Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(21)2023 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-37958979

RESUMEN

Bacterial contamination during space missions is problematic for human health and damages filters and other vital support systems. Staphylococcus aureus is both a human commensal and an opportunistic pathogen that colonizes human tissues and causes acute and chronic infections. Virulence and colonization factors are positively and negatively regulated, respectively, by bacterial cell-to-cell communication (quorum sensing) via the agr (accessory gene regulator) system. When cultured under low-shear modelled microgravity conditions (LSMMG), S. aureus has been reported to maintain a colonization rather than a pathogenic phenotype. Here, we show that the modulation of agr expression via reduced production of autoinducing peptide (AIP) signal molecules was responsible for this behavior. In an LSMMG environment, the S. aureus strains JE2 (methicillin-resistant) and SH1000 (methicillin-sensitive) both exhibited reduced cytotoxicity towards the human leukemia monocytic cell line (THP-1) and increased fibronectin binding. Using S. aureus agrP3::lux reporter gene fusions and mass spectrometry to quantify the AIP concentrations, the activation of agr, which depends on the binding of AIP to the transcriptional regulator AgrC, was delayed in the strains with an intact autoinducible agr system. This was because AIP production was reduced under these growth conditions compared with the ground controls. Under LSMMG, S. aureus agrP3::lux reporter strains that cannot produce endogenous AIPs still responded to exogenous AIPs. Provision of exogenous AIPs to S. aureus USA300 during microgravity culture restored the cytotoxicity of culture supernatants for the THP-1 cells. These data suggest that microgravity does not affect AgrC-AIP interactions but more likely the generation of AIPs.


Asunto(s)
Infecciones Estafilocócicas , Ingravidez , Humanos , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Staphylococcus aureus/metabolismo , Proteínas Quinasas/metabolismo , Percepción de Quorum/genética , Regulación hacia Abajo , Péptidos/metabolismo , Proteínas Bacterianas/metabolismo
2.
Front Chem ; 11: 1113885, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37214482

RESUMEN

Virulence gene expression in the human pathogen, S. aureus is regulated by the agr (accessory gene regulator) quorum sensing (QS) system which is conserved in diverse Gram-positive bacteria. The agr QS signal molecule is an autoinducing peptide (AIP) generated via the initial processing of the AgrD pro-peptide by the transmembrane peptidase AgrB. Since structural information for AgrB and AgrBD interactions are lacking, we used homology modelling and molecular dynamics (MD) annealing to characterise the conformations of AgrB and AgrD in model membranes and in solution. These revealed a six helical transmembrane domain (6TMD) topology for AgrB. In solution, AgrD behaves as a disordered peptide, which binds N-terminally to membranes in the absence and in the presence of AgrB. In silico, membrane complexes of AgrD and dimeric AgrB show non-equivalent AgrB monomers responsible for initial binding and for processing, respectively. By exploiting split luciferase assays in Staphylococcus aureus, we provide experimental evidence that AgrB interacts directly with itself and with AgrD. We confirmed the in vitro formation of an AgrBD complex and AIP production after Western blotting using either membranes from Escherichia coli expressing AgrB or with purified AgrB and T7-tagged AgrD. AgrB and AgrD formed stable complexes in detergent micelles revealed using synchrotron radiation CD (SRCD) and Landau analysis consistent with the enhanced thermal stability of AgrB in the presence of AgrD. Conformational alteration of AgrB following provision of AgrD was observed by small angle X-ray scattering from proteodetergent micelles. An atomistic description of AgrB and AgrD has been obtained together with confirmation of the AgrB 6TMD membrane topology and existence of AgrBD molecular complexes in vitro and in vivo.

3.
Proc Natl Acad Sci U S A ; 120(15): e2208737120, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37011186

RESUMEN

The alarming rise in superbugs that are resistant to drugs of last resort, including vancomycin-resistant enterococci and staphylococci, has become a significant global health hazard. Here, we report the click chemistry synthesis of an unprecedented class of shapeshifting vancomycin dimers (SVDs) that display potent activity against bacteria that are resistant to the parent drug, including the ESKAPE pathogens, vancomycin-resistant Enterococcus (VRE), methicillin-resistant Staphylococcus aureus (MRSA), as well as vancomycin-resistant S. aureus (VRSA). The shapeshifting modality of the dimers is powered by a triazole-linked bullvalene core, exploiting the dynamic covalent rearrangements of the fluxional carbon cage and creating ligands with the capacity to inhibit bacterial cell wall biosynthesis. The new shapeshifting antibiotics are not disadvantaged by the common mechanism of vancomycin resistance resulting from the alteration of the C-terminal dipeptide with the corresponding d-Ala-d-Lac depsipeptide. Further, evidence suggests that the shapeshifting ligands destabilize the complex formed between the flippase MurJ and lipid II, implying the potential for a new mode of action for polyvalent glycopeptides. The SVDs show little propensity for acquired resistance by enterococci, suggesting that this new class of shapeshifting antibiotic will display durable antimicrobial activity not prone to rapidly acquired clinical resistance.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Enterococos Resistentes a la Vancomicina , Vancomicina/farmacología , Antibacterianos/uso terapéutico , Pruebas de Sensibilidad Microbiana
4.
Cell Chem Biol ; 29(7): 1187-1199.e6, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35259345

RESUMEN

As single- and mixed-species biofilms, Staphylococcus aureus and Pseudomonas aeruginosa cause difficult-to-eradicate chronic infections. In P. aeruginosa, pseudomonas quinolone (PQS)-dependent quorum sensing regulates virulence and biofilm development that can be attenuated via antagonists targeting the transcriptional regulator PqsR (MvfR). Here, we exploited a quinazolinone (QZN) library including PqsR agonists and antagonists for their activity against S. aureus alone, when co-cultured with P. aeruginosa, and in combination with the aminoglycoside tobramycin. The PqsR inhibitor, QZN 34 killed planktonic Gram-positives but not Gram-negatives. QZN 34 prevented S. aureus biofilm formation, severely damaged established S. aureus biofilms, and perturbed P. aeruginosa biofilm development. Although P. aeruginosa protected S. aureus from tobramycin in mixed biofilms, the combination of aminoglycoside antibiotic with QZN 34 eradicated the mixed-species biofilm. The mechanism of action of QZN 34 toward Gram-positive bacteria is shown to involve membrane perturbation and dissipation of transmembrane potential.


Asunto(s)
Pseudomonas aeruginosa , Percepción de Quorum , Antibacterianos/farmacología , Proteínas Bacterianas , Biopelículas , Pseudomonas , Staphylococcus aureus , Tobramicina/farmacología
5.
Sensors (Basel) ; 20(15)2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32752273

RESUMEN

Gaussia luciferase (GLuc) is a secreted protein with significant potential for use as a reporter of gene expression in bacterial pathogenicity studies. To date there are relatively few examples of its use in bacteriology. In this study we show that GLuc can be functionally expressed in the human pathogen Staphylococcus aureus and furthermore show that it can be used as a biosensor for the agr quorum sensing (QS) system which employs autoinducing peptides to control virulence. GLuc was linked to the P3 promoter of the S. aureusagr operon. Biosensor strains were validated by evaluation of chemical agent-mediated activation and inhibition of agr. Use of GLuc enabled quantitative assessment of agr activity. This demonstrates the utility of Gaussia luciferase for in vitro monitoring of agr activation and inhibition.


Asunto(s)
Percepción de Quorum , Staphylococcus aureus , Proteínas Bacterianas/genética , Humanos , Luciferasas/genética , Staphylococcus aureus/genética , Transactivadores
6.
J Bacteriol ; 201(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31358609

RESUMEN

Mutations in the polymorphic Staphylococcus aureusagr locus responsible for quorum sensing (QS)-dependent virulence gene regulation occur frequently during host adaptation. In two genomically closely related S. aureus clinical isolates exhibiting marked differences in Panton-Valentine leukocidin production, a mutation conferring an N267I substitution was identified in the cytoplasmic domain of the QS sensor kinase, AgrC. This natural mutation delayed the onset and accumulation of autoinducing peptide (AIP) and showed reduced responsiveness to exogenous AIPs. Other S. aureus strains harboring naturally occurring AgrC cytoplasmic domain mutations were identified, including T247I, I311T, A343T, L245S, and F264C. These mutations were associated with reduced cytotoxicity, delayed/reduced AIP production, and impaired sensitivity to exogenous AIP. Molecular dynamics simulations were used to model the AgrC cytoplasmic domain conformational changes arising. Although mutations were localized in different parts of the C-terminal domain, their impact on molecular structure was manifested by twisting of the leading helical hairpin α1-α2, accompanied by repositioning of the H-box and G-box, along with closure of the flexible loop connecting the two and occlusion of the ATP-binding site. Such conformational rearrangements of key functional subdomains in these mutants highlight the cooperative response of molecular structure involving dimerization and ATP binding and phosphorylation, as well as the binding site for the downstream response element AgrA. These appear to increase the threshold for agr activation via AIP-dependent autoinduction, thus reducing virulence and maintaining S. aureus in an agr-downregulated "colonization" mode.IMPORTANCE Virulence factor expression in Staphylococcus aureus is regulated via autoinducing peptide (AIP)-dependent activation of the sensor kinase AgrC, which forms an integral part of the agr quorum sensing system. In response to bound AIP, the cytoplasmic domain of AgrC (AgrC-cyt) undergoes conformational changes resulting in dimerization, autophosphorylation, and phosphotransfer to the response regulator AgrA. Naturally occurring mutations in AgrC-cyt are consistent with repositioning of key functional domains, impairing dimerization and restricting access to the ATP-binding pocket. Strains harboring specific AgrC-cyt mutations exhibit reduced AIP autoinduction efficiency and a timing-dependent attenuation of cytotoxicity which may confer a survival advantage during established infection by promoting colonization while restricting unnecessary overproduction of exotoxins.


Asunto(s)
Sustitución de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Péptidos Cíclicos/metabolismo , Proteínas Quinasas/química , Proteínas Quinasas/genética , Staphylococcus aureus/metabolismo , Adenosina Trifosfato/metabolismo , Sitios de Unión , Citoplasma/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Fosforilación , Unión Proteica , Dominios Proteicos , Proteínas Quinasas/metabolismo , Multimerización de Proteína , Estructura Secundaria de Proteína , Percepción de Quorum , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Factores de Tiempo , Transactivadores/metabolismo
7.
Genome Biol ; 19(1): 94, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-30021593

RESUMEN

BACKGROUND: Fitness costs imposed on bacteria by antibiotic resistance mechanisms are believed to hamper their dissemination. The scale of these costs is highly variable. Some, including resistance of Staphylococcus aureus to the clinically important antibiotic mupirocin, have been reported as being cost-free, which suggests that there are few barriers preventing their global spread. However, this is not supported by surveillance data in healthy communities, which indicate that this resistance mechanism is relatively unsuccessful. RESULTS: Epistasis analysis on two collections of MRSA provides an explanation for this discord, where the mupirocin resistance-conferring mutation of the ileS gene appears to affect the levels of toxins produced by S. aureus when combined with specific polymorphisms at other loci. Proteomic analysis demonstrates that the activity of the secretory apparatus of the PSM family of toxins is affected by mupirocin resistance. As an energetically costly activity, this reduction in toxicity masks the fitness costs associated with this resistance mutation, a cost that becomes apparent when toxin production becomes necessary. This hidden fitness cost provides a likely explanation for why this mupirocin-resistance mechanism is not more prevalent, given the widespread use of this antibiotic. CONCLUSIONS: With dwindling pools of antibiotics available for use, information on the fitness consequences of the acquisition of resistance may need to be considered when designing antibiotic prescribing policies. However, this study suggests there are levels of depth that we do not understand, and that holistic, surveillance and functional genomics approaches are required to gain this crucial information.


Asunto(s)
Antibacterianos/farmacología , Epistasis Genética , Aptitud Genética/efectos de los fármacos , Genoma Bacteriano , Isoleucina-ARNt Ligasa/genética , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Mupirocina/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/genética , Farmacorresistencia Bacteriana , Evolución Molecular , Sitios Genéticos , Isoleucina-ARNt Ligasa/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/metabolismo , Pruebas de Sensibilidad Microbiana , Mutación , Proteómica/métodos
8.
Methods Mol Biol ; 1673: 89-96, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29130166

RESUMEN

Strains of the Gram-positive pathogen Staphylococcus aureus can be divided into four quorum sensing (QS) groups. Membership of each group is defined by the amino acid sequence of the autoinducing peptide (AIP) QS signal molecule that is encoded within the agrBDCA genetic locus and specifically within agrD. This chapter describes the use of simple, in-cell, lux-based, bio-reporters that can be used to identify/confirm the specific agr group to which a particular S. aureus isolate belongs, as well as to assess the timing and quantity of AIP produced.


Asunto(s)
Bioensayo/métodos , Sitios Genéticos , Péptidos/análisis , Staphylococcus aureus/metabolismo , Secuencia de Aminoácidos , Genes Reporteros , Cinética , Péptidos/química
9.
J Antimicrob Chemother ; 72(3): 744-753, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27999062

RESUMEN

Objectives: Biofilm infections of intravascular catheters caused by Staphylococcus aureus may be treated with catheter lock solutions (CLSs). Here we investigated the antibacterial activity, cytotoxicity and CLS potential of 5-hydroxyethyl-3-tetradecanoyltetramic acid (5HE-C14-TMA) compared with the related compounds 3-tetradecanoyltetronic (C14-TOA) and 3-tetradecanoylthiotetronic (C14-TTA), which are variants of quorum sensing signalling molecules produced by Pseudomonas aeruginosa . Methods: Antibacterial activity and mechanism of action of 5HE-C14-TMA, C14-TOA and C14-TTA were determined via MIC, bacterial killing, membrane potential and permeability assays. Susceptibility of S. aureus biofilms formed in the presence of plasma in vitro was investigated, MTT cytotoxicity testing was undertaken and cytokine release in human blood upon exposure to 5HE-C14-TMA and/or S. aureus biofilms was quantified. The effectiveness of 5HE-C14-TMA as CLS therapy in vivo was assessed using a rat intravascular catheter biofilm infection model. Results: MICs of 5HE-C14-TMA, C14-TOA and C14-TTA ranged from 2 to 4 mg/L. 5HE-C14-TMA and C14-TTA were bactericidal; all three compounds perturbed the staphylococcal membrane by increasing membrane permeability, depolarized the transmembrane potential and caused ATP leakage. Cytotoxicity and haemolytic activity were compound and target cell type-dependent. 5HE-C14-TMA reduced S. aureus biofilm viability in a dose-dependent manner in vitro and in vivo and did not trigger release of cytokines in human blood, but inhibited the high levels of IL-8 and TNF-α induced by S. aureus biofilms. Conclusions: 5HE-C14-TMA, C14-TOA and C14-TTA are membrane-active agents. 5HE-C14-TMA was the most potent, eradicating S. aureus biofilms at 512-1024 mg/L both in vitro and in vivo as a CLS.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones Relacionadas con Catéteres/tratamiento farmacológico , Pirrolidinonas/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Dispositivos de Acceso Vascular/microbiología , Animales , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Infecciones Relacionadas con Catéteres/microbiología , Descubrimiento de Drogas , Humanos , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana , Pseudomonas aeruginosa/metabolismo , Pirrolidinonas/farmacología , Percepción de Quorum , Ratas , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Vancomicina/farmacología
10.
J Med Chem ; 57(6): 2813-9, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24592914

RESUMEN

A series of 3-oxo-C12-HSL, tetramic acid, and tetronic acid analogues were synthesized to gain insights into the structural requirements for quorum sensing inhibition in Staphylococcus aureus. Compounds active against agr were noncompetitive inhibitors of the autoinducing peptide (AIP) activated AgrC receptor, by altering the activation efficacy of the cognate AIP-1. They appeared to act as negative allosteric modulators and are exemplified by 3-tetradecanoyltetronic acid 17, which reduced nasal cell colonization and arthritis in a murine infection model.


Asunto(s)
Antibacterianos/farmacología , Percepción de Quorum/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/síntesis química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/efectos de los fármacos , Línea Celular , Furanos/síntesis química , Furanos/farmacología , Indicadores y Reactivos , Quelantes del Hierro/farmacología , Ratones , Pruebas de Sensibilidad Microbiana , Cavidad Nasal/citología , Péptidos Cíclicos/antagonistas & inhibidores , Proteínas Quinasas/efectos de los fármacos , Pirrolidinonas/síntesis química , Pirrolidinonas/farmacología , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Relación Estructura-Actividad
11.
Arch Microbiol ; 192(12): 1059-67, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20844865

RESUMEN

Abh is a transition state regulator of Bacillus subtilis that controls biofilm formation and the production of several diverse antimicrobial compounds. Using a high-throughput non-biased technique, we show for the first time that Abh influences the sensitivity of B. subtilis to diverse antimicrobial compounds. Following up on these findings with a combination of classical genetics and antibiotic susceptibility assays, we demonstrate that Abh influences cellular processes such as the remodelling of the cell wall. We present data demonstrating that the extracytoplasmic function sigma factor σ(X) controls resistance to ß-lactam antibiotics by activating abh transcription. Downstream from Abh, activation of slrR expression by Abh is responsible for controlling the sensitivity of B. subtilis to such antibiotics due to the role that SlrR plays in regulating autolysin biosynthesis. The abh mutant additionally exhibits increased resistance to aminoglycoside antimicrobials. We confirm that aminoglycoside killing of B. subtilis is likely to be caused by oxidative damage but rule out the possibility that the increased resistance of the abh mutant to aminoglycosides is due to a general increase in resistance to oxidative stress.


Asunto(s)
Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Factor sigma/metabolismo , Aminoglicósidos/farmacología , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Pared Celular/metabolismo , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Genes Reguladores , Pruebas de Sensibilidad Microbiana , Mutación , N-Acetil Muramoil-L-Alanina Amidasa/biosíntesis , Estrés Oxidativo , Resistencia betalactámica , beta-Lactamas/farmacología
12.
J Biol Chem ; 285(50): 39224-38, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20876533

RESUMEN

Arginine decarboxylases (ADCs; EC 4.1.1.19) from four different protein fold families are important for polyamine biosynthesis in bacteria, archaea, and plants. Biosynthetic alanine racemase fold (AR-fold) ADC is widespread in bacteria and plants. We report the discovery and characterization of an ancestral form of the AR-fold ADC in the bacterial Chloroflexi and Bacteroidetes phyla. The ancestral AR-fold ADC lacks a large insertion found in Escherichia coli and plant AR-fold ADC and is more similar to the lysine biosynthetic enzyme meso-diaminopimelate decarboxylase, from which it has evolved. An E. coli acid-inducible ADC belonging to the aspartate aminotransferase fold (AAT-fold) is involved in acid resistance but not polyamine biosynthesis. We report here that the acid-inducible AAT-fold ADC has evolved from a shorter, ancestral biosynthetic AAT-fold ADC by fusion of a response regulator receiver domain protein to the N terminus. Ancestral biosynthetic AAT-fold ADC appears to be limited to firmicute bacteria. The phylogenetic distribution of different forms of ADC distinguishes bacteria from archaea, euryarchaeota from crenarchaeota, double-membraned from single-membraned bacteria, and firmicutes from actinobacteria. Our findings extend to eight the different enzyme forms carrying out the activity described by EC 4.1.1.19. ADC gene clustering reveals that polyamine biosynthesis employs diverse and exchangeable synthetic modules. We show that in Bacillus subtilis, ADC and polyamines are essential for biofilm formation, and this appears to be an ancient, evolutionarily conserved function of polyamines in bacteria. Also of relevance to human health, we found that arginine decarboxylation is the dominant pathway for polyamine biosynthesis in human gut microbiota.


Asunto(s)
Bacillus subtilis/metabolismo , Biopelículas , Carboxiliasas/metabolismo , Poliaminas/química , Alanina Racemasa/metabolismo , Secuencia de Aminoácidos , Clonación Molecular , Escherichia coli/metabolismo , Genómica , Humanos , Intestinos/microbiología , Conformación Molecular , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Filogenia , Homología de Secuencia de Aminoácido
13.
J Bacteriol ; 191(22): 6822-32, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19767430

RESUMEN

A characteristic feature of biofilm formation is the production of a protective extracellular polymeric matrix. In the gram-positive bacterium Bacillus subtilis, the biofilm matrix is synthesized by the products of the epsABCDEFGHIJKLMNO operon (hereafter called the eps operon) and yqxM-sipW-tasA loci. Transcription from these operons is repressed by two key regulators, AbrB and SinR. Relief of inhibition is necessary to allow biofilm formation to proceed. Here we present data indicating that Abh, a sequence and structural homologue of AbrB, regulates biofilm architecture by B. subtilis when colony morphology and pellicle formation are assessed. Data indicating that abh expression is dependent on the environmental signals that stimulate the activity of the extracytoplasmic function sigma-factor sigma(X) are shown. We demonstrate that expression of slrR, the proposed activator of yqxM transcription, is positively controlled by Abh. Furthermore, Abh is shown to activate transcription from the promoter of the eps operon through its control of SlrR. These findings add to the increasingly complex transcriptional network that controls biofilm formation by B. subtilis.


Asunto(s)
Bacillus subtilis/crecimiento & desarrollo , Bacillus subtilis/metabolismo , Proteínas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Proteínas de Unión al ADN/fisiología , Operón/fisiología , Factor sigma/fisiología , Factores de Transcripción/fisiología , Bacillus subtilis/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Huella de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Regulación Bacteriana de la Expresión Génica/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Operón/genética , Factor sigma/genética , Factor sigma/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Microbiology (Reading) ; 155(Pt 1): 1-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19118340

RESUMEN

Bacteria control multicellular behavioural responses, including biofilm formation and swarming motility, by integrating environmental cues through a complex regulatory network. Heterogeneous gene expression within an otherwise isogenic cell population that allows for differentiation of cell fate is an intriguing phenomenon that adds to the complexity of multicellular behaviour. This review focuses on recent data about how DegU, a pleiotropic response regulator, co-ordinates multicellular behaviour in Bacillus subtilis. We review studies that challenge the conventional understanding of the molecular mechanisms underpinning the DegU regulatory system and others that describe novel targets of DegU during activation of biofilm formation by B. subtilis. We also discuss a novel role for DegU in regulating multicellular processes in the food-borne pathogen Listeria monocytogenes.


Asunto(s)
Bacillus subtilis , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes , Bacillus subtilis/genética , Bacillus subtilis/crecimiento & desarrollo , Bacillus subtilis/metabolismo , Bacillus subtilis/fisiología , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Listeria monocytogenes/genética , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/fisiología , Transducción de Señal
15.
J Bacteriol ; 191(1): 100-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18978066

RESUMEN

Biofilm formation is an example of a multicellular process which depends on cooperative behavior and differentiation within a bacterial population. Our findings indicate that there is a complex feedback loop that maintains the stoichiometry of the extracellular matrix and other proteins required for complex colony development by Bacillus subtilis. Analysis of the transcriptional regulation of two DegU-activated genes that are required for complex colony development by B. subtilis revealed additional involvement of global regulators that are central to controlling biofilm formation. Activation of transcription from both the yvcA and yuaB promoters requires DegU approximately phosphate, but transcription is inhibited by direct AbrB binding to the promoter regions. Inhibition of transcription by AbrB is relieved when Spo0A approximately phosphate is generated due to its known role in inhibiting abrB expression. Deletion of SinR, a key coordinator of motility and biofilm formation, enhanced transcription from both loci; however, no evidence of a direct interaction with SinR for either the yvcA or yuaB promoter regions was observed. The enhanced transcription in the sinR mutant background was subsequently demonstrated to be dependent on biosynthesis of the polysaccharide component that forms the major constituent of the B. subtilis biofilm matrix. Together, these findings indicate that a genetic network dependent on activation of both DegU and Spo0A controls complex colony development by B. subtilis.


Asunto(s)
Bacillus subtilis/genética , Proteínas Bacterianas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Antibacterianos/farmacología , Bacillus subtilis/clasificación , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/crecimiento & desarrollo , Biopelículas , Cartilla de ADN , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Genotipo , Operón , Plásmidos , Polisacáridos Bacterianos/metabolismo , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...