Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Infect Immun ; 88(8)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32513855

RESUMO

The serum complement system is a first line of defense against bacterial invaders. Resistance to killing by serum enhances the capacity of Klebsiella pneumoniae to cause infection, but it is an incompletely understood virulence trait. Identifying and characterizing the factors responsible for preventing activation of, and killing by, serum complement could inform new approaches to treatment of K. pneumoniae infections. Here, we used functional genomic profiling to define the genetic basis of complement resistance in four diverse serum-resistant K. pneumoniae strains (NTUH-K2044, B5055, ATCC 43816, and RH201207), and explored their recognition by key complement components. More than 90 genes contributed to resistance in one or more strains, but only three, rfaH, lpp, and arnD, were common to all four strains. Deletion of the antiterminator rfaH, which controls the expression of capsule and O side chains, resulted in dramatic complement resistance reductions in all strains. The murein lipoprotein gene lpp promoted capsule retention through a mechanism dependent on its C-terminal lysine residue; its deletion led to modest reductions in complement resistance. Binding experiments with the complement components C3b and C5b-9 showed that the underlying mechanism of evasion varied in the four strains: B5055 and NTUH-K2044 appeared to bypass recognition by complement entirely, while ATCC 43816 and RH201207 were able to resist killing despite being associated with substantial levels of C5b-9. All rfaH and lpp mutants bound C3b and C5b-9 in large quantities. Our findings show that, even among this small selection of isolates, K. pneumoniae adopts differing mechanisms and utilizes distinct gene sets to avoid complement attack.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Carboxiliases/imunologia , Regulação Bacteriana da Expressão Gênica/imunologia , Genes Bacterianos , Evasão da Resposta Imune , Klebsiella pneumoniae/imunologia , Fatores de Alongamento de Peptídeos/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Atividade Bactericida do Sangue/imunologia , Carboxiliases/deficiência , Carboxiliases/genética , Complemento C3b/genética , Complemento C3b/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Elementos de DNA Transponíveis , Perfilação da Expressão Gênica , Biblioteca Gênica , Humanos , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/patogenicidade , Mutação , Fatores de Alongamento de Peptídeos/deficiência , Fatores de Alongamento de Peptídeos/genética , Análise de Sequência de DNA
2.
Molecules ; 25(8)2020 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-32340372

RESUMO

Green tea-derived galloylated catechins have weak direct antibacterial activity against both Gram-positive and Gram-negative bacterial pathogens and are able to phenotypically transform, at moderate concentrations, methicillin-resistant Staphylococcus aureus (MRSA) clonal pathogens from full ß-lactam resistance (minimum inhibitory concentration 256-512 mg/L) to complete susceptibility (~1 mg/L). Reversible conversion to susceptibility follows intercalation of these compounds into the bacterial cytoplasmic membrane, eliciting dispersal of the proteins associated with continued cell wall peptidoglycan synthesis in the presence of ß-lactam antibiotics. The molecules penetrate deep within the hydrophobic core of the lipid palisade to force a reconfiguration of cytoplasmic membrane architecture. The catechin gallate-induced staphylococcal phenotype is complex, reflecting perturbation of an essential bacterial organelle, and includes prevention and inhibition of biofilm formation, disruption of secretion of virulence-related proteins, dissipation of halotolerance, cell wall thickening and cell aggregation and poor separation of daughter cells during cell division. These features are associated with the reduction of capacity of potential pathogens to cause lethal, difficult-to-treat infections and could, in combination with ß-lactam agents that have lost therapeutic efficacy due to the emergence of antibiotic resistance, form the basis of a new approach to the treatment of staphylococcal infections.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Catequina/análogos & derivados , Chá/química , Antibacterianos/química , Bactérias/metabolismo , Catequina/química , Catequina/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/genética , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Relação Estrutura-Atividade
3.
Infect Immun ; 87(5)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30833331

RESUMO

Gastrointestinal (GI) colonization of 2-day-old (P2) rat pups with Escherichia coli K1 results in translocation of the colonizing bacteria across the small intestine, bacteremia, and invasion of the meninges, with animals frequently succumbing to lethal infection. Infection, but not colonization, is strongly age dependent; pups become progressively less susceptible to infection over the P2-to-P9 period. Colonization leads to strong downregulation of the gene encoding trefoil factor 2 (Tff2), preventing maturation of the protective mucus barrier in the small intestine. Trefoil factors promote mucosal homeostasis. We investigated the contribution of Tff2 to protection of the neonatal rat from E. coli K1 bacteremia and tissue invasion. Deletion of tff2, using clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9, sensitized P9 pups to E. coli K1 bacteremia. There were no differences between tff2-/- homozygotes and the wild type with regard to the dynamics of GI colonization. Loss of the capacity to elaborate Tff2 did not impact GI tract integrity or the thickness of the small-intestinal mucus layer but, in contrast to P9 wild-type pups, enabled E. coli K1 bacteria to gain access to epithelial surfaces in the distal region of the small intestine and exploit an intracellular route across the epithelial monolayer to enter the blood circulation via the mesenteric lymphatic system. Although primarily associated with the mammalian gastric mucosa, we conclude that loss of Tff2 in the developing neonatal small intestine enables the opportunistic neonatal pathogen E. coli K1 to enter the compromised mucus layer in the distal small intestine prior to systemic invasion and infection.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Escherichia coli/patogenicidade , Imunidade Inata/imunologia , Sepse Neonatal/imunologia , Fator Trefoil-2/imunologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Humanos , Ratos
4.
Am J Pathol ; 188(10): 2164-2176, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30036519

RESUMO

Preterm birth is a serious global health problem and the leading cause of infant death before 5 years of age. At least 40% of cases are associated with infection. The most common way for pathogens to access the uterine cavity is by ascending from the vagina. Bioluminescent pathogens have revolutionized the understanding of infectious diseases. We hypothesized that bioluminescent Escherichia coli can be used to track and monitor ascending vaginal infections. Two bioluminescent strains were studied: E. coli K12 MG1655-lux, a nonpathogenic laboratory strain, and E. coli K1 A192PP-lux2, a pathogenic strain capable of causing neonatal meningitis and sepsis in neonatal rats. On embryonic day 16, mice received intravaginal E. coli K12, E. coli K1, or phosphate-buffered saline followed by whole-body bioluminescent imaging. In both cases, intravaginal delivery of E. coli K12 or E. coli K1 led to bacterial ascension into the uterine cavity, but only E. coli K1 induced preterm parturition. Intravaginal administration of E. coli K1 significantly reduced the proportion of pups born alive compared with E. coli K12 and phosphate-buffered saline controls. However, in both groups of viable pups born after bacterial inoculation, there was evidence of comparable brain inflammation by postnatal day 6. This study ascribes specific mechanisms by which exposure to intrauterine bacteria leads to premature delivery and neurologic inflammation in neonates.


Assuntos
Lesões Encefálicas/microbiologia , Nascimento Prematuro/microbiologia , Doenças Vaginais/microbiologia , Animais , Animais Recém-Nascidos , Corioamnionite/microbiologia , Modelos Animais de Doenças , Infecções por Escherichia coli/fisiopatologia , Feminino , Doenças Fetais/microbiologia , Camundongos , Gravidez , Complicações Infecciosas na Gravidez/microbiologia
5.
J Bacteriol ; 200(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29339415

RESUMO

Escherichia coli K1 strains are major causative agents of invasive disease of newborn infants. The age dependency of infection can be reproduced in neonatal rats. Colonization of the small intestine following oral administration of K1 bacteria leads rapidly to invasion of the blood circulation; bacteria that avoid capture by the mesenteric lymphatic system and evade antibacterial mechanisms in the blood may disseminate to cause organ-specific infections such as meningitis. Some E. coli K1 surface constituents, in particular the polysialic acid capsule, are known to contribute to invasive potential, but a comprehensive picture of the factors that determine the fully virulent phenotype has not emerged so far. We constructed a library and constituent sublibraries of ∼775,000 Tn5 transposon mutants of E. coli K1 strain A192PP and employed transposon-directed insertion site sequencing (TraDIS) to identify genes required for fitness for infection of 2-day-old rats. Transposon insertions were lacking in 357 genes following recovery on selective agar; these genes were considered essential for growth in nutrient-replete medium. Colonization of the midsection of the small intestine was facilitated by 167 E. coli K1 gene products. Restricted bacterial translocation across epithelial barriers precluded TraDIS analysis of gut-to-blood and blood-to-brain transits; 97 genes were required for survival in human serum. This study revealed that a large number of bacterial genes, many of which were not previously associated with systemic E. coli K1 infection, are required to realize full invasive potential.IMPORTANCEEscherichia coli K1 strains cause life-threatening infections in newborn infants. They are acquired from the mother at birth and colonize the small intestine, from where they invade the blood and central nervous system. It is difficult to obtain information from acutely ill patients that sheds light on physiological and bacterial factors determining invasive disease. Key aspects of naturally occurring age-dependent human infection can be reproduced in neonatal rats. Here, we employ transposon-directed insertion site sequencing to identify genes essential for the in vitro growth of E. coli K1 and genes that contribute to the colonization of susceptible rats. The presence of bottlenecks to invasion of the blood and cerebrospinal compartments precluded insertion site sequencing analysis, but we identified genes for survival in serum.


Assuntos
Antígenos de Bactérias/genética , Elementos de DNA Transponíveis , Infecções por Escherichia coli/sangue , Escherichia coli/genética , Trato Gastrointestinal/microbiologia , Genoma Bacteriano , Polissacarídeos Bacterianos/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/patogenicidade , Escherichia coli/fisiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Aptidão Genética , Humanos , Viabilidade Microbiana/efeitos dos fármacos , Mutagênese , Mutação , Ratos , Ratos Wistar , Soro/microbiologia , Virulência/genética
6.
Methods ; 127: 62-68, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28522324

RESUMO

In contrast to two-dimensional bioluminescence imaging, three dimensional diffuse light imaging tomography with integrated micro-computed tomography (DLIT-µCT) has the potential to realise spatial variations in infection patterns when imaging experimental animals dosed with derivatives of virulent bacteria carrying bioluminescent reporter genes such as the lux operon from the bacterium Photorhabdus luminescens. The method provides an opportunity to precisely localise the bacterial infection sites within the animal and enables the generation of four-dimensional movies of the infection cycle. Here, we describe the use of the PerkinElmer IVIS SpectrumCT in vivo imaging system to investigate progression of lethal systemic infection in neonatal rats following colonisation of the gastrointestinal tract with the neonatal pathogen Escherichia coli K1. We confirm previous observations that these bacteria stably colonize the colon and small intestine following feeding of the infectious dose from a micropipette; invading bacteria migrate across the gut epithelium into the blood circulation and establish foci of infection in major organs, including the brain. DLIT-µCT revealed novel multiple sites of colonisation within the alimentary canal, including the tongue, oesophagus and stomach, with penetration of the non-keratinised oesophageal epithelial surface, providing strong evidence of a further major site for bacterial dissemination. We highlight technical issues associated with imaging of infections in new born rat pups and show that the whole-body and organ bioburden correlates with disease severity.


Assuntos
Modelos Animais de Doenças , Infecções por Escherichia coli/patologia , Imageamento Tridimensional/métodos , Sepse/patologia , Fatores Etários , Animais , Animais Recém-Nascidos , Progressão da Doença , Genes Reporter , Medições Luminescentes/métodos , Microrganismos Geneticamente Modificados/genética , Ratos , Tomografia Óptica/métodos , Microtomografia por Raio-X/métodos
7.
J Infect Dis ; 214(6): 916-24, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27412582

RESUMO

The genotoxin colibactin, synthesized by Escherichia coli, is a secondary metabolite belonging to the chemical family of hybrid polyketide/nonribosomal peptide compounds. It is produced by a complex biosynthetic assembly line encoded by the pks pathogenicity island. The presence of this large cluster of genes in the E. coli genome is invariably associated with the high-pathogenicity island, encoding the siderophore yersiniabactin, which belongs to the same chemical family as colibactin. The E. coli heat shock protein HtpG (Hsp90Ec) is the bacterial homolog of the eukaryotic molecular chaperone Hsp90, which is involved in the protection of cellular proteins against a variety of environmental stresses. In contrast to eukaryotic Hsp90, the functions and client proteins of Hsp90Ec are poorly known. Here, we demonstrated that production of colibactin and yersiniabactin is abolished in the absence of Hsp90Ec We further characterized an interplay between the Hsp90Ec molecular chaperone and the ClpQ protease involved in colibactin and yersiniabactin synthesis. Finally, we demonstrated that Hsp90Ec is required for the full in vivo virulence of extraintestinal pathogenic E. coli This is the first report highlighting the role of heat shock protein Hps90Ec in the production of two secondary metabolites involved in E. coli virulence.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Mutagênicos/metabolismo , Peptídeos/metabolismo , Fenóis/metabolismo , Policetídeos/metabolismo , Sideróforos/metabolismo , Tiazóis/metabolismo , Animais , Modelos Animais de Doenças , Endopeptidase Clp/metabolismo , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/genética , Feminino , Deleção de Genes , Proteínas de Choque Térmico HSP90/genética , Camundongos Endogâmicos C57BL , Mapeamento de Interação de Proteínas , Ratos Wistar , Virulência
8.
Infect Immun ; 83(12): 4528-40, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26351276

RESUMO

Key features of Escherichia coli K1-mediated neonatal sepsis and meningitis, such as a strong age dependency and development along the gut-mesentery-blood-brain course of infection, can be replicated in the newborn rat. We examined temporal and spatial aspects of E. coli K1 infection following initiation of gastrointestinal colonization in 2-day-old (P2) rats after oral administration of E. coli K1 strain A192PP and a virulent bioluminescent derivative, E. coli A192PP-lux2. A combination of bacterial enumeration in the major organs, two-dimensional bioluminescence imaging, and three-dimensional diffuse light imaging tomography with integrated micro-computed tomography indicated multiple sites of colonization within the alimentary canal; these included the tongue, esophagus, and stomach in addition to the small intestine and colon. After invasion of the blood compartment, the bacteria entered the central nervous system, with restricted colonization of the brain, and also invaded the major organs, in line with increases in the severity of symptoms of infection. Both keratinized and nonkeratinized surfaces of esophagi were colonized to a considerably greater extent in susceptible P2 neonates than in corresponding tissues from infection-resistant 9-day-old rat pups; the bacteria appeared to damage and penetrate the nonkeratinized esophageal epithelium of infection-susceptible P2 animals, suggesting the esophagus represents a portal of entry for E. coli K1 into the systemic circulation. Thus, multimodality imaging of experimental systemic infections in real time indicates complex dynamic patterns of colonization and dissemination that provide new insights into the E. coli K1 infection of the neonatal rat.


Assuntos
Escherichia coli/patogenicidade , Esôfago/patologia , Meningite/patologia , Sepse/patologia , Fatores Etários , Animais , Animais Recém-Nascidos , Translocação Bacteriana , Encéfalo/imunologia , Encéfalo/microbiologia , Encéfalo/patologia , Colo/imunologia , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Esôfago/imunologia , Esôfago/microbiologia , Expressão Gênica , Genes Reporter , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Luciferases/genética , Luciferases/metabolismo , Meningite/imunologia , Meningite/microbiologia , Ratos , Ratos Wistar , Sepse/imunologia , Sepse/microbiologia , Estômago/imunologia , Estômago/microbiologia , Estômago/patologia , Língua/imunologia , Língua/microbiologia , Língua/patologia , Virulência
9.
Infect Immun ; 83(9): 3704-11, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26150540

RESUMO

Escherichia coli strains expressing the K1 capsule are a major cause of sepsis and meningitis in human neonates. The development of these diseases is dependent on the expression of a range of virulence factors, many of which remain uncharacterized. Here, we show that all but 1 of 34 E. coli K1 neonatal isolates carried clbA and clbP, genes contained within the pks pathogenicity island and required for the synthesis of colibactin, a polyketide-peptide genotoxin that causes genomic instability in eukaryotic cells by induction of double-strand breaks in DNA. Inactivation of clbA and clbP in E. coli A192PP, a virulent strain of serotype O18:K1 that colonizes the gastrointestinal tract and translocates to the blood compartment with very high frequency in experimental infection of the neonatal rat, significantly reduced the capacity of A192PP to colonize the gut, engender double-strand breaks in DNA, and cause invasive, lethal disease. Mutation of clbA, which encodes a pleiotropic enzyme also involved in siderophore synthesis, impacted virulence to a greater extent than mutation of clbP, encoding an enzyme specific to colibactin synthesis. Restoration of colibactin gene function by complementation reestablished the fully virulent phenotype. We conclude that colibactin contributes to the capacity of E. coli K1 to colonize the neonatal gastrointestinal tract and to cause invasive disease in the susceptible neonate.


Assuntos
Infecções por Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Peptídeos/metabolismo , Policetídeos/metabolismo , Animais , Animais Recém-Nascidos , Sequência de Bases , Modelos Animais de Doenças , Escherichia coli/genética , Infecções por Escherichia coli/genética , Ilhas Genômicas/genética , Imuno-Histoquímica , Dados de Sequência Molecular , Peptídeos/genética , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Virulência/fisiologia
10.
Mol Microbiol ; 91(6): 1136-47, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24428662

RESUMO

A mixed culture of Pseudomonas fluorescens and Pusillimonas noertemanii, obtained by soil enrichment, elaborated an enzyme (EnvD) which rapidly hydrolysed poly-γ-d-glutamic acid (PDGA), the constituent of the anti-phagocytic capsule conferring virulence on Bacillus anthracis. The EnvD gene is carried on the P. noertemanii genome but co-culture is required for the elaboration of PDGA depolymerase activity. EnvD showed strong sequence homology to dienelactone hydrolases from other Gram-negative bacteria, possessed no general protease activity but cleaved γ-links in both d- and l-glutamic acid-containing polymers. The stability at 37°C was markedly superior to that of CapD, a γ-glutamyltranspeptidase with PDGA depolymerase activity. Recombinant EnvD was recovered from inclusion bodies in soluble form from an Escherichia coli expression vector and the enzyme stripped the PDGA capsule from the surface of B. anthracis Pasteur within 5 min. We conclude from this in vitro study that rEnvD shows promise as a potential therapeutic for the treatment of anthrax.


Assuntos
Bacillus anthracis/química , Cápsulas Bacterianas/metabolismo , Hidrolases/metabolismo , Ácido Poliglutâmico/metabolismo , Pseudomonas/enzimologia , Biotransformação , Estabilidade Enzimática , Hidrolases/química , Hidrolases/isolamento & purificação , Pseudomonas/isolamento & purificação , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Microbiologia do Solo , Temperatura
11.
Antimicrob Agents Chemother ; 59(12): 7687-92, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26438506

RESUMO

Left untreated, inhalation anthrax is usually fatal. Vegetative forms of Bacillus anthracis survive in blood and tissues during infection due to elaboration of a protective poly-γ-D-glutamic acid (PDGA) capsule that permits uncontrolled bacterial growth in vivo, eventually leading to overwhelming bacillosis and death. As a measure to counter threats from multidrug-resistant strains, we are evaluating the prophylactic and therapeutic potential of the PDGA depolymerase EnvD, a stable and potent enzyme which rapidly and selectively removes the capsule from the surface of vegetative cells. Repeated intravenous administration of 10 mg/kg recombinant EnvD (rEnvD) to mice infected with lethal doses of B. anthracis Ames spores by inhalation prevented the emergence of symptoms of anthrax and death; all animals survived the 5-day treatment period, and 70% survived to the end of the 14-day observation period. In contrast to results in sham-treated animals, the lungs and spleen of rEnvD-dosed animals were free of gross pathological changes. We conclude that rEnvD has potential as an agent to prevent the emergence of inhalation anthrax in infected animals and is likely to be effective against drug-resistant forms of the pathogen.


Assuntos
Antraz/prevenção & controle , Antibacterianos/uso terapêutico , Cápsulas Bacterianas/efeitos dos fármacos , Peptídeo Hidrolases/uso terapêutico , Infecções Respiratórias/prevenção & controle , Administração Intravenosa , Aerossóis , Animais , Antibacterianos/administração & dosagem , Bacillus anthracis/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla , Feminino , Meia-Vida , Camundongos Endogâmicos BALB C , Peptídeo Hidrolases/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico
12.
Int J Mol Sci ; 16(8): 16710-27, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26213914

RESUMO

The polyphenol (-)-epicatechin gallate (ECg) inserts into the cytoplasmic membrane (CM) of methicillin-resistant Staphylococcus aureus (MRSA) and reversibly abrogates resistance to ß-lactam antibiotics. ECg elicits an increase in MRSA cell size and induces thickened cell walls. As ECg partially delocalizes penicillin-binding protein PBP2 from the septal division site, reduces PBP2 and PBP2a complexation and induces CM remodelling, we examined the impact of ECg membrane intercalation on phospholipid distribution across the CM and determined if ECg affects the equatorial, orthogonal mode of division. The major phospholipids of the staphylococcal CM, lysylphosphatidylglycerol (LPG), phosphatidylglycerol (PG), and cardiolipin (CL), were distributed in highly asymmetric fashion; 95%-97% of LPG was associated with the inner leaflet whereas PG (~90%) and CL (~80%) were found predominantly in the outer leaflet. ECg elicited small, significant changes in LPG distribution. Atomic force microscopy established that ECg-exposed cells divided in similar fashion to control bacteria, with a thickened band of encircling peptidoglycan representing the most recent plane of cell division, less distinct ribs indicative of previous sites of orthogonal division and concentric rings and "knobbles" representing stages of peptidoglycan remodelling during the cell cycle. Preservation of staphylococcal membrane lipid asymmetry and mode of division in sequential orthogonal planes appear key features of ECg-induced stress.


Assuntos
Catequina/análogos & derivados , Membrana Celular/química , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Fosfolipídeos/química , beta-Lactamas/metabolismo , Cardiolipinas/química , Catequina/farmacologia , Membrana Celular/efeitos dos fármacos , Lisina/química , Lipídeos de Membrana/química , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Microscopia de Força Atômica , Peptidoglicano/química , Fenótipo , Fosfatidilgliceróis/química
13.
Nanotechnology ; 25(28): 285101, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24972373

RESUMO

Surfactant-mediated removal of proteins from biomembranes invariably results in partial or complete loss of function and disassembly of multi-protein complexes. We determined the capacity of styrene-co-maleic acid (SMA) co-polymer to remove components of the cell division machinery from the membrane of drug-resistant staphylococcal cells. SMA-lipid nanoparticles solubilized FtsZ-PBP2-PBP2a complexes from intact cells, demonstrating the close physical proximity of these proteins within the lipid bilayer. Exposure of bacteria to (-)-epicatechin gallate, a polyphenolic agent that abolishes ß-lactam resistance in staphylococci, disrupted the association between PBP2 and PBP2a. Thus, SMA purification provides a means to remove native integral membrane protein assemblages with minimal physical disruption and shows promise as a tool for the interrogation of molecular aspects of bacterial membrane protein structure and function.


Assuntos
Proteínas de Bactérias/química , Proteínas de Membrana/química , Proteínas de Ligação às Penicilinas/química , Peptídeo Sintases/química , Staphylococcus aureus/química , Tensoativos/química , Catequina/análogos & derivados , Catequina/química , Divisão Celular/fisiologia , Bicamadas Lipídicas/química , Maleatos/química , Poliestirenos/química
14.
Tetrahedron ; 70(21): 3485-3490, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24876661

RESUMO

The high-yielding synthesis of enantiomerically pure epicatechin gallate analogues where the A and/or B-ring hydroxylation is reduced or altered has been achieved by optimising routes to the catechin stereochemistry. The B-ring analogues were synthesised by using an electrophilic ring closure onto an enantiomerically enriched epoxide as a key step. The A and B-ring hydroxyl-deleted analogues were synthesised through a Mitsunobu cyclisation. For the B-ring analogues, the anti- (catechin) stereochemistry was converted to the syn- (epicatechin) stereochemistry by a known oxidation/reduction protocol. Absolute stereochemistry was derived from either a Sharpless epoxidation or asymmetric dihydroxylation.

15.
Infect Immun ; 81(9): 3264-75, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23798529

RESUMO

Two-day-old (P2), but not 9-day-old (P9), rat pups are susceptible to systemic infection following gastrointestinal colonization by Escherichia coli K1. Age dependency reflects the capacity of colonizing K1 to translocate from gastrointestinal (GI) tract to blood. A complex GI microbiota developed by P2, showed little variation over P2 to P9, and did not prevent stable K1 colonization. Substantial developmental expression was observed over P2 to P9, including upregulation of genes encoding components of the small intestinal (α-defensins Defa24 and Defa-rs1) and colonic (trefoil factor Tff2) mucus barrier. K1 colonization modulated expression of these peptides: developmental expression of Tff2 was dysregulated in P2 tissues and was accompanied by a decrease in mucin Muc2. Conversely, α-defensin genes were upregulated in P9 tissues. We propose that incomplete development of the mucus barrier during early neonatal life and the capacity of colonizing K1 to interfere with mucus barrier maturation provide opportunities for neuropathogen translocation into the bloodstream.


Assuntos
Escherichia coli/crescimento & desenvolvimento , Escherichia coli/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Animais , Animais Recém-Nascidos , Colo/imunologia , Colo/metabolismo , Colo/microbiologia , Trato Gastrointestinal/metabolismo , Imunidade Inata/imunologia , Intestino Delgado/metabolismo , Microbiota/genética , Microbiota/imunologia , Mucinas/genética , Mucinas/imunologia , Mucinas/metabolismo , Muco/imunologia , Muco/metabolismo , Muco/microbiologia , Peptídeos/genética , Peptídeos/imunologia , Peptídeos/metabolismo , Ratos , Fator Trefoil-2 , Regulação para Cima/genética , Regulação para Cima/imunologia , alfa-Defensinas/genética , alfa-Defensinas/imunologia , alfa-Defensinas/metabolismo
16.
Appl Environ Microbiol ; 79(2): 714-7, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23124233

RESUMO

The poly-γ-d-glutamic acid capsule of Bacillus anthracis is a barrier to infection by B. anthracis-specific bacteriophages. Capsule expression was found to completely inhibit lytic infection by γ phage, an observation supported by the demonstration that this phage does not elaborate a hydrolase that would facilitate penetration through the protective capsule outer layer.


Assuntos
Fagos Bacilares/fisiologia , Bacillus anthracis/virologia , Cápsulas Bacterianas/metabolismo , Bacteriólise , Ácido Poliglutâmico/metabolismo , Fagos Bacilares/enzimologia , Fagos Bacilares/genética , Fagos Bacilares/crescimento & desenvolvimento , Bacillus anthracis/metabolismo , DNA Viral/química , DNA Viral/genética , Hidrolases/genética , Dados de Sequência Molecular , Análise de Sequência de DNA
17.
Nat Commun ; 14(1): 3294, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37322051

RESUMO

Escherichia coli is a leading cause of invasive bacterial infections in humans. Capsule polysaccharide has an important role in bacterial pathogenesis, and the K1 capsule has been firmly established as one of the most potent capsule types in E. coli through its association with severe infections. However, little is known about its distribution, evolution and functions across the E. coli phylogeny, which is fundamental to elucidating its role in the expansion of successful lineages. Using systematic surveys of invasive E. coli isolates, we show that the K1-cps locus is present in a quarter of bloodstream infection isolates and has emerged in at least four different extraintestinal pathogenic E. coli (ExPEC) phylogroups independently in the last 500 years. Phenotypic assessment demonstrates that K1 capsule synthesis enhances E. coli survival in human serum independent of genetic background, and that therapeutic targeting of the K1 capsule re-sensitizes E. coli from distinct genetic backgrounds to human serum. Our study highlights that assessing the evolutionary and functional properties of bacterial virulence factors at population levels is important to better monitor and predict the emergence of virulent clones, and to also inform therapies and preventive medicine to effectively control bacterial infections whilst significantly lowering antibiotic usage.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Humanos , Escherichia coli , Infecções por Escherichia coli/microbiologia , Virulência/genética , Fatores de Virulência/genética , Proteínas de Escherichia coli/genética , Filogenia
18.
Biochemistry ; 51(29): 5860-71, 2012 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-22746203

RESUMO

Various bis-benzimidazole derivatives have been reported to possess activity against Gram-positive pathogens. No mechanism of action has been elucidated to fully account for the antibacterial activity of this class of compounds. A group of symmetric bis-benzimidazoles (BBZ) designed as anticancer agents have previously been shown to possess moderate antiproliferative activity. We sought to assess the antibacterial activity and mechanism of action of BBZ compounds against Staphylococcus aureus. Antibacterial activities were assessed by determination of minimal inhibitory concentrations (MICs), time-kill curves, and scanning electron microscopy. Transcriptional responses to BBZ treatment were determined using whole genome microarrays. Activities against bacterial type II topoisomerases were investigated using in vitro supercoiling, decatenation, DNA binding, and DNA cleavage inhibition assays. MICs for EMRSA-16 were between 0.03 and 0.5 µg/mL. The compounds showed concentration-dependent bactericidal activity and induced cell swelling and lysis. Transcriptional responses to BBZ were consistent with topoisomerase inhibition and DNA damage. A subset of BBZ compounds inhibited S. aureus DNA gyrase supercoiling activity with IC(50) values in the range of 5-10 µM. This inhibition was subsequently shown to operate through both inhibition of binding of DNA gyrase to DNA and accumulation of single-stranded DNA breaks. We conclude that BBZ compounds are potent anti-staphylococcal agents and operate at least in part through DNA gyrase inhibition, leading to the accumulation of single-stranded DNA breaks, and by preventing the binding of gyrase to DNA.


Assuntos
Antibacterianos/farmacologia , Benzimidazóis/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/enzimologia , Inibidores da Topoisomerase II , Antibacterianos/química , Benzimidazóis/química , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , DNA Girase/metabolismo , DNA Topoisomerase IV/antagonistas & inibidores , DNA Topoisomerase IV/metabolismo , DNA Bacteriano/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/enzimologia
19.
Infect Immun ; 80(5): 1690-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22354027

RESUMO

Campylobacter jejuni infection often results in bloody, inflammatory diarrhea, indicating bacterial disruption and invasion of the intestinal epithelium. While C. jejuni infection can be reproduced in vitro using intestinal epithelial cell (IEC) lines, low numbers of bacteria invading IECs do not reflect these clinical symptoms. Performing in vitro assays under atmospheric oxygen conditions neither is optimal for microaerophilic C. jejuni nor reflects the low-oxygen environment of the intestinal lumen. A vertical diffusion chamber (VDC) model system creates microaerobic conditions at the apical surface and aerobic conditions at the basolateral surface of cultured IECs, producing an in vitro system that closely mimics in vivo conditions in the human intestine. Ninefold increases in interacting and 80-fold increases in intracellular C. jejuni 11168H wild-type strain bacteria were observed after 24-h coculture with Caco-2 IECs in VDCs under microaerobic conditions at the apical surface, compared to results under aerobic conditions. Increased bacterial interaction was matched by an enhanced and directional host innate immune response, particularly an increased basolateral secretion of the proinflammatory chemokine interleukin-8 (IL-8). Analysis of the invasive ability of a nonmotile C. jejuni 11168H rpoN mutant in the VDC model system indicates that motility is an important factor in the early stages of bacterial invasion. The first report of the use of a VDC model system for studying the interactions of an invasive bacterial pathogen with IECs demonstrates the importance of performing such experiments under conditions that represent the in vivo situation and will allow novel insights into C. jejuni pathogenic mechanisms.


Assuntos
Campylobacter jejuni/fisiologia , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Mucosa Intestinal/citologia , Oxigênio/farmacologia , Actinas/metabolismo , Aerobiose , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Técnicas Bacteriológicas , Campylobacter jejuni/efeitos dos fármacos , Técnicas de Cocultura , Regulação da Expressão Gênica/fisiologia , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Proteínas de Membrana/metabolismo , Ocludina , Tegafur
20.
J Antimicrob Chemother ; 67(7): 1683-96, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22547662

RESUMO

OBJECTIVES: Pyrrolobenzodiazepine (PBD) dimers, tethered through inert propyldioxy or pentyldioxy linkers, possess potent bactericidal activity against a range of Gram-positive bacteria by virtue of their capacity to cross-link duplex DNA in sequence-selective fashion. Here we attempt to improve the antibacterial activity and cytotoxicity profile of PBD-containing conjugates by extension of dimer linkers and replacement of one PBD unit with phenyl-substituted or benzo-fused heterocycles that facilitate non-covalent interactions with duplex DNA. METHODS: DNase I footprinting was used to identify high-affinity DNA binding sites. A staphylococcal gene microarray was used to assess epidemic methicillin-resistant Staphylococcus aureus 16 phenotypes induced by PBD conjugates. Molecular dynamics simulations were employed to investigate the accommodation of compounds within the DNA helix. RESULTS: Increasing the length of the linker in PBD dimers led to a progressive reduction in antibacterial activity, but not in their cytotoxic capacity. Complex patterns of DNA binding were noted for extended PBD dimers. Modelling of DNA strand cross-linking by PBD dimers indicated distortion of the helix. A majority (26 of 43) of PBD-biaryl conjugates possessed potent antibacterial activity with little or no helical distortion and a more favourable cytotoxicity profile. Bactericidal activity of PBD-biaryl conjugates was determined by inability to excise covalently bound drug molecules from bacterial duplex DNA. CONCLUSIONS: PBD-biaryl conjugates have a superior antibacterial profile compared with PBD dimers such as ELB-21. We have identified six PBD-biaryl conjugates as potential drug development candidates.


Assuntos
Antibacterianos/farmacologia , Benzodiazepinas/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Pirróis/farmacologia , Antibacterianos/metabolismo , Benzodiazepinas/metabolismo , Sítios de Ligação , Pegada de DNA , DNA Bacteriano/metabolismo , Perfilação da Expressão Gênica , Análise em Microsséries , Viabilidade Microbiana/efeitos dos fármacos , Simulação de Dinâmica Molecular , Pirróis/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA