Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Bioorg Chem ; 130: 106226, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36332317

RESUMEN

It is well established that the quorum sensing (QS) in Pseudomonas aeruginosa is primarily responsible for the synthesis and the release of several virulence factors including pyocyanin and are involved in biofilm formation. In the Pseudomonas quinolone signal (PQS) system, autoinducers such as PQS and HHQ bind and activate the transcription regulator protein receptor PqsR (MvfR). Targeting PqsR with competitive inhibitors could be a promising strategy to inhibit QS in P. aeruginosa to overcome antimicrobial resistance. In this study, we have designed and synthesized a series of novel quinazolinone disulfide-containing competitive inhibitor of PqsR. The most potent analogue 8q efficiently inhibited the pqs system with an IC50 value of 4.5 µM. It also showed complete suppression of pyocyanin production and a significant reduction in biofilm formation by P. aeruginosa (PAO1) with low cytotoxicity. Additionally, 8q produced synergy in combination with known antibiotics such as ciprofloxacin and tobramycin. Finally, molecular docking analysis suggested that compound 8q could bind with the ligand-binding domain of PqsR in a similar fashion to the native ligand.


Asunto(s)
Pseudomonas aeruginosa , Percepción de Quorum , Pseudomonas aeruginosa/fisiología , Piocianina , Ligandos , Simulación del Acoplamiento Molecular , Quinazolinonas/farmacología , Quinazolinonas/metabolismo , Disulfuros/farmacología , Biopelículas , Proteínas Bacterianas/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(14): 7021-7026, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30846553

RESUMEN

The opportunistic bacterial pathogen Pseudomonas aeruginosa has a layered acyl-homoserine lactone (AHL) quorum-sensing (QS) system, which controls production of a variety of extracellular metabolites and enzymes. The LasRI system activates genes including those coding for the extracellular protease elastase and for the second AHL QS system, RhlRI. Growth of P. aeruginosa on casein requires elastase production and LasR-mutant social cheats emerge in populations growing on casein. P. aeruginosa colonizes the lungs of individuals with the genetic disease cystic fibrosis (CF), and LasR mutants can be isolated from the colonized lungs; however, unlike laboratory-generated LasR mutants, many of these CF isolates have functioning RhlR-RhlI systems. We show that one such mutant can use the RhlR-RhlI system to activate expression of elastase and grow on casein. We carried out social-evolution experiments by growing this isolate on caseinate and, as with wild-type P. aeruginosa, elastase-negative mutants emerge as cheats, but these are not RhlR mutants; rather, they are mutants that do not produce the non-AHL Pseudomonas quinolone signal (PQS). Furthermore, we generated a RhlRI mutant and showed it had a fitness defect when growing together with the parent. Apparently, RhlR QS and PQS collude to support growth on caseinate in the absence of a functional LasR. Our findings provide a plausible explanation as to why P. aeruginosa LasR mutants, but not RhlR mutants, are common in CF lungs.


Asunto(s)
Acil-Butirolactonas/metabolismo , Proteínas Bacterianas/metabolismo , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum , Transducción de Señal , Transactivadores/metabolismo , Proteínas Bacterianas/genética , Humanos , Pseudomonas aeruginosa/genética , Transactivadores/genética
3.
Appl Environ Microbiol ; 86(9)2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32086305

RESUMEN

The quinolone ring is a common core structure of natural products exhibiting antimicrobial, cytotoxic, and signaling activities. A prominent example is the Pseudomonas quinolone signal (PQS), a quorum-sensing signal molecule involved in the regulation of virulence of Pseudomonas aeruginosa The key reaction to quinolone inactivation and biodegradation is the cleavage of the 3-hydroxy-4(1H)-quinolone ring, catalyzed by dioxygenases (HQDs), which are members of the α/ß-hydrolase fold superfamily. The α/ß-hydrolase fold core domain consists of a ß-sheet surrounded by α-helices, with an active site usually containing a catalytic triad comprising a nucleophilic residue, an acidic residue, and a histidine. The nucleophile is located at the tip of a sharp turn, called the "nucleophilic elbow." In this work, we developed a search workflow for the identification of HQD proteins from databases. Search and validation criteria include an [H-x(2)-W] motif at the nucleophilic elbow, an [HFP-x(4)-P] motif comprising the catalytic histidine, the presence of a helical cap domain, the positioning of the triad's acidic residue at the end of ß-strand 6, and a set of conserved hydrophobic residues contributing to the substrate cavity. The 161 candidate proteins identified from the UniProtKB database originate from environmental and plant-associated microorganisms from all domains of life. Verification and characterization of HQD activity of 9 new candidate proteins confirmed the reliability of the search strategy and suggested residues correlating with distinct substrate preferences. Among the new HQDs, PQS dioxygenases from Nocardia farcinica, N. cyriacigeorgica, and Streptomyces bingchenggensis likely are part of a catabolic pathway for alkylquinolone utilization.IMPORTANCE Functional annotation of protein sequences is a major requirement for the investigation of metabolic pathways and the identification of sought-after biocatalysts. To identify heterocyclic ring-cleaving dioxygenases within the huge superfamily of α/ß-hydrolase fold proteins, we defined search and validation criteria for the primarily motif-based identification of 3-hydroxy-4(1H)-quinolone 2,4-dioxygenases (HQD). HQDs are key enzymes for the inactivation of metabolites, which can have signaling, antimicrobial, or cytotoxic functions. The HQD candidates detected in this study occur particularly in environmental and plant-associated microorganisms. Because HQDs active toward the Pseudomonas quinolone signal (PQS) likely contribute to interactions within microbial communities and modulate the virulence of Pseudomonas aeruginosa, we analyzed the catalytic properties of a PQS-cleaving subset of HQDs and specified characteristics to identify PQS-cleaving dioxygenases within the HQD family.


