Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Cell Mol Life Sci ; 76(20): 4165-4178, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31076805

RESUMEN

Efficient cell-to-cell transfer of Listeria monocytogenes (L. monocytogenes) requires the proper formation of actin-rich membrane protrusions. To date, only the host proteins ezrin, the binding partner of ezrin, CD44, as well as cyclophilin A (CypA) have been identified as crucial components for L. monocytogenes membrane protrusion stabilization and, thus, efficient cell-to-cell movement of the microbes. Here, we examine the classical binding partner of CypA, CD147, and find that this membrane protein is also hijacked by the bacteria for their cellular dissemination. CD147 is enriched at the plasma membrane surrounding the membrane protrusions as well as the resulting invaginations generated in neighboring cells. In cells depleted of CD147, these actin-rich structures appear similar to those generated in CypA depleted cells as they are significantly shorter and more contorted as compared to their straighter counterparts formed in wild-type control cells. The presence of malformed membrane protrusions hampers the ability of L. monocytogenes to efficiently disseminate from CD147-depleted cells. Our findings uncover another important host protein needed for L. monocytogenes membrane protrusion formation and efficient microbial dissemination.


Asunto(s)
Basigina/genética , Membrana Celular/microbiología , Interacciones Huésped-Patógeno/genética , Listeria monocytogenes/fisiología , Shigella flexneri/fisiología , Células A549 , Actinas/genética , Actinas/metabolismo , Animales , Basigina/antagonistas & inhibidores , Basigina/metabolismo , Células CACO-2 , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Ciclofilina A/deficiencia , Ciclofilina A/genética , Endocitosis , Fibroblastos/microbiología , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Listeria monocytogenes/patogenicidad , Listeria monocytogenes/ultraestructura , Ratones , Transporte de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Shigella flexneri/patogenicidad , Shigella flexneri/ultraestructura , Transducción de Señal
2.
J Infect Dis ; 219(1): 145-153, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29733369

RESUMEN

Background: Listeria generate actin-rich tubular protrusions at the plasma membrane that propel the bacteria into neighboring cells. The precise molecular mechanisms governing the formation of these protrusions remain poorly defined. Methods: In this study, we demonstrate that the prolyl cis-trans isomerase (PPIase) cyclophilin A (CypA) is hijacked by Listeria at membrane protrusions used for cell-to-cell spreading. Results: Cyclophilin A localizes within the F-actin of these structures and is crucial for their proper formation, as cells depleted of CypA have extended actin-rich structures that are misshaped and are collapsed due to changes within the F-actin network. The lack of structural integrity within the Listeria membrane protrusions hampers the microbes from spreading from CypA null cells. Conclusions: Our results demonstrate a crucial role for CypA during Listeria infections.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/microbiología , Ciclofilina A/metabolismo , Listeria/metabolismo , Listeriosis/metabolismo , Células A549 , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actinas/ultraestructura , Membrana Celular/metabolismo , Membrana Celular/microbiología , Extensiones de la Superficie Celular/ultraestructura , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Humanos , Listeria/patogenicidad , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Isomerasa de Peptidilprolil/metabolismo
3.
J Proteome Res ; 15(5): 1613-22, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-27018634

RESUMEN

Enteropathogenic and enterohemorrhagic Escherichia coli cause enteric diseases resulting in significant morbidity and mortality worldwide. These pathogens remain extracellular and translocate a set of type III secreted effector proteins into host cells to promote bacterial virulence. Effectors manipulate host cell pathways to facilitate infection by interacting with a variety of host targets, yet the binding partners and mechanism of action of many effectors remain elusive. We performed a mass spectrometry screen to identify host targets for a library of effectors. We found five known effector targets and discovered four novel interactions. Interestingly, we identified multiple effectors that interacted with the microtubule associated protein, ensconsin. Using co-immunoprecipitations, we confirmed that NleB1 and EspL interacted with ensconsin in a region that corresponded to its microtubule binding domain. Ensconsin is an essential cofactor of kinesin-1 that is required for intracellular trafficking, and we demonstrated that intracellular trafficking was severely disrupted during wild type EPEC infections but not during infections with ΔnleB1 or ΔespL mutants. Our findings demonstrate the efficacy of quantitative proteomics for identifying effector-host protein interactions and suggest that vesicular trafficking is a crucial cellular process that may be targeted by NleB1 and EspL through their interaction with ensconsin.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Interacciones Huésped-Patógeno , Sistemas de Secreción Tipo III/metabolismo , Factores de Virulencia/metabolismo , Línea Celular , Humanos , Inmunoprecipitación , Espectrometría de Masas , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Sistemas de Secreción Tipo III/química
4.
Cell Tissue Res ; 363(2): 449-59, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26239909

RESUMEN

Francisella novicida is a surrogate pathogen commonly used to study infections by the potential bioterrorism agent, Francisella tularensis. One of the primary sites of Francisella infections is the liver where >90% of infected cells are hepatocytes. It is known that once Francisella enter cells it occupies a membrane-bound compartment, the Francisella-containing vacuole (FCV), from which it rapidly escapes to replicate in the cytosol. Recent work examining the Francisella disulfide bond formation (Dsb) proteins, FipA and FipB, have demonstrated that these proteins are important during the Francisella infection process; however, details as to how the infections are altered in epithelial cells have remained elusive. To identify the stage of the infections where these Dsbs might act during epithelial infections, we exploited a hepatocyte F. novicida infection model that we recently developed. We found that F. novicida ΔfipA-infected hepatocytes contained bacteria clustered within lysosome-associated membrane protein 1-positive FCVs, suggesting that FipA is involved in the escape of F. novicida from its vacuole. Our morphological evidence provides a tangible link as to how Dsb FipA can influence Francisella infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Francisella/fisiología , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/patología , Animales , Proteínas Bacterianas/genética , Línea Celular , Células Epiteliales/ultraestructura , Francisella/ultraestructura , Hepatocitos/microbiología , Hepatocitos/patología , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones Endogámicos BALB C , Mutación/genética , Vacuolas/metabolismo , Vacuolas/ultraestructura
5.
J Proteome Res ; 14(6): 2520-7, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25907766

RESUMEN

Enteropathogenic Escherichia coli (EPEC) co-opt host signaling pathways and recruit numerous host proteins to motile morphological structures, called pedestals, at sites of bacterial attachment. These pedestals are hallmarks of EPEC-based disease, and the identification and characterization of the functions of pedestal proteins continue to steadily increase. To identify additional constituents in an unbiased manner, we developed a strategy where EPEC pedestals were elongated artificially, severed, and then concentrated prior to their analysis by mass spectrometry (MS)-based proteomics. We identified >90 unique mammalian proteins over multiple experimental trials from our preparations. Seventeen predicted molecules were significantly higher in abundance (p < 0.05) when compared to both the negative controls and sample means. Validation of two identified proteins (cyclophilin A [nonactin-associated] and transgelin [actin-associated]) by immunolocalization was used to confirm our analysis, and both showed enrichment at EPEC pedestals. The EPEC pedestal concentration technique developed here together with the identification of novel pedestal proteins not only provides a resource for the further characterization of molecular components within these structures but also demonstrates that EPEC pedestals can be used as a model system for the identification of novel functions of proteins not normally thought to be at actin-based structures.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Espectrometría de Masas/métodos , Proteómica , Células HeLa , Humanos
6.
Nature ; 449(7164): 827-34, 2007 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17943119

RESUMEN

Bacterial pathogens operate by attacking crucial intracellular pathways in their hosts. These pathogens usually target more than one intracellular pathway and often interact at several points in each of these pathways to commandeer them fully. Although different bacterial pathogens tend to exploit similar pathway components in the host, the way in which they 'hijack' host cells usually differs. Knowledge of how pathogens target distinct cytoskeletal components and immune-cell signalling pathways is rapidly advancing, together with the understanding of bacterial virulence at a molecular level. Studying how these bacterial pathogens subvert host-cell pathways is central to understanding infectious disease.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Interacciones Huésped-Patógeno , Animales , Presentación de Antígeno , Citoesqueleto/metabolismo , Humanos , Transducción de Señal
7.
BMC Microbiol ; 12: 36, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22424399

RESUMEN

BACKGROUND: The spectrin cytoskeleton is emerging as an important host cell target of enteric bacterial pathogens. Recent studies have identified a crucial role for spectrin and its associated proteins during key pathogenic processes of Listeria monocytogenes and Salmonella Typhimurium infections. Here we investigate the involvement of spectrin cytoskeletal components during the pathogenesis of the invasive pathogen Shigella flexneri. RESULTS: Immunofluorescent microscopy reveals that protein 4.1 (p4.1), but not adducin or spectrin, is robustly recruited to sites of S. flexneri membrane ruffling during epithelial cell invasion. Through siRNA-mediated knockdowns, we identify an important role for spectrin and the associated proteins adducin and p4.1 during S. flexneri invasion. Following internalization, all three proteins are recruited to the internalized bacteria, however upon generation of actin-rich comet tails, we observed spectrin recruitment to those structures in the absence of adducin or p4.1. CONCLUSION: These findings highlight the importance of the spectrin cytoskeletal network during S. flexneri pathogenesis and further demonstrate that pathogenic events that were once thought to exclusively recruit the actin cytoskeletal system require additional cytoskeletal networks.


Asunto(s)
Citoesqueleto/metabolismo , Disentería Bacilar/metabolismo , Shigella flexneri/fisiología , Espectrina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/microbiología , Disentería Bacilar/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células HeLa , Humanos
8.
J Infect Dis ; 204(5): 695-703, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21810914

RESUMEN

Enteropathogenic Escherichia coli (EPEC) are primarily extracellular pathogens that generate actin-rich structures known as pedestals during their pathogenesis. Surprising evidence has demonstrated that despite maintaining an extracellular location, EPEC require the endocytic protein, clathrin, for pedestal formation. To evaluate the strategies EPEC use to usurp endocytic machinery, we investigated the roles of a number of clathrin-coated pits components, adaptor protein 2 (AP-2), Eps15 and epsin1, during EPEC infections. We demonstrated that in conjunction with clathrin, pedestal formation also required the recruitment of Eps15 and epsin1 but not AP-2. Because AP-2 orchestrates the recruitment of clathrin, Eps15, and epsin1, as well as other adaptors, during assembly of clathrin-coated pits at the plasma membrane, our findings reveal a novel internalization subversion strategy employed by EPEC. These results further emphasize the recent paradigm that endocytic proteins are important for EPEC-mediated disease.


Asunto(s)
Complejo 2 de Proteína Adaptadora/fisiología , Proteínas Adaptadoras del Transporte Vesicular/fisiología , Proteínas de Unión al Calcio/fisiología , Escherichia coli Enteropatógena/metabolismo , Infecciones por Escherichia coli/metabolismo , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfoproteínas/fisiología , Complejo 2 de Proteína Adaptadora/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Adhesión Bacteriana/fisiología , Sistemas de Secreción Bacterianos/fisiología , Células CACO-2/microbiología , Proteínas de Unión al Calcio/metabolismo , Clatrina/metabolismo , Clatrina/fisiología , Endocitosis , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiología , Células HeLa/microbiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microscopía Fluorescente , Fosfoproteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/fisiología
9.
Infect Immun ; 79(11): 4392-400, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21875964

RESUMEN

To cause disease, Salmonella enterica serovar Typhimurium requires two type III secretion systems that are encoded by Salmonella pathogenicity islands 1 and 2 (SPI-1 and -2). These secretion systems serve to deliver specialized proteins (effectors) into the host cell cytosol. While the importance of these effectors to promote colonization and replication within the host has been established, the specific roles of individual secreted effectors in the disease process are not well understood. In this study, we used an in vivo gallbladder epithelial cell infection model to study the function of the SPI-2-encoded type III effector, SseL. The deletion of the sseL gene resulted in bacterial filamentation and elongation and the unusual localization of Salmonella within infected epithelial cells. Infection with the ΔsseL strain also caused dramatic changes in host cell lipid metabolism and led to the massive accumulation of lipid droplets in infected cells. This phenotype was directly attributable to the deubiquitinase activity of SseL, as a Salmonella strain carrying a single point mutation in the catalytic cysteine also resulted in extensive lipid droplet accumulation. The excessive buildup of lipids due to the absence of a functional sseL gene also was observed in murine livers during S. Typhimurium infection. These results suggest that SseL alters host lipid metabolism in infected epithelial cells by modifying the ubiquitination patterns of cellular targets.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endopeptidasas/metabolismo , Islas Genómicas/fisiología , Metabolismo de los Lípidos/fisiología , Salmonelosis Animal/microbiología , Salmonella typhimurium/metabolismo , Animales , Proteínas Bacterianas/genética , Endopeptidasas/genética , Vesícula Biliar/metabolismo , Vesícula Biliar/microbiología , Regulación Bacteriana de la Expresión Génica , Islas Genómicas/genética , Hígado/metabolismo , Hígado/microbiología , Ratones , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética
10.
Cell Microbiol ; 12(9): 1322-39, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20374249

RESUMEN

Enterohaemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC respectively) are diarrhoeal pathogens that cause the formation of attaching and effacing (A/E) lesions on infected host cells. These pathogens encode a type III secretion system (T3SS) used to inject effector proteins directly into host cells, an essential requirement for virulence. In this study, we identified a function for the type III secreted effector EspZ. Infection with EPEC DeltaespZ caused increased cytotoxicity in HeLa and MDCK cells compared with wild-type EPEC, and expressing espZ in cells abrogated this effect. Using yeast two-hybrid, proteomics, immunofluorescence and co-immunoprecipitation, it was demonstrated that EspZ interacts with the host protein CD98, which contributes to protection against EPEC-mediated cytotoxicity. EspZ enhanced phosphorylation of focal adhesion kinase (FAK) and AKT during infection with EPEC, but CD98 only appeared to facilitate FAK phosphorylation. This study provides evidence that EspZ and CD98 promote host cell survival mechanisms involving FAK during A/E pathogen infection.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/fisiología , Proteína-1 Reguladora de Fusión/metabolismo , Animales , Sistemas de Secreción Bacterianos , Línea Celular , Perros , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Células HeLa , Humanos , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Virulencia
11.
Anat Rec (Hoboken) ; 304(7): 1400-1419, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33099893

RESUMEN

Bacterial pathogens cause disease by subverting the structure and function of their target host cells. Several foodborne agents such as Listeria monocytogenes (L. monocytogenes), Shigella flexneri (S. flexneri), Salmonella enterica serovar Typhimurium (S. Typhimurium) and enteropathogenic Escherichia coli (EPEC) manipulate the host actin cytoskeleton to cause diarrheal (and systemic) infections. During infections, these invasive and adherent pathogens hijack the actin filaments of their host cells and rearrange them into discrete actin-rich structures that promote bacterial adhesion (via pedestals), invasion (via membrane ruffles and endocytic cups), intracellular motility (via comet/rocket tails) and/or intercellular dissemination (via membrane protrusions and invaginations). We have previously shown that actin-rich structures generated by L. monocytogenes contain the host actin cross-linker α-actinin-4. Here we set out to examine α-actinin-4 during other key steps of the L. monocytogenes infectious cycle as well as characterize the subcellular distribution of α-actinin-4 during infections with other model actin-hijacking bacterial pathogens (S. flexneri, S. Typhimurium and EPEC). Although α-actinin-4 is absent at sites of initial L. monocytogenes invasion, we show that it is a new component of the membrane invaginations formed during secondary infections of neighboring host cells. Importantly, we reveal that α-actinin-4 also localizes to the major actin-rich structures generated during cell culture infections with S. flexneri (comet/rocket tails and membrane protrusions), S. Typhimurium (membrane ruffles) and EPEC (pedestals). Taken together, these findings suggest that α-actinin-4 is a host factor that is exploited by an assortment of actin-hijacking bacterial pathogens.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinina/metabolismo , Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Células CACO-2 , Escherichia coli Enteropatógena , Células HeLa , Humanos , Listeria monocytogenes
12.
Anat Rec (Hoboken) ; 304(5): 919-938, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33022122

RESUMEN

The enteric bacterial pathogens Listeria monocytogenes (Listeria) and enteropathogenic Escherichia coli (EPEC) remodel the eukaryotic actin cytoskeleton during their disease processes. Listeria generate slender actin-rich comet/rocket tails to move intracellularly, and later, finger-like membrane protrusions to spread amongst host cells. EPEC remain extracellular, but generate similar actin-rich membranous protrusions (termed pedestals) to move atop the host epithelia. These structures are crucial for disease as diarrheal (and systemic) infections are significantly abrogated during infections with mutant strains that are unable to generate the structures. The current repertoire of host components enriched within these structures is vast and diverse. In this protein catalog, we and others have found that host actin crosslinkers, such as palladin and α-actinin-1, are routinely exploited. To expand on this list, we set out to investigate the distribution of PDLIM1, a scaffolding protein and binding partner of palladin and α-actinin-1, during bacterial infections. We show that PDLIM1 localizes to the site of initial Listeria entry into cells. Following this, PDLIM1 localizes to actin filament clouds surrounding immotile bacteria, and then colocalizes with actin once the comet/rocket tails are generated. Unlike palladin or α-actinin-1, PDLIM1 is maintained within the actin-rich core of membrane protrusions. Conversely, α-actinin-1, but not PDLIM1 (or palladin), is enriched at the membrane invagination that internalizes the Listeria-containing membrane protrusion. We also show that PDLIM1 is a component of the EPEC pedestal core and that its recruitment is dependent on the bacterial effector Tir. Our findings highlight PDLIM1 as another protein present within pathogen-induced actin-rich structures.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Infecciones por Escherichia coli/metabolismo , Proteínas con Dominio LIM/metabolismo , Factores de Transcripción/metabolismo , Escherichia coli Enteropatógena , Células HeLa , Humanos , Listeria monocytogenes
13.
mBio ; 12(6): e0293921, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34781738

RESUMEN

Direct cell-to-cell spreading of Listeria monocytogenes requires the bacteria to induce actin-based finger-like membrane protrusions in donor host cells that are endocytosed through caveolin-rich membrane invaginations by adjacent receiving cells. An actin shell surrounds these endocytic sites; however, its structure, composition, and functional significance remain elusive. Here, we show that the formin mDia1, but surprisingly not the Arp2/3 complex, is enriched at the membrane invaginations generated by L. monocytogenes during HeLa and Jeg-3 cell infections. Electron microscopy reveals a band of linear actin filaments that run along the longitudinal axis of the invagination membrane. Mechanistically, mDia1 expression is vital for the assembly of this F-actin shell. mDia1 is also required for the recruitment of Filamin A, a caveola-associated F-actin cross-linking protein, and caveolin-1 to the invaginations. Importantly, mixed-cell infection assays show that optimal caveolin-based L. monocytogenes cell-to-cell spreading correlates with the formation of the linear actin filament-containing shell by mDia1. IMPORTANCE Listeria monocytogenes spreads from one cell to another to colonize tissues. This cell-to-cell movement requires the propulsive force of an actin-rich comet tail behind the advancing bacterium, which ultimately distends the host plasma membrane into a slender bacterium-containing membrane protrusion. These membrane protrusions induce a corresponding invagination in the membrane of the adjacent host cell. The host cell that receives the protrusion utilizes caveolin-based endocytosis to internalize the structures, and filamentous actin lines these membrane invaginations. Here, we set out to determine the structure and function of this filamentous actin "shell." We demonstrate that the formin mDia1, but not the Arp2/3 complex, localizes to the invaginations. Morphologically, we show that this actin is organized into linear arrays and not branched dendritic networks. Mechanistically, we show that the actin shell is assembled by mDia1 and that mDia1 is required for efficient cell-to-cell transfer of L. monocytogenes.


Asunto(s)
Actinas/metabolismo , Membrana Celular/microbiología , Forminas/metabolismo , Listeria monocytogenes/fisiología , Listeriosis/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiología , Caveolina 1/genética , Caveolina 1/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Filaminas/genética , Filaminas/metabolismo , Forminas/genética , Células HeLa , Humanos , Listeria monocytogenes/genética , Listeriosis/genética , Listeriosis/microbiología
14.
Biochim Biophys Acta ; 1788(4): 832-41, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19059200

RESUMEN

The epithelial barrier is a critical border that segregates luminal material from entering tissues. Essential components of this epithelial fence are physical intercellular structures termed tight junctions. These junctions use a variety of transmembrane proteins coupled with cytoplasmic adaptors, and the actin cytoskeleton, to attach adjacent cells together thereby forming intercellular seals. Breaching of this barrier has profound effects on human health and disease, as barrier deficiencies have been linked with the onset of inflammation, diarrhea generation and pathogenic effects. Although tight junctions efficiently restrict most microbes from penetrating into deeper tissues and contain the microbiota, some pathogens have developed specific strategies to alter or disrupt these structures as part of their pathogenesis, resulting in either pathogen penetration, or other consequences such as diarrhea. Understanding the strategies that microorganisms use to commandeer the functions of tight junctions is an active area of research in microbial pathogenesis. In this review we highlight and overview the tactics bacteria and viruses use to alter tight junctions during disease. Additionally, these studies have identified novel tight junction protein functions by using pathogens and their virulence factors as tools to study the cell biology of junctional structures.


Asunto(s)
Bacterias/patogenicidad , Uniones Estrechas/fisiología , Virus/patogenicidad , Adenoviridae/patogenicidad , Toxina del Cólera/fisiología , Clostridium perfringens/patogenicidad , Enterovirus/patogenicidad , Células Epiteliales/metabolismo , Escherichia coli/patogenicidad , Haptoglobinas , Helicobacter pylori/patogenicidad , Hepacivirus/patogenicidad , Humanos , Precursores de Proteínas , Reoviridae/patogenicidad , Rotavirus/patogenicidad , Salmonella/patogenicidad , Shigella flexneri/patogenicidad
15.
Infect Immun ; 78(8): 3316-22, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20515931

RESUMEN

Enteropathogenic Escherichia coli (EPEC) strains are extracellular pathogens that generate actin-rich structures (pedestals) beneath the adherent bacteria as part of their virulence strategy. Pedestals are hallmarks of EPEC infections, and their efficient formation in vitro routinely requires phosphorylation of the EPEC effector protein Tir at tyrosine 474 (Y474). This phosphorylation results in the recruitment and direct attachment of the host adaptor protein Nck to Tir at Y474, which is utilized for actin nucleation through a downstream N-WASP-Arp2/3-based mechanism. Recently, the endocytic protein clathrin was demonstrated to be involved in EPEC pedestal formation. Here we examine the organization of clathrin in pedestals and report that CD2AP, an endocytosis-associated and cortactin-binding protein, is a novel and important component of EPEC pedestal formation that also utilizes Y474 phosphorylation of EPEC Tir. We also demonstrate the successive recruitment of Nck and then clathrin prior to actin polymerization at pedestals during the Nck-dependent pathway of pedestal formation. This study further demonstrates that endocytic proteins are key components of EPEC pedestals and suggests a novel endocytosis subversion strategy employed by these extracellular bacteria.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adhesión Bacteriana , Proteínas del Citoesqueleto/metabolismo , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/metabolismo , Interacciones Huésped-Patógeno , Receptores de Superficie Celular/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Clatrina/metabolismo , Endocitosis , Humanos , Ratones , Proteínas Oncogénicas/metabolismo , Unión Proteica , Multimerización de Proteína
16.
Cell Microbiol ; 11(2): 351-62, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19016783

RESUMEN

A recently developed model for enterocolitis in mice involves pre-treatment with the antibiotic streptomycin prior to infection with Salmonella enterica serovar Typhimurium (S. Typhimurium). The contribution of Nramp1/Slc11a1 protein, a critical host defence mechanism against S. Typhimurium, to the development of inflammation in this model has not been studied. Here, we analysed the impact of Nramp1 expression on the early development of colitis using isogenic Nramp1(+/+) and Nramp1(-/-) mice. We hypothesized that Nramp1 acts by rapidly inducing an inflammatory response in the gut mucosa creating an antibacterial environment and limiting spread of S. Typhimurium to systemic sites. We observed that Nramp1(+/+) mice showed lower numbers of S. Typhimurium in the caecum compared with Nramp1(-/-) mice at all times analysed. Acute inflammation was much more pronounced in Nramp1(+/+) mice 1 day after infection. The effect of Nramp1 on development of colitis was characterized by higher secretion of the pro-inflammatory cytokines IFN-gamma, TNF-alpha and MIP-1alpha and a massive infiltration of neutrophils and macrophages, compared with Nramp1(-/-) animals. These data show that an early and rapid inflammatory response results in protection against pathological effects of S. Typhimurium infection in Nramp1(+/+) mice.


Asunto(s)
Proteínas de Transporte de Catión/inmunología , Colitis/microbiología , Colitis/patología , Salmonella typhimurium/inmunología , Animales , Proteínas de Transporte de Catión/deficiencia , Ciego/microbiología , Ciego/patología , Quimiocina CCL3/metabolismo , Recuento de Colonia Microbiana , Femenino , Inflamación/patología , Interferón gamma/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Neutrófilos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Infect Dis ; 200(11): 1703-13, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19852670

RESUMEN

The gallbladder is often colonized by Salmonella during typhoid fever, yet little is known about bacterial pathogenesis in this organ. With use of a mouse model of acute typhoid fever, we demonstrate that Salmonella infect gallbladder epithelial cells in vivo. Bacteria in the gallbladder showed a unique behavior as they replicated within gallbladder epithelial cells and remained confined to those cells without translocating to the mucosa. Infected gallbladders showed histopathological damage characterized by destruction of the epithelium and massive infiltration of neutrophils, accompanied by a local increase of proinflammatory cytokines. Damage was determined by the ability of Salmonella to invade gallbladder epithelial cells and was independent of high numbers of replication-competent, although invasion-deficient, bacteria in the lumen. Our results establish gallbladder epithelial cells as a novel niche for in vivo replication of Salmonella and reveal the involvement of these cells in the pathogenesis of Salmonella in the gallbladder during the course of acute typhoid fever.


Asunto(s)
Colecistitis Aguda/microbiología , Vesícula Biliar/microbiología , Salmonelosis Animal/patología , Salmonella typhi/crecimiento & desarrollo , Fiebre Tifoidea/microbiología , Animales , Colecistitis Aguda/patología , Recuento de Colonia Microbiana , Citocinas/metabolismo , Interpretación Estadística de Datos , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Vesícula Biliar/metabolismo , Vesícula Biliar/patología , Histocitoquímica , Inflamación/microbiología , Inflamación/patología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Salmonelosis Animal/microbiología , Fiebre Tifoidea/patología
18.
Anat Rec (Hoboken) ; 303(7): 1859-1864, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31595676

RESUMEN

Klebsiella pneumoniae has become a growing concern within hospitals due to multidrug resistant strains and increasing mortality rates. Recently, we showed that at the subcellular level, K. pneumoniae compromises the integrity of the epithelia by disassembling the microtubule networks of cells through the actions of katanin microtubule severing proteins. In this study, we report on the observation that mitotic cells are targeted by K. pneumoniae and that during infections, the katanin proteins are excluded from the microtubule organizing centers of dividing cells, resulting in the alteration of the microtubule cytoskeleton. Anat Rec, 2019. © 2019 American Association for Anatomy Anat Rec, 303:1859-1864, 2020. © 2019 American Association for Anatomy.


Asunto(s)
Katanina/metabolismo , Klebsiella pneumoniae/metabolismo , Pulmón/microbiología , Microtúbulos/metabolismo , Mitosis/fisiología , Línea Celular , Humanos , Pulmón/citología , Pulmón/metabolismo
19.
mBio ; 11(1)2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31964732

RESUMEN

Listeria monocytogenes moves from one cell to another using actin-rich membrane protrusions that propel the bacterium toward neighboring cells. Despite cholesterol being required for this transfer process, the precise host internalization mechanism remains elusive. Here, we show that caveolin endocytosis is key to this event as bacterial cell-to-cell transfer is severely impaired when cells are depleted of caveolin-1. Only a subset of additional caveolar components (cavin-2 and EHD2) are present at sites of bacterial transfer, and although clathrin and the clathrin-associated proteins Eps15 and AP2 are absent from the bacterial invaginations, efficient L. monocytogenes spreading requires the clathrin-interacting protein epsin-1. We also directly demonstrated that isolated L. monocytogenes membrane protrusions can trigger the recruitment of caveolar proteins in a neighboring cell. The engulfment of these bacterial and cytoskeletal structures through a caveolin-based mechanism demonstrates that the classical nanometer-scale theoretical size limit for this internalization pathway is exceeded by these bacterial pathogens.IMPORTANCEListeria monocytogenes moves from one cell to another as it disseminates within tissues. This bacterial transfer process depends on the host actin cytoskeleton as the bacterium forms motile actin-rich membranous protrusions that propel the bacteria into neighboring cells, thus forming corresponding membrane invaginations. Here, we examine these membrane invaginations and demonstrate that caveolin-1-based endocytosis is crucial for efficient bacterial cell-to-cell spreading. We show that only a subset of caveolin-associated proteins (cavin-2 and EHD2) are involved in this process. Despite the absence of clathrin at the invaginations, the classical clathrin-associated protein epsin-1 is also required for efficient bacterial spreading. Using isolated L. monocytogenes protrusions added onto naive host cells, we demonstrate that actin-based propulsion is dispensable for caveolin-1 endocytosis as the presence of the protrusion/invagination interaction alone triggers caveolin-1 recruitment in the recipient cells. Finally, we provide a model of how this caveolin-1-based internalization event can exceed the theoretical size limit for this endocytic pathway.


Asunto(s)
Caveolina 1/metabolismo , Interacciones Huésped-Patógeno , Listeria monocytogenes/fisiología , Listeriosis/metabolismo , Listeriosis/microbiología , Animales , Biomarcadores , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos
20.
Trends Microbiol ; 16(11): 535-42, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18819802

RESUMEN

Pathogenic microorganisms routinely exploit host cellular functions for their benefit. These alterations often enhance the survival and/or dissemination of the pathogen. However, these effects on the host can be quite debilitating. Consequently, an in-depth understanding of the molecular mechanisms employed by pathogens to manipulate their hosts is crucial. One of the common host phenotypes elicited by enteric pathogens is the generation of diarrhea. Here, we overview the current advances in understanding strategies used by bacterial pathogens to cause diarrheal diseases and discuss how the coordination of various subcellular events can influence disease progression.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Diarrea/microbiología , Animales , Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Tracto Gastrointestinal/microbiología , Interacciones Huésped-Patógeno , Humanos , Uniones Estrechas/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA