Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mBio ; 13(4): e0111922, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35913163

RESUMEN

Enterococcus faecalis is an opportunistic pathogen and a major cause of severe nosocomial infections. Treatment options against enterococcal infections are declining due to the resistance of enterococci to numerous antibiotics. A key risk factor for developing enterococcal infections is treatment with cephalosporin antibiotics, to which enterococci are intrinsically resistant. For susceptible organisms, cephalosporins inhibit bacterial growth by acylating the active site of penicillin-binding proteins (PBPs), key enzymes that catalyze peptidoglycan cross-linking. Two specific PBPs of enterococci, Pbp4(5) and PbpA(2b), exhibit low reactivity toward cephalosporins, allowing these PBPs to cross-link peptidoglycan in the presence of cephalosporins to drive resistance in enterococci, but the mechanisms by which these PBPs are regulated are poorly understood. The CroS/R two-component signal transduction system (TCS) is also required for cephalosporin resistance. Activation of CroS/R by cephalosporins leads to CroR-dependent changes in gene expression. However, the specific genes regulated by CroS/R that are responsible for cephalosporin resistance remain largely unknown. In this study, we characterized CroR-dependent transcriptome remodeling by RNA-seq, identifying pbp4(5) as a CroR regulon member in multiple, diverse lineages of E. faecalis. Through genetic analysis of the pbp4(5) and croR promoters, we uncovered a CroR-dependent regulatory motif. Mutations in this motif to disrupt CroR-dependent upregulation of pbp4(5) in the presence of cell wall stress resulted in a reduction of resistance to cephalosporins in E. faecalis, demonstrating that enhanced production of Pbp4(5) and likely other proteins involved in peptidoglycan biogenesis by the CroS/R system drives enterococcal cephalosporin resistance. IMPORTANCE Investigation into molecular mechanisms used by enterococci to subvert cephalosporin antibiotics is imperative for preventing and treating life-threatening infections. In this study, we used genetic means to investigate the functional output of the CroS/R TCS required for enterococcal resistance to cephalosporins. We found that enhanced production of the penicillin-binding protein Pbp4(5) upon exposure to cell wall stress was mediated by CroS/R and was critical for intrinsic cephalosporin resistance of E. faecalis.


Asunto(s)
Resistencia a las Cefalosporinas , Enterococcus faecalis , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Cefalosporinas/farmacología , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Pruebas de Sensibilidad Microbiana , Proteínas de Unión a las Penicilinas/metabolismo , Peptidoglicano/metabolismo
2.
mSystems ; 7(1): e0023021, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35076278

RESUMEN

The gut microbiome plays an essential role in host energy homeostasis and influences the development of obesity and related conditions. Studies demonstrate that nicotinamide riboside (NR) supplementation for diet-induced obesity (DIO) reduces weight gain and increases energy expenditure in mice. NR is a vitamin B3 derivative and an NAD+ precursor with potential for treating human diseases arising from mitochondrial degeneration, including obesity and type 2 diabetes. Gut bacteria produce vitamin B3 in the colon and are capable of salvaging and metabolizing vitamin B3 and its derivatives. However, it is unknown how dietary supplementation of NR alters the microbiome and if those alterations contribute to deflection of weight gain. In this study, we fed C57BL/6J male mice a high-fat diet (HFD) supplemented with or without NR and performed a fecal material transfer (FMT) to establish a link between NR-conditioned microbiota and NR-induced deflection of weight gain. FMT from NR-treated donors to naive mice fed a HFD was sufficient to deflect weight gain by increasing energy expenditure. We also investigated the effects of NR on the microbiome by using metagenomics sequencing. We found that NR-treated mice displayed an altered gut microbial composition relative to controls and that fecal transplant resulted in a distinct functional metabolic profile characterized by enrichment of butyrate-producing Firmicutes. NR-treated donors and subsequent FMT recipients share a similar enrichment of metagenomic biomarkers relative to controls. These findings suggest that microbial factors contribute to the beneficial effects of dietary NR supplementation, which may be useful to enhance the therapeutic effects of NR. IMPORTANCE With obesity and type 2 diabetes (T2D) at epidemic levels, we need to understand the complex nature of these diseases to design better therapeutics. The underlying causes of both obesity and T2D are complex, but both are thought to develop, in part, based on contributions from the gut microbiota. Nicotinamide riboside is a gut-derived vitamin B3 derivative and NAD+ precursor which has the potential to treat and prevent metabolic disorders by ameliorating mitochondrial dysfunction. Understanding how NR affects the gut microbiome and whether NR-conditioned microbiota contributes to weight loss in the host would (i) improve diagnosis and treatments for obesity and other metabolic pathologies, (ii) tailor treatments to satisfy the needs of each individual moving toward the future of precision medicine, and (iii) benefit other scientific fields that currently investigate the effects of NR in other disease pathologies.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Masculino , Humanos , Animales , Ratones , Dieta Alta en Grasa , NAD/efectos adversos , Ratones Endogámicos C57BL , Aumento de Peso , Obesidad/inducido químicamente , Vitaminas/efectos adversos
3.
J Bacteriol ; 201(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30858297

RESUMEN

Transmembrane Ser/Thr kinases containing extracellular PASTA (penicillin-binding protein [PBP] and Ser/Thr-associated) domains are ubiquitous among Actinobacteria and Firmicutes species. Such PASTA kinases regulate critical bacterial processes, including antibiotic resistance, cell division, cell envelope homeostasis, and virulence, and are sometimes essential for viability. Previous studies of purified PASTA kinase fragments revealed they are capable of autophosphorylation in vitro, typically at multiple sites on the kinase domain. Autophosphorylation of a specific structural element of the kinase known as the activation loop is thought to enhance kinase activity in response to stimuli. However, the role of kinase phosphorylation at other sites is largely unknown. Moreover, the mechanisms by which PASTA kinases are deactivated once their stimulus has diminished are poorly understood. Enterococcus faecalis is a Gram-positive intestinal bacterium and a major antibiotic-resistant opportunistic pathogen. In E. faecalis, the PASTA kinase IreK drives intrinsic resistance to cell wall-active antimicrobials, and such antimicrobials trigger enhanced phosphorylation of IreK in vivo Here we identify multiple sites of phosphorylation on IreK and evaluate their function in vivo and in vitro While phosphorylation of the IreK activation loop is required for kinase activity, we found that phosphorylation at a site distinct from the activation loop reciprocally modulates IreK activity in vivo, leading to diminished activity (and diminished antimicrobial resistance). Moreover, this site is important for deactivation of IreK in vivo upon removal of an activating stimulus. Our results are consistent with a model in which phosphorylation of IreK at distinct sites reciprocally regulates IreK activity in vivo to promote adaptation to cell wall stresses.IMPORTANCE Transmembrane Ser/Thr kinases containing extracellular PASTA domains are ubiquitous among Actinobacteria and Firmicutes species and regulate critical processes, including antibiotic resistance, cell division, and cell envelope homeostasis. Previous studies of PASTA kinase fragments revealed autophosphorylation at multiple sites. However, the functional role of autophosphorylation and the relative impacts of phosphorylation at distinct sites are poorly understood. The PASTA kinase of Enterococcus faecalis, IreK, regulates intrinsic resistance to antimicrobials. Here we identify multiple sites of phosphorylation on IreK and show that modification of IreK at distinct sites reciprocally regulates IreK activity and antimicrobial resistance in vivo Thus, these results provide new insights into the mechanisms by which PASTA kinases can regulate critical physiological processes in a wide variety of bacterial species.


Asunto(s)
Enterococcus faecalis/enzimología , Enterococcus faecalis/metabolismo , Regulación Bacteriana de la Expresión Génica , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Adaptación Fisiológica , Antiinfecciosos/farmacología , Pared Celular/efectos de los fármacos , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/genética , Fosforilación
4.
J Bacteriol ; 200(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29632091

RESUMEN

Two common signal transduction mechanisms used by bacteria to sense and respond to changing environments are two-component systems (TCSs) and eukaryote-like Ser/Thr kinases and phosphatases (eSTK/Ps). Enterococcus faecalis is a Gram-positive bacterium and a serious opportunistic pathogen that relies on both a TCS and an eSTK/P pathway for intrinsic resistance to cell wall-targeting antibiotics. The TCS consists of a histidine kinase (CroS) and a response regulator (CroR) that become activated upon exposure of cells to cell wall-targeting antibiotics, leading to a modulation of gene expression. The eSTK/P pathway consists of a transmembrane kinase (IreK) and its cognate phosphatase (IreP), which act antagonistically to mediate antibiotic resistance through an unknown mechanism. Because both CroS/R and IreK/P contribute to enterococcal resistance toward cell wall-targeting antibiotics, we hypothesized that these signaling systems are intertwined. To test this hypothesis, we analyzed CroR phosphorylation and CroS/R-dependent gene expression to probe the influence of IreK and IreP on CroS/R signaling. In addition, we analyzed the phosphorylation state of CroS, which revealed the IreK-dependent phosphorylation of a Thr residue important for CroS function. Our results are consistent with a model in which IreK positively influences CroR-dependent gene expression through the phosphorylation of CroS to promote antimicrobial resistance in E. faecalisIMPORTANCE Two-component signaling systems (TCSs) and eukaryote-like Ser/Thr kinases (eSTKs) are used by bacteria to sense and adapt to changing environments. Understanding how these pathways are regulated to promote bacterial survival is critical for a more complete understanding of bacterial stress responses and physiology. The opportunistic pathogen Enterococcus faecalis relies on both a TCS (CroS/R) and an eSTK (IreK) for intrinsic resistance to cell wall-targeting antibiotics. We probed the relationship between CroS/R and IreK, revealing the convergence of IreK and the sensor kinase CroS to enhance signaling through CroS/R and increase antimicrobial resistance in E. faecalis This newly described example of eSTK/TCS convergence adds to our understanding of the signaling networks mediating antimicrobial resistance in E. faecalis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Enterococcus faecalis/metabolismo , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Pared Celular/efectos de los fármacos , Pared Celular/enzimología , Pared Celular/genética , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/enzimología , Enterococcus faecalis/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Histidina Quinasa/genética , Fosforilación , Transducción de Señal/efectos de los fármacos
5.
Artículo en Inglés | MEDLINE | ID: mdl-28223383

RESUMEN

Enterococci are serious opportunistic pathogens that are resistant to many cell wall-targeting antibiotics. The CroRS two-component signaling system responds to antibiotic-mediated cell wall stress and is critical for resistance to cell wall-targeting antibiotics in Enterococcus faecalis Here, we identify and characterize an orthologous two-component system found in Enterococcus faecium that is functionally equivalent to the CroRS system of E. faecalis Deletion of croRS in E. faecium resulted in marked susceptibility to cell wall-targeting agents including cephalosporins and bacitracin, as well as moderate susceptibility to ampicillin and vancomycin. As in E. faecalis, exposure to bacitracin and vancomycin stimulates signaling through the CroRS system in E. faecium Moreover, the CroRS system is critical in E. faecium for enhanced beta-lactam resistance mediated by overexpression of Pbp5. Expression of a Pbp5 variant that confers enhanced beta-lactam resistance cannot overcome the requirement for CroRS function. Thus, the CroRS system is a conserved signaling system that responds to cell wall stress to promote intrinsic resistance to important cell wall-targeting antibiotics in clinically relevant enterococci.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/genética , Pared Celular/efectos de los fármacos , Enterococcus faecalis/genética , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/genética , Proteínas Quinasas/genética , Transactivadores/genética , Resistencia betalactámica/genética , Secuencia de Aminoácidos , Ampicilina/farmacología , Bacitracina/farmacología , Cefalosporinas/farmacología , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/metabolismo , Enterococcus faecium/metabolismo , Eliminación de Gen , Pruebas de Sensibilidad Microbiana , Transducción de Señal/genética , Vancomicina/farmacología
6.
J Bacteriol ; 198(8): 1326-36, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26883822

RESUMEN

UNLABELLED: Bacteria use two-component signal transduction systems (TCSs) to sense and respond to environmental changes via a conserved phosphorelay between a sensor histidine kinase and its cognate response regulator. The opportunistic pathogen Enterococcus faecalis utilizes a TCS comprised of the histidine kinase CroS and the response regulator CroR to mediate resistance to cell wall stresses such as cephalosporin antibiotics, but the molecular details by which CroRS promotes cephalosporin resistance have not been elucidated. Here, we analyzed mutants of E. faecalis carrying substitutions in CroR and CroS to demonstrate that phosphorylated CroR drives resistance to cephalosporins, and that CroS exhibits kinase and phosphatase activities to control the level of CroR phosphorylation in vivo. Deletion of croS in various lineages of E. faecalis revealed a CroS-independent mechanism for CroR phosphorylation and led to the identification of a noncognate histidine kinase capable of influencing CroR (encoded by OG1RF_12162; here called cisS). Further analysis of this TCS network revealed that both systems respond to cell wall stress. IMPORTANCE: TCSs allow bacteria to sense and respond to many different environmental conditions. The opportunistic pathogen Enterococcus faecalis utilizes the CroRS TCS to mediate resistance to cell wall stresses, including clinically relevant antibiotics such as cephalosporins and glycopeptides. In this study, we use genetic and biochemical means to investigate the relationship between CroRS signaling and cephalosporin resistance in E. faecalis cells. Through this, we uncovered a signaling network formed between the CroRS TCS and a previously uncharacterized TCS that also responds to cell wall stress. This study provides mechanistic insights into CroRS signaling and cephalosporin resistance in E. faecalis.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Pared Celular/efectos de los fármacos , Cefalosporinas/farmacología , Enterococcus faecalis/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Pared Celular/fisiología , Farmacorresistencia Bacteriana , Regulación Bacteriana de la Expresión Génica , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Mutación , Fosforilación , Transducción de Señal
7.
Antimicrob Agents Chemother ; 58(2): 957-65, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24277024

RESUMEN

Enterococci are ubiquitous inhabitants of the gastrointestinal (GI) tract. However, antibiotic-resistant enterococci are also major causes of hospital-acquired infections. Enterococci are intrinsically resistant to cephalosporins, enabling growth to abnormally high densities in the GI tract in patients during cephalosporin therapy, thereby promoting dissemination to other sites where they cause infection. Despite its importance, many questions about the underlying basis for cephalosporin resistance remain. A specific two-component signaling system, composed of the CroS sensor kinase and its cognate response regulator (CroR), is required for cephalosporin resistance in Enterococcus faecalis, but little is known about the factors that control this signaling system to modulate resistance. To explore the signaling network in which CroR participates to influence cephalosporin resistance, we employed a protein fragment complementation assay to detect protein-protein interactions in E. faecalis cells, revealing a previously unknown association of CroR with the HPr protein of the phosphotransferase system (PTS) responsible for carbohydrate uptake and catabolite control of gene expression. Genetic and physiological analyses indicate that association with HPr restricts the ability of CroR to promote cephalosporin resistance and gene expression in a nutrient-dependent manner. Mutational analysis suggests that the interface used by HPr to associate with CroR is distinct from the interface used to associate with other cellular partners. Our results define a physical and functional connection between a critical nutrient-responsive signaling system (the PTS) and a two-component signaling system that drives antibiotic resistance in E. faecalis, and they suggest a general strategy by which bacteria can integrate their nutritional status with diverse environmental stimuli.


Asunto(s)
Proteínas Bacterianas/metabolismo , Resistencia a las Cefalosporinas/genética , Enterococcus faecalis/genética , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Ceftriaxona/farmacología , Resistencia a las Cefalosporinas/efectos de los fármacos , Medios de Cultivo/química , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/metabolismo , Expresión Génica , Genes Reporteros , Glucosa/metabolismo , Operón Lac , Pruebas de Sensibilidad Microbiana , Mutación , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Proteínas Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...