Asunto(s)
Proteínas Bacterianas/genética , Hidrolasas/genética , Pseudomonas aeruginosa/genética , Quinolonas/metabolismo , Percepción de Quorum , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Hidrolasas/química , Hidrolasas/metabolismo , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/metabolismo , Alineación de Secuencia
4.
J Struct Biol ; 207(3): 287-294, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31228546

RESUMEN

The cofactor-less dioxygenase AqdC of Mycobacteroides abscessus catalyzes the cleavage and thus inactivation of the Pseudomonas quinolone signal (PQS, 2-heptyl-3-hydroxy-4(1H)-quinolone), which plays a central role in the regulation of virulence factor production by Pseudomonas aeruginosa. We present here the crystal structures of AqdC in its native state and in complex with the PQS cleavage product N-octanoylanthranilic acid, and of mutant AqdC proteins in complex with PQS. AqdC possesses an α/ß-hydrolase fold core domain with additional helices forming a cap domain. The protein is traversed by a bipartite tunnel, with a funnel-like entry section leading to an elliptical substrate cavity where PQS positioning is mediated by a combination of hydrophobic interactions and hydrogen bonds, with the substrate's C4 carbonyl and C3 hydroxyl groups tethered by His97 and the catalytic His246, respectively. The side chain of the AqdC-bound product extends deeper into the "alkyl tail section" of the tunnel than PQS, tentatively suggesting product exit via this part of the tunnel. AqdC prefers PQS over congeners with shorter alkyl substituents at C2. Kinetic data confirmed the strict requirement of the active-site base His246 for catalysis, and suggested that evolution of the canonical nucleophile/His/Asp catalytic triad of the hydrolases to an Ala/His/Asp triad is favorable for catalyzing dioxygenolytic PQS ring cleavage.


Asunto(s)
Dioxigenasas/química , Hidrolasas/química , Pseudomonas aeruginosa/metabolismo , Quinolonas/química , Cristalografía por Rayos X , Dioxigenasas/genética , Dioxigenasas/metabolismo , Regulación Bacteriana de la Expresión Génica , Enlace de Hidrógeno , Hidrolasas/genética , Hidrolasas/metabolismo , Cinética , Modelos Moleculares , Mutación , Mycobacterium/enzimología , Mycobacterium/genética , Filogenia , Unión Proteica , Conformación Proteica , Pseudomonas aeruginosa/genética , Quinolonas/metabolismo , Especificidad por Sustrato
5.
Infect Immun ; 87(10)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31308081

