Europe PMC
Nothing Special   »   [go: up one dir, main page]

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Interference with quorum sensing (QS) represents an antivirulence strategy with a significant promise for the treatment of bacterial infections and a new approach to restoring antibiotic tolerance. Over the past two decades, a novel series of studies have reported that quorum quenching approaches and the discovery of quorum sensing inhibitors (QSIs) have a strong impact on the discovery of anti-infective drugs against various types of bacteria. The discovery of QSI was demonstrated to be an appropriate strategy to expand the anti-infective therapeutic approaches to complement classical antibiotics and antimicrobial agents. For the discovery of QSIs, diverse approaches exist and develop in-step with the scale of screening as well as specific QS systems. This review highlights the latest findings in strategies and methodologies for QSI screening, involving activity-based screening with bioassays, chemical methods to seek bacterial QS pathways for QSI discovery, virtual screening for QSI screening, and other potential tools for interpreting QS signaling, which are innovative routes for future efforts to discover additional QSIs to combat bacterial infections.

Free full text 


Logo of jpaLink to Publisher's site
J Pharm Anal. 2022 Feb; 12(1): 1–14.
Published online 2021 Apr 2. https://doi.org/10.1016/j.jpha.2021.03.009
PMCID: PMC9073242
PMID: 35573879

Screening strategies for quorum sensing inhibitors in combating bacterial infections

Abstract

Interference with quorum sensing (QS) represents an antivirulence strategy with a significant promise for the treatment of bacterial infections and a new approach to restoring antibiotic tolerance. Over the past two decades, a novel series of studies have reported that quorum quenching approaches and the discovery of quorum sensing inhibitors (QSIs) have a strong impact on the discovery of anti-infective drugs against various types of bacteria. The discovery of QSI was demonstrated to be an appropriate strategy to expand the anti-infective therapeutic approaches to complement classical antibiotics and antimicrobial agents. For the discovery of QSIs, diverse approaches exist and develop in-step with the scale of screening as well as specific QS systems. This review highlights the latest findings in strategies and methodologies for QSI screening, involving activity-based screening with bioassays, chemical methods to seek bacterial QS pathways for QSI discovery, virtual screening for QSI screening, and other potential tools for interpreting QS signaling, which are innovative routes for future efforts to discover additional QSIs to combat bacterial infections.

Keywords: Quorum sensing inhibitor, Quorum quenching, Anti-infective agent, Screening strategies

1. Introduction

The emergence of antimicrobial resistance constitutes a major global health care concern and poses a significant challenge. Conventional antibiotic therapies targeting the synthesis of protein, DNA, RNA, and folic acid usually exert bacteriostatic or bactericidal effects on multiple targets, resulting in strong selective pressure on bacterial communities and subsequently giving rise to bacterial strains resistant to conventional antibiotics [[1], [2], [3], [4], [5]]. In response to this, alternative strategies to antibiotics for bacterial infections are generating significant interest. Among these, some innovative alternative therapies, such as quorum sensing (QS) disruption, have been intensively studied [[6], [7], [8], [9], [10]]. In terms of the extensive impacts on microbial physiology induced by QS, interference with QS has been demonstrated to be a promising approach to decreasing bacterial virulence and restoring antibiotic effectivity by controlling biofilm production, paving the way for future anti-infective treatment and dressing/coating agents in medical applications [11,12].

In the 1970s, Nealson found that the density of Vibrio fischeri (V. fischeri) and Vibrio harveyi (V. harveyi) was positively correlated with their bioluminescence, and confirmed that this phenomenon is controlled by the QS system in bacteria (first described as QS) [13]. QS is an intercellular chemical communication process in a cell-density-dependent manner in which bacteria coordinate the expression of QS-mediated genes based on the exchange of small signaling molecules defined as quorum sensors or autoinducers (AIs).

Chemically, QS is based on the synthesis, sensing, and uptake of AIs [14]. Once a particular threshold concentration of bacteria is reached, programmed changes that coordinate biological effects including biofilm formation, virulence secretion, swarming ability, sporulation, and protease production are motivated in a density-dependent manner. The signal molecules related to clinical pathogens have been reported and are listed in Table 1 and Fig. 1: 1) acyl homoserine lactones (AHLs), which exist in most Gram-negative bacteria except Vibrio harringiensis and Mucoccus flavus; 2) autoinducer peptides (AIPs), which are produced by most Gram-positive bacteria; 3) autoinducer-2 (AI-2) furanones, which are utilized by more than 40 species of Gram-positive and Gram-negative bacteria for communication and transmission [15,16]; 4) autoinducer-3 (AI-3), which is used by enterohemorrhagic Escherichia coli. Although the AI-3 structure is still not finely characterized, it has been found that epinephrine or norepinephrine could be considered as an alternative to AI-3; and 5) other signal molecules such as the Pseudomonas quinolone signal (Pqs) and ketopyridazines, which also play important roles in intraspecific QS. Some biosynthetic pathways regulating the generation of important QS signal molecules such as AHL and AI-2 involve numerous receptors and enzymes, which are summarized as: S-adenosyl methionine-dependent methyltransferase, LuxS, AHL synthase, LuxR receptor, histidine kinase response regulated LuxN protein, and AI-2 (LuxP and LsrB) receptor. For example, LuxI/LuxR are key regulatory factors in AHL-mediated QS signaling pathways. The AHL synthase (LuxI type) synthesizes AHL derivatives to generate QS signal molecules. The synthesized AHL entities subsequently diffuse in the adjacent environment and accumulate in a cell-density-dependent manner (Fig. 2) [17]. When the AHL level reaches the threshold, the AHL interacts with the LuxR-type receptor to activate target gene transcription in QS signaling pathways [18,19].

Table 1

Classification of autoinducer molecules.

ClassificationSignal moleculesRegulatory factorsMicrobial sources
AHLsC4-HSLRhlR/SwrR/AhyR/AsaRPseudomonas aeruginosa/Serratia liquefaciens/Aeromonas hydrophila/Aeromonas salmonicida
C6-HSLCviR/CepR/SwrR/--/YtbR/PhzRChromobacterium violaceum/Burkholderia cenocepacia/Serratia liquefaciens/Pseudomonas aeruginosa/Yersinia pseudotuberculosis/Pseudomonas aureofaciens
C8-HSLCepR/AinsR/YpsR/CepRBurkholderia cenocepacia/Vibrio fischeri/Yersinia Pseudotuberculosis/B. cepacia
3-OH-C4-HSLLuxNVibrio harveyi
3-OXO-C6-HSLLuxR/Unkown/YpsR/YpeR/CarR/ExpR/EsaRVibrio fischeri/Pseudomonas aeruginosa/Yersinia pseudotuberculosis/Yersinia pestis/Erwinia carotovora/Erwinia chrysanthemi/Erwinia stewartii
3-OXO-C8-HSLTraRAgrobacterium tumefaciens
3-OXO-C10-HSLPpuR/Unkown//YspRPseudomonas putide/Pseudomonas aeruginosa/Yersinia pestis
3-OXO-C12-HSLLasRPseudomonas aeruginosa
7-cis-C14-HSLCerRRhodvacter sphaeroides
AIPsAIP-I, II, III, IVAgrCStaphylococcus aureus/Bacilus subtilis
iAM373Enterococcus faecalis
CSPComDStreptococcus mitis/Streptococcus constellatus/Streptococcus anginosus/Streptococcus thermophilus/Streptococcus milleri/Streptococcus gordonii/Streptococcus pneumoniae
cAM373, cAD1, cCF10, cPD1, iPD1, iAD1, CF10--Enterococcus faecalis
EDFEscherichia coli
CbnB2CbnKCarnobacterium piscicola
ComXRO-E-2Bacillus subtilis
AI-2AI-2LsrR/LsrR/TlpB, CagAVibrio harveyi/Escherichia coli/Helicobacter pylori
AI-2bLsrRVibrio harveyi
OthersPQSPqsRPseudomonas aeruginosa
DKP
IQSIqsRPseudomonas aeruginosa

−: not available. AHLs: acyl homoserine lactones; HSL: homoserine lactone; AIP: autoinducer peptides; CSP: competence stimulating peptide; EDF: extracellular death factor; CbnB2: camobacteriocin B2; AI-2: autoinducer-2; PQS: quinolones; DKP: diketopiperazines; IQS: integrated quorum sensing signal.

Fig. 1

Representation of AI molecules. (A) Acyl homoserine lactones; (B) autoinducer-2 furanones; (C) autoinducer peptides; (D) others. AIs: autoinducers; AIPs: autoinducer peptides; HSL: homoserine lactone; PQS: quinolones; DKP: diketopiperazines; IQS: integrated quorum sensing signal.

Fig. 2

Cyclic process of signal molecules. QS: quorum sensing.

In Gram-negative bacteria, many QS circuits are based on the LuxR-LuxI homologs of V. fischeri that use AHL signal molecules. For example, Pseudomonas aeruginosa (P. aeruginosa) has two key QS systems, LasR-LasI and RhlR-RhlI, which make use of 3-oxo-C12-homoserine lactone and C4-homoserine lactone (HSL) as signal molecules, respectively. The QS is also mediated through the Pqs system, which senses Pqs (2-heptyl-3-hydroxy-4-quinolone). These three systems are hierarchically presented, because the Las system is at the top and LasR controls Rhl and Pqs QS circuits. The Pqs system, which intervenes between the Las and Rhl systems, can interfere with the levels of Las- and Rhl-controlled genes. The Las and Rhl systems coordinate various virulence factors, including exoproteases, siderophores, toxins, and biofilm formation, and apparently exert an influence on host immune systems.

Interference with QS, termed quorum quenching (QQ), was first discovered as an enzyme capable of degrading AHL signals to interrupt QS signaling in Erwinia carotovora [20]. Specifically, QQ can be achieved by preventing bacteria from producing or perceiving AIs using quorum sensing inhibitors (QSIs) screened by different means [21], eliminating AIs by QQ antibodies, or extracellular enzymatic hydrolysis of AIs by QQ enzymes [22]. To date, many QQ enzymes, QSIs, and QQ antibodies have been reported and extensively reviewed [18,[23], [24], [25]]. Because QQ enzymes or antibodies can act remotely and independently without entering the bacterial cells to degrade AHL signals, they are probably less resistance-prone than QSIs, which have to interact with an intracellular target or a receptor on the bacterial outer membrane. However, stability remains a major constraint that usually limits the application of QQ enzymes and antibodies. Moreover, most QQ enzymes discovered such as lactonase are limited in quantity and variety [26]. Compared with that of QQ enzymes, the biochemical nature of purified QSIs, originating from a host of organisms including microbes, plants, fungi, and animals in ecosystems, is highly diverse [27]. For the discovery of QSIs, diverse approaches exist in-step with the scale of screening as well as specific QS systems. Among them, basic activity-based screening strategies consist of screening organisms, cells, and chemical libraries by QS bacterial reporters as well as QS-based phenotypes, including biofilm formation and virulence secretion. However, these natural QSIs may present a weak efficiency or possible toxicity in the targeted environment. A promising solution to overcoming these limitations is most likely to chemically design and synthesize molecules according to natural QSIs. Additionally, alternative strategies involve structure-based screening for target-oriented discovery of QSIs as well as other novel biotechnical tools for interpreting QS signaling. Overall, different methods, relying on the scale of screening as well as specific QS systems, are necessary for conducting QSI screening, which benefits the discovery and utilization of bioactive QSIs and validation of the effects of some QSI candidates. This review highlights the recent findings in strategies and methodologies for QSI screening, which are promising routes for QSI discovery to control bacterial biofilm formation and virulence to combat bacterial infection.

2. Activity-based screening with bioassays

In activity-based screening with bioassays, the detection systems rely on the testing signal generated by certain reporter proteins involved in colorimetric, bioluminescence, chemiluminescence, and fluorescence effects. Regarding QS bacterial reporters, two approaches are currently being applied, including those relying on the natural QS molecule-triggered responses (Fig. 3A) and by constructing engineered strains based on genetic modification (Fig. 3B).

Fig. 3

Activity-based screening with bioassays. (A) The construction of engineering strains based on the naturally observed activity; (B) The construction of engineering strains based on genetic modification. The detection signal produced by QS is observed via colorimetric, fluorimetric, bioluminescent, and chemiluminescent analyses.

2.1. Natural indicator strain for QSI screening

The easiest approaches to potential QSI screening involve colorimetric tests with natural indicator strains such as violacein formation in Chromobacterium violaceum (C. violaceum), bioluminescence in Vibrio Ferdinans and Vibrio Harringtonii, the prodigiosin produced by Serratia marcescens, and the orange promethazine pigments produced by Staphylococcus aureus (S. aureus) (Fig. 3A) [28]. More specifically, C. violaceum, which produces the visible purple pigment violacein regulated by the CviI/CviR system, is a frequently used sensor strain for QSI screening. In addition, V. harveyi is a typical biosensor that regulates an easily observed QS-mediated phenotype termed bioluminescence and has well-characterized QS networks. Recently, it was reported that metabolite extracts of Gram-positive bacteria from diverse marine environments inhibit the bioluminescence produced by V. harveyi.

These studies usually started with an initial screen using an indicator strain or the combined tests against two QS reporter strains to estimate the anti-QS activity of candidates [[29], [30], [31], [32], [33]]. Analysis of QS-controlled phenotypes, such as the virulence factor assay and biofilm formation assay, has significantly decreased the potential candidates.

However, quantitative analyses of bioluminescence and violacein interference do not seem to adequately demonstrate that these inhibitors specifically disrupt QS signaling pathways. Molecular observations, including gene expression and in silico molecular docking, as well as biochemical tests of inhibitor agents, will be needed to explore the mechanisms and action modes of the inhibitory effects.

Notably, for the preparation of extracts for QSI screening, chromatography analysis by liquid chromatography-mass spectrometry or high-performance liquid chromatography (HPLC) fractionation is utilized for the extraction or purification of anti-QS compounds. It is also expected that the extraction approach used for QSI isolation may not be optimal for all types of inhibitory compounds. In some cases, metabolites with high hydrophilicity may not be fully soluble in ethyl acetate. In addition, producing fermentation extracts or isolating metabolites for further biological evaluation is often a laborious process.

It has been found that many microbes generate metabolites or fermentation extracts that regulate QS-dependent phenotypes. In this regard, co-cultivation assays offer a rapid screening approach to testing bacteria that are chemically innovative for QSI discovery. In one instance, an Halobacillus salinus isolate C42 isolated from a co-cultivation assay termed Membrane Overlay was discovered to generate N-(2-phenylethyl)-isobutyramide, which effectively inhibited the QS phenotypes in several Gram-negative biosensors [34]. This active compound represents a promising chemotype for inhibitors of QS phenotypes. These co-cultivation assays were applied to screen a host of organisms to inhibit QS phenotypes.

2.2. Construction of engineering strains for QSI screening

However, these colorimetric assays with natural strains cannot meet the requirement of large-scale QSI screening. To promote large-scale screenings, the QSI selector system, including a plasmid that involves a construct composed of a QS homolog gene such as LasI, LasB, and RhlA, as well as a phenotype or function regulated by a QS-mediated promoter, has been extensively developed [[35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46]].

2.2.1. QSIs-LasI biosensor

The QSIs-LasI biosensor emerged as an innovative strategy for QSI discovery, and diverse QSIs were identified by virtue of this biosensor. Using the P. aeruginosa QSIs-LasI biosensor combined with analysis of QS-controlled phenotypes and HPLC, equisetin in secondary metabolites from the marine fungus Fusarium sp. Z10 and four new alpha-pyrones from the secondary metabolites of Streptomyces sp. were discovered as the bioactive anti-QS components [37,38]. In one instance, Wang et al. designed an efficient screening system involving a QSIs-LasI selector based on the PlasI-sacB reporter. Plasmid pMHLASI harboring PlasI-sacB and Plac-LasR transcriptional fusion was constructed and transformed into a P. aeruginosa LasI-RhlI double mutant PAO-MW1 to generate the selector QSIs-LasI. They tested the crude extracts from 65 marine fungi, and then an isolate, Penicillium Atramentosum QJ012, was selected to exhibit an inhibitory effect on the QS system. The thin-layer chromatography assay of the fungal extracts for bioautographic identification by QSIs-LasI demonstrated that the fungus produced several valuable compounds with anti-QS effects [39].

2.2.2. LasB-gfp and RhlA-gfp biosensor

The LasB-gfp and RhlA-gfp monitor strains contain an LasB promoter regulated by LasR, an RhlA promoter regulated by RhlR, and genes controlling an unstable green fluorescent protein. Using this biosensor, active compounds such as zeaxanthin and evernic acid were discovered to downregulate QS-related genes and inhibit biofilm formation in P. aeruginosa [40,41]. Rasmussen et al. [42] constructed three novel QSI selectors for QSI screening, including QSIs1 phlA encoding the toxic gene product, the expression of which was controlled by LuxR; QSIs2, the LasR- and RhlR-regulating LasB promoter controlled expression of the sacB gene, which led to cell death in the presence of sucrose; and the QSIs3 system, which contained the npt and gfp genes, conferring kanamycin resistance and green fluorescence, respectively. By virtue of these QSIs systems, two potential QSIs, garlic extract and 4-nitro-pyridine-N-oxide, were identified. In addition to in vitro phenotypic assays, GeneChip-based transcriptome analysis and a Caenorhabditis elegans pathogenesis model were also used to evaluate the specificity of QS-regulated virulence genes and virulence in vivo.

2.2.3. AHL-biosensor and others

The application of biosensors for the detection of the inhibitory activity in AHL-mediated QS has been instrumental in the rapid and streamlined screening of the QSI in a broad spectrum of microbes, especially Gram-negative bacteria. The realization that structurally different AHLs exist, possessing diversity in the acyl side chain and having different activation profiles, has prompted the construction of the corresponding biosensors. These reporters could be utilized in further studies to characterize the AHL signals in QS in a broad spectrum of organisms.

Using AHL-biosensors to screen out from the QSI chemical library and AHL-analogs as well as secondary metabolites produced by microbes, marine Streptomyces and Flustra foliacea, a large number of novel QSIs including N,N′-alkylated imidazolium-derivatives, and some secondary metabolites were identified as potential QSIs [[43], [44], [45]].

In another study, coumarin was identified as a potential QSI using a novel screening system composed of three biosensors (Serratia marcescens SP15, C. violaceum DSM30191, and Agrobacterium tumefaciens NTL4 containing a plasmid pZLR4 carrying a traG:lacZ reporter fusion) to detect short, medium, and long AHLs, respectively. Subsequently, coumarin was demonstrated to significantly suppress biofilm formation and protease activity in other microbes and inhibit bioluminescence in Aliivibrio fischeri [46]. All cases mentioned above are shown in Table 2 [[29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46]].

Table 2

Activity-based screening with bioassays.

Strain sourceCompoundsMain routesTarget bacteriaTargets/pathwaysAnti-QS effectsRefs.
Natural indicator strainIsoprenyl caffeateViolacein inhibition assay;
Differential gene expression assay;
In silico docking experiments;
LC-MS; HPLC
C. violaceum
ATCC 12472
vioABCDE operon, involved in violacein biosynthesisInhibition in the violacein biosynthesis as a competitor in VioA and VioD.[29]
Extracts of Tremella fuciformisQuantifying violacein productionC. violaceum CV026[30]
SCS-KFD08Quantitative analysis of violacein productionC. violaceum CV026Inhibitory effects on QS and violacein production[31]
Culture supernatants of a Pseudomonas sp.Virulence factor assay;
Biofilm formation assay;
HPLC
C. violaceum CV026Rhl systemInhibitory effects on Rhl-controlled pyocyanin, rhamnolipid, elastase, protease, and biofilm[32]
Phenethylamides and a cyclic dipeptideTest colony on top assay;
Membrane overlay assay;
Bioassay-guided fractionation;
Lactonase gene analysis
V. harveyi
C. violaceum
Inhibitory effects on bioluminescence production against V. harveyi and violacein production by C. violaceum[33]
Phenethylamide metabolitesCocultivation experiment;
Bioassay-guided fractionation;
Detection of lactonase genes;
Mathematical modeling
V. harveyi
C. violaceum CV026
E. coli JB525
Acting as antagonists of QSInterfering with the bioluminescence, biofilm formation, motility, antibiotic biosynthesis, and virulence production[34]
Construction of engineering strainsLyngbic acidQS assays with V. harveyi JMH626 (ΔluxN luxQ::Tn5 cqsA::Cm) and V. harveyi KM413 (ΔluxS ΔluxM);
16S Illumina tag sequencing Luminescence
V. harveyiFunctioning as a natural antagonist of the CqsS/CAI-1 QS systemInhibitory effect on luminescence and CAI-1-mediated QS[35]
Substitution of AHL moleculesConstruction of QS luxCDABE reporters;
Luminometry
E. coliLuxR, LasR, and RhlRThese sensors can be used to characterize the AHL signal(s) involved in QS[36]
EquisetinQSI screening;
Motility assays;
Transcriptional activity;
Assays for virulence factor production; and biofilm formation; RT-PCR
P. aeruginosaLas, Rhl, and Pqs systemsSuppression in the biofilm formation, swarming motility, and the production of virulence factors[37]
α-pyrones from metabolites of Streptomyces sp.Diversity-oriented synthesis;
Anti-QS activity assay
P. aeruginosaLasIInhibitory effects on the QS-regulated gene expression in P. aeruginosa QSIs-LasI[38]
The crude extracts from 65 marine fungiAgar well-diffusion assay
Bioautographic TLC
P. aeruginosaPlasI-sacB reporterThis fungus produced several QSI compounds other than penicillic acid or patulin[39]
ZeaxanthinMonitor strains (LasB-gfp and RhlA-gfp); qRT-PCR;
In silico molecular docking
P. aeruginosaLas and Rhl systemsDownregulation in the QS-related genes and inhibitory effects on biofilm formation[40]
Evernic acidQSI screenings by LasB-gfp or RhlA-gfp monitor strain; qRT-PCRP. aeruginosaLasB and RhlAQS systems (LasIR and RhlIR) of P. aeruginosa were regulated[41]
Garlic extract and 4-nitro-pyridine-N-oxideQSIs assays; TLC assay; GeneChip-based transcriptome analysis;
Biofilm formation assay;
Caenorhabditis elegans nematode model
P. aeruginosaPhlA, LasR, and RhlRThese QSIs reduced biofilm tolerance to antibiotics and virulence[42]
Secondary metabolites of Flustra foliaceaInhibition assay with AHL-sensor strains including Pseudomonas putida (pKR-C12), P. putida (pAS-C8) and E.coli (pSB403);
Measurement of protease activity;
GC-MS; HPLC
P. aeruginosa PAO1
P. fischeri
LasB: elastase gene;
LasR: encodes the cognate N-3-oxododecanoyl-l-homoserine lactone receptor; cepR: sensitive for N-octanoyl-l-homoserine lactone; luxR: highest sensitivity for the P. fischeri signal N-oxohexanoyl-l-homoserine lactone;
Specifically blocking AHL-regulated gene expression and reducing protease activity[43]
Secondary metabolites produced by marine streptomycesQSI screening; Biofilm production assay; Scanning electron microscopy;
Assessment of virulence factors motility, hemolysin, and urease activity
Proteus mirabilis (uropathogen)Synthesis inhibition by blocking the AHL synthase LuxI-type proteins; Interference with the signal receptors or blocking the formation of AHL/LuxR complexInhibiting QS-regulated prodigiosin biosynthesis and the QS-dependent factors[44]
N,N'-alkylated imidazolium-derivativesPectobacterium AHL-biosensors;
Virulence ssays
P. atrosepticumExpI: encodes the synthase for the biosynthesis of the AHL-signals; RsmA: downregulated by AHLsTargeting virulence genes and decreasing the severity of the symptoms provoked by P. atrosepticum[45]
CoumarinQQ biosensor assay;
β-Galactosidase assay;
Phenazine production;
Assays for biofilm formation and motility;
Bioluminescence of A. fischeri;
Determination of protease activity
A broad spectrum of bacterial strainsRhlI promoter modulating RhlI/R-associated biosurfactant rhamnolipids;
PqsA promoter in Pqs signaling system controlling the production of phenazine
LuxI/R QS system controlling bioluminescence
Presenting abroad spectrum of activity against kinds of AHLs; Inhibiting biofilm, phenazine production and swarming motility[46]

--: not available. TLC: thin-layer chromatography; QS: quorum sensing; QSI: quorum sensing inhibitor; RT-PCR: real time-polymerase chain reaction; GC-MS: gas chromatography-mass spectrometry; HPLC: high performance liquid chromatography; CAI: cholera autoinducer.

2.3. Other activity-based bioassays

Other series of activity-based assays use known QS-mediated mechanisms for functional differentiation. The QS-regulated functions tested in these bioassays involve motility, pyocyanin or pyoverdin production, and biofilm formation in P. aeruginosa. In this regard, similar steps are applied in the preliminary stage, including diffusing potential QSIs into agar; measuring the area of pigment inhibition, which is visualized by the formation of a colorless but visible halo around the well; and the method of microbulion dilution, which quantitatively detects the level of pigment by measuring the optical density using a spectrophotometer [47,48]. Numerous studies have described the discovery of QSIs based on biofilm formation. Commonly used qualitative methods to evaluate the characteristics of the biofilm in these cases involve Congo red agar, the microtiter plate assay, or the crystal violet assay.

Another novel approach used for QSI discovery is proteomic analysis, which is a two-step procedure involving an initial quantitative 2D-difference gel electrophoresis and subsequent protein identification in each spot by mass spectrometry. This method has been proven to be beneficial for the examination of the impact of various factors on a range of proteomic and metabolomic changes mediated by QS in bacteria [49].

Transcriptome analysis provides deep insights into QS modulation in response to anti-QS compounds [50,51]. Comparing real-time polymerase chain reaction used in small-scale genes for QSI screening in early studies, this transcriptome analysis based on high-throughput sequencing methods achieved more quantitative analysis of comprehensive transcription profiles. The analysis included 1) total RNA extraction of bacterial samples, 2) RNA reverse transcription into a complementary DNA library for transcriptome sequencing, and 3) comparative analysis of differentially expressed genes by various genetic techniques. The subsequent differential expression analysis by bioinformatics approaches is beneficial for revealing and finally characterizing global gene expression patterns in response to anti-QS compounds.

In vivo models are widely used to evaluate the efficiency of potential QSIs for antivirulence activity. The Caenorhabditis elegans model achieves in vivo high-throughput screening (HTS) and provides a better understanding of QS modulation and virulence regulation [52]. Additionally, mammalian models with general immune systems are also commonly employed to evaluate the anti-QS activities of potential QSIs on bacterial infections [53].

3. Chemical approaches to interrogating QS pathways for QSI discovery

3.1. Solid-phase synthesis for the identification of small molecule QS modulators by interaction with their target proteins/receptors

Each compound yielded by traditional synthesis strategies requires individual purification, identification, and bio-activity evaluation, rendering the acquisition process cumbersome and time-consuming. Unlike traditional synthesis methods, the update of combinatorial chemistry involving solid-phase synthesis significantly accelerates chemical synthesis and screening. Solid-phase synthesis, in which the reactant molecule is chemically bound to an insoluble material while reagents are added in the solution phase, is extensively applied in the high-throughput synthesis of biological molecules such as peptides, nucleic acids, oligosaccharides, and QSIs [54].

Geske et al. [[55], [56], [57]] constructed a library of AHL-analogs using a solid-phase synthesis technique. They adopted a novel synthetic strategy containing a series of microwave-assisted reactions followed by a CNBr cyclative-cleavage procedure on the polystyrene beads. Furthermore, through systematic screening of these analogs, numerous antagonists were identified to competitively bind to the LuxR-type protein. These compounds were found to attenuate QS-mediated phenotypes in bacterial culture as well as in eukaryotic host infections.

Novel solid-phase synthesis techniques such as macroarrays and microarrays were studied for the discovery of QSIs [[58], [59], [60], [61], [62], [63], [64]]. In small molecule macroarrays, libraries of varying sizes were formed by the spatial synthesis of small molecules on planar polymeric supports. This platform, with advantageous properties such as economics, stability, and compatibility, was proven to be a practical approach to the large-scale synthesis of small molecules including QSIs [65,66].

Not limited to macroarrays, microarray techniques were also investigated and utilized for the combinatorial synthesis of QSIs [67]. Sharing the analogous underlying principle with macroarray synthesis, the size and diversity of microarrays are apparently smaller and simpler [68,69]. Moreover, macroarrays and microarrays simultaneously provide platforms not limited to chemical synthesis, but many on-support biological tests are involved, further facilitating the discovery of bioactive anti-QS molecules.

3.2. Affinity chromatography approaches for identifying QS receptors based on ligand–receptor interactions

Not limited to synthetic AI analogs as QS modulators, ligand–receptor interactions in QS signaling have also been studied for target identification of QS receptors. Affinity chromatography based on ligand–receptor interactions, with its exquisite specificity, has been proven to be a valuable tool for the purification and identification of biomolecules. In affinity chromatography, one molecule is immobilized to a support, whereas its binding counterpart is soaked in the mobile phase. Under alternate conditions, molecular recognition between the two molecules allows the counterpart to be isolated from the eluent and separated from the support. This highly selective separation approach is of significant value in the identification, isolation, purification, pretreatment, or analysis of many biologically relevant molecules.

To identify AHL-binding receptors, Spandl et al. [70] synthesized biotin-labeled N-3-oxo-hexanoyl l-homoserine lactone (OHHL). The biotin tag, with powerful affinity and recognition to avidin, permits the ligand OHHL to be readily immobilized to an avidin-functionalized support. Biological tests and 2D-difference gel electrophoresis have also been pursued to investigate the target for OHHL. In a similar work, affinity resins were utilized to identify putative receptors for AHLs in eukaryotic cells. Two piperazine-modified N-(3-oxo-dodecanoyl)-l-homoserine lactone (OdDHL) derivatives were synthesized and linked to agarose resin [71]. Subsequently, two proteins were successfully obtained by affinity assay by virtue of these OdDHL-modified resins from mammalian cell lysates. Such resins in affinity-based methods could be ultimately valuable for studying novel AHL-binding proteins.

3.3. Antibodies for quenching QS signaling

Another approach that has gained considerable attention for interrupting QS signaling is to apply antibodies to quench QS by competing with or interrupting QS signal molecules [72]. The AIs or derivatives thereof are delivered into an animal to activate monoclonal antibody formation. The yielded antibodies, which are substantially specific for the AI molecules, could catalytically inactivate or effectively sequester AIs by acting as the QS quenchers, resulting in the QS signaling interruption [73]. Usually, generating antibodies such as QQ needs a molecular template (or hapten) to immunize. Small QS molecules such as AHLs, with hydrolytic instability and nonproteinaceous nature, cannot complete antibody-based immune reactions by themselves because they are unlikely to be appropriately presented by the mammalian immune system. The antibody-based strategy has promising applications in future vaccine research and development.

In recent years, numerous studies have reported the design and evaluation of QQ antibodies to sequester AIs in vitro and in vivo [72,74]. Using an immunotherapeutic strategy for QQ, Janda and co-workers [75] pioneered eliciting immune responses against the synthetic 3-oxo-AHL analog RS2 in mice, and isolated an anti-AHL antibody (RS2-1G9). The RS2-1G9 efficiently inhibited QS phenotypes and QS pathways in P. aeruginosa by sequestering 3-oxo-C12-HSL. Furthermore, they utilized this approach to raise antibodies against a similar analog of AIP-IV. In this case, the native thioester in the AIP macrocycle was more hydrolytically stable using an ester bond. The antibody obtained, AP4-24H11, was shown to be an active QSI in S. aureus cultures [76].

Additionally, antibody-based QQ also involved other strategies, such as raising catalytic antibodies for degrading and thus inactivating AIs. Marin et al. [77] utilized this approach to screen and evaluate catalytic antibodies for lactonase activity. An antibody (XYD-11G2) capable of hydrolyzing OdDHL from an antibody library was identified and demonstrated to suppress QS in P. aeruginosa cultures.

3.4. Biotic polymers/polymeric materials

Materials-based approaches have also gained considerable attention for QS modulation. The first strategy is the sequestration of the AI signal by non-native polymeric materials. By computational modeling and prior affinity studies, Piletska et al. [78] selected a series of monomers with binding capacity with an AHL signal named OHHL produced in V. fischeri. Polymers based on one monomer, named itaconic acid (IA), showed higher affinity to OHHL and thus suppressed QS of V. fischeri. Subsequently, they generated a polymer production composed of a template molecule and polymers synthesized by utilizing molecularly imprinted polymers [79]. The formed IA-based polymer production served as high-specificity binding pockets to sequester QS signaling and biofilm formation in P. aeruginosa. In another study, 2-hydroxypropyl-β-cyclodextrin (HP-β-CD) was reported to downregulate the levels of QS-controlled genes in Serratia marcescens and P. aeruginosa. Then, cross-linked HP-β-CD was utilized to form sheets of 2-hydroxypro-pylcellulose to form an AHL-sequestering material. Subsequently, these HP-β-CD-linked sheets were reported to suppress the levels of QS-dependent genes when co-cultured with the two abovementioned strains.

Another strategy is to utilize polymer materials for the spatial accumulation of QS modulators and thus to prevent the release of QS modulators into the surroundings. Poly lactide-co-glycolide is employed as a material to regulate the release and delivery of AHLs. Both the AHL concentration and the time of AHL release could be readily controlled without affecting its biological activity [80]. These studies indicated a novel material-based strategy for modulating QS signaling that can be applied in a series of contexts (e.g., biofilm prevention and coatings on the surface of clinical implants). All those mentioned above are shown in Table 3 [56,57,64,66,69,[76], [77], [78],80].

Table 3

QSI discovery by chemical techniques.

CompoundsRoutesTarget bacteriaReceptorsAnti-QS effectsRefs.
90 non-native AHLsA solid-phase synthetic routeA. tumefaciens
P. Aeruginosa
V. fischeri
TraR
LasR
LuxR
Showing agonistic or antagonistic activity in all species[56]
Majority of the natural AHLs and a small test library of non-natural AHLsA solid-phase synthetic route to both natural and non-natural AHLsP. aeruginosa,
R. leguminosarum
Y. pseudotuberculosis
B. pseudomallei,
S. meliloti,
R. capsulatus, V. fischeri
A. tumefaciens,
V. anguillarum
TraR
LasR
LuxR
Antagonizing LuxR-type protein and TraR[57]
A 39-member library EA microwave-assisted synthetic route to AHLsA. tumefaciens,
P. aeruginosa,
V. fischeri
TraR
LasR LuxR
Antagonists of TraR and LuxR[64]
Macrocyclic peptide-peptoid hybrids (peptomers) as analogs of AIP-IA solid-phase synthetic route includes microwave-assisted reactions followed by a tandem macrocyclization-cleavage stepS. aureusAgrC receptorStimulating biofilm formation[66]
A number of QS analogs3D microarray platform: 3D microarray slides were probed with fluorescently labeled ligand-binding domains of the LuxR homolog CarRSerratia
P. aeruginosa.
LuxR homolog CarRAs potent inhibitors of AHL mediated QS phenotypes in Serratia and P.aeruginosa.[69]
3-oxo-AHL analogue RS2 and AP4-24H11An immunotherapeutic strategy for QQ to elicit immune responses against the synthetic 3-oxo-AHL analogue RS2 and against a close analogue of AIP-IV in miceP. aeruginosa
S. aureus
Sequestration of 3-oxo-C12-HSL, efficiently inhibition in QS phenotypes and QS signaling in vitro.[76]
XYD-11G2Screening catalytic antibodies in an existing library of antibodies known to catalyze the hydrolysis of the insecticide paraoxonP. aeruginosaCapable of hydrolyzing OdDHL and suppression of QS in P. aeruginosa cultures[77]
A set of rationally designed polymers with affinity toward a signal molecule of V. fischeriFunctional monomers were selected based on the computational modeling. All polymers were prepared by thermal polymerizationV. fischeriComputationally designed polymers could sequester a signal molecule of V. fischeri and prevent QS-controlled phenotypes[78]
A synthetic AHL modulator of bacterial QSA polymer-based approach to the release of a synthetic AHLV. fischeriModulating QS in the marine symbiont V. fischeri[80]

--: not available. AIP-I: autoinducing peptide I; QQ: quorum quenching; OdDHL: N-(3-oxo-dodecanoyl)-l-homoserine lactone.

4. Virtual screening for QSI screening

As described above, limited QSIs have been identified using traditional methods from natural resources. Computer-assisted screenings, especially structure-based virtual screening (SB-VS), have been developed as an efficient paradigm for lead discovery that fits in well alongside HTS to promote large-scale screenings, which can complement traditional methods.

The SB-VS approach, which is used extensively in the pharmaceutical industry, has also been applied for novel QSI discovery that antagonized AHL sensing in recent years: 1) against Las, Rhl, and Pqs systems: the screening of natural compounds and recognized drugs/compounds by in-silico docking analysis and SB-VS in P. aeruginosa [81,82]; 2) against the LasR and RhlR receptor proteins: the identification of five active compounds using an SB-VS of 1920 natural compounds [83]; 3) against LasR receptors: two compounds discovered by virtual screening and pharmacophore- and structure-based approaches and six novel potential QSIs screened with parameters including Lipinski's rule, and toxicity as well as absorption, distribution, metabolism, excretion (ADME) from the ZINC database [84,85]; 4) with the crystal structure of LuxP: the screening of cinnamaldehyde derivatives [86], boronic acid derivatives [87], and traditional Chinese medicine (TCM) databases [88] by SB-VS; 5) other targets/systems: the identification of baicalein in 51 bioactive components from TCMs against TraR protein [89], the discovery of eight lead-like molecules from the ZINC database by combined analysis including virtual screening, molecular docking as well as ADME and toxicity studies against FabI which plays the important role of enoyl-acyl carrier protein reductase related to the synthesis of 3-oxo-C12 HSL [90], the screening of most plant compounds and all NSAIDs to find the Z-phytol and lonazolac molecules which can suppress the SdiA as a homolog of LuxR in the QS pathway [91].

To sum up, SB-VS approaches represent an economical and rapid alternative to the traditional methods and HTS approaches. Screening strategies for SB-VS can be limited to commercially available compound libraries to avoid time-consuming and expensive chemical syntheses and utilized to dock compounds from known drugs/natural product libraries to circumvent problems in complicated testing stages, owing to toxicity or poor ADME properties, which would substantially reduce the costs, compared to conventional resource-consuming HTS processes. However, SB-VS approaches, relying on the prediction by the docking software, have one major problem of false positives and false negatives. As more novel and advanced algorithms are developed, false hits may be minimized. Moreover, experimental validation is required to combine SB-VS for in vitro inhibition efficacy.

The SB-VS approaches promote the identification of targeted QSIs, and future directions include extending SB-VS to more novel targets and performing combined tests with quantitative structure-activity relationship studies to promote rational drug design. All cases mentioned above are shown in Table 4 [[81], [82], [83], [84], [85], [86], [87], [88], [89], [90], [91]].

Table 4

Virtual screening for QSI screening.

SystemCompoundsRoutesTargets/pathwaysBioactive componentsTarget bacteriaAnti-QS effectsRefs.
Las, Rhl and Pqs3040 natural compounds and their derivativesSB-VS; Live reporter assay for QS; (iTRAQ)-based proteomic analysis;
Molecular docking; Elastase assay;
AHL competition assay; Glass-slide biofilm assay
LasR and LasB: controlled and encoded elastase; Pqs system: regulates release of extracellular DNA5-imino-4,6-dihydro-3H-1,2,3-triazolo[5,4-d] pyrimidin-7-oneP. aeruginosa
E. coli
Inhibition of Rhl system and Pqs system; Efficient in inhibiting elastase production and eDNA release in P. aeruginosa biofilms[81]
147 recognized drugs/compoundsSB-VS; Molecular docking;
Measurement of virulence factors and biofilm development
LasR, RhlA, PqsASalicylic acid, nifuroxazide, and chlorzoxazoneP. aeruginosa.Significant, dose-dependent inhibition of QS-controlled gene expression and phenotypes[82]
Las and Rhl1,920 natural compounds/drugsSB-VS; Molecular docking studies;
Measurement of virulence factors production; Biofilm inhibition assay;
Swarming assay; Protein degradation assay
LasR and RhlR receptors in Las and Rhl dependent virulence factors productionRosmarinic acid, naringin, chlorogenic acid, morin, and mangiferinP. aeruginosa.Inhibit biofilm related behaviors and virulence factors production[83]
Las122 compounds identified via in silico screeningVirtual screening;
Pharmacophore- and structure-based approaches;
QS inhibition and activation via cell-based assays
LasR, also bind to RhlR and TraRTwo compounds, named ZINC 2060666 and ZINC 2989037P. aeruginosa.Showing in vitro QS inhibition[84]
About 2,603 compounds from ZINC databaseVirtual screening;
Lipinski's rule, ADME and toxicity studies;
Molecular docking
LasRSix novel potential QS inhibiting compoundsP. aeruginosa.[85]
LuxCinnamaldehyde derivativesMolecular docking; e-Pharmacophore based virtual screening;
Bioluminescence and biofilm inhibition assay;
Swimming and swarming assays
LuxR: cognate receptor of AI
LuxI/LuxR QS system controlling a wide range of cellular processes
3-(2,4-dichlorophenyl)-1-(1H-pyrrol-2-yl)-2-propen-1-oneV. harveyiInhibitory effects on the bioluminescence production in a dose dependent manner; Inhibition in biofilm formation and motility in V. harveyi[86]
Five boronic acid derivativesThree docking protocols: RRD, IFD, and QPLD;
Virtual Screening;
Bioluminescence Inhibition Assay;
Biofilm Inhibition Assay;
Swimming and Swarming
LuxP: periplasmic binding protein (LuxP) binds to AI-2 to activates the biosynthetic pathway that is responsible for the production of AIsBicyclo[2,2,1]hept-5-ene-2,3-dicarboxylic acid-2,6-dimethylpyridine 1-oxideV. harveyiDose-dependent inhibition in bioluminescence and biofilm formation[87]
A TCM databaseVirtual screening and molecular docking;
QS inhibitory assay;
Production of extracellular enzymes and siderophores; Swimming and swarming motility;
Impact on AHL and EPS content
LuxI-and LuxR-type proteins
LuxI/LuxR QS system trolling a wide range of cellular processes
Benzyl alcohol, rhodinyl formate and houttuynineP. fluorescens P07Inhibitory effects on swimming and swarming motility, production of extracellular enzymes and siderophores, AHL content and biofilm formation[88]
Others51 bioactive components from TCMsDegradation of TraR protein;
Biofilm formation
The signal receptor TraRBaicaleinP. aeruginosa
E.coli
Inhibiting biofilm formation of P. aeruginosa; Promotion in proteolysis of the signal receptor TraR protein in E.coli[89]
75 natural compounds from the ZINC databaseVirtual screening;
ADME and toxicity studies;
Molecular docking
FabI: the significant role of enoyl-acyl carrier protein reductase in the synthesis of 3-oxo-C12 HSLEight lead-like moleculesP. aeruginosa.[90]
Most plant compounds and all NASIDsMolecular docking; Biofilm formation; Prediction of absorption, solubility and permeability of evaluated compoundsSdiA (a homolog of LuxR)The Z-phytol and lonazolac moleculesSalmonellaInhibition of QS mediated by AI-1 and biofilm formation in Salmonella[91]

--: not available. EPS: extracellular polymeric substances; NSAIDs: nonsteroidal anti-inflammatory drugs; TCM: traditional Chinese medicine; ADME: absorption, distribution, metabolism, excretion; SB-VS: structure-based virtual screening; RRD: rigid receptor docking; IFD: induced fit docking; QPLD: quantum polarized ligand docking.

5. Other potential tools for interpreting QS signaling

5.1. Three-dimensional (3D) printing

A microscopic 3D printing strategy was established to organize multiple populations of bacteria within essentially any 3D geometry. Using this approach, a distinct core–shell arrangement, which was composed of a single species at vastly differing densities as well as polymicrobial communities including motile and non-motile bacteria, was found in microbial populations. For example, Connell et al. [92] also demonstrated that a picoliter-volume aggregate of S. aureus had the ability to produce substantial resistance to β-lactam antibiotics by nesting a unique core–shell arrangement enclosed with a shell composed of P. aeruginosa.

5.2. Immobilized hybrids

Using an operationally simple dip-and-rinse procedure, Gomes et al. [93] synthesized natural product hybrids featuring an AHL inducer as well as a nitrodopamine and subsequently immobilized the hybrids onto biocompatible TiO2 surfaces. The formed immobilized hybrids were demonstrated as effective QS activators by gradually releasing them from the TiO2 surface. This approach is a straightforward and useful strategy for interrupting the QS signaling pathway by the preparation of coated surfaces in medical devices in a wide variety of bacterial species.

5.3. Biological nanofactories

Hebert et al. [94] constructed a biological nanofactory to regulate the behaviors of bacteria on the surface of human epithelial cells. The biological nanofactory construct activated the surface to produce the bacterial QS signaling molecule, AI-2. Biological nanofactories include several functional subassemblies: the cell targeting module with the targeting element CD26 antibody, the fabrication, and the synthesis modules. The biosynthesis component implemented is the fusion protein His6-protein G-LuxS-Pfs-Tyr5 (HGLPT). The HGLPT consisted of both synthesis and fabrication domains, which facilitated flexible self-assembly of the functional units. The HGLPT utilized the substrate S-adenosylhomocysteine to synthesize AI-2 in a two-step process using enzymes such as Pfs and LuxS. This tool would be beneficial for modulating signaling activities associated with the microbiome present in the human gastrointestinal tract and other environments.

6. Challenges in QSI discovery and some criteria for QSIs

To date, many recently authorized patents focused on the discovery of new bioactive agents, analogs, and approaches interfering with QS signaling highlight the application of QS inhibition in the fields of plant pathogens, aquaculture, antifouling, and medical devices (Table 5) [[95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111]]. However, very few QSI compounds have reached human clinical trials. Until now, QSI therapeutic studies for clinical QS-induced bacterial infections are still undergoing preclinical phases and have not been extensively developed. In total, there are only a few clinical trials on azithromycin (a QSI for preventing P. aeruginosa ventilator-associated pneumonia and treatment of cystic fibrosis) (Table 6) [112]. Some beneficial QSI effects, such as the ability to prevent biofilm formation, bacterial virulence, and innovative medical devices equipped with QSI molecules have attracted the most attention. Various inhibitors/agents capable of QS-controlled biofilm formation are ongoing clinical evaluations (Table 7) [112] and have been published with outcomes [[113], [114], [115], [116], [117], [118]]. Moreover, QSIs were primarily designed for functionalizing catheters. The increasing range of QSIs fuel efforts to develop innovative medical devices and diversify their applications.

Table 5

Recently authorized patents on the discovery of new bioactive agents, analogs and approaches interfering with QS signaling.

YearInventionDescription/MethodologyApplicationRefs.
2020Compounds that affect QS in S. aureus and related Staphylococcus speciesModulation includes inhibition or activation of one or more of four AgrC receptorsIn combination with one or more antibiotics to treat bacterial infections[95]
2019A novel brominated furanone derivativeAn effective inhibitory activity of the biofilm formation and QSUseful effects on oral diseases or inflammatory diseases, e.g., periodontal diseases[96]
2019Methods and compositions for the inhibition of biofilm formationA method for the inhibition of biofilm formation with a bifunctional ligand comprising a QS-peptide-binding region and a protease-binding regionInhibition of biofilm formation on the surface[97]
2019Applications of 3,4,5-methyl trihydroxybenzoate in inhibition of the activity of a bacterial QS systemA compound capable of significantly reducing the expression of related pathogenic factors in P. aeruginosa and C. violaceumControlling drug resistance[98]
2019d-galactose in QS inhibitionA composition for inhibiting QSInhibiting QS and treating oral bacterial diseases[99]
2018A preparation method and application of a camphor essential oil-based bacterial QSIThe preparation method comprises preparing essential oil and preparing the bacterial QSI which is a novel antibacterial substance based on bacterial QS inhibition in E.coli, purple bacillus, S. aureus and P. aeruginosaThe raw material is easy to obtain and the preparation method of the novel bacterial QSI is simple and reliable; the bacteriostatic agent does not generate drug resistance and has no toxic effects[100]
2018A QSI comprising at least one of wood particlesWood particles from trees and grassAs feed additive for antibiotic-free prophylaxis of infectious diseases and modulating body temperature under heat stress conditions in farm animals[101]
2018A trackable moiety can be attached to a QS molecule to form a QS modulating conjugateQS modulating conjugates retain their activity for QS manipulation and are able to be detected by imaging techniquesDiagnostic applications by enabling pinpointing of specific bacteria at infection sites[102]
2018Banana pseudostem-based liquid extractsResults in disc diffusion method showed that antibacterial and QS inhibition activity in the extracts, especially in the autoclaved aqueous formsAs an antimicrobial against E. coli, Salmonella typhimurium and S. aureus, and as a QS inhibition agent against P. aeruginosa.[103]
2018A synergist for food biological preservatives and a method of the synergistThe combination of biofilm degrading enzyme and a QSI to interfere with the biofilm formation of food putrefying bacteria and inhibit the QS information exchange between putrefying bacteriaThe target spot of the synergist is the biofilm and QS system of the putrefying bacteria, and the action site is accurate, not a lethal effect[104]
2017Application of pyrimidine derivative in preparing medicine for inhibiting a bacterial QS systemThe pyrimidine derivative can inhibit QS signal molecules to finally reduce bacterial biofilm formation and effectively inhibit pathogenicity of virulence factorsComputer virtual screening is a compound screening method with high efficiency and low cost in studying QS medicine[105]
2017Novel apicidin methods and compositions for the QS inhibitionTreating a staphylococcal infection[106]
2017The modulation of the flora of bacteria in an environment by inhibiting the QS of a specific bacteria by administering a QS control compositionQS control agents include a sorbent material, sorbent mineral or non-porous mineral such as phyllosilicate clays, silica, calcite, zeolites, diatomaceous earth, smectite, activated carbon, a nanoparticle or a combination of any of the foregoingInhibiting the spoilage of food stuff and preventing vibriosis in fish or shell fish[107]
2017Methods for modulating QS in certain Gram-negative bacteria having multiple QS systems including Las, Rhl, and Pqs with associated receptors (LasR, RhlR, and PqsR)Certain combinations of Las, Rhl, and Pqs exhibit improved inhibitory effects on virulenceModulating QS in Pseudomonas and Buckholderia[108]
2017A novel application of lotus plumule extracts in the preparation of QS inhibitory drugsThe lotus plumule extracts provide good inhibition for both P. aeruginosa and drug-resistant P. aeruginosa.Decreasing bacterial virulence and pathogenicity and controlling drug resistance[109]
2017Synthetic cyclic peptide modulators of the AgrC QS system of S.epidermidisCompounds capable of either pan-group or group-selective AgrC receptor inhibition in S. epidermidis were identifiedTreating infections of S. epidermidis and related Staphylococcus by administering a therapeutically effective amount of one or more compounds herein to an individual in need thereof[110]
2017Certain compounds of general formula A-W-HG having various carbocyclic and heterocyclic head groups and various tail groupsThe compounds are useful in methods of modulating QS in Gram-negative bacteria, particularly in Pseudomonas. Compositions including certain RhlR modulators are useful for decreasing the virulence of Gram-negative bacteriaPharmaceutical compositions comprising certain RhlR modulators are useful for treatment of infections of Gram-negative bacteria[111]

--: not available.

Table 6

Quorum-sensing inhibitors in clinical evaluation [112].

NCT No.TitleCondition or diseaseInterventionsPhaseYearStatus
NCT00610623Azithromycin as a QSI for the prevention of P. aeruginosa ventilator-associated pneumoniaPneumonia, ventilator- associated
Pseudomonas infections
Drug: azithromycin
Drug: placebo
II2008Terminated
NCT01201577Biological modulation of bacterial QSSMs, innate and adaptive immunity by antibiotics, probiotics and prebiotics in healthy individualsQS
Prebiotics
Probiotics
Sepsis
Dietary supplement: Bifidobacterium longum BB536
Dietary supplement: active hexose correlated compound
Dietary supplement: Bifidobacterium longum BB536 and active hexose correlated compound
Dietary supplement: corn starch placebo capsule
Drug: azithromycin
I2011Completed

QSSM: quorum sensing signaling molecules.

Table 7

Recent anti-biofilm agents under clinical evaluation [112].

NCT No.Condition or diseaseStatusInterventionPhaseYear
NCT04254835High caries risk patientsRecruitingDrug: cervitec F, ivoclar vivadent - Schaan Liechtenstein
Drug: fluor protector, ivoclar vivadent - Schaan Liechtenstein
IV2020
NCT03683563End-stage kidney disease;
Renal dialysis;
Central venous catheter; Biofilms
UnknownOther: 4% sodium citrate
Other: 30% sodium citrate
2018
NCT03686904Wound infectionActive, not recruitingDrug: benzalkonium gel
Other: standard of care topical gel
Procedure: debridement
Drug: benzalkonium irrigation
Other: saline irrigation (SOC irrigation)
IV2018
NCT03678012Dental cariesCompletedDrug: ferumoxytol/hydrogen peroxide
Drug: hydrogen peroxide;
Drug: water
Early I2018
NCT03213249Breast implant infection;
Mammoplasty
CompletedOther: normal saline; Drug: cefazolin
Procedure: skin biopsy
Procedure: bilateral skin- or nipple-sparing mastectomies
Device: tissue expander; Device: breast implant
Procedure: autologous flap
Other: acellular dermal matrix sling
Drug: gentamicin
Drug: bacitracin
I2017
NCT02946801Biofilms;
Essential oils;
Periodontitis
Not yet recruitingDrug: essential oils
Drug: essential oils without alcohol
Drug: sterile water
IV2016
NCT02946814Biofilms;
Substantivity;
Essential oils,
UnknownDrug: essential oils
Drug: Eessential oils without mouthwash
Drug: sterile water
IV2016
NCT03146390Oral biofilm;
Dental plaque;
Periodontitis
RecruitingDrug: essential oils
Drug: alcohol free essential oils
Other: water
IV2017
NCT02052973Streptococcal infections;
Saliva altered
CompletedOther: propolis varnishI and II2015
NCT02545244GingivitisCompletedDietary supplement: black tea
Dietary supplement: green tea
Drug: 0.12% chlorhexidine mouthwash
III2015
NCT02656251Dental plaqueCompletedDrug: 0.12% clorhexidine with alcohol
Drug: placebo
Drug: 0.12% clorhexidine without alcohol
III2015
NCT02486458Dental cariesCompletedDrug: 5% sodium fluoride varnish
Drug: 1.23% sodium fluoride acidic gel
Early I2015

--: not available. SOC: soil organic carbon.

Unlike antibiotics, QSIs act by interfering with QS-mediated gene expression controlling virulence traits that are not necessary for bacterial growth, which probably produces a limited selective pressure to develop bacterial resistance. However, some studies have pointed out the risk of inducing selective pressure and developing resistance in QS disruption conditions by anti-QS agents [119,120]. However, the literature manifestly indicates that resistance to QS inhibition may emerge, but likely at a much lower level than that which conventional antibiotics could induce with their massive use. To date, there have been few reports regarding the potential QQ-related resistance mechanisms that various bacteria could induce to support this possibility, but it cannot be completely ignored.

At present, only a few QSIs were finely clarified for the anti-QS mechanisms supporting the related anti-QS activity at a subcellular level and functionally evaluated regarding their biological role in vivo. In this regard, QSIs may not be as active as antibiotics, and are in realistic conditions, which becomes a major bottleneck for their potential for antibacterial treatments to reach clinical trials. In some cases, QSIs have been described as having potential medicinal effects on bacterial communities in the human body [121]. The impacts on the hose microbiome were quite likely to influence human health and induce metabolic disturbance [122].

Overall, to prevent the negative influence on hosts and minimize the development of QSI resistance, some criteria for QSIs should be considered: 1) low molecular weight; 2) high stability and specificity; 3) no side effects on hosts and no interference with the host microbiome; and 4) unlikely to develop drug resistance.

7. Conclusions

Over the past two decades, numerous studies have reported that QS circuitry and potential QSIs have significant impact as anti-infective agents against a host of bacteria. The discovery of QSIs and their initial success represent a promising target for the design of novel anti-infective drugs. The discovery of QSIs is appealing in large part to expand the anti-infective therapeutic arsenal to complement classical antibiotics and antimicrobial agents. With regard to QSI discovery, some problems that need to be solved, including the QSI molecular target identification, the molecular targeting delivery, the QSI cytotoxicity evaluation at the organism, cellular, and molecular levels. These queries on the above points are certainly promising trails for further efforts to discover QSIs and diversify their applications.

CRediT author statement

Lan Lu: Project administration, Funding acquisition, Writing - Original draft preparation, Supervision; Mingxing Li: Investigation, Writing - Reviewing and Editing; Guojuan Yi and Li Liao: Conceptualization, Methodology, Software; Qiang Cheng, Jie Zhu and Bin Zhang: Conceptualization, Investigation; Yingying Wang, Yong Chen and Ming Zeng: Visualization.

Declaration of competing interest

The authors declare that there are no conflicts of interest.

Acknowledgments

This study was funded by the National Natural Science Foundation of China (Grant No.: 81803812).

Footnotes

Peer review under responsibility of Xi'an Jiaotong University.

References

1. Olsen I. Biofilm-specific antibiotic tolerance and resistance. Eur. J. Clin. Microbiol. Infect. Dis. 2015;34:877–886. [Abstract] [Google Scholar]
2. Cho H., Uehara T., Bernhardt T.G. Beta-lactam antibiotics induce a lethal malfunctioning of the bacterial cell wall synthesis machinery. Cell. 2014;159:1300–1311. [Europe PMC free article] [Abstract] [Google Scholar]
3. Mina G., Chbib C. Recent progresses on synthesized LuxS inhibitors: a mini-review. Bioorg. Med. Chem. 2019;27:36–42. [Abstract] [Google Scholar]
4. Levy S.B., Marshall B. Antibacterial resistance worldwide: causes, challenges and responses. Nat. Med. 2004;10:S122–S129. [Abstract] [Google Scholar]
5. Roy V., Adams B.L., Bentley W.E. Developing next generation antimicrobials by intercepting AI-2 mediated quorum sensing. Enzyme Microb. Technol. 2011;49:113–123. [Abstract] [Google Scholar]
6. Vadakkan K., Choudhury A.A., Gunasekaran R., et al. Quorum sensing intervened bacterial signaling: pursuit of its cognizance and repression. J. Genet. Eng. Biotechnol. 2018;16:239–252. [Europe PMC free article] [Abstract] [Google Scholar]
7. Subhadra B., Kim D.H., Woo K., et al. Control of biofilm formation in healthcare: recent advances exploiting quorum-sensing interference strategies and multidrug efflux pump inhibitors. Materials (Basel) 2018;11:1676. [Europe PMC free article] [Abstract] [Google Scholar]
8. Soukarieh F., Williams P., Stocks M.J., et al. Pseudomonas aeruginosa quorum sensing systems as drug discovery targets: current position and future perspectives. J. Med. Chem. 2018;61:10385–10402. [Abstract] [Google Scholar]
9. Manner S., Fallarero A. Screening of natural product derivatives identifies two structurally related flavonoids as potent quorum sensing inhibitors against Gram-negative bacteria. Int. J. Mol. Sci. 2018;19:1346. [Europe PMC free article] [Abstract] [Google Scholar]
10. Haque S., Ahmad F., Dar S.A., et al. Developments in strategies for quorum sensing virulence factor inhibition to combat bacterial drug resistance. Microb. Pathog. 2018;121:293–302. [Abstract] [Google Scholar]
11. He X., Lu F., Yuan F., et al. Biofilm formation caused by clinical Acinetobacter baumannii isolates is associated with overexpression of the AdeFGH efflux pump. Antimicrob. Agents Chemother. 2015;59:4817–4825. [Europe PMC free article] [Abstract] [Google Scholar]
12. Defoirdt T. Quorum-sensing systems as targets for antivirulence therapy. Trends Microbiol. 2018;26:313–328. [Abstract] [Google Scholar]
13. Nealson K.H. Autoinduction of bacterial luciferase. Occurrence, mechanism and significance. Arch. Microbiol. 1977;112:73–79. [Abstract] [Google Scholar]
14. Papenfort K., Bassler B.L. Quorum sensing signal-response systems in Gram-negative bacteria. Nat. Rev. Microbiol. 2016;14:576–588. [Europe PMC free article] [Abstract] [Google Scholar]
15. Wynendaele E., Bronselaer A., Nielandt J., et al. Quorumpeps database: chemical space, microbial origin and functionality of quorum sensing peptides. Nucleic Acids Res. 2013;41:D655–D659. [Europe PMC free article] [Abstract] [Google Scholar]
16. Verbeke F., De Craemer S., Debunne N., et al. Peptides as quorum sensing molecules: measurement techniques and obtained levels in vitro and in vivo. Front. Neurosci. 2017;11:183. [Europe PMC free article] [Abstract] [Google Scholar]
17. Welsh M.A., Blackwell H.E. Chemical probes of quorum sensing: from compound development to biological discovery. FEMS Microbiol. Rev. 2016;40:774–794. [Europe PMC free article] [Abstract] [Google Scholar]
18. Kumar M., Saxena M., Saxena A.K., et al. Recent breakthroughs in various antimicrobial resistance induced quorum sensing biosynthetic pathway mediated targets and design of their inhibitors. Comb. Chem. High Throughput Screen. 2020;23:458–476. [Abstract] [Google Scholar]
19. Holm A., Vikström E. Quorum sensing communication between bacteria and human cells: signals, targets, and functions. Front. Plant Sci. 2014;5:309. [Europe PMC free article] [Abstract] [Google Scholar]
20. Dong Y.H., Xu J.L., Li X.Z., et al. AiiA, an enzyme that inactivates the acylhomoserine lactone quorum-sensing signal and attenuates the virulence of Erwinia carotovora. Proc. Natl. Acad. Sci. U S A. 2000;97:3526–3531. [Europe PMC free article] [Abstract] [Google Scholar]
21. Tang K., Zhang X.-H. Quorum quenching agents: resources for antivirulence therapy. Mar. Drugs. 2014;12:3245–3282. [Europe PMC free article] [Abstract] [Google Scholar]
22. Fetzner S. Quorum quenching enzymes. J. Biotechnol. 2015;201:2–14. [Abstract] [Google Scholar]
23. Lu L., Hu W., Tian Z., et al. Developing natural products as potential anti-biofilm agents. Chin. Med. 2019;14:11. [Europe PMC free article] [Abstract] [Google Scholar]
24. Kalia V.C., Patel S.K., Kang Y.C., et al. Quorum sensing inhibitors as antipathogens: biotechnological applications. Biotechnol. Adv. 2019;37:68–90. [Abstract] [Google Scholar]
25. Nandi S. Recent advances in ligand and structure based screening of potent quorum sensing inhibitors against antibiotic resistance induced bacterial virulence. Recent Pat. Biotechnol. 2016;10:195–216. [Abstract] [Google Scholar]
26. Bzdrenga J., Daudé D., Rémy B., et al. Biotechnological applications of quorum quenching enzymes. Chem. Biol. Interact. 2017;267:104–115. [Abstract] [Google Scholar]
27. Grandclément C., Tannières M., Morera S., et al. Quorum quenching: role in nature and applied developments. FEMS Microbiol. Rev. 2016;40:86–116. [Abstract] [Google Scholar]
28. Duran N., Justo G.Z., Duran M., et al. Advances in Chromobacterium violaceum and properties of violacein-its main secondary metabolite: a review. Biotechnol. Adv. 2016;34:1030–1045. [Abstract] [Google Scholar]
29. Gemiarto A.T., Ninyio N.N., Lee S.W., et al. Isoprenyl caffeate, a major compound in manuka propolis, is a quorum-sensing inhibitor in Chromobacterium violaceum. Antonie Van Leeuwenhoek. 2015;108:491–504. [Abstract] [Google Scholar]
30. Zhu H., Sun S.J. Inhibition of bacterial quorum sensing-regulated behaviors by Tremella fuciformis extract. Curr. Microbiol. 2008;57:418–422. [Abstract] [Google Scholar]
31. Kong F.D., Zhou L.M., Ma Q.Y., et al. Metabolites with Gram-negative bacteria quorum sensing inhibitory activity from the marine animal endogenic fungus Penicillium sp. SCS-KFD08. Arch. Pharm. Res. 2017;40:25–31. [Abstract] [Google Scholar]
32. Zhang Y., Yang Y., Wang L., et al. Identification of a Pseudomonas sp. that inhibits RHL system of quorum sensing. Indian J. Microbiol. 2013;53:28–35. [Europe PMC free article] [Abstract] [Google Scholar]
33. Teasdale M.E., Donovan K.A., Forschner-Dancause S.R., et al. Gram-positive marine bacteria as a potential resource for the discovery of quorum sensing inhibitors. Mar. Biotechnol. (NY) 2011;13:722–732. [Abstract] [Google Scholar]
34. Teasdale M.E., Liu J., Wallace J., et al. Secondary metabolites produced by the marine bacterium Halobacillus salinus that inhibit quorum sensing-controlled phenotypes in gram-negative bacteria. Appl. Environ. Microbiol. 2009;75:567–572. [Europe PMC free article] [Abstract] [Google Scholar]
35. Meyer J.L., Gunasekera S.P., Scott R.M., et al. Microbiome shifts and the inhibition of quorum sensing by black band disease cyanobacteria. ISME J. 2016;10:1204–1216. [Europe PMC free article] [Abstract] [Google Scholar]
36. Winson M.K., Swift S., Fish L., et al. Construction and analysis of luxCDABE-based plasmid sensors for investigating N-acyl homoserine lactone-mediated quorum sensing. FEMS Microbiol. Lett. 1998;163:185–192. [Abstract] [Google Scholar]
37. Zhang M., Wang M., Zhu X., et al. Equisetin as potential quorum sensing inhibitor of Pseudomonas aeruginosa. Biotechnol. Lett. 2018;40:865–870. [Abstract] [Google Scholar]
38. Du Y., Sun J., Gong Q., et al. New α-pyridones with quorum-sensing inhibitory activity from diversity-enhanced extracts of a Streptomyces sp. derived from marine algae. J. Agric. Food Chem. 2018;66:1807–1812. [Abstract] [Google Scholar]
39. Wang L., Zou S., Yin S., et al. Construction of an effective screening system for detection of Pseudomonas aeruginosa quorum sensing inhibitors and its application in bioautographic thin-layer chromatography. Biotechnol. Lett. 2011;33:1381–1387. [Abstract] [Google Scholar]
40. Gökalsin B., Aksoydan B., Erman B., et al. Reducing virulence and biofilm of Pseudomonas aeruginosa by potential quorum sensing inhibitor carotenoid: zeaxanthin. Microb. Ecol. 2017;74:466–473. [Abstract] [Google Scholar]
41. Gökalsin B., Sesal N.C. Lichen secondary metabolite evernic acid as potential quorum sensing inhibitor against Pseudomonas aeruginosa, World J. Microbiol. Biotechnol. 2016;32:150. [Abstract] [Google Scholar]
42. Rasmussen T.B., Bjarnsholt T., Skindersoe M.E., et al. Screening for quorum-sensing inhibitors (QSI) by use of a novel genetic system, the QSI selector. J. Bacteriol. 2005;187:1799–1814. [Europe PMC free article] [Abstract] [Google Scholar]
43. Peters L., König G.M., Wright A.D., et al. Secondary metabolites of Flustra foliacea and their influence on bacteria. Appl. Environ. Microbiol. 2003;69:3469–3475. [Europe PMC free article] [Abstract] [Google Scholar]
44. Younis K.M., Usup G., Ahmad A. Secondary metabolites produced by marine streptomyces as antibiofilm and quorum-sensing inhibitor of uropathogen Proteus mirabilis. Environ. Sci. Pollut. Res. Int. 2016;23:4756–4767. [Abstract] [Google Scholar]
45. des Essarts Y.R., Sabbah M., Comte A., et al. N,N'-alkylated Imidazolium-derivatives act as quorum-sensing inhibitors targeting the Pectobacterium atrosepticum-induced symptoms on potato tubers. Int. J. Mol. Sci. 2013;14:19976–19986. [Europe PMC free article] [Abstract] [Google Scholar]
46. Gutiérrez-Barranquero J.A., Reen F.J., McCarthy R.R., et al. Deciphering the role of coumarin as a novel quorum sensing inhibitor suppressing virulence phenotypes in bacterial pathogens. Appl. Microbiol. Biotechnol. 2015;99:3303–3316. [Abstract] [Google Scholar]
47. Deryabin D., Galadzhieva A., Kosyan D., et al. Plant-derived inhibitors of AHL-mediated quorum sensing in bacteria: modes of action. Int. J. Mol. Sci. 2019;20:5588. [Europe PMC free article] [Abstract] [Google Scholar]
48. Joshi C., Kothari V., Patel P. Importance of selecting appropriate wavelength, while quantifying growth and production of quorum sensing regulated pigments in bacteria. Recent Pat. Biotechnol. 2016;10:145–152. [Abstract] [Google Scholar]
49. Swatton J.E., Davenport P.W., Maunders E.A., et al. Impact of azithromycin on the quorum sensing-controlled proteome of pseudomonas aeruginosa. PloS One. 2016;11 [Europe PMC free article] [Abstract] [Google Scholar]
50. Ahmed S., Rudden M., Smyth T.J., et al. Natural quorum sensing inhibitors effectively downregulate gene expression of Pseudomonas aeruginosa virulence factors. Appl. Microbiol. Biotechnol. 2019;103:3521–3535. [Europe PMC free article] [Abstract] [Google Scholar]
51. Asfahl K.L., Schuster M. Additive effects of quorum sensing anti-activators on Pseudomonas aeruginosa virulence traits and transcriptome. Front. Microbiol. 2018;8:2654. [Europe PMC free article] [Abstract] [Google Scholar]
52. Kong C., Eng S.A., Lim M.P., et al. Beyond traditional antimicrobials: a Caenorhabditis elegans model for discovery of novel anti-infectives. Front. Microbiol. 2016;7:1956. [Europe PMC free article] [Abstract] [Google Scholar]
53. van der Worp H.B., Howells D.W., Sena E.S., et al. Can animal models of disease reliably inform human studies? PLoS Med. 2010;7 [Europe PMC free article] [Abstract] [Google Scholar]
54. Boyle N.A., Janda K.D. Formats for combinatorial synthesis: solid-phase, liquid-phase and surface. Curr. Opin. Chem. Biol. 2002;6:339–346. [Abstract] [Google Scholar]
55. Geske G.D., O'Neill J.C., Miller D.M., et al. Modulation of bacterial quorum sensing with synthetic ligands: systematic evaluation of N-acylated homoserine lactones in multiple species and new insights into their mechanisms of action. J. Am. Chem. Soc. 2007;129:13613–13625. [Europe PMC free article] [Abstract] [Google Scholar]
56. Geske G.D., Wezeman R.J., Siegel A.P., et al. Small molecule inhibitors of bacterial quorum sensing and biofilm formation. J. Am. Chem. Soc. 2005;127:12762–12763. [Abstract] [Google Scholar]
57. Geske G.D., Mattmann M.E., Blackwell H.E. Evaluation of a focused library of N-aryl L-homoserine lactones reveals a new set of potent quorum sensing modulators. Bioorg. Med. Chem. Lett. 2008;18:5978–5981. [Europe PMC free article] [Abstract] [Google Scholar]
58. Lin Q., Blackwell H.E. Rapid synthesis of diketopiperazine macroarrays via Ugi four-component reactions on planar solid supports. Chem. Commun. (Camb) 2006:2884–2886. [Europe PMC free article] [Abstract] [Google Scholar]
59. Lin Q., O'Neill J.C., Blackwell H.E. Small molecule macroarray construction via Ugi four-component reactions. Org. Lett. 2005;7:4455–4458. [Abstract] [Google Scholar]
60. Bowman M.D., Jeske R.C., Blackwell H.E. Microwave-accelerated SPOT-synthesis on cellulose supports. Org. Lett. 2004;6:2019–2022. [Abstract] [Google Scholar]
61. Bowman M.D., Jacobson M.M., Blackwell H.E. Discovery of fluorescent cyanopyridine and deazalumazine dyes using small molecule macroarrays. Org. Lett. 2006;8:1645–1648. [Abstract] [Google Scholar]
62. Praneenararat T., Palmer A.G., Blackwell H.E. Chemical methods to interrogate bacterial quorum sensing pathways. Org. Biomol. Chem. 2012;10:8189–8199. [Europe PMC free article] [Abstract] [Google Scholar]
63. Praneenararat T., Geske G.D., Blackwell H.E. Efficient synthesis and evaluation of quorum-sensing modulators using small molecule macroarrays. Org. Lett. 2009;11:4600–4603. [Europe PMC free article] [Abstract] [Google Scholar]
64. Blackwell H.E. Hitting the SPOT: small-molecule macroarrays advance combinatorial synthesis. Curr. Opin. Chem. Biol. 2006;10:203–212. [Abstract] [Google Scholar]
65. Fowler S.A., Stacy D.M., Blackwell H.E. Design and synthesis of macrocyclic peptomers as mimics of a quorum sensing signal from Staphylococcus aureus. Org. Lett. 2008;10:2329–2332. [Abstract] [Google Scholar]
66. Walsh D.P., Chang Y.T. Recent advances in small molecule microarrays: applications and technology. Comb. Chem. High Throughput Screen. 2004;7:557–564. [Abstract] [Google Scholar]
67. Marsden D.M., Nicholson R.L., Ladlow M., et al. 3D small-molecule microarrays. Chem. Commun. (Camb) 2009:7107–7109. [Abstract] [Google Scholar]
68. Marsden D.M., Nicholson R.L., Skindersoe M.E., et al. Discovery of a quorum sensing modulator pharmacophore by 3D small-molecule microarray screening. Org. Biomol. Chem. 2010;8:5313–5323. [Abstract] [Google Scholar]
69. Leslie B.J., Hergenrother P.J. Identification of the cellular targets of bioactive small organic molecules using affinity reagents. Chem. Soc. Rev. 2008;37:1347–1360. [Abstract] [Google Scholar]
70. Spandl R.J., Nicholson R.L., Marsden D.M., et al. Synthesis of a biotin-labeled quorum-sensing molecule: towards a general method for target identification. Synlett. 2008;14:2122–2126. [Google Scholar]
71. Telford G., Wheeler D., Williams P., et al. The Pseudomonas aeruginosa quorum-sensing signal molecule N-(3-oxododecanoyl)-L-homoserine lactone has immunomodulatory activity. Infect. Immun. 1998;66:36–42. [Europe PMC free article] [Abstract] [Google Scholar]
72. Amara N., Krom B.P., Kaufmann G.F., et al. Macromolecular inhibition of quorum sensing: enzymes, antibodies, and beyond. Chem. Rev. 2011;111:195–208. [Abstract] [Google Scholar]
73. Kaufmann G.F., Sartorio R., Lee S.-H., et al. Revisiting quorum sensing: discovery of additional chemical and biological functions for 3-oxo-N-acylhomoserine lactones. Proc. Natl. Acad. Sci. U S A. 2005;102:309–314. [Europe PMC free article] [Abstract] [Google Scholar]
74. Rémy B., Mion S., Plener L., et al. Interference in bacterial quorum sensing: a biopharmaceutical perspective. Front. Pharmacol. 2018;9:203. [Europe PMC free article] [Abstract] [Google Scholar]
75. Kaufmann G.F., Sartorio R., Lee S.-H., et al. Antibody interference with N-acyl homoserine lactone-mediated bacterial quorum sensing. J. Am. Chem. Soc. 2006;128:2802–2803. [Europe PMC free article] [Abstract] [Google Scholar]
76. Park J., Jagasia R., Kaufmann G.F., et al. Infection control by antibody disruption of bacterial quorum sensing signaling. Chem. Biol. 2007;14:1119–1127. [Europe PMC free article] [Abstract] [Google Scholar]
77. De Lamo Marin S., Xu Y., Meijler M.M., et al. Antibody catalyzed hydrolysis of a quorum sensing signal found in Gram-negative bacteria. Bioorg. Med. Chem. Lett. 2007;17:1549–1552. [Abstract] [Google Scholar]
78. Piletska E.V., Stavroulakis G., Karim K., et al. Attenuation of Vibrio fischeri quorum sensing using rationally designed polymers. Biomacromolecules. 2010;11:975–980. [Abstract] [Google Scholar]
79. Piletska E.V., Stavroulakis G., Larcombe L.D., et al. Passive control of quorum sensing: prevention of Pseudomonas aeruginosa biofilm formation by imprinted polymers. Biomacromolecules. 2011;12:1067–1071. [Abstract] [Google Scholar]
80. Breitbach A.S., Broderick A.H., Jewell C.M., et al. Surface-mediated release of a synthetic small-molecule modulator of bacterial quorum sensing: gradual release enhances activity. Chem. Commun. 2011;47:370–372. [Europe PMC free article] [Abstract] [Google Scholar]
81. Tan S.Y., Chua S.L., Chen Y., et al. Identification of five structurally unrelated quorum-sensing inhibitors of Pseudomonas aeruginosa from a natural-derivative database. Antimicrob. Agents Chemother. 2013;57:5629–5641. [Europe PMC free article] [Abstract] [Google Scholar]
82. Yang L., Rybtke M.T., Jakobsen T.H., et al. Computer-aided identification of recognized drugs as Pseudomonas aeruginosa quorum-sensing inhibitors. Antimicrob. Agents. Chemother. 2009;53:2432–2443. [Europe PMC free article] [Abstract] [Google Scholar]
83. Annapoorani A., Umamageswaran V., Parameswari R., et al. Computational discovery of putative quorum sensing inhibitors against LasR and RhlR receptor proteins of Pseudomonas aeruginosa. J. Comput. Aided Mol. Des. 2012;26:1067–1077. [Abstract] [Google Scholar]
84. Skovstrup S., Le Quement S.T., Hansen T., et al. Identification of LasR ligands through a virtual screening approach. ChemMedChem. 2013;8:157–163. [Abstract] [Google Scholar]
85. Kalia M., Singh P.K., Yadav V.K., et al. Structure based virtual screening for identification of potential quorum sensing inhibitors against LasR master regulator in Pseudomonas aeruginosa. Microb. Pathog. 2017;107:136–143. [Abstract] [Google Scholar]
86. Rajamanikandan S., Jeyakanthan J., Srinivasan P. Discovery of potent inhibitors targeting Vibrio harveyi LuxR through shape and e-pharmacophore based virtual screening and its biological evaluation. Microb. Pathog. 2017;103:40–56. [Abstract] [Google Scholar]
87. Rajamanikandan S., Jeyakanthan J., Srinivasan P. Molecular Docking, Molecular dynamics simulations, computational screening to design quorum sensing inhibitors targeting LuxP of Vibrio harveyi and its biological evaluation. Appl. Biochem. Biotechnol. 2017;181:192–218. [Abstract] [Google Scholar]
88. Ding T., Li T., Li J. Identification of natural product compounds as quorum sensing inhibitors in Pseudomonas fluorescens P07 through virtual screening. Bioorg. Med. Chem. 2018;26:4088–4099. [Abstract] [Google Scholar]
89. Zeng Z., Qian L., Cao L., et al. Virtual screening for novel quorum sensing inhibitors to eradicate biofilm formation of Pseudomonas aeruginosa. Appl. Microbiol. Biotechnol. 2008;79:119–126. [Abstract] [Google Scholar]
90. Kalia M., Yadav V.K., Singh P.K., et al. Designing quorum sensing inhibitors of Pseudomonas aeruginosa utilizing FabI: an enzymic drug target from fatty acid synthesis pathway. 3 Biotech. 2019;9:40. [Europe PMC free article] [Abstract] [Google Scholar]
91. de Almeida F.A., Vargas E.L.G., Carneiro D.G., et al. Virtual screening of plant compounds and nonsteroidal anti-inflammatory drugs for inhibition of quorum sensing and biofilm formation in Salmonella. Microb. Pathog. 2018;121:369–388. [Abstract] [Google Scholar]
92. Connell J.L., Ritschdorff E.T., Whiteley M., et al. 3D printing of microscopic bacterial communities. Proc. Natl. Acad. Sci. U S A. 2013;110:18380–18385. [Europe PMC free article] [Abstract] [Google Scholar]
93. Gomes J., Grunau A., Lawrence A.K., et al. Bioinspired, releasable quorum sensing modulators. Chem. Commun. (Camb) 2013;49:155–157. [Abstract] [Google Scholar]
94. Hebert C.G., Gupta A., Fernandes R., et al. Biological nanofactories target and activate epithelial cell surfaces for modulating bacterial quorum sensing and interspecies signaling. ACS Nano. 2010;4:6923–6931. [Abstract] [Google Scholar]
95. Blackwell H., Yftah T.-G., Stacy D., Inventors . 7 May 2020. [Google Scholar]
96. B. Kim, E. Ryu, J. Park, et al., Inventors; Novel brominated furanone derivative, method for preparing same, and pharmaceutical composition containing same as active ingredient, PCT patent WO2019221513A1. 21 November 2019.
97. R. Alarcon, A. Mcnulty, Inventors; Targeted Enzymatic Degradation of Quorum Sensing Peptides, United States Patent US2019298872A1. 3 October 2019.
98. H. Zhu, S. Sun, Y. Wang, et al., Inventors; Applications of 3,4,5-methyl trihydroxybenzoate in inhibition of the activity of bacterial quorum sensing system, Chinese patent CN110692636A. 17 January 2020.
99. B. Choi, E. Ryu, J. Sim, et al., Inventors; Method of Inhibiting Quorum Sensing Using D-Galactose, United States Patent US2019224222A1. 25 July 2019.
100. Y. Li, W. Wang, S. Wu, et al., Inventors; Preparation Method and Application of Camphor Essential Oil-Based Bacterial Quorum Sensing Inhibitor, Chinese patent CN109463402A. 15 March 2019.
101. N. Klaus N, Inventors; Animal feed additive for quorum sensing inhibition and from timbers, PCT patent WO2018153804A1. 30 August 2018.
102. B. Bassler, H. Stone, M. Kim, Inventors; Diagnostic and Therapeutic Quorum-Sensing-Manipulation Molecules that Are Trackable for Healthcare and Industrial Systems, United States patent US2018346525A1. 6 December 2018.
103. B. Marte, Inventor; Formulation of banana pseuostem liquid extract and its usesFormulation of banana pseuostem liquid extract and its uses, 2019 Philippine Patent PH12018000039A1. 14 August 2019.
104. Liu Z., Dong R., Zeng M., Inventors;Synergist for Food Biological Preservative and Use Method of Synergist, Chinese Patent CN109043290A. 21 December 2018.
105. Y. Xiong, Y. Liu, Inventors; Application of Pyrimidine Derivative in Preparing Medicine for Inhibiting Bacterial Quorum Sensing System, Chinese patent CN107019699A. 8 August 2017.
106. C. Pearce, J. Kavanaugh, Parlet C, et al., Inventors; Methods and compositions for the inhibition of quorum sensing in bacterial infections, PCT patent WO2017197303A1. 16 November 2017.
107. S. Naik, S. Ching, J. Scholin, et al., Inventors; Application of Porous Materials for Bacterial Quorum Sensing Inhibition/disruption, United States patent US2017251674A1. 7 September 2017.
108. H. Blackwell, M. Welsh, Inventors; Compound Combinations for Attenuation of Bacterial Virulence, United States patent US2017231962A1. 17 August 2017.
109. G. Zheng, W. Tian, C. Yang, Inventors; Novel Application of lotus Plumule Extracts, Chinese Patent CN107951928A. 24 April 2018.
110. H. Blackwell, T. Yang, Inventors; Peptidic Modulators of Quorum Sensing in staphylococcus Epidermidis, PCT Patent WO2017192442A2. 9 November 2017.
111. H. Blackwell, M. Boursier, Inventors; Synthetic Ligands that Modulate the Activity of the RHLR Quorum Sensing Receptor, PCT patent WO2017190116A1. 2 November 2017.
112. The Database of Privately and Publicly Funded Clinical Studies Conducted around the World, U.S. National Library of Medicine, https://www.clinicaltrials.gov/ct2/home. (Accessed 14 October 2020).
113. Azad M.F., Schwiertz A., Jentsch H.F. Adjunctive use of essential oils following scaling and root planing -a randomized clinical trial. BMC Complement. Altern. Med. 2016;16:171. [Europe PMC free article] [Abstract] [Google Scholar]
114. Salles M.M., Badaró M.M., Arruda C.N., et al. Antimicrobial activity of complete denture cleanser solutions based on sodium hypochlorite and Ricinus communis - a randomized clinical study. J. Appl. Oral Sci. 2015;23:637–642. [Europe PMC free article] [Abstract] [Google Scholar]
115. Goes P., Dutra C.S., Lisboa M.R., et al. Clinical efficacy of a 1% Matricaria chamomile L. mouthwash and 0.12% chlorhexidine for gingivitis control in patients undergoing orthodontic treatment with fixed appliances. J. Oral Sci. 2016;58:569–574. [Abstract] [Google Scholar]
116. Singh I., Gautam L.K., Kaur I.R. Effect of oral cranberry extract (standardized proanthocyanidin-A) in patients with recurrent UTI by pathogenic E. coli: a randomized placebo-controlled clinical research study. Int. Urol. Nephrol. 2016;48:1379–1386. [Abstract] [Google Scholar]
117. de Arruda C.N.F., Salles M.M., Badaró M.M., et al. Effect of sodium hypochlorite and Ricinus communis solutions on control of denture biofilm: A randomized crossover clinical trial. J. Prosthet. Dent. 2017;117:729–734. [Abstract] [Google Scholar]
118. Abdulbaqi H.R., Himratul-Aznita W.H., Baharuddin N.A. Evaluation of Salvadora persica L. and green tea anti-plaque effect: a randomized controlled crossover clinical trial. BMC Complement. Altern. Med. 2016;16:493. [Europe PMC free article] [Abstract] [Google Scholar]
119. Defoirdt T., Boon N., Bossier P. Can bacteria evolve resistance to quorum sensing disruption? PLoS Pathog. 2010;6 [Europe PMC free article] [Abstract] [Google Scholar]
120. Koul S., Prakash J., Mishra A., et al. Potential emergence of multi-quorum sensing inhibitor resistant (MQSIR) bacteria. Indian J. Microbiol. 2016;56:1–18. [Europe PMC free article] [Abstract] [Google Scholar]
121. Hur K.Y., Lee M.S. Gut microbiota and metabolic disorders. Diabetes Metab. J. 2015;39:198–203. [Europe PMC free article] [Abstract] [Google Scholar]
122. Certner R.H., Vollmer S.V. Inhibiting bacterial quorum sensing arrests coral disease development and disease-associated microbes. Environ. Microbiol. 2018;20:645–657. [Abstract] [Google Scholar]

Articles from Journal of Pharmaceutical Analysis are provided here courtesy of Xi'an Jiaotong University

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/128333340
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/128333340

Article citations


Go to all (24) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.

Funding 


Funders who supported this work.

National Natural Science Foundation of China (1)