Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 76(2): 255-267, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31626749

RESUMEN

For decades, mankind has dominated the battle against bacteria, yet the tide is slowly turning. Our antibacterial strategies are becoming less effective, allowing bacteria to get the upper hand. The alarming rise in antibiotic resistance is an important cause of anti-infective therapy failure. However, other factors are at play as well. It is widely recognized that bacterial populations display high levels of heterogeneity. Population heterogeneity generates phenotypes specialized in surviving antibiotic attacks. Nonetheless, the presence of antibiotic-insensitive subpopulations is not considered when initiating treatment. It is therefore time to reevaluate how we combat bacterial infections. We here focus on antibiotic persistence and heteroresistance, phenomena in which small fractions of the population are tolerant (persisters) and resistant to antibiotics, respectively. We discuss molecular mechanisms involved, their clinical importance, and possible therapeutic strategies. Moving forward, we argue that these heterogeneous phenotypes should no longer be ignored in clinical practice and that better diagnostic and therapeutic approaches are urgently needed.


Asunto(s)
Antibacterianos , Bacterias/metabolismo , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/metabolismo , Farmacorresistencia Bacteriana/efectos de los fármacos , Viabilidad Microbiana/efectos de los fármacos , Antibacterianos/efectos adversos , Antibacterianos/uso terapéutico , Bacterias/genética , Infecciones Bacterianas/genética , Infecciones Bacterianas/microbiología , Farmacorresistencia Bacteriana/genética , Humanos
2.
Mol Cell ; 70(5): 763-764, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29883603

RESUMEN

Antibiotic-tolerant persister cells are difficult to eradicate by conventional classes of antibiotics. Kim and colleagues have discovered a new class of synthetic retinoid antibiotics that kill Staphylococcus aureus persisters by disrupting their cytoplasmic membrane.


Asunto(s)
Antibacterianos , Staphylococcus aureus , Humanos , Retinoides , Infecciones Estafilocócicas
3.
Nat Prod Rep ; 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38465694

RESUMEN

Covering: up to October 2023Many bioactive natural products are synthesized by microorganisms that are either difficult or impossible to cultivate under laboratory conditions, or that produce only small amounts of the desired compound. By transferring biosynthetic gene clusters (BGCs) into alternative host organisms that are more easily cultured and engineered, larger quantities can be obtained and new analogues with potentially improved biological activity or other desirable properties can be generated. Moreover, expression of cryptic BGCs in a suitable host can facilitate the identification and characterization of novel natural products. Heterologous expression therefore represents a valuable tool for natural product discovery and engineering as it allows the study and manipulation of their biosynthetic pathways in a controlled setting, enabling innovative applications. Bacillus is a genus of Gram-positive bacteria that is widely used in industrial biotechnology as a host for the production of proteins from diverse origins, including enzymes and vaccines. However, despite numerous successful examples, Bacillus species remain underexploited as heterologous hosts for the expression of natural product BGCs. Here, we review important advantages that Bacillus species offer as expression hosts, such as high secretion capacity, natural competence for DNA uptake, and the increasing availability of a wide range of genetic tools for gene expression and strain engineering. We evaluate different strain optimization strategies and other critical factors that have improved the success and efficiency of heterologous natural product biosynthesis in B. subtilis. Finally, future perspectives for using B. subtilis as a heterologous host are discussed, identifying research gaps and promising areas that require further exploration.

4.
Mol Cell ; 59(1): 9-21, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26051177

RESUMEN

Within bacterial populations, a small fraction of persister cells is transiently capable of surviving exposure to lethal doses of antibiotics. As a bet-hedging strategy, persistence levels are determined both by stochastic induction and by environmental stimuli called responsive diversification. Little is known about the mechanisms that link the low frequency of persisters to environmental signals. Our results support a central role for the conserved GTPase Obg in determining persistence in Escherichia coli in response to nutrient starvation. Obg-mediated persistence requires the stringent response alarmone (p)ppGpp and proceeds through transcriptional control of the hokB-sokB type I toxin-antitoxin module. In individual cells, increased Obg levels induce HokB expression, which in turn results in a collapse of the membrane potential, leading to dormancy. Obg also controls persistence in Pseudomonas aeruginosa and thus constitutes a conserved regulator of antibiotic tolerance. Combined, our findings signify an important step toward unraveling shared genetic mechanisms underlying persistence.


Asunto(s)
Proteínas Bacterianas/genética , Toxinas Bacterianas/biosíntesis , Farmacorresistencia Bacteriana/fisiología , Proteínas de Escherichia coli/biosíntesis , Escherichia coli/genética , Proteínas de Unión al GTP/genética , Pseudomonas aeruginosa/genética , Antibacterianos/farmacología , Toxinas Bacterianas/genética , Membrana Celular/fisiología , Proteínas de Escherichia coli/genética , Potenciales de la Membrana/genética , Pruebas de Sensibilidad Microbiana , Estructura Terciaria de Proteína/genética
5.
Anal Chem ; 94(45): 15781-15789, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36377427

RESUMEN

Partitions in digital PCR (dPCR) assays do not reach the detection threshold at the same time. This heterogeneity in amplification results in intermediate endpoint fluorescence values (i.e., rain) and misclassification of partitions, which has a major impact on the accuracy of nucleic acid quantification. Rain most often results from a reduced amplification efficiency or template inaccessibility; however, exactly how these contribute to rain has not been described. We developed and experimentally validated an analytical model that mechanistically explains the relationship between amplification efficiency, template accessibility, and rain. Using Monte Carlo simulations, we show that a reduced amplification efficiency leads to broader threshold cycle (Ct) distributions that can be fitted using a log-normal probability distribution. From the fit parameters, the amplification efficiency can be calculated. Template inaccessibility, on the other hand, leads to a different rain pattern, in which a distinct exponential tail in the Ct distribution can be observed. Using our model, it is possible to determine if the amplification efficiency, template accessibility, or another source is the main contributor of rain in dPCR assays. We envision that this model will facilitate and speed up dPCR assay optimization and provide an indication for the accuracy of the assay.


Asunto(s)
Lluvia , Reacción en Cadena de la Polimerasa/métodos , Método de Montecarlo
6.
Mol Microbiol ; 112(5): 1593-1608, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31498933

RESUMEN

Obg is a versatile GTPase that plays a pivotal role in bacterial persistence. We previously showed that the Escherichia coli homolog ObgE exerts this activity through transcriptional activation of a toxin-antitoxin module and subsequent membrane depolarization. Here, we assessed the role of G-domain functionality in ObgE-mediated persistence. Through screening of a mutant library, we identified five obgE alleles (with substitutions G166V, D246G, S270I, N283I and I313N) that have lost their persistence function and no longer activate hokB expression. These alleles support viability of a strain otherwise deprived of ObgE, indicating that ObgE's persistence function can be uncoupled from its essential role. Based on the ObgE crystal structure, we designed two additional mutant proteins (T193A and D286Y), one of which (D286Y) no longer affects persistence. Using isothermal titration calorimetry, stopped-flow experiments and kinetics, we subsequently assessed nucleotide binding and GTPase activity in all mutants. With the exception of the S270I mutant that is possibly affected in protein-protein interactions, all mutants that have lost their persistence function display severely reduced binding to GDP or the alarmone ppGpp. However, we find no clear relation between persistence and GTP or pppGpp binding nor with GTP hydrolysis. Combined, our results signify an important step toward understanding biochemical determinants underlying persistence.


Asunto(s)
Toxinas Bacterianas/biosíntesis , Proteínas de Escherichia coli/biosíntesis , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Escherichia coli/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Relación Estructura-Actividad , Activación Transcripcional/genética
7.
Anal Bioanal Chem ; 411(6): 1127-1134, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30637438

RESUMEN

Sulfite is often added to beverages as an antioxidant and antimicrobial agent. In fermented beverages, sulfite is also naturally produced by yeast cells. However, sulfite causes adverse health effects in asthmatic patients and accurate measurement of the sulfite concentration is therefore very important. Current sulfite analysis methods are time- and reagent-consuming and often require costly equipment. Here, we present a system allowing sensitive, ultralow-volume sulfite measurements based on a reusable glass-silicon microdroplet platform on which microdroplet generation, addition of enzymes through chemical-induced emulsion destabilization and pillar-induced droplet merging, emulsion restabilization, droplet incubation, and fluorescence measurements are integrated. In a first step, we developed and verified a fluorescence-based enzymatic assay for sulfite by measuring its analytical performance (LOD, LOQ, the dynamic working range, and the influence of salts, colorant, and sugars) and comparing fluorescent microplate readouts of fermentation samples with standard colorimetric measurements using the 5,5'-dithiobis-(2-nitrobenzoic acid) assay of the standard Gallery Plus Beermaster analysis platform. Next, samples were analyzed on the microdroplet platform, which also showed good correlation with the standard colorimetric analysis. Although the presented platform does not allow stable reinjection of droplets due to the presence of a tight array of micropillars at the fluidics entrances to prevent channel clogging by dust, removing the pillars, and integrating miniaturized pumps and optics in a future design would allow to use this platform for high-throughput, automated, and portable screening of microbes, plant, or mammalian cells. Graphical abstract ᅟ.


Asunto(s)
Bebidas/análisis , Vidrio/química , Técnicas Analíticas Microfluídicas/instrumentación , Silicio/química , Sulfitos/análisis , Diseño de Equipo , Espectrometría de Fluorescencia
8.
Drug Resist Updat ; 38: 12-26, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29857815

RESUMEN

In addition to the well-known strategies of antibiotic resistance and biofilm formation, bacterial populations possess an additional survival strategy to endure hostile environments or antibiotic exposure. A small fraction of transiently antibiotic-tolerant phenotypical variants, called persister cells, is capable of surviving treatment with high doses of antibiotics. When antibiotic pressure drops, persisters that switch back to a normal phenotype can resume growth, ensuring survival of the bacterial population. Persister cells have been identified in every major pathogen, contribute to the antibiotic tolerance observed in biofilms, and are responsible for the recalcitrant nature of chronic infections. Also, evidence is accumulating that persister cells can contribute to the emergence of antibiotic resistance. Consequently, effective treatment of persister cells could greatly improve patient outcome. The small number of persisters and the redundancy in mechanisms of persister formation impede target-based development of anti-persister therapies. Nonetheless, the armory of anti-persister molecules is increasing. This review presents a comprehensive overview of anti-persister molecules and strategies described in literature to date and offers perspectives on potential anti-persistence targets and methods for the development of future therapies. Furthermore, we highlight in vivo model systems for pre-clinical testing and summarize ongoing clinical trials of candidate anti-persister therapeutics.


Asunto(s)
Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Biopelículas/efectos de los fármacos , Farmacorresistencia Bacteriana , Animales , Antibacterianos/efectos adversos , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Diseño de Fármacos , Regulación Bacteriana de la Expresión Génica , Genotipo , Humanos , Viabilidad Microbiana/efectos de los fármacos , Fenotipo , Transducción de Señal/efectos de los fármacos
9.
Int J Mol Sci ; 21(1)2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-31861427

RESUMEN

Even though the Obg protein is essential for bacterial viability, the cellular functions of this universally conserved GTPase remain enigmatic. Moreover, the influence of GTP and GDP binding on the activity of this protein is largely unknown. Previously, we identified a mutant isoform of ObgE (the Obg protein of Escherichia coli) that triggers cell death. In this research we explore the biochemical requirements for the toxic effect of this mutant ObgE* isoform, using cell death as a readily accessible read-out for protein activity. Both the absence of the N-terminal domain and a decreased GTP binding affinity neutralize ObgE*-mediated toxicity. Moreover, a deletion in the region that connects the N-terminal domain to the G domain likewise abolishes toxicity. Taken together, these data indicate that GTP binding by ObgE* triggers a conformational change that is transmitted to the N-terminal domain to confer toxicity. We therefore conclude that ObgE*-GTP, but not ObgE*-GDP, is the active form of ObgE* that is detrimental to cell viability. Based on these data, we speculate that also for wild-type ObgE, GTP binding triggers conformational changes that affect the N-terminal domain and thereby control ObgE function.


Asunto(s)
Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas de Escherichia coli/química , Guanosina Trifosfato/química , Modelos Moleculares , Proteínas de Unión al GTP Monoméricas/química , Proteínas Mutantes , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas , Relación Estructura-Actividad
10.
J Biol Chem ; 292(14): 5871-5883, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28223358

RESUMEN

The Obg protein family belongs to the TRAFAC (translation factor) class of P-loop GTPases and is conserved from bacteria to eukaryotes. Essential roles in many different cellular processes have been suggested for the Obg protein from Escherichia coli (ObgE), and we recently showed that it is a central regulator of bacterial persistence. Here, we report the first crystal structure of ObgE at 1.85-Å resolution in the GDP-bound state, showing the characteristic N-terminal domain and a central G domain that are common to all Obg proteins. ObgE also contains an intrinsically disordered C-terminal domain, and we show here that this domain specifically contributed to GTP binding, whereas it did not influence GDP binding or GTP hydrolysis. Biophysical analysis, using small angle X-ray scattering and multi-angle light scattering experiments, revealed that ObgE is a monomer in solution, regardless of the bound nucleotide. In contrast to recent suggestions, our biochemical analyses further indicate that ObgE is neither activated by K+ ions nor by homodimerization. However, the ObgE GTPase activity was stimulated upon binding to the ribosome, confirming the ribosome-dependent GTPase activity of the Obg family. Combined, our data represent an important step toward further unraveling the detailed molecular mechanism of ObgE, which might pave the way to further studies into how this GTPase regulates bacterial physiology, including persistence.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/química , Proteínas de Unión al GTP Monoméricas/química , Potasio/química , Multimerización de Proteína , Cationes Monovalentes/química , Cationes Monovalentes/metabolismo , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Potasio/metabolismo , Dominios Proteicos
11.
Analyst ; 143(11): 2596-2603, 2018 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-29741175

RESUMEN

Clinical laboratory-based nucleic acid amplification tests (NAT) play an important role in diagnosing viral infections. However, laboratory infrastructure requirements and their failure to diagnose at the point-of-need (PON) limit their clinical utility in both resource-rich and -limited clinical settings. The development of fast and sensitive PON viral NAT may overcome these limitations. The scalability of silicon microchip manufacturing combined with advances in silicon microfluidics present an opportunity for development of rapid and sensitive PON NAT on silicon microchips. In the present study, we present rapid and sensitive NAT for a number of RNA and DNA viruses on the same silicon microchip platform. We first developed sensitive (4 copies per reaction) one-step RT-qPCR and qPCR assays detecting HCV, HIV, Zika, HPV 16, and HPV 18 on a benchtop real-time PCR instrument. A silicon microchip was designed with an etched 1.3 µL meandering microreactor, integrated aluminum heaters, thermal insulation trenches and microfluidic channels; this chip was used in all on-chip experiments. Melting curve analysis confirmed precise and localized heating of the microreactor. Following minimal optimization of reaction conditions, the bench-scale assays were successfully transferred to 1.3 µL silicon microreactors with reaction times of 25 min with no reduction in sensitivity, reproducibility, or reaction efficiencies. Taken together, these results demonstrate that rapid and sensitive detection of multiple viruses on the same silicon microchip platform is feasible. Further development of this technology, coupled with silicon microchip-based nucleic acid extraction solutions, could potentially shift viral nucleic acid detection and diagnosis from centralized clinical laboratories to the PON.


Asunto(s)
ADN Viral/análisis , Técnicas Analíticas Microfluídicas , ARN Viral/análisis , Silicio , Técnicas de Amplificación de Ácido Nucleico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Artículo en Inglés | MEDLINE | ID: mdl-28630188

RESUMEN

Antibiotics typically fail to completely eradicate a bacterial population, leaving a small fraction of transiently antibiotic-tolerant persister cells intact. Persisters are therefore seen to be a major cause of treatment failure and greatly contribute to the recalcitrant nature of chronic infections. The current study focused on Pseudomonas aeruginosa, a Gram-negative pathogen belonging to the notorious ESKAPE group of pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) and, due to increasing resistance against most conventional antibiotics, posing a serious threat to human health. Greatly contributing to the difficult treatment of P. aeruginosa infections is the presence of persister cells, and elimination of these cells would therefore significantly improve patient outcomes. In this study, a small-molecule library was screened for compounds that, in combination with the fluoroquinolone antibiotic ofloxacin, reduced the number of P. aeruginosa persisters compared to the number achieved with treatment with the antibiotic alone. Based on the early structure-activity relationship, 1-((2,4-dichlorophenethyl)amino)-3-phenoxypropan-2-ol (SPI009) was selected for further characterization. Combination of SPI009 with mechanistically distinct classes of antibiotics reduced the number of persisters up to 106-fold in both lab strains and clinical isolates of P. aeruginosa Further characterization of the compound revealed a direct and efficient killing of persister cells. SPI009 caused no erythrocyte damage and demonstrated minor cytotoxicity. In conclusion, we identified a novel antipersister compound active against P. aeruginosa with promising applications for the design of novel, case-specific combination therapies in the fight against chronic infections.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Biopelículas/efectos de los fármacos , Línea Celular , Bacterias Gramnegativas/efectos de los fármacos , Células HEK293 , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Relación Estructura-Actividad
13.
Artículo en Inglés | MEDLINE | ID: mdl-27993858

RESUMEN

The spread of antibiotic resistance and the challenges associated with antiseptics such as chlorhexidine have necessitated a search for new antibacterial agents against oral bacterial pathogens. As a result of failing traditional approaches, drug repurposing has emerged as a novel paradigm to find new antibacterial agents. In this study, we examined the effects of the FDA-approved anticancer agent toremifene against the oral bacteria Porphyromonas gingivalis and Streptococcus mutans We found that the drug was able to inhibit the growth of both pathogens, as well as prevent biofilm formation, at concentrations ranging from 12.5 to 25 µM. Moreover, toremifene was shown to eradicate preformed biofilms at concentrations ranging from 25 to 50 µM. In addition, we found that toremifene prevents P. gingivalis and S. mutans biofilm formation on titanium surfaces. A time-kill study indicated that toremifene is bactericidal against S. mutans Macromolecular synthesis assays revealed that treatment with toremifene does not cause preferential inhibition of DNA, RNA, or protein synthesis pathways, indicating membrane-damaging activity. Biophysical studies using fluorescent probes and fluorescence microscopy further confirmed the membrane-damaging mode of action. Taken together, our results suggest that the anticancer agent toremifene is a suitable candidate for further investigation for the development of new treatment strategies for oral bacterial infections.


Asunto(s)
Antibacterianos/farmacología , Antineoplásicos Hormonales/farmacología , Biopelículas/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Porphyromonas gingivalis/efectos de los fármacos , Streptococcus mutans/efectos de los fármacos , Toremifeno/farmacología , Biopelículas/crecimiento & desarrollo , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular/efectos de los fármacos , Placa Dental/tratamiento farmacológico , Placa Dental/microbiología , Reposicionamiento de Medicamentos , Farmacorresistencia Bacteriana Múltiple/fisiología , Humanos , Pruebas de Sensibilidad Microbiana , Periodontitis/tratamiento farmacológico , Periodontitis/microbiología , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/ultraestructura , Streptococcus mutans/metabolismo , Streptococcus mutans/ultraestructura , Titanio/análisis
14.
Antimicrob Agents Chemother ; 60(8): 4630-7, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27185802

RESUMEN

Health care-associated infections present a major threat to modern medical care. Six worrisome nosocomial pathogens-Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.-are collectively referred to as the "ESKAPE bugs." They are notorious for extensive multidrug resistance, yet persistence, or the phenotypic tolerance displayed by a variant subpopulation, remains underappreciated in these pathogens. Importantly, persistence can prevent eradication of antibiotic-sensitive bacterial populations and is thought to act as a catalyst for the development of genetic resistance. Concentration- and time-dependent aminoglycoside killing experiments were used to investigate persistence in the ESKAPE pathogens. Additionally, a recently developed method for the experimental evolution of persistence was employed to investigate adaptation to high-dose, extended-interval aminoglycoside therapy in vitro We show that ESKAPE pathogens exhibit biphasic killing kinetics, indicative of persister formation. In vitro cycling between aminoglycoside killing and persister cell regrowth, evocative of clinical high-dose extended-interval therapy, caused a 37- to 213-fold increase in persistence without the emergence of resistance. Increased persistence also manifested in biofilms and provided cross-tolerance to different clinically important antibiotics. Together, our results highlight a possible drawback of intermittent, high-dose antibiotic therapy and suggest that clinical diagnostics might benefit from taking into account persistence.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Aminoglicósidos/farmacología , Enterobacter/efectos de los fármacos , Enterococcus faecium/efectos de los fármacos , Klebsiella pneumoniae/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Biopelículas/efectos de los fármacos , Infección Hospitalaria/tratamiento farmacológico , Infección Hospitalaria/microbiología , Humanos
15.
Antimicrob Agents Chemother ; 60(6): 3480-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27021321

RESUMEN

Bacteriophage-encoded endolysins have shown promise as a novel class of antibacterials with a unique mode of action, i.e., peptidoglycan degradation. However, Gram-negative pathogens are generally not susceptible due to their protective outer membrane. Artilysins overcome this barrier. Artilysins are optimized, engineered fusions of selected endolysins with specific outer membrane-destabilizing peptides. Artilysin Art-175 comprises a modified variant of endolysin KZ144 with an N-terminal fusion to SMAP-29. Previously, we have shown the high susceptibility of Pseudomonas aeruginosa to Art-175. Here, we report that Art-175 is highly bactericidal against stationary-phase cells of multidrug-resistant Acinetobacter baumannii, even resulting in a complete elimination of large inocula (≥10(8) CFU/ml). Besides actively dividing cells, Art-175 also kills persisters. Instantaneous killing of A. baumannii upon contact with Art-175 could be visualized after immobilization of the bacteria in a microfluidic flow cell. Effective killing of a cell takes place through osmotic lysis after peptidoglycan degradation. The killing rate is enhanced by the addition of 0.5 mM EDTA. No development of resistance to Art-175 under selection pressure and no cross-resistance with existing resistance mechanisms could be observed. In conclusion, Art-175 represents a highly active Artilysin against both A. baumannii and P. aeruginosa, two of the most life-threatening pathogens of the order Pseudomonadales.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Catelicidinas/farmacología , Endopeptidasas/farmacología , Proteínas Recombinantes de Fusión/farmacología , Infecciones por Acinetobacter/microbiología , Farmacorresistencia Bacteriana Múltiple , Ácido Edético/farmacología , Endopeptidasas/química , Humanos , Pruebas de Sensibilidad Microbiana , Técnicas Analíticas Microfluídicas , Pseudomonas aeruginosa/efectos de los fármacos
16.
J Antimicrob Chemother ; 71(4): 936-45, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26702917

RESUMEN

OBJECTIVES: Biofilm-associated implant infections represent a serious public health problem. Covalent immobilization of antimicrobial agents on titanium (Ti), thereby inhibiting biofilm formation of microbial pathogens, is a solution to this problem. METHODS: Vancomycin (VAN) and caspofungin (CAS) were covalently bound on Ti substrates using an improved processing technique adapted to large-scale coating of implants. Resistance of the VAN-coated Ti (VAN-Ti) and CAS-coated Ti (CAS-Ti) substrates against in vitro biofilm formation of the bacterium Staphylococcus aureus and the fungal pathogen Candida albicans was determined by plate counting and visualized by confocal laser scanning microscopy. The efficacy of the coated Ti substrates was also tested in vivo using an adapted biomaterial-associated murine infection model in which control-Ti, VAN-Ti or CAS-Ti substrates were implanted subcutaneously and subsequently challenged with the respective pathogens. The osseointegration potential of VAN-Ti and CAS-Ti was examined in vitro using human bone marrow-derived stromal cells, and for VAN-Ti also in a rat osseointegration model. RESULTS: In vitro biofilm formation of S. aureus and C. albicans on VAN-Ti and CAS-Ti substrates, respectively, was significantly reduced compared with biofilm formation on control-Ti. In vivo, we observed over 99.9% reduction in biofilm formation of S. aureus on VAN-Ti substrates and 89% reduction in biofilm formation of C. albicans on CAS-Ti substrates, compared with control-Ti substrates. The coated substrates supported osseointegration in vitro and in vivo. CONCLUSIONS: These data demonstrate the clinical potential of covalently bound VAN and CAS on Ti to reduce microbial biofilm formation without jeopardizing osseointegration.


Asunto(s)
Antiinfecciosos/farmacología , Biopelículas/efectos de los fármacos , Candida albicans/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Titanio/farmacología , Animales , Antibacterianos/farmacología , Antifúngicos/farmacología , Candida albicans/fisiología , Caspofungina , Línea Celular , Equinocandinas/farmacología , Femenino , Humanos , Lipopéptidos/farmacología , Ratones , Ratones Endogámicos BALB C , Oseointegración , Prótesis e Implantes/microbiología , Staphylococcus aureus/fisiología , Vancomicina/farmacología
17.
Mol Ecol ; 24(7): 1572-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25721227

RESUMEN

Microbial populations often contain a fraction of slow-growing persister cells that withstand antibiotics and other stress factors. Current theoretical models predict that persistence levels should reflect a stable state in which the survival advantage of persisters under adverse conditions is balanced with the direct growth cost impaired under favourable growth conditions, caused by the nonreplication of persister cells. Based on this direct growth cost alone, however, it remains challenging to explain the observed low levels of persistence (<<1%) seen in the populations of many species. Here, we present data from the opportunistic human pathogen Pseudomonas aeruginosa that can explain this discrepancy by revealing various previously unknown costs of persistence. In particular, we show that in the absence of antibiotic stress, increased persistence is traded off against a lengthened lag phase as well as a reduced survival ability during stationary phase. We argue that these pleiotropic costs contribute to the very low proportions of persister cells observed among natural P. aeruginosa isolates (3 × 10(-8) -3 × 10(-4)) and that they can explain why strains with higher proportions of persister cells lose out very quickly in competition assays under favourable growth conditions, despite a negligible difference in maximal growth rate. We discuss how incorporating these trade-offs could lead to models that can better explain the evolution of persistence in nature and facilitate the rational design of alternative therapeutic strategies for treating infectious diseases.


Asunto(s)
Farmacorresistencia Bacteriana/genética , Aptitud Genética , Pseudomonas aeruginosa/genética , Antibacterianos/farmacología , Evolución Molecular , Variación Genética , Pseudomonas aeruginosa/efectos de los fármacos
18.
Crit Rev Microbiol ; 40(3): 207-24, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23537324

RESUMEN

Obg proteins (also known as ObgE, YhbZ and CgtA) are conserved P-loop GTPases, essential for growth in bacteria. Like other GTPases, Obg proteins cycle between a GTP-bound ON and a GDP-bound OFF state, thereby controlling cellular processes. Interestingly, the in vitro biochemical properties of Obg proteins suggest that they act as sensors for the cellular GDP/GTP pools and adjust their activity according to the cellular energy status. Obg proteins have been attributed a host of cellular functions, including roles in essential cellular processes (DNA replication, ribosome maturation) and roles in different stress adaptation pathways (stringent response, sporulation, general stress response). This review summarizes the current knowledge on Obg activity and function. Furthermore, we present a model that integrates the different functions of Obg by assigning it a fundamental role in cellular physiology, at the hub of protein and DNA synthesis. In particular, we believe that Obg proteins might provide a connection between different global pathways in order to fine-tune cellular processes in response to a given energy status.


Asunto(s)
Bacterias/enzimología , Bacterias/crecimiento & desarrollo , ADN/biosíntesis , GTP Fosfohidrolasas/metabolismo , Biosíntesis de Proteínas , Bacterias/metabolismo , Regulación Bacteriana de la Expresión Génica , Redes Reguladoras de Genes , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Redes y Vías Metabólicas
19.
Bioorg Med Chem Lett ; 24(23): 5404-8, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25453797

RESUMEN

Pseudomonas aeruginosa strains resistant towards all currently available antibiotics are increasingly encountered, raising the need for new anti-pseudomonal drugs. We therefore conducted a medium-throughput screen of a small-molecule collection resulting in the identification of the N-alkylated 3,6-dihalogenocarbazol 1-(sec-butylamino)-3-(3,6-dichloro-9H-carbazol-9-yl)propan-2-ol (MIC = 18.5 µg mL⁻¹). This compound, compound 1, is bacteriostatic towards a broad spectrum of Gram-positive and Gram-negative pathogens, including P. aeruginosa. Importantly, 1 also eradicates mature biofilms of P. aeruginosa. 1 displays no cytotoxicity against various human cell types, pointing to its potential for further development as a novel antibacterial drug.


Asunto(s)
Antibacterianos/uso terapéutico , Carbazoles/química , Pseudomonas aeruginosa/aislamiento & purificación , Biopelículas , Carbazoles/análisis , Humanos , Pruebas de Sensibilidad Microbiana
20.
Appl Microbiol Biotechnol ; 98(10): 4589-602, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24728599

RESUMEN

Rhizobia are soil bacteria that can fix nitrogen in symbiosis with leguminous plants or exist free living in the rhizosphere. Crucial to their complex lifestyle is the ability to sense and respond to diverse environmental stimuli, requiring elaborate signaling pathways. In the majority of bacteria, the nucleotide-based second messenger cyclic diguanosine monophosphate (c-di-GMP) is involved in signal transduction. Surprisingly, little is known about the importance of c-di-GMP signaling in rhizobia. We have analyzed the genome sequences of six well-studied type species (Bradyrhizobium japonicum, Mesorhizobium loti, Rhizobium etli, Rhizobium leguminosarum, Sinorhizobium fredii, and Sinorhizobium meliloti) for proteins possibly involved in c-di-GMP signaling based on the presence of four domains: GGDEF (diguanylate cyclase), EAL and HD-GYP (phosphodiesterase), and PilZ (c-di-GMP sensor). We find that rhizobia possess a high number of these proteins. Conservation analysis suggests that c-di-GMP signaling proteins modulate species-specific pathways rather than ancient rhizobia-specific processes. Two hybrid GGDEF-EAL proteins were selected for functional analysis, R. etli RHE_PD00105 (CdgA) and RHE_PD00137 (CdgB). Expression of cdgA and cdgB is repressed by the alarmone (p)ppGpp. cdgB is significantly expressed on plant roots and free living. Mutation of cdgA, cdgB, or both does not affect plant root colonization, nitrogen fixation capacity, biofilm formation, motility, and exopolysaccharide production. However, heterologous expression of the individual GGDEF and EAL domains of each protein in Escherichia coli strongly suggests that CdgA and CdgB are bifunctional proteins, possessing both diguanylate cyclase and phosphodiesterase activities. Taken together, our results provide a platform for future studies of c-di-GMP signaling in rhizobia.


Asunto(s)
Bradyrhizobium/genética , GMP Cíclico/análogos & derivados , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Mesorhizobium/genética , Rhizobiaceae/genética , Microbiología del Suelo , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Bradyrhizobium/metabolismo , Bradyrhizobium/fisiología , Biología Computacional , GMP Cíclico/metabolismo , Perfilación de la Expresión Génica , Mesorhizobium/metabolismo , Mesorhizobium/fisiología , Fijación del Nitrógeno , Raíces de Plantas/microbiología , Polisacáridos/metabolismo , Rhizobiaceae/metabolismo , Rhizobiaceae/fisiología , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA