Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Nat Immunol ; 13(8): 737-43, 2012 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-22706339

RESUMEN

The innate immune system senses infection by detecting either evolutionarily conserved molecules essential for the survival of microbes or the abnormal location of molecules. Here we demonstrate the existence of a previously unknown innate detection mechanism induced by fusion between viral envelopes and target cells. Virus-cell fusion specifically stimulated a type I interferon response with expression of interferon-stimulated genes, in vivo recruitment of leukocytes and potentiation of signaling via Toll-like receptor 7 (TLR7) and TLR9. The fusion-dependent response was dependent on the stimulator of interferon genes STING but was independent of DNA, RNA and viral capsid. We suggest that membrane fusion is sensed as a danger signal with potential implications for defense against enveloped viruses and various conditions of giant-cell formation.


Asunto(s)
Fusión Celular , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Inmunidad Innata , Interferón Tipo I/biosíntesis , Fusión de Membrana , Proteínas de la Membrana/metabolismo , Animales , Quimiocina CXCL10/metabolismo , Células HEK293 , Células HeLa , Humanos , Leucocitos/inmunología , Leucocitos/metabolismo , Activación de Linfocitos , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Internalización del Virus
2.
J Virol ; 89(3): 1502-11, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25410861

RESUMEN

UNLABELLED: Herpesviruses have a characteristic particle structure comprising an icosahedral capsid, which contains the DNA genome and is, in turn, surrounded by a proteinaceous tegument layer and a lipid envelope. In herpes simplex virus, the interaction between the capsid and tegument is limited to the capsid vertices and involves two minor capsid proteins, pUL17 and pUL25, and the large inner tegument protein pUL36. pUL17 and pUL25 form a heterodimeric structure, the capsid vertex-specific component (CVSC), that lies on top of the peripentonal triplexes, while pUL36 has been reported to connect the CVSC to the penton. In this study, we used virus mutants with deletions in the genes for pUL36 and another inner tegument protein, pUL37, to analyze the contributions of these proteins to CVSC structure. Using electron cryomicroscopy and icosahedral reconstruction of mutants that express pUL17 and pUL25 but not pUL36, we showed that in contrast to accepted models, the CVSC is not formed from pUL17 and pUL25 on their own but requires a contribution from pUL36. In addition, the presence of full-length pUL36 results in weak density that extends the CVSC toward the penton, suggesting either that this extra density is formed directly by pUL36 or that pUL36 stabilizes other components of the vertex-tegument interface. IMPORTANCE: Herpesviruses have complex particles that are formed as a result of a carefully controlled sequence of assembly steps. The nature of the interaction between two of the major particle compartments, the icosahedral capsid and the amorphous tegument, has been extensively studied, but the identity of the interacting proteins and their roles in forming the connections are still unclear. In this study, we used electron microscopy and three-dimensional reconstruction to analyze virus particles formed by mutants that do not express particular interacting proteins. We show that the largest viral protein, pUL36, which occupies the layer of tegument closest to the capsid, is essential for formation of structurally normal connections to the capsid. This demonstrates the importance of pUL36 in the initial stages of tegument addition and provides new insights into the process of virus particle assembly.


Asunto(s)
Proteínas de la Cápside/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas Virales/fisiología , Ensamble de Virus , Animales , Cápside/metabolismo , Línea Celular , Microscopía por Crioelectrón , Eliminación de Gen , Procesamiento de Imagen Asistido por Computador , Proteínas Virales/genética
3.
J Virol ; 89(18): 9407-16, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26136572

RESUMEN

UNLABELLED: The cellular proteins nectin-1 and herpesvirus entry mediator (HVEM) can both mediate the entry of herpes simplex virus 1 (HSV-1). We have recently shown how these receptors contribute to infection of skin by investigating HSV-1 entry into murine epidermis. Ex vivo infection studies reveal nectin-1 as the primary receptor in epidermis, whereas HVEM has a more limited role. Although the epidermis represents the outermost layer of skin, the contribution of nectin-1 and HVEM in the underlying dermis is still open. Here, we analyzed the role of each receptor during HSV-1 entry in murine dermal fibroblasts that were deficient in expression of either nectin-1 or HVEM or both receptors. Because infection was not prevented by the absence of either nectin-1 or HVEM, we conclude that they can act as alternative receptors. Although HVEM was found to be highly expressed on fibroblasts, entry was delayed in nectin-1-deficient cells, suggesting that nectin-1 acts as the more efficient receptor. In the absence of both receptors, entry was strongly delayed leading to a much reduced viral spread and virus production. These results suggest an unidentified cellular component that acts as alternate but inefficient receptor for HSV-1 on dermal fibroblasts. Characterization of the cellular entry mechanism suggests that HSV-1 can enter dermal fibroblasts both by direct fusion with the plasma membrane and via endocytic vesicles and that this is not dependent on the presence or absence of nectin-1. Entry was also shown to require dynamin and cholesterol, suggesting comparable entry pathways in keratinocytes and dermal fibroblasts. IMPORTANCE: Herpes simplex virus (HSV) is a human pathogen which infects its host via mucosal surfaces or abraded skin. To understand how HSV-1 overcomes the protective barrier of mucosa or skin and reaches its receptors in tissue, it is essential to know which receptors contribute to the entry into individual skin cells. Previously, we have explored the contribution of nectin-1 and herpesvirus entry mediator (HVEM) as receptors for HSV-1 entry into murine epidermis, where keratinocytes form the major cell type. Since the underlying dermis consists primarily of fibroblasts, we have now extended our study of HSV-1 entry to dermal fibroblasts isolated from nectin-1- or HVEM-deficient mice or from mice deficient in both receptors. Our results demonstrate a role for both nectin-1 and HVEM as receptors and suggest a further receptor which appears much less efficient.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Fibroblastos/metabolismo , Herpes Simple/metabolismo , Herpesvirus Humano 1/fisiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Internalización del Virus , Animales , Moléculas de Adhesión Celular/genética , Células Cultivadas , Dermis/metabolismo , Dermis/patología , Dermis/virología , Epidermis/metabolismo , Epidermis/patología , Epidermis/virología , Fibroblastos/patología , Fibroblastos/virología , Herpes Simple/genética , Herpes Simple/patología , Humanos , Ratones , Ratones Noqueados , Nectinas , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética
4.
J Virol ; 89(1): 262-74, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25320325

RESUMEN

UNLABELLED: Skin keratinocytes represent a primary entry site for herpes simplex virus 1 (HSV-1) in vivo. The cellular proteins nectin-1 and herpesvirus entry mediator (HVEM) act as efficient receptors for both serotypes of HSV and are sufficient for disease development mediated by HSV-2 in mice. How HSV-1 enters skin and whether both nectin-1 and HVEM are involved are not known. We addressed the impact of nectin-1 during entry of HSV-1 into murine epidermis and investigated the putative contribution of HVEM. Using ex vivo infection of murine epidermis, we showed that HSV-1 entered the basal keratinocytes of the epidermis very efficiently. In nectin-1-deficient epidermis, entry was strongly reduced. Almost no entry was observed, however, in nectin-1-deficient keratinocytes grown in culture. This observation correlated with the presence of HVEM on the keratinocyte surface in epidermis and with the lack of HVEM expression in nectin-1-deficient primary keratinocytes. Our results suggest that nectin-1 is the primary receptor in epidermis, while HVEM has a more limited role. For primary murine keratinocytes, on which nectin-1 acts as a single receptor, electron microscopy suggested that HSV-1 can enter both by direct fusion with the plasma membrane and via endocytic vesicles. Thus, we concluded that nectin-1 directs internalization into keratinocytes via alternative pathways. In summary, HSV-1 entry into epidermis was shown to strongly depend on the presence of nectin-1, but the restricted presence of HVEM can potentially replace nectin-1 as a receptor, illustrating the flexibility employed by HSV-1 to efficiently invade tissue in vivo. IMPORTANCE: Herpes simplex virus (HSV) can cause a range of diseases in humans, from uncomplicated mucocutaneous lesions to life-threatening infections. The skin is one target tissue of HSV, and the question of how the virus overcomes the protective skin barrier and penetrates into the tissue to reach its receptors is still open. Previous studies analyzing entry into cells grown in vitro revealed nectin-1 and HVEM as HSV receptors. To explore the contributions of nectin-1 and HVEM to entry into a natural target tissue, we established an ex vivo infection model. Using nectin-1- or HVEM-deficient mice, we demonstrated the distinct involvement of nectin-1 and HVEM for HSV-1 entry into epidermis and characterized the internalization pathways. Such advances in understanding the involvement of receptors in tissue are essential preconditions for unraveling HSV invasion of skin, which in turn will allow the development of antiviral reagents.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Queratinocitos/virología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Virales/metabolismo , Internalización del Virus , Animales , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Nectinas , Piel/virología
5.
PLoS Pathog ; 9(6): e1003413, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23754946

RESUMEN

Influenza viruses exhibit striking variations in particle morphology between strains. Clinical isolates of influenza A virus have been shown to produce long filamentous particles while laboratory-adapted strains are predominantly spherical. However, the role of the filamentous phenotype in the influenza virus infectious cycle remains undetermined. We used cryo-electron tomography to conduct the first three-dimensional study of filamentous virus ultrastructure in particles budding from infected cells. Filaments were often longer than 10 microns and sometimes had bulbous heads at their leading ends, some of which contained tubules we attribute to M1 while none had recognisable ribonucleoprotein (RNP) and hence genome segments. Long filaments that did not have bulbs were infrequently seen to bear an ordered complement of RNPs at their distal ends. Imaging of purified virus also revealed diverse filament morphologies; short rods (bacilliform virions) and longer filaments. Bacilliform virions contained an ordered complement of RNPs while longer filamentous particles were narrower and mostly appeared to lack this feature, but often contained fibrillar material along their entire length. The important ultrastructural differences between these diverse classes of particles raise the possibility of distinct morphogenetic pathways and functions during the infectious process.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/ultraestructura , Virión/ultraestructura , Animales , Microscopía por Crioelectrón/métodos , Perros , Subtipo H3N2 del Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Virión/fisiología
6.
PLoS Pathog ; 9(6): e1003461, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23818857

RESUMEN

Genetic robustness, or fragility, is defined as the ability, or lack thereof, of a biological entity to maintain function in the face of mutations. Viruses that replicate via RNA intermediates exhibit high mutation rates, and robustness should be particularly advantageous to them. The capsid (CA) domain of the HIV-1 Gag protein is under strong pressure to conserve functional roles in viral assembly, maturation, uncoating, and nuclear import. However, CA is also under strong immunological pressure to diversify. Therefore, it would be particularly advantageous for CA to evolve genetic robustness. To measure the genetic robustness of HIV-1 CA, we generated a library of single amino acid substitution mutants, encompassing almost half the residues in CA. Strikingly, we found HIV-1 CA to be the most genetically fragile protein that has been analyzed using such an approach, with 70% of mutations yielding replication-defective viruses. Although CA participates in several steps in HIV-1 replication, analysis of conditionally (temperature sensitive) and constitutively non-viable mutants revealed that the biological basis for its genetic fragility was primarily the need to coordinate the accurate and efficient assembly of mature virions. All mutations that exist in naturally occurring HIV-1 subtype B populations at a frequency >3%, and were also present in the mutant library, had fitness levels that were >40% of WT. However, a substantial fraction of mutations with high fitness did not occur in natural populations, suggesting another form of selection pressure limiting variation in vivo. Additionally, known protective CTL epitopes occurred preferentially in domains of the HIV-1 CA that were even more genetically fragile than HIV-1 CA as a whole. The extreme genetic fragility of HIV-1 CA may be one reason why cell-mediated immune responses to Gag correlate with better prognosis in HIV-1 infection, and suggests that CA is a good target for therapy and vaccination strategies.


Asunto(s)
Cápside/metabolismo , Proteína p24 del Núcleo del VIH/metabolismo , VIH-1/fisiología , Mutación , Replicación Viral/fisiología , Línea Celular , Femenino , Proteína p24 del Núcleo del VIH/genética , Humanos , Masculino , Estabilidad Proteica , Estructura Terciaria de Proteína
7.
J Virol ; 87(12): 7102-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23596303

RESUMEN

Efficient intracellular transport of the capsid of alphaherpesviruses, such as herpes simplex virus 1 (HSV-1), is known to be dependent upon the microtubule (MT) network. Typically, the MT network radiates from an MT-organizing center (MTOC), which is, in most cases, the centrosome. During herpesvirus egress, it has been assumed that capsids travel first from the nucleus to the centrosome and then from the centrosome to the site of envelopment. Here we report that the centrosome is no longer a primary MTOC in HSV-1-infected cells, but it retains this function in cells infected by another alphaherpesvirus, pseudorabies virus (PrV). As a result, MTs formed at late times after infection with PrV grow from a major, centralized MTOC, while those formed after HSV-1 infection arise from dispersed locations in the cytoplasm, indicating the presence of alternative and minor MTOCs. Thus, loss of the principal MT nucleating center in cells following HSV-1 infection raises questions about the mechanism of HSV-1 capsid egress. It is possible that, rather than passing via the centrosome, capsids may travel directly to the site of envelopment after exiting the nucleus. We suggest that, in HSV-1-infected cells, the disruption of centrosomal functions triggers reorganization of the MT network to favor noncentrosomal MTs and promote efficient viral spread.


Asunto(s)
Centrosoma/metabolismo , Centrosoma/virología , Herpesvirus Humano 1/patogenicidad , Herpesvirus Suido 1/patogenicidad , Animales , Cápside/metabolismo , Línea Celular , Chlorocebus aethiops , Fibroblastos/virología , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Herpesvirus Suido 1/fisiología , Humanos , Microtúbulos/metabolismo , Microtúbulos/virología , Seudorrabia/virología , Células Vero
8.
J Virol ; 87(5): 2857-67, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269794

RESUMEN

Herpes simplex virus 1 (HSV-1) is a neurotropic virus that travels long distances through cells using the microtubule network. Its 125-nm-diameter capsid is a large cargo which efficiently recruits molecular motors for movement. Upon entry, capsids reach the centrosome by minus-end-directed transport. From there, they are believed to reach the nucleus by plus-end-directed transport. Plus-end-directed transport is also important during egress, when capsids leave the nucleus to reach the site of envelopment in the cytoplasm. Although capsid interactions with dynein and kinesins have been described in vitro, the actual composition of the cellular machinery recruited by herpesviruses for capsid transport in infected cells remains unknown. Here, we identify the spectraplakin protein, dystonin/BPAG1, an important cytoskeleton cross-linker involved in microtubule-based transport, as a binding partner of the HSV-1 protein pUL37, which has been implicated in capsid transport. Viral replication is delayed in dystonin-depleted cells, and, using video microscopy of living infected cells, we show that dystonin depletion strongly inhibits capsid movement in the cytoplasm during egress. This study provides new insights into the cellular requirements for HSV-1 capsid transport and identifies dystonin as a nonmotor protein part of the transport machinery.


Asunto(s)
Cápside/fisiología , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Proteínas de la Cápside/metabolismo , Proteínas Portadoras/genética , Línea Celular , Chlorocebus aethiops , Cricetinae , Proteínas del Citoesqueleto/genética , Distonina , Células HEK293 , Herpes Simple/metabolismo , Humanos , Microtúbulos/virología , Proteínas del Tejido Nervioso/genética , Estructura Terciaria de Proteína , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño , Células Vero , Liberación del Virus , Replicación Viral
9.
J Virol ; 87(20): 11008-18, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23903849

RESUMEN

During infection by herpes simplex virus 1 (HSV-1), the viral capsid is transported around the cytoplasm along the microtubule (MT) network. Although molecular motors have been implicated in this process, the composition of the molecular machinery required for efficient directional transport is unknown. We previously showed that dystonin (BPAG1) is recruited to HSV-1 capsids by the capsid-bound tegument protein pUL37 to promote efficient cytoplasmic transport of capsids during egress. Dystonin is a cytoskeleton cross-linker which localizes at MT plus ends and has roles in retrograde and anterograde transport in neurons. In this study, we investigated the role of dystonin during the entry stages of HSV-1 infection. Because of the way in which the MT network is organized, capsids are required to change their direction of motion along the MTs as they travel from the point of entry to the nucleus, where replication takes place. Thus, capsids first travel to the centrosome (the principal microtubule organizing center) by minus-end-directed transport and then switch polarity and travel to the nucleus by plus-end-directed transport. We observed that transport of capsids toward the centrosome was slowed, but not blocked, by dystonin depletion. However, transport of capsids away from the centrosome was significantly impaired, causing them to accumulate in the vicinity of the centrosome and reducing the numbers reaching the nucleus. We conclude that, during entry of HSV-1, dystonin has a specific role in plus-ended transport of capsids from the centrosome to the nucleus.


Asunto(s)
Cápside/metabolismo , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Internalización del Virus , Animales , Línea Celular , Distonina , Humanos
10.
PLoS Pathog ; 8(10): e1002961, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055933

RESUMEN

Herpes viruses are prevalent and well characterized human pathogens. Despite extensive study, much remains to be learned about the structure of the genome packaging and release machinery in the capsids of these large and complex double-stranded DNA viruses. However, such machinery is well characterized in tailed bacteriophage, which share a common evolutionary origin with herpesvirus. In tailed bacteriophage, the genome exits from the virus particle through a portal and is transferred into the host cell by a complex apparatus (i.e. the tail) located at the portal vertex. Here we use electron cryo-tomography of human herpes simplex type-1 (HSV-1) virions to reveal a previously unsuspected feature at the portal vertex, which extends across the HSV-1 tegument layer to form a connection between the capsid and the viral membrane. The location of this assembly suggests that it plays a role in genome release into the nucleus and is also important for virion architecture.


Asunto(s)
Cápside/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/ultraestructura , Virión/ultraestructura , Internalización del Virus , Animales , Bacteriófagos , Cápside/química , Cricetinae , Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Herpesvirus Humano 1/metabolismo , Humanos , Virión/metabolismo
11.
J Immunol ; 187(10): 5268-76, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21998456

RESUMEN

Autophagy has been established as a player in host defense against viruses. The mechanisms by which the host induces autophagy during infection are diverse. In the case of HSV type 1 (HSV-1), dsRNA-dependent protein kinase is essential for induction of autophagy in fibroblasts through phosphorylation of eukaryotic initiation factor 2α (eIF2α). HSV-1 counteracts autophagy via ICP34.5, which dephosphorylates eIF2α and inhibits Beclin 1. Investigation of autophagy during HSV-1 infection has largely been conducted in permissive cells, but recent work suggests the existence of a eIF2α-independent autophagy-inducing pathway in nonpermissive cells. To clarify and further characterize the existence of a novel autophagy-inducing pathway in nonpermissive cells, we examined different HSV and cellular components in murine myeloid cells for their role in autophagy. We demonstrate that HSV-1-induced autophagy does not correlate with phosphorylation of eIF2α, is independent of functional dsRNA-dependent protein kinase, and is not antagonized by ICP34.5. Autophagy was activated independent of viral gene expression, but required viral entry. Importantly, we found that the presence of genomic DNA in the virion was essential for induction of autophagy and, conversely, that transfection of HSV-derived DNA induced microtubule-associated protein 1 L chain II formation, a marker of autophagy. This occurred through a mechanism dependent on stimulator of IFN genes, an essential component for the IFN response to intracellular DNA. Finally, we observed that HSV-1 DNA was present in the cytosol devoid of capsid material following HSV-1 infection of dendritic cells. Thus, our data suggest that HSV-1 genomic DNA induces autophagy in nonpermissive cells in a stimulator of IFN gene-dependent manner.


Asunto(s)
Autofagia/inmunología , Citosol/virología , ADN Viral , Herpesvirus Humano 1/inmunología , Proteínas de la Membrana/fisiología , Células Mieloides/inmunología , Células Mieloides/virología , Animales , Autofagia/genética , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/virología , Línea Celular , Citosol/inmunología , ADN Viral/genética , Células Dendríticas/inmunología , Células Dendríticas/virología , Femenino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Células Mieloides/citología
12.
J Virol ; 84(9): 4252-63, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181717

RESUMEN

The herpes simplex virus type 1 (HSV-1) UL25 gene encodes a minor capsid protein, pUL25, that is essential for packaging the full-length viral genome. Six regions which contain disordered residues have been identified in the high-resolution structure of pUL25. To investigate the significance of these flexible regions, a panel of plasmids was generated encoding mutant proteins, with each member lacking the disordered residues in one of the six regions. In addition, UL25 constructs were produced, which specified proteins that contained missense mutations individually affecting two of the four regions on the surface of pUL25 predicted from evolutionary trace analysis to be important in protein-protein interactions. The impacts of these mutations on viral DNA packaging, virus assembly, and growth were examined. Of the nine mutant proteins analyzed, five failed to complement the growth of a UL25 deletion mutant in Vero cells. These noncomplementing proteins fell into three classes. Proteins in one class did not alter the DNA packaging phenotype of an HSV-1 UL25 deletion mutant, whereas proteins from the other two classes allowed the UL25 null mutant to package full-length viral DNA. Subsequent analysis of the latter classes of mutant proteins demonstrated that one class enabled the null virus to release enveloped virus particles from U2OS cells, whereas the other class prevented egress of mature HSV-1 capsids from the nucleus. These findings reveal a new role for pUL25 in virion assembly, consistent with its flexible structure and location on the capsid.


Asunto(s)
Proteínas de la Cápside/fisiología , Empaquetamiento del ADN , Herpesvirus Humano 1/fisiología , Ensamble de Virus , Liberación del Virus , Animales , Proteínas de la Cápside/genética , Línea Celular , Chlorocebus aethiops , ADN Viral/metabolismo , Eliminación de Gen , Prueba de Complementación Genética , Herpesvirus Humano 1/genética , Humanos , Microscopía Electrónica de Transmisión , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Estructura Terciaria de Proteína , Virión/ultraestructura
13.
J Gen Virol ; 91(Pt 9): 2145-51, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20505007

RESUMEN

Secondary envelopment of herpes simplex virus type 1 has been demonstrated as taking place at the trans-Golgi network (TGN). The inner tegument proteins pUL36 and pUL37 and the envelope glycoproteins gD and gE are known to be important for secondary envelopment. We compared the cellular localizations of capsids from a virus mutant lacking the UL37 gene with those of a virus mutant lacking the genes encoding gD and gE. Although wild-type capsids accumulated at the TGN, capsids of the pUL37(-) mutant were distributed throughout the cytoplasm and showed no association with TGN-derived vesicles. This was in contrast to capsids from a gD(-)gE(-) mutant, which accumulated in the vicinity of TGN vesicles, but did not colocalize with them, suggesting that they were transported to the TGN but were unable to undergo envelopment. We conclude that the inner tegument protein pUL37 is required for directing capsids to the TGN, where secondary envelopment occurs.


Asunto(s)
Herpesvirus Humano 1/fisiología , Proteínas Estructurales Virales/fisiología , Transporte Biológico Activo , Cápside/fisiología , Proteínas de la Cápside/genética , Proteínas de la Cápside/fisiología , Línea Celular , Genes Virales , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Herpesvirus Humano 1/genética , Humanos , Mutación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Estructurales Virales/genética , Ensamble de Virus/genética , Ensamble de Virus/fisiología , Red trans-Golgi/virología
14.
J Virol ; 83(13): 6610-23, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19386703

RESUMEN

After penetrating the host cell, the herpesvirus capsid is transported to the nucleus along the microtubule network and docks to the nuclear pore complex before releasing the viral DNA into the nucleus. The viral and cellular interactions involved in the docking process are poorly characterized. However, the minor capsid protein pUL25 has recently been reported to be involved in viral DNA uncoating. Here we show that herpes simplex virus type 1 (HSV-1) capsids interact with the nucleoporin CAN/Nup214 in infected cells and that RNA silencing of CAN/Nup214 delays the onset of viral DNA replication in the nucleus. We also show that pUL25 interacts with CAN/Nup214 and another nucleoporin, hCG1, and binds to the pUL36 and pUL6 proteins, two other components of the herpesvirus particle that are known to be important for the initiation of infection and viral DNA release. These results identify CAN/Nup214 as being a nuclear receptor for the herpesvirus capsid and pUL25 as being an interface between incoming capsids and the nuclear pore complex and as being a triggering element for viral DNA release into the nucleus.


Asunto(s)
Proteínas de la Cápside/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/virología , Proteínas Virales/metabolismo , Animales , Chlorocebus aethiops , Cricetinae , ADN Viral/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Células Vero , Replicación Viral
15.
J Virol ; 83(1): 105-16, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18971278

RESUMEN

Studies with herpes simplex virus type 1 (HSV-1) have shown that secondary envelopment and virus release are blocked in mutants deleted for the tegument protein gene UL36 or UL37, leading to the accumulation of DNA-containing capsids in the cytoplasm of infected cells. The failure to assemble infectious virions has meant that the roles of these genes in the initial stages of infection could not be investigated. To circumvent this, cells infected at a low multiplicity were fused to form syncytia, thereby allowing capsids released from infected nuclei access to uninfected nuclei without having to cross a plasma membrane. Visualization of virus DNA replication showed that a UL37-minus mutant was capable of transmitting infection to all the nuclei within a syncytium as efficiently as the wild-type HSV-1 strain 17(+) did, whereas infection by UL36-minus mutants failed to spread. Thus, these inner tegument proteins have differing functions, with pUL36 being essential during both the assembly and uptake stages of infection, while pUL37 is needed for the formation of virions but is not required during the initial stages of infection. Analysis of noninfectious enveloped particles (L-particles) further showed that pUL36 and pUL37 are dependent on each other for incorporation into tegument.


Asunto(s)
Herpesvirus Humano 1/fisiología , Proteínas Virales/metabolismo , Proteínas Estructurales Virales/metabolismo , Internalización del Virus , Células Cultivadas , Eliminación de Gen , Herpesvirus Humano 1/genética , Humanos , Unión Proteica , Proteínas Virales/genética , Proteínas Estructurales Virales/genética , Ensamble de Virus
16.
J Virol ; 83(7): 3187-99, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19176621

RESUMEN

Axonal transport of herpes simplex virus (HSV-1) is essential for viral infection and spread in the peripheral nervous system of the host. Therefore, the virus probably utilizes existing active transport and targeting mechanisms in neurons for virus assembly and spread from neurons to skin. In the present study, we used transmission immunoelectron microscopy to investigate the nature and origin of vesicles involved in the anterograde axonal transport of HSV-1 tegument and envelope proteins and of vesicles surrounding partially and fully enveloped capsids in growth cones. This study aimed to elucidate the mechanism of virus assembly and exit from axons of human fetal dorsal root ganglia neurons. We demonstrated that viral tegument and envelope proteins can travel in axons independently of viral capsids and were transported to the axon terminus in two types of transport vesicles, tubulovesicular membrane structures and large dense-cored vesicles. These vesicles and membrane carriers were derived from the trans-Golgi network (TGN) and contained key proteins, such as Rab3A, SNAP-25, GAP-43, and kinesin-1, involved in the secretory and exocytic pathways in axons. These proteins were also observed on fully and partially enveloped capsids in growth cones and on extracellular virions. Our findings provide further evidence to the subassembly model of separate transport in axons of unenveloped capsids from envelope and tegument proteins with final virus assembly occurring at the axon terminus. We postulate that HSV-1 capsids invaginate tegument- and envelope-bearing TGN-derived vesicles and utilize the large secretory vesicle pathway of exocytosis for exit from axons.


Asunto(s)
Axones/virología , Exocitosis , Conos de Crecimiento/virología , Herpesvirus Humano 1/fisiología , Vesículas Secretoras/virología , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus , Axones/ultraestructura , Línea Celular , Proteína GAP-43/análisis , Conos de Crecimiento/ultraestructura , Humanos , Cinesinas/análisis , Microscopía Inmunoelectrónica , Vesículas Secretoras/química , Proteína 25 Asociada a Sinaptosomas/análisis , Proteína de Unión al GTP rab3A/análisis
17.
PLoS Comput Biol ; 2(10): e146, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-17069457

RESUMEN

Efforts in structural biology have targeted the systematic determination of all protein structures through experimental determination or modeling. In recent years, 3-D electron cryomicroscopy (cryoEM) has assumed an increasingly important role in determining the structures of these large macromolecular assemblies to intermediate resolutions (6-10 A). While these structures provide a snapshot of the assembly and its components in well-defined functional states, the resolution limits the ability to build accurate structural models. In contrast, sequence-based modeling techniques are capable of producing relatively robust structural models for isolated proteins or domains. In this work, we developed and applied a hybrid modeling approach, utilizing cryoEM density and ab initio modeling to produce a structural model for the core domain of a herpesvirus structural protein, VP26. Specifically, this method, first tested on simulated data, utilizes the cryoEM density map as a geometrical constraint in identifying the most native-like models from a gallery of models generated by ab initio modeling. The resulting model for the core domain of VP26, based on the 8.5-A resolution herpes simplex virus type 1 (HSV-1) capsid cryoEM structure and mutational data, exhibited a novel fold. Additionally, the core domain of VP26 appeared to have a complementary interface to the known upper-domain structure of VP5, its cognate binding partner. While this new model provides for a better understanding of the assembly and interactions of VP26 in HSV-1, the approach itself may have broader applications in modeling the components of large macromolecular assemblies.


Asunto(s)
Absorciometría de Fotón/métodos , Proteínas de la Cápside/química , Proteínas de la Cápside/ultraestructura , Microscopía por Crioelectrón/métodos , Modelos Químicos , Modelos Moleculares , Secuencia de Aminoácidos , Simulación por Computador , Cristalografía/métodos , Datos de Secuencia Molecular , Conformación Proteica , Estructura Terciaria de Proteína
18.
Virus Res ; 117(1): 90-104, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16490275

RESUMEN

Herpesviruses comprise an abundant, widely distributed group of large DNA viruses of humans and other vertebrates, and overall are among the most extensively studied large DNA viruses. Many herpesvirus genome sequences have been determined, and interpreted in terms of gene contents to give detailed views of both ubiquitous and lineage-specific functions. Availability of gene sequences has also enabled evaluations of evolutionary relationships. For herpesviruses of mammals, a robust phylogenetic tree has been constructed, which shows many features characteristic of synchronous development of virus and host lineages over large evolutionary timespans. It has also emerged that three distinct groupings of herpesviruses exist: the first containing viruses with mammals, birds and reptiles as natural hosts; the second containing viruses of amphibians and fish; and the third consisting of a single invertebrate herpesvirus. Within each of the first two groups, the genomes show clear evidence of descent from a common ancestor, but relationships between the three groups are extremely remote. Detailed analyses of capsid structures provide the best evidence for a common origin of the three groups. At a finer level, the structure of the capsid shell protein further suggests an element of common origin between herpesviruses and tailed DNA bacteriophages.


Asunto(s)
Evolución Molecular , Genómica , Herpesviridae/genética , Animales , Cápside/química , Humanos , Modelos Moleculares
19.
Biochim Biophys Acta ; 1571(1): 1-8, 2002 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-12031284

RESUMEN

An investigation has been performed into the biophysical properties of the enveloped mammalian virus, herpes simplex virus type 1 (HSV-1). The dielectrophoretic behaviour of the virus particles was measured as a function of applied frequency (over the range 100 kHz-20 MHz) and conductivity of the suspending medium (over the range 1-100 mS m(-1)). The dielectric properties of the virus were determined from the dielectrophoretic data using the smeared-out shell model. The data suggest that the intact particle has a surface conductance of 0.3 nS, an internal and membrane permittivity of 75varepsilon(o) and 7.5varepsilon(o), respectively, an internal conductivity of approximately 0.1 S m(-1) and a zeta potential of 70 mV. The dielectric properties were measured for intact, fresh virus particles and also for particles following exposure to various modifying agents, such as treatment with enzymes, ionophores and ageing. It is shown that the observed changes in the dielectrophoretic spectrum, and the variations in the dielectric properties of the virus concur with the expected physiological effects of these agents.


Asunto(s)
Herpesvirus Humano 1/química , Virión/química , Fenómenos Biofísicos , Biofisica , Frío , Conductividad Eléctrica , Electroforesis/métodos , Saponinas , Tripsina , Valinomicina
20.
Adv Protein Chem ; 64: 379-408, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-13677054

RESUMEN

In this article we have attempted to describe some structural aspects of large viruses. Although this may seem a straightforward task, it is complicated by the fact that large viruses do not represent a distinctive class of organisms and any grouping under this heading will include a range of unrelated viruses with different structures, replication strategies, and host types. To simplify matters we limited our definition to dsDNA viruses with genomes of 100 kbp or larger. However, even this restricted grouping includes viruses with diverse and seemingly unrelated structures. Furthermore, few if any structural features are exclusive to large viruses and most of what appears distinctive about their structure or assembly can also be found in smaller, and usually better characterized, viruses. Therefore we have not attempted to provide a comprehensive catalog of the properties of large viruses but have tried to illustrate particular structural points with examples from a few of the better known forms, notably herpes simplex virus (HSV) and phage T4. The two techniques used to provide rigorous analyses of virus structures are X-ray crystallography and electron cryomicroscopy with computer-assisted reconstruction. To date, X-ray crystallography has been successful only with smaller viruses, and what is known about the structures of these large viruses has come primarily from electron cryomicroscopy. However, with the notable exception of the HSV capsid, such studies have been limited in extent and of relatively low resolution, and the information obtained has been confined largely to describing the spatial distributions and relationships between the subunits. Nevertheless, these studies have given us our clearest insights into the biology of these complex particles and increases in resolution promise to extend these insights by bridging the gap between gross and atomic structures, as exemplified by the identification and mapping of secondary structural elements in the HSV capsid.


Asunto(s)
Virus/química , Virus/ultraestructura , Genoma Viral , Humanos , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/metabolismo , Virión/ultraestructura , Fenómenos Fisiológicos de los Virus , Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA