Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 187(9): 2236-2249.e17, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38614100

RESUMEN

Unlike those of double-stranded DNA (dsDNA), single-stranded DNA (ssDNA), and ssRNA viruses, the mechanism of genome packaging of dsRNA viruses is poorly understood. Here, we combined the techniques of high-resolution cryoelectron microscopy (cryo-EM), cellular cryoelectron tomography (cryo-ET), and structure-guided mutagenesis to investigate genome packaging and capsid assembly of bluetongue virus (BTV), a member of the Reoviridae family of dsRNA viruses. A total of eleven assembly states of BTV capsid were captured, with resolutions up to 2.8 Å, with most visualized in the host cytoplasm. ATPase VP6 was found underneath the vertices of capsid shell protein VP3 as an RNA-harboring pentamer, facilitating RNA packaging. RNA packaging expands the VP3 shell, which then engages middle- and outer-layer proteins to generate infectious virions. These revealed "duality" characteristics of the BTV assembly mechanism reconcile previous contradictory co-assembly and core-filling models and provide insights into the mysterious RNA packaging and capsid assembly of Reoviridae members and beyond.


Asunto(s)
Virus de la Lengua Azul , Proteínas de la Cápside , Cápside , Microscopía por Crioelectrón , ARN Viral , Empaquetamiento del Genoma Viral , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/fisiología , Virus de la Lengua Azul/metabolismo , Cápside/metabolismo , Cápside/ultraestructura , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/química , Animales , ARN Viral/metabolismo , ARN Viral/genética , Genoma Viral/genética , Ensamble de Virus , Tomografía con Microscopio Electrónico , Virión/metabolismo , Virión/genética , Virión/ultraestructura , Modelos Moleculares , Línea Celular , Cricetinae
2.
Biomolecules ; 13(6)2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37371457

RESUMEN

At least 12 serotypes of 'atypical' bluetongue virus (BTV-25 to BTV-36) have been identified to date. These atypical serotypes fail to infect/replicate in Culicoides-derived cell lines and/or adult Culicoides vectors and hence can no longer be transmitted by these vectors. They appear to be horizontally transmitted from infected to in-contact ruminants, although the route(s) of infection remain to be identified. Viral genome segments 1, 2 and 3 (Seg-1, Seg2 and Seg-3) of BTV-26 were identified as involved in blocking virus replication in KC cells. We have developed Culicoides-specific expression plasmids, which we used in transfected insect cells to assess the stability of viral mRNAs and protein expression from full-length open reading frames of Seg-1, -2 and -3 of BTV-1 (a Culicoides-vectored BTV) or BTV-26. Our results indicate that the blocked replication of BTV-26 in KC cells is not due to an RNAi response, which would lead to rapid degradation of viral mRNAs. A combination of degradation/poor expression and/or modification of the proteins encoded by these segments appears to drive the failure of BTV-26 core/whole virus-particles to assemble and replicate effectively in Culicoides cells.


Asunto(s)
Virus de la Lengua Azul , Ceratopogonidae , Animales , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/metabolismo , Ceratopogonidae/genética , Serogrupo , Genoma Viral , Línea Celular , Replicación Viral/genética
3.
Nucleic Acids Res ; 51(10): 5210-5227, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37070191

RESUMEN

How multi-segmented double-stranded RNA (dsRNA) viruses correctly incorporate their genomes into their capsids remains unclear for many viruses, including Bluetongue virus (BTV), a Reoviridae member, with a genome of 10 segments. To address this, we used an RNA-cross-linking and peptide-fingerprinting assay (RCAP) to identify RNA binding sites of the inner capsid protein VP3, the viral polymerase VP1 and the capping enzyme VP4. Using a combination of mutagenesis, reverse genetics, recombinant proteins and in vitro assembly, we validated the importance of these regions in virus infectivity. Further, to identify which RNA segments and sequences interact with these proteins, we used viral photo-activatable ribonucleoside crosslinking (vPAR-CL) which revealed that the larger RNA segments (S1-S4) and the smallest segment (S10) have more interactions with viral proteins than the other smaller segments. Additionally, using a sequence enrichment analysis we identified an RNA motif of nine bases that is shared by the larger segments. The importance of this motif for virus replication was confirmed by mutagenesis followed by virus recovery. We further demonstrated that these approaches could be applied to a related Reoviridae member, rotavirus (RV), which has human epidemic impact, offering the possibility of novel intervention strategies for a human pathogen.


Asunto(s)
Virus de la Lengua Azul , Cápside , ARN Viral , Proteínas Virales , Animales , Humanos , Virus de la Lengua Azul/química , Virus de la Lengua Azul/metabolismo , Cápside/química , Cápside/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Replicación Viral , Proteínas Virales/química , Proteínas Virales/metabolismo
4.
Virol Sin ; 37(4): 521-530, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35513266

RESUMEN

ISG20 is an interferon-inducible exonuclease that inhibits virus replication. Although ISG20 is thought to degrade viral RNA, the antiviral mechanism and specificity of ISG20 remain unclear. In this study, the antiviral role of ovine ISG20 (oISG20) in bluetongue virus â€‹(BTV) infection was investigated. It was found that BTV infection up-regulated the transcription of ovine ISG20 (oISG20) in a time- and BTV multiplicity of infection (MOI)-dependent manner. Overexpression of oISG20 suppressed the production of BTV genome, proteins, and virus titer, whereas the knockdown of oISG20 increased viral replication. oISG20 was found to co-localize with BTV proteins VP4, VP5, VP6, and NS2, but only directly interacted with VP4. Exonuclease defective oISG20 significantly decreased the inhibitory effect on BTV replication. In addition, the interaction of mutant oISG20 and VP4 was weakened, suggesting that binding to VP4 was associated with the inhibition of BTV replication. The present data characterized the anti-BTV effect of oISG20, and provides a novel clue for further exploring the inhibition mechanism of double-stranded RNA virus by ISG20.


Asunto(s)
Virus de la Lengua Azul , Lengua Azul , Animales , Antivirales/farmacología , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/metabolismo , Exonucleasas/genética , Exonucleasas/metabolismo , Exonucleasas/farmacología , Ovinos , Replicación Viral
5.
Viruses ; 14(2)2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35215776

RESUMEN

Bluetongue virus (BTV) is the etiologic agent of a non-contagious arthropod-borne disease transmitted to wild and domestic ruminants. BTV induces a large panel of clinical manifestations ranging from asymptomatic infection to lethal hemorrhagic fever. Despite the fact that BTV has been studied extensively, we still have little understanding of the molecular determinants of BTV virulence. In our report, we have performed a comparative yeast two-hybrid (Y2H) screening approach to search direct cellular targets of the NS4 virulence factor encoded by two different serotypes of BTV: BTV8 and BTV27. This led to identifying Wilms' tumor 1-associated protein (WTAP) as a new interactor of the BTV-NS4. In contrast to BTV8, 1, 4 and 25, NS4 proteins from BTV27 and BTV30 are unable to interact with WTAP. This interaction with WTAP is carried by a peptide of 34 amino acids (NS422-55) within its putative coil-coiled structure. Most importantly, we showed that binding to WTAP is restored with a chimeric protein where BTV27-NS4 is substituted by BTV8-NS4 in the region encompassing residue 22 to 55. We also demonstrated that WTAP silencing reduces viral titers and the expression of viral proteins, suggesting that BTV-NS4 targets a cellular function of WTAP to increase its viral replication.


Asunto(s)
Virus de la Lengua Azul/metabolismo , Lengua Azul/metabolismo , Lengua Azul/virología , Enfermedades de los Bovinos/metabolismo , Factores de Empalme de ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Lengua Azul/genética , Virus de la Lengua Azul/química , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/patogenicidad , Bovinos , Enfermedades de los Bovinos/genética , Enfermedades de los Bovinos/virología , Interacciones Huésped-Patógeno , Unión Proteica , Factores de Empalme de ARN/genética , Alineación de Secuencia , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Factores de Virulencia/genética , Replicación Viral
6.
Nat Microbiol ; 6(11): 1424-1432, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34702979

RESUMEN

Bluetongue virus (BTV) is a non-enveloped virus and causes substantial morbidity and mortality in ruminants such as sheep. Fashioning a receptor-binding protein (VP2) and a membrane penetration protein (VP5) on the surface, BTV releases its genome-containing core (VP3 and VP7) into the host cell cytosol after perforation of the endosomal membrane. Unlike enveloped ones, the entry mechanisms of non-enveloped viruses into host cells remain poorly understood. Here we applied single-particle cryo-electron microscopy, cryo-electron tomography and structure-guided functional assays to characterize intermediate states of BTV cell entry in endosomes. Four structures of BTV at the resolution range of 3.4-3.9 Å show the different stages of structural rearrangement of capsid proteins on exposure to low pH, including conformational changes of VP5, stepwise detachment of VP2 and a small shift of VP7. In detail, sensing of the low-pH condition by the VP5 anchor domain triggers three major VP5 actions: projecting the hidden dagger domain, converting a surface loop to a protonated ß-hairpin that anchors VP5 to the core and stepwise refolding of the unfurling domains into a six-helix stalk. Cryo-electron tomography structures of BTV interacting with liposomes show a length decrease of the VP5 stalk from 19.5 to 15.5 nm after its insertion into the membrane. Our structures, functional assays and structure-guided mutagenesis experiments combined indicate that this stalk, along with dagger domain and the WHXL motif, creates a single pore through the endosomal membrane that enables the viral core to enter the cytosol. Our study unveils the detailed mechanisms of BTV membrane penetration and showcases general methods to study cell entry of other non-enveloped viruses.


Asunto(s)
Virus de la Lengua Azul/metabolismo , Lengua Azul/virología , Proteínas de la Cápside/metabolismo , Endosomas/virología , Animales , Virus de la Lengua Azul/química , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/ultraestructura , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Microscopía por Crioelectrón , Endosomas/química , Concentración de Iones de Hidrógeno , Modelos Moleculares , Ovinos , Enfermedades de las Ovejas/virología , Internalización del Virus
7.
Viruses ; 13(2)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33540654

RESUMEN

Bluetongue virus (BTV), an arbovirus transmitted by Culicoides biting midges, is a major concern of wild and domestic ruminants. While BTV induces type I interferon (alpha/beta interferon [IFN-α/ß]) production in infected cells, several reports have described evasion strategies elaborated by this virus to dampen this intrinsic, innate response. In the present study, we suggest that BTV VP3 is a new viral antagonist of the IFN-ß synthesis. Indeed, using split luciferase and coprecipitation assays, we report an interaction between VP3 and both the mitochondrial adapter protein MAVS and the IRF3-kinase IKKε. Overall, this study describes a putative role for the BTV structural protein VP3 in the control of the antiviral response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Virus de la Lengua Azul/metabolismo , Lengua Azul/metabolismo , Proteína 58 DEAD Box/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas del Núcleo Viral/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Lengua Azul/genética , Lengua Azul/virología , Virus de la Lengua Azul/genética , Proteína 58 DEAD Box/genética , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Unión Proteica , Receptores Inmunológicos/genética , Transducción de Señal , Proteínas del Núcleo Viral/genética
8.
Med Sci Monit ; 27: e930634, 2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33507885

RESUMEN

BACKGROUND The bluetongue virus (BTV) is the prototype virus in the genus Orbivirus within the family Reoviridae. Recent studies indicate that BTVs are capable of infecting and selectively lysing human hepatic carcinoma cells (Hep-3B) and prostate carcinoma cells (pc-3). This study was designed to evaluate the oncolytic potential of BTV in experimental models of human renal cancer in vitro and in vivo. MATERIAL AND METHODS Five human renal cancer cell lines, ACHN, CAKI-1, OS-RC-2, 786-O, and A498, were used in this study to analyze BTV replication. These cells were lysed by oncolysis compared to normal control. Xenograft models were used to assess the efficacy and toxicity of BTVs in vivo. Data were analyzed by one-way ANOVA or two-sided unpaired t tests. RESULTS The results showed HPTEC cells to be relatively resistant to cytotoxic effects of BTVs and exhibited normal growth rate even at high dose of BTVs. Nonetheless, the renal cancer cells showed a remarkably higher sensitivity to BTVs. Moreover, the ultramicroscopic subcellular changes were also detected in the renal cells. The viral particles were observed in all the RCC cell lines, but not in HPTEC cells. Intratumoral injections of BTVs significantly decreased the tumor volume as compared to animals that received no virus treatment. Infection with BTVs significantly increased the percentage of apoptotic renal cancer cells but not the HPTEC cells. Moreover, BTV triggered apoptosis in renal cancer cells via a mitochondria-mediated pathway. CONCLUSIONS This study for the first time demonstrated the oncolytic potential of BTV in experimental models of human renal cancer. BTV exhibits the potential to inhibit human renal cancer cell growth in vitro and in vivo.


Asunto(s)
Virus de la Lengua Azul/metabolismo , Neoplasias Renales/terapia , Neoplasias Renales/virología , Animales , Apoptosis/fisiología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/terapia , Carcinoma de Células Renales/virología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Humanos , Neoplasias Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Vet Microbiol ; 254: 108986, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33486325

RESUMEN

Previous studies have pointed out that bluetongue virus (BTV) down-regulates the expression levels of type Ⅰ interferon (IFN-Ⅰ) and inhibits IFN-Ⅰ signaling by targeting on the Janus tyrosine kinase (JAK)-signal transducer and activator of transcription protein (STAT) pathway. However, individual viral protein could not effectively block IFN-Ⅰ signaling. There is a need to explore the underlying mechanisms by which viral proteins of BTV coordinate to antagonize the IFN-Ⅰ signaling. We investigated the coordinative role of BTV-1 nonstructural protein 3 (NS3) and NS4 in counteracting IFN-Ⅰ signaling in the JAK-STAT pathway by directly interacting with STAT1. The NS3 and NS4 targeted the SH2 domain of STAT1 to inhibit its phosphorylation, heterodimerization, nuclear translocation, as well as activation of downstream genes of the JAK-STAT pathway. NS3 and NS4 impaired STAT1 phosphorylation induced by IFN-Ⅰ in a dose dependent manner. Overall, this study confirmed that NS3 and NS4 of BTV participate in interfering with IFN-Ⅰ signaling process. Also, a new mechanism employed by BTV to evade host innate immune responses was revealed.


Asunto(s)
Virus de la Lengua Azul/genética , Virus de la Lengua Azul/metabolismo , Interacciones Huésped-Patógeno , Interferón Tipo I/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Proteínas no Estructurales Virales/metabolismo , Línea Celular , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Fosforilación , Factor de Transcripción STAT1/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
10.
PLoS Pathog ; 16(10): e1009015, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33075107

RESUMEN

Recent developments on extracellular vesicles (EVs) containing multiple virus particles challenge the rigid definition of non-enveloped viruses. However, how non-enveloped viruses hijack cell machinery to promote non-lytic release in EVs, and their functional roles, remain to be clarified. Here we used Bluetongue virus (BTV) as a model of a non-enveloped arthropod-borne virus and discovered that the majority of viruses are released in EVs. Based on the cellular proteins detected in these EVs, and use of inhibitors targeting the cellular degradation process, we demonstrated that these extracellular vesicles are derived from secretory lysosomes, in which the acidic pH is neutralized upon the infection. Moreover, we report that secreted EVs are more efficient than free-viruses for initiating infections, but that they trigger super-infection exclusion that only free-viruses can overcome.


Asunto(s)
Virus de la Lengua Azul/metabolismo , Vesículas Extracelulares/metabolismo , Lisosomas/metabolismo , Animales , Arbovirus/patogenicidad , Virus de la Lengua Azul/genética , Línea Celular , Vesículas Extracelulares/fisiología , Modelos Biológicos , Vesículas Secretoras , Virión , Replicación Viral
11.
Curr Top Med Chem ; 20(19): 1720-1732, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32416694

RESUMEN

BACKGROUND: The capsid coated protein of Bluetongue virus (BTV) VP2 is responsible for BTV transmission by the Culicoides vector to vertebrate hosts. Besides, VP2 is responsible for BTV entry into permissive cells and hence plays a major role in disease progression. However, its mechanism of action is still unknown. OBJECTIVE: The present investigation aimed to predict the 3D structure of Viral Protein 2 of the bluetongue virus assisted by Optimized Potential for Liquid Simulations (OPLS), structure validation, and an active site prediction. METHODS: The 3D structure of the VP2 protein was built using a Python-based Computational algorithm. The templates were identified using Smith waterman's Local alignment. The VP2 protein structure validated using PROCHECK. Molecular Dynamics Simulation (MDS) studies were performed using an academic software Desmond, Schrodinger dynamics, for determining the stability of a model protein. The Ligand-Binding site was predicted by structure comparison using homology search and proteinprotein network analysis to reveal their stability and inhibition mechanism, followed by the active site identification. RESULTS: The secondary structure of the VP2 reveals that the protein contains 220 alpha helix atoms, 40 310 helix, 151 beta sheets, 134 coils and 424 turns, whereas the 3D structure of Viral Protein 2 of BTV has been found to have 15774 total atoms in the structure. However, 961 amino acids were found in the final model. The dynamical cross-correlation matrix (DCCM) analysis tool identifies putative protein domains and also confirms the stability of the predicted model and their dynamical behavior difference with the correlative fluctuations in motion. CONCLUSION: The biological interpretation of the Viral Protein 2 was carried out. DCCM maps were calculated, using a different coordinate reference frame, through which, protein domain boundaries and protein domain residue constituents were identified. The obtained model shows good reliability. Moreover, we anticipated that this research should play a promising role in the identification of novel candidates with the target protein to inhibit their functional significance.


Asunto(s)
Virus de la Lengua Azul/química , Proteínas de la Cápside/química , Diseño Asistido por Computadora , Simulación de Dinámica Molecular , Virus de la Lengua Azul/metabolismo , Proteínas de la Cápside/metabolismo , Ligandos , Filogenia
12.
Soft Matter ; 15(38): 7663-7671, 2019 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-31490506

RESUMEN

Clear understanding of the principles that control the arrangement of proteins and their self-assembly into viral shells is very important for the development of antiviral strategies. Here we consider the structural peculiarities and hidden symmetry of the anomalous bluetongue virus (BTV) capsid. Each of its three concentric shells violates the paradigmatic geometrical model of Caspar and Klug, which is otherwise well suited to describe most of the known icosahedral viral shells. As we show, three icosahedral spherical lattices, which are commensurate with each other and possess locally hexagonal (primitive or honeycomb) order, underlie the proteinaceous shells of the BTV capsid. This interpretation of the multishelled envelope allows us to discuss the so-called "symmetry mismatch" between its layers. We also analyze the structural stability of the considered spherical lattices on the basis of the classical theory of spherical packing and relate the proximity of the outer spherical lattice to destabilization with the fact that during infection of the cell VP2 trimers are detached from the surface of the BTV capsid. An electrostatic mechanism that can assist in this detachment is discussed in detail.


Asunto(s)
Virus de la Lengua Azul/química , Proteínas de la Cápside/química , Cápside/química , Modelos Biológicos , Virosis/metabolismo , Virus de la Lengua Azul/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Concentración de Iones de Hidrógeno , Conformación Proteica , Multimerización de Proteína , Electricidad Estática , Termodinámica
13.
Nat Microbiol ; 4(5): 837-845, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30778144

RESUMEN

Bluetongue virus (BTV) non-structural protein 1 (NS1) regulates viral protein synthesis and exists as tubular and non-tubular forms in infected cells, but how tubules assemble and how protein synthesis is regulated are unknown. Here, we report near-atomic resolution structures of two NS1 tubular forms determined by cryo-electron microscopy. The two tubular forms are different helical assemblies of the same NS1 monomer, consisting of an amino-terminal foot, a head and body domains connected to an extended carboxy-terminal arm, which wraps atop the head domain of another NS1 subunit through hydrophobic interactions. Deletion of the C terminus prevents tubule formation but not viral replication, suggesting an active non-tubular form. Two zinc-finger-like motifs are present in each NS1 monomer, and tubules are disrupted by divalent cation chelation and restored by cation addition, including Zn2+, suggesting a regulatory role of divalent cations in tubule formation. In vitro luciferase assays show that the NS1 non-tubular form upregulates BTV mRNA translation, whereas zinc-finger disruption decreases viral mRNA translation, tubule formation and virus replication, confirming a functional role for the zinc-fingers. Thus, the non-tubular form of NS1 is sufficient for viral protein synthesis and infectious virus replication, and the regulatory mechanism involved operates through divalent cation-dependent conversion between the non-tubular and tubular forms.


Asunto(s)
Virus de la Lengua Azul/metabolismo , Lengua Azul/virología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Animales , Virus de la Lengua Azul/química , Virus de la Lengua Azul/genética , Línea Celular , Cricetinae , Biosíntesis de Proteínas , Dominios Proteicos , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Zinc/química , Zinc/metabolismo , Dedos de Zinc
14.
J Virol ; 93(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30518645

RESUMEN

Bluetongue virus (BTV), in the family Reoviridae, is an insect-borne, double-capsid virus causing hemorrhagic disease in livestock around the world. Here, we elucidate how outer capsid proteins VP2 and VP5 coordinate cell entry of BTV. To identify key functional residues, we used atomic-level structural data to guide mutagenesis of VP2 and VP5 and a series of biological and biochemical approaches, including site-directed mutagenesis, reverse genetics-based virus recovery, expression and characterization of individual recombinant mutant proteins, and various in vitro and in vivo assays. We demonstrate the dynamic nature of the conformational change process, revealing that a unique zinc finger (CCCH) in VP2 acts as the major low pH sensor, coordinating VP2 detachment, subsequently allowing VP5 to sense low pH via specific histidine residues at key positions. We show that single substitution of only certain histidine residues has a lethal effect, indicating that the location of histidine in VP5 is critical to inducing changes in VP5 conformation that facilitates membrane penetration. Further, we show that the VP5 anchoring domain alone recapitulates sensing of low pH. Our data reveal a novel, multiconformational process that overcomes entry barriers faced by this multicapsid nonenveloped virus.IMPORTANCE Virus entry into a susceptible cell is the first step of infection and a significant point at which infection can be prevented. To enter effectively, viruses must sense the cellular environment and, when appropriate, initiate a series of changes that eventually jettison the protective shell and deposit virus genes into the cytoplasm. Many viruses sense pH, but how this happens and the events that follow are often poorly understood. Here, we address this question for a large multilayered bluetongue virus. We show key residues in outer capsid proteins, a pH-sensing histidine of a zinc finger within the receptor-binding VP2 protein, and certain histidine residues in the membrane-penetrating VP5 protein that detect cellular pH, leading to irreversible changes and propel the virus through the cell membrane. Our data reveal a novel mechanism of cell entry for a nonenveloped virus and highlight mechanisms which may also be used by other viruses.


Asunto(s)
Virus de la Lengua Azul/genética , Virus de la Lengua Azul/metabolismo , Interacciones Microbiota-Huesped/fisiología , Virus de la Lengua Azul/patogenicidad , Proteínas de la Cápside/genética , Línea Celular , Membrana Celular/metabolismo , Virus ADN/genética , Concentración de Iones de Hidrógeno , Unión Proteica/fisiología , Reoviridae/genética , Virión/genética , Internalización del Virus
15.
Protein Expr Purif ; 147: 85-93, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29551716

RESUMEN

Bluetongue virus (BTV) is an Orbivirus that has a profound economic impact due to direct loss of livestock as well as movement bans in an attempt to prevent the spread of the disease to susceptible areas. BTV VP7, along with VP3, forms the inner capsid core of the virus where it acts as the barrier between the outer layer and the inner core housing the genetic material. Purification of BTV VP7 has proven to be problematic and expensive mainly due to its insolubility is several expression systems. To overcome this, in this paper we present a protocol for the solubilisation of BTV VP7 from inclusion bodies expressed in E.coli, and subsequent purification using nickel affinity chromatography. The purified protein was then characterised using native PAGE, far ultraviolet circular dichroism (far-UV CD) and intrinsic fluorescence and found to have both secondary and tertiary structure even in the presence of 5 M urea. Both tertiary and secondary structure was further shown to be to be maintained at least to 42 °C in 5 M urea.


Asunto(s)
Virus de la Lengua Azul/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas del Núcleo Viral/metabolismo , Virus de la Lengua Azul/genética , Dicroismo Circular , Escherichia coli/genética , Escherichia coli/metabolismo , Desplegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Solubilidad , Espectrometría de Fluorescencia , Temperatura , Proteínas del Núcleo Viral/química , Proteínas del Núcleo Viral/genética
16.
Virus Res ; 232: 80-95, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28167267

RESUMEN

VP7 is the major core protein of orbiviruses and is essential for virion assembly. African horse sickness virus (AHSV) VP7 self-assembles into highly insoluble crystalline particles - an attribute that may be related to the role of AHSV VP7 in virus assembly but also prevents crystallization. Given that this inherent insolubility is unique to AHSV VP7, we use amino acid sequence conservation analysis between AHSV VP7 and other orbiviruses to identify putative key residues that drive AHSV VP7 self-assembly. A homology model of the AHSV VP7 trimer was generated to analyze surface properties of the trimer and to identify surface residues as candidates for the AHSV VP7 trimer-trimer interactions that drive AHSV VP7 self-assembly. Nine regions were identified as candidate residues for future site-directed mutagenesis experiments that will likely result in a soluble AHSV VP7 protein. Additionally, we identified putative residues that function in the intermolecular interactions within the AHSV VP7 trimer as well as several epitopes. Given the many previous efforts of solubilizing AHSV VP7, we propose a useful strategy that will yield a soluble AHSV VP7 that can be used to study AHSV assembly and increase yield of recombinant vaccine preparations.


Asunto(s)
Virus de la Enfermedad Equina Africana/química , Anticuerpos Monoclonales/química , Antígenos Virales/química , Virus de la Lengua Azul/química , Proteínas del Núcleo Viral/química , Virus de la Enfermedad Equina Africana/inmunología , Virus de la Enfermedad Equina Africana/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales/metabolismo , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Sitios de Unión , Virus de la Lengua Azul/inmunología , Virus de la Lengua Azul/metabolismo , Mapeo Epitopo , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Alineación de Secuencia , Homología Estructural de Proteína , Proteínas del Núcleo Viral/inmunología , Proteínas del Núcleo Viral/metabolismo , Ensamble de Virus
17.
Virol J ; 13: 119, 2016 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-27368544

RESUMEN

BACKGROUND: Bluetongue virus (BTV) and African horse sickness virus (AHSV) are distinct arthropod borne virus species in the genus Orbivirus (Reoviridae family), causing the notifiable diseases Bluetongue and African horse sickness of ruminants and equids, respectively. Reverse genetics systems for these orbiviruses with their ten-segmented genome of double stranded RNA have been developed. Initially, two subsequent transfections of in vitro synthesized capped run-off RNA transcripts resulted in the recovery of BTV. Reverse genetics has been improved by transfection of expression plasmids followed by transfection of ten RNA transcripts. Recovery of AHSV was further improved by use of expression plasmids containing optimized open reading frames. RESULTS: Plasmids containing full length cDNA of the 10 genome segments for T7 promoter-driven production of full length run-off RNA transcripts and expression plasmids with optimized open reading frames (ORFs) were used. BTV and AHSV were rescued using reverse genetics. The requirement of each expression plasmid and capping of RNA transcripts for reverse genetics were studied and compared for BTV and AHSV. BTV was recovered by transfection of VP1 and NS2 expression plasmids followed by transfection of a set of ten capped RNAs. VP3 expression plasmid was also required if uncapped RNAs were transfected. Recovery of AHSV required transfection of VP1, VP3 and NS2 expression plasmids followed by transfection of capped RNA transcripts. Plasmid-driven expression of VP4, 6 and 7 was also needed when uncapped RNA transcripts were used. Irrespective of capping of RNA transcripts, NS1 expression plasmid was not needed for recovery, although NS1 protein is essential for virus propagation. Improvement of reverse genetics for AHSV was clearly demonstrated by rescue of several mutants and reassortants that were not rescued with previous methods. CONCLUSIONS: A limited number of expression plasmids is required for rescue of BTV or AHSV using reverse genetics, making the system much more versatile and generally applicable. Optimization of reverse genetics enlarge the possibilities to rescue virus mutants and reassortants, and will greatly benefit the control of these important diseases of livestock and companion animals.


Asunto(s)
Virus de la Enfermedad Equina Africana/genética , Enfermedad Equina Africana/virología , Virus de la Lengua Azul/genética , Lengua Azul/virología , Genética Inversa/métodos , Virus de la Enfermedad Equina Africana/metabolismo , Animales , Virus de la Lengua Azul/metabolismo , Genoma Viral , Caballos , Plásmidos/genética , Plásmidos/metabolismo , ARN Viral/genética , Rumiantes/virología
18.
J Biol Chem ; 291(23): 12408-19, 2016 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-27036941

RESUMEN

The entry of viruses into host cells is one of the key processes of infection. The mechanisms of cellular entry for enveloped virus have been well studied. The fusion proteins as well as the facilitating cellular lipid factors involved in the viral fusion entry process have been well characterized. The process of non-enveloped virus cell entry, in comparison, remains poorly defined, particularly for large complex capsid viruses of the family Reoviridae, which comprises a range of mammalian pathogens. These viruses enter cells without the aid of a limiting membrane and thus cannot fuse with host cell membranes to enter cells. Instead, these viruses are believed to penetrate membranes of the host cell during endocytosis. However, the molecular mechanism of this process is largely undefined. Here we show, utilizing an in vitro liposome penetration assay and cell biology, that bluetongue virus (BTV), an archetypal member of the Reoviridae, utilizes the late endosome-specific lipid lysobisphosphatidic acid for productive membrane penetration and viral entry. Further, we provide preliminary evidence that lipid lysobisphosphatidic acid facilitates pore expansion during membrane penetration, suggesting a mechanism for lipid factor requirement of BTV. This finding indicates that despite the lack of a membrane envelope, the entry process of BTV is similar in specific lipid requirements to enveloped viruses that enter cells through the late endosome. These results are the first, to our knowledge, to demonstrate that a large non-enveloped virus of the Reoviridae has specific lipid requirements for membrane penetration and host cell entry.


Asunto(s)
Virus de la Lengua Azul/metabolismo , Cápside/metabolismo , Endosomas/metabolismo , Lisofosfolípidos/metabolismo , Monoglicéridos/metabolismo , Internalización del Virus , Animales , Western Blotting , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/fisiología , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Endocitosis , Endosomas/virología , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Liposomas/metabolismo , Microscopía Confocal , Mutación , Células Sf9 , Spodoptera
19.
J Gen Virol ; 96(11): 3280-3293, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26290332

RESUMEN

Viruses have often evolved overlapping reading frames in order to maximize their coding capacity. Until recently, the segmented dsRNA genome of viruses of the Orbivirus genus was thought to be monocistronic, but the identification of the bluetongue virus (BTV) NS4 protein changed this assumption. A small ORF in segment 10, overlapping the NS3 ORF in the +1 position, is maintained in more than 300 strains of the 27 different BTV serotypes and in more than 200 strains of the phylogenetically related African horse sickness virus (AHSV). In BTV, this ORF (named S10-ORF2 in this study) encodes a putative protein 50-59 residues in length and appears to be under strong positive selection. HA- or GFP-tagged versions of S10-ORF2 expressed from transfected plasmids localized within the nucleoli of transfected cells, unless a putative nucleolar localization signal was mutated. S10-ORF2 inhibited gene expression, but not RNA translation, in transient transfection reporter assays. In both mammalian and insect cells, BTV S10-ORF2 deletion mutants (BTV8ΔS10-ORF2) displayed similar replication kinetics to wt virus. In vivo, S10-ORF2 deletion mutants were pathogenic in mouse models of disease. Although further evidence is required for S10-ORF2 expression during infection, the data presented provide an initial characterization of this ORF.


Asunto(s)
Virus de la Lengua Azul/genética , Lengua Azul/virología , Genoma Viral , Sistemas de Lectura Abierta , Proteínas Virales/genética , Animales , Virus de la Lengua Azul/clasificación , Virus de la Lengua Azul/metabolismo , Línea Celular , Ratones , Filogenia , Proteínas Virales/metabolismo
20.
J Virol ; 89(20): 10467-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246581

RESUMEN

UNLABELLED: Bluetongue virus (BTV) is an arbovirus transmitted to livestock by midges of the Culicoides family and is the etiological agent of a hemorrhagic disease in sheep and other ruminants. In mammalian cells, BTV particles are released primarily by virus-induced cell lysis, while in insect cells they bud from the plasma membrane and establish a persistent infection. BTV possesses a ten-segmented double-stranded RNA genome, and NS3 proteins are encoded by segment 10 (Seg-10). The viral nonstructural protein 3 (NS3) plays a key role in mediating BTV egress as well as in impeding the in vitro synthesis of type I interferon in mammalian cells. In this study, we asked whether genetically distant NS3 proteins can alter BTV-host interactions. Using a reverse genetics approach, we showed that, depending on the NS3 considered, BTV replication kinetics varied in mammals but not in insects. In particular, one of the NS3 proteins analyzed harbored a proline at position 24 that leads to its rapid intracellular decay in ovine but not in Culicoides cells and to the attenuation of BTV virulence in a mouse model of disease. Overall, our data reveal that the genetic variability of Seg-10/NS3 differentially modulates BTV replication kinetics in a host-specific manner and highlight the role of the host-specific variation in NS3 protein turnover rate. IMPORTANCE: BTV is the causative agent of a severe disease transmitted between ruminants by biting midges of Culicoides species. NS3, encoded by Seg-10 of the BTV genome, fulfills key roles in BTV infection. As Seg-10 sequences from various BTV strains display genetic variability, we assessed the impact of different Seg-10 and NS3 proteins on BTV infection and host interactions. In this study, we revealed that various Seg-10/NS3 proteins alter BTV replication kinetics in mammals but not in insects. Notably, we found that NS3 protein turnover may vary in ovine but not in Culicoides cells due to a single amino acid residue that, most likely, leads to rapid and host-dependent protein degradation. Overall, this study highlights that genetically distant BTV Seg-10/NS3 influence BTV biological properties in a host-specific manner and increases our understanding of how NS3 proteins contribute to the outcome of BTV infection.


Asunto(s)
Virus de la Lengua Azul/genética , Células Endoteliales/virología , Regulación Viral de la Expresión Génica , Genoma Viral , Proteínas no Estructurales Virales/genética , Replicación Viral/genética , Secuencia de Aminoácidos , Animales , Aorta/metabolismo , Aorta/patología , Aorta/virología , Virus de la Lengua Azul/química , Virus de la Lengua Azul/metabolismo , Línea Celular Transformada , Ceratopogonidae , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Plexo Coroideo/virología , Cricetulus , Células Endoteliales/metabolismo , Células Endoteliales/patología , Especificidad del Huésped , Ratones , Datos de Secuencia Molecular , Cultivo Primario de Células , Estabilidad Proteica , Proteolisis , Genética Inversa , Ovinos , Transducción de Señal , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Liberación del Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...