RESUMEN

The nosocomial pathogen Pseudomonas aeruginosa regulates its virulence via a complex quorum sensing network, which, besides N-acylhomoserine lactones, includes the alkylquinolone signal molecules 2-heptyl-3-hydroxy-4(1H)-quinolone (Pseudomonas quinolone signal [PQS]) and 2-heptyl-4(1H)-quinolone (HHQ). Mycobacteroides abscessus subsp. abscessus, an emerging pathogen, is capable of degrading the PQS and also HHQ. Here, we show that although M. abscessus subsp. abscessus reduced PQS levels in coculture with P. aeruginosa PAO1, this did not suffice for quenching the production of the virulence factors pyocyanin, pyoverdine, and rhamnolipids. However, the levels of these virulence factors were reduced in cocultures of P. aeruginosa PAO1 with recombinant M. abscessus subsp. massiliense overexpressing the PQS dioxygenase gene aqdC of M. abscessus subsp. abscessus, corroborating the potential of AqdC as a quorum quenching enzyme. When added extracellularly to P. aeruginosa cultures, AqdC quenched alkylquinolone and pyocyanin production but induced an increase in elastase levels. When supplementing P. aeruginosa cultures with QsdA, an enzyme from Rhodococcus erythropolis which inactivates N-acylhomoserine lactone signals, rhamnolipid and elastase levels were quenched, but HHQ and pyocyanin synthesis was promoted. Thus, single quorum quenching enzymes, targeting individual circuits within a complex quorum sensing network, may also elicit undesirable regulatory effects. Supernatants of P. aeruginosa cultures grown in the presence of AqdC, QsdA, or both enzymes were less cytotoxic to human epithelial lung cells than supernatants of untreated cultures. Furthermore, the combination of both aqdC and qsdA in P. aeruginosa resulted in a decline of Caenorhabditis elegans mortality under P. aeruginosa exposure.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Dioxigenasas/genética , Regulación Bacteriana de la Expresión Génica , Mycobacterium abscessus/genética , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/genética , Células A549 , Animales , Antibiosis/genética , Caenorhabditis elegans/microbiología , Hidrolasas de Éster Carboxílico/metabolismo , Hidrolasas de Éster Carboxílico/farmacología , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Dioxigenasas/metabolismo , Dioxigenasas/farmacología , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Mycobacterium abscessus/enzimología , Oligopéptidos/genética , Oligopéptidos/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Piocianina/genética , Piocianina/metabolismo , Quinolonas/metabolismo , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
6.
Molecules ; 23(2)2018 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-29382099

RESUMEN

Pseudomonas aeruginosa is a major opportunistic pathogen in cystic fibrosis, wound and nosocomial infections, posing a serious burden to public health, due to its antibiotic resistance. The P. aeruginosa Pseudomonas Quinolone System (pqs) quorum sensing system, driven by the activation of the transcriptional regulator, PqsR (MvfR) by alkylquinolone (AQ) signal molecules, is a key player in the regulation of virulence and a potential target for the development of novel antibacterial agents. In this study, we performed in silico docking analysis, coupled with screening using a P. aeruginosa mCTX::PpqsA-lux chromosomal promoter fusion, to identify a series of new PqsR antagonists. The hit compounds inhibited pyocyanin and alkylquinolone signal molecule production in P. aeruginosa PAO1-L and PA14 strains. The inhibitor Ia, which showed the highest activity in PA14, reduced biofilm formation in PAO1-L and PA14, increasing their sensitivity to tobramycin. Furthermore, the hepatic and plasma stabilities for these compounds were determined in both rat and human in vitro microsomal assays, to gain a further understanding of their therapeutic potential. This work has uncovered a new class of P. aeruginosa PqsR antagonists with potential for hit to lead optimisation in the search for quorum sensing inhibitors for future anti-infective drug discovery programs.


Asunto(s)
Antibacterianos/química , Biopelículas , Simulación del Acoplamiento Molecular , Pseudomonas aeruginosa/fisiología , Quinolonas/metabolismo , Percepción de Quorum/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
J Biol Chem ; 291(13): 6610-24, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26811339

RESUMEN

Pseudomonas aeruginosaproduces a number of alkylquinolone-type secondary metabolites best known for their antimicrobial effects and involvement in cell-cell communication. In the alkylquinolone biosynthetic pathway, the ß-ketoacyl-(acyl carrier protein) synthase III (FabH)-like enzyme PqsBC catalyzes the condensation of octanoyl-coenzyme A and 2-aminobenzoylacetate (2-ABA) to form the signal molecule 2-heptyl-4(1H)-quinolone. PqsBC, a potential drug target, is unique for its heterodimeric arrangement and an active site different from that of canonical FabH-like enzymes. Considering the sequence dissimilarity between the subunits, a key question was how the two subunits are organized with respect to the active site. In this study, the PqsBC structure was determined to a 2 Å resolution, revealing that PqsB and PqsC have a pseudo-2-fold symmetry that unexpectedly mimics the FabH homodimer. PqsC has an active site composed of Cys-129 and His-269, and the surrounding active site cleft is hydrophobic in character and approximately twice the volume of related FabH enzymes that may be a requirement to accommodate the aromatic substrate 2-ABA. From physiological and kinetic studies, we identified 2-aminoacetophenone as a pathway-inherent competitive inhibitor of PqsBC, whose fluorescence properties could be used forin vitrobinding studies. In a time-resolved setup, we demonstrated that the catalytic histidine is not involved in acyl-enzyme formation, but contributes to an acylation-dependent increase in affinity for the second substrate 2-ABA. Introduction of Asn into the PqsC active site led to significant activity toward the desamino substrate analog benzoylacetate, suggesting that the substrate 2-ABA itself supplies the asparagine-equivalent amino function that assists in catalysis.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/química , 4-Quinolonas/química , Acilcoenzima A/química , Aminobenzoatos/química , Proteínas Bacterianas/química , Pseudomonas aeruginosa/química , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/antagonistas & inhibidores , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , 4-Quinolonas/metabolismo , Acetofenonas/química , Acilcoenzima A/metabolismo , Secuencia de Aminoácidos , Aminobenzoatos/metabolismo , Antibacterianos/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Sitios de Unión , Unión Competitiva , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Escherichia coli/genética , Escherichia coli/metabolismo , Cinética , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Pseudomonas aeruginosa/enzimología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alineación de Secuencia
8.
Infect Immun ; 85(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28630072

RESUMEN

Pseudomonasaeruginosa causes lung infections in patients with cystic fibrosis (CF). The Pseudomonas quinolone signal (PQS) compound is a secreted P. aeruginosa virulence factor that contributes to the pathogenicity of P. aeruginosa We were able to detect PQS in sputum samples from CF patients infected with P. aeruginosa but not in samples from uninfected patients. We then tested the hypothesis that PQS induces oxidative stress in host cells by determining the ability of PQS to induce the production of reactive oxygen species (ROS) in lung epithelial cells (A549 and primary normal human bronchial epithelial [NHBE]) cells and macrophages (J774A.1 and THP-1). ROS production induced by PQS was detected with fluorescent probes (dichlorodihydrofluorescein diacetate, dihydroethidium, and MitoSOX Red) in conjunction with confocal microscopy and flow cytometry. PQS induced ROS production in lung epithelial (A549 and NHBE) cells and macrophages (J774A.1 and THP-1 cells). NHBE cells were sensitive to PQS concentrations as low as 500 ng/ml. PQS significantly induced early apoptosis (P < 0.05, n = 6) in lung epithelial cells, as measured by annexin/propidium iodide detection by flow cytometry. However, no change in apoptosis upon PQS treatment was seen in J774A.1 cells. Heme oxygenase-1 (HO-1) protein is an antioxidant enzyme usually induced by oxidative stress. Interestingly, incubation with PQS significantly reduced HO-1 and NrF2 expression in A549 and NHBE cells but increased HO-1 expression in J774A.1 cells (P < 0.05, n = 3), as determined by immunoblotting and densitometry. These PQS effects on host cells could play an important role in the pathogenicity of P. aeruginosa infections.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Células Epiteliales/efectos de los fármacos , Hemo-Oxigenasa 1/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Estrés Oxidativo , Quinolonas/metabolismo , Animales , Línea Celular , Células Epiteliales/química , Células Epiteliales/enzimología , Citometría de Flujo , Humanos , Macrófagos/química , Macrófagos/enzimología , Ratones , Microscopía Confocal , Especies Reactivas de Oxígeno/análisis
9.
Chembiochem ; 18(20): 2045-2055, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-28834007

RESUMEN

Pseudomonas aeruginosa, a prevalent pathogen in nosocomial infections and a major burden in cystic fibrosis, uses three interconnected quorum-sensing systems to coordinate virulence processes. At variance with other Gram-negative bacteria, one of these systems relies on 2-alkyl-4(1H)-quinolones (Pseudomonas quinolone signal, PQS) and might hence be an attractive target for new anti-infective agents. Here we report crystal structures of the N-terminal domain of anthranilate-CoA ligase PqsA, the first enzyme of PQS biosynthesis, in complex with anthraniloyl-AMP and with 6-fluoroanthraniloyl-AMP (6FABA-AMP) at 1.4 and 1.7 Šresolution. We find that PqsA belongs to an unrecognized subfamily of anthranilate-CoA ligases that recognize the amino group of anthranilate through a water-mediated hydrogen bond. The complex with 6FABA-AMP explains why 6FABA, an inhibitor of PQS biosynthesis, is a good substrate of PqsA. Together, our data might pave a way to new pathoblockers in P. aeruginosa infections.


Asunto(s)
Ligasas/química , Ligasas/metabolismo , Pseudomonas aeruginosa/citología , Pseudomonas aeruginosa/metabolismo , Quinolonas/metabolismo , Percepción de Quorum , ortoaminobenzoatos/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Modelos Moleculares , Pseudomonas aeruginosa/enzimología , ortoaminobenzoatos/química
10.
J Basic Microbiol ; 55(5): 566-78, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25283438

RESUMEN

Pathogenicity of opportunistic Pseudomonas aeruginosa is mediated through expression of different virulence determinants, most of which are under the control of quorum sensing. Besides acylhomoserine lactones, P. aeruginosa produces Pseudomonas quinolone signal (PQS) molecules which co-regulate expression of overlapping subset of genes. In the present study, effect of mutations in the pqs genes on the production of virulence factors, biofilm, and membrane vesicles (MVs) was studied using standard strain and isogenic pqs mutants of P. aeruginosa. Mutations in pqs genes severely reduced elastase, pyocyanin, siderophores, biofilm formation, and production of MVs. Further, effect of synthetic PQS on virulence of P. aeruginosa and its correlation with MVs was investigated. Supplementation of PQS resulted in enhancement of phenotypic expression of virulence factors and biofilm forming capacity of these strains. Restoration of virulent phenotype of mutants in presence of PQS indicated that PQS system play an important role in the virulence of P. aeruginosa. In addition, PQS also induced substantial release of MVs in all strains. When vesicles containing natural PQS were added to the mutants, significant increase in production of virulence factors was observed. This augmentation of the virulence traits may be associated with the efficient delivery of PQS among bacterial cells, which could be one possible mechanism of pqs system contributing to the overall virulence of P. aeruginosa.


Asunto(s)
Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Quinolonas/metabolismo , Transducción de Señal , Factores de Virulencia/metabolismo , Biopelículas/crecimiento & desarrollo , Vías Biosintéticas/genética , Vesículas Extracelulares/metabolismo , Mutación , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Virulencia
11.
Int J Med Microbiol ; 304(8): 1199-208, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25240873

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen which employs quorum sensing system to regulate several genes required for its survival and pathogenicity within the host. Besides acylhomoserine lactone (AHL) mediated las and rhl systems, this organism possesses Pseudomonas quinolone signalling (PQS) system based on alkyl quinolone signal molecules. The quinolone system represents another layer of sophistication in the complex quorum sensing cascade. Therefore, in the present study, we evaluated the contribution of the PQS system in the establishment of acute urinary tract infection (UTI) in the mouse model. For this, wild-type parent strain of P. aeruginosa MPAO1 and its isogenic single transposon mutant strains pqsH and pqsA were employed to induce UTI in mice. PQS molecules in the tissue homogenates of mice were detected by high performance thin layer chromatography (HP-TLC) method. Virulence of strains was assessed in terms of bacteriological count, histopathological lesions in the renal and bladder tissue and generation of pathological index markers like reactive nitrogen intermediates and malondialdehyde. HP-TLC analysis showed presence of PQS molecules in the renal and bladder tissue of mice infected with MPAO1 while no PQS was detected in case of pqsH and pqsA mutant strains. Results indicated that MPAO1 possessing fully functional PQS biosynthetic genes was highly virulent and caused acute pyelonephritis with severe inflammation and tissue destruction. On the contrary, significant reduction in the log count, mild tissue damage and declined levels of pathological markers were observed in mice infected with mutant strains as compared to MPAO1. Further among mutants, all these parameters were maximally impaired in the pqsA mutant in which synthesis of alkyl quinolones was completely abolished due to the transposon mutation in respective gene. Virulence of the pqsH mutant strain was lesser than that of the MPAO1 but higher than pqsA mutant. In addition, the levels of locally generated pro- and anti-inflammatory cytokines were also found to be low in the renal homogenates of mice infected with the mutant strains. Further, supplementation of strains with PQS resulted in significant enhancement in the virulence as indicated by increased bacterial load, severe histopathological damage and enhanced levels of pro-inflammatory cytokines. These findings provide a new insight into the relevant importance of the Pseudomonas quinolone signalling system in the acute UTI caused by P. aeruginosa. This system can be a potential target for futuristic anti-infective approach against this organism.


Asunto(s)
Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Quinolonas/metabolismo , Percepción de Quorum , Transducción de Señal , Infecciones Urinarias/microbiología , Animales , Carga Bacteriana , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Histocitoquímica , Riñón/microbiología , Riñón/patología , Ratones , Mutagénesis Insercional , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/crecimiento & desarrollo , Índice de Severidad de la Enfermedad , Vejiga Urinaria/microbiología , Vejiga Urinaria/patología , Infecciones Urinarias/patología , Virulencia
12.
J Basic Microbiol ; 54(7): 633-43, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23996096

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen capable of group behaviors including swarming motility and biofilm formation. Swarming motility plays an important role in the bacterium's spread to new environments, attachment to surfaces, and biofilm formation. Bacterial biofilm is associated with many persistent infections and increased resistance to antibiotics. In this study, we tested the effect of a 2-alkyl-4(1H)-quinolone (AHQ) signal, the Pseudomonas quinolone signal (PQS) on P. aeruginosa swarming and biofilm formation. Our results show that PQS repressed the swarming motility of P. aeruginosa PAO1. Such repression was independent of its cognate receptor PqsR and was not related to changes in the flagellae, type IV pili or the production of the surface-wetting agent rhamnolipid surfactant. While PQS did not affect twitching motility in PAO1, a pqsR deletion abolished twitching motility, indicating that pqsR is required for twitching motility. Our results also indicate that the enhancement of biofilm formation by PQS is at least partially dependent on the GacAS-Rsm regulatory pathway but does not involve the las or rhl QS systems.


Asunto(s)
Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica , Pseudomonas aeruginosa/efectos de los fármacos , Quinolonas/farmacología , Receptores de Superficie Celular/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Fimbrias Bacterianas/fisiología , Flagelos/efectos de los fármacos , Flagelos/fisiología , Eliminación de Gen , Glucolípidos/biosíntesis , Movimiento/efectos de los fármacos , Movimiento/fisiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Quinolonas/metabolismo , Percepción de Quorum , Receptores de Superficie Celular/metabolismo , Transducción de Señal
13.
J Infect Public Health ; 17(2): 329-338, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38194764

RESUMEN

BACKGROUND: Because of the rise in antibiotic resistance and the control of pathogenicity, polymicrobial bacterial biofilms exacerbate wound infections. Since bacterial quorum sensing (QS) signals can dysregulate biofilm development, they are interesting therapeutic treatments. In this study, Pseudomonas Quinolone Signal (PQS) was used to treat an animal model of a wound that had both Staphylococcus aureus and Pseudomonas aeruginosa co-infection. METHODS: S. aureus and P. aeruginosa mono- and co-infection models were developed in vitro on the L-929 cell line and in an animal model of wound infection. Moreover, PQS was extracted and purified using liquid chromatography. Then, the mono- and co-infection models were treated by PQS in vitro and in vivo. RT-PCR analysis was used to look into changes in biofilm, QS, tissue regeneration, and apoptosis genes after the treatment. RESULTS: PQS significantly disrupted established biofilm up to 90% in both in vitro and in vivo models. Moreover, a 93% reduction in the viability of S. aureus and P. aeruginosa was detected during the 10 days of treatment in comparison to control groups. In addition, the biofilm-encoding and QS-regulating genes were down-regulated to 75% in both microorganisms. Also, fewer epithelial cells died when treated with PQS compared to control groups in both mono- and co-infection groups. CONCLUSION: According to this study, PQS may facilitate wound healing by stimulating the immune system and reducing apoptosis. It seems to be a potential medication to use in conjunction with antibiotics to treat infections that are difficult to treat.


Asunto(s)
Coinfección , Pseudomonas aeruginosa , Quinolonas , Animales , Staphylococcus aureus , Coinfección/tratamiento farmacológico , Percepción de Quorum , Biopelículas , Modelos Animales , Proteínas Bacterianas/genética
14.
Biochim Biophys Acta Gen Subj ; 1867(2): 130269, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36379399

RESUMEN

Pseudomonas quinolone signal (PQS) is a quorum-sensing molecule associated with Pseudomonas aeruginosa that regulates quorum sensing, extracellular vesicle biogenesis, iron acquisition, and the secretion of virulence factors. PQS has been shown to have immunomodulatory effects on the host. It induces oxidative stress, modulates cytokine levels, and activates regulated cell death in the host. In this study, we investigated the effects of PQS (10 µM) on host organelle dynamics and dysfunction in human macrophages at the interphase of endoplasmic reticulum (ER), mitochondria, and lysosome. This study showed that PQS increases cytosolic Ca+2 levels and elevates ER stress, as evidenced by increased expression of BiP and activation of the PERK-CHOP axis of unfolded protein response (UPR). Moreover, PQS also negatively affects mitochondria by disrupting mitochondrial membrane potential and increasing mitochondrial ROS generation (mROS). Additionally, PQS stimulation decreased the number of acridine orange-positive lysosomes, indicating lysosomal destabilization. Furthermore, PQS-induced lysosomal destabilization also induces overexpression of the lysosomal stress-responsive gene TFEB. Besides organelle dysfunction, PQS dysregulates inflammation-related genes by upregulating NLRC4, TMS1, and Caspase 1 while downregulating NLRP3 and IL-1ß. Also, PQS increases gene expression of pro-inflammatory cytokines (IL-6, TNF-α, and IFN-γ). In conclusion, our findings suggest that PQS negatively affects human macrophages by interfering with organelle function and dysregulating inflammatory response. Consequently, this study provides crucial insight into PQS-driven macrophage dysfunction and may contribute to a better understanding of Pseudomonas aeruginosa-associated infections.


Asunto(s)
Infecciones por Pseudomonas , Pseudomonas aeruginosa , Humanos , Orgánulos , Macrófagos , Inflamación
15.
mLife ; 2(1): 28-42, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38818333

RESUMEN

Pseudomonas aeruginosa is one of the leading nosocomial pathogens that causes both severe acute and chronic infections. The strong capacity of P. aeruginosa to form biofilms can dramatically increase its antibiotic resistance and lead to treatment failure. The biofilm resident bacterial cells display distinct gene expression profiles and phenotypes compared to their free-living counterparts. Elucidating the genetic determinants of biofilm formation is crucial for the development of antibiofilm drugs. In this study, a high-throughput transposon-insertion site sequencing (Tn-seq) approach was employed to identify novel P. aeruginosa biofilm genetic determinants. When analyzing the novel biofilm regulatory genes, we found that the cell division factor ZapE (PA4438) controls the P. aeruginosa pqs quorum sensing system. The ∆zapE mutant lost fitness against the wild-type PAO1 strain in biofilms and its production of 2-heptyl-3-hydroxy-4(1H)-quinolone (PQS) had been reduced. Further biochemical analysis showed that ZapE interacts with PqsH, which encodes the synthase that converts 2-heptyl-4-quinolone (HHQ) to PQS. In addition, site-directed mutagenesis of the ATPase active site of ZapE (K72A) abolished the positive regulation of ZapE on PQS signaling. As ZapE is highly conserved among the Pseudomonas group, our study suggests that it is a potential drug target for the control of Pseudomonas infections.

16.
Biomolecules ; 12(2)2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35204671

RESUMEN

The opportunistic pathogen Pseudomonas aeruginosa employs quorum sensing to govern the production of many virulence factors. Interference with quorum sensing signaling has therefore been put forward as an attractive approach to disarm this pathogen. Here, we analyzed the quorum quenching properties of natural and engineered (2-alkyl-)3-hydroxy-4(1H)-quinolone 2,4-dioxygenases (HQDs) that inactivate the P. aeruginosa signal molecule PQS (Pseudomonas quinolone signal; 2-heptyl-3-hydroxy-4(1H)-quinolone). When added exogenously to P. aeruginosa cultures, all HQDs tested significantly reduced the levels of PQS and other alkylquinolone-type secondary metabolites deriving from the biosynthetic pathway, such as the respiratory inhibitor 2-heptyl-4-hydroxyquinoline N-oxide. HQDs from Nocardia farcinica and Streptomyces bingchenggensis, which combine low KM values for PQS with thermal stability and resilience in the presence of P. aeruginosa exoproducts, respectively, attenuated production of the virulence factors pyocyanin and pyoverdine. A delay in mortality was observed when Galleria mellonella larvae were infected with P. aeruginosa suspensions treated with the S. bingchenggensis HQD or with inhibitors of alkylquinolone biosynthesis. Our data indicate that quenching of PQS signaling has potential as an anti-virulence strategy; however, an efficient anti-virulence therapy against P. aeruginosa likely requires a combination of agents addressing multiple targets.


Asunto(s)
Dioxigenasas , Quinolonas , Proteínas Bacterianas/metabolismo , Dioxigenasas/metabolismo , Pseudomonas aeruginosa/metabolismo , Piocianina , Quinolonas/farmacología , Percepción de Quorum
17.
Front Microbiol ; 13: 896687, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36187967

RESUMEN

The aryl hydrocarbon receptor (AhR) protein senses microbial-secreted metabolites to trigger the host's innate immune system. The Pseudomonas quinolone signal (PQS) and Mycobacterium tuberculosis (MTb) metabolite phthiocol (Pht) are both ligands of AhR with similar chemical structures. As PQS is an essential quorum-sensing molecule that regulates a wide range of virulence factors in Pseudomonas aeruginosa, we hypothesized that Pht and its analogs are potential P. aeruginosa quorum-sensing inhibitors (QSIs) with immune-modulating functions. In this study, we demonstrated that Pht was able to inhibit the P. aeruginosa pqs QS system and reduce both biofilm formation and the production of pyocyanin. Molecular docking analysis suggested that Pht competes with PQS at the binding site of its receptor, PqsR. An electrophoretic mobility shift assay confirmed the Pht-PqsR interaction and showed that Pht attenuated PqsR from binding to the pqsA promoter. Proteomic analysis showed that synthesis of the key pqs QS proteins decreased upon the addition of Pht to the bacterial cultures. Furthermore, Pht analogs vitamins K1 (Phylloquinone), K2 (Menaquinones), and K3 (Menadione) were also showed to inhibit the P. aeruginosa pqs QS system while able to activate the AhR signaling pathways. Our study suggests that the AhR ligands Pht and its vitamin K analogs are promising QSIs for the alternative treatment of P. aeruginosa infections.

18.
Pharmaceuticals (Basel) ; 15(4)2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35455414

RESUMEN

Biofilm formation is considered a major cause of therapeutic failure because bacteria in biofilms have higher protection against antimicrobials. Thus, biofilm-related infections are extremely challenging to treat and pose major concerns for public health, along with huge economic impacts. Pseudomonas aeruginosa, in particular, is a "critical priority" pathogen, responsible for severe infections, especially in cystic fibrosis patients because of its capacity to form resistant biofilms. Therefore, new therapeutic approaches are needed to complete the pipeline of molecules offering new targets and modes of action. Biofilm formation is mainly controlled by Quorum Sensing (QS), a communication system based on signaling molecules. In the present study, we employed a molecular docking approach (Autodock Vina) to assess two series of chromones-based compounds as possible ligands for PqsR, a LuxR-type receptor. Most compounds showed good predicted affinities for PqsR, higher than the PQS native ligand. Encouraged by these docking results, we synthesized a library of 34 direct and 25 retro chromone carboxamides using two optimized routes from 2-chromone carboxylic acid as starting material for both series. We evaluated the synthesized carboxamides for their ability to inhibit the biofilm formation of P. aeruginosa in vitro. Overall, results showed several chromone 2-carboxamides of the retro series are potent inhibitors of the formation of P. aeruginosa biofilms (16/25 compound with % inhibition ≥ 50% at 50 µM), without cytotoxicity on Vero cells (IC50 > 1.0 mM). The 2,4-dinitro-N-(4-oxo-4H-chromen-2-yl) benzamide (6n) was the most promising antibiofilm compound, with potential for hit to lead optimization.

19.
Sci Total Environ ; 806(Pt 4): 151403, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-34742801

RESUMEN

Quorum sensing signal molecules can be used to regulate the formation of biofilm, but it has not been reported that outer membrane vesicles (OMVs) can package and mediate signal molecules to regulate biofilm. We isolated and purified OMVs packaged with Pseudomonas quinolone signal (PQS) released by Pseudomonas aeruginosa and studied the effects of OMV-mediated PQS on the formation and structure of biofilms. OMV-mediated PQS promoted the growth of biofilm, and the cells in the biofilm were stretched, deformed and "bridged" with the surrounding cells. Raman spectrometry showed that the structure and components of the extracellular polymeric substances of P. aeruginosa changed; moreover extracellular proteins rather than polysaccharides played the dominant role in the formation of P. aeruginosa biofilms when regulated by OMV-mediated PQS. In the combination biofilm formed by P. aeruginosa and Staphylococcus aureus, the mediation of OMVs enhanced the inhibitory effect of PQS to the growth of S. aureus, resulting a decrease in EPS produced by the two bacteria. OMV-mediated PQS led to changes in the biodiversity, richness and structure of the microbial community in biofilms formed by active sludge. This work reveals the mechanism of OMVs mediated signal molecules regulating biofilm, which lays a new theoretical and practical foundation for guiding the operation of low-level of biofouling MBRs.


Asunto(s)
Percepción de Quorum , Staphylococcus aureus , Biopelículas , Pseudomonas aeruginosa
20.
Anal Chim Acta ; 1199: 339574, 2022 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-35227378

RESUMEN

Here a highly selective molecular imprinting polymer was developed to attenuate biofilm formation of the multidrug-resistant pathogen Pseudomonas aeruginosa by disrupting the intermolecular signaling system. Firstly, a dummy template molecular imprinting polymer (MIP) was rationally designed through molecular modeling to capture 2-heptyl-3-hydroxy-4-quinolone (Pseudomonas quinolone signal). This multifunctional signaling molecule interferes with the pathogenicity of P. aeruginosa as an auto-inducer. Then, the synthesized MIP and the non-imprinted polymer (NIP) as reference polymer were evaluated for their binding capacity and biofilm inhibition. The results indicated a significant difference in biofilm inhibition (∼56%) between imprinted (∼67%) and non-imprinted (∼11%) polymer, which is an impressive level, especially for the treatment of various surfaces affected by P. aeruginosa. These results open a new window in the special biological application of MIPs as a promising candidate to reduce concerns in clinical or industrial issues by preventing microbial infections.


Asunto(s)
Impresión Molecular , Percepción de Quorum , Biopelículas , Impresión Molecular/métodos , Polímeros/química , Pseudomonas aeruginosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA