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












Base de datos
Intervalo de año de publicación
1.
Science ; 370(6518): 856-860, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33082293

RESUMEN

The causative agent of coronavirus disease 2019 (COVID-19) is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). For many viruses, tissue tropism is determined by the availability of virus receptors and entry cofactors on the surface of host cells. In this study, we found that neuropilin-1 (NRP1), known to bind furin-cleaved substrates, significantly potentiates SARS-CoV-2 infectivity, an effect blocked by a monoclonal blocking antibody against NRP1. A SARS-CoV-2 mutant with an altered furin cleavage site did not depend on NRP1 for infectivity. Pathological analysis of olfactory epithelium obtained from human COVID-19 autopsies revealed that SARS-CoV-2 infected NRP1-positive cells facing the nasal cavity. Our data provide insight into SARS-CoV-2 cell infectivity and define a potential target for antiviral intervention.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/virología , Neuropilina-1/metabolismo , Neumonía Viral/virología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Monoclonales/inmunología , Betacoronavirus/genética , COVID-19 , Células CACO-2 , Femenino , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Pulmón/metabolismo , Masculino , Nanopartículas del Metal , Ratones , Ratones Endogámicos C57BL , Mutación , Neuropilina-1/química , Neuropilina-1/genética , Neuropilina-1/inmunología , Neuropilina-2/metabolismo , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/virología , Pandemias , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica , Dominios Proteicos , Mucosa Respiratoria/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/química
2.
Science ; 370(6518): 861-865, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33082294

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), uses the viral spike (S) protein for host cell attachment and entry. The host protease furin cleaves the full-length precursor S glycoprotein into two associated polypeptides: S1 and S2. Cleavage of S generates a polybasic Arg-Arg-Ala-Arg carboxyl-terminal sequence on S1, which conforms to a C-end rule (CendR) motif that binds to cell surface neuropilin-1 (NRP1) and NRP2 receptors. We used x-ray crystallography and biochemical approaches to show that the S1 CendR motif directly bound NRP1. Blocking this interaction by RNA interference or selective inhibitors reduced SARS-CoV-2 entry and infectivity in cell culture. NRP1 thus serves as a host factor for SARS-CoV-2 infection and may potentially provide a therapeutic target for COVID-19.


Asunto(s)
Betacoronavirus/fisiología , Neuropilina-1/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Enzima Convertidora de Angiotensina 2 , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , COVID-19 , Células CACO-2 , Infecciones por Coronavirus/virología , Cristalografía por Rayos X , Furina/metabolismo , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/química , Neuropilina-1/genética , Pandemias , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética
3.
Annu Rev Biochem ; 89: 21-43, 2020 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-32569520

RESUMEN

My coworkers and I have used animal viruses and their interaction with host cells to investigate cellular processes difficult to study by other means. This approach has allowed us to branch out in many directions, including membrane protein characterization, endocytosis, secretion, protein folding, quality control, and glycobiology. At the same time, our aim has been to employ cell biological approaches to expand the fundamental understanding of animal viruses and their pathogenic lifestyles. We have studied mechanisms of host cell entry and the uncoating of incoming viruses as well as the synthesis, folding, maturation, and intracellular movement of viral proteins and molecular assemblies. I have had the privilege to work in institutions in four different countries. The early years in Finland (the University of Helsinki) were followed by 6 years in Germany (European Molecular Biology Laboratory), 16 years in the United States (Yale School of Medicine), and 16 years in Switzerland (ETH Zurich).


Asunto(s)
Calnexina/genética , Calreticulina/genética , Interacciones Huésped-Patógeno/genética , Virus de la Influenza A/genética , Picornaviridae/genética , Proteínas Virales/genética , Virología/historia , Animales , Calnexina/química , Calnexina/metabolismo , Calreticulina/química , Calreticulina/metabolismo , Línea Celular , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Endosomas/metabolismo , Endosomas/virología , Regulación de la Expresión Génica , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Virus de la Influenza A/metabolismo , Picornaviridae/metabolismo , Pliegue de Proteína , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/metabolismo , Vesiculovirus/genética , Vesiculovirus/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Internalización del Virus
4.
Nat Microbiol ; 4(4): 578-586, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30692667

RESUMEN

Influenza A virus is a pathogen of great medical impact. To develop novel antiviral strategies, it is essential to understand the molecular aspects of virus-host cell interactions in detail. During entry, the viral ribonucleoproteins (vRNPs) that carry the RNA genome must be released from the incoming particle before they can enter the nucleus for replication. The uncoating process is facilitated by histone deacetylase 6 (ref.1). However, the precise mechanism of shell opening and vRNP debundling is unknown. Here, we show that transportin 1, a member of the importin-ß family proteins, binds to a PY-NLS2 sequence motif close to the amino terminus of matrix protein (M1) exposed during acid priming of the viral core. It promotes the removal of M1 and induces disassembly of vRNP bundles. Next, the vRNPs interact with importin-α/ß and enter the nucleus. Thus, influenza A virus uses dual importin-ßs for distinct steps in host cell entry.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , beta Carioferinas/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/virología , Humanos , Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/virología , Ribonucleoproteínas/genética , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/genética , Replicación Viral
5.
J Mol Biol ; 430(13): 1853-1862, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29709571

RESUMEN

Research over a period of more than half a century has provided a reasonably accurate picture of mechanisms involved in animal virus entry into their host cells. Successive steps in entry include binding to receptors, endocytosis, passage through one or more membranes, targeting to specific sites within the cell, and uncoating of the genome. For some viruses, the molecular interactions are known in great detail. However, as more viruses are analyzed, and as the focus shifts from tissue culture to in vivo experiments, it is evident that viruses display considerable redundancy and flexibility in receptor usage, endocytic mechanism, location of penetration, and uncoating mechanism. For many viruses, the picture is still elusive because the interactions that they engage in rely on sophisticated adaptation to complex cellular functions and defense mechanisms.


Asunto(s)
Fenómenos Fisiológicos de los Virus , Animales , Endocitosis , Humanos , Polisacáridos/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus
7.
Proc Natl Acad Sci U S A ; 113(50): E8069-E8078, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27834731

RESUMEN

Caveolae are invaginated plasma membrane domains involved in mechanosensing, signaling, endocytosis, and membrane homeostasis. Oligomers of membrane-embedded caveolins and peripherally attached cavins form the caveolar coat whose structure has remained elusive. Here, purified Cavin1 60S complexes were analyzed structurally in solution and after liposome reconstitution by electron cryotomography. Cavin1 adopted a flexible, net-like protein mesh able to form polyhedral lattices on phosphatidylserine-containing vesicles. Mutating the two coiled-coil domains in Cavin1 revealed that they mediate distinct assembly steps during 60S complex formation. The organization of the cavin coat corresponded to a polyhedral nano-net held together by coiled-coil segments. Positive residues around the C-terminal coiled-coil domain were required for membrane binding. Purified caveolin 8S oligomers assumed disc-shaped arrangements of sizes that are consistent with the discs occupying the faces in the caveolar polyhedra. Polygonal caveolar membrane profiles were revealed in tomograms of native caveolae inside cells. We propose a model with a regular dodecahedron as structural basis for the caveolae architecture.


Asunto(s)
Caveolas/química , Caveolas/metabolismo , Caveolina 1/química , Caveolina 1/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Animales , Caveolas/ultraestructura , Células HEK293 , Células HeLa , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Electrónica de Transmisión , Modelos Biológicos , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Dominios Proteicos , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eliminación de Secuencia
8.
Annu Rev Cell Dev Biol ; 32: 197-222, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27298089

RESUMEN

Transport of newly synthesized proteins from the endoplasmic reticulum (ER) to the Golgi complex is highly selective. As a general rule, such transport is limited to soluble and membrane-associated secretory proteins that have reached properly folded and assembled conformations. To secure the efficiency, fidelity, and control of this crucial transport step, cells use a combination of mechanisms. The mechanisms are based on selective retention of proteins in the ER to prevent uptake into transport vesicles, on selective capture of proteins in COPII carrier vesicles, on inclusion of proteins in these vesicles by default as part of fluid and membrane bulk flow, and on selective retrieval of proteins from post-ER compartments by retrograde vesicle transport.


Asunto(s)
Vías Secretoras , Animales , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Humanos , Transporte de Proteínas , Vesículas Transportadoras/metabolismo
9.
PLoS Pathog ; 12(3): e1005508, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27030971

RESUMEN

Phosphoinositide-3-kinases have been shown to be involved in influenza virus pathogenesis. They are targeted directly by virus proteins and are essential for efficient viral replication in infected lung epithelial cells. However, to date the role of PI3K signaling in influenza infection in vivo has not been thoroughly addressed. Here we show that one of the PI3K subunits, p110γ, is in fact critically required for mediating the host's antiviral response. PI3Kγ deficient animals exhibit a delayed viral clearance and increased morbidity during respiratory infection with influenza virus. We demonstrate that p110γ is required for the generation and maintenance of potent antiviral CD8+ T cell responses through the developmental regulation of pulmonary cross-presenting CD103+ dendritic cells under homeostatic and inflammatory conditions. The defect in lung dendritic cells leads to deficient CD8+ T cell priming, which is associated with higher viral titers and more severe disease course during the infection. We thus identify PI3Kγ as a novel key host protective factor in influenza virus infection and shed light on an unappreciated layer of complexity concerning the role of PI3K signaling in this context.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/citología , Pulmón/virología , Infecciones por Orthomyxoviridae/virología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Linfocitos T CD8-positivos/citología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Células Epiteliales/virología , Pulmón/inmunología , Activación de Linfocitos/inmunología , Ratones , Replicación Viral/fisiología
10.
J Vis Exp ; (109): e53909, 2016 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-27077390

RESUMEN

Acid-triggered molecular processes closely control cell entry of many viruses that enter through the endocytic system. In the case of influenza A virus (IAV), virus fusion with the endosomal membrane as well as the subsequent disassembly of the viral capsid, called uncoating, is governed by the ionic conditions inside endocytic vesicles. The early steps in the virus life cycle are hard to study because endosomes cannot be directly accessed experimentally, creating the need for an in vitro approach. Here, we describe a method based on velocity gradient centrifugation of purified virions through a two-layer glycerol gradient, which enables analysis of the IAV core and its stability. The gradient contains a non-ionic detergent (NP-40) in its lower layer to remove the viral membrane by solubilization as the virus sediments toward the bottom. At neutral pH, viral cores are pelleted as stable structures. The major core components, matrix protein (M1) and the viral ribonucleoproteins (vRNPs), can be clearly identified in the pellet fraction by SDS-PAGE. Decreasing the pH to 6.0 or lower in the bottom layer selectively removes M1 from the pellet followed by release of vRNPs at more acidic conditions. Viral protein bands on Coomassie-stained gels can be subjected to densitometric quantification to monitor intermediate states of IAV disassembly. Besides pH, other factors that influence viral core stability can be assessed, such as salt concentration and putative viral uncoating factors, simply by modifying the detergent-containing glycerol layer accordingly. Taken together, the presented technique allows highly reproducible and quantitative analysis of viral uncoating in vitro. It can be applied to other enveloped viruses that undergo complex uncoating processes.


Asunto(s)
Virus de la Influenza A , Virión/aislamiento & purificación , Virología/métodos , Internalización del Virus , Cápside , Electroforesis en Gel de Poliacrilamida , Proteínas Virales/aislamiento & purificación
11.
Elife ; 5: e13841, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-27008177

RESUMEN

Cullin-3 (CUL3)-based ubiquitin ligases regulate endosome maturation and trafficking of endocytic cargo to lysosomes in mammalian cells. Here, we report that these functions depend on SPOPL, a substrate-specific CUL3 adaptor. We find that SPOPL associates with endosomes and is required for both the formation of multivesicular bodies (MVBs) and the endocytic host cell entry of influenza A virus. In SPOPL-depleted cells, endosomes are enlarged and fail to acquire intraluminal vesicles (ILVs). We identify a critical substrate ubiquitinated by CUL3-SPOPL as EPS15, an endocytic adaptor that also associates with the ESCRT-0 complex members HRS and STAM on endosomes. Indeed, EPS15 is ubiquitinated in a SPOPL-dependent manner, and accumulates with HRS in cells lacking SPOPL. Together, our data indicates that a CUL3-SPOPL E3 ubiquitin ligase complex regulates endocytic trafficking and MVB formation by ubiquitinating and degrading EPS15 at endosomes, thereby influencing influenza A virus infection as well as degradation of EGFR and other EPS15 targets.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Cullin/metabolismo , Endocitosis , Endosomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transporte Biológico , Línea Celular , Humanos , Virus de la Influenza A/fisiología , Internalización del Virus
12.
Traffic ; 17(4): 351-68, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26650385

RESUMEN

Human cytomegalovirus (HCMV) is an important and widespread pathogen in the human population. While infection by this ß-herpesvirus in endothelial, epithelial and dendritic cells depends on endocytosis, its entry into fibroblasts is thought to occur by direct fusion of the viral envelope with the plasma membrane. To characterize individual steps during entry in primary human fibroblasts, we employed quantitative assays as well as electron, fluorescence and live cell microscopy in combination with a variety of inhibitory compounds. Our results showed that while infectious entry was pH- and clathrin-independent, it required multiple, endocytosis-related factors and processes. The virions were found to undergo rapid internalization into large vacuoles containing internalized fluid and endosome markers. The characteristics of the internalization process fulfilled major criteria for macropinocytosis. Moreover, we found that soon after addition to fibroblasts the virus rapidly triggered the formation of circular dorsal ruffles in the host cell followed by the generation of large macropinocytic vacuoles. This distinctive form of macropinocytosis has been observed especially in primary cells but has not previously been reported in response to virus stimulation.


Asunto(s)
Citomegalovirus/fisiología , Fibroblastos/virología , Pinocitosis , Internalización del Virus , Células Cultivadas , Citomegalovirus/patogenicidad , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Humanos
13.
Traffic ; 16(8): 814-31, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25869659

RESUMEN

The prototypic poxvirus, vaccinia virus (VACV), occurs in two infectious forms, mature virions (MVs) and extracellular virions (EVs). Both enter HeLa cells by inducing macropinocytic uptake. Using confocal microscopy, live-cell imaging, targeted RNAi screening and perturbants of endosome maturation, we analyzed the properties and maturation pathway of the macropinocytic vacuoles containing VACV MVs in HeLa cells. The vacuoles first acquired markers of early endosomes [Rab5, early endosome antigen 1 and phosphatidylinositol(3)P]. Prior to release of virus cores into the cytoplasm, they contained markers of late endosomes and lysosomes (Rab7a, lysosome-associated membrane protein 1 and sorting nexin 3). RNAi screening of endocytic cell factors emphasized the importance of late compartments for VACV infection. Follow-up perturbation analysis showed that infection required Rab7a and PIKfyve, confirming that VACV is a late-penetrating virus dependent on macropinosome maturation. VACV EV infection was inhibited by depletion of many of the same factors, indicating that both infectious particle forms share the need for late vacuolar conditions for penetration.


Asunto(s)
Fagocitosis , Fagosomas/metabolismo , Virus Vaccinia/patogenicidad , Endosomas/metabolismo , Endosomas/virología , Células HeLa , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Fagosomas/virología , Nexinas de Clasificación/genética , Nexinas de Clasificación/metabolismo , Virus Vaccinia/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo , Proteínas de Unión a GTP rab7
14.
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
15.
Science ; 346(6208): 473-7, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25342804

RESUMEN

During cell entry, capsids of incoming influenza A viruses (IAVs) must be uncoated before viral ribonucleoproteins (vRNPs) can enter the nucleus for replication. After hemagglutinin-mediated membrane fusion in late endocytic vacuoles, the vRNPs and the matrix proteins dissociate from each other and disperse within the cytosol. Here, we found that for capsid disassembly, IAV takes advantage of the host cell's aggresome formation and disassembly machinery. The capsids mimicked misfolded protein aggregates by carrying unanchored ubiquitin chains that activated a histone deacetylase 6 (HDAC6)-dependent pathway. The ubiquitin-binding domain was essential for recruitment of HDAC6 to viral fusion sites and for efficient uncoating and infection. That other components of the aggresome processing machinery, including dynein, dynactin, and myosin II, were also required suggested that physical forces generated by microtubule- and actin-associated motors are essential for IAV entry.


Asunto(s)
Cápside/metabolismo , Histona Desacetilasas/fisiología , Virus de la Influenza A/fisiología , Gripe Humana/virología , Internalización del Virus , Animales , Línea Celular Tumoral , Núcleo Celular/virología , Complejo Dinactina , Dineínas/metabolismo , Técnicas de Inactivación de Genes , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Interacciones Huésped-Patógeno , Humanos , Gripe Humana/genética , Gripe Humana/metabolismo , Fusión de Membrana/genética , Fusión de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Miosina Tipo II/metabolismo , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Interferencia de ARN , Ribonucleoproteínas/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo , Replicación Viral
16.
Cell Host Microbe ; 16(3): 403-11, 2014 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-25211080

RESUMEN

In addition to classically defined immune mechanisms, cell-intrinsic processes can restrict virus infection and have shaped virus evolution. The details of this virus-host interaction are still emerging. Following a genome-wide siRNA screen for host factors affecting replication of Semliki Forest virus (SFV), a positive-strand RNA (+RNA) virus, we found that depletion of nonsense-mediated mRNA decay (NMD) pathway components Upf1, Smg5, and Smg7 led to increased levels of viral proteins and RNA and higher titers of released virus. The inhibitory effect of NMD was stronger when virus replication efficiency was impaired by mutations or deletions in the replicase proteins. Consequently, depletion of NMD components resulted in a more than 20-fold increase in production of these attenuated viruses. These findings indicate that a cellular mRNA quality control mechanism serves as an intrinsic barrier to the translation of early viral proteins and the amplification of +RNA viruses in animal cells.


Asunto(s)
Infecciones por Alphavirus/virología , Proteínas Portadoras/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido , Virus de los Bosques Semliki/fisiología , Transactivadores/metabolismo , Replicación Viral , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/metabolismo , Proteínas Portadoras/genética , Interacciones Huésped-Patógeno , Humanos , ARN Helicasas , Virus de los Bosques Semliki/genética , Transactivadores/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Liberación del Virus
17.
J Virol ; 88(22): 13029-46, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25165113

RESUMEN

UNLABELLED: Influenza A virus (IAV) uses the low pH in late endocytic vacuoles as a cue for penetration by membrane fusion. Here, we analyzed the prefusion reactions that prepare the core for uncoating after it has been delivered to the cytosol. We found that this priming process occurs in two steps that are mediated by the envelope-embedded M2 ion channel. The first weakens the interactions between the matrix protein, M1, and the viral ribonucleoprotein bundle. It involves a conformational change in a linker sequence and the C-terminal domain of M1 after exposure to a pH below 6.5. The second step is triggered by a pH of <6.0 and by the influx of K(+) ions. It causes additional changes in M1 as well as a loss of stability in the viral ribonucleoprotein bundle. Our results indicate that both the switch from Na(+) to K(+) in maturing endosomes and the decreasing pH are needed to prime IAV cores for efficient uncoating and infection of the host cell. IMPORTANCE: The entry of IAV involves several steps, including endocytosis and fusion at late endosomes. Entry also includes disassembly of the viral core, which is composed of the viral ribonucleoproteins and the RNA genome. We have found that the uncoating process of IAV is initiated long before the core is delivered into the cytosol. M2, an ion channel in the viral membrane, is activated when the virus passes through early endosomes. Here, we show that protons entering the virus through M2 cause a conformational change in the matrix protein, M1. This weakens interactions between M1 and the viral ribonucleoproteins. A second change was found to occur when the virus enters late endosomes. The preacidified core is then exposed to a high concentration of K(+), which affects the interactions between the ribonucleoproteins. Thus, when cores are finally delivered to the cytosol, they are already partially destabilized and, therefore, uncoating competent and infectious.


Asunto(s)
Endosomas/metabolismo , Endosomas/virología , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Potasio/metabolismo , Proteínas de la Matriz Viral/metabolismo , Desencapsidación Viral/efectos de los fármacos , Animales , Humanos , Concentración de Iones de Hidrógeno , Unión Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos
18.
Artículo en Inglés | MEDLINE | ID: mdl-25085912

RESUMEN

Of the many pathogens that infect humans and animals, a large number use cells of the host organism as protected sites for replication. To reach the relevant intracellular compartments, they take advantage of the endocytosis machinery and exploit the network of endocytic organelles for penetration into the cytosol or as sites of replication. In this review, we discuss the endocytic entry processes used by viruses and bacteria and compare the strategies used by these dissimilar classes of pathogens.


Asunto(s)
Endocitosis/fisiología , Células Eucariotas/microbiología , Modelos Biológicos , Internalización del Virus , Adhesión Bacteriana , Células Eucariotas/citología , Redes y Vías Metabólicas
19.
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
20.
J Virol ; 88(15): 8565-78, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24850728

RESUMEN

UNLABELLED: The Bunyaviridae constitute a large family of enveloped animal viruses, many of which are important emerging pathogens. How bunyaviruses enter and infect mammalian cells remains largely uncharacterized. We used two genome-wide silencing screens with distinct small interfering RNA (siRNA) libraries to investigate host proteins required during infection of human cells by the bunyavirus Uukuniemi virus (UUKV), a late-penetrating virus. Sequence analysis of the libraries revealed that many siRNAs in the screens inhibited infection by silencing not only the intended targets but additional genes in a microRNA (miRNA)-like manner. That the 7-nucleotide seed regions in the siRNAs can cause a perturbation in infection was confirmed by using synthetic miRNAs (miRs). One of the miRs tested, miR-142-3p, was shown to interfere with the intracellular trafficking of incoming viruses by regulating the v-SNARE VAMP3, a strong hit shared by both siRNA screens. Inactivation of VAMP3 by the tetanus toxin led to a block in infection. Using fluorescence-based techniques in fixed and live cells, we found that the viruses enter VAMP3(+) endosomal vesicles 5 min after internalization and that colocalization was maximal 15 min thereafter. At this time, LAMP1 was associated with the VAMP3(+) virus-containing endosomes. In cells depleted of VAMP3, viruses were mainly trapped in LAMP1-negative compartments. Together, our results indicated that UUKV relies on VAMP3 for penetration, providing an indication of added complexity in the trafficking of viruses through the endocytic network. IMPORTANCE: Bunyaviruses represent a growing threat to humans and livestock globally. Unfortunately, relatively little is known about these emerging pathogens. We report here the first human genome-wide siRNA screens for a bunyavirus. The screens resulted in the identification of 562 host cell factors with a potential role in cell entry and virus replication. To demonstrate the robustness of our approach, we confirmed and analyzed the role of the v-SNARE VAMP3 in Uukuniemi virus entry and infection. The information gained lays the basis for future research into the cell biology of bunyavirus infection and new antiviral strategies. In addition, by shedding light on serious caveats in large-scale siRNA screening, our experimental and bioinformatics procedures will be valuable in the comprehensive analysis of past and future high-content screening data.


Asunto(s)
Silenciador del Gen , Interacciones Huésped-Patógeno , ARN Interferente Pequeño/análisis , Virus Uukuniemi/fisiología , Proteína 3 de Membrana Asociada a Vesículas/metabolismo , Internalización del Virus , Endosomas/química , Endosomas/virología , Células Epiteliales/virología , Pruebas Genéticas , Células HeLa , Humanos , Proteínas de Membrana de los Lisosomas/análisis , ARN Interferente Pequeño/genética , Factores de Tiempo , Proteína 3 de Membrana Asociada a Vesículas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...