Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Microbiol ; 21(1): 247, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34525965

RESUMEN

BACKGROUND: Infants with cystic fibrosis (CF) suffer from gastrointestinal (GI) complications, including pancreatic insufficiency and intestinal inflammation, which have been associated with impaired nutrition and growth. Recent evidence identified altered fecal microbiota taxonomic compositions in infants with CF relative to healthy infants that were characterized by differences in the abundances of taxa associated with GI health and nutrition. Furthermore, these taxonomic differences were more pronounced in low length infants with CF, suggesting a potential link to linear growth failure. We hypothesized that these differences would entail shifts in the microbiome's functional capacities that could contribute to inflammation and nutritional failure in infants with CF. RESULTS: To test this hypothesis, we compared fecal microbial metagenomic content between healthy infants and infants with CF, supplemented with an analysis of fecal metabolomes in infants with CF. We identified notable differences in CF fecal microbial functional capacities, including metabolic and environmental response functions, compared to healthy infants that intensified during the first year of life. A machine learning-based longitudinal metagenomic age analysis of healthy and CF fecal metagenomic functional profiles further demonstrated that these differences are characterized by a CF-associated delay in the development of these functional capacities. Moreover, we found metagenomic differences in functions related to metabolism among infants with CF that were associated with diet and antibiotic exposure, and identified several taxa as potential drivers of these functional differences. An integrated metagenomic and metabolomic analysis further revealed that abundances of several fecal GI metabolites important for nutrient absorption, including three bile acids, correlated with specific microbes in infants with CF. CONCLUSIONS: Our results highlight several metagenomic and metabolomic factors, including bile acids and other microbial metabolites, that may impact nutrition, growth, and GI health in infants with CF. These factors could serve as promising avenues for novel microbiome-based therapeutics to improve health outcomes in these infants.


Asunto(s)
Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Disbiosis/complicaciones , Heces/microbiología , Enfermedades Gastrointestinales/etiología , Metaboloma , Metagenoma , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/fisiopatología , Humanos , Lactante , Estudios Longitudinales , Metabolómica/métodos , Estudios Prospectivos
2.
Commun Biol ; 4(1): 817, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34188171

RESUMEN

Multi-resistant bacteria are a major threat in modern medicine. The gram-negative coccobacillus Acinetobacter baumannii currently leads the WHO list of pathogens in critical need for new therapeutic development. The maintenance of lipid asymmetry (MLA) protein complex is one of the core machineries that transport lipids from/to the outer membrane in gram-negative bacteria. It also contributes to broad-range antibiotic resistance in several pathogens, most prominently in A. baumannii. Nonetheless, the molecular details of its role in lipid transport has remained largely elusive. Here, we report the cryo-EM maps of the core MLA complex, MlaBDEF, from the pathogen A. baumannii, in the apo-, ATP- and ADP-bound states, revealing multiple lipid binding sites in the cytosolic and periplasmic side of the complex. Molecular dynamics simulations suggest their potential trajectory across the membrane. Collectively with the recently-reported structures of the E. coli orthologue, this data also allows us to propose a molecular mechanism of lipid transport by the MLA system.


Asunto(s)
Acinetobacter baumannii/química , Lípidos de la Membrana/química , Adenosina Trifosfato/química , Sitios de Unión , Membrana Celular/química , Microscopía por Crioelectrón , Simulación de Dinámica Molecular
3.
J Pediatr Gastroenterol Nutr ; 73(3): 395-402, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34016873

RESUMEN

OBJECTIVES: To identify factors that increase the risk of gastrointestinal-related (GI-related) hospitalization of infants with cystic fibrosis (CF) during the first year of life. METHODS: The Baby Observational and Nutrition Study was a longitudinal, observational cohort of 231 infants diagnosed with CF by newborn screening. We performed a post-hoc assessment of the frequency and indications for GI-related admissions during the first year of life. RESULTS: Sixty-five participants had at least one admission in the first 12 months of life. High pancreatic enzyme replacement therapy (PERT) dosing (>2000 lipase units/kg per meal; hazard ratio [HR] = 14.75, P = 0.0005) and use of acid suppressive medications (HR = 4.94, P = 0.01) during the study period were positively associated with subsequent GI-related admissions. High levels of fecal calprotectin (fCP) (>200 µg/g) and higher relative abundance of fecal Klebsiella pneumoniae were also positively associated with subsequent GI-related admissions (HR = 2.64, P = 0.033 and HR = 4.49, P = 0.002, respectively). During the first 12 months of life, participants with any admission had lower weight-for-length z scores (WLZ) (P = 0.01). The impact of admission on WLZ was particularly evident in participants with a GI-related admission (P < 0.0001). CONCLUSIONS: Factors associated with a higher risk for GI-related admission during the first 12 months include high PERT dosing, exposure to acid suppressive medications, higher fCP levels, and/or relative abundance of fecal K pneumoniae early in life. Infants with CF requiring GI-related hospitalization had lower WLZ at 12 months of age than those not admitted as well as those admitted for non-GI-related indications.


Asunto(s)
Fibrosis Quística , Estudios de Cohortes , Fibrosis Quística/complicaciones , Fibrosis Quística/tratamiento farmacológico , Terapia de Reemplazo Enzimático , Hospitalización , Humanos , Lactante , Recién Nacido , Tamizaje Neonatal
4.
Thorax ; 75(9): 780-790, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32631930

RESUMEN

RATIONALE: The most common antibiotic used to treat people with cystic fibrosis (PWCF) is inhaled tobramycin, administered as maintenance therapy for chronic Pseudomonas aeruginosa lung infections. While the effects of inhaled tobramycin on P. aeruginosa abundance and lung function diminish with continued therapy, this maintenance treatment is known to improve long-term outcomes, underscoring how little is known about why antibiotics work in CF infections, what their effects are on complex CF sputum microbiomes and how to improve these treatments. OBJECTIVES: To rigorously define the effect of maintenance tobramycin on CF sputum microbiome characteristics. METHODS AND MEASUREMENTS: We collected sputum from 30 PWCF at standardised times before, during and after a single month-long course of maintenance inhaled tobramycin. We used traditional culture, quantitative PCR and metagenomic sequencing to define the dynamic effects of this treatment on sputum microbiomes, including abundance changes in both clinically targeted and untargeted bacteria, as well as functional gene categories. MAIN RESULTS: CF sputum microbiota changed most markedly by 1 week of antibiotic therapy and plateaued thereafter, and this shift was largely driven by changes in non-dominant taxa. The genetically conferred functional capacities (ie, metagenomes) of subjects' sputum communities changed little with antibiotic perturbation, despite taxonomic shifts, suggesting functional redundancy within the CF sputum microbiome. CONCLUSIONS: Maintenance treatment with inhaled tobramycin, an antibiotic with demonstrated long-term mortality benefit, primarily impacted clinically untargeted bacteria in CF sputum, highlighting the importance of monitoring the non-canonical effects of antibiotics and other treatments to accurately define and improve their clinical impact.


Asunto(s)
Antibacterianos/farmacología , Bacterias , Fibrosis Quística/microbiología , Microbiota/efectos de los fármacos , Esputo/microbiología , Tobramicina/farmacología , Administración por Inhalación , Adolescente , Adulto , Anciano , Antibacterianos/uso terapéutico , Bacterias/genética , Bacterias/aislamiento & purificación , Infecciones Bacterianas/prevención & control , Niño , Fibrosis Quística/fisiopatología , Volumen Espiratorio Forzado , Humanos , Quimioterapia de Mantención , Metagenoma/efectos de los fármacos , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Factores de Tiempo , Tobramicina/uso terapéutico , Adulto Joven
5.
Cell Host Microbe ; 27(6): 851-853, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32526179

RESUMEN

In this issue of Cell Host and Microbe, Lee et al. define the glycan binding specificity of a variant of typhoid toxin produced by a non-typhoidal Salmonellae serotype. The authors elegantly demonstrate that tissue and host specificity of the toxin are related to specific glycan binding characteristics of the toxin.


Asunto(s)
Fiebre Tifoidea , Humanos , Lectinas , Especificidad de Órganos , Polisacáridos , Salmonella , Virulencia
6.
mBio ; 11(1)2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32047135

RESUMEN

The Salmonella enterica subsp. enterica serovar Typhimurium PhoPQ two-component system is activated within the intracellular phagosome environment, where it promotes remodeling of the outer membrane and resistance to innate immune antimicrobial peptides. Maintenance of the PhoPQ-regulated outer membrane barrier requires PbgA, an inner membrane protein with a transmembrane domain essential for growth, and a periplasmic domain required for PhoPQ-activated increases in outer membrane cardiolipin. Here, we report the crystal structure of cardiolipin-bound PbgA, adopting a novel transmembrane fold that features a cardiolipin binding site in close proximity to a long and deep cleft spanning the lipid bilayer. The end of the cleft extends into the periplasmic domain of the protein, which is structurally coupled to the transmembrane domain via a functionally critical C-terminal helix. In conjunction with a conserved putative catalytic dyad situated at the middle of the cleft, our structural and mutational analyses suggest that PbgA is a multifunction membrane protein that mediates cardiolipin transport, a function essential for growth, and perhaps catalysis of an unknown enzymatic reaction.IMPORTANCE Gram-negative bacteria cause many types of infections and have become increasingly resistant to available antibiotic drugs. The outer membrane serves as an important barrier that protects bacteria against antibiotics and other toxic compounds. This outer membrane barrier function is regulated when bacteria are in host environments, and the protein PbgA contributes significantly to this increased barrier function by transporting cardiolipin to the outer membrane. We determined the crystal structure of PbgA in complex with cardiolipin and propose a model for its function. Knowledge of the mechanisms of outer membrane assembly and integrity can greatly contribute to the development of new and effective antibiotics, and this structural information may be useful in this regard.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Cardiolipinas/química , Salmonella typhimurium/química , Animales , Cardiolipinas/genética , Membrana Celular/química , Células Cultivadas , Cristalización , Femenino , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Salmonella typhimurium/genética
7.
PLoS Pathog ; 16(1): e1008251, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961914

RESUMEN

Patients with cystic fibrosis (CF) have altered fecal microbiomes compared to those of healthy controls. The magnitude of this dysbiosis correlates with measures of CF gastrointestinal (GI) disease, including GI inflammation and nutrient malabsorption. However, whether this dysbiosis is caused by mutations in the CFTR gene, the underlying defect in CF, or whether CF-associated dysbiosis augments GI disease was not clear. To test the relationships between CFTR dysfunction, microbes, and intestinal health, we established a germ-free (GF) CF mouse model and demonstrated that CFTR gene mutations are sufficient to alter the GI microbiome. Furthermore, flow cytometric analysis demonstrated that colonized CF mice have increased mesenteric lymph node and spleen TH17+ cells compared with non-CF mice, suggesting that CFTR defects alter adaptive immune responses. Our findings demonstrate that CFTR mutations modulate both the host adaptive immune response and the intestinal microbiome.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/microbiología , Disbiosis/microbiología , Microbioma Gastrointestinal , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Fibrosis Quística/genética , Fibrosis Quística/inmunología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , Modelos Animales de Enfermedad , Disbiosis/genética , Disbiosis/inmunología , Femenino , Humanos , Intestinos/inmunología , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación
8.
Nat Med ; 26(2): 215-221, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31959989

RESUMEN

Most infants with cystic fibrosis (CF) have pancreatic exocrine insufficiency that results in nutrient malabsorption and requires oral pancreatic enzyme replacement. Newborn screening for CF has enabled earlier diagnosis, nutritional intervention and enzyme replacement for these infants, allowing most infants with CF to achieve their weight goals by 12 months of age1. Nevertheless, most infants with CF continue to have poor linear growth during their first year of life1. Although this early linear growth failure is associated with worse long-term respiratory function and survival2,3, the determinants of body length in infants with CF have not been defined. Several characteristics of the CF gastrointestinal (GI) tract, including inflammation, maldigestion and malabsorption, may promote intestinal dysbiosis4,5. As GI microbiome activities are known to affect endocrine functions6,7, the intestinal microbiome of infants with CF may also impact growth. We identified an early, progressive fecal dysbiosis that distinguished infants with CF and low length from infants with CF and normal length. This dysbiosis included altered abundances of taxa that perform functions that are important for GI health, nutrient harvest and growth hormone signaling, including decreased abundance of Bacteroidetes and increased abundance of Proteobacteria. Thus, the GI microbiota represent a potential therapeutic target for the correction of low linear growth in infants with CF.


Asunto(s)
Fibrosis Quística/microbiología , Disbiosis/microbiología , Heces/microbiología , Trastornos del Crecimiento/etiología , Tamaño Corporal , Estudios de Casos y Controles , Femenino , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Humanos , Lactante , Recién Nacido , Inflamación , Estudios Longitudinales , Masculino , Análisis Multivariante , Mutación , Tamizaje Neonatal , Estudios Prospectivos , Análisis de Secuencia de ADN
9.
Curr Biol ; 29(20): R1086-R1088, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31639355

RESUMEN

Many intracellular pathogens reside in host-membrane-encased vacuoles, but the mechanism initiating xenophagic targeting of these vacuoles was unknown. A recent study identifies the host vacuolar-ATPase as essential to xenophagic clearance and the Salmonellae effector SopF that inhibits bacterial clearance by its ADP-ribosylation.


Asunto(s)
ATPasas de Translocación de Protón Vacuolares , Vacuolas , Bacterias , Citoplasma , Macroautofagia
10.
Cell Microbiol ; 21(11): e13116, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31509644

RESUMEN

Salmonellae are important enteric pathogens that cause gastroenteritis and systemic illnesses. Macrophages are important components of both the innate and acquired immune system, acting as phagocytes with significant antimicrobial killing activities that present antigen to the adaptive immune system. Macrophages can also be cultured from a variety of sites as primary cells, and the study of the survival and interactions of Salmonellae with these cells is a very early model of infection and cellular microbiology. This review traces the history of discoveries made using Salmonellae infection of macrophages and addresses the possibility of future research in this area, in particular with regards to understanding the complexity of individual bacteria and macrophage cell variability and how such heterogeneity may alter the outcome of infection.


Asunto(s)
Macrófagos/microbiología , Infecciones por Salmonella/metabolismo , Salmonella/metabolismo , Inmunidad Adaptativa , Historia del Siglo XIX , Historia del Siglo XX , Inmunidad Innata , Macrófagos/inmunología , Técnicas Microbiológicas/instrumentación , Técnicas Microbiológicas/métodos , Microbiología/historia , Fagocitosis/inmunología , Salmonella/genética , Salmonella/inmunología , Salmonella/patogenicidad
11.
Cell Rep ; 27(7): 2147-2156.e5, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31091452

RESUMEN

Intracellular Salmonella use a type III secretion system (TTSS) to translocate effector proteins across the phagosome membrane and thus promote vacuole membrane tubulation, resulting in intracellular survival. This work demonstrates that the effector SseJ binds the eukaryotic lipid transporter oxysterol binding protein 1 (OSBP1). SseJ directs OSBP1 to the endosomal compartment in a manner dependent on the TTSS located on Salmonella pathogenicity island 2 (SPI2). OSBP1 localization is mediated by both SseJ and another OSBP1-binding SPI2 translocated effector, the deubiquitinase SseL. Deletion of both SseJ and SseL reduced vacuolar integrity with increased bacteria released into the eukaryotic cytoplasm of epithelial cells, indicating that their combined activities are necessary for vacuole membrane stability. Cells knocked down for OSBP1 or deleted for the OSBP1-binding proteins VAPA/B also demonstrate loss of vacuole integrity, consistent with the hypothesis that OSBP1 recruitment is required for SPI2-mediated alterations that promote vacuolar integrity of salmonellae.


Asunto(s)
Membranas Intracelulares/metabolismo , Fagosomas/metabolismo , Receptores de Esteroides/metabolismo , Salmonella typhimurium/metabolismo , Vacuolas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células HeLa , Humanos , Membranas Intracelulares/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fagosomas/genética , Fagosomas/microbiología , Receptores de Esteroides/genética , Salmonella typhimurium/genética , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Vacuolas/genética , Vacuolas/microbiología
12.
Proc Natl Acad Sci U S A ; 116(13): 6335-6340, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30862737

RESUMEN

Salmonella Typhimurium can invade and survive within macrophages where the bacterium encounters a range of host environmental conditions. Like many bacteria, S. Typhimurium rapidly responds to changing environments by the use of second messengers such as cyclic di-GMP (c-di-GMP). Here, we generate a fluorescent biosensor to measure c-di-GMP concentrations in thousands of individual bacteria during macrophage infection and to define the sensor enzymes important to c-di-GMP regulation. Three sensor phosphodiesterases were identified as critical to maintaining low c-di-GMP concentrations generated after initial phagocytosis by macrophages. Maintenance of low c-di-GMP concentrations by these phosphodiesterases was required to promote survival within macrophages and virulence for mice. Attenuation of S Typhimurium virulence was due to overproduction of c-di-GMP-regulated cellulose, as deletion of the cellulose synthase machinery restored virulence to a strain lacking enzymatic activity of the three phosphodiesterases. We further identified that the cellulose-mediated reduction in survival was constrained to a slow-replicating persister population of S. Typhimurium induced within the macrophage intracellular environment. As utilization of glucose has been shown to be required for S. Typhimurium macrophage survival, one possible hypothesis is that this persister population requires the glucose redirected to the synthesis of cellulose to maintain a slow-replicating, metabolically active state.


Asunto(s)
GMP Cíclico/análogos & derivados , Citoplasma/metabolismo , Citoplasma/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Salmonella typhimurium/patogenicidad , Animales , Técnicas Biosensibles/métodos , Celulosa/metabolismo , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fluorescencia , Glucosiltransferasas , Interacciones Huésped-Patógeno/fisiología , Ratones , Ratones Endogámicos BALB C , Viabilidad Microbiana , Fagocitosis , Hidrolasas Diéster Fosfóricas/metabolismo , Salmonella typhimurium/metabolismo , Virulencia
13.
Cell Rep ; 26(8): 2227-2240.e5, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30784601

RESUMEN

Metagenomic sequencing is a promising approach for identifying and characterizing organisms and their functional characteristics in complex, polymicrobial infections, such as airway infections in people with cystic fibrosis. These analyses are often hampered, however, by overwhelming quantities of human DNA, yielding only a small proportion of microbial reads for analysis. In addition, many abundant microbes in respiratory samples can produce large quantities of extracellular bacterial DNA originating either from biofilms or dead cells. We describe a method for simultaneously depleting DNA from intact human cells and extracellular DNA (human and bacterial) in sputum, using selective lysis of eukaryotic cells and endonuclease digestion. We show that this method increases microbial sequencing depth and, consequently, both the number of taxa detected and coverage of individual genes such as those involved in antibiotic resistance. This finding underscores the substantial impact of DNA from sources other than live bacteria in microbiological analyses of complex, chronic infection specimens.


Asunto(s)
Infecciones Bacterianas/microbiología , Código de Barras del ADN Taxonómico/métodos , Metagenoma , Metagenómica/métodos , Microbiota , Esputo/microbiología , Infecciones Bacterianas/diagnóstico , Humanos , Técnicas de Diagnóstico Molecular/métodos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología
14.
Elife ; 82019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30638443

RESUMEN

The outer membrane (OM) of Gram-negative bacteria serves as a selective permeability barrier that allows entry of essential nutrients while excluding toxic compounds, including antibiotics. The OM is asymmetric and contains an outer leaflet of lipopolysaccharides (LPS) or lipooligosaccharides (LOS) and an inner leaflet of glycerophospholipids (GPL). We screened Acinetobacter baumannii transposon mutants and identified a number of mutants with OM defects, including an ABC transporter system homologous to the Mla system in E. coli. We further show that this opportunistic, antibiotic-resistant pathogen uses this multicomponent protein complex and ATP hydrolysis at the inner membrane to promote GPL export to the OM. The broad conservation of the Mla system in Gram-negative bacteria suggests the system may play a conserved role in OM biogenesis. The importance of the Mla system to Acinetobacter baumannii OM integrity and antibiotic sensitivity suggests that its components may serve as new antimicrobial therapeutic targets.


Asunto(s)
Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Glicerofosfolípidos/metabolismo , Lipopolisacáridos/metabolismo , Acinetobacter baumannii/genética , Adenosina Trifosfato/química , Transporte Biológico , Biología Computacional , Microscopía por Crioelectrón , Elementos Transponibles de ADN , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Genoma Bacteriano , Hidrólisis , Conformación Molecular , Mutagénesis , Mutación , Fenotipo
15.
Mol Cell ; 73(1): 157-165.e5, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30449724

RESUMEN

Efforts to battle antimicrobial resistance (AMR) are generally focused on developing novel antibiotics. However, history shows that resistance arises regardless of the nature or potency of new drugs. Here, we propose and provide evidence for an alternate strategy to resolve this problem: inhibiting evolution. We determined that the DNA translocase Mfd is an "evolvability factor" that promotes mutagenesis and is required for rapid resistance development to all antibiotics tested across highly divergent bacterial species. Importantly, hypermutator alleles that accelerate AMR development did not arise without Mfd, at least during evolution of trimethoprim resistance. We also show that Mfd's role in AMR development depends on its interactions with the RNA polymerase subunit RpoB and the nucleotide excision repair protein UvrA. Our findings suggest that AMR development can be inhibited through inactivation of evolvability factors (potentially with "anti-evolution" drugs)-in particular, Mfd-providing an unexplored route toward battling the AMR crisis.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/genética , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Evolución Molecular , Factores de Transcripción/genética , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Bacterias/crecimiento & desarrollo , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Células CACO-2 , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Diseño de Fármacos , Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Mutagénesis/efectos de los fármacos , Unión Proteica , Especificidad de la Especie , Factores de Tiempo , Factores de Transcripción/metabolismo
16.
Chembiochem ; 20(3): 394-407, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30395379

RESUMEN

The bacterial second messenger cyclic diguanosine monophosphate (c-di-GMP) is a key regulator of cellular motility, the cell cycle, and biofilm formation with its resultant antibiotic tolerance, which can make chronic infections difficult to treat. Therefore, diguanylate cyclases, which regulate the spatiotemporal production of c-di-GMP, might be attractive drug targets for control of biofilm formation that is part of chronic infections. We present a FRET-based biochemical high-throughput screening approach coupled with detailed structure-activity studies to identify synthetic small-molecule modulators of the diguanylate cyclase DgcA from Caulobacter crescentus. We identified a set of seven small molecules that regulate DgcA enzymatic activity in the low-micromolar range. Subsequent structure-activity studies on selected scaffolds revealed a remarkable diversity of modulatory behavior, including slight chemical substitutions that reverse the effects from allosteric enzyme inhibition to activation. The compounds identified represent new chemotypes and are potentially developable into chemical genetic tools for the dissection of c-di-GMP signaling networks and alteration of c-di-GMP-associated phenotypes. In sum, our studies underline the importance of detailed mechanism-of-action studies for inhibitors of c-di-GMP signaling and demonstrate the complex interplay between synthetic small molecules and the regulatory mechanisms that control the activity of diguanylate cyclases.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas de Escherichia coli/antagonistas & inhibidores , Liasas de Fósforo-Oxígeno/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Regulación Alostérica/efectos de los fármacos , Caulobacter crescentus/enzimología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Proteínas de Escherichia coli/metabolismo , Estructura Molecular , Liasas de Fósforo-Oxígeno/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
17.
Sci Signal ; 11(558)2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30482849

RESUMEN

The outer membranes of Gram-negative bacteria and mitochondria contain proteins with a distinct ß-barrel tertiary structure that could function as a molecular pattern recognized by the innate immune system. Here, we report that purified outer membrane proteins (OMPs) from different bacterial and mitochondrial sources triggered the induction of autophagy-related endosomal acidification, LC3B lipidation, and p62 degradation. Furthermore, OMPs reduced the phosphorylation and therefore activation of the multiprotein complex mTORC2 and its substrate Akt in macrophages and epithelial cells. The cell surface receptor SlamF8 and the DNA-protein kinase subunit XRCC6 were required for these OMP-specific responses in macrophages and epithelial cells, respectively. The addition of OMPs to mouse bone marrow-derived macrophages infected with Salmonella Typhimurium facilitated bacterial clearance. These data identify a specific cellular response mediated by bacterial and mitochondrial OMPs that can alter inflammatory responses and influence the killing of pathogens.


Asunto(s)
Autofagia , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Celular/patología , Diana Mecanicista del Complejo 2 de la Rapamicina/antagonistas & inhibidores , Membranas Mitocondriales/patología , Monocitos/patología , Infecciones por Salmonella/patología , Animales , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Membranas Mitocondriales/metabolismo , Monocitos/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/aislamiento & purificación , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-29891596

RESUMEN

Acinetobacter baumannii is a Gram-negative organism that is a cause of hospital-acquired multidrug-resistant (MDR) infections. A. baumannii has a unique cell surface compared to those of many other Gram-negative pathogens in that it can live without lipopolysaccharide (LPS) and it has a high content of cardiolipin in the outer membrane. Therefore, to better understand the cell envelope and mechanisms of MDR A. baumannii, we screened a transposon library for mutants with defective permeability barrier function, defined as a deficiency in the ability to exclude the phosphatase chromogenic substrate 5-bromo-4-chloro-3-indolylphosphate (XP). We identified multiple mutants with mutations in the ABUW_0982 gene, predicted to encode a permease broadly present in A. baumannii isolates with increased susceptibility to the ribosome-targeting antibiotic chloramphenicol (CHL). Moreover, compared to other known CHL resistance genes, such as chloramphenicol acyltransferase genes, we found that ABUW_0982 is the primary determinant of intrinsic CHL resistance in A. baumannii strain 5075 (Ab5075), an important isolate responsible for severe MDR infections in humans. Finally, studies measuring the efflux of chloramphenicol and expression of ABUW_0982 in CHL-susceptible Escherichia coli support the conclusion that ABUW_0982 encodes a single-component efflux protein with specificity for small, hydrophobic molecules, including CHL.


Asunto(s)
Acinetobacter baumannii/genética , Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Resistencia al Cloranfenicol/genética , Cloranfenicol/metabolismo , Farmacorresistencia Bacteriana Múltiple/genética , Proteínas de Transporte de Membrana/genética , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/aislamiento & purificación , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Transporte Biológico , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cloranfenicol/farmacología , Compuestos Cromogénicos/química , Clonación Molecular , Elementos Transponibles de ADN , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Biblioteca de Genes , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Indoles/química , Proteínas de Transporte de Membrana/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
PLoS Negl Trop Dis ; 12(1): e0006156, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29329299

RESUMEN

Non-typhoidal Salmonella (NTS) is a leading cause of bloodstream infections in Africa, but the various contributions of host susceptibility versus unique pathogen virulence factors are unclear. We used data from a population-based surveillance platform (population ~25,000) between 2007-2014 and NTS genome-sequencing to compare host and pathogen-specific factors between individuals presenting with NTS bacteremia and those presenting with NTS diarrhea. Salmonella Typhimurium ST313 and Salmonella Enteritidis ST11 were the most common isolates. Multi-drug resistant strains of NTS were more commonly isolated from patients presenting with NTS bacteremia compared to NTS diarrhea. This relationship was observed in patients under age five [aOR = 15.16, 95% CI (2.84-81.05), P = 0.001], in patients five years and older, [aOR = 6.70 95% CI (2.25-19.89), P = 0.001], in HIV-uninfected patients, [aOR = 21.61, 95% CI (2.53-185.0), P = 0.005], and in patients infected with Salmonella serogroup B [aOR = 5.96, 95% CI (2.28-15.56), P < 0.001] and serogroup D [aOR = 14.15, 95% CI (1.10-182.7), P = 0.042]. Thus, multi-drug-resistant NTS was strongly associated with bacteremia compared to diarrhea among children and adults. This association was seen in HIV-uninfected individuals infected with either S. Typhimurium or S. Enteritidis. Risk of developing bacteremia from NTS infection may be driven by virulence properties of the Salmonella pathogen.


Asunto(s)
Bacteriemia/epidemiología , Farmacorresistencia Bacteriana Múltiple , Infecciones por Salmonella/epidemiología , Salmonella enterica/efectos de los fármacos , Salmonella enterica/aislamiento & purificación , Adolescente , Adulto , Anciano , Bacteriemia/microbiología , Niño , Preescolar , ADN Bacteriano/química , ADN Bacteriano/genética , Diarrea/epidemiología , Diarrea/microbiología , Femenino , Humanos , Lactante , Kenia/epidemiología , Masculino , Persona de Mediana Edad , Infecciones por Salmonella/microbiología , Salmonella enterica/genética , Análisis de Secuencia de ADN , Adulto Joven
20.
Proc Natl Acad Sci U S A ; 115(7): 1605-1610, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29378945

RESUMEN

The mature human gut microbiota is established during the first years of life, and altered intestinal microbiomes have been associated with several human health disorders. Escherichia coli usually represents less than 1% of the human intestinal microbiome, whereas in cystic fibrosis (CF), greater than 50% relative abundance is common and correlates with intestinal inflammation and fecal fat malabsorption. Despite the proliferation of E. coli and other Proteobacteria in conditions involving chronic gastrointestinal tract inflammation, little is known about adaptation of specific characteristics associated with microbiota clonal expansion. We show that E. coli isolated from fecal samples of young children with CF has adapted to growth on glycerol, a major component of fecal fat. E. coli isolates from different CF patients demonstrate an increased growth rate in the presence of glycerol compared with E. coli from healthy controls, and unrelated CF E. coli strains have independently acquired this growth trait. Furthermore, CF and control E. coli isolates have differential gene expression when grown in minimal media with glycerol as the sole carbon source. While CF isolates display a growth-promoting transcriptional profile, control isolates engage stress and stationary-phase programs, which likely results in slower growth rates. Our results indicate that there is selection of unique characteristics within the microbiome of individuals with CF, which could contribute to individual disease outcomes.


Asunto(s)
Fibrosis Quística/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/patogenicidad , Heces/microbiología , Microbioma Gastrointestinal/genética , Intestinos/microbiología , Estudios de Casos y Controles , Preescolar , Fibrosis Quística/genética , Fibrosis Quística/patología , Grasas de la Dieta/metabolismo , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/patología , Redes Reguladoras de Genes , Glicerol/metabolismo , Humanos , Lactante , Fosfolípidos/metabolismo , Filogenia , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA