Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Discov ; 4: 24, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29844921

RESUMEN

Successful inter-host transmission of most apicomplexan parasites requires the formation of infective sporozoites within the oocysts. Unlike all other infective stages that are strictly intracellular and depend on host resources, the sporozoite stage develops outside the host cells, but little is known about its self-governing metabolism. This study deployed Eimeria falciformis, a parasite infecting the mouse as its natural host, to investigate the process of phospholipid biogenesis in sporozoites. Lipidomic analyses demonstrated the occurrence of prototypical phospholipids along with abundant expression of at least two exclusive lipids, phosphatidylthreonine (PtdThr) and inositol phosphorylceramide with a phytosphingosine backbone, in sporozoites. To produce them de novo, the parasite harbors nearly the entire biogenesis network, which is an evolutionary mosaic of eukaryotic-type and prokaryotic-type enzymes. Notably, many have no phylogenetic counterpart or functional equivalent in the mammalian host. Using Toxoplasma gondii as a gene-tractable surrogate to examine Eimeria enzymes, we show a highly compartmentalized network of lipid synthesis spread primarily in the apicoplast, endoplasmic reticulum, mitochondrion, and Golgi complex. Likewise, trans-genera complementation of a Toxoplasma mutant with the PtdThr synthase from Eimeria reveals a convergent role of PtdThr in fostering the lytic cycle of coccidian parasites. Taken together, our work establishes a model of autonomous membrane biogenesis involving significant inter-organelle cooperation and lipid trafficking in sporozoites. Phylogenetic divergence of certain pathways offers attractive drug targets to block the sporulation and subsequent transmission. Not least, our results vindicate the possession of an entire de novo lipid synthesis network in a representative protist adapted to an obligate intracellular parasitic lifestyle.

2.
J Struct Biol ; 197(2): 172-180, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27345930

RESUMEN

Enveloped viruses enter their host cells by membrane fusion. The process of attachment and fusion in retroviruses is mediated by a single viral envelope glycoprotein (Env). Conformational changes of Env in the course of fusion are a focus of intense studies. Here we provide further insight into the changes occurring in retroviral Env during its initial interaction with the cell, employing murine leukemia virus (MLV) as model system. We first determined the structure of both natively membrane anchored MLV Env and MLV Env tagged with YFP in the proline rich region (PRR) by electron cryo tomography (cET) and sub-volume averaging. At a resolution of ∼20Å, native MLV Env presents as a hollow trimer (height ∼85Å, diameter ∼120Å) composed of step-shaped protomers. The major difference to the YFP-tagged protein was in regions outside of the central trimer. Next, we focused on elucidating the changes in MLV Env upon interaction with a host cell. Virus interaction with the plasma membrane occurred over a large surface and Env clustering on the binding site was observed. Sub-volume averaging did yield a low-resolution structure of Env interacting with the cell, which had lost its threefold symmetry and was elongated by ∼35Å in comparison to the unbound protein. This indicates a major rearrangement of Env upon host cell binding. At the site of virus interaction, the otherwise clearly defined bilayer structure of the host cell plasma membrane was much less evident, indicative of integral membrane protein accumulation and/or a change in membrane lipid composition.


Asunto(s)
Microscopía por Crioelectrón/métodos , Virus de la Leucemia Murina/ultraestructura , Proteínas del Envoltorio Viral/ultraestructura , Sitios de Unión , Virus de la Leucemia Murina/fisiología , Conformación Proteica
3.
Viruses ; 8(10)2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27754319

RESUMEN

The serine-threonine protein kinase encoded by US3 gene (pUS3) of alphaherpesviruses was shown to modulate actin reorganization, cell-to-cell spread, and virus egress in a number of virus species. However, the role of the US3 orthologues of equine herpesvirus type 1 and 4 (EHV-1 and EHV-4) has not yet been studied. Here, we show that US3 is not essential for virus replication in vitro. However, growth rates and plaque diameters of a US3-deleted EHV-1 and a mutant in which the catalytic active site was destroyed were significantly reduced when compared with parental and revertant viruses or a virus in which EHV-1 US3 was replaced with the corresponding EHV-4 gene. The reduced plaque sizes were consistent with accumulation of primarily enveloped virions in the perinuclear space of the US3-negative EHV-1, a phenotype that was also rescued by the EHV-4 orthologue. Furthermore, actin stress fiber disassembly was significantly more pronounced in cells infected with parental EHV-1, revertant, or the recombinant EHV-1 expressing EHV-4 US3. Finally, we observed that deletion of US3 in EHV-1 did not affect the expression of adhesion molecules on the surface of infected cells.


Asunto(s)
Actinas/metabolismo , Herpesvirus Équido 1/enzimología , Herpesvirus Équido 1/fisiología , Interacciones Huésped-Patógeno , Proteínas Serina-Treonina Quinasas/metabolismo , Liberación del Virus , Línea Celular , Técnicas de Inactivación de Genes , Herpesvirus Équido 1/crecimiento & desarrollo , Humanos , Proteínas Serina-Treonina Quinasas/genética , Ensayo de Placa Viral , Proteínas Virales/genética , Proteínas Virales/metabolismo
4.
J Virol ; 88(21): 12802-15, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25165105

RESUMEN

UNLABELLED: Equine herpesvirus type 1 (EHV-1) downregulates cell surface expression of major histocompatibility complex class I (MHC-I) in infected cells. We have previously shown that pUL56 encoded by the EHV-1 ORF1 gene regulates the process (G. Ma, S. Feineis, N. Osterrieder, and G. R. Van de Walle, J. Virol. 86:3554-3563, 2012, doi:http://dx.doi.org/10.1128/JVI.06994-11). Here, we report that cell surface MHC-I in EHV-1-infected cells is internalized and degraded in the lysosomal compartment in a pUL56-dependent fashion. pUL56-induced MHC-I endocytosis required dynamin and tyrosine kinase but was independent of clathrin and caveolin-1, the main constituents of the clathrin- and raft/caveola-mediated endocytosis pathways, respectively. Downregulation of cell surface MHC-I was significantly inhibited by the ubiquitin-activating enzyme E1 inhibitor PYR41, indicating that ubiquitination is essential for the process. Finally, we show that downregulation is not specific for MHC-I and that other molecules, including CD46 and CD63, are also removed from the cell surface in a pUL56-dependent fashion. IMPORTANCE: We show that alphaherpesvirus induces MHC-I downregulation through endocytosis, which is mediated by pUL56. The dynamin-dependent endocytic pathway is responsible for MHC-I internalization in infected cells. Furthermore, we discovered that this endocytic process can be disrupted by the inhibiting ubiquitin-activating E1 enzyme, which is indispensable for ubiquitination. Finally, pUL56 action extends to a number of cell surface molecules that are significant for host immunity. Therefore, the protein may exert a more general immunomodulatory effect.


Asunto(s)
Dinaminas/metabolismo , Endocitosis , Herpesvirus Équido 1/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Proteínas Virales/metabolismo , Animales , Línea Celular , Regulación hacia Abajo , Fibroblastos/inmunología , Fibroblastos/virología , Caballos , Humanos
5.
Nanomedicine ; 10(5): 981-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24589930

RESUMEN

Macrophages represent an important cellular target of HIV-1. Interestingly, they are also believed to play a potential role counteracting its infection. However, HIV-1 is known to impair macrophage immune functions such as antibody-mediated phagocytosis. Here, we present immunoliposomes that can bind HIV-1 virus-like particles (HIV-VLPs) while being specifically phagocytosed by macrophages, thus allowing the co-internalization of HIV-VLPs. These liposomes are decorated with anti-Env antibodies and contain phosphatidylserine (PS). PS mediates liposome internalization by macrophages via a mechanism not affected by HIV-1. Hence, PS-liposomes mimic apoptotic cells and are internalized into the macrophages due to specific recognition, carrying the previously bound HIV-VLPs. With a combination of flow cytometry, confocal live-cell imaging and electron microscopy we demonstrate that the PS-immunoliposomes presented here are able to elicit efficient HIV-VLPs phagocytosis by macrophages and might represent a new nanotechnological approach to enhance HIV-1 antigen presentation and reduce the ongoing inflammation processes. FROM THE CLINICAL EDITOR: This team of authors demonstrate that specific phosphatidylserin immunoliposomes are able to elicit efficient phagocytosis of HIV-virus-like particle by macrophages and might represent a new nanomedicine approach to enhance HIV-1 antigen presentation and reduce ongoing inflammation processes.


Asunto(s)
Anticuerpos/química , Liposomas/química , Liposomas/farmacología , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Fosfatidilserinas/química , Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Línea Celular , Productos del Gen env/inmunología , Infecciones por VIH , Humanos , Microscopía Fluorescente
6.
Traffic ; 15(4): 362-82, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24423236

RESUMEN

For membrane-bound intracellular pathogens, the surrounding vacuole is the portal of communication with the host cell. The parasitophorous vacuole (PV) harboring intrahepatocytic Plasmodium parasites satisfies the parasites' needs of nutrition and protection from host defenses to allow the rapid parasite growth that occurs during the liver stage of infection. In this study, we visualized the PV membrane (PVM) and the associated tubovesicular network (TVN) through fluorescent tagging of two PVM-resident Plasmodium berghei proteins, UIS4 and IBIS1. This strategy revealed previously unrecognized dynamics with which these membranes extend throughout the host cell. We observed dynamic vesicles, elongated clusters of membranes and long tubules that rapidly extend and contract from the PVM in a microtubule-dependent manner. Live microscopy, correlative light-electron microscopy and fluorescent recovery after photobleaching enabled a detailed characterization of these membranous features, including velocities, the distribution of UIS4 and IBIS1, and the connectivity of PVM and TVN. Labeling of host cell compartments revealed association of late endosomes and lysosomes with the elongated membrane clusters. Moreover, the signature host autophagosome protein LC3 was recruited to the PVM and TVN and colocalized with UIS4. Together, our data demonstrate that the membranes surrounding intrahepatic Plasmodium are involved in active remodeling of host cells.


Asunto(s)
Hígado/parasitología , Plasmodium/metabolismo , Animales , Membrana Celular/metabolismo , Interacciones Huésped-Parásitos , Plasmodium/patogenicidad
7.
Plant Cell Physiol ; 55(3): 645-57, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24449654

RESUMEN

Arabidopsis HEMA1 and HEMA2 encode glutamyl-tRNA reductase (GluTR) 1 and 2, the two isoforms of the initial enzyme of tetrapyrrole biosynthesis. HEMA1 is dominantly expressed in photosynthetic tissue, while HEMA2 shows low constitutive expression and is induced upon stress treatments. We introduce a new HEMA1 knockout mutant which grows only heterotrophically on MS (Murashige and Skoog) medium at low light, indicating that the remaining GluTR2 does not sufficiently compensate for the extensive needs of metabolic precursors for Chl. While hema1 accumulates low amounts of Chl, it contains more than half of the wild-type heme content. The functional diversity of the two GluTR isoforms was analyzed by means of complementation studies of the hema1 mutant by expression of pHEMA1::HEMA2 and p35S::HEMA1, respectively. Expression of both transgenes complements hema1, indicating that GluTR2 can likewise be involved in the synthesis of Chl and is not exclusively assigned to heme synthesis. In comparison with p35S::HEMA1-complemented hema1 and the wild type, GluTR2 expression under control of the HEMA1 promoter (pHEMA1) in pHEMA1::HEMA2-complemented hema1 mutants causes elevated protochlorophyllide levels under extended dark periods as well as in short-day-grown adult plants, resulting in the formation of necrotic leaf tissue. Although both GluTR isoforms have similar activity and contribute to 5-aminolevulinic acid synthesis for adequate accumulation of Chl and heme, it is proposed that the two proteins experience a different post-translational control in darkness and light. While GluTR2 continues 5-aminolevulinic acid synthesis in darkness, GluTR1 is efficiently inactivated by the interaction with the FLU (FLUORESCENT) protein, thereby preventing an accumulation of protochlorophyllide.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Arabidopsis/metabolismo , Aldehído Oxidorreductasas/genética , Ácido Aminolevulínico/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Clorofila/metabolismo , Regulación de la Expresión Génica de las Plantas/genética , Regulación de la Expresión Génica de las Plantas/fisiología , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/metabolismo , Protoclorofilida/metabolismo , Tetrapirroles/metabolismo
8.
J Virol ; 87(10): 5937-48, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23514881

RESUMEN

Herpesviruses enter cells either by direct fusion at the plasma membrane or from within endosomes, depending on the cell type and receptor(s). We investigated two closely related herpesviruses of horses, equine herpesvirus type 1 (EHV-1) and EHV-4, for which the cellular and viral determinants routing virus entry are unknown. We show that EHV-1 enters equine epithelial cells via direct fusion at the plasma membrane, while EHV-4 does so via an endocytic pathway, which is dependent on dynamin II, cholesterol, caveolin 1, and tyrosine kinase activity. Exchange of glycoprotein H (gH) between EHV-1 and EHV-4 resulted in rerouting of EHV-1 to the endocytic pathway, as did blocking of α4ß1 integrins on the cell surface. Furthermore, a point mutation in the SDI integrin-binding motif of EHV-1 gH also directed EHV-1 to the endocytic pathway. Cumulatively, we show that viral gH and cellular α4ß1 integrins are important determinants in the choice of alphaherpesvirus cellular entry pathways.


Asunto(s)
Herpesvirus Équido 1/fisiología , Herpesvirus Équido 4/fisiología , Interacciones Huésped-Patógeno , Integrina alfa4beta1/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Línea Celular , Caballos
9.
Virol J ; 9: 299, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23194647

RESUMEN

BACKGROUND: Epithelio- and endotheliotropic viruses often exert polarized entry and release that may be responsible for viral spread and dissemination. Hantaviruses, mostly rodent-borne members of the Bunyaviridae family infect epithelial and endothelial cells of different organs leading to organ dysfunction or even failure. Endothelial and renal epithelial cells belong to the target cells of Old World hantavirus. Therefore, we examined the release of hantaviruses in several renal epithelial cell culture models. We used Vero cells that are commonly used in hantavirus studies and primary human renal epithelial cells (HREpC). In addition, we analyzed MDCKII cells, an epithelial cell line of a dog kidney, which represents a widely accepted in vitro model of polarized monolayers for their permissiveness for hantavirus infection. RESULTS: Vero C1008 and primary HREpCs were grown on porous-support filter inserts for polarization. Monolayers were infected with hantavirus Hantaan (HTNV) and Puumala (PUUV) virus. Supernatants from the apical and basolateral chamber of infected cells were analyzed for the presence of infectious particles by re-infection of Vero cells. Viral antigen and infectious particles of HTNV and PUUV were exclusively detected in supernatants collected from the apical chamber of infected Vero C1008 cells and HREpCs. MDCKII cells were permissive for hantavirus infection and polarized MDCKII cells released infectious hantaviral particles from the apical surface corresponding to the results of Vero and primary human epithelial cells. CONCLUSIONS: Pathogenic Old World hantaviruses are released from the apical surface of different polarized renal epithelial cells. We characterized MDCKII cells as a suitable polarized cell culture model for hantavirus infection studies.


Asunto(s)
Células Epiteliales/virología , Túbulos Renales/citología , Túbulos Renales/virología , Orthohantavirus/fisiología , Liberación del Virus , Animales , Antígenos CD55/metabolismo , Línea Celular , Chlorocebus aethiops , Perros , Virus Hantaan/fisiología , Humanos , Integrina alfaVbeta3/metabolismo , Virus Puumala/fisiología , Receptores Virales/metabolismo , Células Vero , Internalización del Virus , Replicación Viral
10.
J Biol Chem ; 287(24): 20197-207, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22535959

RESUMEN

The obligate intracellular apicomplexan parasites, e.g. Toxoplasma gondii and Plasmodium species, induce an IFNγ-driven induction of host indoleamine 2,3-dioxygenase (IDO), the first and rate-limiting enzyme of tryptophan catabolism in the kynurenine pathway. Induction of IDO1 supposedly depletes cellular levels of tryptophan in host cells, which is proposed to inhibit the in vitro growth of auxotrophic pathogens. In vivo function of IDO during infections, however, is not clear, let alone controversial. We show that Eimeria falciformis, an apicomplexan parasite infecting the mouse caecum, induces IDO1 in the epithelial cells of the organ, and the enzyme expression coincides with the parasite development. The absence or inhibition of IDO1/2 and of two downstream enzymes in infected animals is detrimental to the Eimeria growth. The reduced parasite yield is not due to a lack of an immunosuppressive effect of IDO1 in the parasitized IDO1(-/-) or inhibitor-treated mice because they did not show an accentuated Th1 and IFNγ response. Noticeably, the parasite development is entirely rescued by xanthurenic acid, a by-product of tryptophan catabolism inducing exflagellation in male gametes of Plasmodium in the mosquito mid-gut. Our data demonstrate a conceptual subversion of the host defense (IFNγ, IDO) by an intracellular pathogen for progression of its natural life cycle. Besides, we show utility of E. falciformis, a monoxenous parasite of a well appreciated host, i.e. mouse, to identify in vivo factors underlying the parasite-host interactions.


Asunto(s)
Coccidiosis/metabolismo , Eimeria/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Células TH1/metabolismo , Triptófano/metabolismo , Animales , Coccidiosis/genética , Coccidiosis/inmunología , Culicidae/parasitología , Eimeria/genética , Eimeria/inmunología , Inhibidores Enzimáticos/farmacología , Hipolipemiantes/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células TH1/inmunología , Triptófano/genética , Triptófano/inmunología , Xanturenatos/farmacología
11.
Mol Microbiol ; 83(6): 1229-43, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22329949

RESUMEN

The importance of pathogen-induced host cell remodelling has been well established for red blood cell infection by the human malaria parasite Plasmodium falciparum. Exported parasite-encoded proteins, which often possess a signature motif, termed Plasmodium export element (PEXEL) or host-targeting (HT) signal, are critical for the extensive red blood cell modifications. To what extent remodelling of erythrocyte membranes also occurs in non-primate hosts and whether it is in fact a hallmark of all mammalian Plasmodium parasites remains elusive. Here we characterize a novel Plasmodium berghei PEXEL/HT-containing protein, which we term IBIS1. Temporal expression and spatial localization determined by fluorescent tagging revealed the presence of IBIS1 at the parasite/host interface during both liver and blood stages of infection. Targeted deletion of the IBIS1 protein revealed a mild impairment of intra-erythrocytic growth indicating a role for these structures in the rapid expansion of the parasite population in the blood in vivo. In red blood cells, the protein localizes to dynamic, punctate structures external to the parasite. Biochemical and microscopic data revealed that these intra-erythrocytic P. berghei-induced structures (IBIS) are membranous indicating that P. berghei, like P. falciparum, creates an intracellular membranous network in infected red blood cells.


Asunto(s)
Membrana Celular/química , Eritrocitos/química , Malaria/parasitología , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Membrana Celular/parasitología , Eritrocitos/parasitología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/química , Plasmodium berghei/genética , Transporte de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética
12.
PLoS One ; 5(10): e13172, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20949027

RESUMEN

RNA interference (RNAi) has been considered as an efficient therapeutic approach against the human immunodeficiency virus type 1 (HIV-1). However, to establish a durable inhibition of HIV-1, multiple effective short hairpin RNAs (shRNAs) need to be stably expressed to prevent the emergence of viral escape variants. In this study, we engineered a randomized lentiviral H1-promoter driven shRNA-library against the viral genome. Potent HIV-1 specific shRNAs were selected by ganciclovir treatment of cell lines stably expressing the cDNA of Herpes Simplex Virus thymidine kinase (HSV-TK) fused to HIV-1 nucleotide sequences. More than 50% of 200 selected shRNAs inhibited an HIV-1 based luciferase reporter assay by more than 70%. Stable expression of some of those shRNAs in an HIV-1 permissive HeLa cell line inhibited infection of wild-type HIV-1 by more than 90%. The combination of a randomized shRNA-library directed against HIV-1 with a live cell selection procedure yielded non-toxic and highly efficient HIV-1 specific inhibitory sequences that could serve as valuable candidates for gene therapy studies.


Asunto(s)
VIH-1/genética , ARN Viral/genética , Secuencia de Bases , Northern Blotting , Western Blotting , Cartilla de ADN , ADN Complementario , Células HeLa , Humanos , Regiones Promotoras Genéticas , Interferencia de ARN
13.
Cytometry A ; 77(1): 52-63, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19899129

RESUMEN

The actin cytoskeleton modulates a large variety of physiological and disease-related processes in the cell. For example, actin has been shown to be a crucial host factor for successful infection by HIV-1, but the underlying mechanistic details are still unknown. Automated approaches open up the perspective to clarify such an issue by processing many samples in a high-throughput manner. To analyze the alterations in the actin cytoskeleton within an automated setting, large-scale image acquisition and analysis were established for JC-53 cells stained for actin. As a quantitative measure in such an automated approach, we suggest a parameter called image coherency. We successfully benchmarked our analysis by calculating coherency for both a biophysical model of the actin cytoskeleton and for cells whose actin architecture had been disturbed pharmacologically by latrunculin B or cytochalasin D. We then tested the influence of HIV-1 infection on actin coherency, but observed no significant differences between uninfected and infected cells.


Asunto(s)
Actinas/ultraestructura , Citoesqueleto/ultraestructura , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Células Cultivadas , Citocalasina D/farmacología , Citoesqueleto/efectos de los fármacos , Infecciones por VIH , VIH-1/efectos de los fármacos , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Biológicos , Tiazolidinas/farmacología
14.
Biotechnol J ; 5(1): 39-49, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20013946

RESUMEN

RNA interference (RNAi) has emerged as a powerful technique for studying loss-of-function phenotypes by specific down-regulation of gene expression, allowing the investigation of virus-host interactions by large-scale high-throughput RNAi screens. Here we present a robust and sensitive small interfering RNA screening platform consisting of an experimental setup, single-cell image and statistical analysis as well as bioinformatics. The workflow has been established to elucidate host gene functions exploited by viruses, monitoring both suppression and enhancement of viral replication simultaneously by fluorescence microscopy. The platform comprises a two-stage procedure in which potential host factors are first identified in a primary screen and afterwards re-tested in a validation screen to confirm true positive hits. Subsequent bioinformatics allows the identification of cellular genes participating in metabolic pathways and cellular networks utilised by viruses for efficient infection. Our workflow has been used to investigate host factor usage by the human immunodeficiency virus-1 (HIV-1), but can also be adapted to other viruses. Importantly, we expect that the description of the platform will guide further screening approaches for virus-host interactions. The ViroQuant-CellNetworks RNAi Screening core facility is an integral part of the recently founded BioQuant centre for systems biology at the University of Heidelberg and will provide service to external users in the near future.


Asunto(s)
Biología Computacional/métodos , VIH-1/genética , Interferencia de ARN , ARN Mensajero/genética , Análisis de Secuencia de ARN/métodos , Replicación Viral/genética , Células HeLa , Humanos
15.
Retrovirology ; 6: 84, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19765276

RESUMEN

BACKGROUND: Most retroviruses enter their host cells by fusing the viral envelope with the plasma membrane. Although the protein machinery promoting fusion has been characterized extensively, the dynamics of the process are largely unknown. RESULTS: We generated human immunodeficiency virus-1 (HIV-1) particles pseudotyped with the envelope (Env) protein of ecotropic murine leukemia virus eMLV to study retrovirus entry at the plasma membrane using live-cell microscopy. This Env protein mediates highly efficient pH independent fusion at the cell surface and can be functionally tagged with a fluorescent protein. To detect fusion events, double labeled particles carrying one fluorophor in Env and the other in the matrix (MA) domain of Gag were generated and characterized. Fusion events were defined as loss of Env signal after virus-cell contact. Single particle tracking of >20,000 individual traces in two color channels recorded 28 events of color separation, where particles lost the Env protein, with the MA layer remaining stable at least for a short period. Fourty-five events were detected where both colors were lost simultaneously. Importantly, the first type of event was never observed when particles were pseudotyped with a non-fusogenic Env. CONCLUSION: These results reveal rapid retroviral fusion at the plasma membrane and permit studies of the immediate post-fusion events.


Asunto(s)
VIH-1/fisiología , Microscopía/métodos , Internalización del Virus , Animales , Antígenos VIH/genética , Antígenos VIH/metabolismo , VIH-1/genética , Humanos , Virus de la Leucemia Murina/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Coloración y Etiquetado/métodos , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
16.
Biotechnol J ; 4(6): 826-37, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19507150

RESUMEN

Over millions of years pathogens have coevolved with their respective hosts utilizing host cell functions for survival and replication. Despite remarkable progress in developing antibiotics and vaccination strategies in the last century, infectious diseases still remain a severe threat to human health. Meanwhile, genomic research offers a new era of data-generating platforms that will dramatically enhance our knowledge of pathogens and the diseases they cause. Improvements in gene knockdown studies by RNA interference (RNAi) combined with recent developments in instrumentation and image analysis enable the use of high-throughput screening approaches to elucidate host gene functions exploited by pathogens. Although only a few RNAi-based screens focusing on host genes have been reported so far, these studies have already uncovered hundreds of genes not previously known to be involved in pathogen infection. This review describes recent progress in RNAi screening approaches, highlighting both the limitations and the tremendous potential of RNAi-based screens for the identification of essential host cell factors during infection.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Genómica/métodos , Interacciones Huésped-Patógeno/genética , Interferencia de ARN , Animales , Humanos , Infecciones/microbiología , Infecciones/parasitología , ARN Interferente Pequeño/genética
18.
Nat Cell Biol ; 9(3): 310-5, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17293854

RESUMEN

The spread of retroviruses between cells is estimated to be 2-3 orders of magnitude more efficient when cells can physically interact with each other. The underlying mechanism is largely unknown, but transfer is believed to occur through large-surface interfaces, called virological or infectious synapses. Here, we report the direct visualization of cell-to-cell transmission of retroviruses in living cells. Our results reveal a mechanism of virus transport from infected to non-infected cells, involving thin filopodial bridges. These filopodia originate from non-infected cells and interact, through their tips, with infected cells. A strong association of the viral envelope glycoprotein (Env) in an infected cell with the receptor molecules in a target cell generates a stable bridge. Viruses then move along the outer surface of the filopodial bridge toward the target cell. Our data suggest that retroviruses spread by exploiting an inherent ability of filopodia to transport ligands from cell to cell.


Asunto(s)
Comunicación Celular/fisiología , Células Eucariotas/virología , Seudópodos/virología , Retroviridae/fisiología , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Antígenos CD4/genética , Antígenos CD4/metabolismo , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Endocitosis/fisiología , Células Eucariotas/metabolismo , VIH-1/fisiología , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Mutación , Seudópodos/ultraestructura , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
19.
J Biol Chem ; 281(28): 19618-30, 2006 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-16687395

RESUMEN

The Nef protein is a key determinant of human immunodeficiency virus (HIV) pathogenicity that, among other activities, sensitizes T-lymphocytes for optimal virus production. The initial events by which Nef modulates the T-cell receptor (TCR) cascade are poorly understood. TCR engagement triggers actin rearrangements that control receptor clustering for signal initiation and dynamic organization of signaling protein complexes to form an immunological synapse. Here we report that Nef potently interferes with cell spreading and formation of actin-rich circumferential rings in T-lymphocytes upon surface-supported TCR stimulation. These effects were conserved among Nef proteins from different lentiviruses and occurred in HIV-1-infected primary human T-lymphocytes. This novel Nef activity critically depended on its Src homology 3 domain binding motif and required efficient association with Pak2 activity. Notably, whereas overall signaling microcluster formation immediately following TCR engagement occurred normally in Nef-expressing cells, the viral protein inhibited the concomitant activation of the actin organizer N-Wasp. During the subsequent maturation phase of the stimulatory contact, Nef interfered with the translocation of N-Wasp to the cell periphery, the overall induction of tyrosine phosphorylation, and the selective recruitment of phosphorylated LAT to stimulatory contacts. Consistent with such a critical role of N-Wasp in this process, Nef also blocked morphological changes induced by the known N-Wasp regulators Rac1 and Cdc42. Together, our results demonstrate that Nef alters both the amount and composition of signaling microclusters. We propose modulation of actin dynamics as an important mechanism for Nef-induced alterations of TCR signaling.


Asunto(s)
Productos del Gen nef/fisiología , VIH-1/metabolismo , Linfocitos T/metabolismo , Linfocitos T/virología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Actinas/química , Productos del Gen nef/metabolismo , Humanos , Células Jurkat , Fosforilación , Unión Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Sinapsis/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA