Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Pathog ; 10(7): e1004295, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25078082

RESUMEN

Intracellular acting protein exotoxins produced by bacteria and plants are important molecular determinants that drive numerous human diseases. A subset of these toxins, the cytolethal distending toxins (CDTs), are encoded by several Gram-negative pathogens and have been proposed to enhance virulence by allowing evasion of the immune system. CDTs are trafficked in a retrograde manner from the cell surface through the Golgi apparatus and into the endoplasmic reticulum (ER) before ultimately reaching the host cell nucleus. However, the mechanism by which CDTs exit the ER is not known. Here we show that three central components of the host ER associated degradation (ERAD) machinery, Derlin-2 (Derl2), the E3 ubiquitin-protein ligase Hrd1, and the AAA ATPase p97, are required for intoxication by some CDTs. Complementation of Derl2-deficient cells with Derl2:Derl1 chimeras identified two previously uncharacterized functional domains in Derl2, the N-terminal 88 amino acids and the second ER-luminal loop, as required for intoxication by the CDT encoded by Haemophilus ducreyi (Hd-CDT). In contrast, two motifs required for Derlin-dependent retrotranslocation of ERAD substrates, a conserved WR motif and an SHP box that mediates interaction with the AAA ATPase p97, were found to be dispensable for Hd-CDT intoxication. Interestingly, this previously undescribed mechanism is shared with the plant toxin ricin. These data reveal a requirement for multiple components of the ERAD pathway for CDT intoxication and provide insight into a Derl2-dependent pathway exploited by retrograde trafficking toxins.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Toxinas Bacterianas/farmacología , Degradación Asociada con el Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adenosina Trifosfatasas/genética , Animales , Western Blotting , Células CHO , Membrana Celular/metabolismo , Chancroide/metabolismo , Chancroide/microbiología , Chancroide/patología , Cricetinae , Cricetulus , Regulación de la Expresión Génica/efectos de los fármacos , Aparato de Golgi/metabolismo , Haemophilus ducreyi/crecimiento & desarrollo , Haemophilus ducreyi/patogenicidad , Células HeLa , Humanos , Inmunoprecipitación , Inmunosupresores/farmacología , Proteínas de la Membrana/genética , Proteínas Nucleares/genética , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitina-Proteína Ligasas/genética
2.
Proc Natl Acad Sci U S A ; 110(50): E4904-12, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24191014

RESUMEN

Pathogenic microorganisms and toxins have evolved a variety of mechanisms to gain access to the host-cell cytosol and thereby exert virulent effects upon the host. One common mechanism of cellular entry requires trafficking to an acidified endosome, which promotes translocation across the host membrane. To identify small-molecule inhibitors that block this process, a library of 30,000 small molecules was screened for inhibitors of anthrax lethal toxin. Here we report that 4-bromobenzaldehyde N-(2,6-dimethylphenyl)semicarbazone, the most active compound identified in the screen, inhibits intoxication by lethal toxin and blocks the entry of multiple other acid-dependent bacterial toxins and viruses into mammalian cells. This compound, which we named EGA, also delays lysosomal targeting and degradation of the EGF receptor, indicating that it targets host-membrane trafficking. In contrast, EGA does not block endosomal recycling of transferrin, retrograde trafficking of ricin, phagolysosomal trafficking, or phagosome permeabilization by Franciscella tularensis. Furthermore, EGA does not neutralize acidic organelles, demonstrating that its mechanism of action is distinct from pH-raising agents such as ammonium chloride and bafilomycin A1. EGA is a powerful tool for the study of membrane trafficking and represents a class of host-targeted compounds for therapeutic development to treat infectious disease.


Asunto(s)
Toxinas Bacterianas/antagonistas & inhibidores , Endosomas/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Semicarbazonas/farmacología , Internalización del Virus/efectos de los fármacos , Aminas , Animales , Transporte Biológico/fisiología , Caspasa 1/metabolismo , Cromatografía Liquida , Endosomas/fisiología , Citometría de Flujo , Células HeLa , Humanos , Macrófagos , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Estructura Molecular , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Semicarbazonas/química , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
3.
J Biol Chem ; 288(11): 7492-7505, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23306199

RESUMEN

The cytolethal distending toxins (CDTs) compose a subclass of intracellularly acting genotoxins produced by many Gram-negative pathogenic bacteria that disrupt the normal progression of the eukaryotic cell cycle. Here, the intoxication mechanisms of CDTs from Escherichia coli (Ec-CDT) and Haemophilus ducreyi (Hd-CDT), which share limited amino acid sequence homology, were directly compared. Ec-CDT and Hd-CDT shared comparable in vitro DNase activities of the CdtB subunits, saturable cell surface binding with comparable affinities, and the requirement for an intact Golgi complex to induce cell cycle arrest. In contrast, disruption of endosome acidification blocked Hd-CDT-mediated cell cycle arrest and toxin transport to the endoplasmic reticulum and nucleus, while having no effects on Ec-CDT. Phosphorylation of the histone protein H2AX, as well as nuclear localization, was inhibited for Hd-CdtB, but not Ec-CdtB, in cells expressing dominant negative Rab7 (T22N), suggesting that Hd-CDT, but not Ec-CDT, is trafficked through late endosomal vesicles. In support of this idea, significantly more Hd-CdtB than Ec-CdtB co-localized with Rab9, which is enriched in late endosomal compartments. Competitive binding studies suggested that Ec-CDT and Hd-CDT bind to discrete cell surface determinants. These results suggest that Ec-CDT and Hd-CDT are transported within cells by distinct pathways, possibly mediated by their interaction with different receptors at the cell surface.


Asunto(s)
Toxinas Bacterianas/metabolismo , Escherichia coli/metabolismo , Haemophilus ducreyi/metabolismo , Animales , Biotinilación , Células CHO , Células CACO-2 , Ciclo Celular , Núcleo Celular/metabolismo , Clonación Molecular , Cricetinae , Desoxirribonucleasas/metabolismo , Regulación Bacteriana de la Expresión Génica , Células HeLa , Histonas/química , Histonas/metabolismo , Humanos , Transporte de Proteínas , Proteínas Recombinantes/química
4.
J Biol Chem ; 285(24): 18199-207, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20385557

RESUMEN

Cytolethal distending toxins (CDTs) are tripartite protein exotoxins produced by a diverse group of pathogenic Gram-negative bacteria. Based on their ability to induce DNA damage, cell cycle arrest, and apoptosis of cultured cells, CDTs are proposed to enhance virulence by blocking cellular division and/or directly killing epithelial and immune cells. Despite the widespread distribution of CDTs among several important human pathogens, our understanding of how these toxins interact with host cells is limited. Here we demonstrate that CDTs from Haemophilus ducreyi, Aggregatibacter actinomycetemcomitans, Escherichia coli, and Campylobacter jejuni differ in their abilities to intoxicate host cells with defined defects in host factors previously implicated in CDT binding, including glycoproteins, and glycosphingolipids. The absence of cell surface sialic acid sensitized cells to intoxication by three of the four CDTs tested. Surprisingly, fucosylated N-linked glycans and glycolipids, previously implicated in CDT-host interactions, were not required for intoxication by any of the CDTs tested. Finally, altering host-cellular cholesterol, also previously implicated in CDT binding, affected intoxication by only a subset of CDTs tested. The findings presented here provide insight into the molecular and cellular basis of CDT-host interactions.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Colesterol/química , Polisacáridos/química , Animales , Células CHO , Campylobacter jejuni/metabolismo , Colesterol/metabolismo , Cricetinae , Cricetulus , Daño del ADN , Escherichia coli/metabolismo , Glucolípidos/química , Bacterias Gramnegativas/metabolismo , Haemophilus ducreyi/metabolismo , Células HeLa , Humanos , Ratones , Células 3T3 NIH , Unión Proteica
5.
Artículo en Inglés | MEDLINE | ID: mdl-23061054

RESUMEN

The cytolethal distending toxins (CDTs) comprise a family of intracellular-acting bacterial protein toxins whose actions upon eukaryotic cells result in several consequences, the most characteristic of which is the induction of G(2)/M cell cycle arrest. Most CDTs are hetero-tripartite assemblies of CdtA, CdtB, and CdtC, with CdtB required for CDT-mediated cell cycle arrest. Several lines of evidence indicate that CdtA and CdtC are required for the optimal intracellular activity of CdtB, although the exact functional roles of CdtA and CdtC remain poorly understood. The genes encoding the CDTs have been identified in a diverse array of Gram-negative pathogenic bacteria. More recently, the genes encoding several CdtB subunits have been associated with alternatively linked subunits resembling the B-subunits of pertussis toxin. Although the CDTs are generally considered to all function as bacterial genotoxins, the extent to which individual members of the CDTs employ similar mechanisms of cell surface binding, uptake, and trafficking within sensitive cells is poorly understood. Recently, data have begun to emerge suggesting differences in the molecular basis by which individual CDTs interact with and enter host cells, suggesting the possibility that CDTs possess properties reflecting the specific niches idiosyncratic to those CDT bacterial pathogens that produce them. The extent to which functional differences between individual CDTs reflect the specific requirements for intoxicating cells and tissues within the diverse range of host microenvironments colonized by CDT-producing pathogenic bacteria remains to be experimentally explored.


Asunto(s)
Toxinas Bacterianas/toxicidad , Ciclo Celular/efectos de los fármacos , Células Eucariotas/efectos de los fármacos , Bacterias Gramnegativas/patogenicidad , Unión Proteica , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA