Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 293(39): 15316-15329, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30049795

RESUMEN

The closely related type III secretion system zinc metalloprotease effector proteins GtgA, GogA, and PipA are translocated into host cells during Salmonella infection. They then cleave nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) transcription factor subunits, dampening activation of the NF-κB signaling pathway and thereby suppressing host immune responses. We demonstrate here that GtgA, GogA, and PipA cleave a subset of NF-κB subunits, including p65, RelB, and cRel but not NF-κB1 and NF-κB2, whereas the functionally similar type III secretion system effector NleC of enteropathogenic and enterohemorrhagic Escherichia coli cleaved all five NF-κB subunits. Mutational analysis of NF-κB subunits revealed that a single nonconserved residue in NF-κB1 and NF-κB2 that corresponds to the P1' residue Arg-41 in p65 prevents cleavage of these subunits by GtgA, GogA, and PipA, explaining the observed substrate specificity of these enzymes. Crystal structures of GtgA in its apo-form and in complex with the p65 N-terminal domain explained the importance of the P1' residue. Furthermore, the pattern of interactions suggested that GtgA recognizes NF-κB subunits by mimicking the shape and negative charge of the DNA phosphate backbone. Moreover, structure-based mutational analysis of GtgA uncovered amino acids that are required for the interaction of GtgA with p65, as well as those that are required for full activity of GtgA in suppressing NF-κB activation. This study therefore provides detailed and critical insight into the mechanism of substrate recognition by this family of proteins important for bacterial virulence.


Asunto(s)
Escherichia coli/química , Metaloproteasas/química , Infecciones por Salmonella/genética , Salmonella enterica/química , Secuencia de Aminoácidos/genética , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/patogenicidad , Células HeLa , Humanos , Inmunidad Celular , Metaloproteasas/genética , FN-kappa B/química , Conformación Proteica , Infecciones por Salmonella/microbiología , Salmonella enterica/genética , Salmonella enterica/patogenicidad , Transducción de Señal , Relación Estructura-Actividad , Factor de Transcripción ReIA/química , Sistemas de Secreción Tipo III/química , Sistemas de Secreción Tipo III/genética , Zinc/química
2.
J Gen Virol ; 96(Pt 6): 1380-1388, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25701818

RESUMEN

Hepatitis C virus (HCV) infects the liver and hepatocytes are the major cell type supporting viral replication. Hepatocytes and cholangiocytes derive from a common hepatic progenitor cell that proliferates during inflammatory conditions, raising the possibility that cholangiocytes may support HCV replication and contribute to the hepatic reservoir. We screened cholangiocytes along with a panel of cholangiocarcinoma-derived cell lines for their ability to support HCV entry and replication. While primary cholangiocytes were refractory to infection and lacked expression of several entry factors, two cholangiocarcinoma lines, CC-LP-1 and Sk-ChA-1, supported efficient HCV entry; furthermore, Sk-ChA-1 cells supported full virus replication. In vivo cholangiocarcinomas expressed all of the essential HCV entry factors; however, cholangiocytes adjacent to the tumour and in normal tissue showed a similar pattern of receptor expression to ex vivo isolated cholangiocytes, lacking SR-BI expression, explaining their inability to support infection. This study provides the first report that HCV can infect cholangiocarcinoma cells and suggests that these heterogeneous tumours may provide a reservoir for HCV replication in vivo.


Asunto(s)
Células Epiteliales/virología , Hepacivirus/fisiología , Tropismo Viral , Línea Celular Tumoral , Hepacivirus/crecimiento & desarrollo , Humanos , Internalización del Virus , Replicación Viral
3.
Cell Host Microbe ; 27(1): 41-53.e6, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31862381

RESUMEN

Many Gram-negative bacterial pathogens antagonize anti-bacterial immunity through translocated effector proteins that inhibit pro-inflammatory signaling. In addition, the intracellular pathogen Salmonella enterica serovar Typhimurium initiates an anti-inflammatory transcriptional response in macrophages through its effector protein SteE. However, the target(s) and molecular mechanism of SteE remain unknown. Here, we demonstrate that SteE converts both the amino acid and substrate specificity of the host pleiotropic serine/threonine kinase GSK3. SteE itself is a substrate of GSK3, and phosphorylation of SteE is required for its activity. Remarkably, phosphorylated SteE then forces GSK3 to phosphorylate the non-canonical substrate signal transducer and activator of transcription 3 (STAT3) on tyrosine-705. This results in STAT3 activation, which along with GSK3 is required for SteE-mediated upregulation of the anti-inflammatory M2 macrophage marker interleukin-4Rα (IL-4Rα). Overall, the conversion of GSK3 to a tyrosine-directed kinase represents a tightly regulated event that enables a bacterial virulence protein to reprogram innate immune signaling and establish an anti-inflammatory environment.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Macrófagos/microbiología , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Salmonella typhimurium , Animales , Proteínas Bacterianas/metabolismo , Células HEK293 , Células HeLa , Interacciones Microbiota-Huesped/inmunología , Humanos , Interleucina-4/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas/metabolismo , Salmonella typhimurium/inmunología , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Virulencia/inmunología
4.
Cell Host Microbe ; 22(2): 217-231, 2017 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-28799907

RESUMEN

Serovars of Salmonella enterica cause both gastrointestinal and systemic diseases in a broad range of mammalian hosts, including humans. Salmonella virulence depends in part on its pathogenicity island 2 type III secretion system (SPI-2 T3SS), which is required to translocate at least 28 effector proteins from vacuolar-resident bacteria into host cells. Comparative genomic analysis reveals that all serovars encode a subset of "core" effectors, suggesting that they are critical for virulence in different hosts. An additional subset of effectors is found sporadically throughout different serovars, and several inhibit activation of the innate immune system. In this Review, we summarize the biochemical activities, host cell interaction partners, and physiological functions of SPI-2 T3SS effectors in the context of the selective pressures encountered by S. enterica in vivo. We also consider some of the remaining challenges to achieve a unified understanding of how effector activities work together to promote Salmonella virulence.


Asunto(s)
Salmonella enterica/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Factores de Virulencia/metabolismo , Citoesqueleto de Actina , Inmunidad Adaptativa , Animales , Proteínas Bacterianas/metabolismo , Citosol , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Salmonella/microbiología , Salmonella enterica/genética , Salmonella enterica/patogenicidad , Vacuolas/microbiología , Factores de Virulencia/genética
5.
Nat Commun ; 7: 13292, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27808091

RESUMEN

Sensing bacterial products in the cytosol of mammalian cells by NOD-like receptors leads to the activation of caspase-1 inflammasomes, and the production of the pro-inflammatory cytokines interleukin (IL)-18 and IL-1ß. In addition, mouse caspase-11 (represented in humans by its orthologs, caspase-4 and caspase-5) detects cytosolic bacterial LPS directly. Activation of caspase-1 and caspase-11 initiates pyroptotic host cell death that releases potentially harmful bacteria from the nutrient-rich host cell cytosol into the extracellular environment. Here we use single cell analysis and time-lapse microscopy to identify a subpopulation of host cells, in which growth of cytosolic Salmonella Typhimurium is inhibited independently or prior to the onset of cell death. The enzymatic activities of caspase-1 and caspase-11 are required for growth inhibition in different cell types. Our results reveal that these proteases have important functions beyond the direct induction of pyroptosis and proinflammatory cytokine secretion in the control of growth and elimination of cytosolic bacteria.


Asunto(s)
Caspasa 1/inmunología , Caspasas/inmunología , Citosol/inmunología , Piroptosis/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Células 3T3 , Animales , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasas/genética , Caspasas/metabolismo , Caspasas Iniciadoras , Citosol/enzimología , Citosol/microbiología , Modelos Animales de Enfermedad , Espacio Extracelular/microbiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Inflamasomas/metabolismo , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Salmonella/microbiología , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Análisis de la Célula Individual , Imagen de Lapso de Tiempo
6.
Cell Host Microbe ; 20(5): 584-595, 2016 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-27832589

RESUMEN

The SPI-2 type III secretion system (T3SS) of intracellular Salmonella enterica translocates effector proteins into mammalian cells. Infection of antigen-presenting cells results in SPI-2 T3SS-dependent ubiquitination and reduction of surface-localized mature MHC class II (mMHCII). We identify the effector SteD as required and sufficient for this process. In Mel Juso cells, SteD localized to the Golgi network and vesicles containing the E3 ubiquitin ligase MARCH8 and mMHCII. SteD caused MARCH8-dependent ubiquitination and depletion of surface mMHCII. One of two transmembrane domains and the C-terminal cytoplasmic region of SteD mediated binding to MARCH8 and mMHCII, respectively. Infection of dendritic cells resulted in SteD-dependent depletion of surface MHCII, the co-stimulatory molecule B7.2, and suppression of T cell activation. SteD also accounted for suppression of T cell activation during Salmonella infection of mice. We propose that SteD is an adaptor, forcing inappropriate ubiquitination of mMHCII by MARCH8 and thereby suppressing T cell activation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Evasión Inmune , Salmonella typhimurium/patogenicidad , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Línea Celular , Células Dendríticas/microbiología , Interacciones Huésped-Patógeno , Humanos , Activación de Linfocitos , Ratones , Unión Proteica , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA