Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 171(6): 1354-1367.e20, 2017 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-29103614

RESUMEN

A number of bacterial cell processes are confined functional membrane microdomains (FMMs), structurally and functionally similar to lipid rafts of eukaryotic cells. How bacteria organize these intricate platforms and what their biological significance is remain important questions. Using the pathogen methicillin-resistant Staphylococcus aureus (MRSA), we show here that membrane-carotenoid interaction with the scaffold protein flotillin leads to FMM formation, which can be visualized using super-resolution array tomography. These membrane platforms accumulate multimeric protein complexes, for which flotillin facilitates efficient oligomerization. One of these proteins is PBP2a, responsible for penicillin resistance in MRSA. Flotillin mutants are defective in PBP2a oligomerization. Perturbation of FMM assembly using available drugs interferes with PBP2a oligomerization and disables MRSA penicillin resistance in vitro and in vivo, resulting in MRSA infections that are susceptible to penicillin treatment. Our study demonstrates that bacteria possess sophisticated cell organization programs and defines alternative therapies to fight multidrug-resistant pathogens using conventional antibiotics.


Asunto(s)
Microdominios de Membrana/metabolismo , Staphylococcus aureus Resistente a Meticilina/fisiología , Infecciones Estafilocócicas/microbiología , Animales , Proteínas Bacterianas/metabolismo , Carotenoides/metabolismo , Membrana Celular/metabolismo , Femenino , Microdominios de Membrana/química , Proteínas de la Membrana/metabolismo , Staphylococcus aureus Resistente a Meticilina/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Proteínas de Unión a las Penicilinas/metabolismo , Xantófilas/metabolismo
2.
Cell Chem Biol ; 24(7): 845-857.e6, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28669526

RESUMEN

Scaffold proteins are ubiquitous chaperones that bind proteins and facilitate physical interaction of multi-enzyme complexes. Here we used a biochemical approach to dissect the scaffold activity of the flotillin-homolog protein FloA of the multi-drug-resistant human pathogen Staphylococcus aureus. We show that FloA promotes oligomerization of membrane protein complexes, such as the membrane-associated RNase Rny, which forms part of the RNA-degradation machinery called the degradosome. Cells lacking FloA had reduced Rny function and a consequent increase in the targeted sRNA transcripts that negatively regulate S. aureus toxin expression. Small molecules that altered FloA oligomerization also reduced Rny function and decreased the virulence potential of S. aureus in vitro, as well as in vivo, using invertebrate and murine infection models. Our results suggest that flotillin assists in the assembly of protein complexes involved in S. aureus virulence, and could thus be an attractive target for the development of new antimicrobial therapies.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Staphylococcus aureus/patogenicidad , Virulencia , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Femenino , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Fosforilcolina/análogos & derivados , Fosforilcolina/química , Fosforilcolina/farmacología , Fosforilcolina/uso terapéutico , Multimerización de Proteína/efectos de los fármacos , ARN Bacteriano/genética , ARN Bacteriano/aislamiento & purificación , ARN Bacteriano/metabolismo , ARN Ribosómico 5S/genética , ARN Ribosómico 5S/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/mortalidad , Infecciones Estafilocócicas/patología , Staphylococcus aureus/metabolismo , Tasa de Supervivencia , Técnicas del Sistema de Dos Híbridos , Virulencia/efectos de los fármacos , Virulencia/genética
3.
Curr Opin Microbiol ; 36: 76-84, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28237903

RESUMEN

Recent studies show that internal organization of bacterial cells is more complex than previously appreciated. A clear example of this is the assembly of the nanoscale membrane platforms termed functional membrane microdomains. The lipid composition of these regions differs from that of the surrounding membrane; these domains confine a set of proteins involved in specific cellular processes such as protease secretion and signal transduction. It is currently thought that functional membrane microdomains act as oligomerization platforms and promote efficient oligomerization of interacting protein partners in bacterial membranes. In this review, we highlight the most noteworthy achievements, challenges and controversies of this emerging research field over the past five years.


