Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Microbiol ; 121(6): 1148-1163, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38646792

RESUMEN

Enterococcal infections frequently show high levels of antibiotic resistance, including to cell envelope-acting antibiotics like daptomycin (DAP). While we have a good understanding of the resistance mechanisms, less is known about the control of such resistance genes in enterococci. Previous work unveiled a bacitracin resistance network, comprised of the sensory ABC transporter SapAB, the two-component system (TCS) SapRS and the resistance ABC transporter RapAB. Interestingly, components of this system have recently been implicated in DAP resistance, a role usually regulated by the TCS LiaFSR. To better understand the regulation of DAP resistance and how this relates to mutations observed in DAP-resistant clinical isolates of enterococci, we here explored the interplay between these two regulatory pathways. Our results show that SapR regulates an additional resistance operon, dltXABCD, a known DAP resistance determinant, and show that LiaFSR regulates the expression of sapRS. This regulatory structure places SapRS-target genes under dual control, where expression is directly controlled by SapRS, which itself is up-regulated through LiaFSR. The network structure described here shows how Enterococcus faecalis coordinates its response to cell envelope attack and can explain why clinical DAP resistance often emerges via mutations in regulatory components.


Asunto(s)
Antibacterianos , Bacitracina , Proteínas Bacterianas , Daptomicina , Farmacorresistencia Bacteriana , Enterococcus faecalis , Regulación Bacteriana de la Expresión Génica , Operón , Daptomicina/farmacología , Enterococcus faecalis/genética , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/metabolismo , Bacitracina/farmacología , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Pared Celular/metabolismo , Pared Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética
2.
Mol Microbiol ; 120(3): 408-424, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37475106

RESUMEN

Antimicrobial tolerance is the ability of a microbial population to survive, but not proliferate, during antimicrobial exposure. Significantly, it has been shown to precede the development of bona fide antimicrobial resistance. We have previously identified the two-component system CroRS as a critical regulator of tolerance to antimicrobials like teixobactin in the bacterial pathogen Enterococcus faecalis. To understand the molecular mechanism of this tolerance, we have carried out RNA-seq analyses in the E. faecalis wild-type and isogenic ∆ croRS mutant to determine the teixobactin-induced CroRS regulon. We identified a 132 gene CroRS regulon and demonstrate that CroRS upregulates biosynthesis of all major components of the enterococcal cell envelope in response to teixobactin. This suggests a coordinating role of this regulatory system in maintaining integrity of the multiple layers of the enterococcal envelope during antimicrobial stress, likely contributing to bacterial survival. Using experimental evolution, we observed that truncation of HppS, a key enzyme in the synthesis of the quinone electron carrier demethylmenaquinone, was sufficient to rescue tolerance in the croRS deletion strain. This highlights a key role for isoprenoid biosynthesis in antimicrobial tolerance in E. faecalis. Here, we propose a model of CroRS acting as a master regulator of cell envelope biogenesis and a gate-keeper between isoprenoid biosynthesis and respiration to ensure tolerance against antimicrobial challenge.


Asunto(s)
Antibacterianos , Antiinfecciosos , Antibacterianos/farmacología , Enterococcus faecalis/genética , Proteínas Bacterianas/genética , Homeostasis , Terpenos , Pruebas de Sensibilidad Microbiana
3.
Mol Microbiol ; 115(1): 157-174, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32955745

RESUMEN

Bacteria closely control gene expression to ensure optimal physiological responses to their environment. Such careful gene expression can minimize the fitness cost associated with antibiotic resistance. We previously described a novel regulatory logic in Bacillus subtilis enabling the cell to directly monitor its need for detoxification. This cost-effective strategy is achieved via a two-component regulatory system (BceRS) working in a sensory complex with an ABC-transporter (BceAB), together acting as a flux-sensor where signaling is proportional to transport activity. How this is realized at the molecular level has remained unknown. Using experimentation and computation we here show that the histidine kinase is activated by piston-like displacements in the membrane, which are converted to helical rotations in the catalytic core via an intervening HAMP-like domain. Intriguingly, the transporter was not only required for kinase activation, but also to actively maintain the kinase in its inactive state in the absence of antibiotics. Such coupling of kinase activity to that of the transporter ensures the complete control required for transport flux-dependent signaling. Moreover, we show that the transporter likely conserves energy by signaling with sub-maximal sensitivity. These results provide the first mechanistic insights into transport flux-dependent signaling, a unique strategy for energy-efficient decision making.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Bacillus subtilis/metabolismo , Histidina Quinasa/metabolismo , Transportadoras de Casetes de Unión a ATP/fisiología , Antibacterianos/farmacología , Bacillus subtilis/genética , Bacitracina/metabolismo , Bacitracina/farmacología , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/genética , Histidina Quinasa/fisiología , Proteínas de Transporte de Membrana/metabolismo , Transducción de Señal/efectos de los fármacos
4.
Microb Cell Fact ; 21(1): 57, 2022 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-35392905

RESUMEN

BACKGROUND: Subtilin is a peptide antibiotic (lantibiotic) natively produced by Bacillus subtilis ATCC6633. It is encoded in a gene cluster spaBTCSIFEGRK (spa-locus) consisting of four transcriptional units: spaS (subtilin pre-peptide), spaBTC (modification and export), spaIFEG (immunity) and spaRK (regulation). Despite the pioneer understanding on subtilin biosynthesis, a robust platform to facilitate subtilin research and improve subtilin production is still a poorly explored spot. RESULTS: In this work, the intact spa-locus was successfully integrated into the chromosome of Bacillus subtilis W168, which is the by far best-characterized Gram-positive model organism with powerful genetics and many advantages in industrial use. Through systematic analysis of spa-promoter activities in B. subtilis W168 wild type and mutant strains, our work demonstrates that subtilin is basally expressed in B. subtilis W168, and the transition state regulator AbrB strongly represses subtilin biosynthesis in a growth phase-dependent manner. The deletion of AbrB remarkably enhanced subtilin gene expression, resulting in comparable yield of bioactive subtilin production as for B. subtilis ATCC6633. However, while in B. subtilis ATCC6633 AbrB regulates subtilin gene expression via SigH, which in turn activates spaRK, AbrB of B. subtilis W168 controls subtilin gene expression in SigH-independent manner, except for the regulation of spaBTC. Furthermore, the work shows that subtilin biosynthesis in B. subtilis W168 is regulated by the two-component regulatory system SpaRK and strictly relies on subtilin itself as inducer to fulfill the autoregulatory circuit. In addition, by incorporating the subtilin-producing system (spa-locus) and subtilin-reporting system (PpsdA-lux) together, we developed "online" reporter strains to efficiently monitor the dynamics of subtilin biosynthesis. CONCLUSIONS: Within this study, the model organism B. subtilis W168 was successfully established as a novel platform for subtilin biosynthesis and the underlying regulatory mechanism was comprehensively characterized. This work will not only facilitate genetic (engineering) studies on subtilin, but also pave the way for its industrial production. More broadly, this work will shed new light on the heterologous production of other lantibiotics.


Asunto(s)
Bacillus subtilis , Bacteriocinas , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Péptidos/metabolismo
5.
Microbiology (Reading) ; 167(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33881981

RESUMEN

Micro-organisms contribute to Earth's mineral deposits through a process known as bacteria-induced mineral precipitation (BIMP). It is a complex phenomenon that can occur as a result of a variety of physiological activities that influence the supersaturation state and nucleation catalysis of mineral precipitation in the environment. There is a good understanding of BIMP induced by bacterial metabolism through the control of metal redox states and enzyme-mediated reactions such as ureolysis. However, other forms of BIMP often cannot be attributed to a single pathway but rather appear to be a passive result of bacterial activity, where minerals form as a result of metabolic by-products and surface interactions within the surrounding environment. BIMP from such processes has formed the basis of many new innovative biotechnologies, such as soil consolidation, heavy metal remediation, restoration of historic buildings and even self-healing concrete. However, these applications to date have primarily incorporated BIMP-capable bacteria sampled from the environment, while detailed investigations of the underpinning mechanisms have been lagging behind. This review covers our current mechanistic understanding of bacterial activities that indirectly influence BIMP and highlights the complexity and connectivity between the different cellular and metabolic processes involved. Ultimately, detailed insights will facilitate the rational design of application-specific BIMP technologies and deepen our understanding of how bacteria are shaping our world.


Asunto(s)
Bacterias/metabolismo , Minerales/química , Minerales/metabolismo , Bacterias/genética , Precipitación Química , Oxidación-Reducción , Suelo/química
6.
Microb Cell Fact ; 20(1): 214, 2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34794448

RESUMEN

BACKGROUND: Microbially induced calcite precipitation (MICP) is an ancient property of bacteria, which has recently gained considerable attention for biotechnological applications. It occurs as a by-product of bacterial metabolism and involves a combination of chemical changes in the extracellular environment, e.g. pH increase, and presence of nucleation sites on the cell surface or extracellular substances produced by the bacteria. However, the molecular mechanisms underpinning MICP and the interplay between the contributing factors remain poorly understood, thus placing barriers to the full biotechnological and synthetic biology exploitation of bacterial biomineralisation. RESULTS: In this study, we adopted a bottom-up approach of systematically engineering Bacillus subtilis, which has no detectable intrinsic MICP activity, for biomineralisation. We showed that heterologous production of urease can induce MICP by local increases in extracellular pH, and this can be enhanced by co-expression of urease accessory genes for urea and nickel uptake, depending on environmental conditions. MICP can be strongly enhanced by biofilm-promoting conditions, which appeared to be mainly driven by production of exopolysaccharide, while the protein component of the biofilm matrix was dispensable. Attempts to modulate the cell surface charge of B. subtilis had surprisingly minor effects, and our results suggest this organism may intrinsically have a very negative cell surface, potentially predisposing it for MICP activity. CONCLUSIONS: Our findings give insights into the molecular mechanisms driving MICP in an application-relevant chassis organism and the genetic elements that can be used to engineer de novo or enhanced biomineralisation. This study also highlights mutual influences between the genetic drivers and the chemical composition of the surrounding environment in determining the speed, spatial distribution and resulting mineral crystals of MICP. Taken together, these data pave the way for future rational design of synthetic precipitator strains optimised for specific applications.


Asunto(s)
Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Biomineralización , Carbonato de Calcio/metabolismo , Polisacáridos Bacterianos/metabolismo , Ureasa/metabolismo , Biopelículas , Carbonato de Calcio/química , Precipitación Química , ADN Bacteriano , Ingeniería Genética , Concentración de Iones de Hidrógeno , Microbiología Industrial/métodos , Níquel/metabolismo , Eliminación de Secuencia , Urea/metabolismo
7.
Artículo en Inglés | MEDLINE | ID: mdl-31871088

RESUMEN

Resistance against cell wall-active antimicrobial peptides in bacteria is often mediated by transporters. In low-GC-content Gram-positive bacteria, a common type of such transporters is BceAB-like systems, which frequently provide high-level resistance against peptide antibiotics that target intermediates of the lipid II cycle of cell wall synthesis. How a transporter can offer protection from drugs that are active on the cell surface, however, has presented researchers with a conundrum. Multiple theories have been discussed, ranging from removal of the peptides from the membrane and internalization of the drug for degradation to removal of the cellular target rather than the drug itself. To resolve this much-debated question, we here investigated the mode of action of the transporter BceAB of Bacillus subtilis We show that it does not inactivate or import its substrate antibiotic bacitracin. Moreover, we present evidence that the critical factor driving transport activity is not the drug itself but instead the concentration of drug-target complexes in the cell. Our results, together with previously reported findings, lead us to propose that BceAB-type transporters act by transiently freeing lipid II cycle intermediates from the inhibitory grip of antimicrobial peptides and thus provide resistance through target protection of cell wall synthesis. Target protection has so far only been reported for resistance against antibiotics with intracellular targets, such as the ribosome. However, this mechanism offers a plausible explanation for the use of transporters as resistance determinants against cell wall-active antibiotics in Gram-positive bacteria where cell wall synthesis lacks the additional protection of an outer membrane.


Asunto(s)
Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Pared Celular/efectos de los fármacos , Bacillus subtilis/efectos de los fármacos , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos
8.
Appl Environ Microbiol ; 86(7)2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31980427

RESUMEN

Microbially induced calcite precipitation (MICP) has not only helped to shape our planet's geological features but is also a promising technology to address environmental concerns in civil engineering applications. However, limited understanding of the biomineralization capacity of environmental bacteria impedes application. We therefore surveyed the environment for different mechanisms of precipitation across bacteria. The most fundamental difference was in ureolytic ability, where urease-positive bacteria caused rapid, widespread increases in pH, whereas nonureolytic strains produced such changes slowly and locally. These pH shifts correlated well with patterns of precipitation on solid medium. Strikingly, while both mechanisms led to high levels of precipitation, we observed clear differences in the precipitate. Ureolytic bacteria produced homogenous, inorganic fine crystals, whereas the crystals of nonureolytic strains were larger and had a mixed organic/inorganic composition. When representative strains were tested in application for crack healing in cement mortars, nonureolytic bacteria gave robust results, while ureolytic strains showed more variation. This may be explained by our observation that urease activity differed between growth conditions or by the different natures and therefore different material performances of the precipitates. Our results shed light on the breadth of biomineralization activity among environmental bacteria, an important step toward the rational design of bacterially based engineering solutions.IMPORTANCE Biomineralization triggered by bacteria is important in the natural environment and has many applications in industry and in civil and geotechnical engineering. The diversity in biomineralization capabilities of environmental bacteria is, however, not well understood. This study surveyed environmental bacteria for their ability to precipitate calcium carbonate minerals and investigated both the mechanisms and the resulting crystals. We show that while urease activity leads to the fastest precipitation, it is by no means essential. Importantly, the same quantities of calcium carbonate are produced by nonureolytic bacteria, and the resulting crystals appear to have larger volumes and more organic components, which are likely beneficial in specific applications. Testing both precipitation mechanisms in a self-healing concrete application showed that nonureolytic bacteria delivered more robust results. Here, we performed a systematic study of the fundamental differences in biomineralization between environmental bacteria, and we provide important information for the design of bacterially based engineering solutions.


Asunto(s)
Bacterias/metabolismo , Carbonato de Calcio/metabolismo , Microbiología Ambiental , Carbonato de Calcio/química , Precipitación Química
9.
Appl Environ Microbiol ; 86(14)2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32414796

RESUMEN

Bce-like systems mediate resistance against antimicrobial peptides in Firmicutes bacteria. Lactobacillus casei BL23 encodes an "orphan" ABC transporter that, based on homology to BceAB-like systems, was proposed to contribute to antimicrobial peptide resistance. A mutant lacking the permease subunit was tested for sensitivity against a collection of peptides derived from bacteria, fungi, insects, and humans. Our results show that the transporter specifically conferred resistance against insect-derived cysteine-stabilized αß defensins, and it was therefore renamed DerAB for defensin resistance ABC transporter. Surprisingly, cells lacking DerAB showed a marked increase in resistance against the lantibiotic nisin. This could be explained by significantly increased expression of the antimicrobial peptide resistance determinants regulated by the Bce-like systems PsdRSAB (formerly module 09) and ApsRSAB (formerly module 12). Bacterial two-hybrid studies in Escherichia coli showed that DerB could interact with proteins of the sensory complex in the Psd resistance system. We therefore propose that interaction of DerAB with this complex in the cell creates signaling interference and reduces the cell's potential to mount an effective nisin resistance response. In the absence of DerB, this negative interference is relieved, leading to the observed hyperactivation of the Psd module and thus increased resistance to nisin. Our results unravel the function of a previously uncharacterized Bce-like orphan resistance transporter with pleiotropic biological effects on the cell.IMPORTANCE Antimicrobial peptides (AMPs) play an important role in suppressing the growth of microorganisms. They can be produced by bacteria themselves-to inhibit competitors-but are also widely distributed in higher eukaryotes, including insects and mammals, where they form an important component of innate immunity. In low-GC-content Gram-positive bacteria, BceAB-like transporters play a crucial role in AMP resistance but have so far been primarily associated with interbacterial competition. Here, we show that the orphan transporter DerAB from the lactic acid bacterium Lactobacillus casei is crucial for high-level resistance against insect-derived AMPs. It therefore represents an important mechanism for interkingdom defense. Furthermore, our results support a signaling interference from DerAB on the PsdRSAB module that might prevent the activation of a full nisin response. The Bce modules from L. casei BL23 illustrate a biological paradox in which the intrinsic nisin detoxification potential only arises in the absence of a defensin-specific ABC transporter.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Antibiosis , Proteínas Bacterianas/genética , Defensinas/química , Proteínas de Insectos/química , Lacticaseibacillus casei/genética , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Lacticaseibacillus casei/metabolismo
10.
EMBO J ; 34(16): 2162-81, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26157010

RESUMEN

The epithelial to mesenchymal transition (EMT) is a biological process in which cells lose cell-cell contacts and become motile. EMT is used during development, for example, in triggering neural crest migration, and in cancer metastasis. Despite progress, the dynamics of JNK signaling, its role in genomewide transcriptional reprogramming, and involved downstream effectors during EMT remain largely unknown. Here, we show that JNK is not required for initiation, but progression of phenotypic changes associated with EMT. Such dependency resulted from JNK-driven transcriptional reprogramming of critical EMT genes and involved changes in their chromatin state. Furthermore, we identified eight novel JNK-induced transcription factors that were required for proper EMT. Three of these factors were also highly expressed in invasive cancer cells where they function in gene regulation to maintain mesenchymal identity. These factors were also induced during neuronal development and function in neuronal migration in vivo. These comprehensive findings uncovered a kinetically distinct role for the JNK pathway in defining the transcriptome that underlies mesenchymal identity and revealed novel transcription factors that mediate these responses during development and disease.


Asunto(s)
Diferenciación Celular , Redes Reguladoras de Genes , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Mesodermo/fisiología , Ciclo Celular , Línea Celular , Perfilación de la Expresión Génica , Humanos , Imagen de Lapso de Tiempo , Factores de Transcripción/metabolismo
11.
Microbiology (Reading) ; 165(4): 475-487, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30777814

RESUMEN

Bacitracin is a cell wall targeting antimicrobial with clinical and agricultural applications. With the growing mismatch between antimicrobial resistance and development, it is essential we understand the molecular mechanisms of resistance in order to prioritize and generate new effective antimicrobials. BcrR is a unique membrane-bound one-component system that regulates high-level bacitracin resistance in Enterococcus faecalis. In the presence of bacitracin, BcrR activates transcription of the bcrABD operon conferring resistance through a putative ATP-binding cassette (ABC) transporter (BcrAB). BcrR has three putative functional domains, an N-terminal helix-turn-helix DNA-binding domain, an intermediate oligomerization domain and a C-terminal transmembrane domain. However, the molecular mechanisms of signal transduction remain unknown. Random mutagenesis of bcrR was performed to generate loss- and gain-of-function mutants using transcriptional reporters fused to the target promoter PbcrA. Fifteen unique mutants were isolated across all three proposed functional domains, comprising 14 loss-of-function and one gain-of-function mutant. The gain-of-function variant (G64D) mapped to the putative dimerization domain of BcrR, and functional analyses indicated that the G64D mutant constitutively expresses the PbcrA-luxABCDE reporter. DNA-binding and membrane insertion were not affected in the five mutants chosen for further characterization. Homology modelling revealed putative roles for two key residues (R11 and S33) in BcrR activation. Here we present a new model of BcrR activation and signal transduction, providing valuable insight into the functional characterization of membrane-bound one-component systems and how they can coordinate critical bacterial responses, such as antimicrobial resistance.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Bacitracina/metabolismo , Membrana Celular/metabolismo , Farmacorresistencia Bacteriana , Transducción de Señal/fisiología , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/genética , Bacitracina/farmacología , Farmacorresistencia Bacteriana/genética , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/genética , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos/genética , Mutación , Operón , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos
12.
Arch Gynecol Obstet ; 299(1): 239-246, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30357498

RESUMEN

BACKGROUND: Tumor-infiltrating lymphocytes influence the prognosis of solid tumors, including ovarian cancer (OC). The immunoregulatory transcription factor (IRF4) is mainly expressed in plasma cells and regulates immunoglobulin class switch recombination as well as plasma cell differentiation. Therefore, we analyzed the impact of IRF4 expression in a consecutive cohort of OC patients. METHODS: IRF4 expression was evaluated by immunostaining. Differences in IRF4 expression among the subgroups of the established clinical-pathological features like age, histological subtype, tumor stage, histological grading, postoperative tumor burden, and completeness of chemotherapy were determined by χ2 test. The impact of IRF4 expression on progression-free survival (PFS) and overall survival (OS) was examined by univariate and multivariate Cox analysis adjusted for established clinical-pathological factors and Kaplan-Meier survival analysis. RESULTS: 114 patients entered this study. IRF4 was expressed in 51.7% of the entire cohort. 72.3% patients with high-grade serous OC showed IRF4 expression compared to 37.3% patients with a non-high-grade serous OC (p < 0.001). Univariate Cox-regression analysis revealed no prognostic impact of IRF4 expression in terms of PFS (p = 0.35) and OS (p = 0.98). Kaplan-Meier plots failed to show any prognostic impact for PFS (p = 0.35) and OS (p = 0.98), too. Established clinical-pathological factors retained their prognostic impact as tumor stage in terms of PFS (< 0.001) and as postoperative residual tumor burden (p = 0.04), tumor stage (< 0.001), histological grade (p = 0.02), and completeness of chemotherapy (p < 0.001) in terms of OS, respectively. CONCLUSION: Immunohistochemically determined IRF4 expression correlated with high-grade serous OC. However, it failed to show any prognostic impact in this cohort of 114 patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Factores Reguladores del Interferón/sangre , Neoplasias Ováricas/patología , Adulto , Anciano , Carcinoma Epitelial de Ovario , Estudios de Cohortes , Cistadenocarcinoma Seroso/sangre , Cistadenocarcinoma Seroso/patología , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Persona de Mediana Edad , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Pronóstico , Estudios Retrospectivos
13.
Mol Microbiol ; 104(1): 1-15, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28152228

RESUMEN

Transporters are essential players in bacterial growth and survival, since they are key for uptake of nutrients on the one hand, and for defence against endogenous and environmental stresses on the other hand. Remarkably, in addition to their primary role in substrate translocation, it has become clear that some transporters have acquired a secondary function as sensors and information processors in signalling pathways. In this review, we describe recent advances in our understanding of the role of transporters in such signalling cascades, and discuss some of the emergent dynamic behaviour found in hallmark examples. A particular focus is placed on new insights into mechanistic details of information transfer between transporters and regulatory proteins. Quantitative considerations reveal that these signalling complexes can implement a remarkable diversity of regulatory logic functions, where the transporter can act as activity switch, as positive or negative reporter of transport flux, or as a signalling hub for the integration of multiple inputs. Such a dual use of transport proteins not only enables efficient substrate translocation but is also an elegant strategy to integrate important information about the cell's external conditions with its current physiological state.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/fisiología , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Transducción de Señal/fisiología
14.
Mol Microbiol ; 104(1): 16-31, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27997719

RESUMEN

BceRS and PsdRS are paralogous two-component systems in Bacillus subtilis controlling the response to antimicrobial peptides. In the presence of extracellular bacitracin and nisin, respectively, the two response regulators (RRs) bind their target promoters, PbceA or PpsdA , resulting in a strong up-regulation of target gene expression and ultimately antibiotic resistance. Despite high sequence similarity between the RRs BceR and PsdR and their known binding sites, no cross-regulation has been observed between them. We therefore investigated the specificity determinants of PbceA and PpsdA that ensure the insulation of these two paralogous pathways at the RR-promoter interface. In vivo and in vitro analyses demonstrate that the regulatory regions within these two promoters contain three important elements: in addition to the known (main) binding site, we identified a linker region and a secondary binding site that are crucial for functionality. Initial binding to the high-affinity, low-specificity main binding site is a prerequisite for the subsequent highly specific binding of a second RR dimer to the low-affinity secondary binding site. In addition to this hierarchical cooperative binding, discrimination requires a competition of the two RRs for their respective binding site mediated by only slight differences in binding affinities.


Asunto(s)
Bacillus subtilis/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Antibacterianos/farmacología , Bacillus subtilis/genética , Bacitracina/metabolismo , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Sitios de Unión , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Datos de Secuencia Molecular , Nisina/metabolismo , Regiones Promotoras Genéticas , Unión Proteica/genética , Secuencias Reguladoras de Ácidos Nucleicos , Regulación hacia Arriba
15.
Mol Microbiol ; 100(4): 607-20, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26815905

RESUMEN

Protection against antimicrobial peptides (AMPs) often involves the parallel production of multiple, well-characterized resistance determinants. So far, little is known about how these resistance modules interact and how they jointly protect the cell. Here, we studied the interdependence between different layers of the envelope stress response of Bacillus subtilis when challenged with the lipid II cycle-inhibiting AMP bacitracin. The underlying regulatory network orchestrates the production of the ABC transporter BceAB, the UPP phosphatase BcrC and the phage-shock proteins LiaIH. Our systems-level analysis reveals a clear hierarchy, allowing us to discriminate between primary (BceAB) and secondary (BcrC and LiaIH) layers of bacitracin resistance. Deleting the primary layer provokes an enhanced induction of the secondary layer to partially compensate for this loss. This study reveals a direct role of LiaIH in bacitracin resistance, provides novel insights into the feedback regulation of the Lia system, and demonstrates a pivotal role of BcrC in maintaining cell wall homeostasis. The compensatory regulation within the bacitracin network can also explain how gene expression noise propagates between resistance layers. We suggest that this active redundancy in the bacitracin resistance network of B. subtilis is a general principle to be found in many bacterial antibiotic resistance networks.


Asunto(s)
Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/genética , Bacitracina/farmacología , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana , Pared Celular/metabolismo , Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
16.
Microbiology (Reading) ; 162(1): 164-176, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26364265

RESUMEN

When faced with carbon source limitation, the Gram-positive soil organism Bacillus subtilis initiates a survival strategy called sporulation, which leads to the formation of highly resistant endospores that allow B. subtilis to survive even long periods of starvation. In order to avoid commitment to this energy-demanding and irreversible process, B. subtilis employs another strategy called 'cannibalism' to delay sporulation as long as possible. Cannibalism involves the production and secretion of two cannibalism toxins, sporulation delaying protein (SDP) and sporulation killing factor (SKF), which are able to lyse sensitive siblings. The lysed cells are thought to then provide nutrients for the cannibals to slow down or even prevent them from entering sporulation. In this study, we uncovered the role of the cell envelope stress response (CESR), especially the Bce-like antimicrobial peptide detoxification modules, in the cannibalism stress response during the stationary phase. SDP and SKF specifically induce Bce-like systems and some extracytoplasmic function σ factors in stationary-phase cultures, but only the latter provide some degree of protection. A full Bce response is only triggered by mature toxins, and not by toxin precursors. Our study provides insights into the close relationship between stationary-phase survival and the CESR of B. subtilis.


Asunto(s)
Bacillus subtilis/fisiología , Bacillus subtilis/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Estrés Fisiológico
17.
J Biol Chem ; 289(40): 27899-910, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25118291

RESUMEN

Resistance against antimicrobial peptides in many Firmicutes bacteria is mediated by detoxification systems that are composed of a two-component regulatory system (TCS) and an ATP-binding cassette (ABC) transporter. The histidine kinases of these systems depend entirely on the transporter for sensing of antimicrobial peptides, suggesting a novel mode of signal transduction where the transporter constitutes the actual sensor. The aim of this study was to investigate the molecular mechanisms of this unusual signaling pathway in more detail, using the bacitracin resistance system BceRS-BceAB of Bacillus subtilis as an example. To analyze the proposed communication between TCS and the ABC transporter, we characterized their interactions by bacterial two-hybrid analyses and could show that the permease BceB and the histidine kinase BceS interact directly. In vitro pulldown assays confirmed this interaction, which was found to be independent of bacitracin. Because it was unknown whether BceAB-type transporters could detect their substrate peptides directly or instead recognized the peptide-target complex in the cell envelope, we next analyzed substrate binding by the transport permease, BceB. Direct and specific binding of bacitracin by BceB was demonstrated by surface plasmon resonance spectroscopy. Finally, in vitro signal transduction assays indicated that complex formation with the transporter influenced the autophosphorylation activity of the histidine kinase. Taken together, our findings clearly show the existence of a sensory complex composed of TCS and ABC transporters and provide the first functional insights into the mechanisms of stimulus perception, signal transduction, and antimicrobial resistance employed by Bce-like detoxification systems.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Antibacterianos/metabolismo , Bacillus subtilis/metabolismo , Bacitracina/metabolismo , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Proteínas de Transporte de Membrana/metabolismo , Proteínas Quinasas/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/enzimología , Bacillus subtilis/genética , Bacitracina/farmacología , Proteínas Bacterianas/genética , Histidina Quinasa , Proteínas de Transporte de Membrana/genética , Unión Proteica , Proteínas Quinasas/genética
18.
J Bacteriol ; 196(19): 3472-81, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25049090

RESUMEN

Bacterial uptake of phosphate is usually accomplished via high-affinity transporters that are commonly regulated by two-component systems, which are activated when the concentration of phosphate is low. Mycobacterium smegmatis possesses two such transporters, the widely distributed PstSCAB system and PhnDCE, a transporter that in other bacteria mediates the uptake of alternative phosphorus sources. We previously reported that the transcriptional regulator PhnF controls the production of the Phn system, acting as a repressor under high-phosphate conditions. Here we show that the phnDCE genes are common among environmental mycobacteria, where they are often associated with phnF-like genes. In contrast, pathogenic mycobacteria were not found to encode Phn-like systems but instead were found to possess multiple copies of the pst genes. A detailed biochemical analysis of PhnF binding to its identified binding sites in the phnD-phnF intergenic region of M. smegmatis has allowed us to propose a quantitative model for repressor binding, which shows that a PhnF dimer binds independently to each site. We present the crystal structure of M. smegmatis PhnF at 1.8-Å resolution, showing a homodimer with a helix-turn-helix N-terminal domain and a C-terminal domain with a UbiC transcription regulator-associated fold. The C-terminal domain crystallized with a bound sulfate ion instead of the so far unidentified physiological ligand, allowing the identification of residues involved in effector binding. Comparison of the positioning of the DNA binding domains in PhnF with that in homologous proteins suggests that its DNA binding activity is regulated via a conformational change in the linker region, triggering a movement of the N-terminal domains.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas de Transporte de Membrana/genética , Mycobacterium smegmatis/metabolismo , Fosfatos/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteínas Bacterianas/genética , Sitios de Unión , Transporte Biológico , Cristalografía por Rayos X , Regulación Bacteriana de la Expresión Génica , Proteínas de Transporte de Membrana/metabolismo , Familia de Multigenes , Mycobacterium smegmatis/química , Mycobacterium smegmatis/genética , Operón , Unión Proteica , Factores de Transcripción/genética
19.
Antimicrob Agents Chemother ; 58(3): 1425-33, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24342648

RESUMEN

Resistance of Enterococcus faecalis against antimicrobial peptides, both of host origin and produced by other bacteria of the gut microflora, is likely to be an important factor in the bacterium's success as an intestinal commensal. The aim of this study was to identify proteins with a role in resistance against the model antimicrobial peptide bacitracin. Proteome analysis of bacitracin-treated and untreated cells showed that bacitracin stress induced the expression of cell wall-biosynthetic proteins and caused metabolic rearrangements. Among the proteins with increased production, an ATP-binding cassette (ABC) transporter with similarity to known peptide antibiotic resistance systems was identified and shown to mediate resistance against bacitracin. Expression of the transporter was dependent on a two-component regulatory system and a second ABC transporter, which were identified by genome analysis. Both resistance and the regulatory pathway could be functionally transferred to Bacillus subtilis, proving the function and sufficiency of these components for bacitracin resistance. Our data therefore show that the two ABC transporters and the two-component system form a resistance network against antimicrobial peptides in E. faecalis, where one transporter acts as the sensor that activates the TCS to induce production of the second transporter, which mediates the actual resistance.


Asunto(s)
Antibacterianos/farmacología , Bacitracina/farmacología , Enterococcus faecalis/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/fisiología , Farmacorresistencia Bacteriana , Enterococcus faecalis/genética , Enterococcus faecalis/fisiología , Regulación Bacteriana de la Expresión Génica , Pruebas de Sensibilidad Microbiana , Regiones Promotoras Genéticas/genética , Proteoma
20.
Environ Microbiol ; 16(5): 1225-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24548478

RESUMEN

The Firmicutes constitute a phylum of bacteria that can be found in a wide variety of habitats, from soil to the gastrointestinal tract of animals, where they have to thrive in complex communities. Competition in these communities usually involves the production of compounds such as antimicrobial peptides (AMPs) to eliminate competitor organisms. Animals and plants also produce AMPs to control their associated microbiota. In turn, defence mechanisms have evolved to prevent the action of these compounds. The close association of some Firmicutes with humans as prominent pathogens or commensal organisms has driven a considerable research effort on defence mechanisms used by these bacteria against antimicrobial compounds. This review focuses on the most recent advances on two well-characterized defence mechanisms against AMPs: the modification of the cell wall by D-alanylation and the role of peptide antibiotic-specific adenosine triphosphate-binding cassette transporters.


Asunto(s)
Antibacterianos/metabolismo , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/metabolismo , Bacterias Grampositivas/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Antibacterianos/química , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Pared Celular/química , Pared Celular/metabolismo , Farmacorresistencia Bacteriana , Bacterias Grampositivas/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Ácidos Teicoicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA