Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 16(8): e1008845, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32866210

RESUMEN

Modified vaccinia virus Ankara (MVA) is an approved smallpox vaccine and a promising vaccine vector for other pathogens as well as for cancer therapeutics with more than 200 current or completed clinical trials. MVA was derived by passaging the parental Ankara vaccine virus hundreds of times in chick embryo fibroblasts during which it lost the ability to replicate in human and most other mammalian cells. Although this replication deficiency is an important safety feature, the genetic basis of the host restriction is not understood. Here, an unbiased human genome-wide RNAi screen in human A549 cells revealed that the zinc-finger antiviral protein (ZAP), previously shown to inhibit certain RNA viruses, is a host restriction factor for MVA, a DNA virus. Additional studies demonstrated enhanced MVA replication in several human cell lines following knockdown of ZAP. Furthermore, CRISPR-Cas9 knockout of ZAP in human A549 cells increased MVA replication and spread by more than one log but had no effect on a non-attenuated strain of vaccinia virus. The intact viral C16 protein, which had been disrupted in MVA, antagonized ZAP by binding and sequestering the protein in cytoplasmic punctate structures. Studies aimed at exploring the mechanism by which ZAP restricts MVA replication in the absence of C16 showed that knockout of ZAP had no discernible effect on viral DNA or individual mRNA or protein species as determined by droplet digital polymerase chain reaction, deep RNA sequencing and mass spectrometry, respectively. Instead, inactivation of ZAP reduced the number of aberrant, dense, spherical particles that typically form in MVA-infected human cells, suggesting that ZAP has a novel role in interfering with a late step in the assembly of infectious MVA virions in the absence of the C16 protein.


Asunto(s)
Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Virus Vaccinia/fisiología , Replicación Viral/fisiología , Células A549 , Animales , Pollos , Citoplasma/metabolismo , Citoplasma/virología , ADN Viral/genética , ADN Viral/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , RNA-Seq , Proteínas Represoras/genética
2.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32669330

RESUMEN

Unlike RNA viruses, most DNA viruses replicate their genomes with high-fidelity polymerases that rarely make base substitution errors. Nevertheless, experimental evolution studies have revealed rapid acquisition of adaptive mutations during serial passage of attenuated vaccinia virus (VACV). One way in which adaptation can occur is by an accordion mechanism in which the gene copy number increases followed by base substitutions and, finally, contraction of the gene copy number. Here, we show rapid acquisition of multiple adaptive mutations mediated by a gene-inactivating frameshift mechanism during passage of an attenuated VACV. Attenuation had been achieved by exchanging the VACV A8R intermediate transcription factor gene with the myxoma virus ortholog. A total of seven mutations in six different genes occurred in three parallel passages of the attenuated virus. The most frequent mutations were single-nucleotide insertions or deletions within runs of five to seven As or Ts, although a deletion of 11 nucleotides also occurred, leading to frameshifts and premature stop codons. During 10 passage rounds, the attenuated VACV was replaced by the mutant viruses. At the end of the experiment, virtually all remaining viruses had one fixed mutation and one or more additional mutations. Although nucleotide substitutions in the transcription apparatus accounted for two low-frequency mutations, frameshifts in genes encoding protein components of the mature virion, namely, A26L, G6R, and A14.5L, achieved 74% to 98% fixation. The adaptive role of the mutations was confirmed by making recombinant VACV with A26L or G6R or both deleted, which increased virus replication levels and decreased particle/PFU ratios.IMPORTANCE Gene inactivation is considered to be an important driver of orthopoxvirus evolution. Whereas cowpox virus contains intact orthologs of genes present in each orthopoxvirus species, numerous genes are inactivated in all other members of the genus. Inactivation of additional genes can occur upon extensive passaging of orthopoxviruses in cell culture leading to attenuation in vivo, a strategy for making vaccines. Whether inactivation of multiple viral genes enhances replication in the host cells or has a neutral effect is unknown in most cases. Using an experimental evolution protocol involving serial passages of an attenuated vaccinia virus, rapid acquisition of inactivating frameshift mutations occurred. After only 10 passage rounds, the starting attenuated vaccinia virus was displaced by viruses with one fixed mutation and one or more additional mutations. The high frequency of multiple inactivating mutations during experimental evolution simulates their acquisition during normal evolution and extensive virus passaging to make vaccine strains.


Asunto(s)
Adaptación Biológica/genética , Mutación del Sistema de Lectura , Myxoma virus/genética , Factores de Transcripción/genética , Virus Vaccinia/genética , Proteínas Virales/genética , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Codón sin Sentido , Células Epiteliales/metabolismo , Células Epiteliales/virología , Dosificación de Gen , Aptitud Genética , Myxoma virus/metabolismo , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinación Genética , Factores de Transcripción/metabolismo , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Secuenciación Completa del Genoma
3.
J Virol ; 93(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31092566

RESUMEN

Infectious human papillomavirus 16 (HPV16) L1/L2 pseudovirions were found to remain largely intact during vesicular transport to the nucleus. By electron microscopy, capsids with a diameter of 50 nm were clearly visible within small vesicles attached to mitotic chromosomes and to a lesser extent within interphase nuclei, implying nuclear disassembly. By confocal analysis, it was determined that nuclear entry of assembled L1 is dependent upon the presence of the minor capsid protein, L2, but independent of encapsidated DNA. We also demonstrate that L1 nuclear localization and mitotic chromosome association can occur in vivo in the murine cervicovaginal challenge model of HPV16 infection. These findings challenge the prevailing concepts of PV uncoating and disassembly. More generally, they document that a largely intact viral capsid can enter the nucleus within a transport vesicle, establishing a novel mechanism by which a virus accesses the nuclear cellular machinery.IMPORTANCE Papillomaviruses (PVs) comprise a large family of nonenveloped DNA viruses that include HPV16, among other oncogenic types, the causative agents of cervical cancer. Delivery of the viral DNA into the host cell nucleus is necessary for establishment of infection. This was thought to occur via a subviral complex following uncoating of the larger viral capsid. In this study, we demonstrate that little disassembly of the PV capsid occurs prior to nuclear delivery. These surprising data reveal a previously unrecognized viral strategy to access the nuclear replication machinery. Understanding viral entry mechanisms not only increases our appreciation of basic cell biological pathways but also may lead to more effective antiviral interventions.


Asunto(s)
Proteínas de la Cápside/metabolismo , Núcleo Celular/virología , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/metabolismo , Internalización del Virus , Animales , Cápside/metabolismo , Cápside/ultraestructura , Línea Celular , Modelos Animales de Enfermedad , Papillomavirus Humano 16/ultraestructura , Humanos , Microscopía Electrónica , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología
4.
PLoS Pathog ; 15(5): e1007710, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31145755

RESUMEN

Modified vaccinia virus Ankara (MVA) is the leading poxvirus vector for development of vaccines against diverse infectious diseases. This distinction is based on high expression of proteins and good immunogenicity despite an inability to assemble infectious progeny in human cells, which together promote efficacy and safety. Nevertheless, the basis for the host-range restriction is unknown despite past systematic attempts to identify the relevant missing viral gene(s). The search for host-range factors is exacerbated by the large number of deletions, truncations and mutations that occurred during the long passage history of MVA in chicken embryo fibroblasts. By whole genome sequencing of a panel of recombinant host-range extended (HRE) MVAs generated by marker rescue with 40 kbp segments of vaccinia virus DNA, we identified serine protease inhibitor 1 (SPI-1) as one of several candidate host-range factors present in those viruses that gained the ability to replicate in human cells. Electron microscopy revealed that the interruption of morphogenesis in human cells infected with MVA occurred at a similar stage as that of a vaccinia virus strain WR SPI-1 deletion mutant. Moreover, the introduction of the SPI-1 gene into the MVA genome led to more than a 2-log enhancement of virus spread in human diploid MRC-5 cells, whereas deletion of the gene diminished the spread of HRE viruses by similar extents. Furthermore, MRC-5 cells stably expressing SPI-1 also enhanced replication of MVA. A role for additional host range genes was suggested by the restoration of MVA replication to a lower level relative to HRE viruses, particularly in other human cell lines. Although multiple sequence alignments revealed genetic changes in addition to SPI-1 common to the HRE MVAs, no evidence for their host-range function was found by analysis thus far. Our finding that SPI-1 is host range factor for MVA should simplify use of high throughput RNAi or CRISPR/Cas single gene methods to identify additional viral and human restriction elements.


Asunto(s)
Especificidad del Huésped/inmunología , Inhibidores de Serina Proteinasa/inmunología , Virus Vaccinia/fisiología , Vaccinia/virología , Vacunas Virales/inmunología , Replicación Viral , Células A549 , Vectores Genéticos/inmunología , Humanos , Inhibidores de Serina Proteinasa/genética , Vaccinia/inmunología , Vaccinia/prevención & control
5.
Nat Immunol ; 20(5): 602-612, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30886418

RESUMEN

Despite intense interest in antiviral T cell priming, the routes by which virions move in lymph nodes (LNs) are imperfectly understood. Current models fail to explain how virus-infected cells rapidly appear within the LN interior after viral infection. To better understand virion trafficking in the LN, we determined the locations of virions and infected cells after administration to mice of vaccinia virus or Zika virus. Notably, many rapidly infected cells in the LN interior were adjacent to LN conduits. Through the use of confocal and electron microscopy, we clearly visualized virions within conduits. Functionally, CD8+ T cells rapidly and preferentially associated with vaccinia virus-infected cells in the LN paracortex, which led to T cell activation in the LN interior. These results reveal that it is possible for even large virions to flow through LN conduits and infect dendritic cells within the T cell zone to prime CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Virión/inmunología , Animales , Linfocitos T CD8-positivos/virología , Femenino , Ganglios Linfáticos/ultraestructura , Ganglios Linfáticos/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Virus Vaccinia/inmunología , Virus Vaccinia/fisiología , Virión/fisiología , Virión/ultraestructura , Virosis/inmunología , Virosis/virología , Virus Zika/inmunología , Virus Zika/fisiología
6.
Proc Natl Acad Sci U S A ; 114(51): E11001-E11009, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29203656

RESUMEN

The long-standing inability to visualize connections between poxvirus membranes and cellular organelles has led to uncertainty regarding the origin of the viral membrane. Indeed, there has been speculation that viral membranes form de novo in cytoplasmic factories. Another possibility, that the connections are too short-lived to be captured by microscopy during a normal infection, motivated us to identify and characterize virus mutants that are arrested in assembly. Five conserved vaccinia virus proteins, referred to as Viral Membrane Assembly Proteins (VMAPs), that are necessary for formation of immature virions were found. Transmission electron microscopy studies of two VMAP deletion mutants had suggested retention of connections between viral membranes and the endoplasmic reticulum (ER). We now analyzed cells infected with each of the five VMAP deletion mutants by electron tomography, which is necessary to validate membrane continuity, in addition to conventional transmission electron microscopy. In all cases, connections between the ER and viral membranes were demonstrated by 3D reconstructions, supporting a role for the VMAPs in creating and/or stabilizing membrane scissions. Furthermore, coexpression of the viral reticulon-like transmembrane protein A17 and the capsid-like scaffold protein D13 was sufficient to form similar ER-associated viral structures in the absence of other major virion proteins. Determination of the mechanism of ER disruption during a normal VACV infection and the likely participation of both viral and cell proteins in this process may provide important insights into membrane dynamics.


Asunto(s)
Retículo Endoplásmico/metabolismo , Imagenología Tridimensional , Virus Vaccinia/fisiología , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus , Cápside/metabolismo , Cápside/ultraestructura , Tomografía con Microscopio Electrónico , Retículo Endoplásmico/ultraestructura , Mutación , Eliminación de Secuencia , Virus Vaccinia/ultraestructura , Proteínas de la Matriz Viral/genética , Virión
7.
J Virol ; 90(19): 8891-905, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27466413

RESUMEN

UNLABELLED: The anterograde pathway, from the endoplasmic reticulum through the trans-Golgi network to the cell surface, is utilized by trans-membrane and secretory proteins. The retrograde pathway, which directs traffic in the opposite direction, is used following endocytosis of exogenous molecules and recycling of membrane proteins. Microbes exploit both routes: viruses typically use the anterograde pathway for envelope formation prior to exiting the cell, whereas ricin and Shiga-like toxins and some nonenveloped viruses use the retrograde pathway for cell entry. Mining a human genome-wide RNA interference (RNAi) screen revealed a need for multiple retrograde pathway components for cell-to-cell spread of vaccinia virus. We confirmed and extended these results while discovering that retrograde trafficking was required for virus egress rather than entry. Retro-2, a specific retrograde trafficking inhibitor of protein toxins, potently prevented spread of vaccinia virus as well as monkeypox virus, a human pathogen. Electron and confocal microscopy studies revealed that Retro-2 prevented wrapping of virions with an additional double-membrane envelope that enables microtubular transport, exocytosis, and actin polymerization. The viral B5 and F13 protein components of this membrane, which are required for wrapping, normally colocalize in the trans-Golgi network. However, only B5 traffics through the secretory pathway, suggesting that F13 uses another route to the trans-Golgi network. The retrograde route was demonstrated by finding that F13 was largely confined to early endosomes and failed to colocalize with B5 in the presence of Retro-2. Thus, vaccinia virus makes novel use of the retrograde transport system for formation of the viral wrapping membrane. IMPORTANCE: Efficient cell-to-cell spread of vaccinia virus and other orthopoxviruses depends on the wrapping of infectious particles with a double membrane that enables microtubular transport, exocytosis, and actin polymerization. Interference with wrapping or subsequent steps results in severe attenuation of the virus. Some previous studies had suggested that the wrapping membrane arises from the trans-Golgi network, whereas others suggested an origin from early endosomes. Some nonenveloped viruses use retrograde trafficking for entry into the cell. In contrast, we provided evidence that retrograde transport from early endosomes to the trans-Golgi network is required for the membrane-wrapping step in morphogenesis of vaccinia virus and egress from the cell. The potent in vitro inhibition of this step by the drug Retro-2 suggests that derivatives with enhanced pharmacological properties might serve as useful antipoxviral agents.


Asunto(s)
Endosomas/metabolismo , Virus Vaccinia/fisiología , Virión/metabolismo , Liberación del Virus , Red trans-Golgi/metabolismo , Transporte Biológico , Membrana Celular , Exocitosis , Células HeLa , Humanos , Microscopía Confocal , Microscopía Electrónica , Monkeypox virus/metabolismo , Monkeypox virus/fisiología , Virus Vaccinia/metabolismo
8.
Cell Rep ; 14(9): 2084-2091, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26923595

RESUMEN

Poxviruses are enveloped DNA viruses that replicate within the cytoplasm. The first viral structures are crescents and spherical particles, with a lipoprotein membrane bilayer, that are thought to be derived from the ER. We determined that A17, a conserved viral transmembrane protein essential for crescent formation, forms homo-oligomers and shares topological features with cellular reticulon-like proteins. The latter cell proteins promote membrane curvature and contribute to the tubular structure of the ER. When the purified A17 protein was incorporated into liposomes, 25 nm diameter vesicles and tubules formed at low and high A17 concentrations, respectively. In addition, intracellular expression of A17 in the absence of other viral structural proteins transformed the ER into aggregated three-dimensional (3D) tubular networks. We suggest that A17 is a viral reticulon-like protein that contributes to curvature during biogenesis of the poxvirus membrane.


Asunto(s)
Estructuras de la Membrana Celular/ultraestructura , Poxviridae/genética , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Estructuras de la Membrana Celular/virología , Chlorocebus aethiops , Secuencia Conservada , Retículo Endoplásmico/ultraestructura , Retículo Endoplásmico/virología , Proteínas Virales/química
9.
Virology ; 452-453: 59-66, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24606683

RESUMEN

Some orthopoxviruses including cowpox virus embed virus particles in dense bodies, comprised of the A-type inclusion (ATI) protein, which may provide long-term environmental protection. This strategy could be beneficial if the host population is sparse or spread is inefficient or indirect. However, the formation of ATI may be neutral or disadvantageous for orthopoxviruses that rely on direct respiratory spread. Disrupted ATI open reading frames in orthopoxviruses such as variola virus, the agent of smallpox, and monkeypox virus suggests that loss of this feature provided positive selection. To test this hypothesis, we constructed cowpox virus mutants with deletion of the ATI gene or another gene required for embedding virions. The ATI deletion mutant caused greater weight loss and higher replication in the respiratory tract than control viruses, supporting our hypothesis. Deletion of the gene for embedding virions had a lesser effect, possibly due to known additional functions of the encoded protein.


Asunto(s)
Evolución Biológica , Virus de la Viruela Vacuna/fisiología , Viruela Vacuna/virología , Eliminación de Gen , Orthopoxvirus/genética , Proteínas Virales/genética , Replicación Viral , Animales , Virus de la Viruela Vacuna/genética , Humanos , Cuerpos de Inclusión Viral/virología , Ratones , Ratones Endogámicos BALB C , Orthopoxvirus/fisiología , Proteínas Virales/metabolismo
10.
J Virol ; 87(22): 12313-26, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24027302

RESUMEN

Crescents consisting of a single lipoprotein membrane with an external protein scaffold comprise the initial structural elements of poxvirus morphogenesis. Crescents enlarge to form spherical immature virions, which enclose viroplasm consisting of proteins destined to form the cores of mature virions. Previous studies suggest that the L2 protein participates in the recruitment of endoplasmic reticulum (ER)-derived membranes to form immature virions within assembly sites of cytoplasmic factories. Here we show that L2 interacts with the previously uncharacterized 42-amino-acid A30.5 protein. An open reading frame similar in size to the one encoding A30.5 is at the same genome location in representatives of all chordopoxvirus genera. A30.5 has a putative transmembrane domain and colocalized with markers of the endoplasmic reticulum and with L2. By constructing a complementing cell line expressing A30.5, we isolated a deletion mutant virus that exhibits a defect in morphogenesis in normal cells. Large electron-dense cytoplasmic inclusions and clusters of scaffold protein-coated membranes that resemble crescents and immature virions devoid of viroplasm were seen in place of normal structures. Crescent-shaped membranes were continuous with the endoplasmic reticulum membrane and oriented with the convex scaffold protein-coated side facing the lumen, while clusters of completed spherical immature-virion-like forms were trapped within the expanded lumen. Immature-virion-like structures were more abundant in infected RK-13 cells than in BS-C-1 or HeLa cells, in which cytoplasmic inclusions were decorated with scaffold protein-coated membrane arcs. We suggest that the outer surface of the poxvirus virion is derived from the luminal side of the ER membrane.


Asunto(s)
Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Virus Vaccinia/fisiología , Vaccinia/metabolismo , Proteínas Virales/metabolismo , Virión/fisiología , Ensamble de Virus , Replicación Viral , Secuencia de Aminoácidos , Animales , Western Blotting , Células Cultivadas , Chlorocebus aethiops , Citoplasma/metabolismo , Citoplasma/virología , Células HeLa , Humanos , Inmunoprecipitación , Riñón/citología , Riñón/metabolismo , Riñón/virología , Espectrometría de Masas , Datos de Secuencia Molecular , Conejos , Homología de Secuencia de Aminoácido , Vaccinia/virología
11.
J Virol ; 87(18): 10195-206, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864611

RESUMEN

The apparent de novo formation of viral membranes within cytoplasmic factories is a mysterious, poorly understood first step in poxvirus morphogenesis. Genetic studies identified several viral proteins essential for membrane formation and the assembly of immature virus particles. Their repression results in abortive replication with the accumulation of dense masses of viroplasm. In the present study, we further characterized one of these proteins, A11, and investigated its association with cellular and viral membranes under normal and abortive replication conditions. We discovered that A11 colocalized in cytoplasmic factories with the endoplasmic reticulum (ER) and L2, another viral protein required for morphogenesis. Confocal microscopy and subcellular fractionation indicated that A11 was not membrane associated in uninfected cells, whereas L2 still colocalized with the ER. Cell-free transcription and translation experiments indicated that both A11 and L2 are tail-anchored proteins that associate posttranslationally with membranes and likely require specific cytoplasmic targeting chaperones. Transmission electron microscopy indicated that A11, like L2, associated with crescent membranes and immature virions during normal infection and with vesicles and tubules near masses of dense viroplasm during abortive infection in the absence of the A17 or A14 protein component of viral membranes. When the synthesis of A11 was repressed, "empty" immature-virion-like structures formed in addition to masses of viroplasm. The immature-virion-like structures were labeled with antibodies to A17 and to the D13 scaffold protein and were closely associated with calnexin-labeled ER. These studies revealed similarities and differences between A11 and L2, both of which may be involved in the recruitment of the ER for virus assembly.


Asunto(s)
Retículo Endoplásmico/virología , Virus Vaccinia/fisiología , Proteínas Virales/metabolismo , Ensamble de Virus , Replicación Viral , Línea Celular , Humanos , Microscopía Electrónica de Transmisión , Unión Proteica , Virión/ultraestructura
12.
J Virol ; 87(19): 10700-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23885081

RESUMEN

The A19L open reading frame of vaccinia virus encodes a 9-kDa protein that is conserved in all sequenced chordopoxviruses, yet until now it has not been specifically characterized in any species. We appended an epitope tag after the start codon of the A19L open reading frame without compromising infectivity. The protein was synthesized after viral DNA replication and was phosphorylated independently of the vaccinia virus F10 kinase. The A19 protein was present in purified virions and was largely resistant to nonionic detergent extraction, suggesting a location within the core. A conditional lethal mutant virus was constructed by placing the A19 open reading frame under the control of the Escherichia coli lac repressor system. A19 synthesis and infectious virus formation were dependent on inducer. In the absence of inducer, virion morphogenesis was interrupted, and spherical dense particles that had greatly reduced amounts of the D13 scaffold accumulated in place of barrel-shaped mature virions. The infectivity of purified A19-deficient particles was more than 2 log units less than that of A19-containing virions. Nevertheless, the A19-deficient particles contained DNA, and except for the absence of A19 and decreased core protein processing, they appeared to have a similar protein composition as A19-containing virions. Thus, the A19 protein participates in the maturation of immature vaccinia virus virions to infectious particles.


Asunto(s)
Virus Vaccinia/patogenicidad , Vaccinia/virología , Proteínas Virales/metabolismo , Virión/patogenicidad , Ensamble de Virus , Animales , Western Blotting , Chlorocebus aethiops , Replicación del ADN , ADN Viral , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Riñón/citología , Riñón/metabolismo , Riñón/virología , Mutación/genética , Sistemas de Lectura Abierta , Fosforilación , Vaccinia/genética , Vaccinia/metabolismo , Proteínas Virales/genética , Replicación Viral
13.
J Virol ; 87(3): 1861-71, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23192873

RESUMEN

Assembly of the poxvirus immature virion (IV) membrane is a poorly understood event that occurs within the cytoplasm. At least eight viral proteins participate in formation of the viral membrane. Of these, A14, A17, and D13 are structural components whereas A6, A11, F10, H7, and L2 participate in membrane biogenesis. L2, the object of this study, is conserved in all chordopoxviruses, expressed early in infection, and associated with the endoplasmic reticulum (ER) throughout the cell and at the edges of crescent-shaped IV precursors. Previous studies with an inducible L2 mutant revealed abortive formation of the crescent membrane. However, possible low-level L2 synthesis under nonpermissive conditions led to ambiguity in interpretation. Here, we constructed a cell line that expresses L2, which allowed the creation of an L2-deletion mutant. In noncomplementing cells, replication was aborted prior to formation of mature virions and two types of aberrant structures were recognized. One consisted of short crescents, at the surface of dense masses of viroplasm, which were labeled with antibodies to the A11, A14, A17, and D13 proteins. The other structure consisted of "empty" IV-like membranes, also labeled with antibodies to the viral proteins, which appeared to be derived from adjacent calnexin-containing ER. A subset of 25 proteins examined, exemplified by components of the entry-fusion complex, were greatly diminished in amount. The primary role of L2 may be to recruit ER and modulate its transformation to viral membranes in juxtaposition with the viroplasm, simultaneously preventing the degradation of viral proteins dependent on viral membranes for stability.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Virus Vaccinia/fisiología , Ensamble de Virus , Línea Celular , Humanos , Eliminación de Secuencia , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
14.
PLoS Pathog ; 7(12): e1002446, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22194690

RESUMEN

For many viruses, one or two proteins allow cell attachment and entry, which occurs through the plasma membrane or following endocytosis at low pH. In contrast, vaccinia virus (VACV) enters cells by both neutral and low pH routes; four proteins mediate cell attachment and twelve that are associated in a membrane complex and conserved in all poxviruses are dedicated to entry. The aim of the present study was to determine the roles of cellular and viral proteins in initial stages of entry, specifically fusion of the membranes of the mature virion and cell. For analysis of the role of cellular components, we used well characterized inhibitors and measured binding of a recombinant VACV virion containing Gaussia luciferase fused to a core protein; viral and cellular membrane lipid mixing with a self-quenching fluorescent probe in the virion membrane; and core entry with a recombinant VACV expressing firefly luciferase and electron microscopy. We determined that inhibitors of tyrosine protein kinases, dynamin GTPase and actin dynamics had little effect on binding of virions to cells but impaired membrane fusion, whereas partial cholesterol depletion and inhibitors of endosomal acidification and membrane blebbing had a severe effect at the later stage of core entry. To determine the role of viral proteins, virions lacking individual membrane components were purified from cells infected with members of a panel of ten conditional-lethal inducible mutants. Each of the entry protein-deficient virions had severely reduced infectivity and except for A28, L1 and L5 greatly impaired membrane fusion. In addition, a potent neutralizing L1 monoclonal antibody blocked entry at a post-membrane lipid-mixing step. Taken together, these results suggested a 2-step entry model and implicated an unprecedented number of viral proteins and cellular components involved in signaling and actin rearrangement for initiation of virus-cell membrane fusion during poxvirus entry.


Asunto(s)
Proteínas de la Membrana/metabolismo , Virus Vaccinia/fisiología , Proteínas Virales/metabolismo , Virión/fisiología , Internalización del Virus , Animales , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/virología , Endocitosis , Humanos , Concentración de Iones de Hidrógeno , Fusión de Membrana , Proteínas de la Membrana/genética , Virus Vaccinia/metabolismo , Virión/metabolismo , Virión/patogenicidad
15.
J Virol ; 85(23): 12431-41, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21917978

RESUMEN

The initial step in poxvirus morphogenesis, the formation of crescent membranes, occurs within cytoplasmic factories. L2 is one of several vaccinia virus proteins known to be necessary for formation of crescents and the only one synthesized early in infection. Virus replication was unaffected when the L2R open reading frame was replaced by L2R containing an N-terminal epitope tag while retaining the original promoter. L2 colocalized with the endoplasmic reticulum (ER) protein calnexin throughout the cytoplasm of infected and transfected cells. Topological studies indicated that the N terminus of L2 is exposed to the cytoplasm with the hydrophobic C terminus anchored in the ER. Using immunogold labeling and electron microscopy, L2 was detected in tubular membranes outside factories and inside factories near crescents and close to the edge or rim of crescents; a similar labeling pattern was found for the ER luminal protein disulfide isomerase (PDI). The phenotype of L2 conditional lethal mutants and the localization of L2 suggest that it participates in elongation of crescents by the addition of ER membrane to the growing edge. Small amounts of L2 and PDI were detected within immature and mature virions, perhaps trapped during assembly. The repression of L2, as well as A11 and A17, two other proteins that are required for viral crescent formation, profoundly decreased the stability of a subset of viral membrane proteins including those comprising the entry-fusion complex. To avoid degradation, these unstable membrane proteins may need to directly insert into the viral membrane or be rapidly shunted there from the ER.


Asunto(s)
Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Virus Vaccinia/patogenicidad , Vaccinia/virología , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo , Virión/crecimiento & desarrollo , Replicación Viral , Western Blotting , Citoplasma/metabolismo , Citoplasma/virología , Células HeLa , Humanos , Estabilidad Proteica , Vaccinia/metabolismo , Virión/metabolismo , Virión/patogenicidad , Ensamble de Virus
16.
PLoS One ; 6(2): e17248, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21347205

RESUMEN

Vaccinia virus (VACV), a member of the chordopox subfamily of the Poxviridae, abortively infects insect cells. We have investigated VACV infection of Drosophila S2 cells, which are useful for protein expression and genome-wide RNAi screening. Biochemical and electron microscopic analyses indicated that VACV entry into Drosophila S2 cells depended on the VACV multiprotein entry-fusion complex but appeared to occur exclusively by a low pH-dependent endocytic mechanism, in contrast to both neutral and low pH entry pathways used in mammalian cells. Deep RNA sequencing revealed that the entire VACV early transcriptome, comprising 118 open reading frames, was robustly expressed but neither intermediate nor late mRNAs were made. Nor was viral late protein synthesis or inhibition of host protein synthesis detected by pulse-labeling with radioactive amino acids. Some reduction in viral early proteins was noted by Western blotting. Nevertheless, synthesis of the multitude of early proteins needed for intermediate gene expression was demonstrated by transfection of a plasmid containing a reporter gene regulated by an intermediate promoter. In addition, expression of a reporter gene with a late promoter was achieved by cotransfection of intermediate genes encoding the late transcription factors. The requirement for transfection of DNA templates for intermediate and late gene expression indicated a defect in viral genome replication in VACV-infected S2 cells, which was confirmed by direct analysis. Furthermore, VACV-infected S2 cells did not support the replication of a transfected plasmid, which occurs in mammalian cells and is dependent on all known viral replication proteins, indicating a primary restriction of DNA synthesis.


Asunto(s)
Replicación del ADN , ADN Viral/biosíntesis , Drosophila/citología , Endocitosis , Transcripción Genética , Virus Vaccinia/fisiología , Internalización del Virus , Animales , Drosophila/virología , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Concentración de Iones de Hidrógeno , Análisis de Secuencia de ARN , Transcriptoma , Virus Vaccinia/genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética
17.
J Virol ; 85(6): 2504-11, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21228235

RESUMEN

Morphogenesis of vaccinia virus begins with the appearance of crescent-shaped membrane precursors of immature virions in cytoplasmic factories. During the initial characterization of the product of the L2R reading frame, we discovered that it plays an important role in crescent formation. The L2 protein was expressed early in infection and was associated with the detergent-soluble membrane fraction of mature virions, consistent with two potential membrane-spanning domains. All chordopoxviruses have L2 homologs, suggesting an important function. Indeed, we were unable to isolate an infectious L2R deletion mutant. Consequently, we constructed an inducible mutant with a conditional lethal phenotype. When L2 expression was repressed, proteolytic processing of the major core proteins and the A17 protein, which is an essential component of the immature virion membrane, failed to occur, suggesting an early block in viral morphogenesis. At 8 h after infection in the presence of inducer, immature and mature virions were abundantly seen by electron microscopy. In contrast, those structures were rare in the absence of inducer and were replaced by large, dense aggregates of viroplasm. A minority of these aggregates had short spicule-coated membranes, which resembled the beginnings of crescent formation, at their periphery. These short membrane segments at the edge of the dense viroplasm increased in number at later times, and some immature virions were seen. Although the L2 protein was not detected under nonpermissive conditions, minute amounts could account for stunted and delayed viral membrane formation. These findings suggested that L2 is required for the formation or elongation of crescent membranes.


Asunto(s)
Virus Vaccinia/química , Virus Vaccinia/fisiología , Proteínas Virales/metabolismo , Virión/química , Virión/metabolismo , Ensamble de Virus , Secuencia de Aminoácidos , Línea Celular , Eliminación de Gen , Genes Esenciales , Genes Virales , Humanos , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Alineación de Secuencia , Virus Vaccinia/genética , Ensayo de Placa Viral , Proteínas Virales/genética
18.
J Virol ; 84(15): 7592-602, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20484506

RESUMEN

Some orthopoxviruses, e.g., the cowpox, ectromelia, and raccoonpox viruses, form large, discrete cytoplasmic inclusions within which mature virions (MVs) are embedded by a process called occlusion. These inclusions, which may protect occluded MVs in the environment, are composed of aggregates of the A-type inclusion protein (ATIp), which is truncated in orthopoxviruses such as vaccinia virus (VACV) and variola virus that fail to form inclusions. In addition to an intact ATIp, occlusion requires the A26 protein (A26p). Although VACV contains a functional A26p, determined by complementation of a cowpox virus occlusion-defective mutant, its role in occlusion was unknown. We found that restoration of the full-length ATI gene was sufficient for VACV inclusion formation and the ensuing occlusion of MVs. A26p was present in inclusions even when virion assembly was inhibited, suggesting a direct interaction of A26p with ATIp. Analysis of a panel of ATIp mutants indicated that the C-terminal repeat region was required for inclusion formation and the N-terminal domain for interaction with A26p and occlusion. A26p is tethered to MVs via interaction with the A27 protein (A27p); A27p was not required for association of A26p with ATIp but was necessary for occlusion. In addition, the C-terminal domain of A26p, which mediates A26p-A27p interactions, was necessary but insufficient for occlusion. Taken together, the data suggest a model for occlusion in which A26p has a bridging role between ATIp and A27p, and A27p provides a link to the MV membrane.


Asunto(s)
Proteínas Portadoras/metabolismo , Cuerpos de Inclusión Viral , Mapeo de Interacción de Proteínas , Virus Vaccinia/fisiología , Proteínas Virales de Fusión/metabolismo , Proteínas Virales/metabolismo , Ensamble de Virus , Animales , Línea Celular , Chlorocebus aethiops , Humanos , Proteínas de la Membrana , Unión Proteica
19.
J Virol ; 83(18): 9140-50, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570860

RESUMEN

Infectious poxvirus particles are unusual in that they are brick shaped and lack symmetry. Nevertheless, an external honeycomb lattice comprised of a capsid-like protein dictates the spherical shape and size of immature poxvirus particles. In the case of vaccinia virus, trimers of 63-kDa D13 polypeptides form the building blocks of the lattice. In the present study, we addressed two questions: how D13, which has no transmembrane domain, associates with the immature virion (IV) membrane to form the lattice structure and how this scaffold is removed during the subsequent stage of morphogenesis. Interaction of D13 with the A17 membrane protein was demonstrated by immunoaffinity purification and Western blot analysis. In addition, the results of immunogold electron microscopy indicated a close association of A17 and D13 in crescents, as well as in vesicular structures when crescent formation was prevented. Further studies indicated that binding of A17 to D13 was abrogated by truncation of the N-terminal segment of A17. The N-terminal region of A17 was also required for the formation of crescent and IV structures. Disassembly of the D13 scaffold correlated with the processing of A17 by the I7 protease. When I7 expression was repressed, D13 was retained on aberrant virus particles. Furthermore, the morphogenesis of IVs to mature virions was blocked by mutation of the N-terminal but not the C-terminal cleavage site on A17. Taken together, these data indicate that A17 and D13 interactions regulate the assembly and disassembly of the IV scaffold.


Asunto(s)
Proteínas de la Cápside/metabolismo , Virus Vaccinia/fisiología , Virión/metabolismo , Ensamble de Virus , Cápside/química , Cápside/metabolismo , Proteínas de la Cápside/química , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Tamaño de la Partícula , Unión Proteica , Proteínas Virales/metabolismo , Virión/química
20.
Virology ; 386(2): 478-85, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19217136

RESUMEN

The vaccinia virus E6R gene (VACVWR062) is conserved in all members of the poxvirus family and encodes a protein associated with the mature virion. We confirmed this association and provided evidence for an internal location. An inducible mutant that conditionally expresses E6 was constructed. In the absence of inducer, plaque formation and virus production were severely inhibited in several cell lines, whereas some replication occurred in others. This difference could be due to variation in the stringency of repression, since we could not isolate a stable deletion mutant even in the more "permissive" cells. Under non-permissive conditions, viral late proteins were synthesized but processing of core proteins was inefficient, indicative of an assembly block. Transmission electron microscopy of sections of cells infected with the mutant in the absence of inducer revealed morphogenetic defects with crescents and empty immature virions adjacent to dense inclusions of viroplasm. Mature virions were infrequent and cores appeared to have lucent centers.


Asunto(s)
Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo , Virión/ultraestructura , Ensamble de Virus , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Secuencia Conservada , ADN Viral/genética , Regulación Viral de la Expresión Génica , Genes Esenciales , Células HeLa , Humanos , Datos de Secuencia Molecular , Virus Vaccinia/genética , Virus Vaccinia/fisiología , Virus Vaccinia/ultraestructura , Células Vero , Proteínas Virales/genética , Virión/genética , Virión/metabolismo , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...