Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 19(11): e1011677, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37917600

RESUMEN

Candida albicans, the primary etiology of human mycoses, is well-adapted to catabolize proline to obtain energy to initiate morphological switching (yeast to hyphal) and for growth. We report that put1-/- and put2-/- strains, carrying defective Proline UTilization genes, display remarkable proline sensitivity with put2-/- mutants being hypersensitive due to the accumulation of the toxic intermediate pyrroline-5-carboxylate (P5C), which inhibits mitochondrial respiration. The put1-/- and put2-/- mutations attenuate virulence in Drosophila and murine candidemia models and decrease survival in human neutrophils and whole blood. Using intravital 2-photon microscopy and label-free non-linear imaging, we visualized the initial stages of C. albicans cells infecting a kidney in real-time, directly deep in the tissue of a living mouse, and observed morphological switching of wildtype but not of put2-/- cells. Multiple members of the Candida species complex, including C. auris, are capable of using proline as a sole energy source. Our results indicate that a tailored proline metabolic network tuned to the mammalian host environment is a key feature of opportunistic fungal pathogens.


Asunto(s)
Candida albicans , Saccharomyces cerevisiae , Animales , Ratones , Humanos , Virulencia , Saccharomyces cerevisiae/genética , Prolina/metabolismo , Candida , Mamíferos
2.
Methods Mol Biol ; 2220: 189-200, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32975776

RESUMEN

Listeria monocytogenes is a model intracellular pathogen that can invade the cytoplasm of host mammalian cells. Cellular invasion can be measured using standard techniques, such as the classical gentamicin protection assay, based on the quantification of colony-forming units from lysates of infected cells. In addition, there are methods based on immunofluorescence microscopy which allow for assaying invasion in a medium- to high-throughput manner. In the following sections, we detail two different assays that can be used alone or in combination to quantify the internalization of L. monocytogenes in host cells.


Asunto(s)
Listeria monocytogenes/fisiología , Listeriosis/patología , Carga Bacteriana/métodos , Recuento de Colonia Microbiana/métodos , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Listeriosis/microbiología , Coloración y Etiquetado/métodos
3.
Pathog Dis ; 76(8)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30445439

RESUMEN

Listeria monocytogenes is a pathogenic bacterium that invades epithelial cells by activating host signaling cascades, which promote bacterial engulfment within a phagosome. The pore-forming toxin listeriolysin O (LLO), which is required for bacteria phagosomal escape, has also been associated with the activation of several signaling pathways when secreted by extracellular bacteria, including Ca2+ influx and promotion of L. monocytogenes entry. Quantitative host surfaceome analysis revealed significant quantitative remodeling of a defined set of cell surface glycoproteins upon LLO treatment, including a subset previously identified to play a role in the L. monocytogenes infection process. Our data further shows that the lysosomal-associated membrane proteins LAMP-1 and LAMP-2 are translocated to the cellular surface and those LLO-induced Ca2+ fluxes are required to trigger the surface relocalization of LAMP-1. Finally, we identify late endosomes/lysosomes as the major donor compartments of LAMP-1 upon LLO treatment and by perturbing their function, we suggest that these organelles participate in L. monocytogenes invasion.


Asunto(s)
Toxinas Bacterianas/metabolismo , Endocitosis , Células Epiteliales/microbiología , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Interacciones Huésped-Patógeno , Listeria monocytogenes/fisiología , Proteínas de la Membrana/análisis , Proteoma/análisis , Endosomas/metabolismo , Endosomas/microbiología , Células HeLa , Humanos , Listeria monocytogenes/metabolismo , Lisosomas/metabolismo , Lisosomas/microbiología
4.
J Immunol ; 201(11): 3383-3391, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30348734

RESUMEN

Toll-like receptor 4 plays an important role in the regulation of the innate and adaptive immune response. The majority of TLR4 activators currently in clinical use are derivatives of its prototypic ligand LPS. The discovery of innovative TLR4 activators has the potential of providing new therapeutic immunomodulators and adjuvants. We used computational design methods to predict and optimize a total of 53 cyclic and linear peptides targeting myeloid differentiation 2 (MD2) and cluster of differentiation 14 (CD14), both coreceptors of human TLR4. Activity of the designed peptides was first assessed using NF-κB reporter cell lines expressing either TLR4/MD2 or TLR4/CD14 receptors, then binding to CD14 and MD2 confirmed and quantified using MicroScale Thermophoresis. Finally, we incubated select peptides in human whole blood and observed their ability to induce cytokine production, either alone or in synergy with LPS. Our data demonstrate the advantage of computational design for the discovery of new TLR4 peptide activators with little structural resemblance to known ligands and indicate an efficient strategy with which to identify TLR4 targeting peptides that could be used as easy-to-produce alternatives to LPS-derived molecules in a variety of settings.


Asunto(s)
Anticuerpos Biespecíficos/genética , Sitios de Unión de Anticuerpos/genética , Receptores de Lipopolisacáridos/inmunología , Antígeno 96 de los Linfocitos/inmunología , Receptor Toll-Like 4/agonistas , Anticuerpos Biespecíficos/metabolismo , Células Cultivadas , Biología Computacional , Humanos , Ligandos , Estructura Molecular , FN-kappa B/metabolismo , Unión Proteica , Conformación Proteica , Ingeniería de Proteínas , Transducción de Señal
5.
Microorganisms ; 5(2)2017 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-28590443

RESUMEN

The human skin is commonly colonized by diverse fungal species. Some Candida species, especially C. albicans, do not only reside on the skin surface as commensals, but also cause infections by growing into the colonized tissue. However, defense mechanisms at the skin barrier level are very efficient, involving residential non-immune and immune cells as well as immune cells specifically recruited to the site of infection. Therefore, the skin is an effective barrier against fungal infection. While most studies about commensal and pathogenic interaction of Candida species with host epithelia focus on the interaction with mucosal surfaces such as the vaginal and gastrointestinal epithelia, less is known about the mechanisms underlying Candida interaction with the skin. In this review, we focus on the ecology and molecular pathogenesis of Candida species on the skin and give an overview of defense mechanisms against C. albicans in this context. We also discuss new research avenues in dermal infection, including the involvement of neurons, fibroblasts, and commensal bacteria in both mouse and human model systems.

6.
J Infect Dis ; 215(11): 1742-1752, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28368492

RESUMEN

The fungal pathogen Candida albicans colonizes basically all human epithelial surfaces, including the skin. Under certain conditions, such as immunosuppression, invasion of the epithelia occurs. Not much is known about defense mechanisms against C. albicans in subepithelial layers such as the dermis. Using immune cell-supplemented 3D skin models we defined a new role for fibroblasts in the dermis and identified a minimal set of cell types for skin protection against C. albicans invasion. Dual RNA sequencing of individual host cell populations and C. albicans revealed that dermal invasion is directly impeded by dermal fibroblasts. They are able to integrate signals from the pathogen and CD4+ T cells and shift toward an antimicrobial phenotype with broad specificity that is dependent on Toll-like receptor 2 and interleukin 1ß. These results highlight a central function of dermal fibroblasts for skin protection, opening new possibilities for treatment of infectious diseases.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Dermis , Fibroblastos , Modelos Biológicos , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Dermis/citología , Dermis/inmunología , Dermis/microbiología , Fibroblastos/citología , Fibroblastos/inmunología , Fibroblastos/microbiología , Humanos , Interleucina-1beta , Queratinocitos/citología , Queratinocitos/inmunología , Queratinocitos/microbiología , Masculino , Transducción de Señal/inmunología , Receptor Toll-Like 2
7.
Methods Mol Biol ; 1508: 439-449, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27837520

RESUMEN

Human skin is a niche for various fungal species which either colonize the surface of this tissue as commensals or, primarily under conditions of immunosuppression, invade the skin and cause infection. Here we present a method for generation of a human in vitro skin model supplemented with immune cells of choice. This model represents a complex yet amenable tool to study molecular mechanisms of host-fungi interactions at human skin.


Asunto(s)
Candidiasis/microbiología , Piel/microbiología , Animales , Candida albicans/fisiología , Candidiasis/inmunología , Técnicas de Cultivo de Célula , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Modelos Biológicos , Piel/inmunología , Piel/patología
8.
J Control Release ; 229: 163-171, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27012977

RESUMEN

Cationic antimicrobial peptides (AMPs) are part of the innate immunity, and act against a wide variety of pathogenic microorganisms by perturbation of the microorganism's plasma membrane. Although attractive for clinical applications, these agents suffer from limited stability and activity in vivo, as well as non-specific interaction with host biological membranes, leading to cytotoxic adverse effects. We hypothesized that encapsulation of AMPs within liposomes could result in reduced cytotoxicity, and with enhanced stability as well as bioactivity against herpes simplex virus 1 (HSV-1). We formulated nano-sized liposomal formulations of LL-37 and indolicidin, and their physicochemical properties, cellular uptake, in vitro cytotoxicity and antiviral efficacy have been determined. Lower cytotoxicity of LL-37 liposomes was found in comparison to indolicidin liposomes attributed to the superior physicochemical properties, and to the different degree of interaction with the liposomal membrane. The disc-like shaped LL-37 liposomes (106.8±10.1nm, shelf-life stability of >1year) were taken up more rapidly and to a significantly higher extent than the free peptide by human keratinocyte cell line (HaCaT), remained intact within the cells, followed by release of the active peptide within the cytoplasm and migration of the vesicles' lipids to the plasma membrane. LL-37 liposomes were found significantly less toxic than both the free agent and liposomal indolicidin. In the new 3D epidermis model (immortalized primary keratinocytes) liposomal LL-37 treatment (>20µM), but not free LL-37, efficiently protected the epidermis, inhibiting HSV-1 infection. This positive antiviral effect was obtained with no cytotoxicity even at very high concentrations (400µM). Thus, the antiviral activity of encapsulated LL-37 was significantly improved, expanding its therapeutic window. Liposomal LL-37 appears to be a promising delivery system for HSV therapy.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/administración & dosificación , Antivirales/administración & dosificación , Herpesvirus Humano 1/efectos de los fármacos , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Epidermis/virología , Prepucio/citología , Humanos , Queratinocitos/virología , Lípidos/química , Liposomas , Masculino , Catelicidinas
9.
mBio ; 6(3): e00598-15, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25991686

RESUMEN

UNLABELLED: Listeria monocytogenes enters nonphagocytic cells by a receptor-mediated mechanism that is dependent on a clathrin-based molecular machinery and actin rearrangements. Bacterial intra- and intercellular movements are also actin dependent and rely on the actin nucleating Arp2/3 complex, which is activated by host-derived nucleation-promoting factors downstream of the cell receptor Met during entry and by the bacterial nucleation-promoting factor ActA during comet tail formation. By genome-wide small interfering RNA (siRNA) screening for host factors involved in bacterial infection, we identified diverse cellular signaling networks and protein complexes that support or limit these processes. In addition, we could precise previously described molecular pathways involved in Listeria invasion. In particular our results show that the requirements for actin nucleators during Listeria entry and actin comet tail formation are different. Knockdown of several actin nucleators, including SPIRE2, reduced bacterial invasion while not affecting the generation of comet tails. Most interestingly, we observed that in contrast to our expectations, not all of the seven subunits of the Arp2/3 complex are required for Listeria entry into cells or actin tail formation and that the subunit requirements for each of these processes differ, highlighting a previously unsuspected versatility in Arp2/3 complex composition and function. IMPORTANCE: Listeria is a bacterial pathogen that induces its internalization within the cytoplasm of human cells and has been used for decades as a major molecular tool to manipulate cells in order to explore and discover cellular functions. We have inactivated individually, for the first time in epithelial cells, all the genes of the human genome to investigate whether each gene modifies positively or negatively the Listeria infectious process. We identified novel signaling cascades that have never been associated with Listeria infection. We have also revisited the role of the molecular complex Arp2/3 involved in the polymerization of the actin cytoskeleton, which was shown previously to be required for Listeria entry and movement inside host cells, and we demonstrate that contrary to the general dogma, some subunits of the complex are dispensable for both Listeria entry and bacterial movement.


Asunto(s)
Actinas/metabolismo , Endocitosis , Interacciones Huésped-Patógeno , Listeria monocytogenes/fisiología , Transducción de Señal , Células Epiteliales/microbiología , Silenciador del Gen , Pruebas Genéticas , Células HeLa , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
10.
Biochim Biophys Acta ; 1851(6): 911-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25241942

RESUMEN

Phosphoinositides control key cellular processes including vesicular trafficking and actin polymerization. Intracellular bacterial pathogens manipulate phosphoinositide metabolism in order to promote their uptake by target cells and to direct in some cases the biogenesis of their replication compartments. In this chapter, we review the molecular strategies that major pathogens including Listeria, Mycobacterium, Shigella, Salmonella, Legionella and Yersinia use to hijack phosphoinositides during infection. This article is part of a Special Issue entitled Phosphoinositides.


Asunto(s)
Infecciones Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Fosfatidilinositoles/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Expresión Génica , Humanos , Legionella/inmunología , Legionella/metabolismo , Listeria/inmunología , Listeria/metabolismo , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Fosfatidilinositoles/inmunología , Unión Proteica , Salmonella/inmunología , Salmonella/metabolismo , Shigella/inmunología , Shigella/metabolismo , Vesículas Transportadoras/metabolismo , Yersinia/inmunología , Yersinia/metabolismo
11.
BMC Genomics ; 15: 1162, 2014 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-25534632

RESUMEN

BACKGROUND: Large-scale RNAi screening has become an important technology for identifying genes involved in biological processes of interest. However, the quality of large-scale RNAi screening is often deteriorated by off-targets effects. In order to find statistically significant effector genes for pathogen entry, we systematically analyzed entry pathways in human host cells for eight pathogens using image-based kinome-wide siRNA screens with siRNAs from three vendors. We propose a Parallel Mixed Model (PMM) approach that simultaneously analyzes several non-identical screens performed with the same RNAi libraries. RESULTS: We show that PMM gains statistical power for hit detection due to parallel screening. PMM allows incorporating siRNA weights that can be assigned according to available information on RNAi quality. Moreover, PMM is able to estimate a sharedness score that can be used to focus follow-up efforts on generic or specific gene regulators. By fitting a PMM model to our data, we found several novel hit genes for most of the pathogens studied. CONCLUSIONS: Our results show parallel RNAi screening can improve the results of individual screens. This is currently particularly interesting when large-scale parallel datasets are becoming more and more publicly available. Our comprehensive siRNA dataset provides a public, freely available resource for further statistical and biological analyses in the high-content, high-throughput siRNA screening field.


Asunto(s)
Genómica/métodos , Interferencia de ARN , ARN Interferente Pequeño/genética , Línea Celular , Biblioteca de Genes , Genómica/normas , Ensayos Analíticos de Alto Rendimiento , Interacciones Huésped-Patógeno/genética , Humanos , Curva ROC , Reproducibilidad de los Resultados
12.
PLoS Pathog ; 10(5): e1004162, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24874089

RESUMEN

A two-step, high-throughput RNAi silencing screen was used to identify host cell factors required during human papillomavirus type 16 (HPV16) infection. Analysis of validated hits implicated a cluster of mitotic genes and revealed a previously undetermined mechanism for import of the viral DNA (vDNA) into the nucleus. In interphase cells, viruses were endocytosed, routed to the perinuclear area, and uncoated, but the vDNA failed to be imported into the nucleus. Upon nuclear envelope perforation in interphase cells HPV16 infection occured. During mitosis, the vDNA and L2 associated with host cell chromatin on the metaphase plate. Hence, we propose that HPV16 requires nuclear envelope breakdown during mitosis for access of the vDNA to the nucleoplasm. The results accentuate the value of genes found by RNAi screens for investigation of viral infections. The list of cell functions required during HPV16 infection will, moreover, provide a resource for future virus-host cell interaction studies.


Asunto(s)
Papillomavirus Humano 16 , Mitosis/fisiología , Membrana Nuclear/metabolismo , Proteínas Oncogénicas Virales/genética , Interferencia de ARN , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Células Cultivadas , ADN Viral/genética , Papillomavirus Humano 16/genética , Humanos
13.
Methods Mol Biol ; 1157: 167-78, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24792557

RESUMEN

Listeria monocytogenes is a model intracellular pathogen that can invade the cytoplasm of host mammalian cells. Cellular invasion can be measured using standard techniques such as the classical gentamicin protection assay, based on the quantification of colony-forming units from lysates of infected cells. In addition, there are methods based on immunofluorescence microscopy which allow for assaying invasion in a medium- to high-throughput manner. In the following sections we detail two different assays that can be used alone or in combination to quantify the internalization of L. monocytogenes in host cells.


Asunto(s)
Interacciones Huésped-Patógeno , Listeria monocytogenes/citología , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Listeriosis/patología , Recuento de Colonia Microbiana/métodos , Células HeLa/microbiología , Humanos , Microscopía Fluorescente/métodos , Coloración y Etiquetado/métodos
14.
mBio ; 5(2): e00969-14, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24667708

RESUMEN

For nearly 3 decades, listeriologists and immunologists have used mainly three strains of the same serovar (1/2a) to analyze the virulence of the bacterial pathogen Listeria monocytogenes. The genomes of two of these strains, EGD-e and 10403S, were released in 2001 and 2008, respectively. Here we report the genome sequence of the third reference strain, EGD, and extensive genomic and phenotypic comparisons of the three strains. Strikingly, EGD-e is genetically highly distinct from EGD (29,016 single nucleotide polymorphisms [SNPs]) and 10403S (30,296 SNPs), and is more related to serovar 1/2c than 1/2a strains. We also found that while EGD and 10403S strains are genetically very close (317 SNPs), EGD has a point mutation in the transcriptional regulator PrfA (PrfA*), leading to constitutive expression of several major virulence genes. We generated an EGD-e PrfA* mutant and showed that EGD behaves like this strain in vitro, with slower growth in broth and higher invasiveness in human cells than those of EGD-e and 10403S. In contrast, bacterial counts in blood, liver, and spleen during infection in mice revealed that EGD and 10403S are less virulent than EGD-e, which is itself less virulent than EGD-e PrfA*. Thus, constitutive expression of PrfA-regulated virulence genes does not appear to provide a significant advantage to the EGD strain during infection in vivo, highlighting the fact that in vitro invasion assays are not sufficient for evaluating the pathogenic potential of L. monocytogenes strains. Together, our results pave the way for deciphering unexplained differences or discrepancies in experiments using different L. monocytogenes strains. IMPORTANCE Over the past 3 decades, Listeria has become a model organism for host-pathogen interactions, leading to critical discoveries in a broad range of fields, including bacterial gene regulation, cell biology, and bacterial pathophysiology. Scientists studying Listeria use primarily three pathogenic strains: EGD, EGD-e, and 10403S. Despite many studies on EGD, it is the only one of the three strains whose genome has not been sequenced. Here we report the sequence of its genome and a series of important genomic and phenotypic differences between the three strains, in particular, a critical mutation in EGD's PrfA, the main regulator of Listeria virulence. Our results show that the three strains display differences which may play an important role in the virulence differences observed between the strains. Our findings will be of critical relevance to listeriologists and immunologists who have used or may use Listeria as a tool to study the pathophysiology of listeriosis and immune responses.


Asunto(s)
ADN Bacteriano/química , ADN Bacteriano/genética , Variación Genética , Genoma Bacteriano , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Animales , Sangre/microbiología , Modelos Animales de Enfermedad , Humanos , Listeriosis/microbiología , Listeriosis/patología , Hígado/microbiología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Bazo/microbiología , Virulencia
15.
J Vis Exp ; (79): e51043, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-24084755

RESUMEN

Bacterial intracellular pathogens can be conceived as molecular tools to dissect cellular signaling cascades due to their capacity to exquisitely manipulate and subvert cell functions which are required for the infection of host target tissues. Among these bacterial pathogens, Listeria monocytogenes is a Gram positive microorganism that has been used as a paradigm for intracellular parasitism in the characterization of cellular immune responses, and which has played instrumental roles in the discovery of molecular pathways controlling cytoskeletal and membrane trafficking dynamics. In this article, we describe a robust microscopical assay for the detection of late cellular infection stages of L. monocytogenes based on the fluorescent labeling of InlC, a secreted bacterial protein which accumulates in the cytoplasm of infected cells; this assay can be coupled to automated high-throughput small interfering RNA screens in order to characterize cellular signaling pathways involved in the up- or down-regulation of infection.


Asunto(s)
Proteínas Bacterianas/análisis , Proteínas Bacterianas/metabolismo , Listeria monocytogenes/patogenicidad , Proteínas Bacterianas/química , Células HeLa , Humanos , Listeria monocytogenes/química , Listeria monocytogenes/genética , Listeriosis/microbiología , Microscopía Fluorescente/métodos , ARN Interferente Pequeño/genética , Transfección
16.
Artículo en Inglés | MEDLINE | ID: mdl-23125201

RESUMEN

Listeria monocytogenes is a bacterial pathogen that promotes its internalization into host epithelial cells. Interaction between the bacterial surface molecules InlA and InlB and their cellular receptors E-cadherin and Met, respectively, triggers the recruitment of endocytic effectors, the subversion of the phosphoinositide metabolism, and the remodeling of the actin cytoskeleton that lead to bacterial engulfment. Additional bacterial surface and secreted virulence factors also contribute to entry, albeit to a lesser extent. Here we review the increasing number of signaling effectors that are reported as being subverted by L. monocytogenes during invasion of cultured cell lines. We also update the current knowledge of the early steps of in vivo cellular infection, which, as shown recently, challenges previous concepts generated from in vitro data.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Interacciones Huésped-Patógeno , Listeria monocytogenes/fisiología , Proteínas de la Membrana/metabolismo , Animales , Adhesión Bacteriana , Cadherinas/metabolismo , Membrana Celular/metabolismo , Clatrina/metabolismo , Citoesqueleto/metabolismo , Endocitosis , Humanos , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Proteínas Proto-Oncogénicas c-met/metabolismo
17.
Traffic ; 13(12): 1653-66, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22984946

RESUMEN

Invasive bacterial pathogens often target cellular proteins involved in adhesion as a first event during infection. For example, Listeria monocytogenes uses the bacterial protein InlA to interact with E-cadherin, hijack the host adherens junction (AJ) machinery and invade non-phagocytic cells by a clathrin-dependent mechanism. Here, we investigate a potential role for clathrin in cell-cell adhesion. We observed that the initial steps of AJ formation trigger the phosphorylation of clathrin, and its transient localization at forming cell-cell contacts. Furthermore, we show that clathrin serves as a hub for the recruitment of proteins that are necessary for the actin rearrangements that accompany the maturation of AJs. Using an InlA/E-cadherin chimera, we show that adherent cells expressing the chimera form AJs with cells expressing E-cadherin. We demonstrate that non-adherent cells expressing the InlA chimera, as bacteria, can be internalized by E-cadherin-expressing adherent cells. Together these results reveal that a common clathrin-mediated machinery may regulate internalization and cell adhesion and that the relative mobility of one of the interacting partners plays an important role in the commitment to either one of these processes.


Asunto(s)
Uniones Adherentes/microbiología , Clatrina/metabolismo , Endocitosis , Uniones Adherentes/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Cadherinas/metabolismo , Adhesión Celular , Perros , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Listeria monocytogenes/patogenicidad , Células de Riñón Canino Madin Darby , Fosforilación
18.
J Biol Chem ; 287(16): 13128-36, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22351770

RESUMEN

Listeria monocytogenes is a bacterial pathogen that induces its own entry into a broad range of mammalian cells through interaction of the bacterial surface protein InlB with the cellular receptor Met, promoting an actin polymerization/depolymerization process that leads to pathogen engulfment. Phosphatidylinositol bisphosphate (PI[4,5]P(2)) and trisphosphate (PI[3,4,5]P(3)) are two major phosphoinositide species that function as molecular scaffolds, recruiting cellular effectors that regulate actin dynamics during L. monocytogenes infection. Because the phosphatidylinositol 5'-phosphatase OCRL dephosphorylates PI(4,5)P(2) and to a lesser extent PI(3,4,5)P(3), we investigated whether this phosphatase modulates cell invasion by L. monocytogenes. Inactivation of OCRL by small interfering RNA (siRNA) leads to an increase in the internalization levels of L. monocytogenes in HeLa cells. Interestingly, OCRL depletion does not increase but rather decreases the surface expression of the receptor Met, suggesting that OCRL controls bacterial internalization by modulating signaling cascades downstream of Met. Immuno-fluorescence microscopy reveals that endogenous and overexpressed OCRL are present at L. monocytogenes invasion foci; live-cell imaging additionally shows that actin depolymerization coincides with EGFP-OCRL-a accumulation around invading bacteria. Together, these observations suggest that OCRL promotes actin depolymerization during L. monocytogenes infection; in agreement with this hypothesis, OCRL depletion leads to an increase in actin, PI(4,5)P(2), and PI(3,4,5)P(3) levels at bacterial internalization foci. Furthermore, in cells knocked down for OCRL, transfection of enzymatically active EGFP-OCRL-a (but not of a phosphatase-dead enzyme) decreases the levels of intracellular L. monocytogenes and of actin associated with invading bacteria. These results demonstrate that through its phosphatase activity, OCRL restricts L. monocytogenes invasion by modulating actin dynamics at bacterial internalization sites.


Asunto(s)
Actinas/metabolismo , Listeria monocytogenes/metabolismo , Listeriosis/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiología , Proteínas Bacterianas/metabolismo , Células HeLa , Humanos , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Proteínas de la Membrana/metabolismo , Fagocitosis/fisiología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , ARN Interferente Pequeño/genética , Virulencia
19.
Cell Host Microbe ; 10(1): 75-88, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21767814

RESUMEN

During natural transmission, bunyaviruses are introduced into the skin through arthropod bites, and dermal dendritic cells (DCs) are the first to encounter incoming viruses. DC-SIGN is a C-type lectin highly expressed on the surface of dermal DCs. We found that several arthropod-borne phleboviruses (Bunyaviridae), including Rift Valley fever and Uukuniemi viruses, exploit DC-SIGN to infect DCs and other DC-SIGN-expressing cells. DC-SIGN binds the virus directly via interactions with high-mannose N-glycans on the viral glycoproteins and is required for virus internalization and infection. In live cells, virus-induced clustering of cell surface DC-SIGN could be visualized. An endocytosis-defective mutant of DC-SIGN was unable to mediate virus uptake, indicating that DC-SIGN is an authentic receptor required for both attachment and endocytosis. After internalization, viruses separated from DC-SIGN and underwent trafficking to late endosomes. Our study provides real-time visualization of virus-receptor interactions on the cell surface and establishes DC-SIGN as a phlebovirus entry receptor.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/virología , Lectinas Tipo C/metabolismo , Phlebovirus/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Infecciones por Bunyaviridae/metabolismo , Moléculas de Adhesión Celular/genética , Células Dendríticas/metabolismo , Endocitosis/fisiología , Endosomas/metabolismo , Células HeLa/virología , Interacciones Huésped-Patógeno , Humanos , Lectinas Tipo C/genética , Mutación , Phlebovirus/patogenicidad , Polisacáridos/química , Polisacáridos/metabolismo , Receptores de Superficie Celular/genética , Virus Uukuniemi/metabolismo , Virus Uukuniemi/patogenicidad , Internalización del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA