Open access peer-reviewed chapter

Natural Compounds Inhibiting Pseudomonas aeruginosa Biofilm Formation by Targeting Quorum Sensing Circuitry

Written By

Julie Carette, Amandine Nachtergael, Pierre Duez, Mondher El Jaziri and Tsiry Rasamiravaka

Submitted: 06 September 2019 Reviewed: 11 December 2019 Published: 29 January 2020

DOI: 10.5772/intechopen.90833

From the Edited Volume

Bacterial Biofilms

Edited by Sadik Dincer, Melis Sümengen Özdenefe and Afet Arkut

Chapter metrics overview

1,132 Chapter Downloads

View Full Metrics

Abstract

The biofilm lifestyle mode certainly represents one of the most successful behaviors to facilitate bacterial survival in diverse inhospitable environments. Conversely, the ability of bacteria to develop effective biofilms represents one of the major obstacles in the fight against bacterial infections. In Pseudomonas aeruginosa, the biofilm formation is intimately connected to the quorum sensing (QS) mechanisms, a mode of cell-to-cell communication that allows many bacteria to detect their population density in order to coordinate common actions. In this chapter, we propose an overview (i) on P. aeruginosa QS mechanisms and their implication in biofilm formation, and (ii) on natural products that are known to interfere with these QS mechanisms, subsequently disrupting biofilm formation. The concluding remarks focus on perspectives of these compounds as possible antibiotherapy adjuvants.

Keywords

  • biofilm
  • las
  • natural products
  • PQS
  • pseudomonas
  • quorum sensing
  • rhl

1. Introduction

Bacterial infections are mainly related to the ability of bacteria to invade and disseminate through their hosts by using different types of motility, by releasing a myriad of virulence factors, by building structured biofilm which lead to host cell and tissue damage but also allow bacteria to evade the immune system and conventional antimicrobial agents [1]. For decades, antibiotics, although less effective in biofilm-growing bacteria [2], have represented our best weapon against bacterial diseases. However, the on-going emergence and worldwide spreading of resistant bacteria is considerably reducing the antibiotic pallet available for the treatment of bacterial infections [3]. This alarming situation forces researchers to consider other strategies to combat bacterial infections, notably the use of phages [4] or the use of alternative agents, such as essential oils [5], silver nanoparticles [6], bacteriocins [7], and antimicrobial peptides [8]. Some interesting strategies propose original compounds that disrupt biofilm formation without affecting the viability of invading bacteria; this strategy is expected (i) to reduce the bacterial aptitude to build protective barriers, but without exerting a selective pressure per se [4]; (ii) to allow sufficient time for the immune defenses to effectively destroy invaders; and (iii) to minimize the use of effective antibiotics.

In most bacteria, the expressions of virulence factors are coordinated by quorum sensing (QS) mechanisms, a cell-to-cell communication which allows bacteria to detect their population density by producing and perceiving diffusible signal molecules to synchronize common actions [9]. This cell-to-cell communication has been largely investigated in Pseudomonas aeruginosa, an opportunistic pathogen which mainly affects people who are severely immunocompromised, in part due to its ability to evade from both innate and acquired immune defenses through adhesion, colonization, and biofilm forming and to produce various virulence factors that cause significant tissue damage [10, 11]. In this bacterium, QS regulates virulence factors production, motilities and, in particular, biofilm formation for which QS is one of the relevant key actors. Interestingly, within the two past decades, study papers reporting natural and synthetic compounds that interfere with QS and/or biofilm formation are regularly published; QS circuitry and biofilm formation control mechanisms indeed constitute promising targets to struggle against P. aeruginosa infection with potential huge clinical interests [12]. The present chapter covers the scope of natural compounds from both prokaryote and eukaryote organisms that have been identified to disrupt the biofilm lifestyle cycle in P. aeruginosa via modulation of QS mechanisms. An overview of the entanglement between QS circuitry and biofilm formation is reported as a prerequisite for a better understanding of the mechanisms of action proposed for some of the identified compounds. The concluding remarks focus on the perspectives of these compounds as possible antibiotherapy adjuvants for possible eradication of resistant infections caused by P. aeruginosa.

Advertisement

2. P. aeruginosa biofilm lifestyle

Like most bacteria, P. aeruginosa can develop two distinct lifestyles, planktonic and sessile cells. The planktonic state is encountered when P. aeruginosa evolves freely in a liquid suspension, whereas on natural or synthetic surfaces, P. aeruginosa can form sticky clusters in permanent rearrangements characterized by the secretion of an adhesive and protective matrix [13]. Defined as “biofilm,” this set of bacterial community adherent to a surface appears as an adaptive response to an environment more or less unsuited to growth in planktonic form [14].

The biofilm formation can be delimited in five main stages (Figure 1, image A). A first reversible phase corresponds to the initial adhesion of bacteria to surfaces; this adhesion becomes irreversible in the second stage (image B). Then, thanks to a proliferation period corresponding to the third stage, microcolonies are built concomitantly with the production of extracellular matrix (image C), leading to the fourth stage of biofilm structuration and organization in which the growth of three dimensional communities is observed with amplified extracellular matrix production (image D). This biofilm cycle is completed by a dispersion step (image E) [12].

Figure 1.

Sketch of the different steps of a biofilm development (A) [15]. Several representative scanning electron microscopy (SEM-JEOL JSM-7200F) images of the P. aeruginosa biofilm at different steps of development and with different magnifications (B = reversible and irreversible stages at 8 h growth, C = microcolonies stage at 30 h growth, D = mature biofilm stage at 120 h growth, and E = dispersion stage at 144 h growth). P. aeruginosa PAO1 colonies were grown at 37°C with Centers for Disease Control and Prevention (CDC) biofilm reactor (biosurface technologies, MT) on tryptone soy broth (TSB).

The secreted extracellular matrix mainly consists of proteins, nucleic acids, lipids, and exopolysaccharides (EPS). These account for 50–90% of total organic matter [16]. P. aeruginosa produces at least three types of EPS that are required for biofilm formation and architecture [17]. (i) Alginate a linear polysaccharide composed of L-guluronic and D-mannuronic acids linked by β-1,4 bonds [18], (ii) Pel polysaccharide, a glucose-rich matrix material, with unclarified composition, and (iii) Psl polysaccharide, a repeating pentasaccharide consisting of D-mannose, L-rhamnose, and D-glucose. In mucoid strains, EPS are predominantly characterized by the presence of alginate. The alginate participates in the structuring of the biofilm [19], but its real importance is still controversial since some authors claim that it is not essential; indeed architecture and antibiotic resistance profiles of wild-type and alginate-deficient biofilms are identical [20, 21]. Nevertheless, the overexpression of alginate was shown to protect P. aeruginosa from phagocytosis and host responses [22]. In “nonmucoid” P. aeruginosa strains, such as the PAO1 strain isolated from an infected wound [23], alginate is even considered poorly produced at the expense of exopolysaccharides rich in glucose and mannose [24], Pel and Psl, which have been described as being more important in the formation and maintenance of the biofilm [25].

Extracellular DNA (eDNA) is an important component of P. aeruginosa biofilm matrix, which particularly intervenes in the establishment, maintenance, and perpetuation of structured biofilms [26]. Its importance has been demonstrated since P. aeruginosa biofilm formation is prevented by exposition to DNase I [27] and biofilms that are deficient in eDNA have been shown to be more sensitive to the detergent sodium dodecyl sulfate [28]. It has been established that eDNA plays roles in bacterial adhesion and in the structural stability of biofilms by maintaining coherent cell alignments [29]; interestingly, its contribution to antimicrobial resistance has also been proposed as eDNA, a highly anionic polymer, is believed to bind cationic antibiotics, such as aminoglycosides and antimicrobial peptides [30].

Advertisement

3. QS mechanisms and their implication in biofilm formation

The complex regulation of biofilm formation involves multiple bacterial machineries including the QS systems. In P. aeruginosa, this mechanism is involved in the development of various common bacterial behaviors, including virulence factors expression and biofilm formation, which are mostly implicated in infection success. Three QS systems have been clearly characterized: (i) the las system and the rhl system, two LuxI/R type systems using the signal molecules of the family of acyl-homoserine lactones (AHLs); and (ii) the PQS (pseudomonas quinolone signal) system based on molecules of the 2-alkyl-4-quinolone class [10, 31]. The mechanisms of QS in P. aeruginosa are summarized in Figure 2 while the main functions regulated by QS systems and involved in the pathogenesis of P. aeruginosa are presented in Figure 3.

Figure 2.

Systems involved in P. aeruginosa QS circuitry. The main QS systems in P. aeruginosa are the las, rhl, and PQS systems. The las system consists of a lasR regulatory gene coding for the LasR protein, a lasI gene coding for a LasI synthase involved in the synthesis of a signal molecule of the acyl-homoserine lactone (AHL) family, the 3-oxo-C12-HSL. The LasR/3-oxo-C12-HSL complex is a transcriptional activator of virulence genes (protease, elastase, and exotoxin) and lasI gene. According to the same model, the rhl system consists of rhlR, rhlI genes, and another AHL, the C4-HSL. This system activates genes in common with the las system and also specific genes, such as those coding for the synthesis of rhamnolipids, pyocyanin, and swarming/twitching motilities. The las system controls the rhl system. The third PQS system is interposed between the two main systems. The PqsABCDE operon produces the precursor 2-heptyl-4-quinolone (HHQ), and PqsH catalyzes conversion of HHQ to 2-heptyl-3-hydroxy-4-quinolone (PQS), detected by the receptor PqsR [10, 31].

Figure 3.

Functions positively regulated by QS in P. aeruginosa [10, 31].

Evidence that the las system is implicated in biofilm formation has been firstly established when Davies et al. [32] demonstrated that the biofilm formed by lasI mutant appears flat, undifferentiated, and quickly dispersed from the surface upon exposure to sodium dodecyl sulfate, compared to wild type biofilms.

Furthermore, Gilbert et al. [33] observed the binding of the QS regulator LasR to the promoter region of the psl operon, suggesting that the psl expression may be regulated by the QS. Considering that the psl operon is implied in biofilm modulation, the QS then plays a role in the biofilm formation and architecture. The transcription of the pel operon seems to be reduced in rhlI mutant, suggesting that the rhl system plays a biofilm formation role in P. aeruginosa by modulating the biosynthesis of the Pel polysaccharide [34]. The pqsA mutant produces a biofilm with less eDNA than the wild type biofilm, suggesting that the PQS system also plays a role in biofilm formation, more particularly in the eDNA releasing [34].

Notably, the production of rhamnolipids and lectins is under QS control, indicating a further indirect link between biofilm formation/degradation and QS.

Indeed, the rhl system controls the production of rhamnolipids [35], that play multiple roles in P. aeruginosa biofilm formation: (i) as biosurfactant and virulence factor [36]; (ii) in the formation of microcolonies [37]; (iii) in the maintenance of open channel structures necessary for nutrient circulation [38]; (iv) in the development of biofilm mushroom-shaped structures [37]; and (v) in cell dispersion from the biofilm [39]. Indeed, a hyper-detaching property has been observed in the P. aeruginosa mutants that produce more rhamnolipids compared to wild type strains [40]. Moreover, the rhl system also controls the expression of the cytotoxic virulence factors LecA and LecB. Data obtained on mutant strains indicate that these galactophilic lectins probably contribute to the biofilm development [41, 42]. Similarly, two types of P. aeruginosa motilities implicated in biofilm formation are also QS-regulated. The first movement, swarming motility, accomplishes an organized surface translocation, dependent on cell-to-cell contacts and extensive flagellation [43]; this has been observed during the first stage of P. aeruginosa biofilm development and seems to be regulated by the rhl system [44]. Flat and uniform biofilms are formed when the strains grow under conditions promoting swarming motility, for example, a growth medium with glutamate or succinate as carbon sources; by contrast, a biofilm without confluent cell aggregates is formed by strains with limited swarming motility [45]. The second movement, a flagella-independent form of translocation, is described as a successive extension and retraction of polar type IV pili [46]. This kind of movement, regulated by the rhl system on a Fe-limited minimal medium [47], is necessary to assemble bacteria in monolayers that form microcolonies [38].

Advertisement

4. Other mechanisms implied in biofilm formation

The QS systems are not the sole key actors intervening in biofilm formation by P. aeruginosa. Indeed, the complex regulation of biofilm formation involves multiple bacterial machineries that also include the membrane-bound sensor kinase GacS, the transcriptional response regulator GacA (GacS/GacA two-component regulatory system), and the intracellular second messenger bis-(3′-5′)-cyclic dimeric guanosine monophosphate (c-di-GMP). Briefly, the GacS/GacA system acts as a super-regulator of the las and rhl systems [48], whereas c-di-GMP is important for the biosynthesis of alginate and Pel polysaccharides and for the switch from planktonic to biofilm lifestyle [49].

Advertisement

5. Natural products that affect QS and biofilm formation by Pseudomonas aeruginosa

5.1 From prokaryotes

5.1.1 Enzymes

Microorganisms known to have the ability to produce anti-QS enzymes are still limited to a few bacteria from the families of (i) Actinobacteria (Rhodococcus and Streptomyces); (ii) Firmicutes-Arthrobacter (Bacillus and Oceanobacillus); (iii) Cyanobacteria (Anabaena); (iv) Bacteroidetes (Tenacibaculum); (v) Proteobacteria (Acinetobacter, Agrobacterium tumefaciens, Alteromonas, Comomonas, Halomonas, Hyphomonas, Klebsiella pneumoniae, P. aeruginosa, Ralstonia, Stappia, and Variovorax paradoxus) [5056].

Four types of enzymes are known to degrade AHLs [57, 58], a phenomenon sometimes described as “quorum quenching” (QQ) [59]; these include AHL-lactonases and decarboxylases that attack the lactone ring (Bacillus indicus, B. pumilus, and B. sp. SS4 cause significant inhibition of QS-dependent activities in Gram-negative bacteria such as P. aeruginosa PAO1, Serratia marcescens, and Vibrio), AHL-acylases that cleave the acyl side chain (B. pumilus S8-07 degrades 3-oxo-C12-HSL into the corresponding lauric acid [60]), and deaminases that separate the lactone ring from the acyl side chain. Recently, lactonases and acylases were identified in Erythrobacter, Labrenzia, and Bacterioplanes found in Red Sea sediments; these both degrade AHLs of different acyl chain lengths, particularly the 3-oxo-C12-HSL, and inhibit the formation P. aeruginosa PAO1 biofilm [59].

Mycobacteroides abscessus subspecies, emerging pathogens, are capable of degrading both PQS and HHQ . M. abscessus subsp. abscessus, in coculture with P. aeruginosa PAO1, reduced PQS levels through a PQS dioxygenase (encoded by the aqdC gene), M. abscessus subsp. massiliense, a recombinant strain overexpressing the aqdC gene, reduces the level of the virulence factors pyocyanin, pyoverdine, and rhamnolipids, suggesting that AqdC is a QQ enzyme [61]. Its impact on biofilm formation would have been interesting to investigate as another dioxygenase, the 2-alkyl-3-hydroxy-4(1H)-quinolone 2,4-dioxygenase (HodC), was described to cleave PQS, attenuate the production of virulence factors but conversely increase the viable biomass, in both newly formed and established biofilms, by increasing iron availability [62].

5.1.2 Organic acids

The acetic and phenyl lactic acids, found in the supernatant of probiotic strains Lactobacillus paracasei subsp. paracasei CMGB isolated from newborn feces, were shown to inhibit, at nonbacteriostatic/bactericide levels, the expression of QS genes in P. aeruginosa, preventing adherence of bacteria to an inert substratum [63, 64]. Similarly, the lactic acid produced by a potential probiotic Pediococcus acidilactici M7 strain, also isolated from newborn feces, inhibits the production of P. aeruginosa short-chain AHLs, elastase, protease, pyocyanin, and biofilm as well as the swarming-swimming-twitching motilities [65].

5.2 From fungi

5.2.1 Antibiotics and mycotoxins

Penicillin produced by Penicillium spp. has been shown to be effective in controlling a bacterial infection. Recently, about 33 Penicillium spp. have been recognized as producers of QS inhibitors such as the small lactone mycotoxins patulin and penicillic acid. The use of patulin can significantly reduce lung infection caused by P. aeruginosa on a mouse model. Interestingly, a synergy has been shown in vitro between patulin and tobramycin toward P. aeruginosa PAO1 biofilms, whereas patulin alone does not affect the development of biofilm [66]. Although the anti-infective property of patulin has been demonstrated, its genotoxicity and potential carcinogenic properties [67] probably preclude clinical applications.

Erythromycin, a macrolide antibiotic isolated from Saccharopolyspora erythraea, has been recently demonstrated to reduce virulence factors in P. aeruginosa PAO1, including various motilities, biofilm formation, and production of rhamnolipids, total protease, elastase, and pyocyanin at nonmicrobicidal level (1.6 μg/mL) [68]. Comparably, the erythromycin derivate, azithromycin, shows a strong P. aeruginosa QS and biofilm inhibitory effect [6971] with inhibition of alginate synthesis [69], a reduction of each type of bacteria movement [72] and a diminution of gacA gene expression [73]. At weak antibiotic concentration (2 μg/mL), a biofilm inhibition is observed, probably explained by a lower production of both AHL signal molecules, C4-HSL and 3-oxo-C12-HSL, and of virulence factors [7476].

5.2.2 Alkylcyclopentanone

Recently, Kim et al. [77] indicated that the alkylcyclopentanone terrein, isolated from Aspergillus terreus, reduced virulence factors (elastase, pyocyanin, and rhamnolipids) and biofilm formation via antagonizing QS receptors without affecting P. aeruginosa cell growth. Beyond a negative impact on the production of QS signaling molecules and expression of QS-related genes, terrein also reduced c-di-GMP levels, an important secondary messenger for the switch from planktonic to biofilm lifestyle mode, by decreasing the activity of a diguanylate cyclase required for c-di-GMP biosynthesis [78].

5.3 From Plants

5.3.1 Derivatives of shikimic acid, phenols, and polyphenols

Many phenolic compounds and derivatives with anti-QS and antibiofilm activities have been isolated from plants [79]. Cinnamaldehyde [the dominant compound of certain essential oils, in particular Cinnamomum camphora (L.) J. Presl] and its derivatives modulate a wide range of anti-QS and antibiofilm activities of P. aeruginosa [8082]. Curcuma longa L. produces curcumin, which inhibits the expression of virulence genes of P. aeruginosa PA01 [83].

Ellagic acid derivatives from Terminalia chebula Retz. downregulate lasIR and rhlIR genes expression and decrease AHLs production, leading to an attenuation of virulence factor production and to an enhanced sensitivity of biofilm facing a tobramycin treatment [84].

Flavonoids have been investigated for their roles as QS modulating compounds. From these, naringenin and taxifolin reduced the expression of several QS-controlled genes (i.e., lasI, lasR, rhlI, rhlR, lasA, lasB, phzA1, and rhlA) in P. aeruginosa PAO1. Similarly, the flavan-3-ol catechin, extracted from the bark of Combretum albiflorum (Tul.) Jongkind, reduces the production of QS-dependent virulence factors, such as pyocyanin, elastase, and the formation of biofilm by P. aeruginosa PAO1 [85]. Interestingly, baicalin, an active natural compound extracted from the traditional Chinese medicinal Scutellaria baicalensis, has been demonstrated to inhibit the formation of P. aeruginosa biofilms and enhance the bactericidal effects of antibiotics such as amikacin. Moreover, at sub-minimal inhibitory concentration (256 μg/mL), this flavonoid has been shown to reduce LasA protease, LasB elastase, pyocyanin, rhamnolipids, and exotoxin A production and to downregulate the three QS-regulatory genes, including lasI, lasR, rhlI, rhlR, pqsR, and pqsA [86]. Consistently, in vivo experiments indicated that baicalin treatment reduces P. aeruginosa pathogenicity in Caenorhabditis elegans and enhances the clearance of P. aeruginosa from the peritoneal implants of infected mice.

Furocoumarins from grapefruit can inhibit the QS signaling (AHLs and AI-2) of V. harveyi BB886 and BB170 strains as well as biofilm formation in pathogens such as E. coli O157:H7, Salmonella typhimurium and P. aeruginosa [87]. These purified furocoumarins (dihydroxybergamottin and bergamottin), tested at the concentration of 1 μg/mL, cause 94% inhibition of autoinducers (AHLs) without affecting bacterial viability. Biofilm inhibition was up to 58.3 and 72%, respectively, for E. coli O157:H7 but modest for P. aeruginosa (27.3 and 18.1%, respectively).

Malabaricone C, a diarylnonanoid isolated from the bark of Myristica cinnamomea King inhibited the QS-regulated pyocyanin production and biofilm formation in P. aeruginosa PAO1 [88].

A screening of various herbs revealed that a clove extract [Syzygium aromaticum (L.) Merr. Et Perry] inhibits QS-controlled gene expression (las and PQS systems) in P. aeruginosa with eugenol as major active constituent [89]. Recently, the effects of eugenol and its nanoemulsion on P. aeruginosa QS-mediated virulence factors and biofilm formation have been identified by Lou et al. [90] at a 0.2 mg/mL concentration. Similarly, the anthraquinone emodin from Rheum palmatum L., a traditional Chinese medicinal plant, was found to inhibit the P. aeruginosa biofilm formation at 20 μM, increasing the antibiotic activity of ampicillin [91]. Finally, the 6-gingerol, isolated from fresh ginger oil, reduces the production of several virulence factors, decreasing the mortality induced in mice by P. aeruginosa. A DNA microarray analysis revealed that the application of the 6-gingerol on biofilm-encapsulated cells down-regulates several QS-related genes, notably those involved in the production of rhamnolipids, elastase, pyocyanin, all of which are involved in biofilm formation [92].

5.3.2 Alkaloids

Recently, caffeine (a purine alkaloid) has been shown to inhibit AHLs production and swarming mobility in P. aeruginosa PAO1 without causing AHLs degradation [93].

5.3.3 Terpenoids and Triterpenoids

The pentacyclic triterpenoid ursolic acid was identified as an inhibitor of biofilm formation from Diospyros dendo Welw, the tree used for ebony from Gabon, Africa [94]. Tested at a dose of 10 μg/mL, ursolic acid reduces biofilm formation by 79% in E. coli and 57–95% in V. harveyi and P. aeruginosa PAO1. Similarly, oleanolic acid inhibits the in vitro biofilm formation by S. aureus and P. aeruginosa [95]. However, these triterpenoids showed no inhibitory effect on QS mechanisms contrarily to triterpenoid coumarate esters isolated from Dalbergia trichocarpa, a tropical legume from Madagascar. Indeed, oleanolic aldehyde coumarate at 200 μM inhibits the formation/maintenance of P. aeruginosa PAO1 biofilm and the expression of the las and rhl QS systems as well as gacA gene [96]. Consequently, the production of QS-controlled virulence factors, including, rhamnolipids, pyocyanin, elastase, and extracellular polysaccharides, as well as twitching and swarming motilities is reduced. Other African plants harbor terpenoids and triterpenoids with antivirulence properties. Indeed, cassipourol and β-sitosterol (both at 100 μM), isolated from Platostoma rotundifolium (Briq.) A. J. Paton, a Burundian medicinal plant, inhibit quorum sensing-regulated and -regulatory gene expression in las and rhl systems. These triterpenoids can still disrupt the formation of biofilms at concentrations down to 12.5 and 50 μM [97].

5.3.4 Isothiocyanates and organosulfur compounds

Isothiocyanates produced by many plants are also QS inhibitors in P. aeruginosa PAO1. For example, iberin, isolated from horseradish (Armoracia rusticana G. Gaertn et al.), specifically blocks the expression of QS-regulated genes in P. aeruginosa PAO1 at the concentration of 100 μM; its impact on biofilm formation has not been investigated [98]. Sulforaphane and erucin, two isothiocyanates isolated from broccoli, inhibit the P. aeruginosa PAO1 las and rhl system as well as biofilm formation at concentrations of 50 and 100 μM, respectively [99].

A further compound known to affect the QS-regulated genes in P. aeruginosa, including the rhamnolipids production, is ajoene, an allyl sulfide isolated from Allium sativum L. Ajoene, at the concentration of 100 μg/mL and combined with the antibiotic tobramycin, leads to killing of biofilm-encapsulated P. aeruginosa. In a mouse model of pulmonary infection, this synergy improves the clearance of P. aeruginosa from lungs [100]. The S-phenyl-L-cysteine sulfoxide and its derivatives, notably diphenyl disulfide, have shown a significant impact on the biofilm formation by P. aeruginosa [101]; the sulfoxide derivative seems to interfere with both las and rhl systems whereas the diphenyl sulfide only disturbs the las system.

5.4 From marine organisms

5.4.1 Furanones

A series of studies have indicated that marine organisms are a potential source of anti-QS [102104]. The halogenated furanones produced by the red alga Delisea pulchra inhibit QS-induced activities in bacteria by competing with AHL signals related to their receptor site (LuxR) [104]. This protein-ligand binding is destabilized, causing rapid receptor recycling [102]. Inspired from natural compounds, the halogenated furanones C-30 and C-56 have been demonstrated to exhibit biofilm reduction and target the las and rhl systems in P. aeruginosa [105].

5.4.2 Terpenoids

Following a screening of 284 extracts from the marine sponge Luffariella variabilis, 36 extracts were revealed as inhibitors of P. aeruginosa QS, targeting the las system [103]; from these, the sesterterpenoids manoalide displays antibiofilm activities. Note that this molecule does not generate bactericidal effects on P. aeruginosa [103], but presents an antibiotic activity against Gram-positive bacteria [106].

5.5 From animals and human

5.5.1 Enzymes

Type I porcine kidney acylase inactivates QS signals such as C6-HSL and 3-oxo-C12-HSL but not C4-HSL [50]. This type I acylase moderately reduces biofilm formation in Aeromonas hydrophila, P. putida, and probably P. aeruginosa [107]. This degradation is dependent on the length of the acyl chain, since only C6-HSL and 3-oxo-C12-HSL are degraded [108].

Mammalian cells release enzymes called paraoxonases 1 (extracted from human and murine sera) that have lactonase activity; degrading P. aeruginosa AHLs. They prevent, in an indirect way, QS and biofilm formation [109]. Similarly, human epithelial cells and particularly human respiratory epithelia have the capacity to inactivate a P. aeruginosa QS signal by inactivating AHLs (3-oxo-C12HSL) produced by P. aeruginosa [108, 110]. However, the enzyme or enzyme-like compound involved in acyl-homoserine lactone inactivation have not been identified and characterized yet. Recently, Losa et al. [111] demonstrated that polarized airway epithelial monolayers, in contrast to nonpolarized cells, are also able to degrade 3-oxo-C12-HSL using membrane-associated paraoxonase 2 that catalyzes the opening of the lactone ring.

5.5.2 Alkaloids

The P. aeruginosa pyocyanin production is inhibited by a molecule found and isolated from the ant Solenopsis invicta, the piperidine alkaloid Solenopsin A alkaloid. The biofilm formation is also reduced in a dose-dependent manner. This molecule probably disrupts the signals from the rhl system [112].

Advertisement

6. Concluding remarks

This review presents natural compounds reported to exhibit anti-QS and antibiofilm properties against P. aeruginosa (summarized in Table 1); these highlight the great potentiality of living organisms as reservoir of compounds susceptible to modulate virulence mechanisms without affecting bacterial viability. Overall, it appears that prokaryotes as well as animals and humans are sources for enzymes that degrade or antagonize AHLs, whereas plants harbor larger panels of anti-QS and antibiofilm compounds with very diverse chemical structures, including alkaloids, organosulfurs, phenolics, and terpenoids. Contrarily to animals and humans, plants are not able to deploy elaborate defense through humoral and cell-mediated immunity (antibodies and phagocytes) to struggle against bacterial invasions [113]. Plants immune defenses rely on the secretion of antibacterial compounds (bactericide and/or bacteriostatic agents [114]), including resistance modulating compounds [115] (e.g., inhibitors of efflux pumps [116]), and mostly on their ability to recognize molecules released from pathogens through plant cell surface receptors. This recognition triggers specific signaling cascades, activating series of defense responses, including the synthesis of antimicrobial lytic proteins, enzymes, phytoalexins, and other secondary metabolites. Some of these exert nonmicrobicidal antivirulence properties [117, 118]. Finally, marine organisms and fungi produce also bioactive secondary metabolites (halogenated furanones and antibiotics, respectively) and other original and promising compounds, such as terrein which was identified as the first dual inhibitor of QS and c-di-GMP signaling at 30 μM.

Origin Compounds (class) Target (QS) Synergy with antibiotics
Prokaryotes Bacillus indicus, B. pumilus, B. sp. [60]; Erythrobacter, Labrenzia, Bacterioplanes [59] AHL-acylase (Enzyme) AHL degradation NC
AHL-lactonase (Enzyme) NC
Lactobacillus paracasei subsp. Paracasei [64]; Pediococcus acidilactici M7 [65] Acetic acid, lactic acid, phenyl lactic acid AHL antagonist NC
Fungi Penicillium species [66] Penicillic acid (Furanone) LasR and RhlR NC
Patulin (Furopyranone) LasR and RhlR ǂ + 1
Saccharopolyspora erythraea [68] Erythromycin (Macrolide) rhl system and GacA NC
Aspergillus terreus [77] Terrein (alkylcyclopentanone) LasR and RhlR antagonist;
c-di-GMP
NC
marine organisms Delisea pulchra [102104] halogenated furanones and derivative AHL antagonist + 1
Luffariella variabilis (Polejaeff, 1884) [103] Manoalide (Sesterterpenoid) las system NC
Plants Platostoma rotundifolium (Briq,) A, J, Paton [97] Cassipourol (terpenoid), β-sitosterol (terpenoid) las and rhl systems + 1
Combretum albiflorum (Tul.) Jongkind [85] Catechin (Flavonoid) las and rhl systems NC
Dalbergia trichocarpa Baker. [96] Oleanolic aldehyde Coumarate (Phenolic compound) las and rhl systems + 1
Allium sativum L. [100] Ajoene (Organosulfur) las and rhl systems + 1
Armoracia rusticana G. Gaertn et al. [98] Iberin (Isothiocyanate) las and rhl systems NC
Terminalia chebula Retz. [84] Ellagic acid derivatives (Phenolic compound) las and rhl systems + 1
Syzygium aromaticum (L.) Merr. Et Perry [8990] Eugenol (Phenylpropanoid) las and PQS systems NC
Curcuma longa L. [83] Curcumin (Phenolic compound) AHLs inhibition NC
Citrus paradisi Macfad. (Rio Red and Marsh White grapefruits) [87] Bergamottin and dihydroxybergamottin (Furocoumarins) AHLs inhibition NC
Rheum palmatum L. [91] Emodin (Anthraquinone) docking traR * + 2
Scutellaria baicalensis Georgi. [86] Baicalin (Flavonoid) las, rhl and PQS systems + 1
Zingiber officinale Rosc. [92] 6-gingerol (Phenolic compound) docking lasR NC
Animals and Human Porcine kidney [50107] Type I acylase AHL degradation NC
Human and murine sera [109, 110] Paraoxonases 1 Enzyme (lactonase) AHL degradation NC
Solenopsis invicta (insect; ant) [112] Solenopsin A (Alkaloid) rhl system NC

Table 1.

Natural compounds inhibiting P. aeruginosa QS and biofilm formation.

Patulin alone does not affect the development of biofilm.


LuxR-type transcription factor of Agrobacterium tumefaciens.


Aminoglycosides.


Ampicillin.


+, yes; NC, not communicated.

The increasing presence of antibiotic-resistant bacteria certainly pushes scientists to reorient the strategy of fight against bacterial infections to defer entry into a post-antibiotic era where major antibiotics would not be effective even for banal infections. Antivirulence approaches and antivirulence drugs are being increasingly considered as potential therapeutic alternatives and/or adjuvants to currently failing antibiotics. For example, oleanolic aldehyde coumarate and cassipourol, anti-QS compounds, exert interesting antibiofilm properties, restoring the effectiveness of the antibiotic tobramycin in the clearance of biofilm-encapsulated P. aeruginosa (Figure 4); also the association between biofilm formation and antimicrobial resistance has been highlighted in carbapenem-resistant P. aeruginosa [119]. Such nonmicrobicidal drugs inhibit virulence factors essential for establishing infection and pathogenesis through targeting nonessential metabolic pathways which should not lead to activation of bacterial evasion mechanisms. This approach should reduce the selective pressure and consequently could slow down the development of resistance. Compounds that target QS may be particularly interesting as they impact planktonic and biofilm lifestyles, by reducing at the same time the production of virulence factors and the generation of biofilms. This should lead to less severe infections at levels that can be cleared by the host’s immune defense and with increased activity of antibiotics.

Figure 4.

P. aeruginosa biofilm phenotypes and effectiveness of tobramycin treatment in presence of dimethyl sulfoxide (DMSO 1%) or, cassipourol (CAS: 100 μM) or oleanolic aldehyde coumarate (OALC: 200 μM). (a) After 1 day of incubation, P. aeruginosa fails to form structured confluent aggregate in presence of CAS or OALC as compared to DMSO treatment. (b) CAS and OALC considerably increase the susceptibility of P. aeruginosa to tobramycin (100 μg/mL), as shown by the increased proportion of dead cells compared with DMSO. Similar results are observed when tobramycin is added simultaneously with CAS or OALC to one-day old untreated biofilms. The bacterial viability was assessed by staining the cells with SYTO-9 (green areas zones—live living bacteria) and propidium iodide (red areas zones—dead bacteria) furnished in the LIVE/DEAD BacLight kit. Cells were visualized using a LeicaDMIRE2 inverted fluorescence microscope using equipped with a 40× objective lens and colored images were assembled using Adobe Photoshop.

Despite these important prospects, however, the big breakthrough in antibacterial strategies is still out of reach. This is probably due to a very complex entanglement between different QS systems, to the ability of Pseudomonas to compensate deficient systems and to the intervention of key actors involved in biofilm formation, outside of QS circuitry [12]. Millenia of coevolution between plants and bacteria have led to complex defense strategies, with plants producing cocktails of bioactive compounds with multiple targets [114] and/or compounds such as terrein that impact dual inhibitory targets. In the current state of research, much remains to be done in understanding these mechanisms and the real impact of such combinations before arriving at a commercial use. Nevertheless, following a combined approach for “adjuvant antibiotherapy” and “combined antibiotherapy” will undeniably lead to a renewed concept of “complex drugs for complex diseases,” a well-known presupposed in traditional medicines [120].

Advertisement

Acknowledgments

The authors would like to thank ARES (Académie de Recherche et d’Enseignement Supérieur, Belgium) for financial support throughout PRD projects.

Advertisement

Conflict of interest

The authors declare that there is no conflict of interests regarding the publication of this paper.

References

  1. 1. Wu H-J, Wang AHJ, Jennings MP. Discovery of virulence factors of pathogenic bacteria. Current Opinion in Chemical Biology. 2008;12:93-101
  2. 2. Ciofu O, Tolker-Nielsen T. Tolerance and resistance of Pseudomonas aeruginosa biofilms to antimicrobial agents-how P. aeruginosa can escape antibiotics. Frontiers in Microbiology. 2019;10:913
  3. 3. Pashang R, Yusuf F, Zhao S, Deljoomanesh S, Gilbride KA. Widespread detection of antibiotic-resistant bacteria from natural aquatic environments in southern Ontario. Canadian Journal of Microbiology. 2018;65(4):322-331
  4. 4. Torres-Barceló C, Hochberg ME. Evolutionary rationale for Phages as complements of antibiotics. Trends in Microbiology. 2016;24(4):249-256. DOI: 10.1016/j.tim.2015.12.011
  5. 5. Valdivieso-Ugarte M, Gomez-Llorente C, Plaza-Díaz J, Gil Á. Antimicrobial, antioxidant, and Immunomodulatory properties of essential oils: A systematic review. Nutrients. 2019;11(11):2786. DOI: 10.3390/nu11112786
  6. 6. Rai M, Paralikar P, Jogee P, Agarkar G, Ingle AP, Derita M, et al. Synergistic antimicrobial potential of essential oils in combination with nanoparticles: Emerging trends and future perspectives. International Journal of Pharmaceutics. 2017;519(1-2):67-78. DOI: 10.1016/j.ijpharm. 2017.01.013
  7. 7. Cotter PD, Ross RP, Hill C. Bacteriocins- a viable alternative to antibiotics? Nature Reviews Microbiology. 2013;11(2):95-105. DOI: 10.1038/nrmicro2937
  8. 8. Nuti R, Goud NS, Saraswati AP, Alvala R, Alvala M. Antimicrobial peptides: A Promissing therapeutic strategy in tackling antimicrobial resistance. Current Medicinal Chemistry. 2017;24(38):4303-4314. DOI: 10.2174/0929867324666170815102441
  9. 9. Rutherford ST, Bassler BL. Bacterial quorum sensing: Its role in virulence and possibilities for its control. Cold Spring Harb, Perspect, Med. 2012;2:a012427
  10. 10. Turkina MV, Vikström E. Bacteria-host crosstalk: Sensing of the quorum in the context of Pseudomonas aeruginosa infections. Journal of Innate Immunity. 2019;11(3):263-279
  11. 11. Bricha S, Ounine K, Oulkheir S, Haloui N, Attarassi B. Virulence factors and epidemiology related to Pseudomonas aeruginosa. Tunisian Journal of Infectious Diseases. 2009;2:7-14
  12. 12. 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 Research International 2015; 2015: a759348
  13. 13. Filloux A, Vallet I. [biofilm: Set-up and organization of a bacterial community] article in french. Médecine Sciences. 2003;19:77-83. DOI: 10,1051/medsci/200319177
  14. 14. Vu B, Chen M, Crawford RJ, Ivanova EP. Bacterial extracellular polysaccharides involved in biofilm formation. Molecules. 2009;14(7):2535-2554. DOI: 10.3390/molecules14072535
  15. 15. Sauer K. The genomics and proteomics of biofilm formation. Genome Biology. 2003;4:219. DOI: 10.1186/gb-2003-4-6-219
  16. 16. Häussler S, Parsek MR. Biofilms 2009: New perspectives at the heart of surface-associated microbial communities. Journal of Bacteriology. 2010;192(12):2941-2949. DOI: 10.1128/JB.00332-10
  17. 17. Ghafoor A, Hay ID, Rehm BHA. Role of exopolysaccharides in Pseudomonas aeruginosa biofilm formation and architecture. Applied and Environmental Microbiology. 2011;77(15):5238-5246
  18. 18. Evans LR, Production LA. Characterization of the slime polysaccharide of Pseudomonas aeruginosa. Journal of Bacteriology. 1973;116(2):915-924
  19. 19. Hentzer M, Wu H, Andersen JB. Attenuation of Pseudomonas aeruginosa virulence by quorum sensing inhibitors. EMBO Journal. 2003;22(15):3803-3815
  20. 20. Wozniak DJ, Wyckoff TJO, Starkey M, Keyser R, Azadi P, O’Toole GA, et al. Alginate is not a significant component of the extracellular polysaccharide matrix of PA14 and PAO1 Pseudomonas aeruginosa biofilms. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(13):7907-7912
  21. 21. Stapper AP, Narasimhan G, Ohman DE, Barakat J, Hentzer M, Molin S, et al. Alginate production affects Pseudomonas aeruginosa biofilm development and architecture, but is not essential for biofilm formation. Journal of Medical Microbiology. 2004;53(7):679-690
  22. 22. Leid JG, Willson CJ, Shirtliff ME, Hassett DJ, Parsek MR, Jeffers AF. The exopolysaccharide alginate protects Pseudomonas aeruginosa biofilm Bacteria from IFN-γ-mediated macrophage killing. Journal of Immunology. 2005;175(11):7512-7518
  23. 23. Friedman L, Kolter R. Genes involved in matrix formation in Pseudomonas aeruginosa PA14 biofilms. Molecular Microbiology. 2004;51(3):675-690
  24. 24. Holloway BW, Krishnapillai V, Morgan AF. Chromosomal genetics of pseudomonas. Microbiological Reviews. 1975;43(1):73-102
  25. 25. Yang L, Liu Y, Wu H. Combating biofilms. FEMS Immunology & Medical Microbiology. 2012;65(2):146-157
  26. 26. Okshevsky M, Meyer RL. The role of extracellular DNA in the establishment, maintenance and perpetuation of bacterial biofilms. Critical Reviews in Microbiology. 2015;41(3):341-352
  27. 27. Swartjes JJTM, Das T, Sharifi S. A functional DNase coating to prevent adhesion of bacteria and the formation of biofilm. Advanced Functional Materials. 2013;23(22):2843-2849
  28. 28. Yang L, Barken KB, Skindersoe ME, Christensen AB, Givskov M, Tolker-Nielsen T. Effects of iron on DNA release and biofilm development by Pseudomonas aeruginosa. Microbiology. 2007;153(5):1318-1328
  29. 29. Gloag ES, Turnbull L, Huang A. Self-organization of bacterial biofilms is facilitated by extracellular DNA. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(28):11541-11546
  30. 30. Mulcahy H, Charron-Mazenod L, Lewenza S. Extracellular DNA chelates cations and induces antibiotic resistance in Pseudomonas aeruginosa biofilms. PLoS Pathogens. 2008;4(11):e1000213
  31. 31. Jimenez PN, Koch G, Thompson JA, Xavier KB, Cool RH, Quax WJ. The multiple signaling systems regulating virulence in Pseudomonas aeruginosa. Microbiology and Molecular Biology Reviews. 2012;76(1):46-65
  32. 32. Davies DG, Parsek MR, Pearson JP, Iglewski BH, Costerton JW, Greenberg EP. The involvement of cell-to-cell signals in the development of a bacterial biofilm. Science. 1998;280(5361):295-298
  33. 33. Gilbert KB, Kim TH, Gupta R, Greenberg EP, Schuster M. Global position analysis of the Pseudomonas aeruginosa quorum-sensing transcription factor LasR. Molecular Microbiology. 2009;73(6):1072-1085
  34. 34. Sakuragi Y, Kolter R. Quorum-sensing regulation of the biofilm matrix genes (pel) of Pseudomonas aeruginosa. Journal of Bacteriology. 2007;189(14):5383-5386
  35. 35. Davey ME, Caiazza NC, O’Toole GA. Rhamnolipid surfactant production affects biofilm architecture in Pseudomonas aeruginosa PAO1. Journal of Bacteriology. 2003;185(3):1027-1036
  36. 36. Pamp SJ, Tolker-Nielsen T. Multiple roles of biosurfactants in structural biofilm development by Pseudomonas aeruginosa. Journal of Bacteriology. 2007;189(6):2531-2539
  37. 37. Dusane DH, Zinjarde SS, Venugopalan VP, Mclean RJC, Weber MM, Rahman PKSM. Quorum sensing: Implications on Rhamnolipid biosurfactant production. Biotechnology and Genetic Engineering Reviews. 2010;27:159-184
  38. 38. O’Toole GA, Kolter R. Flagellar and twitching motility are necessary for Pseudomonas aeruginosa biofilm development. Molecular Microbiology. 1998;30(2):295-304
  39. 39. Boles BR, Thoendel M, Singh PK. Rhamnolipids mediate detachment of Pseudomonas aeruginosa from biofilms. Molecular Microbiology. 2005;57(5):1210-1223
  40. 40. Schooling SR, Charaf UK, Allison DG, Gilbert P. A role for rhamnolipid in biofilm dispersion. Biofilms. 2004;1:91-99
  41. 41. Diggle SP, Stacey RE, Dodd C, Camara M, Williams P, Winzer K. The galactophilic lectin, LecA, contributes to biofilm development in Pseudomonas aeruginosa. Environmental Microbiology. 2006;8(6):1095-1104
  42. 42. Tielker D, Hacker S, Loris R. Pseudomonas aeruginosa lectin LecB is located in the outer membrane and is involved in biofilm formation. Microbiology. 2005;151(5):1313-1323
  43. 43. Fraser GM, Hughes C. Swarming motility. Current Opinion in Microbiology. 1999;2(6):630-635
  44. 44. Daniels R, Vanderleyden J, Michiels J. Quorum sensing and swarming migration in bacteria. FEMS Microbiology Reviews. 2004;28(3):261-289
  45. 45. Shrout JD, Chopp DL, Just CL, Hentzer M, Givskov M, Parsek MR. The impact of quorum sensing and swarming motility on Pseudomonas aeruginosa biofilm formation is nutritionally conditional. Molecular Microbiology. 2006;62(5):1264-1277
  46. 46. Mattick JS. Type IV pili and twitching motility. Annual Review of Microbiology. 2002;56:289-314
  47. 47. Patriquin GM, Banin E, Gilmour C, Tuchman R, Greenberg EP, Poole K. Influence of quorum sensing and iron on twitching motility and biofilm formation in Pseudomonas aeruginosa. Journal of Bacteriology. 2008;190(2):662-671
  48. 48. Parkins MD, Ceri H, Storey DG. Pseudomonas aeruginosa GacA, a factor in multihost virulence, is also essential for biofilm formation. Molecular Microbiology. 2001;40(5):1215-1226
  49. 49. Merighi M, Lee VT, Hyodo M, Hayakawa J, 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. Molecular Microbiology. 2007;65(4):876-895
  50. 50. Dong YH, Zhang LH. Quorum sensing and quorum-quenching enzymes. The Journal of Microbiology. 2005;43(Spec):101-109
  51. 51. Huma N, Pratap S, Jyoti K, Ashish B, Jayadev J, Tanmoy M, et al. Diversity and polymorphism in AHL-Lactonase gene (aiiA) of bacillus. Journal of Microbiology and Biotechnology. 2011;21(10):1001-1011
  52. 52. Kalia VC, Purohit HJ. Quenching the quorum sensing system: Potential antibacterial drug targets. Critical Reviews in Microbiology. 2011;37(2):121-140
  53. 53. Kang Y, Durfee T, Glasner JD, Qiu Y, Frisch D, Winterberg KM, et al. Systematic mutagenesis of the Escherichia coli genome. Journal of Bacteriology. 2004;186:4921-4930
  54. 54. Park JK, Jung JY, Park YH. Cellulose production by Gluconacetobacter hansenii in a medium containing ethanol. Biotechnology Letters. 2003;25(24):2055-2059
  55. 55. Romero D, Aguilar C, Losick R, Kolter R. Amyloid fibers provide structural integrity to Bacillus subtilis biofilms. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:2230-2234
  56. 56. Uroz S, Dessaux Y, Oger P. Quorum sensing and quorum quenching : The Yin and Yang of bacterial communication. Chembiochem. 2009;10(2):205-216
  57. 57. Musthafa KS, Ravi AV, Annapoorani A, Packiavathy ISV, Pandian SK. Evaluation of anti-quorum-sensing activity of edible plants and fruits through inhibition of theN-acyl-homoserine lactone system in Chromobacterium violaceum and Pseudomonas aeruginosa. Chemotherapy. 2010;56:333-339
  58. 58. Nithya C, Pandian SK. The in vitro antibiofilm activity of selected marine bacterial culture supernatants against vibrio spp. Archives of Microbiology. 2010;192:10
  59. 59. Rehman ZU, Leiknes T. Quorum quenching bacteria isolated from Red Sea sediments reduces biofilm formation by Pseudomonas aeruginosa. Frontiers in Microbiology. 2018;9:1354
  60. 60. Nithya C, Begum MF, Pandian SK. Marine bacterial isolates inhibit biofilm formation and disrupt mature biofilms of Pseudomonas aeruginosa PAO1. Applied Microbiology and Biotechnology. 2010;88(1):341-358
  61. 61. Birmes FS, Säring R, Hauke MC, Ritzmann NH, Drees SL, Daniel J, et al. Interference with Pseudomonas aeruginosa quorum sensing and virulence by the mycobacterial pseudomonas quinolone signal Dioxygenase AqdC in combination with the N-Acylhomoserine lactone Lactonase QsdA. Infection and Immunity. 2018;87(10):e00278-e00219
  62. 62. Tettmann B, Niewerth C, Kirschhöfer F, Neidig A, Dötsch A, Brenner-Weiss G, et al. Enzyme-mediated quenching of the pseudomonas quinolone signal (PQS) promotes biofilm formation of Pseudomonas aeruginosa by increasing iron availability. Frontiers in Microbiology. 2016;7:1978
  63. 63. Cotar A, Chifiriuc MC, Dinu S, Pelinescu D, Banu O. Quantitative real-time PCR study of the influence of probiotic culture soluble fraction on the expression of Pseudomonas aeruginosa quorum sensing genes. Roumanian Archives of Microbiology and Immunology. 2010;69:213-223
  64. 64. Chifiriuc MC, Ditu ML, Banu O, Bleotu C, Dracea O. Subinhibitory concentrations of phenyl lactic acid interfere with the expression of virulence factors in Staphylococcus aureus and Pseudomonas aeruginosa clinical strains. Roumanian Archives of Microbiology and Immunology. 2009;68:27-33
  65. 65. Kiymaci ME, Altanlar N, Gumustas M, Ozkan SA, Akin A. Quorum sensing signals and related virulence inhibition of Pseudomonas aeruginosa by a potential probiotic strain’s organic acid. Microbial Pathogenesis. 2018;121:190-197
  66. 66. Rasmussen TB, Skindersoe ME, Bjarnsholt T. Identity and effects of quorum-sensing inhibitors produced by Penicillium species. Microbiology. 2005;151(5):1325-1340
  67. 67. Glasser N. Patulin: Mechanism of genotoxicity. Food and Chemical Toxicology. 2012;50(5):1796-1801. DOI: 10.1016/j.fct.2012.02.096
  68. 68. Shusaku S, Yoko M, Katsumi F, Nobuhiko F. Effects of long- term, low-dose macrolide treatment on Pseudomonas aeruginosa PAO1 virulence factors In vitro. Archives in Clinical Microbiology. 2017;8(4):50. DOI: 10.21767/1989-8436.100050
  69. 69. Skindersoe ME, Alhede M, Phipps R. Effects of antibiotics on quorum sensing in Pseudomonas aeruginosa. Antimicrobial Agents and Chemotherapy. 2008;52(10):3648-3663
  70. 70. Tateda K, Comte R, Pechere J-C, Köhler T, Yamaguchi K, van Delden C. Azithromycin inhibits quorum sensing in Pseudomonas aeruginosa. Antimicrobial Agents and Chemotherapy. 2001;45(6):1930-1933
  71. 71. Ichimiya T, Takeoka K, Hiramatsu K, Hirai K, Yamasaki T, Nasu M. The influence of azithromycin on the biofilm formation of Pseudomonas aeruginosa in vitro. Chemotherapy. 1996;42(3):186-191
  72. 72. Bala A, Kumar R, Harjai K. Inhibition of quorum sensing in Pseudomonas aeruginosa by azithromycin and its effectiveness in urinary tract infections. Journal of Medical Microbiology. 2011;60(3):300-306
  73. 73. Pérez-Martìnez I, Haas D. Azithromycin inhibits expression of the GacA-dependent small RNAs RsmY and RsmZ in Pseudomonas aeruginosa. Antimicrobial Agents and Chemotherapy. 2011;55(7):3399-3405
  74. 74. Pechère J-C. Azithromycin reduces the production of virulence factors in Pseudomonas aeruginosa by inhibiting quorum sensing. Japanese Journal of Antibiotics. 2001;54:87-89
  75. 75. Sofer D, Gilboa-Garber N, Belz A, Garber NC. ‘Subinhibitory’ erythromycin represses production of Pseudomonas aeruginosa lectins, autoinducer and virulence factors. Chemotherapy. 1999;45(5):335-341
  76. 76. Favre-Bonté S, Köhler T, van Delden C. Biofilm formation by Pseudomonas aeruginosa: Role of the C4-HSL cell-to-cell signal and inhibition by azithromycin. Journal of Antimicrobial Chemotherapy. 2003;52(4):598-604
  77. 77. Kim B, Park J-S, Choi HY, Yoon SS, Kim WG. Terrein is an inhibitor of quorum sensing and c-di-GMP in Pseudomonas aeruginosa: A connection between quorum sensing and c-di-GMP. Scientific Reports. 2018;8(1):8617
  78. 78. Merighi M, Lee VT, Hyodo M, Hayakawa Y, Lory S. The second messenger bis-(3′-5′)-cyclic-GMP and its PilZ domain containing receptorAlg44 are required for alginate biosynthesis in Pseudomonas aeruginosa. Molecular Microbiology. 2007;65(4):876-895
  79. 79. Silva LN, Zimmer KR, Macedo AJ, Trentin DS. Plant natural products targeting bacterial virulence factors. Chemical Reviews. 2016;116:9162-9236
  80. 80. Brackman G, Defoirdt T, Miyamoto C, Bossier P, Van Calenbergh S, Nelis H, et al. Cinnamaldehyde and cinnamaldehyde derivatives reduce virulence in vibrio spp, by decreasing the DNA-binding activity of the quorum sensing response regulator LuxR. BMC Microbiology. 2008;8:149. DOI: 10.1186/1471-2180-8-149
  81. 81. Niu C, Gilbert ES. Colorimetric method for identifying plant essential oil components that affect biofilm formation and structure. Applied and Environmental Microbiology. 2004;70(12):6951-6956
  82. 82. Niu C, Afre S, Gilbert ES. Subinhibitory concentrations of cinnamaldehyde interfere with quorum sensing. Letters in Applied Microbiology. 2006;43(5):489-494
  83. 83. Rudrappa T, Bais HP. Curcumin, a known phenolic from Curcuma longa, attenuates the virulence of Pseudomonas aeruginosa PAO1 in whole plant and animal pathogenicity models. Journal of Agricultural and Food Chemistry. 2008;56(6):1955-1962
  84. 84. Sarabhai S, Sharma P, Capalash N. Ellagic acid derivatives from Terminalia chebula Retz, Downregulate the expression of quorum sensing genes to attenuate Pseudomonas aeruginosa PAO1 virulence. PLoS One. 2013;8(1):e53441
  85. 85. Vandeputte OM, Kiendrebeogo M, Rajaonson S, Diallo B, Mol A, El Jaziri M, et al. Identification of catechin as one of the flavonoids from Combretum albiflorum bark extract that reduces the production of quorum-sensing-controlled virulence factors in Pseudomonas aeruginosa PAQ1. Applied and Environmental Microbiology. 2010;76(1):243-253
  86. 86. Luo J, Dong B, Wang K, Cai S, Liu T, Cheng X, et al. Baicalin inhibits biofilm formation, attenuates the quorum sensing-controlled virulence and enhances Pseudomonas aeruginosa clearance in a mouse peritoneal implant infection model. PLoS One. 2017;12(4):e0176883
  87. 87. Girennavar B, Cepeda ML, Soni KA. Grapefruit juice and its furocoumarins inhibits autoinducer signaling and biofilm formation in bacteria. International Journal of Food Microbiology. 2008;125(2):204-208
  88. 88. Chong YM, Yin WF, Ho CY, Mustafa MR, Hadi AH, Awang K. Malabaricone C from Myristica cinnamomea exhibits anti-quorum sensing activity. Journal of Natural Products. 2011;74:2261-2264
  89. 89. Zhou L, Zheng H, Tang Y, Yu W, Gong Q . Eugenol inhibits quorum sensing at sub-inhibitory concentrations. Biotechnology Letters. 2013;35(4):631-637
  90. 90. Lou Z, Letsididi KS, Yu F, Pei Z, Wang H, Letsididi R. Inhibitive effect of eugenol and its nanoemulsion on quorum sensing–mediated virulence factors and biofilm formation by pseudomonas aeruginosa. Journal of Food Protection. 2019;82(3):379-389
  91. 91. Ding X, Yin B, Qian L. Screening for novel quorum sensing inhibitors to interfere with the formation of Pseudomonas aeruginosa biofilm. Journal of Medical Microbiology. 2011;60(12):1827-1834
  92. 92. Kim H-S, Lee S-H, Byun Y, Park H-D. 6-Gingerol reduces Pseudomonas aeruginosa biofilm formation and virulence via quorum sensing inhibition. Scientific Reports. 2015;5:8656
  93. 93. Maisarah Norizan SN, Chan KG, Yin W-F, Ping TS, Nafiah MA. The study of caffeine as novel quorum sensing inhibitor. The Open Conference Proceedings Journal. 2013;4:185. DOI: 10.2174/22102892013040100185
  94. 94. Ren D, Zuo R, Gonzàlez-Barrios AF. Differential gene expression for investigation of Escherichia coli biofilm inhibition by plant extract ursolic acid. Applied and Environmental Microbiology. 2005;71(7):4022-4034
  95. 95. Kiplimo JJ, Koorbanally NA, Chenia HY. Triterpenoids from Vernonia auriculifera Hiern exhibit antimicrobial activity. African Journal of Pharmacy and Pharmacology. 2011;5:1150-1156
  96. 96. Rasamiravaka T, Vandeputte OM, Pottier L, Huet J, Rabemanantsoa C, Kiendrebeogo M, et al. Pseudomonas aeruginosa biofilm formation and persistence, along with the production of quorum sensing dependent virulence factors, are disrupted by a triterpenoid coumarate ester isolated from Dalbergia trichocarpa, a tropical “legume”. PLoS One. 2015;10:e0132791
  97. 97. Rasamiravaka T, Ngezahayo J, Pottier L, Oliveira Ribeiro S, Souard F, Hari L, et al. Terpenoids from Platostoma rotundifolium (Briq,) a, J, Paton Alter the expression of quorum sensing-related virulence factors and the formation of biofilm in Pseudomonas aeruginosa PAO1. International Journal of Molecular Sciences. 2017;18:1270
  98. 98. Jakobsen TH, Bragason SK, Phipps RK, Christensen LD, van Gennip M, Alhede M, et al. Food as a source for quorum sensing inhibitors: Iberin from horseradish revealed as a quorum sensing inhibitor of Pseudomonas aeruginosa. Applied Environmental Microbiology. 2012;78(7):2410-2421
  99. 99. Ganin H, Rayo J, Amara N, Levy N, Krief P, Meijler MM. Sulforaphane and erucin, natural isothiocyanates from broccoli, inhibit bacterial quorum sensing. Medicinal Chemistry Communications. 2013;4:175-179
  100. 100. Jakobsen TH, van Gennip M, Phipps RK. Ajoene, a sulfur-rich molecule from garlic, inhibits genes controlled by quorum sensing. Antimicrobial Agents and Chemotherapy. 2012;56(5):2314-2325
  101. 101. Cady NC, McKean KA, Behnke J. Inhibition of biofilm formation, quorum sensing and infection in Pseudomonas aeruginosa by natural products-inspired organosulfur compounds. PLoS One. 2012;7(6):e38492
  102. 102. Manefield M, de Nys R, Naresh K, Roger R, Givskov M, Peter S, et al. Evidence that halogenated furanones from Delisea pulchra inhibit acylated homoserine lactone (AHL)-mediated gene expression by displacing the AHL signal from its receptor protein. Microbiology. 1999;145(2):283-291
  103. 103. Skindersoe ME, Ettinger-Epstein P, Rasmussen TB, Bjarnsholt T, de Nys R, Givskov M. Quorum sensing antagonism from marine organisms. Marine Biotechnology. 2008;10(1):56-63
  104. 104. Manefield M, Rasmussen TB, Henzter M, Andersen JB, Steinberg P, Kjelleberg S, et al. Halogenated furanones inhibit quorum sensing through accelerated LuxR turnover. Microbiology. 2002;148(4):1119-1127
  105. 105. Hentzer M, Riedel K, Rasmussen TB. Inhibition of quorum sensing in Pseudomonas aeruginosa biofilm bacteria by a halogenated furanone compound. Microbiology. 2002;148(1):87-102
  106. 106. Ebada SS, Lin W, Proksch P. Bioactive sesterterpenes and triterpenes from marine sponges: Occurrence and pharmacological significance. Marine Drugs. 2010;8(2):313-346
  107. 107. Paul D, Kim YS, Ponnusamy K, Kweon JH. Application of quorum quenching to inhibit biofilm formation. Environmental Engineering Science. 2009;26(8):1319-1324
  108. 108. Chun CK, Ozer EA, Welsh MJ, Zabner J, Greenberg EP. Inactivation of a Pseudomonas aeruginosa quorum-sensing signal by human airway epithelia. Proceedings of the National Academy of Sciences. 2004;101(10):3587-3590
  109. 109. Ozer EA, Pezzulo A, Shih DM. Human and murine paraoxonase 1 are host modulators of Pseudomonas aeruginosa quorum-sensing. FEMS Microbiology Letters. 2005;253(1):29-37
  110. 110. Stoltz DA, Ozer EA, Ng CJ, Yu JM, Reddy ST, Lusis AJ, et al. Paraoxonase-2 deficiency enhances Pseudomonas aeruginosa quorum sensing in murine tracheal epithelia. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2007;292:852-860
  111. 111. Losa D, Kohler T, Bacchetta M, Saab JB, Frieden M, van Delden C, et al. Airway epithelial cell integrity protects from cytotoxicity of Pseudomonas aeruginosa quorum-sensing signals. American Journal of Respiratory Cell and Molecular Biology. 2015;53(2):265-275
  112. 112. Park J, Kaufmann GF, Bowen JP, Arbiser JM, Janda KD. Solenopsin a, a venom alkaloid from the fire ant Solenopsis invicta, inhibits quorum-sensing signaling in Pseudomonas aeruginosa. Journal of Infectious Diseases. 2008;198(8):1198-1201
  113. 113. Villena J, Kitazawa H, Van Wees SC, Pieterse CM, Takahashi H. Receptors and signaling pathways for recognition of bacteria in livestock and crops: Prospects for beneficial microbes in healthy growth strategies. Frontiers in Immunology. 2018;9:2223. DOI: 10.3389/fimmu.2018.02223
  114. 114. Ngezahayo J, Pottier L, Ribeiro SO, Delporte C, Fontaine V, Hari L, et al. Plastotoma rotundifolium aerial tissue extract has antibacterial activities. Industrial Crops and Products. 2016;86:301-310
  115. 115. Okusa PN, Penge O, Devleeschouwer M, Duez P. Direct and indirect antimicrobial effects and antioxidant activity of Cordia gilletii De wild (Boraginaceae). Journal of Ethnopharmacology. 2007;112(3):476-481
  116. 116. Okusa PN, Stévigny C, Névraumont M, Gelbcke M, Van Antwerpen P, Braekman J-C, et al. Ferulaldehyde and lupeol as direct and indirect antimicrobial compounds from Cordia gilletii (Boraginaceae) root barks. Natural Product Communications. 2014;9(5):619-622
  117. 117. Nobori T, Mine A, Tsuda K. Molecular networks in plant–pathogen holobiont. FEBS Letters. 2018;592(12):1937-1953
  118. 118. Ahmed SA, Rudden M, Smyth TJ, Dooley JS, Marchant R, Banat IM. Natural quorum sensing inhibitors effectively downregulate gene expression of Pseudomonas aeruginosa virulence factors. Applied Microbiology and Biotechnology. 2019;103(8):3521-3535
  119. 119. Cho HH, Kwon KC, Kim S, Park Y, Koo SH. Association between biofilm formation and antimicrobial resistance in carbapenem-resistant Pseudomonas aeruginosa. Annals of Clinical and Laboratory Science. 2018;48(3):363-368
  120. 120. Xu Q , Bauer R, Hendry BM, Fan T-P, Zhao Z, Duez P, et al. The quest for modernisation of traditional Chinese medicine. BMC Complementary and Alternative Medicine. 2013;13:132-143

Written By

Julie Carette, Amandine Nachtergael, Pierre Duez, Mondher El Jaziri and Tsiry Rasamiravaka

Submitted: 06 September 2019 Reviewed: 11 December 2019 Published: 29 January 2020