Asunto(s)
Archaea/fisiología , Membrana Celular/metabolismo , Bacterias Gramnegativas/fisiología , Bacterias Grampositivas/fisiología , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Archaea/genética , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Membrana Celular/química , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/genética , Bacterias Grampositivas/metabolismo , Microdominios de Membrana/genética , Proteínas de la Membrana/química , Transporte de Proteínas , Transducción de Señal
4.
PLoS Genet ; 11(4): e1005140, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25909364

RESUMEN

Lipid rafts are membrane microdomains specialized in the regulation of numerous cellular processes related to membrane organization, as diverse as signal transduction, protein sorting, membrane trafficking or pathogen invasion. It has been proposed that this functional diversity would require a heterogeneous population of raft domains with varying compositions. However, a mechanism for such diversification is not known. We recently discovered that bacterial membranes organize their signal transduction pathways in functional membrane microdomains (FMMs) that are structurally and functionally similar to the eukaryotic lipid rafts. In this report, we took advantage of the tractability of the prokaryotic model Bacillus subtilis to provide evidence for the coexistence of two distinct families of FMMs in bacterial membranes, displaying a distinctive distribution of proteins specialized in different biological processes. One family of microdomains harbors the scaffolding flotillin protein FloA that selectively tethers proteins specialized in regulating cell envelope turnover and primary metabolism. A second population of microdomains containing the two scaffolding flotillins, FloA and FloT, arises exclusively at later stages of cell growth and specializes in adaptation of cells to stationary phase. Importantly, the diversification of membrane microdomains does not occur arbitrarily. We discovered that bacterial cells control the spatio-temporal remodeling of microdomains by restricting the activation of FloT expression to stationary phase. This regulation ensures a sequential assembly of functionally specialized membrane microdomains to strategically organize signaling networks at the right time during the lifespan of a bacterium.


Asunto(s)
Bacillus subtilis/genética , Microdominios de Membrana/genética , Proteínas de la Membrana/genética , Regulación Bacteriana de la Expresión Génica , Proteínas de la Membrana/biosíntesis , Transporte de Proteínas/genética , Transducción de Señal/genética
5.
Mol Cancer ; 14: 38, 2015 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-25742883

RESUMEN

BACKGROUND: The metastasis-associated in colon cancer 1 (MACC1) gene has been identified as prognostic biomarker for colorectal cancer (CRC). Here, we aimed at the refinement of risk assessment by separate and combined survival analyses of MACC1 expression with any of the markers KRAS mutated in codon 12 (KRAS G12) or codon 13 (KRAS G13), BRAF V600 mutation and MSI status in a retrospective study of 99 CRC patients with tumors UICC staged I, II and III. FINDINGS: We showed that only high MACC1 expression (HR: 6.09, 95% CI: 2.50-14.85, P < 0.001) and KRAS G13 mutation (HR: 5.19, 95% CI: 1.06-25.45, P = 0.042) were independent prognostic markers for shorter metastasis-free survival (MFS). Accordingly, Cox regression analysis revealed that patients with high MACC1 expression and KRAS G13 mutation exhibited the worst prognosis (HR: 14.48, 95% CI: 3.37-62.18, P < 0.001). Patients were classified based on their molecular characteristics into four clusters with significant differences in MFS (P = 0.003) by using the SPSS 2-step cluster function and Kaplan-Meier survival analysis. CONCLUSION: According to our results, patients with high MACC1 expression and mutated KRAS G13 exhibited the highest risk for metachronous metastases formation. Moreover, we demonstrated that the "Traditional pathway" with an intermediate risk for metastasis formation can be further subdivided by assessing MACC1 expression into a low and high risk group with regard to MFS prognosis. This is the first report showing that identification of CRC patients at high risk for metastasis is possible by assessing MACC1 expression in combination with KRAS G13 mutation.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Expresión Génica , Mutación , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Proteínas ras/genética , Adulto , Anciano , Análisis por Conglomerados , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Femenino , Humanos , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras) , Transactivadores , Factores de Transcripción/metabolismo
6.
Cell ; 158(5): 1060-1071, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171407

RESUMEN

Antibiotic resistance is a key medical concern, with antibiotic use likely being an important cause. However, here we describe an alternative route to clinically relevant antibiotic resistance that occurs solely due to competitive interactions among bacterial cells. We consistently observe that isolates of Methicillin-resistant Staphylococcus aureus diversify spontaneously into two distinct, sequentially arising strains. The first evolved strain outgrows the parent strain via secretion of surfactants and a toxic bacteriocin. The second is resistant to the bacteriocin. Importantly, this second strain is also resistant to intermediate levels of vancomycin. This so-called VISA (vancomycin-intermediate S. aureus) phenotype is seen in many hard-to-treat clinical isolates. This strain diversification also occurs during in vivo infection in a mouse model, which is consistent with the fact that both coevolved phenotypes resemble strains commonly found in clinic. Our study shows how competition between coevolving bacterial strains can generate antibiotic resistance and recapitulate key clinical phenotypes.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/clasificación , Staphylococcus aureus Resistente a Meticilina/genética , Infecciones Estafilocócicas/microbiología , Secuencia de Aminoácidos , Animales , Antibacterianos/farmacología , Bacteriocinas/genética , Bacteriocinas/metabolismo , Biopelículas/efectos de los fármacos , Evolución Biológica , Femenino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/fisiología , Ratones Endogámicos BALB C , Fenómenos Microbiológicos , Datos de Secuencia Molecular , Pigmentación , Alineación de Secuencia , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/clasificación , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/fisiología , Vancomicina/farmacología
7.
FEMS Microbiol Lett ; 356(1): 62-70, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24863934

RESUMEN

Bacterial communication via the secretion of small diffusible compounds allows microorganisms to regulate gene expression in a coordinated manner. As many virulence traits are regulated in this fashion, disruption of chemical communication has been proposed as novel antimicrobial therapy. Quorum-quenching enzymes have been a promising discovery in this field as they interfere with the communication of Gram-negative bacteria. AHL-lactonases and AHL-acylases have been described in a variety of bacterial strains; however, usually only one of these two groups of enzymes has been described in a single species. We report here the presence of a member of each group of enzymes in the extremophile bacterium Deinococcus radiodurans. Co-occurrence of both enzymes in a single species increases the chance of inactivating foreign AHL signals under different conditions. We demonstrate that both enzymes are able to degrade the quorum-sensing molecules of various pathogens subsequently affecting virulence gene expression. These studies add the quorum-quenching enzymes of D. radiodurans to the list of potent quorum-quenchers and highlight the idea that quorum quenching could have evolved in some bacteria as a strategy to gain a competitive advantage by altering gene expression in other species.


Asunto(s)
Amidohidrolasas/fisiología , Proteínas Bacterianas/fisiología , Hidrolasas de Éster Carboxílico/fisiología , Deinococcus/enzimología , Percepción de Quorum , Acil-Butirolactonas/metabolismo , Amidohidrolasas/química , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Caenorhabditis elegans/microbiología , Hidrolasas de Éster Carboxílico/química , Datos de Secuencia Molecular , Pseudomonas aeruginosa/fisiología
8.
J Bacteriol ; 196(14): 2681-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24816606

RESUMEN

The iron binding siderophore pyoverdine constitutes a major adaptive factor contributing to both virulence and survival in fluorescent pseudomonads. For decades, pyoverdine production has allowed the identification and classification of fluorescent and nonfluorescent pseudomonads. Here, we demonstrate that PvdP, a periplasmic enzyme of previously unknown function, is a tyrosinase required for the maturation of the pyoverdine chromophore in Pseudomonas aeruginosa. PvdP converts the nonfluorescent ferribactin, containing two iron binding groups, into a fluorescent pyoverdine, forming a strong hexadentate complex with ferrous iron, by three consecutive oxidation steps. PvdP represents the first characterized member of a small family of tyrosinases present in fluorescent pseudomonads that are required for siderophore maturation and are capable of acting on large peptidic substrates.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Monofenol Monooxigenasa/metabolismo , Oligopéptidos/metabolismo , Pseudomonas aeruginosa/enzimología , Proteínas Bacterianas/genética , Dominio Catalítico , Regulación Bacteriana de la Expresión Génica/fisiología , Modelos Moleculares , Monofenol Monooxigenasa/genética , Oligopéptidos/genética , Filogenia , Conformación Proteica , Transporte de Proteínas , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
9.
Methods Mol Biol ; 1149: 681-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24818942

RESUMEN

Pseudomonas aeruginosa is an opportunistic human pathogen responsible for severe to deadly infections in patients suffering from cystic fibrosis, AIDS, undergoing immune suppressing therapies or suffering from severe burns. In the recent years there has been an increasing interest in exploring animal infection models that, to a certain extent, could mimic human infections. Here we describe the use of the larvae of the greater wax moth Galleria mellonella as a non-expensive, easy-to-use, and easy-to-obtain animal model to study P. aeruginosa infections.


Asunto(s)
Bioensayo/métodos , Interacciones Huésped-Patógeno , Mariposas Nocturnas/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Animales , Antiinfecciosos/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Insulina , Mamíferos , Pseudomonas aeruginosa/efectos de los fármacos , Virulencia/efectos de los fármacos
10.
Appl Environ Microbiol ; 80(13): 3868-78, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24747904

RESUMEN

Protein localization has been traditionally explored in unicellular organisms, whose ease of genetic manipulation facilitates molecular characterization. The two rod-shaped bacterial models Escherichia coli and Bacillus subtilis have been prominently used for this purpose and have displaced other bacteria whose challenges for genetic manipulation have complicated any study of cell biology. Among these bacteria is the spherical pathogenic bacterium Staphylococcus aureus. In this report, we present a new molecular toolbox that facilitates gene deletion in staphylococci in a 1-step recombination process and additional vectors that facilitate the insertion of diverse reporter fusions into newly identified neutral loci of the S. aureus chromosome. Insertion of the reporters does not add any antibiotic resistance genes to the chromosomes of the resultant strains, thereby making them amenable for further genetic manipulations. We used this toolbox to reconstitute the expression of mreB in S. aureus, a gene that encodes an actin-like cytoskeletal protein which is absent in coccal cells and is presumably lost during the course of speciation. We observed that in S. aureus, MreB is organized in discrete structures in association with the membrane, leading to an unusual redistribution of the cell wall material. The production of MreB also caused cell enlargement, but it did not revert staphylococcal shape. We present interactions of MreB with key staphylococcal cell wall-related proteins. This work facilitates the use S. aureus as a model system in exploring diverse aspects of cellular microbiology.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Expresión Génica , Biología Molecular/métodos , Plásmidos , Staphylococcus aureus/genética , Fusión Artificial Génica , Eliminación de Gen , Genes Reporteros , Genética Microbiana/métodos , Recombinación Genética
11.
Proc Natl Acad Sci U S A ; 111(4): 1568-73, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24474783

RESUMEN

The use of enzymes to interfere with quorum sensing represents an attractive strategy to fight bacterial infections. We used PvdQ, an effective quorum-quenching enzyme from Pseudomonas aeruginosa, as a template to generate an acylase able to effectively hydrolyze C8-HSL, the major communication molecule produced by the Burkholderia species. We discovered that the combination of two single mutations leading to variant PvdQ(Lα146W,Fß24Y) conferred high activity toward C8-HSL. Exogenous addition of PvdQ(Lα146W,Fß24Y) dramatically decreased the amount of C8-HSL present in Burkholderia cenocepacia cultures and inhibited a quorum sensing-associated phenotype. The efficacy of this PvdQ variant to combat infections in vivo was further confirmed by its ability to rescue Galleria mellonella larvae upon infection, demonstrating its potential as an effective agent toward Burkholderia infections. Kinetic analysis of the enzymatic activities toward 3-oxo-C12-L-HSL and C8-L-HSL corroborated a substrate switch. This work demonstrates the effectiveness of quorum-quenching acylases as potential novel antimicrobial drugs. In addition, we demonstrate that their substrate range can be easily switched, thereby paving the way to selectively target only specific bacterial species inside a complex microbial community.


Asunto(s)
Amidohidrolasas/metabolismo , Burkholderia cenocepacia/patogenicidad , Percepción de Quorum , Amidohidrolasas/química , Animales , Burkholderia cenocepacia/enzimología , Cinética , Larva/microbiología , Modelos Moleculares , Mariposas Nocturnas/crecimiento & desarrollo , Mariposas Nocturnas/microbiología , Especificidad por Sustrato , Virulencia
12.
Mol Microbiol ; 86(2): 457-71, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22882210

RESUMEN

Biofilm formation in Bacillus subtilis requires the differentiation of a subpopulation of cells responsible for the production of the extracellular matrix that structures the biofilm. Differentiation of matrix-producing cells depends, among other factors, on the FloT and YqfA proteins. These proteins are present exclusively in functional membrane microdomains of B. subtilis and are homologous to the eukaryotic lipid raft-specific flotillin proteins. In the absence of FloT and YqfA, diverse proteins normally localized to the membrane microdomains of B. subtilis are not functional. Here we show that the absence of FloT and YqfA reduces the level of the septal-localized protease FtsH. The flotillin homologues FloT and YqfA are occasionally present at the midcell in exponentially growing cells and the absence of FloT and YqfA negatively affects FtsH concentration. Biochemical experiments indicate a direct interaction between FloT/YqfA and FtsH. Moreover, FtsH is essential for the differentiation of matrix producers and hence, biofilm formation. This molecular trigger of biofilm formation may therefore be used as a target for the design of new biofilm inhibitors. Accordingly, we show that the small protein SpoVM, known to bind to and inhibit FtsH activity, inhibits biofilm formation in B. subtilis and other distantly related bacteria.


Asunto(s)
Bacillus subtilis/enzimología , Proteínas Bacterianas/metabolismo , Biopelículas , Proteínas de la Membrana/genética , Péptido Hidrolasas/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Proteínas de la Membrana/metabolismo , Mutación , Péptido Hidrolasas/genética , Unión Proteica
13.
Microbiol Mol Biol Rev ; 76(1): 46-65, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22390972

RESUMEN

Cell-to-cell communication is a major process that allows bacteria to sense and coordinately react to the fluctuating conditions of the surrounding environment. In several pathogens, this process triggers the production of virulence factors and/or a switch in bacterial lifestyle that is a major determining factor in the outcome and severity of the infection. Understanding how bacteria control these signaling systems is crucial to the development of novel antimicrobial agents capable of reducing virulence while allowing the immune system of the host to clear bacterial infection, an approach likely to reduce the selective pressures for development of resistance. We provide here an up-to-date overview of the molecular basis and physiological implications of cell-to-cell signaling systems in Gram-negative bacteria, focusing on the well-studied bacterium Pseudomonas aeruginosa. All of the known cell-to-cell signaling systems in this bacterium are described, from the most-studied systems, i.e., N-acyl homoserine lactones (AHLs), the 4-quinolones, the global activator of antibiotic and cyanide synthesis (GAC), the cyclic di-GMP (c-di-GMP) and cyclic AMP (cAMP) systems, and the alarmones guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp), to less-well-studied signaling molecules, including diketopiperazines, fatty acids (diffusible signal factor [DSF]-like factors), pyoverdine, and pyocyanin. This overview clearly illustrates that bacterial communication is far more complex than initially thought and delivers a clear distinction between signals that are quorum sensing dependent and those relying on alternative factors for their production.


Asunto(s)
Pseudomonas aeruginosa/patogenicidad , Animales , Comunicación Celular/genética , Comunicación Celular/fisiología , Humanos , Transducción de Señal/genética , Transducción de Señal/fisiología , Virulencia/genética , Virulencia/fisiología
14.
Biol Chem ; 392(3): 169-87, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21194378

RESUMEN

The discovery of the DP4-related enzymes DP8 and DP9 raised controversial discussion regarding the physiological and pathophysiological function of distinct members of the DP4 family. Particularly with regard to their potential relevance in regulating immune functions, it is of interest to know which role the subcellular distribution of the enzymes play. Synthetic substrates as well as low molecular weight inhibitors are widely used as tools, but little is yet known regarding their features in cell experiments, such as their plasma membrane penetration capacity. The fluorogenic substrates Gly-Pro-AMC or (Ala-Pro)2-R110 predominantly detect plasma membrane-bound activities of viable cells (less than 0.1% of fluorochromes R110 or AMC inside viable cells after 1 h incubation). Additionally, the selective and non-selective DP8/9 inhibitors allo-Ile-isoindoline and Lys[Z(NO2)]-pyrrolidide were found to be incapable of passing the plasma membrane easily. This suggests that previously reported cellular effects are not due to inhibition of the cytosolic enzymes DP8 or DP9. Moreover, our enzymatic studies with viable cells provided evidence that DP8 and/or DP9 are also present on the surface of immune cells under certain circumstances and could gain relevance particularly in the absence of DP4 expression. In summary, in cells which do express DP4 on the surface, this archetypical member of the DP4 family is the most relevant peptidase in the regulation of cellular functions.


Asunto(s)
Dipeptidasas/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Espacio Intracelular/metabolismo , Animales , Línea Celular , ADN/biosíntesis , Dipeptidil Peptidasa 4/genética , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Colorantes Fluorescentes/metabolismo , Humanos , Indoles/metabolismo , Isoleucina/análogos & derivados , Isoleucina/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Ratones , Ratones Noqueados , Neuropéptido Y/metabolismo , Pirrolidinas/metabolismo , Especificidad por Sustrato , Linfocitos T/enzimología
15.
Environ Microbiol Rep ; 2(3): 433-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23766117

RESUMEN

Pyoverdine biosynthesis in fluorescent Pseudomonas spp. and especially in the opportunistic human pathogen Pseudomonas aeruginosa has been extensively studied. The acylase PvdQ is required for a maturation step in pyoverdine biosynthesis but also has been proven to be effective in degrading long-chain N-acyl homoserine lactones (AHLs). These molecules are used as quorum-sensing molecules by Gram-negative bacteria such as Pseudomonads themselves. Interestingly, the pvdQ gene is part of a pyoverdine cluster in P. aeruginosa and P. syringae but not in other fluorescent Pseudomonas spp. In this study we have compared the activities of PvdQ orthologues from various species and provide evidence for conserved functions in Pseudomonas fluorescens PfO-1, P. putida KT2440 and P. aeruginosa PA14. Despite large differences in genomic organization, expression of each of these pvdQ orthologues is regulated by iron availability. Moreover, PvdQ and its orthologues have conserved substrate specificity for AHLs and play a role in pyoverdine production in all tested Pseudomonas species. These data strongly suggest that the role of PvdQ in pyoverdine biosynthesis is conserved among Pseudomonas spp., while the control that PvdQ exerts in P. aeruginosa over its own quorum-sensing signals seems to be unique to this bacterium.

16.
Microbiology (Reading) ; 156(Pt 1): 49-59, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19778968

RESUMEN

PvdQ, an acylase from Pseudomonas aeruginosa PAO1, has been shown to have at least two functions. It can act as a quorum quencher due to its ability to degrade long-chain N-acylhomoserine lactones (AHLs), e.g. 3-oxo-C12-HSL, leading to a decrease in virulence factors. In addition, PvdQ is involved in iron homeostasis by playing a role in the biosynthesis of pyoverdine, the major siderophore of P. aeruginosa. In accordance with earlier studies on RNA level, we could show at the protein level that PvdQ is only expressed when iron is present at very low concentrations. We therefore set out to investigate the two functions of PvdQ under iron-limiting conditions. Gene deletion of pvdQ does not affect growth of P. aeruginosa but abrogates pyoverdine production, and results in an accumulation of 3-oxo-C12-HSL. Phenotypic analyses of our DeltapvdQ mutant at low iron concentrations revealed that this mutant is impaired in swarming motility and biofilm formation. Additionally, a plant and a Caenorhabditis elegans infection model demonstrated that the deletion of pvdQ resulted in reduced virulence. None of the phenotypes in the present study could be linked to the presence or absence of AHLs. These results clearly indicate that under iron-limiting conditions PvdQ plays a major role in swarming motility, in biofilm development and in infection that is more likely to be linked to the pyoverdine pathway rather than the LasI/LasR/3-oxo-C12-HSL quorum-sensing circuit.


Asunto(s)
Amidohidrolasas/metabolismo , Proteínas Bacterianas/metabolismo , Hierro/metabolismo , Pseudomonas aeruginosa/genética , 4-Butirolactona/análogos & derivados , 4-Butirolactona/biosíntesis , Amidohidrolasas/genética , Animales , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Caenorhabditis elegans/microbiología , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Homoserina/análogos & derivados , Homoserina/biosíntesis , Oligopéptidos/biosíntesis , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/patogenicidad , Solanum tuberosum/microbiología , Virulencia
17.
Antimicrob Agents Chemother ; 53(11): 4891-7, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19721066

RESUMEN

The Pseudomonas aeruginosa PAO1 gene pvdQ encodes an acyl-homoserine lactone (AHL) acylase capable of degrading N-(3-oxododecanoyl)-L-homoserine lactone by cleaving the AHL amide. PvdQ has been proven to function as a quorum quencher in vitro in a number of phenotypic assays. To address the question of whether PvdQ also shows quorum-quenching properties in vivo, an infection model based on the nematode Caenorhabditis elegans was explored. In a fast-acting paralysis assay, strain PAO1(pMEpvdQ), which overproduces PvdQ, was shown to be less virulent than the wild-type strain. More than 75% of the nematodes exposed to PAO1(pMEpvdQ) survived and continued to grow when using this strain as a food source. Interestingly, in a slow-killing assay monitoring the survival of the nematodes throughout a 4-day course, strain PAO1-Delta pvdQ was shown to be more virulent than the wild-type strain, confirming the role of PvdQ as a virulence-reducing agent. It was observed that larval stage 1 (L1) to L3-stage larvae benefit much more from protection by PvdQ than L4 worms. Finally, purified PvdQ protein was added to C. elegans worms infected with wild-type PAO1, and this resulted in reduced pathogenicity and increased the life span of the nematodes. From our observations we can conclude that PvdQ might be a strong candidate for antibacterial therapy against Pseudomonas infections.


Asunto(s)
Acil-Butirolactonas/metabolismo , Amidohidrolasas/fisiología , Caenorhabditis elegans/microbiología , Pseudomonas aeruginosa/enzimología , Percepción de Quorum , 4-Butirolactona/análogos & derivados , 4-Butirolactona/análisis , Animales , Caenorhabditis elegans/efectos de los fármacos , Modelos Animales de Enfermedad , Homoserina/análogos & derivados , Homoserina/análisis , Cianuro de Hidrógeno/toxicidad , Pseudomonas aeruginosa/patogenicidad , Virulencia
18.
Clin Chem Lab Med ; 47(3): 253-61, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19327105

RESUMEN

BACKGROUND: In the past, different research groups could show that treatment of immune cells with inhibitors of post-proline splitting dipeptidyl aminopeptidases leads to functional changes in the immune system consistent with immunosuppression. This is due to the inhibition of proliferation of lymphocytes and the production of inflammatory cytokines of the TH1, TH2, and TH17, cells as well as the induction of immunosuppressive cytokines, such as transforming growth factor-beta1 (TGF-beta1) and interleukin (IL)-1RA. Until recently, most of the effects of these inhibitors on immune functions were attributed to the inhibition of dipeptidyl aminopeptidase IV (DPIV/CD26). With the identification of new peptidases of the DPIV family (DASH) with the same or similar substrate specificity [fibroblast activation protein (FAP), DP8/9], the question arose whether and to what extent the inhibition of intracellularly localized enzymes, DP8 and DP9, contribute to the observed immunosuppression. In addition, members of the aminopeptidase N (APN) family are also involved in the regulation of immune functions. Hence, the concept of a combined targeting of both families of peptidases for treatment of inflammatory diseases is a promising strategy. RESULTS/CONCLUSIONS: Summarizing data obtained from the usage of different non-selective and selective inhibitors of DPIV, DP8/9, FAP, and DPII, this review provides evidence that in addition to DPIV, DP8/9 also regulate the immune response via modulation of cell cycle progression and cytokine production. The strongest and most consistent effects in vitro were, however, observed with non-selective inhibitors for the suppression of DNA synthesis and cytokine production. Similar effects were provoked by APN inhibitors, which were also found to suppress DNA synthesis and the production of inflammatory cytokines in vitro. However, different mechanisms and signaling pathways appear to mediate the cellular effects resulting from the inhibition of either APN or DPIV family members. In particular, members of the APN family uniquely influence the function of CD4+CD25+ regulatory T-cells. Consequently, the concomitant inhibition of both APN and DPIV enzyme families by means of two separate inhibitors or by binary inhibitors with specificity for both enzyme families (PETIR, peptidase targeted immunoregulation) synergistically affects immune cells on the level of cell cycle regulation, suppression of TH1, TH2, and TH17 cytokines as well as the activation of regulatory T-cells. Besides leukocytes, dermal cells as sebocytes, keratinocytes, and fibroblasts are also targeted by these inhibitors. This strongly suggests a broad potential of the multiple anti-inflammatory effects of PETIR in treatment of chronic inflammatory diseases, such as autoimmune diseases, allergies, and transplant rejections, as well as of inflammatory skin diseases, such as acne, psoriasis, rosacea or atopic dermatitis. The first active dual inhibitor, IP10.C8, has been developed by IMTM for the treatment of inflammatory skin diseases and has just entered the first phase II study.


Asunto(s)
Antígenos CD13/inmunología , Dipeptidil Peptidasa 4/inmunología , Animales , Inhibidores de la Dipeptidil-Peptidasa IV , Humanos , Inhibidores de Proteasas/farmacología , Enfermedades de la Piel/tratamiento farmacológico , Enfermedades de la Piel/inmunología
19.
FEBS J ; 274(21): 5600-10, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17922842

RESUMEN

There is strong interest in creating an enzyme that can deacylate natural cephalosporins such as cephalosporin C in order to efficiently acquire the starting compound for the industrial production of semisynthetic cephalosporin antibiotics. In this study, the active site of the glutaryl acylase from Pseudomonas SY-77 was randomized rationally. Several mutations that were found in previous studies to enhance the activity of the enzyme towards adipyl-7-aminodesacetoxycephalosporanic acid (ADCA) and cephalosporin C have now been combined, and libraries have been made in which random amino acid substitutions at these positions are joined. The mutants were expressed in a leucine-deficient Escherichia coli strain and subjected to growth selection with adipyl-leucine or amino-adipyl-leucine as sole leucine source. The mutants growing on these media were selected and purified, and their hydrolysis activities towards adipyl-7-ADCA and cephalosporin C were tested. Several mutants with highly improved activities towards the desired substrates were found in these rationally randomized libraries. The best mutant was selected from a library of totally randomized residues: 178, 266, and 375. This mutant comprises two mutations, Y178F + F375H, which synergistically improve the catalytic efficiency towards adipyl-7-ADCA 36-fold. The activity of this mutant towards adipyl-7-ADCA is 50% of the activity of the wild-type enzyme towards the preferred substrate glutaryl-7-aminocephalosporanic acid, and therefore the characteristics of this mutant approach those needed for industrial application.


Asunto(s)
Cefalosporinas/metabolismo , Penicilina Amidasa/química , Sitios de Unión , Catálisis , Cefalosporinas/química , Ingeniería Genética , Modelos Moleculares , Mutagénesis , Mutación , Penicilina Amidasa/genética , Penicilina Amidasa/aislamiento & purificación , Penicilina Amidasa/metabolismo , Pseudomonas/enzimología , Especificidad por Sustrato
20.
Mol Microbiol ; 60(6): 1474-89, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16796682

RESUMEN

Quorum sensing, the population density-dependent regulation mediated by N-acylhomoserine lactones (AHSL), is essential for the control of virulence in the plant pathogen Erwinia carotovora ssp. carotovora (Ecc). In Erwinia carotovora ssp. the AHSL signal with an acyl chain of either 6 or 8 carbons is generated by an AHSL synthase, the expI gene product. This work demonstrates that the AHSL receptor, ExpR1, of Ecc strain SCC3193 has strict specificity for the cognate AHSL 3-oxo-C8-HSL. We have also identified a second AHSL receptor (ExpR2) and demonstrate a novel quorum sensing mechanism, where ExpR2 acts synergistically with the previously described ExpR1 to repress virulence gene expression in Ecc. We show that this repression is released by addition of AHSLs and appears to be largely mediated via the negative regulator RsmA. Additionally we show that ExpR2 has the novel property to sense AHSLs with different acyl chain lengths. The expI expR1 double mutant is able to act in response to a number of different AHSLs, while the expI expR2 double mutant can only respond to the cognate signal of Ecc strain SCC3193. These results suggest that Ecc is able to react both to the cognate AHSL signal and the signals produced by other bacterial species.


Asunto(s)
Proteínas Bacterianas/fisiología , Regulación Bacteriana de la Expresión Génica , Pectobacterium carotovorum/patogenicidad , Enfermedades de las Plantas/microbiología , Proteínas Represoras/fisiología , 4-Butirolactona/análogos & derivados , 4-Butirolactona/metabolismo , 4-Butirolactona/farmacología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Pared Celular/enzimología , Celulasa/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Homoserina/análogos & derivados , Homoserina/metabolismo , Homoserina/farmacología , Datos de Secuencia Molecular , Pectobacterium carotovorum/genética , Fenotipo , Proteínas de Plantas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Transducción de Señal/genética , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...