Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cell ; 186(23): 5098-5113.e19, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37918395

ABSTRACT

Drug-resistant Pseudomonas aeruginosa (PA) poses an emerging threat to human health with urgent need for alternative therapeutic approaches. Here, we deciphered the B cell and antibody response to the virulence-associated type III secretion system (T3SS) in a cohort of patients chronically infected with PA. Single-cell analytics revealed a diverse B cell receptor repertoire directed against the T3SS needle-tip protein PcrV, enabling the production of monoclonal antibodies (mAbs) abrogating T3SS-mediated cytotoxicity. Mechanistic studies involving cryoelectron microscopy identified a surface-exposed C-terminal PcrV epitope as the target of highly neutralizing mAbs with broad activity against drug-resistant PA isolates. These anti-PcrV mAbs were as effective as treatment with conventional antibiotics in vivo. Our study reveals that chronically infected patients represent a source of neutralizing antibodies, which can be exploited as therapeutics against PA.


Subject(s)
Antibodies, Bacterial , Antibodies, Neutralizing , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Antibodies, Bacterial/pharmacology , Cryoelectron Microscopy , Immunoglobulins/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas Infections/drug therapy
2.
Antimicrob Agents Chemother ; : e0069424, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269189

ABSTRACT

Treatment of Pseudomonas aeruginosa infection is challenging due to its intrinsic and acquired antibiotic resistance. As the number of current therapeutic options for P. aeruginosa infections is limited, developing novel treatments against the pathogen is an urgent clinical priority. The suppression of virulence of P. aeruginosa could be a new therapeutic option, and the type III secretion system (T3SS), which enables the bacteria to translocate various kinds of toxins into host cells and inhibits cellular functions, is considered as one possible target. In this report, we examined T3SS inhibition by COT-143/INFEX702, a humanized monoclonal antibody against PcrV, T3SS component, and present the crystal structure of the antibody-PcrV complex. COT-143 inhibited T3SS-dependent cytotoxicity and protected mice from the mortality caused by P. aeruginosa infection. The inhibition of cytotoxicity coincided with inhibition of translocon formation in a host cell membrane, which is necessary for T3SS intoxication. COT-143 protected murine neutrophils and facilitated phagocytosis of P. aeruginosa. These results suggest that COT-143 facilitates P. aeruginosa clearance by protecting neutrophil via inhibition of T3SS-dependent toxin translocation. This is the first report to show that an anti-PcrV antibody directly interferes with translocon formation to inhibit intoxication of host cells.

3.
Microbiology (Reading) ; 169(10)2023 10.
Article in English | MEDLINE | ID: mdl-37819040

ABSTRACT

Pseudomonas aeruginosa is a widespread γ-proteobacterium and an important opportunistic pathogen. The genetically diverse P. aeruginosa phylogroup 3 strains are characterized by producing the pore-forming ExlA toxin and by their lack of a type III secretion system. However, like all strains of this species, they produce several virulence-associated traits, such as elastase, rhamnolipids and pyocyanin, which are regulated by quorum sensing (QS). The P. aeruginosa QS response comprises three systems (Las, Rhl and Pqs, respectively) that hierarchically regulate these virulence factors. The Pqs QS system is composed of the PqsR transcriptional factor, which, coupled with the alkyl-quinolones HHQ or PQS, activates the transcription of the pqsABCDE operon. The products of the first four genes of this operon produce HHQ, which is then converted to PQS by PqsH, while PqsE forms a complex with RhlR and stabilizes it. In this study we report that mutations affecting the Pqs system are particularly common in phylogroup 3 strains. To better understand QS in phylogroup 3 strains we studied strain MAZ105 isolated from tomato rhizosphere and showed that it contains mutations in the central QS transcriptional regulator, LasR, and in the gene encoding the PqsA enzyme involved in the synthesis of PQS. However, it can still produce QS-regulated virulence factors and is virulent in Galleria mellonella and mildly pathogenic in the mouse abscess/necrosis model; our results show that this may be due to the expression of pqsE from a different PqsR-independent promoter than the pqsA promoter. Our results indicate that using anti-virulence therapy based on targeting the PQS system will not be effective against infections by P. aeruginosa phylogroup 3 strains.


Subject(s)
Quorum Sensing , Solanum lycopersicum , Animals , Mice , Quorum Sensing/genetics , Pseudomonas aeruginosa/metabolism , Rhizosphere , Signal Transduction/genetics , Virulence Factors/genetics , Virulence Factors/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial
4.
Microb Pathog ; 183: 106281, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37541553

ABSTRACT

Metformin (MeT) is an FDA-approved drug with a myriad of health benefits. Besides being used as an anti-diabetic drug, MeT is also effective against various cancers, liver-, cardiovascular-, and renal diseases. This study was undertaken to examine its unique potential as an anti-virulence drug against an opportunistic bacterial pathogen, Pseudomonas aeruginosa. Due to the menace of multidrug resistance in pathogenic microorganisms, many novel or repurposed drugs with anti-virulence prospects are emerging as next-generation therapies with the aim to overshadow the application of existing antimicrobial regimens. The quorum sensing (QS) mechanisms of P. aeruginosa are an attractive drug target for attenuating bacterial virulence. In this context, the anti-QS potential of MeT was scrutinized using biosensor assays. MeT was comprehensively evaluated for its effects on different motility phenotypes, virulence factor production (phenotypic and genotypic expression) along with biofilm development in P. aeruginosa in vitro. At sub-lethal concentrations, MeT displayed prolific quorum quenching (QQ) ability and remarkably inhibited AHL biosynthesis in P. aeruginosa. Moreover, MeT (1/8 MIC) effectively downregulated the expression levels of various QS- and virulence genes in P. aeruginosa, which coincided with a notable reduction in the levels of alginate, hemolysin, pyocyanin, pyochelin, elastase, and protease production. In silico analysis through molecular docking also predicted strong associations between MeT and QS receptors of P. aeruginosa. MeT also compromised the motility phenotypes and successfully abrogated biofilm formation by inhibiting EPS production in P. aeruginosa. Hence, MeT may be repurposed as an anti-virulence drug against P. aeruginosa in clinical settings.


Subject(s)
Metformin , Pseudomonas aeruginosa , Molecular Docking Simulation , Metformin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Virulence/genetics , Biofilms , Quorum Sensing , Virulence Factors/metabolism
5.
EMBO J ; 37(17)2018 09 03.
Article in English | MEDLINE | ID: mdl-30037823

ABSTRACT

New anti-infective approaches are much needed to control multi-drug-resistant (MDR) pathogens, such as methicillin-resistant Staphylococcus aureus (MRSA). Here, we found for the first time that a recombinant protein derived from the cell wall binding domain (CBD) of the bacteriophage lysin PlyV12, designated as V12CBD, could attenuate S. aureus virulence and enhance host immune defenses via multiple manners. After binding with V12CBD, S. aureus became less invasive to epithelial cells and more susceptible to macrophage killing. The expressions of multiple important virulence genes of S. aureus were reduced 2.4- to 23.4-fold as response to V12CBD More significantly, V12CBD could activate macrophages through NF-κB pathway and enhance phagocytosis against S. aureus As a result, good protections of the mice from MRSA infections were achieved in therapeutic and prophylactic models. These unique functions of V12CBD would render it a novel alternative molecule to control MDRS. aureus infections.


Subject(s)
Macrophage Activation , Macrophages/immunology , Methicillin-Resistant Staphylococcus aureus , Staphylococcus Phages/immunology , Viral Proteins/immunology , Virulence Factors/immunology , Animals , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Macrophages/microbiology , Macrophages/pathology , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology , Staphylococcus Phages/genetics , Virulence Factors/genetics
6.
Chimia (Aarau) ; 76(5): 402-408, 2022 May 25.
Article in English | MEDLINE | ID: mdl-38069711

ABSTRACT

As resistance to clinically available antibiotics persistently increases, applying new strategies to target pathogenic bacteria are paramount to design effective drugs. Bacterial proteases play vital roles in cell viability and stress response, contributing to the pathogenicity of the resistant bacteria. Targeting these extracellular enzymes by antivirulence therapy is a prominent strategy in combating multi-drug resistant bacteria. By preventing the colonization and infiltration of the host, this method can lower selection pressure and reduce resistance development significantly. Here, we review the role of bacterial proteases, the rise of antivirulence therapy and we report on the development of novel antivirulence agents targeting two key virulence factors: elastase B (LasB) from Pseudomonas aeruginosa and collagenase H (ColH) from Clostridium histolyticum.

7.
Adv Sci (Weinh) ; 11(15): e2306070, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38350718

ABSTRACT

Anti-virulence therapy that interferes with bacterial communication, known as "quorum sensing (QS)", is a promising strategy for circumventing bacterial resistance. Using nanomaterials to regulate bacterial QS in anti-virulence therapy has attracted much attention, which is mainly attributed to unique physicochemical properties and excellent designability of nanomaterials. However, bacterial QS is a dynamic and multistep process, and there are significant differences in the specific regulatory mechanisms and related influencing factors of nanomaterials in different steps of the QS process. An in-depth understanding of the specific regulatory mechanisms and related influencing factors of nanomaterials in each step can significantly optimize QS regulatory activity and enhance the development of novel nanomaterials with better comprehensive performance. Therefore, this review focuses on the mechanisms by which nanomaterials regulate bacterial QS in the signal supply (including signal synthesis, secretion, and accumulation) and signal transduction cascade (including signal perception and response) processes. Moreover, based on the two key influencing factors (i.e., the nanomaterial itself and the environment), optimization strategies to enhance the QS regulatory activity are comprehensively summarized. Collectively, applying nanomaterials to regulate bacterial QS is a promising strategy for anti-virulence therapy. This review provides reference and inspiration for further research on the anti-virulence application of nanomaterials.


Subject(s)
Bacteria , Quorum Sensing , Virulence , Signal Transduction
8.
Pharmaceuticals (Basel) ; 16(11)2023 Nov 03.
Article in English | MEDLINE | ID: mdl-38004425

ABSTRACT

MbtI from Mycobacterium tuberculosis (Mtb) is a Mg2+-dependent salicylate synthase, belonging to the chorismate-utilizing enzyme (CUE) family. As a fundamental player in iron acquisition, MbtI promotes the survival and pathogenicity of Mtb in the infected host. Hence, it has emerged in the last decade as an innovative, potential target for the anti-virulence therapy of tuberculosis. In this context, 5-phenylfuran-2-carboxylic acids have been identified as potent MbtI inhibitors. The first co-crystal structure of MbtI in complex with a member of this class was described in 2020, showing the enzyme adopting an open configuration. Due to the high mobility of the loop adjacent to the binding pocket, large portions of the amino acid chain were not defined in the electron density map, hindering computational efforts aimed at structure-driven ligand optimization. Herein, we report a new, high-resolution co-crystal structure of MbtI with a furan-based derivative, in which the closed configuration of the enzyme allowed tracing the entirety of the active site pocket in the presence of the bound inhibitor. Moreover, we describe a new crystal structure of MbtI in open conformation and in complex with the known inhibitor methyl-AMT, suggesting that in vitro potency is not related to the observed enzyme conformation. These findings will prove fundamental to enhance the potency of this series via rational structure-based drug-design approaches.

9.
Life Sci ; 313: 121267, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36481167

ABSTRACT

AIMS: This study scrutinized α-Terpineol (α-T) for its anti-virulence and anti-fouling potential against P. aeruginosa PAO1 in conjunction with gentamicin (GeN) using in-vitro, in-silico, and in-vivo approaches. MAIN METHODS: The quorum quenching (QQ) potential of the drug combination was studied using a quorum sensing (QS) biosensor strain and tested for synergy using chequerboard and time-kill kinetics assays. The effect of α-T and GeN on bacterial motility, QS-regulated virulence factor production, and biofilm formation was assessed in P. aeruginosa PAO1 along with molecular docking analysis. The protective effects of α-T-GeN combination were also examined in a Caenorhabditis elegans infection model through slow-killing (SK) assays. KEY FINDINGS: The drug combination displayed synergy, enhanced QQ activity, and suppressed AHL production in PAO1. At sub-inhibitory concentrations, the drug combination suppressed the expression of genes regulating QS and pseudomonal virulence, thereby inhibiting the production of virulence factors in PAO1. The drug combination compromised all forms of pseudomonal motility, strongly inhibited biofilm formation, and successfully eradicated preformed biofilms. Based on these findings, it is concluded that GeN (alone) does not harbor any QQ properties, but enhances the QQ potential of α-T. Moreover, combinational treatment protected C. elegans from pseudomonal infection and improved survival rates by 73 % at 96 h. SIGNIFICANCE: For the first time, the molecular mechanism responsible for the anti-QS activity of α-T was unraveled through a comprehensive investigation, thereby asserting its potential as an anti-virulent drug against P. aeruginosa.


Subject(s)
Pseudomonas Infections , Quorum Sensing , Animals , Caenorhabditis elegans/metabolism , Gentamicins/pharmacology , Molecular Docking Simulation , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Biofilms , Virulence Factors/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Pseudomonas aeruginosa
10.
Front Cell Infect Microbiol ; 12: 899566, 2022.
Article in English | MEDLINE | ID: mdl-35782125

ABSTRACT

The quorum sensing (QS) circuitry of Pseudomonas aeruginosa represents an attractive target to attenuate bacterial virulence and antibiotic resistance. In this context, phytochemicals harboring anti-virulent properties have emerged as an alternative medicine to combat pseudomonal infections. Hence, this study was undertaken to investigate the synergistic effects and quorum quenching (QQ) potential of cinnamaldehyde (CiNN) in combination with gentamicin (GeN) against P. aeruginosa. The QQ activity of this novel combination was evaluated using a QS reporter strain and synergism was studied using chequerboard assays. Further, the genotypic and phenotypic expression of pseudomonal virulence factors was examined alongside biofilm formation. The combination of CiNN and GeN exhibited synergy and promising anti-QS activity. This drug combination was shown to suppress AHL production and downregulate the expression of critical QS genes in P. aeruginosa PAO1. Molecular docking revealed strong interactions between the QS receptors and CiNN, asserting its QQ potential. Bacterial motility was compromised along with a significant reduction in pyocyanin (72.3%), alginate (58.7%), rhamnolipid (33.6%), hemolysin (82.6%), protease (70.9%), and elastase (63.9%) production. The drug combination successfully eradicated preformed biofilms and inhibited biofilm formation by abrogating EPS production. Our findings suggest that although GeN alone could not attenuate QS, but was able to augment the anti-QS potential of CiNN. To validate our results using an infection model, we quantified the survival rates of Caenorhabditis elegans following PAO1 challenge. The combination significantly rescued C. elegans from PAO1 infection and improved its survival rate by 54% at 96 h. In summary, this study is the first to elucidate the mechanism behind the QQ prospects of CiNN (augmented in presence of GeN) by abrogating AHL production and increasing the survival rate of C. elegans, thereby highlighting its anti-virulent properties.


Subject(s)
Pseudomonas Infections , Acrolein/analogs & derivatives , Animals , Caenorhabditis elegans , Gentamicins , Molecular Docking Simulation , Pseudomonas aeruginosa , Quorum Sensing
11.
Drug Discov Today ; 26(9): 2164-2172, 2021 09.
Article in English | MEDLINE | ID: mdl-33781954

ABSTRACT

Virulence factor, sortase A (SrtA), has crucial roles in the pathogenesis of Gram-positive superbugs. SrtA is a bacterial cell membrane enzyme that anchors crucial virulence factors to the cell wall surface of Gram-positive bacteria. SrtA is not necessary for bacterial growth and viability and is conveniently accessible in the cell membrane; therefore, it is an ideal target for antivirulence drug development. In this review, we focus on antimicrobial resistance (AMR)-expressing bacteria and SrtA as a potential target for overcoming AMR. The mechanism of action of SrtA and its inhibition by various types of inhibitors, such as synthetic small molecules, peptides, and natural products, are provided. Future SrtA research perspectives for alternative drug development to antibiotics are also proposed.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Drug Resistance, Bacterial , Gram-Positive Bacterial Infections/drug therapy , Aminoacyltransferases/chemistry , Aminoacyltransferases/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biological Products/therapeutic use , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Humans , Peptides/therapeutic use
12.
Antibiotics (Basel) ; 10(2)2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33494538

ABSTRACT

While antimicrobial resistance (AMR) is seen in both Neisseria gonorrhoeae and Neisseria meningitidis, the former has become resistant to commonly available over-the-counter antibiotic treatments. It is imperative then to develop new therapies that combat current AMR isolates whilst also circumventing the pathways leading to the development of AMR. This review highlights the growing research interest in developing anti-virulence therapies (AVTs) which are directed towards inhibiting virulence factors to prevent infection. By targeting virulence factors that are not essential for gonococcal survival, it is hypothesized that this will impart a smaller selective pressure for the emergence of resistance in the pathogen and in the microbiome, thus avoiding AMR development to the anti-infective. This review summates the current basis of numerous anti-virulence strategies being explored for N. gonorrhoeae.

13.
Antibiotics (Basel) ; 9(4)2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32244277

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen and a common cause of skin infection. S. aureus also plays a role in the pathogenesis of the chronic inflammatory skin disease, atopic dermatitis. S. aureus virulence involves activation of the quorum sensing agr operon. In this paper, we show that the diterpene carnosic acid, present in R. officinalis L. (rosemary) leaves, is a specific inhibitor of S. aureus agr expression as low as 5 µM. Carnosol and rosmarinic acid are two other phytochemicals present in rosemary leaves. Carnosol, but not rosmarinic acid, is also a potent agr expression inhibitor. Natural rosemary extracts containing carnosic acid and carnosol inhibit S. aureus agr expression, both in luciferase reporter strains and in wild type strains isolated from patients with atopic dermatitis. Specific inhibition of S. aureus virulence using topical formulations of rosemary extract may offer a practical approach to preventing and treating flares of atopic dermatitis.

14.
Biomaterials ; 217: 119310, 2019 10.
Article in English | MEDLINE | ID: mdl-31271860

ABSTRACT

Achieving remote and reversible control of bacterial cell-cell interactions associated with interference with pathological processes in living systems remains a challenge owing to the complexity of the in vivo microenvironment and the lack of regulation systems. We present, for the first time, the development of a versatile platform to achieve NIR-driven reversible bacterial clustering both in vitro and in vivo. This platform consisted of ß-CD modified UCNP (UCNP-CD) and photochromic azobenzene glycoconjugates (azo-man), which could dynamically display d-mannose bioactive ligands. Specifically, by virtue of the noncovalent yet strong multivalent interactions between bacteria and nanosystems, robust bacterial clusters could be formed even in vivo within 1 h. Upon NIR stimulation, the upconverted emissions from UCNPs triggered the continuous isomerization of azo-man, leading to dissociation of the nanosystems and dispersion of bacterial clusters. Moreover, in vivo pathogenic infection process could be interfered with the NIR-switched bacterial agglutination. Most importantly, the noninvasive and deep-tissue-penetrating nature of NIR made it possible for dynamically regulation of cellular interactions with minimized influence to both normal cells and nature bacteria flora. This strategy would bring new perspectives for anti-virulence therapeutics and in-depth investigations of specific physiological phenomena.


Subject(s)
Escherichia coli/radiation effects , Infrared Rays , Nanoparticles/chemistry , Animals , Male , Mice , NIH 3T3 Cells , Nanoparticles/ultrastructure , beta-Cyclodextrins/chemistry
15.
Article in English | MEDLINE | ID: mdl-31001485

ABSTRACT

Antimicrobial resistance constitutes one of the major challenges facing humanity in the Twenty-First century. The spread of resistant pathogens has been such that the possibility of returning to a pre-antibiotic era is real. In this scenario, innovative therapeutic strategies must be employed to restrict resistance. Among the innovative proposed strategies, anti-virulence therapy has been envisioned as a promising alternative for effective control of the emergence and spread of resistant pathogens. This review presents some of the anti-virulence strategies that are currently being developed, it will cover strategies focused on quench pathogen quorum sensing (QS) systems, disassemble of bacterial functional membrane microdomains (FMMs), disruption of biofilm formation and bacterial toxin neutralization.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Biofilms/drug effects , Drug Discovery/trends , Membrane Microdomains/drug effects , Quorum Sensing/drug effects , Virulence/drug effects , Anti-Bacterial Agents/isolation & purification , Bacteria/pathogenicity , Virulence Factors/antagonists & inhibitors
16.
Front Microbiol ; 10: 2657, 2019.
Article in English | MEDLINE | ID: mdl-31798568

ABSTRACT

Pseudomonas aeruginosa is an opportunistic bacterium associated with healthcare infections in intensive care units (ICUs), ventilator-associated pneumonia (VAP), surgical site infections, and burns. This bacterium causes 75% of death in burned patients, since it can develop a persistent biofilm associated with infections, express several virulence factors, and antibiotic-resistance mechanisms. Some of these virulence factors are proteases such as elastase and alkaline protease, or toxic metabolites such as pyocyanin and is one of the few microorganisms able to produce cyanide, which inhibits the cytochrome oxidase of host cells. These virulence factors are controlled by quorum sensing (QS). In this work, 30 P. aeruginosa clinical strains isolated from burned patients from a tertiary hospital in Mexico City were studied. Antibiotic susceptibility tests were done, and virulence factors (elastase, alkaline protease, HCN, and pyocyanin) were determined in presence of an N-acylhomoserine lactonase, AiiM able to hydrolyze a wide range of acyl homoserine lactones. The treatment reduced significantly the activities of elastase and alkaline protease, and the production of pyocyanin and HCN in all producer strains but not the secretion of toxins through the type III secretion system. Our work suggests that AiiM treatment may be an effective therapy to combat P. aeruginosa infection in burn patients.

17.
Front Microbiol ; 10: 1701, 2019.
Article in English | MEDLINE | ID: mdl-31428062

ABSTRACT

Elastase B (lasB) is a multifunctional metalloenzyme secreted by the gram-negative pathogen Pseudomonas aeruginosa, and this enzyme orchestrates several physiopathological events during bacteria-host interplays. LasB is considered to be a potential target for the development of an innovative chemotherapeutic approach, especially against multidrug-resistant strains. Recently, our group showed that 1,10-phenanthroline-5,6-dione (phendione), [Ag(phendione)2]ClO4 (Ag-phendione) and [Cu(phendione)3](ClO4)2.4H2O (Cu-phendione) had anti-P. aeruginosa action against both planktonic- and biofilm-growing cells. In the present work, we have evaluated the effects of these compounds on the (i) interaction with the lasB active site using in silico approaches, (ii) lasB proteolytic activity by using a specific fluorogenic peptide substrate, (iii) lasB gene expression by real time-polymerase chain reaction, (iv) lasB protein secretion by immunoblotting, (v) ability to block the damages induced by lasB on a monolayer of lung epithelial cells, and (vi) survivability of Galleria mellonella larvae after being challenged with purified lasB and lasB-rich bacterial secretions. Molecular docking analyses revealed that phendione and its Ag+ and Cu2+ complexes were able to interact with the amino acids forming the active site of lasB, particularly Cu-phendione which exhibited the most favorable interaction energy parameters. Additionally, the test compounds were effective inhibitors of lasB activity, blocking the in vitro cleavage of the peptide substrate, aminobenzyl-Ala-Gly-Leu-Ala-p-nitrobenzylamide, with Cu-phendione having the best inhibitory action (K i = 90 nM). Treating living bacteria with a sub-inhibitory concentration (½ × MIC value) of the test compounds caused a significant reduction in the expression of the lasB gene as well as its mature protein production/secretion. Further, Ag-phendione and Cu-phendione offered protective action for lung epithelial cells, reducing the A549 monolayer damage by approximately 32 and 42%, respectively. Interestingly, Cu-phendione mitigated the toxic effect of both purified lasB molecules and lasB-containing bacterial secretions in the in vivo model, increasing the survival time of G. mellonella larvae. Collectively, these data reinforce the concept of lasB being a veritable therapeutic target and phendione-based compounds (mainly Cu-phendione) being prospective anti-virulence drugs against P. aeruginosa.

18.
Eur J Med Chem ; 152: 370-376, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29738955

ABSTRACT

Emergence of antibiotic-resistant bacteria constitutes an increasing threat to human health. For example, treatment options for Staphylococcus aureus infections is declining with the worldwide spreading of highly virulent community-associated methicillin-resistant S. aureus (CA-MRSA) strains. Anti-virulence therapy has been proposed as an alternative treatment strategy, as it typically involves inhibition of expression of virulence factors rather than direct bacterial killing, thereby attenuating the risk of resistance development. An intriguing target is the agr quorum-sensing system, which is a major inducer of virulence in CA-MRSA upon activation by agr-encoded staphylococcal autoinducing peptides (AIPs). In the present work a previously identified lactam hybrid analogue based on the marine depsipeptide solonamide B and the general structure of AIPs was investigated with respect to structure-function relationships. An array of 27 analogues exploring expansion of ring size, type of side chain, amino acid substitutions, and stereochemistry was designed and tested for AgrC-inhibitory activity. Interestingly, it was found that an analogue derived from the mirror image of the original hit proved to be the hitherto most efficient AgrC inhibitor resembling solonamide B in amino acid sequence. This and closely related compounds were 20- to 40-fold more potent in AgrC inhibition than the starting hit compound.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/pharmacology , Depsipeptides/pharmacology , Lactams/pharmacology , Peptides, Cyclic/pharmacology , Protein Kinase Inhibitors/pharmacology , Bacterial Proteins/chemical synthesis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Depsipeptides/chemical synthesis , Depsipeptides/chemistry , Dose-Response Relationship, Drug , Lactams/chemistry , Molecular Conformation , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinases/metabolism , Structure-Activity Relationship , beta-Lactamases/metabolism
19.
Article in English | MEDLINE | ID: mdl-30805311

ABSTRACT

Faced with the global health threat of increasing resistance to antibiotics, researchers are exploring interventions that target bacterial virulence factors. Quorum sensing is a particularly attractive target because several bacterial virulence factors are controlled by this mechanism. Furthermore, attacking the quorum-sensing signaling network is less likely to select for resistant strains than using conventional antibiotics. Strategies that focus on the inhibition of quorum-sensing signal production are especially attractive because the enzymes involved are expressed in bacterial cells but are not present in their mammalian counterparts. We review here various approaches that are being taken to interfere with quorum-sensing signal production via the inhibition of autoinducer-2 synthesis, PQS synthesis, peptide autoinducer synthesis, and N-acyl-homoserine lactone synthesis. We expect these approaches will lead to the discovery of new quorum-sensing inhibitors that can help to stem the tide of antibiotic resistance.


Subject(s)
Adaptation, Physiological/drug effects , Anti-Bacterial Agents/isolation & purification , Bacteria/drug effects , Gene Expression Regulation, Bacterial/drug effects , Quorum Sensing/drug effects , Virulence/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/pathogenicity , Drug Evaluation, Preclinical/methods , Virulence Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL