa on endoscope channel material. SSRN Electron. J. 2025, 10, 5288165. [Google Scholar] [CrossRef]
  266. Kuik, C.; van Hoogstraten, S.W.; Arts, J.J.; Honing, M.; Cillero-Pastor, B. Matrix-assisted laser desorption/ionization mass spectrometry imaging for quorum sensing. AMB Express 2024, 14, 45. [Google Scholar] [CrossRef]
  267. Ngai, Y.T.; Lau, D.; Mittal, P.; Hoffmann, P. Mini review: Highlight of recent advances and applications of MALDI mass spectrometry imaging in 2024. Anal. Sci. Adv. 2025, 6, e70016. [Google Scholar] [CrossRef]
  268. Ameer, S.; Ibrahim, H.; Yaseen, M.U.; Kulsoom, F.; Cinti, S.; Sher, M. Electrochemical impedance spectroscopy-based sensing of biofilms: A comprehensive review. Biosensors 2023, 13, 777. [Google Scholar] [CrossRef] [PubMed]
  269. Haghighian, N.; Kataky, R. Advances in electrochemical detection of bacterial biofilm metabolites. Curr. Opin. Electrochem. 2024, 46, 101486. [Google Scholar] [CrossRef]
  270. Fisher, O.J.; Wang, Y.; Ahmed, A. Making waves: Transforming biofilm-based wastewater treatment using machine learning-driven real-time monitoring. Water Res. 2025, 287, 124491. [Google Scholar] [CrossRef]
  271. Abouhagger, A.; Celiešiūtė-Germanienė, R.; Bakute, N.; Stirke, A.; Melo, W.C. Electrochemical biosensors on microfluidic chips as promising tools to study microbial biofilms: A review. Front. Cell. Infect. Microbiol. 2024, 14, 1419570. [Google Scholar] [CrossRef]
  272. Blanco-Cabra, N.; López-Martínez, M.J.; Arévalo-Jaimes, B.V.; Martin-Gómez, M.T.; Samitier, J.; Torrents, E. A new BiofilmChip device for testing biofilm formation and antibiotic susceptibility. npj Biofilms Microbiomes 2021, 7, 62. [Google Scholar] [CrossRef] [PubMed]
  273. Plebani, R.; Potla, R.; Soong, M.; Bai, H.; Izadifar, Z.; Jiang, A.; Travis, R.N.; Belgur, C.; Dinis, A.; Cartwright, M.J. Modeling pulmonary cystic fibrosis in a human lung airway-on-a-chip. J. Cyst. Fibros. 2022, 21, 606–615. [Google Scholar] [CrossRef]
  274. Weaver, A.A.; Shrout, J.D. Use of analytical strategies to understand spatial chemical variation in bacterial surface communities. J. Bacteriol. 2025, 207, e00402-24. [Google Scholar] [CrossRef]
  275. Jain, A.; Dabburu, G.R.; Samanta, B.; Singhal, N.; Kumar, M. An explainable machine learning pipeline for prediction of antimicrobial resistance in Pseudomonas aeruginosa. Bioinform. Adv. 2025, 5, vbaf190. [Google Scholar] [CrossRef]
  276. Vergauwe, F.; De Waele, G.; Sass, A.; Highmore, C.; Hanrahan, N.; Cook, Y.; Lichtenberg, M.; Cnockaert, M.; Vandamme, P.; Mahajan, S. Harnessing machine learning to predict antibiotic susceptibility in Pseudomonas aeruginosa biofilms. bioRxiv 2025, 11, 205. [Google Scholar] [CrossRef] [PubMed]
  277. Bilal, H.; Khan, M.N.; Khan, S.; Shafiq, M.; Fang, W.; Khan, R.U.; Rahman, M.U.; Li, X.; Lv, Q.-L.; Xu, B. The role of artificial intelligence and machine learning in predicting and combating antimicrobial resistance. Comput. Struct. Biotechnol. J. 2025, 27, 423–439. [Google Scholar] [CrossRef]
  278. Poh, W.H.; Rice, S.A. Recent developments in nitric oxide donors and delivery for antimicrobial and anti-biofilm applications. Molecules 2022, 27, 674. [Google Scholar] [CrossRef] [PubMed]
  279. ISO 10993-1:2018; Biological Evaluation of Medical Devices—Part 1: Evaluation and Testing Within a Risk Management Process. International Organization for Standardization (ISO): Geneva, Switzerland, 2018. Available online: https://www.iso.org/standard/68936.html (accessed on 23 October 2025).
  280. ISO 10993-5:2009; Biological Evaluation of Medical Devices—Part 5: Tests for In Vitro Cytotoxicity. International Organization for Standardization (ISO): Geneva, Switzerland, 2009. Available online: https://www.x-cellr8.com/wp-content/uploads/2020/07/CT-060_01-07_20-ISO-Cytotoxicity.pdf (accessed on 23 October 2025).
  281. FDA. Use of International Standard ISO 10993-1, “Biological Evaluation of Medical Devices—Part 1: Evaluation and Testing Within a Risk Management Process”: Guidance for Industry and Food and Drug Administration Staff; U.S. Food and Drug Administration (FDA): Silver Spring, MD, USA, 2020. Available online: https://www.fda.gov/media/142959/download (accessed on 23 October 2025).
  282. ISO/TR 10993-22:2017; Biological Evaluation of Medical Devices—Part 22: Guidance on Nanomaterials. International Organization for Standardization (ISO): Geneva, Switzerland, 2017. Available online: https://www.iso.org/standard/65918.html (accessed on 23 October 2025).
  283. Gruber, S.; Nickel, A. Toxic or not toxic? The specifications of the standard ISO 10993-5 are not explicit enough to yield comparable results in the cytotoxicity assessment of an identical medical device. Front. Med. Technol. 2023, 5, 1195529. [Google Scholar] [CrossRef]
  284. ISO/TR 10993-55:2023; Biocompatibility Assessment of Medical Devices. eFOR Group: Paris, France, 2023. Available online: https://efor-group.com/en/biocompatibility-assessment-of-md-iso-tr-10993-552023/ (accessed on 24 October 2025).
  285. Kanďárová, H.; Pôbiš, P. The “Big Three” in biocompatibility testing of medical devices: Implementation of alternatives to animal experimentation—Are we there yet? Front. Toxicol. 2024, 5, 1337468. [Google Scholar] [CrossRef]
  286. ISO 10993-18:2020; Biological Evaluation of Medical Devices—Part 18: Chemical Characterization of Medical Device Materials Within a Risk Management Process. International Organization for Standardization (ISO): Geneva, Switzerland, 2020. Available online: https://www.iso.org/standard/64750.html (accessed on 24 October 2025).
  287. Kalia, V.C.; Wood, T.K.; Kumar, P. Evolution of resistance to quorum-sensing inhibitors. Microb. Ecol. 2014, 68, 13–23. [Google Scholar] [CrossRef]
  288. Maiga, A.; Ampomah-Wireko, M.; Li, H.; Fan, Z.; Lin, Z.; Zhen, H.; Kpekura, S.; Wu, C. Multidrug-resistant bacteria quorum-sensing inhibitors: A particular focus on Pseudomonas aeruginosa. Eur. J. Med. Chem. 2025, 281, 117008. [Google Scholar] [CrossRef]
  289. Hetta, H.F.; Ramadan, Y.N.; Rashed, Z.I.; Alharbi, A.A.; Alsharef, S.; Alkindy, T.T.; Alkhamali, A.; Albalawi, A.S.; Battah, B.; Donadu, M.G. Quorum sensing inhibitors: An alternative strategy to win the battle against multidrug-resistant (MDR) bacteria. Molecules 2024, 29, 3466. [Google Scholar] [CrossRef] [PubMed]
  290. Kim, M.K.; Chen, Q.; Echterhof, A.; Pennetzdorfer, N.; McBride, R.C.; Banaei, N.; Burgener, E.B.; Milla, C.E.; Bollyky, P.L. A blueprint for broadly effective bacteriophage-antibiotic cocktails against bacterial infections. Nat. Commun. 2024, 15, 9987. [Google Scholar] [CrossRef]
  291. Cai, Y.-M.; Zhang, Y.-D.; Yang, L. NO donors and NO delivery methods for controlling biofilms in chronic lung infections. Appl. Microbiol. Biotechnol. 2021, 105, 3931–3954. [Google Scholar] [CrossRef]
  292. ClinicalTrials.gov. Inhaled Sodium Nitrite As an Antimicrobial for Cystic Fibrosis (Identifier: NCT02694393); U.S. National Library of Medicine: Bethesda, MD, USA, 2024. Available online: https://clinicaltrials.gov/study/NCT02694393 (accessed on 24 October 2025).
  293. Debast, S.; van den Bos-Kromhout, M.; de Vries-van Rossum, S.; Abma-Blatter, S.; Notermans, D.; Kluytmans, J.; Immeker, B.; Zuur, J.; Hijmering, M.; Bergwerff, A. Aquatic reservoir-associated outbreaks of multidrug-resistant bacteria: A hospital outbreak report of Pseudomonas aeruginosa in perspective from the Dutch national surveillance databases. J. Hosp. Infect. 2025, 162, 310–318. [Google Scholar] [CrossRef]
  294. Querin, B.; Danjean, M.; Jolivet, S.; Couturier, J.; Oubbéa, S.; Jouans, C.; Lazare, C.; Montagne, T.; Chamming’s, A.; Luce, S. Protracted outbreaks of VIM-producing Pseudomonas aeruginosa in a surgical intensive care unit in France, January 2018 to June 2024. Antimicrob. Resist. Infect. Control 2025, 14, 95. [Google Scholar] [CrossRef]
  295. Rath, A.; Kieninger, B.; Fritsch, J.; Caplunik-Pratsch, A.; Blaas, S.; Ochmann, M.; Pfeifer, M.; Hartl, J.; Holzmann, T.; Schneider-Brachert, W. Whole-genome sequencing reveals two prolonged simultaneous outbreaks involving Pseudomonas aeruginosa high-risk strains ST111 and ST235 with resistance to quaternary ammonium compounds. J. Hosp. Infect. 2024, 145, 155–164. [Google Scholar] [CrossRef] [PubMed]
  296. Hayward, C.; Ross, K.; Brown, M.; Bentham, R.; Hinds, J.; Whiley, H. Drinking water plumbing systems are a hot spot for antimicrobial-resistant pathogens. J. Hosp. Infect. 2025, 159, 62–70. [Google Scholar] [CrossRef]
  297. Nguyen, N.T.; Kurisu, F.; Furumai, H.; Kasuga, I. Antimicrobial Resistance profiles of Bacterial Community in Premise Plumbing before and after Water Stagnation. J. Water Environ. Technol. 2023, 21, 19–29. [Google Scholar] [CrossRef]
  298. Malcom, H.B.; Bowes, D.A. Use of Wastewater to Monitor Antimicrobial Resistance Trends in Communities and Implications for Wastewater-Based Epidemiology: A Review of the Recent Literature. Microorganisms 2025, 13, 2073. [Google Scholar] [CrossRef]
  299. Clarke, L.M.; O’Brien, J.W.; Murray, A.K.; Gaze, W.H.; Thomas, K.V. A review of wastewater-based epidemiology for antimicrobial resistance surveillance. J. Environ. Expo. 2024, 3, 7. [Google Scholar] [CrossRef]
  300. WHO. Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report 2024; World Health Organization: Geneva, Switzerland, 2024. Available online: https://www.who.int/initiatives/glass (accessed on 24 October 2025).
  301. WHO. Global Antibiotic Resistance Surveillance Report 2025; World Health Organization: Geneva, Switzerland, 2025. Available online: https://www.who.int/publications/i/item/9789240116337 (accessed on 24 October 2025).
  302. WHO. Sharp Global Rise in Antibiotic-Resistant Infections in Hospitals, WHO Finds; The Guardian: London, UK, 2025. Available online: https://www.theguardian.com/world/2025/oct/13/sharp-global-rise-in-antibiotic-resistant-infections-in-hospitals-who-finds? (accessed on 24 October 2025).
Figure 1. The structural code of P. aeruginosa biofilms. A conceptual overview that summarizes how biofilms form, strengthen, and disperse. The panel on the left presents the staged progression from reversible attachment to maturation and active dispersal. These stages are guided by genetic checkpoints that include sadB, pel, psl, algD, and bdlA. The central panel illustrates EPS architecture, which contains Psl that mediates cell-to-cell and cell-to-surface adhesion, Pel that contributes to cohesion and tolerance, alginate that forms the mucoid matrix, CdrA that crosslinks with Psl, and eDNA that provides mechanical reinforcement. The panel on the right shows how oxygen and nutrient gradients create stratified physiology. These gradients support phenazine-linked redox activity and favor the development of drug-tolerant persister and VBNC subpopulations.
Figure 1. The structural code of P. aeruginosa biofilms. A conceptual overview that summarizes how biofilms form, strengthen, and disperse. The panel on the left presents the staged progression from reversible attachment to maturation and active dispersal. These stages are guided by genetic checkpoints that include sadB, pel, psl, algD, and bdlA. The central panel illustrates EPS architecture, which contains Psl that mediates cell-to-cell and cell-to-surface adhesion, Pel that contributes to cohesion and tolerance, alginate that forms the mucoid matrix, CdrA that crosslinks with Psl, and eDNA that provides mechanical reinforcement. The panel on the right shows how oxygen and nutrient gradients create stratified physiology. These gradients support phenazine-linked redox activity and favor the development of drug-tolerant persister and VBNC subpopulations.
Microorganisms 14 00109 g001
Figure 2. c-di-GMP network dynamics in P. aeruginosa. Environmental and host-related signals that include nitric oxide, temperature changes, and hypoxia or redox-linked cues sensed by PAS domain regulators converge on diguanylate cyclases that include WspR, sadC, and RoeA, and on phosphodiesterases such as BifA and RbdA. These enzymes set intracellular c-di-GMP levels and shape biofilm behavior. High c-di-GMP promotes matrix assembly. PelD activates Pel polysaccharide synthesis, and Alg44 promotes alginate polymerization. Low c-di-GMP, produced through PDE activity or nitric oxide-mediated reduction, favors motility and detachment. The schematic reads from left to right and shows inputs, enzymes, the c-di-GMP pool, and downstream effectors or phenotypes. The figure also highlights therapeutic intervention points at DGCs, PDEs, and c-di-GMP-dependent effectors. Abbreviations: DGC, diguanylate cyclase; PDE, phosphodiesterase; EPS, extracellular polymeric substance.
Figure 2. c-di-GMP network dynamics in P. aeruginosa. Environmental and host-related signals that include nitric oxide, temperature changes, and hypoxia or redox-linked cues sensed by PAS domain regulators converge on diguanylate cyclases that include WspR, sadC, and RoeA, and on phosphodiesterases such as BifA and RbdA. These enzymes set intracellular c-di-GMP levels and shape biofilm behavior. High c-di-GMP promotes matrix assembly. PelD activates Pel polysaccharide synthesis, and Alg44 promotes alginate polymerization. Low c-di-GMP, produced through PDE activity or nitric oxide-mediated reduction, favors motility and detachment. The schematic reads from left to right and shows inputs, enzymes, the c-di-GMP pool, and downstream effectors or phenotypes. The figure also highlights therapeutic intervention points at DGCs, PDEs, and c-di-GMP-dependent effectors. Abbreviations: DGC, diguanylate cyclase; PDE, phosphodiesterase; EPS, extracellular polymeric substance.
Microorganisms 14 00109 g002
Figure 3. Quorum-sensing and c-di-GMP networks reinforce antimicrobial tolerance in P. aeruginosa biofilms. QS hierarchies that include Las, Rhl, and Pqs, together with the second messenger c-di-GMP, function as coordinated regulatory hubs that strengthen tolerance in biofilm communities. Elevated c-di-GMP acts as a master lifestyle regulator and promotes EPS synthesis through Psl, Pel, and alginate, and increases eDNA accumulation, which restricts antibiotic penetration and buffers cationic agents. In parallel, QS and c-di-GMP enhance the expression of efflux regulators brlR and sagS, and RND pumps mexAB-oprM and mexEF-oprN, which improve intracellular detoxification. These combined interactions favor the formation of persister and VBNC populations and help maintain horizontal gene transfer and plasmid stability within the matrix. Together, this layered regulatory architecture produces a biofilm tolerance phenotype in which MBIC and MBEC values rise well above conventional MIC thresholds.
Figure 3. Quorum-sensing and c-di-GMP networks reinforce antimicrobial tolerance in P. aeruginosa biofilms. QS hierarchies that include Las, Rhl, and Pqs, together with the second messenger c-di-GMP, function as coordinated regulatory hubs that strengthen tolerance in biofilm communities. Elevated c-di-GMP acts as a master lifestyle regulator and promotes EPS synthesis through Psl, Pel, and alginate, and increases eDNA accumulation, which restricts antibiotic penetration and buffers cationic agents. In parallel, QS and c-di-GMP enhance the expression of efflux regulators brlR and sagS, and RND pumps mexAB-oprM and mexEF-oprN, which improve intracellular detoxification. These combined interactions favor the formation of persister and VBNC populations and help maintain horizontal gene transfer and plasmid stability within the matrix. Together, this layered regulatory architecture produces a biofilm tolerance phenotype in which MBIC and MBEC values rise well above conventional MIC thresholds.
Microorganisms 14 00109 g003
Figure 4. Mechanistic overview of network disruption strategies targeting the P. aeruginosa biofilm code. Four complementary therapeutic modules converge to dismantle biofilms and resensitize bacteria to antibiotics. Quorum-sensing inhibition uses small-molecule antagonists, signal-degrading enzymes, and CRISPR interference that targets lasI, rhlI, and pqsR to mute virulence and community signaling. Furthermore, c-di-GMP modulators, including PDE activators, DGC inhibitors, and nitric oxide donors, lower intracellular c-di-GMP and promote dispersal with a shift toward motile states. Matrix disruption relies on nanoparticles, depolymerases such as PslG, and mesoporous silica carriers to degrade EPS and improve antibiotic penetration. Host-directed and immunomodulatory approaches employ monoclonal antibodies such as MEDI3902, vaccines, and immune modulators to enhance clearance. Together, these strategies collapse the biofilm tolerance architecture and restore the effectiveness of conventional antibiotics.
Figure 4. Mechanistic overview of network disruption strategies targeting the P. aeruginosa biofilm code. Four complementary therapeutic modules converge to dismantle biofilms and resensitize bacteria to antibiotics. Quorum-sensing inhibition uses small-molecule antagonists, signal-degrading enzymes, and CRISPR interference that targets lasI, rhlI, and pqsR to mute virulence and community signaling. Furthermore, c-di-GMP modulators, including PDE activators, DGC inhibitors, and nitric oxide donors, lower intracellular c-di-GMP and promote dispersal with a shift toward motile states. Matrix disruption relies on nanoparticles, depolymerases such as PslG, and mesoporous silica carriers to degrade EPS and improve antibiotic penetration. Host-directed and immunomodulatory approaches employ monoclonal antibodies such as MEDI3902, vaccines, and immune modulators to enhance clearance. Together, these strategies collapse the biofilm tolerance architecture and restore the effectiveness of conventional antibiotics.
Microorganisms 14 00109 g004
Table 1. QS systems and regulatory integration in P. aeruginosa.
Table 1. QS systems and regulatory integration in P. aeruginosa.
QS SystemSignal(s)Synthase/ReceptorMajor Regulatory LinksPrincipal Outputs/TraitsEnvironmental ModulatorsRepresentative Drug TargetsReference
Las3 oxo C12 HSLLasI and LasRActivates rhl and pqs and functions as a central regulator in the QS hierarchyElastase (lasB) and proteases, exotoxin A, and initiation of biofilm formationNutrient sufficiency and oxygen availabilityLasR antagonists and LasI inhibitors[16,102]
RhlC4 HSLRhlI and RhlRRepresses pqsABCDE during phosphate stress and forms a functional partnership with PqsERhamnolipids (rhlAB) and hydrogen cyanide, and control of motilityPhosphate limitationRhlR antagonists and compounds that disrupt the RhlR and PqsE interaction[60,65]
Pqs (MvfR)HHQ and PQSPqsABCDH and PqsR (MvfR)Regulates phenazine production as well as eDNA and OMV release, and provides feedback to QSPyocyanin and other phenazines and eDNA, OMVs, and promotion of dispersalIron availability and redox-related stressPqsR antagonists and inhibitors of PQS biosynthesis[103,104,105]
IqsIQSAmbBCDE and IqsR are responsive to PhoBLinks phosphate stress through PhoB to activation of rhl and pqsStress-responsive activation of virulence programsLow phosphateInhibitors of the PhoB IQS signaling pathway[61,106]
Global integrationMultipleRhlR with PqsE and AlgU with Alg44 and c-di-GMP and Gac and Rsm and VfrCoordinates QS and c-di-GMP cross-talk and Alg44 activation and higher-level regulationExpression of psl, pel, and alginate, and formation of chronic biofilms and persistenceHost-related stresses and mucoid conditionsEnzymes that control c-di-GMP and modulators of AlgU and Alg44[18,74,107]
Table 2. Representative c-di-GMP enzymes in P. aeruginosa inputs, phenotypes, and mechanisms.
Table 2. Representative c-di-GMP enzymes in P. aeruginosa inputs, phenotypes, and mechanisms.
Enzyme (Protein/Gene)Domain ArchitecturePrimary Input/StimulusDominant PhenotypeMechanism/NotesReference
WspR (wspR)GGDEF response regulator DGCSurface engagement through WspA and WspE and cell envelope perturbationIncreased biofilm formation and reduced motilityPhosphorylation-dependent subcellular clustering enhances DGC activity[111,112]
SadC (sadC)Membrane-associated GGDEFFlagellar stator load through MotC interactionReduced motility and increased biofilm formationMotC engages sadC to stimulate c-di-GMP synthesis[114]
RoeA (roeA)Inner membrane GGDEFControl of matrix productionIncreased Pel-related EPS synthesisChannels c-di-GMP to Pel machinery and functions separately from sadC control of motility[34,115]
TdcA (tdcA)GGDEF thermosensory DGCTemperature increase at a strong rate across a ten-degree rangeTemperature-dependent motility, biofilm formation, and virulenceThermosensory activity of TdcA produces marked increases in c-di-GMP with warming[130]
BifA (bifA)EAL domain PDEHomeostatic degradation of c-di-GMPActive BifA supports lower biofilm levels and greater swarming motilityDeletion of bifA elevates c-di-GMP and produces a hyperbiofilm and poor swarming state[116]
RbdA (rbdA)PAS GGDEF EAL PDERedox and oxygen-linked signalsIncreased dispersal through PDE activationAllosteric activation of the EAL domain by GTP binding to the GGDEF domain[117]
DipA (dipA)Multidomain EAL PDENutrient shifts and dispersion cuesIncreased dispersal that depends on PDE activityEssential for induced dispersion with rising PDE activity during dispersal[33,132]
ProE (proE)GGDEF EAL hybrid with active PDERedox and nutrient inputs with local EPS controlRestraint of the EPS gene expression when activeHighly active PDE that suppresses EPS transcription and shows polar localization[133]
Table 3. Therapeutic modalities targeting P. aeruginosa biofilms: mechanisms, models, outcomes, and translational status.
Table 3. Therapeutic modalities targeting P. aeruginosa biofilms: mechanisms, models, outcomes, and translational status.
ModalityMolecular Target(s) andMechanismRepresentative Agent(s)Primary Model(s) TestedKey Antibiofilm OutcomesTranslational ReadinessReference
QSIsAntagonize AHL-mediated QS through Las and Rhl; reduce QS-regulated virulence and EPS without strong growth inhibitionHalogenated furanones; ajoeneIn vitro flow cells; mammalian infection modelsReduced Las-controlled transcription, lower pyocyanin and rhamnolipids, decreased biofilm-associated virulence at sub-MIC levelsLead stage with in vivo proof-of-concept[174,176]
Natural QS antagonistsInhibit LasR and RhlR activation and downstream QS outputsMethyl gallateIn vitro multi-strain P. aeruginosa panels; docking and biochemical assaysReduced QS phenotypes, including pyocyanin, motility, and rhamnolipids, at sub-MIC levelsPreclinical discovery and optimization[178,227]
Quorum quenching enzymesEnzymatic degradation of AHL signalsPvdQ acylase; AHL lactonases and acylasesMurine lung infection models; in vitro biofilmsAttenuated QS activity and decreased biofilm burden; adjuvant potential with antibioticsEnzyme engineering in progress[180,181,228]
CRISPRi against QS nodesProgrammable repression of lasI, rhlI, and pqsRdCas9-based CRISPRi constructsIn vitro systems; emerging in vivo modelsTargeted QS knockdown with reduced virulence and biofilm formationPlatform stage[187,229]
c-di-GMP modulatorsLower c-di-GMP through PDE activation or DGC inhibition; promotes dispersalH6-335 class; H6-335-P1In vitro biofilms; murine implant infection modelsDispersal of established biofilms and enhanced clearance with antibioticsStrong preclinical signal[188]
NO-based dispersal triggersActivate PDEs and transiently reduce c-di-GMPLow-dose NO donorsFlow-cell and airway modelsRapid dispersal and increased antibiotic susceptibilityAdjunct concept with growing clinical interest[21,230]
Metal/metal-oxide NPsGenerate reactive species, disrupt membranes and proteins, enhance penetrationAgNPs; ZnO; TiO2In vitro biofilms; device coatings; preclinical studiesSynergy with antibiotics; antibiofilm surface formationVariable; dose and irradiation windows critical[231,232]
Phages with depolymerasesMatrix digestion and lytic killing; improve penetrationPhage cocktails; polysaccharide depolymerasesIn vitro, zebrafish, murine airway/implant modelsStrong biofilm reduction; synergy with antibioticsAdvancing through preclinical development[213,233,234,235]
Monoclonal antibodiesNeutralize adhesins and secretion components; enhance opsonophagocytosisMEDI3902 (anti-Psl and anti-PcrV)Phase 1 studies; preclinical pneumonia/bacteremia modelsEstablished safety and pharmacokinetics; protective effectsClinical-stage monoclonal antibody[221,236]
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

Elbehiry, A.; Marzouk, E.; Edrees, H.M.; Ibrahem, M.; Alzahrani, S.; Anagreyyah, S.; Abualola, H.; Alghamdi, A.; Alzahrani, A.; Jaber, M.; et al. Understanding Pseudomonas aeruginosa Biofilms: Quorum Sensing, c-di-GMP Signaling, and Emerging Antibiofilm Approaches. Microorganisms 2026, 14, 109. https://doi.org/10.3390/microorganisms14010109

AMA Style

Elbehiry A, Marzouk E, Edrees HM, Ibrahem M, Alzahrani S, Anagreyyah S, Abualola H, Alghamdi A, Alzahrani A, Jaber M, et al. Understanding Pseudomonas aeruginosa Biofilms: Quorum Sensing, c-di-GMP Signaling, and Emerging Antibiofilm Approaches. Microorganisms. 2026; 14(1):109. https://doi.org/10.3390/microorganisms14010109

Chicago/Turabian Style

Elbehiry, Ayman, Eman Marzouk, Husam M. Edrees, Mai Ibrahem, Safiyah Alzahrani, Sulaiman Anagreyyah, Hussain Abualola, Abdulaziz Alghamdi, Ahmed Alzahrani, Mahmoud Jaber, and et al. 2026. "Understanding Pseudomonas aeruginosa Biofilms: Quorum Sensing, c-di-GMP Signaling, and Emerging Antibiofilm Approaches" Microorganisms 14, no. 1: 109. https://doi.org/10.3390/microorganisms14010109

APA Style

Elbehiry, A., Marzouk, E., Edrees, H. M., Ibrahem, M., Alzahrani, S., Anagreyyah, S., Abualola, H., Alghamdi, A., Alzahrani, A., Jaber, M., & Abu-Okail, A. (2026). Understanding Pseudomonas aeruginosa Biofilms: Quorum Sensing, c-di-GMP Signaling, and Emerging Antibiofilm Approaches. Microorganisms, 14(1), 109. https://doi.org/10.3390/microorganisms14010109

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