Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Virol ; 98(3): e0153623, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38315014

RESUMEN

African swine fever (ASF) is a highly contagious viral disease that affects domestic and wild pigs. The causative agent of ASF is African swine fever virus (ASFV), a large double-stranded DNA virus with a complex virion structure. Among the various proteins encoded by ASFV, A137R is a crucial structural protein associated with its virulence. However, the structure and molecular mechanisms underlying the functions of A137R remain largely unknown. In this study, we present the structure of A137R determined by cryogenic electron microscopy single-particle reconstruction, which reveals that A137R self-oligomerizes to form a dodecahedron-shaped cage composed of 60 polymers. The dodecahedron is literally equivalent to a T = 1 icosahedron where the icosahedral vertexes are located in the center of each dodecahedral facet. Within each facet, five A137R protomers are arranged in a head-to-tail orientation with a long N-terminal helix forming the edge through which adjacent facets stitch together to form the dodecahedral cage. Combining structural analysis and biochemical evidence, we demonstrate that the N-terminal domain of A137R is crucial and sufficient for mediating the assembly of the dodecahedron. These findings imply the role of A137R cage as a core component in the icosahedral ASFV virion and suggest a promising molecular scaffold for nanotechnology applications. IMPORTANCE: African swine fever (ASF) is a lethal viral disease of pigs caused by African swine fever virus (ASFV). No commercial vaccines and antiviral treatments are available for the prevention and control of the disease. A137R is a structural protein of ASFV that is associated with its virulence. The discovery of the dodecahedron-shaped cage structure of A137R in this study is of great importance in understanding ASFV pathogenicity. This finding sheds light on the molecular mechanisms underlying the functions of A137R. Furthermore, the dodecahedral cage formed by A137R shows promise as a molecular scaffold for nanoparticle vectors. Overall, this study provides valuable insights into the structure and function of A137R, contributing to our understanding of ASFV and potentially opening up new avenues for the development of vaccines or treatments for ASF.


Asunto(s)
Virus de la Fiebre Porcina Africana , Porcinos , Proteínas Estructurales Virales , Animales , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/química , Virus de la Fiebre Porcina Africana/crecimiento & desarrollo , Virus de la Fiebre Porcina Africana/patogenicidad , Virus de la Fiebre Porcina Africana/ultraestructura , Microscopía por Crioelectrón , Relación Estructura-Actividad , Porcinos/virología , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/metabolismo , Proteínas Estructurales Virales/ultraestructura , Virión/química , Virión/metabolismo , Virión/ultraestructura , Virulencia
2.
Viruses ; 13(9)2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34578379

RESUMEN

A variety of immunolabeling procedures for both light and electron microscopy were used to examine the cellular origins of the host membranes supporting the SARS-CoV-2 replication complex. The endoplasmic reticulum has long been implicated as a source of membrane for the coronavirus replication organelle. Using dsRNA as a marker for sites of viral RNA synthesis, we provide additional evidence supporting ER as a prominent source of membrane. In addition, we observed a rapid fragmentation of the Golgi apparatus which is visible by 6 h and complete by 12 h post-infection. Golgi derived lipid appears to be incorporated into the replication organelle although protein markers are dispersed throughout the infected cell. The mechanism of Golgi disruption is undefined, but chemical disruption of the Golgi apparatus by brefeldin A is inhibitory to viral replication. A search for an individual SARS-CoV-2 protein responsible for this activity identified at least five viral proteins, M, S, E, Orf6, and nsp3, that induced Golgi fragmentation when expressed in eukaryotic cells. Each of these proteins, as well as nsp4, also caused visible changes to ER structure as shown by correlative light and electron microscopy (CLEM). Collectively, these results imply that specific disruption of the Golgi apparatus is a critical component of coronavirus replication.


Asunto(s)
Retículo Endoplásmico/virología , Aparato de Golgi/virología , SARS-CoV-2/fisiología , Replicación Viral , Animales , Chlorocebus aethiops , Proteínas M de Coronavirus/fisiología , Proteínas M de Coronavirus/ultraestructura , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/ultraestructura , Humanos , Membranas Intracelulares/ultraestructura , Membranas Intracelulares/virología , Microscopía Electrónica , SARS-CoV-2/ultraestructura , Células Vero , Proteínas Estructurales Virales/fisiología , Proteínas Estructurales Virales/ultraestructura
3.
Int J Mol Sci ; 22(16)2021 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-34445741

RESUMEN

(1) Background: coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been linked to hematological dysfunctions, but there are little experimental data that explain this. Spike (S) and Nucleoprotein (N) proteins have been putatively associated with these dysfunctions. In this work, we analyzed the recruitment of hemoglobin (Hb) and other metabolites (hemin and protoporphyrin IX-PpIX) by SARS-Cov2 proteins using different approaches. (2) Methods: shotgun proteomics (LC-MS/MS) after affinity column adsorption identified hemin-binding SARS-CoV-2 proteins. The parallel synthesis of the peptides technique was used to study the interaction of the receptor bind domain (RBD) and N-terminal domain (NTD) of the S protein with Hb and in silico analysis to identify the binding motifs of the N protein. The plaque assay was used to investigate the inhibitory effect of Hb and the metabolites hemin and PpIX on virus adsorption and replication in Vero cells. (3) Results: the proteomic analysis by LC-MS/MS identified the S, N, M, Nsp3, and Nsp7 as putative hemin-binding proteins. Six short sequences in the RBD and 11 in the NTD of the spike were identified by microarray of peptides to interact with Hb and tree motifs in the N protein by in silico analysis to bind with heme. An inhibitory effect in vitro of Hb, hemin, and PpIX at different levels was observed. Strikingly, free Hb at 1mM suppressed viral replication (99%), and its interaction with SARS-CoV-2 was localized into the RBD region of the spike protein. (4) Conclusions: in this study, we identified that (at least) five proteins (S, N, M, Nsp3, and Nsp7) of SARS-CoV-2 recruit Hb/metabolites. The motifs of the RDB of SARS-CoV-2 spike, which binds Hb, and the sites of the heme bind-N protein were disclosed. In addition, these compounds and PpIX block the virus's adsorption and replication. Furthermore, we also identified heme-binding motifs and interaction with hemin in N protein and other structural (S and M) and non-structural (Nsp3 and Nsp7) proteins.


Asunto(s)
COVID-19/etiología , Hemoglobinas/metabolismo , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas Estructurales Virales/metabolismo , COVID-19/sangre , Hemina/metabolismo , Hemoglobinas/ultraestructura , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , Dominios Proteicos , Proteómica , Protoporfirinas/metabolismo , SARS-CoV-2/patogenicidad , Proteínas no Estructurales Virales/ultraestructura , Proteínas Estructurales Virales/ultraestructura , Acoplamiento Viral , Replicación Viral
4.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34143202

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a causative agent of the coronavirus disease (COVID-19), is a part of the $\beta $-Coronaviridae family. The virus contains five major protein classes viz., four structural proteins [nucleocapsid (N), membrane (M), envelop (E) and spike glycoprotein (S)] and replicase polyproteins (R), synthesized as two polyproteins (ORF1a and ORF1ab). Due to the severity of the pandemic, most of the SARS-CoV-2-related research are focused on finding therapeutic solutions. However, studies on the sequences and structure space throughout the evolutionary time frame of viral proteins are limited. Besides, the structural malleability of viral proteins can be directly or indirectly associated with the dysfunctionality of the host cell proteins. This dysfunctionality may lead to comorbidities during the infection and may continue at the post-infection stage. In this regard, we conduct the evolutionary sequence-structure analysis of the viral proteins to evaluate their malleability. Subsequently, intrinsic disorder propensities of these viral proteins have been studied to confirm that the short intrinsically disordered regions play an important role in enhancing the likelihood of the host proteins interacting with the viral proteins. These interactions may result in molecular dysfunctionality, finally leading to different diseases. Based on the host cell proteins, the diseases are divided in two distinct classes: (i) proteins, directly associated with the set of diseases while showing similar activities, and (ii) cytokine storm-mediated pro-inflammation (e.g. acute respiratory distress syndrome, malignancies) and neuroinflammation (e.g. neurodegenerative and neuropsychiatric diseases). Finally, the study unveils that males and postmenopausal females can be more vulnerable to SARS-CoV-2 infection due to the androgen-mediated protein transmembrane serine protease 2.


Asunto(s)
COVID-19/genética , Genoma Viral/genética , Conformación Proteica , SARS-CoV-2/ultraestructura , COVID-19/virología , Proteínas de la Envoltura de Coronavirus/genética , Proteínas de la Envoltura de Coronavirus/ultraestructura , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/ultraestructura , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/ultraestructura , Proteinas del Complejo de Replicasa Viral/genética , Proteinas del Complejo de Replicasa Viral/ultraestructura , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/ultraestructura
5.
PLoS Pathog ; 17(3): e1009396, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33730056

RESUMEN

Mosquito viruses cause unpredictable outbreaks of disease. Recently, several unassigned viruses isolated from mosquitoes, including the Omono River virus (OmRV), were identified as totivirus-like viruses, with features similar to those of the Totiviridae family. Most reported members of this family infect fungi or protozoans and lack an extracellular life cycle stage. Here, we identified a new strain of OmRV and determined high-resolution structures for this virus using single-particle cryo-electron microscopy. The structures feature an unexpected protrusion at the five-fold vertex of the capsid. Disassociation of the protrusion could result in several conformational changes in the major capsid. All these structures, together with some biological results, suggest the protrusions' associations with the extracellular transmission of OmRV.


Asunto(s)
Microscopía por Crioelectrón/métodos , Virus ARN Bicatenario/ultraestructura , Proteínas Estructurales Virales/ultraestructura , Aedes/virología , Animales
6.
Nat Commun ; 10(1): 4840, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31649265

RESUMEN

Assembly of tailed bacteriophages and herpesviruses starts with formation of procapsids (virion precursors without DNA). Scaffolding proteins (SP) drive assembly by chaperoning the major capsid protein (MCP) to build an icosahedral lattice. Here we report near-atomic resolution cryo-EM structures of the bacteriophage SPP1 procapsid, the intermediate expanded procapsid with partially released SPs, and the mature capsid with DNA. In the intermediate state, SPs are bound only to MCP pentons and to adjacent subunits from hexons. SP departure results in the expanded state associated with unfolding of the MCP N-terminus and straightening of E-loops. The newly formed extensive inter-capsomere bonding appears to compensate for release of SPs that clasp MCP capsomeres together. Subsequent DNA packaging instigates bending of MCP A domain loops outwards, closing the hexons central opening and creating the capsid auxiliary protein binding interface. These findings provide a molecular basis for the sequential structural rearrangements during viral capsid maturation.


Asunto(s)
Bacteriófagos/ultraestructura , Proteínas de la Cápside/ultraestructura , Cápside/ultraestructura , Ensamble de Virus , Bacteriófagos/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Proteínas Estructurales Virales/metabolismo , Proteínas Estructurales Virales/ultraestructura
7.
J Gen Virol ; 98(11): 2837-2849, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29035172

RESUMEN

Inside the virions of α-herpesviruses, tegument protein pUL25 anchors the tegument to capsid vertices through direct interactions with tegument proteins pUL17 and pUL36. In addition to promoting virion assembly, both pUL25 and pUL36 are critical for intracellular microtubule-dependent capsid transport. Despite these essential roles during infection, the stoichiometry and precise organization of pUL25 and pUL36 on the capsid surface remain controversial due to the insufficient resolution of existing reconstructions from cryo-electron microscopy (cryoEM). Here, we report a three-dimensional (3D) icosahedral reconstruction of pseudorabies virus (PRV), a varicellovirus of the α-herpesvirinae subfamily, obtained by electron-counting cryoEM at 4.9 Å resolution. Our reconstruction resolves a dimer of pUL25 forming a capsid-associated tegument complex with pUL36 and pUL17 through a coiled coil helix bundle, thus correcting previous misinterpretations. A comparison between reconstructions of PRV and the γ-herpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV) reinforces their similar architectures and establishes important subfamily differences in the capsid-tegument interface.


Asunto(s)
Herpesvirus Suido 1/química , Herpesvirus Suido 1/ultraestructura , Multimerización de Proteína , Proteínas Estructurales Virales/análisis , Proteínas Estructurales Virales/ultraestructura , Virión/química , Virión/ultraestructura , Microscopía por Crioelectrón , Imagenología Tridimensional , Unión Proteica
8.
J Invertebr Pathol ; 144: 18-23, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28057460

RESUMEN

Bombyx mori bidensiovirus (BmBDV) is a species of Bidensovirus that has been was placed into a new genus within the new family Bidnaviridae by the International Committee on Taxonomy of Viruses. BmBDV causes fatal flacherie disease in silkworms, which causes large losses to the sericulture industry. BmBDV contains two sets of complementary linear single-stranded DNAs of approximately 6.5kb (viral DNA 1, VD1) and 6.0kb (viral DNA 2, VD2). VD1 and VD2 are encapsidated in separate icosahedral non-enveloped capsids, which are similar in size and shape. However, the strategies used to express BmBDV structural proteins remains unclear. In this work, a total of six structural proteins were separated by two-dimensional electrophoresis and shown to be encoded by the BmBDV VP gene via mass spectrometry. The transmission electron microscopy results showed that co-expression of the BmBDV VP and SP structural proteins in Spodoptera frugiperda sf9 cells resulted in the formation of 22-24nm virus-like particles. Furthermore, a mutation of the major structural protein-encoding VP gene, in which the second in-frame ATG codon was mutated to GCG, abrogated the production of several structural proteins, indicating that this strategy of expressing BmBDV VP is dependent on a leaky scanning translation mechanism.


Asunto(s)
Densovirus/fisiología , Proteínas Estructurales Virales/fisiología , Secuencia de Aminoácidos , Animales , Western Blotting , Bombyx/virología , Electroforesis en Gel Bidimensional , Microscopía Electrónica de Transmisión , Células Sf9 , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteínas Estructurales Virales/ultraestructura
9.
Trends Microbiol ; 25(6): 438-446, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28094093

RESUMEN

Recent developments of rational strategies for the design of antiviral therapies, including monoclonal antibodies (mAbs), have naturally relied extensively on available viral structural information. As new strategies continue to be developed, it is equally important to continue to refine our understanding and interpretation of viral structural data. There are known limitations to the traditional (Caspar-Klug) theory for describing virus capsid structures that involves subdividing a capsid into triangular subunits. In this context, we describe a more general polyhedral framework for describing virus capsid structures that is able to account for many of these limitations, including a more thorough characterization of intersubunit interfaces. Additionally, our use of pentagonal subunits instead of triangular ones accounts for the intrinsic chirality observed in all capsids. In conjunction with the existing theory, the framework presented here provides a more complete picture of a capsid's structure and therefore can help contribute to the development of more effective antiviral strategies.


Asunto(s)
Cápside/química , Conformación Proteica , Proteínas Estructurales Virales/química , Cápside/ultraestructura , Proteínas de la Cápside/química , Modelos Biológicos , Proteínas Estructurales Virales/ultraestructura , Ensamble de Virus
10.
Nature ; 533(7603): 346-52, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27193680

RESUMEN

Several systems, including contractile tail bacteriophages, the type VI secretion system and R-type pyocins, use a multiprotein tubular apparatus to attach to and penetrate host cell membranes. This macromolecular machine resembles a stretched, coiled spring (or sheath) wound around a rigid tube with a spike-shaped protein at its tip. A baseplate structure, which is arguably the most complex part of this assembly, relays the contraction signal to the sheath. Here we present the atomic structure of the approximately 6-megadalton bacteriophage T4 baseplate in its pre- and post-host attachment states and explain the events that lead to sheath contraction in atomic detail. We establish the identity and function of a minimal set of components that is conserved in all contractile injection systems and show that the triggering mechanism is universally conserved.


Asunto(s)
Bacteriófago T4/química , Bacteriófago T4/ultraestructura , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/ultraestructura , Microscopía por Crioelectrón , Cristalografía por Rayos X , Modelos Moleculares , Conformación Proteica
11.
J Struct Biol ; 192(1): 88-99, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26291392

RESUMEN

Polyhedra represent an ancient system used by a number of insect viruses to protect virions during long periods of environmental exposure. We present high resolution crystal structures of polyhedra for seven previously uncharacterised types of cypoviruses, four using ab initio selenomethionine phasing (two of these required over 100 selenomethionine crystals each). Approximately 80% of residues are structurally equivalent between all polyhedrins (pairwise rmsd ⩽ 1.5 Å), whilst pairwise sequence identities, based on structural alignment, are as little as 12%. These structures illustrate the effect of 400 million years of evolution on a system where the crystal lattice is the functionally conserved feature in the face of massive sequence variability. The conservation of crystal contacts is maintained across most of the molecular surface, except for a dispensable virus recognition domain. By spreading the contacts over so much of the protein surface the lattice remains robust in the face of many individual changes. Overall these unusual structural constraints seem to have skewed the molecule's evolution so that surface residues are almost as conserved as the internal residues.


Asunto(s)
Virus de Insectos/ultraestructura , Proteínas Estructurales Virales/química , Secuencia de Aminoácidos , Sitios de Unión , Secuencia Conservada , Citidina Trifosfato/química , Evolución Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Proteínas Estructurales Virales/ultraestructura
12.
J Virol ; 89(1): 676-87, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25355879

RESUMEN

UNLABELLED: A total of 2,691 mosquitoes representing 17 species was collected from eight locations in southwest Cameroon and screened for pathogenic viruses. Ten isolates of a novel reovirus (genus Dinovernavirus) were detected by culturing mosquito pools on Aedes albopictus (C6/36) cell cultures. A virus that caused overt cytopathic effects was isolated, but it did not infect vertebrate cells or produce detectable disease in infant mice after intracerebral inoculation. The virus, tentatively designated Fako virus (FAKV), represents the first 9-segment, double-stranded RNA (dsRNA) virus to be isolated in nature. FAKV appears to have a broad mosquito host range, and its detection in male specimens suggests mosquito-to-mosquito transmission in nature. The structure of the T=1 FAKV virion, determined to subnanometer resolution by cryoelectron microscopy (cryo-EM), showed only four proteins per icosahedral asymmetric unit: a dimer of the major capsid protein, one turret protein, and one clamp protein. While all other turreted reoviruses of known structures have at least two copies of the clamp protein per asymmetric unit, FAKV's clamp protein bound at only one conformer of the major capsid protein. The FAKV capsid architecture and genome organization represent the most simplified reovirus described to date, and phylogenetic analysis suggests that it arose from a more complex ancestor by serial loss-of-function events. IMPORTANCE: We describe the detection, genetic, phenotypic, and structural characteristics of a novel Dinovernavirus species isolated from mosquitoes collected in Cameroon. The virus, tentatively designated Fako virus (FAKV), is related to both single-shelled and partially double-shelled viruses. The only other described virus in this genus was isolated from cultured mosquito cells. It was previously unclear whether the phenotypic characteristics of that virus were reflective of this genus in nature or were altered during serial passaging in the chronically infected cell line. FAKV is a naturally occurring single-shelled reovirus with a unique virion architecture that lacks several key structural elements thought to stabilize a single-shelled reovirus virion, suggesting what may be the minimal number of proteins needed to form a viable reovirus particle. FAKV evolved from more complex ancestors by losing a genome segment and several virion proteins.


Asunto(s)
Culicidae/virología , Genoma Viral , Reoviridae/genética , Reoviridae/aislamiento & purificación , Animales , Camerún , Línea Celular , Análisis por Conglomerados , Microscopía por Crioelectrón , Efecto Citopatogénico Viral , Evolución Molecular , Especificidad del Huésped , Sustancias Macromoleculares/ultraestructura , Masculino , Ratones , Datos de Secuencia Molecular , Filogenia , ARN Viral/genética , Reoviridae/fisiología , Reoviridae/ultraestructura , Análisis de Secuencia de ADN , Proteínas Estructurales Virales/ultraestructura , Virión/ultraestructura , Cultivo de Virus
13.
PLoS One ; 9(7): e102511, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25019511

RESUMEN

Cytosolic lipid droplets are central organelles in the Hepatitis C Virus (HCV) life cycle. The viral capsid protein core localizes to lipid droplets and initiates the production of viral particles at lipid droplet-associated ER membranes. Core is thought to encapsidate newly synthesized viral RNA and, through interaction with the two envelope proteins E1 and E2, bud into the ER lumen. Here, we visualized the spatial distribution of HCV structural proteins core and E2 in vicinity of small lipid droplets by three-color 3D super-resolution microscopy. We observed and analyzed small areas of colocalization between the two structural proteins in HCV-infected cells with a diameter of approximately 100 nm that might represent putative viral assembly sites.


Asunto(s)
Hepacivirus/metabolismo , Gotas Lipídicas/virología , Proteínas Estructurales Virales/metabolismo , Gotas Lipídicas/ultraestructura , Microscopía Confocal , Microscopía Fluorescente/métodos , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/ultraestructura
14.
J Struct Biol ; 184(2): 193-202, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24036281

RESUMEN

Single-particle electron cryomicroscopy is undergoing a technical revolution due to the recent developments of direct detectors. These new recording devices detect electrons directly (i.e. without conversion into light) and feature significantly improved detective quantum efficiencies and readout rates as compared to photographic films or CCDs. We evaluated here the potential of one such detector (Gatan K2 Summit) to enable the achievement of near-atomic resolution reconstructions of biological specimens when coupled to a widely used, mid-range transmission electron microscope (FEI TF20 Twin). Compensating for beam-induced motion and stage drift provided a 4.4Å resolution map of Sulfolobus turreted icosahedral virus (STIV), which we used as a test particle in this study. Several motion correction and dose fractionation procedures were explored and we describe their influence on the resolution of the final reconstruction. We also compared the quality of this data to that collected with a FEI Titan Krios microscope equipped with a Falcon I direct detector, which provides a benchmark for data collected using a high-end electron microscope.


Asunto(s)
Microscopía por Crioelectrón/instrumentación , Microscopía Electrónica de Transmisión/instrumentación , Rudiviridae/ultraestructura , Modelos Moleculares , Rudiviridae/química , Sulfolobus/virología , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/ultraestructura , Virión/química , Virión/ultraestructura
15.
PLoS Pathog ; 9(5): e1003374, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696739

RESUMEN

The genus Orthobunyavirus within the family Bunyaviridae constitutes an expanding group of emerging viruses, which threaten human and animal health. Despite the medical importance, little is known about orthobunyavirus structure, a prerequisite for understanding virus assembly and entry. Here, using electron cryo-tomography, we report the ultrastructure of Bunyamwera virus, the prototypic member of this genus. Whilst Bunyamwera virions are pleomorphic in shape, they display a locally ordered lattice of glycoprotein spikes. Each spike protrudes 18 nm from the viral membrane and becomes disordered upon introduction to an acidic environment. Using sub-tomogram averaging, we derived a three-dimensional model of the trimeric pre-fusion glycoprotein spike to 3-nm resolution. The glycoprotein spike consists mainly of the putative class-II fusion glycoprotein and exhibits a unique tripod-like arrangement. Protein-protein contacts between neighbouring spikes occur at membrane-proximal regions and intra-spike contacts at membrane-distal regions. This trimeric assembly deviates from previously observed fusion glycoprotein arrangements, suggesting a greater than anticipated repertoire of viral fusion glycoprotein oligomerization. Our study provides evidence of a pH-dependent conformational change that occurs during orthobunyaviral entry into host cells and a blueprint for the structure of this group of emerging pathogens.


Asunto(s)
Virus Bunyamwera/ultraestructura , Glicoproteínas/ultraestructura , Proteínas Estructurales Virales/ultraestructura , Virión/ultraestructura , Animales , Virus Bunyamwera/metabolismo , Línea Celular , Cricetinae , Glicoproteínas/química , Humanos , Estructura Cuaternaria de Proteína , Proteínas Estructurales Virales/metabolismo , Virión/metabolismo
16.
PLoS Pathog ; 9(3): e1003239, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23555252

RESUMEN

Cell-to-cell transmission of vaccinia virus can be mediated by enveloped virions that remain attached to the outer surface of the cell or those released into the medium. During egress, the outer membrane of the double-enveloped virus fuses with the plasma membrane leaving extracellular virus attached to the cell surface via viral envelope proteins. Here we report that F-actin nucleation by the viral protein A36 promotes the disengagement of virus attachment and release of enveloped virus. Cells infected with the A36(YdF) virus, which has mutations at two critical tyrosine residues abrogating localised actin nucleation, displayed a 10-fold reduction in virus release. We examined A36(YdF) infected cells by transmission electron microscopy and observed that during release, virus appeared trapped in small invaginations at the plasma membrane. To further characterise the mechanism by which actin nucleation drives the dissociation of enveloped virus from the cell surface, we examined recombinant viruses by super-resolution microscopy. Fluorescently-tagged A36 was visualised at sub-viral resolution to image cell-virus attachment in mutant and parental backgrounds. We confirmed that A36(YdF) extracellular virus remained closely associated to the plasma membrane in small membrane pits. Virus-induced actin nucleation reduced the extent of association, thereby promoting the untethering of virus from the cell surface. Virus release can be enhanced via a point mutation in the luminal region of B5 (P189S), another virus envelope protein. We found that the B5(P189S) mutation led to reduced contact between extracellular virus and the host membrane during release, even in the absence of virus-induced actin nucleation. Our results posit that during release virus is tightly tethered to the host cell through interactions mediated by viral envelope proteins. Untethering of virus into the surrounding extracellular space requires these interactions be relieved, either through the force of actin nucleation or by mutations in luminal proteins that weaken these interactions.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Virus Vaccinia/fisiología , Vaccinia/transmisión , Proteínas del Envoltorio Viral/metabolismo , Proteínas Estructurales Virales/metabolismo , Liberación del Virus/fisiología , Citoesqueleto de Actina/química , Citoesqueleto de Actina/ultraestructura , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Membrana Celular/ultraestructura , Membrana Celular/virología , Chlorocebus aethiops , Ensayo Cometa , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Fibroblastos/virología , Interacciones Huésped-Patógeno , Ratones , Microscopía Electrónica de Transmisión , Células 3T3 NIH , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Virus Vaccinia/ultraestructura , Células Vero , Proteínas del Envoltorio Viral/ultraestructura , Proteínas Estructurales Virales/ultraestructura
17.
PLoS Pathog ; 9(1): e1003132, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23341770

RESUMEN

Rabbit hemorrhagic disease, first described in China in 1984, causes hemorrhagic necrosis of the liver. Its etiological agent, rabbit hemorrhagic disease virus (RHDV), belongs to the Lagovirus genus in the family Caliciviridae. The detailed molecular structure of any lagovirus capsid has yet to be determined. Here, we report a cryo-electron microscopic (cryoEM) reconstruction of wild-type RHDV at 6.5 Å resolution and the crystal structures of the shell (S) and protruding (P) domains of its major capsid protein, VP60, each at 2.0 Å resolution. From these data we built a complete atomic model of the RHDV capsid. VP60 has a conserved S domain and a specific P2 sub-domain that differs from those found in other caliciviruses. As seen in the shell portion of the RHDV cryoEM map, which was resolved to ~5.5 Å, the N-terminal arm domain of VP60 folds back onto its cognate S domain. Sequence alignments of VP60 from six groups of RHDV isolates revealed seven regions of high variation that could be mapped onto the surface of the P2 sub-domain and suggested three putative pockets might be responsible for binding to histo-blood group antigens. A flexible loop in one of these regions was shown to interact with rabbit tissue cells and contains an important epitope for anti-RHDV antibody production. Our study provides a reliable, pseudo-atomic model of a Lagovirus and suggests a new candidate for an efficient vaccine that can be used to protect rabbits from RHDV infection.


Asunto(s)
Cápside/ultraestructura , Virus de la Enfermedad Hemorrágica del Conejo/ultraestructura , Proteínas Estructurales Virales/ultraestructura , Secuencia de Aminoácidos , Animales , Infecciones por Caliciviridae/veterinaria , Infecciones por Caliciviridae/virología , Proteínas de la Cápside , Microscopía por Crioelectrón , Cristalografía por Rayos X , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Conejos/virología , Alineación de Secuencia , Proteínas Estructurales Virales/química
18.
Protein Eng Des Sel ; 25(11): 789-95, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23081839

RESUMEN

Viral protein 4 (VP4) is a serine protease that catalyzes the hydrolysis of polyprotein pVP2-VP4-VP3 of infectious bursal disease virus. In this report, the recombinant VP4 with a His-tag and three mutants (VP4-S652A, VP4-K692A and VP4-S652A.K692A) were expressed in Escherichia coli. Soluble VP4 was purified using immobilized metal-ion affinity chromatography or sucrose density gradient following with gel-filtration chromatography. The purified VP4 has a tubular structure with 25-30 nm in width and ∼300 nm in length, as observed by transmission electron microscope. A similar tubular structure was also found for these three mutants. The endopeptidase activity of these VP4 tubules was characterized by fluorescence resonance energy transfer using a synthetic fluorogenic oligopeptide as a substrate. The results show that the tubule-like VP4 is a functional enzyme with K(m) of 43 ± 2 µM and k(cat) of 0.04 ± 0.01 min⁻¹; however, k(cat) of three mutants were significantly reduced. This is the first report to demonstrate that VP4 protein expressed in E. coli can self-assemble into functional tubule-like particles and its activity can be completely inhibited by 1 mM of Ni⁺² ions.


Asunto(s)
Endopeptidasas/metabolismo , Virus de la Enfermedad Infecciosa de la Bolsa/enzimología , Proteínas Estructurales Virales/metabolismo , Infecciones por Birnaviridae/virología , Cromatografía de Afinidad , Clonación Molecular , Endopeptidasas/genética , Endopeptidasas/aislamiento & purificación , Endopeptidasas/ultraestructura , Escherichia coli/genética , Virus de la Enfermedad Infecciosa de la Bolsa/genética , Cinética , Níquel/metabolismo , Mutación Puntual , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/aislamiento & purificación , Proteínas Estructurales Virales/ultraestructura
19.
J Virol ; 86(11): 6123-37, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22457528

RESUMEN

Herpes simplex virus 1 (HSV-1) enters neurons primarily by fusion of the viral envelope with the host cell plasma membrane, leading to the release of the capsid into the cytosol. The capsid travels via microtubule-mediated retrograde transport to the nuclear membrane, where the viral DNA is released for replication in the nucleus. In the present study, the composition and kinetics of incoming HSV-1 capsids during entry and retrograde transport in axons of human fetal and dissociated rat dorsal root ganglia (DRG) neurons were examined by wide-field deconvolution microscopy and transmission immunoelectron microscopy (TIEM). We show that HSV-1 tegument proteins, including VP16, VP22, most pUL37, and some pUL36, dissociated from the incoming virions. The inner tegument proteins, including pUL36 and some pUL37, remained associated with the capsid during virus entry and transit to the nucleus in the neuronal cell body. By TIEM, a progressive loss of tegument proteins, including VP16, VP22, most pUL37, and some pUL36, was observed, with most of the tegument dissociating at the plasma membrane of the axons and the neuronal cell body. Further dissociation occurred within the axons and the cytosol as the capsids moved to the nucleus, resulting in the release of free tegument proteins, especially VP16, VP22, pUL37, and some pUL36, into the cytosol. This study elucidates ultrastructurally the composition of HSV-1 capsids that encounter the microtubules in the core of human axons and the complement of free tegument proteins released into the cytosol during virus entry.


Asunto(s)
Ganglios Espinales/virología , Herpesvirus Humano 1/fisiología , Neuronas/virología , Proteínas Estructurales Virales/metabolismo , Internalización del Virus , Animales , Células Cultivadas , Herpesvirus Humano 1/ultraestructura , Humanos , Microscopía , Ratas , Ratas Wistar , Proteínas Estructurales Virales/ultraestructura , Virión/ultraestructura
20.
Vet Immunol Immunopathol ; 145(1-2): 522-6, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22209204

RESUMEN

Goose parvovirus (GPV), a small non-enveloped ssDNA virus, can cause Derzsy's disease, a highly contagious and lethal disease in goslings and muscovy ducklings, leading to a huge economic loss. However, little is known about the localization of B-cell epitopes on GPV structural protein. To address the issue, the structural protein of GPV was dissected into sets of partially overlapping fragments and expressed in Escherichia coli. Then Western blot reactivity of these glutathione S-transferase (GST) fusion short peptides to viral infected sera was surveyed. The results showed linear immunodominant epitopes, which were found in seven fragments covering amino acid residues 35-71, 123-198, 423-444, 474-491, 531-566, 616-669, 678-732. Our findings may provide the basis for the development of immunity-based prophylactic, therapeutic, and diagnostic clinical techniques for Derzsy's disease.


Asunto(s)
Epítopos de Linfocito B/inmunología , Gansos/virología , Infecciones por Parvoviridae/veterinaria , Parvovirus/inmunología , Enfermedades de las Aves de Corral/virología , Proteínas Estructurales Virales/inmunología , Secuencia de Aminoácidos , Animales , Western Blotting/veterinaria , Mapeo Epitopo/veterinaria , Gansos/inmunología , Datos de Secuencia Molecular , Infecciones por Parvoviridae/inmunología , Infecciones por Parvoviridae/virología , Parvovirus/ultraestructura , Enfermedades de las Aves de Corral/inmunología , Alineación de Secuencia/veterinaria , Proteínas Estructurales Virales/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...