Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(43): 26868-26875, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33055207

RESUMEN

Horizontal gene transfer (HGT) confers the rapid acquisition of novel traits and is pervasive throughout microbial evolution. Despite the central role of HGT, the evolutionary forces that drive the dynamics of HGT alleles in evolving populations are poorly understood. Here, we show that HGT alters the evolutionary dynamics of genetic variation, so that deleterious genetic variants, including antibiotic resistance genes, can establish in populations without selection. We evolve antibiotic-sensitive populations of the human pathogen Helicobacter pylori in an environment without antibiotic but with HGT from an antibiotic-resistant isolate of H. pylori We find that HGT increases the rate of adaptation, with most horizontally transferred genetic variants establishing at a low frequency in the population. When challenged with antibiotic, this low-level variation potentiates adaptation, with HGT populations flourishing in conditions where nonpotentiated populations go extinct. By extending previous models of evolution under HGT, we evaluated the conditions for the establishment and spread of HGT-acquired alleles into recipient populations. We then used our model to estimate parameters of HGT and selection from our experimental evolution data. Together, our findings show how HGT can act as an evolutionary force that facilitates the spread of nonselected genetic variation and expands the adaptive potential of microbial populations.


Asunto(s)
Adaptación Fisiológica/genética , Evolución Biológica , Farmacorresistencia Bacteriana/genética , Transferencia de Gen Horizontal , Helicobacter pylori/genética , Antibacterianos , Flujo Génico , Aptitud Genética , Variación Genética , Metronidazol , Selección Genética
2.
J Enzyme Inhib Med Chem ; 34(1): 1660-1667, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31530039

RESUMEN

Ethoxzolamide (EZA), acetazolamide, and methazolamide are clinically used sulphonamide drugs designed to treat non-bacteria-related illnesses (e.g. glaucoma), but they also show antimicrobial activity against the gastric pathogen Helicobacter pylori. EZA showed the highest activity, and was effective against clinical isolates resistant to metronidazole, clarithromycin, and/or amoxicillin, suggesting that EZA kills H. pylori via mechanisms different from that of these antibiotics. The frequency of single-step spontaneous resistance acquisition by H. pylori was less than 5 × 10-9, showing that resistance to EZA does not develop easily. Resistance was associated with mutations in three genes, including the one that encodes undecaprenyl pyrophosphate synthase, a known target of sulphonamides. The data indicate that EZA impacts multiple targets in killing H. pylori. Our findings suggest that developing the approved anti-glaucoma drug EZA into a more effective anti-H. pylori agent may offer a faster and cost-effective route towards new antimicrobials with a novel mechanism of action.


Asunto(s)
Antibacterianos/farmacología , Etoxzolamida/farmacología , Helicobacter pylori/efectos de los fármacos , Antibacterianos/síntesis química , Antibacterianos/química , Relación Dosis-Respuesta a Droga , Etoxzolamida/síntesis química , Etoxzolamida/química , Helicobacter pylori/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad
3.
Artículo en Inglés | MEDLINE | ID: mdl-29468142

RESUMEN

The Gram-negative bacterium, Helicobacter pylori, causes chronic gastritis, peptic ulcers, and gastric cancer in humans. Although the gastric epithelium is the primary site of H. pylori colonization, H. pylori can gain access to deeper tissues. Concurring with this notion, H. pylori has been found in the vicinity of endothelial cells in gastric submucosa. Endothelial cells play crucial roles in innate immune response, wound healing and tumorigenesis. This study examines the molecular mechanisms by which H. pylori interacts with and triggers inflammatory responses in endothelial cells. We observed that H. pylori infection of primary human endothelial cells stimulated secretion of the key inflammatory cytokines, interleukin-6 (IL-6) and interleukin-8 (IL-8). In particular, IL-8, a potent chemokine and angiogenic factor, was secreted by H. pylori-infected endothelial cells to levels ~10- to 20-fold higher than that typically observed in H. pylori-infected gastric epithelial cells. These inflammatory responses were triggered by the H. pylori type IV secretion system (T4SS) and the T4SS-associated adhesin CagL, but not the translocation substrate CagA. Moreover, in contrast to integrin α5ß1 playing an essential role in IL-8 induction by H. pylori upon infection of gastric epithelial cells, both integrin α5ß1 and integrin αvß3 were dispensable for IL-8 induction in H. pylori-infected endothelial cells. However, epidermal growth factor receptor (EGFR) is crucial for mediating the potent H. pylori-induced IL-8 response in endothelial cells. This study reveals a novel mechanism by which the H. pylori T4SS and its adhesin subunit, CagL, may contribute to H. pylori pathogenesis by stimulating the endothelial innate immune responses, while highlighting EGFR as a potential therapeutic target for controlling H. pylori-induced inflammation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno , Sistemas de Secreción Tipo IV/metabolismo , Adhesinas Bacterianas/metabolismo , Biomarcadores , Línea Celular , Citocinas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Receptores ErbB/metabolismo , Infecciones por Helicobacter/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Fosforilación , Transporte de Proteínas
4.
Sci Rep ; 7(1): 16140, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29170397

RESUMEN

The Helicobacter pylori phase variable gene modH, typified by gene HP1522 in strain 26695, encodes a N6-adenosine type III DNA methyltransferase. Our previous studies identified multiple strain-specific modH variants (modH1 - modH19) and showed that phase variation of modH5 in H. pylori P12 influenced expression of motility-associated genes and outer membrane protein gene hopG. However, the ModH5 DNA recognition motif and the mechanism by which ModH5 controls gene expression were unknown. Here, using comparative single molecule real-time sequencing, we identify the DNA site methylated by ModH5 as 5'-Gm6ACC-3'. This motif is vastly underrepresented in H. pylori genomes, but overrepresented in a number of virulence genes, including motility-associated genes, and outer membrane protein genes. Motility and the number of flagella of H. pylori P12 wild-type were significantly higher than that of isogenic modH5 OFF or ΔmodH5 mutants, indicating that phase variable switching of modH5 expression plays a role in regulating H. pylori motility phenotypes. Using the flagellin A (flaA) gene as a model, we show that ModH5 modulates flaA promoter activity in a GACC methylation-dependent manner. These findings provide novel insights into the role of ModH5 in gene regulation and how it mediates epigenetic regulation of H. pylori motility.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Proteínas Bacterianas/genética , Epigénesis Genética/genética , Regulación Bacteriana de la Expresión Génica/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Helicobacter pylori/genética
5.
Microbiologyopen ; 6(6)2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29055967

RESUMEN

Helicobacter pylori is a gram-negative bacterial pathogen that chronically inhabits the human stomach. To survive and maintain advantage, it has evolved unique host-pathogen interactions mediated by Helicobacter-specific proteins in the bacterial outer membrane. These outer membrane proteins (OMPs) are anchored to the cell surface via a C-terminal ß-barrel domain, which requires their assembly by the ß-barrel assembly machinery (BAM). Here we have assessed the complexity of the OMP C-terminal ß-barrel domains employed by H. pylori, and characterized the H. pyloriBAM complex. Around 50 Helicobacter-specific OMPs were assessed with predictive structural algorithms. The data suggest that H. pylori utilizes a unique ß-barrel architecture that might constitute H. pylori-specific Type V secretions system. The structural and functional diversity in these proteins is encompassed by their extramembrane domains. Bioinformatic and biochemical characterization suggests that the low ß-barrel-complexity requires only minimalist assembly machinery. The H. pylori proteins BamA and BamD associate to form a BAM complex, with features of BamA enabling an oligomerization that might represent a mechanism by which a minimalist BAM complex forms a larger, sophisticated machinery capable of servicing the outer membrane proteome of H. pylori.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Helicobacter pylori/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Cristalografía por Rayos X , Helicobacter pylori/química , Helicobacter pylori/genética , Modelos Moleculares , Conformación Proteica , Dominios Proteicos , Pliegue de Proteína
6.
Toxins (Basel) ; 9(8)2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28788072

RESUMEN

Cytotoxin-associated gene product A (CagA) is a major virulence factor secreted by Helicobacter pylori. CagA activity in the gastric epithelium is associated with higher risk of gastric cancer development. Bacterial type IV secretion system (T4SS)-mediated translocation of CagA into the cytosol of human epithelial cells occurs via a poorly understood mechanism that requires CagA interaction with the host membrane lipid phosphatidylserine (PS) and host cell receptor integrin α5ß1. Here we have characterized the isolated recombinant middle fragment of CagA (CagA-M) that contains the positively-charged PS-binding region (aa 613-636) and a putative ß1 integrin binding site, but lacks the EPIYA region, secretion signal peptide and the CagA multimerization motif. We show that CagA-M, when immobilized on latex beads, is capable of binding to, and triggering its own uptake into, gastric epithelial cells in the absence of infection with cagA-positive H. pylori. Using site-directed mutagenesis, fluorescent and electron microscopy, and highly-specific inhibitors, we demonstrate that the cell-binding and endocytosis-like internalization of CagA-M are dependent on (1) binding to PS; (2) ß1 integrin activity; and (3) actin dynamics. Interaction of CagA-M with the host cells is accompanied by the development of long filopodia-like protrusions (macrospikes). This novel morphology is different from the hummingbird phenotype induced by the translocation of full-length CagA. The determinants within CagA-M and within the host that are important for endocytosis-like internalization into host cells are very similar to those observed for T4SS-mediated internalization of full-length CagA, suggesting that the latter may involve an endocytic pathway.


Asunto(s)
Actinas/metabolismo , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno , Antígenos Bacterianos/química , Proteínas Bacterianas/química , Células Cultivadas , Endocitosis , Mucosa Gástrica/citología , Humanos , Proteínas Inmovilizadas/metabolismo , Proteínas Recombinantes/metabolismo
7.
Cell Stress Chaperones ; 21(3): 523-33, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26928021

RESUMEN

Bacterial infections typically elicit a strong Heat Shock Response (HSR) in host cells. However, the gastric pathogen Helicobacter pylori has the unique ability to repress this response, the mechanism of which has yet to be elucidated. This study sought to characterize the underlying mechanisms by which H. pylori down-modulates host HSP expression upon infection. Examination of isogenic mutant strains of H. pylori defective in components of the type IV secretion system (T4SS), identified the secretion substrate, CagA, to be essential for down-modulation of the HSPs HSPH1 (HSP105), HSPA1A (HSP72), and HSPD1 (HSP60) upon infection of the AGS gastric adenocarcinoma cell line. Ectopic expression of CagA by transient transfection was insufficient to repress HSP expression in AGS or HEK293T cells, suggesting that additional H. pylori factors are required for HSP repression. RT-qPCR analysis of HSP gene expression in AGS cells infected with wild-type H. pylori or isogenic cagA-deletion mutant found no significant change to account for reduced HSP levels. In summary, this study identified CagA to be an essential bacterial factor for H. pylori-mediated suppression of host HSP expression. The novel finding that HSPH1 is down-modulated by H. pylori further highlights the unique ability of H. pylori to repress the HSR within host cells. Elucidation of the mechanism by which H. pylori achieves HSP repression may prove to be beneficial in the identification of novel mechanisms to inhibit the HSR pathway and provide further insight into the interactions between H. pylori and the host gastric epithelium.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Choque Térmico/biosíntesis , Helicobacter pylori/genética , Interacciones Huésped-Patógeno/genética , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Proteínas de Choque Térmico/genética , Respuesta al Choque Térmico/genética , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Humanos , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología
8.
J Infect Dis ; 213(12): 1927-31, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-26908724

RESUMEN

Previous studies suggest overrepresentation of particular polymorphisms within the Helicobacter pylori CagL hypervariable motif (CagLHM) in gastric cancer-associated isolates. However, these disease correlations were geographically variable and ambiguous. We compared the disease correlation of several hundred geographically diverse CagL sequences and identified 33 CagLHM sequence combinations with disparate geographical distribution, revealing substantial worldwide CagLHM diversity, particularly within Asian countries. Notably, polymorphisms E59 and I60 were significantly overrepresented, whereas D58 and E62 were underrepresented, in gastric cancer-associated H. pylori isolates worldwide. Thus, CagLHM regional diversity may contribute to the varied prevalence of H. pylori-related gastric cancer observed in diverse populations.


Asunto(s)
Proteínas Bacterianas/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Polimorfismo Genético/genética , Neoplasias Gástricas/microbiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Antígenos Bacterianos/genética , Asia/epidemiología , Salud Global , Infecciones por Helicobacter/epidemiología , Helicobacter pylori/aislamiento & purificación , Interacciones Huésped-Patógeno , Humanos , Prevalencia , Neoplasias Gástricas/epidemiología
9.
Infect Immun ; 81(10): 3880-93, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918779

RESUMEN

The natural immune response to Helicobacter pylori neither clears infection nor prevents reinfection. However, the ability of secretory antibodies to influence the course of H. pylori infection has not been determined. We compared the natural progression of H. pylori infection in wild-type C57BL/6 mice with that in mice lacking the polymeric immunoglobulin receptor (pIgR) that is essential for the secretion of polymeric antibody across mucosal surfaces. H. pylori SS1-infected wild-type and pIgR knockout (KO) mice were sampled longitudinally for gastrointestinal bacterial load, antibody response, and histological changes. The gastric bacterial loads of wild-type and pIgR KO mice remained constant and comparable at up to 3 months postinfection (mpi) despite SS1-reactive secretory IgA in the intestinal contents of wild-type mice at that time. Conversely, abundant duodenal colonization of pIgR KO animals contrasted with the near-total eradication of H. pylori from the intestine of wild-type animals by 3 mpi. H. pylori was cultured only from the duodenum of those animals in which colonization in the distal gastric antrum was of sufficient density for immunohistological detection. By 6 mpi, the gastric load of H. pylori in wild-type mice was significantly lower than in pIgR KO animals. While there was no corresponding difference between the two mouse strains in gastric pathology results at 6 mpi, reductions in gastric bacterial load correlated with increased gastric inflammation together with an intestinal secretory antibody response in wild-type mice. Together, these results suggest that naturally produced secretory antibodies can modulate the progress of H. pylori infection, particularly in the duodenum.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Inmunidad Mucosa/fisiología , Mucosa Intestinal/metabolismo , Animales , Western Blotting , Regulación de la Expresión Génica/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/metabolismo , Inmunoglobulina G/sangre , Inmunoglobulina G/metabolismo , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Inmunoglobulina Polimérica/genética , Receptores de Inmunoglobulina Polimérica/metabolismo
10.
Cell Microbiol ; 15(4): 554-70, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23107019

RESUMEN

The type IV secretion system (T4SS) of Helicobacter pylori triggers massive inflammatory responses during gastric infection by mechanisms that are poorly understood. Here we provide evidence for a novel pathway by which the T4SS structural component, CagL, induces secretion of interleukin-8 (IL-8) independently of CagA translocation and peptidoglycan-sensing nucleotide-binding oligomerization domain 1 (NOD1) signalling. Recombinant CagL was sufficient to trigger IL-8 secretion, requiring activation of α5 ß1 integrin and the arginine-glycine-aspartate (RGD) motif in CagL. Mutation of the encoded RGD motif to arginine-glycine-alanine (RGA) in the cagL gene of H. pylori abrogated its ability to induce IL-8. Comparison of IL-8 induction between H. pylori ΔvirD4 strains bearing wild-type or mutant cagL indicates that CagL-dependent IL-8 induction can occur independently of CagA translocation. In line with this notion, exogenous CagL complemented H. pylori ΔcagL mutant in activating NF-κB and inducing IL-8 without restoring CagA translocation. The CagA translocation-independent, CagL-dependent IL-8 induction involved host signalling via integrin α5 ß1 , Src kinase, the mitogen-activated protein kinase (MAPK) pathway and NF-κB but was independent of NOD1. Our findings reveal a novel pathway whereby CagL, via interaction with host integrins, can trigger pro-inflammatory responses independently of CagA translocation or NOD1 signalling.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Helicobacter pylori/inmunología , Interleucina-8/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteínas Bacterianas/genética , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Integrina alfa5beta1/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Proteínas Mutantes/metabolismo , Mutación , FN-kappa B/metabolismo , Transducción de Señal
11.
PLoS One ; 6(12): e27569, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22162751

RESUMEN

Many host-adapted bacterial pathogens contain DNA methyltransferases (mod genes) that are subject to phase-variable expression (high-frequency reversible ON/OFF switching of gene expression). In Haemophilus influenzae and pathogenic Neisseria, the random switching of the modA gene, associated with a phase-variable type III restriction modification (R-M) system, controls expression of a phase-variable regulon of genes (a "phasevarion"), via differential methylation of the genome in the modA ON and OFF states. Phase-variable type III R-M systems are also found in Helicobacter pylori, suggesting that phasevarions may also exist in this key human pathogen. Phylogenetic studies on the phase-variable type III modH gene revealed that there are 17 distinct alleles in H. pylori, which differ only in their DNA recognition domain. One of the most commonly found alleles was modH5 (16% of isolates). Microarray analysis comparing the wild-type P12modH5 ON strain to a P12ΔmodH5 mutant revealed that six genes were either up- or down-regulated, and some were virulence-associated. These included flaA, which encodes a flagella protein important in motility and hopG, an outer membrane protein essential for colonization and associated with gastric cancer. This study provides the first evidence of this epigenetic mechanism of gene expression in H. pylori. Characterisation of H. pylori modH phasevarions to define stable immunological targets will be essential for vaccine development and may also contribute to understanding H. pylori pathogenesis.


Asunto(s)
Epigénesis Genética , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/genética , Algoritmos , Alelos , Proteínas Bacterianas/genética , ADN/genética , Metilación de ADN , Metilasas de Modificación del ADN/genética , Epigenómica , Variación Genética , Modelos Genéticos , Reacción en Cadena de la Polimerasa , Estructura Terciaria de Proteína , Regulón , Análisis de Secuencia de ADN , Programas Informáticos
12.
Infect Immun ; 77(11): 5116-29, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19737910

RESUMEN

Studies of active immunization against Helicobacter pylori indicate that antibodies play a minor role in immunity. There is also evidence, however, that the translocation of antibodies in the stomach may be insufficient to achieve functional antibody levels in the gastric lumen. We have used a suckling mouse model of passive immunity to determine if perorally delivered antibodies can protect against infection with H. pylori. Female C57BL/6 mice were immunized parenterally with formalin-fixed cells of three clinical isolates of H. pylori (3HP) or the mouse-adapted H. pylori strain SS1 before mating. Their pups were challenged with the SS1 strain at 4 days of age and left to suckle before determination of bacterial loads 14 days later. Compared to age-matched controls, pups suckled by 3HP-vaccinated dams were significantly protected against infection (>95% reduction in median bacterial load; P<0.0001). Pups suckled by SS1-vaccinated dams were also significantly protected in terms of both median bacterial load (>99.5% reduction; P<0.0001) and the number of culture-negative pups (28% versus 2% for immune and nonimmune cohorts, respectively; P<0.0001). Similar results were obtained with pups suckled by dams immunized with a urease-deficient mutant of SS1. Fostering experiments demonstrated that protection was entirely attributable to suckling from an immunized dam, and antibody isotype analysis suggested that protection was mediated by the immunoglobulin G fraction of immune milk. Analysis of the bacterial loads in pups sampled before and after weaning confirmed that infection had been prevented in culture-negative animals. These data indicate that antibodies can prevent colonization by H. pylori and suppress the bacterial loads in animals that are colonized.


Asunto(s)
Anticuerpos Antibacterianos/administración & dosificación , Anticuerpos Antibacterianos/inmunología , Infecciones por Helicobacter/prevención & control , Inmunidad Materno-Adquirida/inmunología , Leche/inmunología , Animales , Animales Lactantes , Femenino , Infecciones por Helicobacter/sangre , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Inmunoglobulina A/análisis , Inmunoglobulina A/inmunología , Inmunoglobulina G/análisis , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Leche/química
13.
FEMS Immunol Med Microbiol ; 44(2): 213-9, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15866218

RESUMEN

Helicobacter pylori populations recovered from the human stomach display extensive recombination and quasispecies development, and this suggests frequent exchange of DNA between different strains in vivo. In vitro, however, most H. pylori strains display restriction to the uptake of non-self DNA, as measured using selectable markers, regardless of their competency for transformation with self DNA. We have examined the effect of different selectable markers on double-crossover recombination efficiencies in three reference strains (1061, 26695 & SS1) and one clinical isolate (CHP1) of H. pylori. All strains were efficiently transformable to kanamycin or chloramphenicol resistance by using self-genomic DNA from isogenic mutants bearing the aphA3 or cat cassettes, respectively. However, strains 26695 and CHP1 showed a 3-5-log reduction in transformation efficiency by non-self recombinant DNA containing aphA3, when compared to cat. Strain 1061 readily accepted either cassette, and strain SS1 was poorly tolerant of any non-self DNA. Genome-wide random mutagenesis of these strains was only achievable with a selectable marker that allowed high transformation efficiency. Digestion of 32P-labelled cassettes by H. pylori lysates mirrored the transformation results and indicated that in some strains these cassettes are the targets of enzymatic restriction.


Asunto(s)
Proteínas Bacterianas/metabolismo , Enzimas de Restricción del ADN/metabolismo , ADN Bacteriano/metabolismo , Farmacorresistencia Bacteriana/genética , Helicobacter pylori/genética , Transformación Bacteriana , Proteínas Bacterianas/genética , Cloranfenicol/farmacología , ADN Bacteriano/genética , Marcadores Genéticos , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/metabolismo , Humanos , Kanamicina/farmacología , Mutagénesis , Recombinación Genética
14.
J Virol ; 76(21): 11113-22, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12368353

RESUMEN

Picornavirus RNA replication requires the formation of replication complexes (RCs) consisting of virus-induced vesicles associated with viral nonstructural proteins and RNA. Brefeldin A (BFA) has been shown to strongly inhibit RNA replication of poliovirus but not of encephalomyocarditis virus (EMCV). Here, we demonstrate that the replication of parechovirus 1 (ParV1) is partly resistant to BFA, whereas echovirus 11 (EV11) replication is strongly inhibited. Since BFA inhibits COPI-dependent steps in endoplasmic reticulum (ER)-Golgi transport, we tested a hypothesis that different picornaviruses may have differential requirements for COPI in the formation of their RCs. Using immunofluorescence and cryo-immunoelectron microscopy we examined the association of a COPI component, beta-COP, with the RCs of EMCV, ParV1, and EV11. EMCV RCs did not contain beta-COP. In contrast, beta-COP appeared to be specifically distributed to the RCs of EV11. In ParV1-infected cells beta-COP was largely dispersed throughout the cytoplasm, with some being present in the RCs. These results suggest that there are differences in the involvement of COPI in the formation of the RCs of various picornaviruses, corresponding to their differential sensitivity to BFA. EMCV RCs are likely to be formed immediately after vesicle budding from the ER, prior to COPI association with membranes. ParV1 RCs are formed from COPI-containing membranes but COPI is unlikely to be directly involved in their formation, whereas formation of EV11 RCs appears to be dependent on COPI association with membranes.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Proteína Coat de Complejo I/metabolismo , Virus de la Encefalomiocarditis/genética , Enterovirus Humano B/genética , Parechovirus/genética , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Replicación Viral , Animales , Antivirales/farmacología , Brefeldino A/farmacología , Línea Celular , Chlorocebus aethiops , Proteína Coatómero/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/fisiología , Virus de la Encefalomiocarditis/ultraestructura , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/fisiología , Enterovirus Humano B/ultraestructura , Humanos , Parechovirus/efectos de los fármacos , Parechovirus/fisiología , Parechovirus/ultraestructura
15.
Proc Natl Acad Sci U S A ; 99(10): 7066-71, 2002 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-12011463

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) is an enteric pathogen that causes cholera-like diarrhea in humans and animals. ETEC secretes a heat-labile enterotoxin (LT), which resembles cholera toxin, but the actual mechanism of LT secretion is presently unknown. We have identified a previously unrecognized type II protein secretion pathway in the prototypic human ETEC strain, H10407 (serotype O78:H11). The genes for this pathway are absent from E. coli K-12, although examination of the K-12 genome suggests that it probably once possessed them. The secretory pathway bears significant homology at the amino acid level to the type II protein secretory pathway required by Vibrio cholerae for the secretion of cholera toxin. With this in mind, we determined whether the homologous pathway of E. coli H10407 played a role in the secretion of LT. To this end, we inactivated the pathway by inserting a kanamycin-resistance gene into one of the genes (gspD) of the type II secretion pathway by homologous recombination. LT secretion by E. coli H10407 and the gspD mutant was assayed by enzyme immunoassay, and its biological activity was assessed by using Y-1 adrenal cells. This investigation showed that the protein secretory pathway is functional and necessary for the secretion of LT by ETEC. Our findings have revealed the mechanism for the secretion of LT by ETEC, which previously was unknown, and provide further evidence of close biological similarities of the LT and cholera toxin.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Enterotoxinas/metabolismo , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Secuencia de Bases , ADN Bacteriano , Enterotoxinas/genética , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Genes Bacterianos , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...