Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Cell ; 172(3): 618-628.e13, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29307492

RESUMEN

Peptides have great potential to combat antibiotic resistance. While many platforms can screen peptides for their ability to bind to target cells, there are virtually no platforms that directly assess the functionality of peptides. This limitation is exacerbated when identifying antimicrobial peptides because the phenotype, death, selects against itself and has caused a scientific bottleneck that confines research to a few naturally occurring classes of antimicrobial peptides. We have used this seeming dissonance to develop Surface Localized Antimicrobial Display (SLAY), a platform that allows screening of unlimited numbers of peptides of any length, composition, and structure in a single tube for antimicrobial activity. Using SLAY, we screened ∼800,000 random peptide sequences for antimicrobial function and identified thousands of active sequences, dramatically increasing the number of known antimicrobial sequences. SLAY hits present with different potential mechanisms of peptide action and access to areas of antimicrobial physicochemical space beyond what nature has evolved. VIDEO ABSTRACT.


Asunto(s)
Antibacterianos/farmacología , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Biblioteca de Péptidos , Animales , Antibacterianos/química , Escherichia coli , Ratones
2.
Cell ; 149(2): 358-70, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22500802

RESUMEN

The function of the Vibrio 7(th) pandemic island-1 (VSP-1) in cholera pathogenesis has remained obscure. Utilizing chromatin immunoprecipitation sequencing and RNA sequencing to map the regulon of the master virulence regulator ToxT, we identify a TCP island-encoded small RNA that reduces the expression of a previously unrecognized VSP-1-encoded transcription factor termed VspR. VspR modulates the expression of several VSP-1 genes including one that encodes a novel class of di-nucleotide cyclase (DncV), which preferentially synthesizes a previously undescribed hybrid cyclic AMP-GMP molecule. We show that DncV is required for efficient intestinal colonization and downregulates V. cholerae chemotaxis, a phenotype previously associated with hyperinfectivity. This pathway couples the actions of previously disparate genomic islands, defines VSP-1 as a pathogenicity island in V. cholerae, and implicates its occurrence in 7(th) pandemic strains as a benefit for host adaptation through the production of a regulatory cyclic di-nucleotide.


Asunto(s)
AMP Cíclico/biosíntesis , Nucleótidos Cíclicos/metabolismo , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidad , Animales , Proteínas Bacterianas , Secuencia de Bases , Regulación Viral de la Expresión Génica , Islas Genómicas , Humanos , Intestinos/microbiología , Redes y Vías Metabólicas , Ratones , Datos de Secuencia Molecular , Liasas de Fósforo-Oxígeno , ARN no Traducido/metabolismo , ARN Viral/metabolismo , Alineación de Secuencia , Factores de Transcripción , Vibrio cholerae/genética , Virulencia
3.
Appl Environ Microbiol ; 89(5): e0033523, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37078870

RESUMEN

Small proteins perform a diverse array of functions, from microbial competition, to endocrine signaling, to building biomaterials. Microbial systems that can produce recombinant small proteins enable discovery of new effectors, exploration of sequence activity relationships, and have the potential for in vivo delivery. However, we lack simple systems for controlling small-protein secretion from Gram-negative bacteria. Microcins are small-protein antibiotics secreted by Gram-negative bacteria that inhibit the growth of neighboring microbes. They are exported from the cytosol to the environment in a one-step process through a specific class of type I secretion systems (T1SSs). However, relatively little is known about substrate requirements for small proteins exported through microcin T1SSs. Here, we investigate the prototypic microcin V T1SS from Escherichia coli and show that it can export a remarkably wide range of natural and synthetic small proteins. We demonstrate that secretion is largely independent of the cargo protein's chemical properties and appears to be constrained only by protein length. We show that a varied range of bioactive sequences, including an antibacterial protein, a microbial signaling factor, a protease inhibitor, and a human hormone, can all be secreted and elicit their intended biological effect. Secretion through this system is not limited to E. coli, and we demonstrate its function in additional Gram-negative species that can inhabit the gastrointestinal tract. Our findings uncover the highly promiscuous nature of small-protein export through the microcin V T1SS, which has implications for native-cargo capacity and the use of this system in Gram-negative bacteria for small-protein research and delivery. IMPORTANCE Type I secretion systems for microcin export in Gram-negative bacteria transport small antibacterial proteins from the cytoplasm to the extracellular environment in a single step. In nature, each secretion system is generally paired with a specific small protein. We know little about the export capacity of these transporters and how cargo sequence influences secretion. Here, we investigate the microcin V type I system. Remarkably, our studies show that this system can export small proteins of diverse sequence composition and is only limited by protein length. Furthermore, we demonstrate that a wide range of bioactive small proteins can be secreted and that this system can be used in Gram-negative species that colonize the gastrointestinal tract. These findings expand our understanding of secretion through type I systems and their potential uses in a variety of small-protein applications.


Asunto(s)
Escherichia coli , Sistemas de Secreción Tipo I , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Bacterias Gramnegativas/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(44): 27620-27626, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33087568

RESUMEN

The extracellular polysaccharide capsule of Klebsiella pneumoniae resists penetration by antimicrobials and protects the bacteria from the innate immune system. Host antimicrobial peptides are inactivated by the capsule as it impedes their penetration to the bacterial membrane. While the capsule sequesters most peptides, a few antimicrobial peptides have been identified that retain activity against encapsulated K. pneumoniae, suggesting that this bacterial defense can be overcome. However, it is unclear what factors allow peptides to avoid capsule inhibition. To address this, we created a peptide analog with strong antimicrobial activity toward several K. pneumoniae strains from a previously inactive peptide. We characterized the effects of these two peptides on K. pneumoniae, along with their physical interactions with K. pneumoniae capsule. Both peptides disrupted bacterial cell membranes, but only the active peptide displayed this activity against capsulated K. pneumoniae Unexpectedly, the active peptide showed no decrease in capsule binding, but did lose secondary structure in a capsule-dependent fashion compared with the inactive parent peptide. We found that these characteristics are associated with capsule-peptide aggregation, leading to disruption of the K. pneumoniae capsule. Our findings reveal a potential mechanism for disrupting the protective barrier that K. pneumoniae uses to avoid the immune system and last-resort antibiotics.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Cápsulas Bacterianas/efectos de los fármacos , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Animales , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Cápsulas Bacterianas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple , Femenino , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/citología , Ratones , Pruebas de Sensibilidad Microbiana , Polisacáridos Bacterianos/metabolismo
5.
Appl Environ Microbiol ; 88(23): e0148622, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36394322

RESUMEN

Microcins are a class of antimicrobial peptides produced by certain Gram-negative bacterial species to kill or inhibit the growth of competing bacteria. Only 10 unique, experimentally validated class II microcins have been identified, and the majority of these come from Escherichia coli. Although the current representation of microcins is sparse, they exhibit a diverse array of molecular functionalities, uptake mechanisms, and target specificities. This broad diversity from such a small representation suggests that microcins may have untapped potential for bioprospecting peptide antibiotics from genomic data sets. We used a systematic bioinformatics approach to search for verified and novel class II microcins in E. coli and other species within its family, Enterobacteriaceae. Nearly one-quarter of the E. coli genome assemblies contained one or more microcins, where the prevalence of hits to specific microcins varied by isolate phylogroup. E. coli isolates from human extraintestinal and poultry meat sources were enriched for microcins, while those from freshwater were depleted. Putative microcins were found in various abundances across all five distinct phylogenetic lineages of Enterobacteriaceae, with a particularly high prevalence in the "Klebsiella" clade. Representative genome assemblies from species across the Enterobacterales order, as well as a few outgroup species, also contained putative microcin sequences. This study suggests that microcins have a complicated evolutionary history, spanning far beyond our limited knowledge of the currently validated microcins. Efforts to functionally characterize these newly identified microcins have great potential to open a new field of peptide antibiotics and microbiome modulators and elucidate the ways in which bacteria compete with each other. IMPORTANCE Class II microcins are small bacteriocins produced by strains of Gram-negative bacteria in the Enterobacteriaceae. They are generally understood to play a role in interbacterial competition, although direct evidence of this is limited, and they could prove informative in developing new peptide antibiotics. However, few examples of verified class II microcins exist, and novel microcins are difficult to identify due to their sequence diversity, making it complicated to study them as a group. Here, we overcome this limitation by developing a bioinformatics pipeline to detect microcins in silico. Using this pipeline, we demonstrate that both verified and novel class II microcins are widespread within and outside the Enterobacteriaceae, which has not been systematically shown previously. The observed prevalence of class II microcins suggests that they are ecologically important, and the elucidation of novel microcins provides a resource that can be used to expand our knowledge of the structure and function of microcins as antibacterials.


Asunto(s)
Bacteriocinas , Escherichia coli , Antibacterianos/farmacología , Antibacterianos/química , Bacterias , Bacteriocinas/genética , Bacteriocinas/farmacología , Bacteriocinas/química , Enterobacteriaceae , Escherichia coli/genética , Péptidos/genética , Filogenia
6.
Antimicrob Agents Chemother ; 65(10): e0044321, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34280019

RESUMEN

Otilonium bromide is a poorly absorbed oral medication used to control irritable bowel syndrome. It is thought to act as a muscle relaxant in the intestine. Here, we show that otilonium bromide has broad-spectrum antibacterial and antifungal activity, including against multidrug-resistant strains. Our results suggest otilonium bromide acts on enteric pathogens and may offer a new scaffold for poorly absorbed intestinal antimicrobial therapy.


Asunto(s)
Síndrome del Colon Irritable , Humanos , Intestinos , Síndrome del Colon Irritable/tratamiento farmacológico , Compuestos de Amonio Cuaternario
7.
Proc Natl Acad Sci U S A ; 115(38): E8968-E8976, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30126994

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) is a global diarrheal pathogen that utilizes adhesins and secreted enterotoxins to cause disease in mammalian hosts. Decades of research on virulence factor regulation in ETEC has revealed a variety of environmental factors that influence gene expression, including bile, pH, bicarbonate, osmolarity, and glucose. However, other hallmarks of the intestinal tract, such as low oxygen availability, have not been examined. Further, determining how ETEC integrates these signals in the complex host environment is challenging. To address this, we characterized ETEC's response to the human host using samples from a controlled human infection model. We found ETEC senses environmental oxygen to globally influence virulence factor expression via the oxygen-sensitive transcriptional regulator fumarate and nitrate reduction (FNR) regulator. In vitro anaerobic growth replicates the in vivo virulence factor expression profile, and deletion of fnr in ETEC strain H10407 results in a significant increase in expression of all classical virulence factors, including the colonization factor antigen I (CFA/I) adhesin operon and both heat-stable and heat-labile enterotoxins. These data depict a model of ETEC infection where FNR activity can globally influence virulence gene expression, and therefore proximity to the oxygenated zone bordering intestinal epithelial cells likely influences ETEC virulence gene expression in vivo. Outside of the host, ETEC biofilms are associated with seasonal ETEC epidemics, and we find FNR is a regulator of biofilm production. Together these data suggest FNR-dependent oxygen sensing in ETEC has implications for human infection inside and outside of the host.


Asunto(s)
Escherichia coli Enterotoxigénica/patogenicidad , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Proteínas Hierro-Azufre/genética , Adulto , Biopelículas , Diarrea/epidemiología , Diarrea/microbiología , Diarrea/prevención & control , Células Epiteliales/microbiología , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Proteínas de Escherichia coli/metabolismo , Vacunas contra Escherichia coli/administración & dosificación , Femenino , Voluntarios Sanos , Humanos , Intestinos/citología , Intestinos/microbiología , Proteínas Hierro-Azufre/metabolismo , Masculino , Persona de Mediana Edad , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Adulto Joven
8.
Inorganica Chim Acta ; 5172021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33568836

RESUMEN

Three new coinage metal carbene complexes of silver and gold were synthesized from a thiamine inspired proligand. The compounds were characterized by HRMS, NMR spectroscopy (1H, 19F, 31P and 13C), FT-IR and elemental analysis. The coordination environment around the metal centers was correlated to the diffusion coefficients obtained from DOSY-NMR experiments and was in agreement with the nuclearity observed in the solid-state by single crystal X-ray crystallography. The silver and gold carbene compounds were subjected to MIC studies against a panel of pathogenic bacteria, including multidrug resistant strains, with the gold carbene derivative showing the most potent antimicrobial activity against Gram-positive methicillin resistant Staphylococcus aureus (MRSA).

9.
Proc Natl Acad Sci U S A ; 113(41): E6228-E6237, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27681618

RESUMEN

The Gram-negative bacterial outer membrane fortifies the cell against environmental toxins including antibiotics. Unique glycolipids called lipopolysaccharide/lipooligosaccharide (LPS/LOS) are enriched in the cell-surface monolayer of the outer membrane and promote antimicrobial resistance. Colistin, which targets the lipid A domain of LPS/LOS to lyse the cell, is the last-line treatment for multidrug-resistant Gram-negative infections. Lipid A is essential for the survival of most Gram-negative bacteria, but colistin-resistant Acinetobacter baumannii lacking lipid A were isolated after colistin exposure. Previously, strain ATCC 19606 was the only A. baumannii strain demonstrated to subsist without lipid A. Here, we show that other A. baumannii strains can also survive without lipid A, but some cannot, affording a unique model to study endotoxin essentiality. We assessed the capacity of 15 clinical A. baumannii isolates including 9 recent clinical isolates to develop colistin resistance through inactivation of the lipid A biosynthetic pathway, the products of which assemble the LOS precursor. Our investigation determined that expression of the well-conserved penicillin-binding protein (PBP) 1A, prevented LOS-deficient colony isolation. The glycosyltransferase activity of PBP1A, which aids in the polymerization of the peptidoglycan cell wall, was lethal to LOS-deficient A. baumannii Global transcriptomic analysis of a PBP1A-deficient mutant and four LOS-deficient A. baumannii strains showed a concomitant increase in transcription of lipoproteins and their transporters. Examination of the LOS-deficient A. baumannii cell surface demonstrated that specific lipoproteins were overexpressed and decorated the cell surface, potentially compensating for LOS removal. This work expands our knowledge of lipid A essentiality and elucidates a drug resistance mechanism.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Colistina/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Lipopolisacáridos/deficiencia , Proteínas de Unión a las Penicilinas/metabolismo , Acinetobacter baumannii/genética , Membrana Celular/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Genes Bacterianos , Lipopolisacáridos/biosíntesis , Pruebas de Sensibilidad Microbiana , Análisis de Secuencia de ARN , Transcripción Genética/efectos de los fármacos
10.
J Am Chem Soc ; 140(10): 3768-3774, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29466660

RESUMEN

Peptide oligomers offer versatile scaffolds for the formation of potent antimicrobial agents due to their high sequence versatility, inherent biocompatibility, and chemical tunability. Though many methods exist for the formation of peptide-based macrocycles (MCs), increasingly pervasive in commercial antimicrobial therapeutics, the introduction of multiple looped structures into a single peptide oligomer remains a significant challenge. Herein, we report the utilization of dynamic hydrazone condensation for the versatile formation of single-, double-, and triple-loop peptide MCs using simple dialdehyde or dihydrazide small-molecule cross-linkers, as confirmed by MALDI-TOF MS, HPLC, and SDS-PAGE. Furthermore, incorporation of aldehyde-containing side chains onto peptides synthesized from hydrazide C-terminal resins resulted in tunable peptide MC assemblies formed directly upon resin cleavage post solid-phase peptide synthesis. Both of these types of dynamic covalent assemblies produced significant enhancements to overall antimicrobial properties when introduced into a known antimicrobial peptide, buforin II, when compared to the original unassembled sequence.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Péptidos/química , Péptidos/farmacología , Secuencia de Aminoácidos , Antibacterianos/síntesis química , Bacterias/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Técnicas de Química Sintética/métodos , Humanos , Compuestos Macrocíclicos/síntesis química , Péptidos/síntesis química , Proteínas/farmacología
11.
PLoS Pathog ; 12(4): e1005570, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27070545

RESUMEN

The virulence regulator ToxR initiates and coordinates gene expression needed by Vibrio cholerae to colonize the small intestine and cause disease. Despite its prominence in V. cholerae virulence, our understanding of the direct ToxR regulon is limited to four genes: toxT, ompT, ompU and ctxA. Here, we determine ToxR's genome-wide DNA-binding profile and demonstrate that ToxR is a global regulator of both progenitor genome-encoded genes and horizontally acquired islands that encode V. cholerae's major virulence factors and define pandemic lineages. We show that ToxR shares more than a third of its regulon with the histone-like nucleoid structuring protein H-NS, and antagonizes H-NS binding at shared binding locations. Importantly, we demonstrate that this regulatory interaction is the critical function of ToxR in V. cholerae colonization and biofilm formation. In the absence of H-NS, ToxR is no longer required for V. cholerae to colonize the infant mouse intestine or for robust biofilm formation. We further illustrate a dramatic difference in regulatory scope between ToxR and other prominent virulence regulators, despite similar predicted requirements for DNA binding. Our results suggest that factors in addition to primary DNA structure influence the ability of ToxR to recognize its target promoters.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Unión al ADN/genética , Regulación Bacteriana de la Expresión Génica/genética , Factores de Transcripción/genética , Vibrio cholerae/patogenicidad , Virulencia/genética , Animales , Secuencia de Bases , Northern Blotting , Cólera/genética , Inmunoprecipitación de Cromatina , Transferencia de Gen Horizontal , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Virulencia/genética
12.
Artículo en Inglés | MEDLINE | ID: mdl-28193670

RESUMEN

Enterococcus faecium has emerged as a major opportunistic pathogen for 2 decades with the spread of hospital-adapted multidrug-resistant clones. As members of the intestinal microbiota, they are subjected to numerous bacterial stresses, including antibiotics at subinhibitory concentrations (SICs). Since fluoroquinolones are extensively prescribed, SICs are very likely to occur in vivo, with potential effects on bacterial metabolism with subsequent modulation of opportunistic traits. The aim of this study was to evaluate globally the impact of SICs of ciprofloxacin on antimicrobial resistance and pathogenicity of E. faecium Transcriptomic analysis was performed by RNA sequencing (RNA-seq) (HiSeq 2500; Illumina) using the vanB-positive reference strain E. faecium Aus0004 in the absence or presence of ciprofloxacin SIC (0.38 mg/liter, i.e., 1/8 of the MIC). Several genetic and phenotypic tests were used for validation. In the presence of ciprofloxacin SIC, 196 genes were significantly induced, whereas 286 genes were significantly repressed, meaning that 16.8% of the E. faecium genome was altered. Among upregulated genes, EFAU004_02294 (fold change, 14.3) encoded a protein (Qnr of E. faecium [EfmQnr]) homologue of Qnr proteins involved in quinolone resistance in Gram-negative bacilli. Its implication in intrinsic and adaptive fluoroquinolone (FQ) resistance in E. faecium was experimentally ascertained. Moreover, EFAU004_02292, coding for the collagen adhesin Acm, was also induced by the SIC of ciprofloxacin (fold change, 8.2), and higher adhesion capabilities were demonstrated phenotypically. Both EfmQnr and Acm determinants may play an important role in the transition from a commensal to a pathogenic state of E. faecium that resides in the gut of patients receiving fluoroquinolone therapy.


Asunto(s)
Adhesinas Bacterianas/genética , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Ciprofloxacina/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Enterococcus faecium/efectos de los fármacos , Colágeno/metabolismo , Farmacorresistencia Bacteriana/genética , Enterococcus faecium/genética , Genoma Bacteriano/genética , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Quinolonas/farmacología , Transcriptoma/genética
13.
Biochem Biophys Res Commun ; 484(3): 612-617, 2017 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-28153719

RESUMEN

The product of the human C21orf57 (huYBEY) gene is predicted to be a homologue of the highly conserved YbeY proteins found in nearly all bacteria. We show that, like its bacterial and chloroplast counterparts, the HuYbeY protein is an RNase and that it retains sufficient function in common with bacterial YbeY proteins to partially suppress numerous aspects of the complex phenotype of an Escherichia coli ΔybeY mutant. Expression of HuYbeY in Saccharomyces cerevisiae, which lacks a YbeY homologue, results in a severe growth phenotype. This observation suggests that the function of HuYbeY in human cells is likely regulated through specific interactions with partner proteins similarly to the way YbeY is regulated in bacteria.


Asunto(s)
Cloroplastos/química , Cloroplastos/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Evolución Molecular , Metaloproteínas/química , Metaloproteínas/genética , Ribonucleasas/química , Ribonucleasas/genética , Homología de Secuencia de Aminoácido , Secuencia de Aminoácidos , Secuencia de Bases , Secuencia Conservada/genética , Datos de Secuencia Molecular
14.
Mol Cell ; 36(5): 845-60, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005847

RESUMEN

Hydroxyurea (HU) specifically inhibits class I ribonucleotide reductase (RNR), depleting dNTP pools and leading to replication fork arrest. Although HU inhibition of RNR is well recognized, the mechanism by which it leads to cell death remains unknown. To investigate the mechanism of HU-induced cell death, we used a systems-level approach to determine the genomic and physiological responses of E. coli to HU treatment. Our results suggest a model by which HU treatment rapidly induces a set of protective responses to manage genomic instability. Continued HU stress activates iron uptake and toxins MazF and RelE, whose activity causes the synthesis of incompletely translated proteins and stimulation of envelope stress responses. These effects alter the properties of one of the cell's terminal cytochrome oxidases, causing an increase in superoxide production. The increased superoxide production, together with the increased iron uptake, fuels the formation of hydroxyl radicals that contribute to HU-induced cell death.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Escherichia coli/efectos de los fármacos , Hidroxiurea/farmacología , Toxinas Bacterianas/metabolismo , Membrana Celular/efectos de los fármacos , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Endorribonucleasas/metabolismo , Endorribonucleasas/fisiología , Escherichia coli/citología , Escherichia coli/fisiología , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiología , Genoma Bacteriano , Modelos Biológicos , Superóxidos/metabolismo , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
15.
PLoS Pathog ; 10(6): e1004175, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24901994

RESUMEN

YbeY, a highly conserved protein, is an RNase in E. coli and plays key roles in both processing of the critical 3' end of 16 S rRNA and in 70 S ribosome quality control under stress. These central roles account for YbeY's inclusion in the postulated minimal bacterial genome. However, YbeY is not essential in E. coli although loss of ybeY severely sensitizes it to multiple physiological stresses. Here, we show that YbeY is an essential endoribonuclease in Vibrio cholerae and is crucial for virulence, stress regulation, RNA processing and ribosome quality control, and is part of a core set of RNases essential in most representative pathogens. To understand its function, we analyzed the rRNA and ribosome profiles of a V. cholerae strain partially depleted for YbeY and other RNase mutants associated with 16 S rRNA processing; our results demonstrate that YbeY is also crucial for 16 S rRNA 3' end maturation in V. cholerae and that its depletion impedes subunit assembly into 70 S ribosomes. YbeY's importance to V. cholerae pathogenesis was demonstrated by the complete loss of mice colonization and biofilm formation, reduced cholera toxin production, and altered expression levels of virulence-associated small RNAs of a V. cholerae strain partially depleted for YbeY. Notably, the ybeY genes of several distantly related pathogens can fully complement an E. coli ΔybeY strain under various stress conditions, demonstrating the high conservation of YbeY's activity in stress regulation. Taken together, this work provides the first comprehensive exploration of YbeY's physiological role in a human pathogen, showing its conserved function across species in essential cellular processes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endorribonucleasas/metabolismo , Procesamiento de Término de ARN 3' , ARN Bacteriano/metabolismo , ARN Ribosómico/metabolismo , Estrés Fisiológico , Vibrio cholerae/enzimología , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Cólera/enzimología , Cólera/inmunología , Cólera/metabolismo , Cólera/microbiología , Toxina del Cólera/biosíntesis , Secuencia Conservada , Endorribonucleasas/química , Endorribonucleasas/genética , Regulación Bacteriana de la Expresión Génica , Inmunidad Mucosa , Mucosa Intestinal/crecimiento & desarrollo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Mutación , Filogenia , Vibrio cholerae/inmunología , Vibrio cholerae/patogenicidad , Vibrio cholerae/fisiología , Virulencia , Factores de Virulencia/biosíntesis
16.
Proc Natl Acad Sci U S A ; 108(30): 12467-72, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21750152

RESUMEN

ChIP coupled with next-generation sequencing (ChIP-seq) has revolutionized whole-genome mapping of DNA-binding protein sites. Although ChIP-seq rapidly gained support in eukaryotic systems, it remains underused in the mapping of bacterial transcriptional regulator-binding sites. Using the virulence-required iron-responsive ferric uptake regulator (Fur), we report a simple, broadly applicable ChIP-seq method in the pathogen Vibrio cholerae. Combining our ChIP-seq results with available microarray data, we clarify direct and indirect Fur regulation of known iron-responsive genes. We validate a subset of Fur-binding sites in vivo and show a common motif present in all Fur ChIP-seq peaks that has enhanced binding affinity for purified V. cholerae Fur. Further analysis shows that V. cholerae Fur directly regulates several additional genes associated with Fur-binding sites, expanding the role of this transcription factor into the regulation of ribosome formation, additional transport functions, and unique sRNAs.


Asunto(s)
Proteínas Bacterianas/genética , Genes Bacterianos , Regulón , Proteínas Represoras/genética , Vibrio cholerae/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Inmunoprecipitación de Cromatina , Mapeo Cromosómico , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Regulación Bacteriana de la Expresión Génica , Hierro/metabolismo , ARN Bacteriano/genética , Proteínas Represoras/metabolismo , Vibrio cholerae/metabolismo
17.
ACS Infect Dis ; 10(6): 2151-2160, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38712889

RESUMEN

Antimicrobial peptides (AMPs) are presented as potential scaffolds for antibiotic development due to their desirable qualities including broad-spectrum activity, rapid action, and general lack of susceptibility to current resistance mechanisms. However, they often lose antibacterial activity under physiological conditions and/or display mammalian cell toxicity, which limits their potential use. Identification of AMPs that overcome these barriers will help develop rules for how this antibacterial class can be developed to treat infection. Here we describe the development of our novel synthetic AMP, from discovery through in vivo application. Our evolved AMP, DTr18-dab, has broad-spectrum antibacterial activity and is nonhemolytic. It is active against planktonic bacteria and biofilm, is unaffected by colistin resistance, and importantly is active in both human serum and a Galleria mellonella infection model. Several modifications, including the incorporation of noncanonical amino acids, were used to arrive at this robust sequence. We observed that the impact on antibacterial activity with noncanonical amino acids was dependent on assay conditions and therefore not entirely predictable. Overall, our results demonstrate how a relatively weak lead can be developed into a robust AMP with qualities important for potential therapeutic translation.


Asunto(s)
Antibacterianos , Péptidos Antimicrobianos , Biopelículas , Pruebas de Sensibilidad Microbiana , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Animales , Humanos , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/síntesis química , Biopelículas/efectos de los fármacos , Mariposas Nocturnas/efectos de los fármacos , Colistina/farmacología , Colistina/química , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/síntesis química
18.
Nat Biomed Eng ; 8(7): 842-853, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39085646

RESUMEN

Many antimicrobial peptides directly disrupt bacterial membranes yet can also damage mammalian membranes. It is therefore central to their therapeutic use that rules governing the membrane selectivity of antimicrobial peptides be deciphered. However, this is difficult even for short peptides owing to the large combinatorial space of amino acid sequences. Here we describe a method for measuring the loss or maintenance of antimicrobial-peptide activity for thousands of peptide-sequence variants simultaneously, and its application to Protegrin-1, a potent yet toxic antimicrobial peptide, to determine the positional importance and flexibility of residues across its sequence while identifying variants with changes in membrane selectivity. More bacterially selective variants maintained a membrane-bound secondary structure while avoiding aromatic residues and cysteine pairs. A machine-learning model trained with our datasets accurately predicted membrane-specific activities for over 5.7 million Protegrin-1 variants, and identified one variant that showed substantially reduced toxicity and retention of activity in a mouse model of intraperitoneal infection. The high-throughput methodology may help elucidate sequence-structure-function relationships in antimicrobial peptides and inform the design of peptide-based synthetic drugs.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Aprendizaje Automático , Animales , Ratones , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/genética , Secuencia de Aminoácidos , Mutación , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/química , Pruebas de Sensibilidad Microbiana , Relación Estructura-Actividad , Femenino
19.
PLoS Pathog ; 7(2): e1001295, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21379340

RESUMEN

Ingested Vibrio cholerae pass through the stomach and colonize the small intestines of its host. Here, we show that V. cholerae requires at least two types of DNA repair systems to efficiently compete for colonization of the infant mouse intestine. These results show that V. cholerae experiences increased DNA damage in the murine gastrointestinal tract. Agreeing with this, we show that passage through the murine gut increases the mutation frequency of V. cholerae compared to liquid culture passage. Our genetic analysis identifies known and novel defense enzymes required for detoxifying reactive nitrogen species (but not reactive oxygen species) that are also required for V. cholerae to efficiently colonize the infant mouse intestine, pointing to reactive nitrogen species as the potential cause of DNA damage. We demonstrate that potential reactive nitrogen species deleterious for V. cholerae are not generated by host inducible nitric oxide synthase (iNOS) activity and instead may be derived from acidified nitrite in the stomach. Agreeing with this hypothesis, we show that strains deficient in DNA repair or reactive nitrogen species defense that are defective in intestinal colonization have decreased growth or increased mutation frequency in acidified nitrite containing media. Moreover, we demonstrate that neutralizing stomach acid rescues the colonization defect of the DNA repair and reactive nitrogen species defense defective mutants suggesting a common defense pathway for these mutants.


Asunto(s)
Cólera/prevención & control , Daño del ADN , Reparación del ADN , Intestinos/microbiología , Especies de Nitrógeno Reactivo/metabolismo , Vibrio cholerae/genética , Vibrio cholerae/patogenicidad , Animales , Animales Recién Nacidos , Adhesión Bacteriana , Cólera/genética , Cólera/microbiología , Ratones , Vibrio cholerae/crecimiento & desarrollo
20.
Access Microbiol ; 5(2)2023.
Artículo en Inglés | MEDLINE | ID: mdl-36910511

RESUMEN

Klebsiella pneumoniae biofilm formation is associated with chronic and relapsing infections. Scanning electron microscopy (SEM) is a powerful tool for characterizing biofilm structure and studying their formation. Reliable visualization of biofilm structure requires careful sample preservation, otherwise there may be loss of non-covalent interactions that are susceptible to damage during the dehydration and washing preparation steps. However, no standard procedure has been adopted in the literature to fix K. pneumoniae biofilm for scanning electron microscopy studies. This lack of standardization makes it challenging to compare results between studies and determine the degree to which native structures have been preserved. To advance this critical area of study, we investigated different scanning electron microscopy fixation methods for K. pneumoniae biofilm preservation. Our study reveals the impact preparation steps can have on retaining in biofilm architecture observed using scanning electron microscopy. Using fixation methods developed through our studies, we show that although species that overproduce capsular extracellular polysaccharides produced more robust biofilms, K. pneumoniae can form a developed biofilm in the absence of capsular polysaccharides.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA