Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 95(14): e0012321, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-33952643

RESUMO

African swine fever virus (ASFV) causes a virulent, deadly infection in wild and domestic swine and is currently causing a pandemic covering a contiguous geographical area from Central and Eastern Europe to Asia. No commercial vaccines are available to prevent African swine fever (ASF), resulting in devastating economic losses to the swine industry. The most advanced vaccine candidates are live attenuated strains developed using a genetically modified virulent parental virus. Recently, we developed a vaccine candidate, ASFV-G-ΔI177L, by deleting the I177L gene from the genome of the highly virulent ASFV pandemic strain Georgia (ASFV-G). ASFV-G-ΔI177L is safe and highly efficacious in challenge studies using parental ASFV-G. Large-scale production of ASFV-G-ΔI177L has been limited because it can replicate efficiently only in primary swine macrophages. Here, we present the development of an ASFV-G-ΔI177L derivative strain, ASFV-G-ΔI177L/ΔLVR, that replicates efficiently in a stable porcine cell line. In challenge studies, ASFV-G-ΔI177L/ΔLVR maintained the same level of attenuation, immunogenic characteristics, and protective efficacy as ASFV-G-ΔI177L. ASFV-G-ΔI177L/ΔLVR is the first rationally designed ASF vaccine candidate that can be used for large-scale commercial vaccine manufacture. IMPORTANCE African swine fever is currently causing a pandemic resulting in devastating losses to the swine industry. Experimental ASF vaccines rely on the production of vaccine in primary swine macrophages, which are difficult to use for the production of a vaccine on a commercial level. Here, we report a vaccine for ASFV with a deletion in the left variable region (LVR). This deletion allows for growth in stable cell cultures while maintaining the potency and efficacy of the parental vaccine strain. This discovery will allow for the production of an ASF vaccine on a commercial scale.


Assuntos
Vírus da Febre Suína Africana/imunologia , Febre Suína Africana/prevenção & controle , Vacinas Virais/imunologia , Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Animais , Técnicas de Cultura de Células , Linhagem Celular , Imunogenicidade da Vacina , Macrófagos/virologia , Pandemias , Deleção de Sequência , Suínos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/genética , Cultura de Vírus/métodos , Replicação Viral
2.
J Virol ; 95(12)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33827941

RESUMO

The classical swine fever virus (CSFV) glycoprotein E2 is the major structural component of the virus particle. E2 is involved in several functions, such as virus adsorption to the cell, the elicitation of protective immune responses, and virus virulence in swine. Using a yeast two-hybrid system, we previously identified the swine host protein Torsin-1A, an ATPase protein residing in the endoplasmic reticulum and inner nucleus membrane of the cell, as a specific binding partner for E2. The interaction between Torsin-1A and E2 proteins was confirmed to occur in CSFV-infected swine cells using three independent methods: coimmunoprecipitation, confocal microscopy, and proximity ligation assay (PLA). Furthermore, the E2 residue critical to mediate the protein-protein interaction with Torsin-1A was identified by a reverse yeast two-hybrid assay using a randomly mutated E2 library. A recombinant CSFV E2 mutant protein with a Q316L substitution failed to bind swine Torsin-1A in the yeast two-hybrid model. In addition, a CSFV infectious clone harboring the E2 Q316L substitution, although expressing substantial levels of E2 protein, repetitively failed to produce virus progeny when the corresponding RNA was transfected into susceptible SK6 cells. Importantly, PLA analysis of the transfected cells demonstrated an abolishment of the interaction between E2 Q316L and Torsin-1A, indicating a critical role for that interaction during CSFV replication.IMPORTANCE Structural glycoprotein E2 is an important structural component of the CSFV particle. E2 is involved in several virus functions, particularly virus-host interactions. Here, we characterized the interaction between CSFV E2 and swine protein Torsin-1A during virus infection. The critical amino acid residue in E2 mediating the interaction with Torsin-1A was identified and the effect of disrupting the E2-Torsin-1A protein-protein interaction was studied using reverse genetics. It is shown that the amino acid substitution abrogating E2-Torsin-1A interaction constitutes a lethal mutation, demonstrating that this virus-host protein-protein interaction is a critical factor during CSFV replication. This highlights the potential importance of the E2-Torsin-1A protein-protein interaction during CSFV replication and provides a potential pathway toward blocking virus replication, an important step toward the potential development of novel virus countermeasures.


Assuntos
Vírus da Febre Suína Clássica/fisiologia , Chaperonas Moleculares/metabolismo , Proteínas do Envelope Viral/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular , Vírus da Febre Suína Clássica/metabolismo , Interações Hospedeiro-Patógeno , Chaperonas Moleculares/genética , Mutação , Ligação Proteica , Proteínas Recombinantes/metabolismo , Suínos , Técnicas do Sistema de Duplo-Híbrido , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Replicação Viral
3.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597779

RESUMO

The E2 protein in classical swine fever (CSF) virus (CSFV) is the major virus structural glycoprotein and is an essential component of the viral particle. E2 has been shown to be involved in several functions, including virus adsorption, induction of protective immunity, and virulence in swine. Using the yeast two-hybrid system, we previously identified a swine host protein, dynactin subunit 6 (DCTN6) (a component of the cell dynactin complex), as a specific binding partner for E2. We confirmed the interaction between DCTN6 and E2 proteins in CSFV-infected swine cells by using two additional independent methodologies, i.e., coimmunoprecipitation and proximity ligation assays. E2 residues critical for mediating the protein-protein interaction with DCTN6 were mapped by a reverse yeast two-hybrid approach using a randomly mutated E2 library. A recombinant CSFV mutant, E2ΔDCTN6v, harboring specific substitutions in those critical residues was developed to assess the importance of the E2-DCTN6 protein-protein interaction for virus replication and virulence in swine. CSFV E2ΔDCTN6v showed reduced replication, compared with the parental virus, in an established swine cell line (SK6) and in primary swine macrophage cultures. Remarkably, animals infected with CSFV E2ΔDCTN6v remained clinically normal during the 21-day observation period, which suggests that the ability of CSFV E2 to bind host DCTN6 protein efficiently during infection may play a role in viral virulence.IMPORTANCE Structural glycoprotein E2 is an important component of CSFV due to its involvement in many virus activities, particularly virus-host interactions. Here, we present the description and characterization of the protein-protein interaction between E2 and the swine host protein DCTN6 during virus infection. The E2 amino acid residues mediating the interaction with DCTN6 were also identified. A recombinant CSFV harboring mutations disrupting the E2-DCTN6 interaction was created. The effect of disrupting the E2-DCTN6 protein-protein interaction was studied using reverse genetics. It was shown that the same amino acid substitutions that abrogated the E2-DCTN6 interaction in vitro constituted a critical factor in viral virulence in the natural host, domestic swine. This highlights the potential importance of the E2-DCTN6 protein-protein interaction in CSFV virulence and provides possible mechanisms of virus attenuation for the development of improved CSF vaccines.


Assuntos
Vírus da Febre Suína Clássica/genética , Peste Suína Clássica/virologia , Complexo Dinactina/genética , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Proteínas do Envelope Viral/genética , Animais , Sítios de Ligação , Linhagem Celular , Peste Suína Clássica/mortalidade , Peste Suína Clássica/patologia , Vírus da Febre Suína Clássica/metabolismo , Vírus da Febre Suína Clássica/patogenicidade , Complexo Dinactina/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Biblioteca Gênica , Macrófagos/metabolismo , Macrófagos/virologia , Mutação , Cultura Primária de Células , Ligação Proteica , Transdução de Sinais , Análise de Sobrevida , Suínos , Técnicas do Sistema de Duplo-Híbrido , Proteínas do Envelope Viral/metabolismo , Replicação Viral
5.
Virology ; 526: 38-44, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30340154

RESUMO

Envelope glycoprotein E2 of Classical Swine Fever Virus (CSFV) is involved in several critical virus functions. To analyze the role of E2 in virus replication, a series of recombinant CSFVs harboring chimeric forms of E2 CSFV and Bovine viral diarrhea virus (BVDV) were created and tested for their ability to infect swine or bovine cell lines. Substitution of native CSFV E2 by BVDV E2 abrogates virus replication in both cell lines. Substitution of individual domains in CSFV Brescia E2 by the homologous from BVDV produces chimeras that efficiently replicate in SK6 cells with the exception of a chimera harboring BVDV E2 residues 93-168. Further mapping revealed a critical area in E2 required for CSFV replication in SK6 cells between protein residues 136-156. This is the first report categorically defining a discrete portion of E2 as essential to pestivirus infection in susceptible cells.


Assuntos
Vírus da Febre Suína Clássica/fisiologia , Vírus da Diarreia Viral Bovina/fisiologia , Infecções por Pestivirus/virologia , Domínios Proteicos/genética , Proteínas do Envelope Viral/química , Replicação Viral/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Bovinos , Linhagem Celular , Vírus da Febre Suína Clássica/genética , Vírus da Febre Suína Clássica/patogenicidade , Vírus da Diarreia Viral Bovina/genética , Vírus da Diarreia Viral Bovina/patogenicidade , Especificidade de Hospedeiro , Vírus Reordenados/genética , Vírus Reordenados/patogenicidade , Vírus Reordenados/fisiologia , Suínos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
6.
J Virol ; 92(23)2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30232178

RESUMO

Nonstructural protein 2B of foot-and-mouth disease (FMD) virus (FMDV) is comprised of a small, hydrophobic, 154-amino-acid protein. Structure-function analyses demonstrated that FMDV 2B is an ion channel-forming protein. Infrared spectroscopy measurements using partially overlapping peptides that spanned regions between amino acids 28 and 147 demonstrated the adoption of helical conformations in two putative transmembrane regions between residues 60 and 78 and between residues 119 and 147 and a third transmembrane region between residues 79 and 106, adopting a mainly extended structure. Using synthetic peptides, ion channel activity measurements in planar lipid bilayers and imaging of single giant unilamellar vesicles (GUVs) revealed the existence of two sequences endowed with membrane-porating activity: one spanning FMDV 2B residues 55 to 82 and the other spanning the C-terminal region of 2B from residues 99 to 147. Mapping the latter sequence identified residues 119 to 147 as being responsible for the activity. Experiments to assess the degree of insertion of the synthetic peptides in bilayers and the inclination angle adopted by each peptide regarding the membrane plane normal confirm that residues 55 to 82 and 119 to 147 of 2B actively insert as transmembrane helices. Using reverse genetics, a panel of 13 FMD recombinant mutant viruses was designed, which harbored nonconservative as well as alanine substitutions in critical amino acid residues in the area between amino acid residues 28 and 147. Alterations to any of these structures interfered with pore channel activity and the capacity of the protein to permeabilize the endoplasmic reticulum (ER) to calcium and were lethal for virus replication. Thus, FMDV 2B emerges as the first member of the viroporin family containing two distinct pore domains.IMPORTANCE FMDV nonstructural protein 2B is able to insert itself into cellular membranes to form a pore. This pore allows the passage of ions and small molecules through the membrane. In this study, we were able to show that both current and small molecules are able to pass though the pore made by 2B. We also discovered for the first time a virus with a pore-forming protein that contains two independent functional pores. By making mutations in our infectious clone of FMDV, we determined that mutations in either pore resulted in nonviable virus. This suggests that both pore-forming functions are independently required during FMDV infection.


Assuntos
Permeabilidade da Membrana Celular , Vírus da Febre Aftosa/metabolismo , Febre Aftosa/metabolismo , Bicamadas Lipídicas/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Animais , Células Cultivadas , Cricetinae , Febre Aftosa/genética , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Transporte de Íons , Mutação , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Domínios Proteicos , Homologia de Sequência , Proteínas não Estruturais Virais/genética
7.
J Virol ; 90(22): 10299-10308, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27605674

RESUMO

E2, the major envelope glycoprotein of classical swine fever virus (CSFV), is involved in several critical virus functions, including cell attachment, host range susceptibility, and virulence in natural hosts. Functional structural analysis of E2 based on a Wimley-White interfacial hydrophobicity distribution predicted the involvement of a loop (residues 864 to 881) stabilized by a disulfide bond (869CKWGGNWTCV878, named FPII) in establishing interactions with the host cell membrane. This loop further contains an 872GG873 dipeptide, as well as two aromatic residues (871W and 875W) accessible to solvent. Reverse genetics utilizing a full-length infectious clone of the highly virulent CSFV strain Brescia (BICv) was used to evaluate how amino acid substitutions within FPII may affect replication of BICv in vitro and virus virulence in swine. Recombinant CSFVs containing mutations in different residues of FPII were constructed. A particular construct, harboring amino acid substitutions W871T, W875D, and V878T (FPII.2), demonstrated a significantly decreased ability to replicate in a swine cell line (SK6) and swine macrophage primary cell cultures. Interestingly, mutated virus FPII.2 was completely attenuated in pigs. Also, animals infected with FPII.2 virus were protected against virulent challenge with Brescia virus at 21 days postvaccination. Supporting a role for the E2 the loop from residues 864 to 881 in membrane fusion, only synthetic peptides that were based on the native E2 functional sequence were competent for insertion into model membranes and perturbation of their integrity, and this functionality was lost in synthetic peptides harboring amino acid substitutions W871T, W875D, and V878T in FPII.2. IMPORTANCE: This report describes the identification and characterization of a putative fusion peptide (FP) in the major structural protein E2 of classical swine fever virus (CSFV). The FP identification was performed by functional structural analysis of E2. We characterized the functional significance of this FP by using artificial membranes. Replacement of critical amino acid residues within the FP radically alters how it interacts with the artificial membranes. When we introduced the same mutations into the viral sequence, there was a reduction in replication in cell cultures, and when we infected domestic swine, the natural host of CSFV host, we observed that the virus was now completely attenuated in swine. In addition, the virus mutant that was attenuated in vivo efficiently protected pigs against wild-type virus. These results provide the proof of principle to support as a strategy for vaccine development the discovery and manipulation of FPs.


Assuntos
Vírus da Febre Suína Clássica/genética , Glicoproteínas/genética , Peptídeos/genética , Virulência/genética , Replicação Viral/genética , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Peste Suína Clássica/virologia , Peptídeos e Proteínas de Sinalização Intercelular , Mutação/genética , Suínos , Proteínas do Envelope Viral/genética
8.
Virus Res ; 213: 165-171, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26656424

RESUMO

African swine fever virus (ASFV) is the etiological agent of a contagious and often lethal viral disease of domestic pigs. There are no vaccines to control Africa swine fever (ASF). Experimental vaccines have been developed using genetically modified live attenuated ASFVs obtained by specifically deleting virus genes involved in virulence, including the thymidine kinase (TK) gene. TK has been shown to be involved in the virulence of several viruses, including ASFV. Here we report the construction of a recombinant virus (ASFV-G/V-ΔTK) obtained by deleting the TK gene in a virulent strain of ASFV Georgia adapted to replicate in Vero cells (ASFV-G/VP30). ASFV-G/P-ΔTK demonstrated decreased replication both in primary swine macrophage cell cultures and in Vero cells compared with ASFV-G/VP30. In vivo, intramuscular administration of up to 10(6) TCID50 of ASFV-G/V-ΔTK does not result in ASF disease. However, these animals are not protected when challenged with the virulent parental Georgia strain.


Assuntos
Vírus da Febre Suína Africana/enzimologia , Vírus da Febre Suína Africana/patogenicidade , Febre Suína Africana/patologia , Deleção de Genes , Timidina Quinase/genética , Fatores de Virulência/genética , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/fisiologia , Animais , Chlorocebus aethiops , Células Epiteliais/virologia , Injeções Intramusculares , Macrófagos/virologia , Suínos , Timidina Quinase/metabolismo , Células Vero , Virulência , Fatores de Virulência/metabolismo , Replicação Viral
9.
Virology ; 471-473: 13-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25461526

RESUMO

Controlling classical swine fever (CSF) involves vaccination in endemic regions and preemptive slaughter of infected swine herds during epidemics. Live attenuated marker vaccines that confer effective protection against the disease and allow differentiation between infected and vaccinated animals (DIVA) could impact CSF control policies. Previously, we reported the development of FlagT4 virus (FlagT4v), a rationally designed live attenuated marker vaccine. During its vaccine assessment, FlagT4v reverted to a virulent virus during successive passages in piglets. Sequence analysis revealed deletions and substitutions almost exclusively in the areas of E1 and E2. To improve genetic stability of FlagT4v, we introduced changes in the codon usage in those areas. The newly developed virus, FlagT4Gv, was shown to retain the attenuated phenotype after successive passages in piglets. As observed with FlagT4v, the newly developed FlagT4Gv conferred effective protection against challenge with virulent CSFV at early (7 days) and at late (28 days) times post-vaccination.


Assuntos
Vírus da Febre Suína Clássica/genética , Vírus da Febre Suína Clássica/imunologia , Peste Suína Clássica/prevenção & controle , Vacinas Virais/imunologia , Animais , Linhagem Celular , Vírus da Febre Suína Clássica/patogenicidade , Feminino , Suínos , Vacinas Atenuadas/imunologia , Viremia , Virulência
10.
Virology ; 468-470: 185-196, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25216088

RESUMO

Foot-and-mouth disease virus (FMDV) produces a disease in cattle characterized by vesicular lesions and a persistent infection with asymptomatic low-level production of virus in pharyngeal tissues. Here we describe the establishment of a persistently infected primary cell culture derived from bovine pharynx tissue (PBPT) infected with FMDV serotype O1 Manisa, where surviving cells were serially passed until a persistently infected culture was generated. Characterization of the persistent virus demonstrated changes in its plaque size, ability to grow in different cell lines, and change in the use of integrins as receptors, when compared with the parental virus. These results demonstrate the establishment of persistently infected PBPT cell cultures where co-adaptation has taken place between the virus and host cells. This in vitro model for FMDV persistence may help further understanding of the molecular mechanisms of the cattle carrier state.


Assuntos
Vírus da Febre Aftosa/fisiologia , Faringe/citologia , Animais , Bovinos , Células , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Fatores de Tempo , Replicação Viral
11.
Virology ; 460-461: 173-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25010283

RESUMO

Classical swine fever virus (CSFV) Core protein is involved in virus RNA protection, transcription regulation and virus virulence. To discover additional Core protein functions a yeast two-hybrid system was used to identify host proteins that interact with Core. Among the identified host proteins, the osteosarcoma amplified 9 protein (OS9) was further studied. Using alanine scanning mutagenesis, the OS9 binding site in the CSFV Core protein was identified, between Core residues (90)IAIM(93), near a putative cleavage site. Truncated versions of Core were used to show that OS9 binds a polypeptide representing the 12 C-terminal Core residues. Cells transfected with a double-fluorescent labeled Core construct demonstrated that co-localization of OS9 and Core occurred only on unprocessed forms of Core protein. A recombinant CSFV containing Core protein where residues (90)IAIM(93) were substituted by alanines showed no altered virulence in swine, but a significant decreased ability to replicate in cell cultures.


Assuntos
Vírus da Febre Suína Clássica/metabolismo , Peste Suína Clássica/metabolismo , Degradação Associada com o Retículo Endoplasmático , Proteínas de Neoplasias/metabolismo , Proteínas do Core Viral/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Peste Suína Clássica/genética , Peste Suína Clássica/virologia , Vírus da Febre Suína Clássica/química , Vírus da Febre Suína Clássica/genética , Vírus da Febre Suína Clássica/patogenicidade , Interações Hospedeiro-Patógeno , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Ligação Proteica , Suínos , Técnicas do Sistema de Duplo-Híbrido , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Virulência
12.
Virology ; 456-457: 121-30, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24889231

RESUMO

E2, along with E(rns) and E1, is an envelope glycoprotein of Classical Swine Fever Virus (CSFV). E2 is involved in several virus functions: cell attachment, host range susceptibility and virulence in natural hosts. Here we evaluate the role of a specific E2 region, (818)CPIGWTGVIEC(828), containing a putative fusion peptide (FP) sequence. Reverse genetics utilizing a full-length infectious clone of the highly virulent CSFV strain Brescia (BICv) was used to evaluate how individual amino acid substitutions within this region of E2 may affect replication of BICv. A synthetic peptide representing the complete E2 FP amino acid sequence adopted a ß-type extended conformation in membrane mimetics, penetrated into model membranes, and perturbed lipid bilayer integrity in vitro. Similar peptides harboring amino acid substitutions adopted comparable conformations but exhibited different membrane activities. Therefore, a preliminary characterization of the putative FP (818)CPIGWTGVIEC(828) indicates a membrane fusion activity and a critical role in virus replication.


Assuntos
Vírus da Febre Suína Clássica/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Substituição de Aminoácidos , Animais , Linhagem Celular , Membrana Celular/metabolismo , Vírus da Febre Suína Clássica/genética , Lipossomos/metabolismo , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformação Proteica , Genética Reversa , Suínos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
13.
J Virol ; 88(5): 2737-47, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24352458

RESUMO

UNLABELLED: Nonstructural protein 3A of foot-and-mouth disease virus (FMDV) is a partially conserved protein of 153 amino acids in most FMDVs examined to date. The role of 3A in virus growth and virulence within the natural host is not well understood. Using a yeast two-hybrid approach, we identified cellular protein DCTN3 as a specific host binding partner for 3A. DCTN3 is a subunit of the dynactin complex, a cofactor for dynein, a motor protein. The dynactin-dynein duplex has been implicated in several subcellular functions involving intracellular organelle transport. The 3A-DCTN3 interaction identified by the yeast two-hybrid approach was further confirmed in mammalian cells. Overexpression of DCTN3 or proteins known to disrupt dynein, p150/Glued and 50/dynamitin, resulted in decreased FMDV replication in infected cells. We mapped the critical amino acid residues in the 3A protein that mediate the protein interaction with DCTN3 by mutational analysis and, based on that information, we developed a mutant harboring the same mutations in O1 Campos FMDV (O1C3A-PLDGv). Although O1C3A-PLDGv FMDV and its parental virus (O1Cv) grew equally well in LFBK-αvß6, O1C3A-PLDGv virus exhibited a decreased ability to replicate in primary bovine cell cultures. Importantly, O1C3A-PLDGv virus exhibited a delayed disease in cattle compared to the virulent parental O1Campus (O1Cv). Virus isolated from lesions of animals inoculated with O1C3A-PLDGv virus contained amino acid substitutions in the area of 3A mediating binding to DCTN3. Importantly, 3A protein harboring similar amino acid substitutions regained interaction with DCTN3, supporting the hypothesis that DCTN3 interaction likely contributes to virulence in cattle. IMPORTANCE: The objective of this study was to understand the possible role of a FMD virus protein 3A, in causing disease in cattle. We have found that the cellular protein, DCTN3, is a specific binding partner for 3A. It was shown that manipulation of DCTN3 has a profound effect in virus replication. We developed a FMDV mutant virus that could not bind DCTN3. This mutant virus exhibited a delayed disease in cattle compared to the parental strain highlighting the role of the 3A-DCTN3 interaction in virulence in cattle. Interestingly, virus isolated from lesions of animals inoculated with mutant virus contained mutations in the area of 3A that allowed binding to DCTN3. This highlights the importance of the 3A-DCTN3 interaction in FMD virus virulence and provides possible mechanisms of virus attenuation for the development of improved FMD vaccines.


Assuntos
Vírus da Febre Aftosa/fisiologia , Febre Aftosa/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Bovinos , Linhagem Celular , Complexo Dinactina , Vírus da Febre Aftosa/patogenicidade , Expressão Gênica , Humanos , Espaço Intracelular/metabolismo , Proteínas Associadas aos Microtúbulos/química , Dados de Sequência Molecular , Mutação , Ligação Proteica , Mapeamento de Interação de Proteínas , Alinhamento de Sequência , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Virulência , Replicação Viral
14.
J Virol ; 87(12): 6794-803, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23576498

RESUMO

Foot-and-mouth disease virus (FMDV), the causative agent of foot-and-mouth disease, is an Aphthovirus within the Picornaviridae family. During infection with FMDV, several host cell membrane rearrangements occur to form sites of viral replication. FMDV protein 2C is part of the replication complex and thought to have multiple roles during virus replication. To better understand the role of 2C in the process of virus replication, we have been using a yeast two-hybrid approach to identify host proteins that interact with 2C. We recently reported that cellular Beclin1 is a natural ligand of 2C and that it is involved in the autophagy pathway, which was shown to be important for FMDV replication. Here, we report that cellular vimentin is also a specific host binding partner for 2C. The 2C-vimentin interaction was further confirmed by coimmunoprecipitation and immunofluorescence staining to occur in FMDV-infected cells. It was shown that upon infection a vimentin structure forms around 2C and that this structure is later resolved or disappears. Interestingly, overexpression of vimentin had no effect on virus replication; however, overexpression of a truncated dominant-negative form of vimentin resulted in a significant decrease in viral yield. Acrylamide, which causes disruption of vimentin filaments, also inhibited viral yield. Alanine scanning mutagenesis was used to map the specific amino acid residues in 2C critical for vimentin binding. Using reverse genetics, we identified 2C residues that are necessary for virus growth, suggesting that the interaction between FMDV 2C and cellular vimentin is essential for virus replication.


Assuntos
Proteínas de Transporte/metabolismo , Células Epiteliais/virologia , Vírus da Febre Aftosa/fisiologia , Vimentina/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Proteínas de Transporte/genética , Linhagem Celular , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/metabolismo , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Técnicas do Sistema de Duplo-Híbrido , Vimentina/genética , Proteínas não Estruturais Virais/genética
15.
J Virol ; 86(22): 12080-90, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22933281

RESUMO

Foot-and-mouth disease virus (FMDV), the causative agent of foot-and-mouth disease, is an Apthovirus within the Picornaviridae family. Replication of the virus occurs in association with replication complexes that are formed by host cell membrane rearrangements. The largest viral protein in the replication complex, 2C, is thought to have multiple roles during virus replication. However, studies examining the function of FMDV 2C have been rather limited. To better understand the role of 2C in the process of virus replication, we used a yeast two-hybrid approach to identify host proteins that interact with 2C. We report here that cellular Beclin1 is a specific host binding partner for 2C. Beclin1 is a regulator of the autophagy pathway, a metabolic pathway required for efficient FMDV replication. The 2C-Beclin1 interaction was further confirmed by coimmunoprecipitation and confocal microscopy to actually occur in FMDV-infected cells. Overexpression of either Beclin1 or Bcl-2, another important autophagy factor, strongly affects virus yield in cell culture. The fusion of lysosomes to autophagosomes containing viral proteins is not seen during FMDV infection, a process that is stimulated by Beclin1; however, in FMDV-infected cells overexpressing Beclin1 this fusion occurs, suggesting that 2C would bind to Beclin1 to prevent the fusion of lysosomes to autophagosomes, allowing for virus survival. Using reverse genetics, we demonstrate here that modifications to the amino acids in 2C that are critical for interaction with Beclin1 are also critical for virus growth. These results suggest that interaction between FMDV 2C and host protein Beclin1 could be essential for virus replication.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Vírus da Febre Aftosa/metabolismo , Proteínas de Membrana/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Animais , Autofagia , Proteína Beclina-1 , Bovinos , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae/metabolismo , Células Epiteliais/citologia , Vírus da Febre Aftosa/genética , Biblioteca Gênica , Humanos , Glândulas Mamárias Humanas/metabolismo , Modelos Genéticos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Plasmídeos/metabolismo , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido
16.
Virology ; 412(1): 68-74, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21262517

RESUMO

Here we show that IQGAP1, a cellular protein that plays a pivotal role as a regulator of the cytoskeleton interacts with Classical Swine Fever Virus (CSFV) Core protein. Sequence analyses identified residues within CSFV Core protein (designated as areas I, II, III and IV) that maintain homology to regions within the matrix protein of Moloney Murine Leukemia Virus (MMLV) that mediate binding to IQGAP1 [EMBO J, 2006 25:2155]. Alanine-substitution within Core regions I, II, III and IV identified residues that specifically mediate the Core-IQGAP1 interaction. Recombinant CSFV viruses harboring alanine substitutions at residues (207)ATI(209) (I), (210)VVE(212) (II), (213)GVK(215) (III), or (232)GLYHN(236) (IV) have defective growth in primary swine macrophage cultures. In vivo, substitutions of residues in areas I and III yielded viruses that were completely attenuated in swine. These data shows that the interaction of Core with an integral component of cytoskeletal regulation plays a role in the CSFV cycle.


Assuntos
Vírus da Febre Suína Clássica/patogenicidade , Interações Hospedeiro-Patógeno , Mapeamento de Interação de Proteínas , Proteínas do Core Viral/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Células Cultivadas , Peste Suína Clássica/patologia , Peste Suína Clássica/virologia , Vírus da Febre Suína Clássica/genética , Macrófagos/virologia , Dados de Sequência Molecular , Vírus da Leucemia Murina de Moloney/genética , Mutagênese Sítio-Dirigida , Ligação Proteica , Homologia de Sequência de Aminoácidos , Suínos , Proteínas do Core Viral/genética , Virulência
17.
Virology ; 407(1): 129-36, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-20800867

RESUMO

Here we have identified host cell proteins involved with the cellular SUMOylation pathway, SUMO-1 (small ubiquitin-like modifier) and UBC9, a SUMO-1 conjugating enzyme that interact with classical swine fever virus (CSFV) Core protein. Five highly conserved lysine residues (K179, K180, K220, K221, and K246) within the CSFV Core were identified as putative SUMOylation sites. Analysis of these interactions showed that K179A, K180A, and K221A substitutions disrupt Core-SUMO-1 binding, while K220A substitution precludes Core-UBC9 binding. In vivo, Core mutant viruses (K179A, K180A, K220A, K221A) and (K220A, K221A) harboring those substitutions were attenuated in swine. These data shows a clear correlation between the disruption of Core protein binding to SUMO-1 and UBC9 and CSFV attenuation. Overall, these data suggest that the interaction of Core with the cellular SUMOylation pathway plays a significant role in the CSFV growth cycle in vivo.


Assuntos
Vírus da Febre Suína Clássica/patogenicidade , Mapeamento de Interação de Proteínas , Proteína SUMO-1/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Proteínas do Core Viral/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Peste Suína Clássica/patologia , Peste Suína Clássica/virologia , Lisina/genética , Lisina/metabolismo , Dados de Sequência Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Alinhamento de Sequência , Suínos , Virulência
18.
J Virol ; 84(3): 1536-49, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19923180

RESUMO

NS4B is one of the nonstructural proteins of classical swine fever virus (CSFV), the etiological agent of a severe, highly lethal disease of swine. Protein domain analysis of the predicted amino acid sequence of the NS4B protein of highly pathogenic CSFV strain Brescia (BICv) identified a putative Toll/interleukin-1 receptor (TIR)-like domain. This TIR-like motif harbors two conserved domains, box 1 and box 2, also observed in other members of the TIR superfamily, including Toll-like receptors (TLRs). Mutations within the BICv NS4B box 2 domain (V2566A, G2567A, I2568A) produced recombinant virus NS4B.VGIv, with an altered phenotype displaying enhanced transcriptional activation of TLR-7-induced genes in swine macrophages, including a significant sustained accumulation of interleukin-6 (IL-6) mRNA. Transfection of swine macrophages with the wild-type NS4B gene partially blocked the TLR-7-activating effect of imiquimod (R837), while transfection with the NS4B gene harboring mutations in either of the putative boxes displayed decreased blocking activity. NS4B.VGIv showed an attenuated phenotype in swine, displaying reduced replication in the oronasal cavity and limited spread from the inoculation site to secondary target organs. Furthermore, the level and duration of IL-6 production in the tonsils of pigs intranasally inoculated with NS4B.VGIv were significantly higher than those for animals infected with BICv. The peak of IL-6 production in infected animals paralleled the ability of animals infected with NS4B.VGIv to resist challenge with virulent BICv. Interestingly, treatment of peripheral blood mononuclear cell cultures with recombinant porcine IL-6 results in a significant decrease in BICv replication.


Assuntos
Vírus da Influenza A Subtipo H1N1/patogenicidade , Mutação , Proteínas não Estruturais Virais/fisiologia , Virulência/fisiologia , Sequência de Aminoácidos , Aminoquinolinas/farmacologia , Animais , Sequência de Bases , Primers do DNA , Imiquimode , Interleucina-6/genética , Macrófagos/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Suínos , Receptores Toll-Like/fisiologia , Transfecção , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
19.
Virology ; 384(1): 106-13, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19046591

RESUMO

Until recently strategies for controlling Classical Swine Fever Virus (CSFV) involve either prophylactic vaccination or non-vaccination with elimination of infected herds depending on the epidemiological situation of the affected geographical area. Marker vaccines allowing distinction between naturally infected from vaccinated swine could complement "stamping out" measures. Here we developed a double antigenic marker live attenuated CSFV strain FlagT4v obtained by combining two genetic determinants of attenuation. FlagT4v harbors a positive antigenic marker, synthetic Flag(R) epitope, introduced via a 19mer insertion in E1 glycoprotein; and a negative marker resulting from mutations of the binding site of monoclonal antibody WH303 (mAbWH303) epitope in E2 glycoprotein. Intranasal or intramuscular administration of FlagT4v protected swine against virulent CSFV Brescia strain at early (2 or 3 days), and late (28 days) time post-inoculation. FlagT4v induced antibody response in pigs reacted strongly against the Flag(R) epitope but failed to inhibit binding of mAbWH303 to a synthetic peptide representing the WH303 epitope. These results constitute a proof-of-concept for rationally designing a CSFV antigenically marked live attenuated virus.


Assuntos
Antígenos Virais/análise , Vírus da Febre Suína Clássica/imunologia , Peste Suína Clássica/imunologia , Vacinas Atenuadas , Vacinas Virais , Administração Intranasal , Sequência de Aminoácidos , Animais , Sequência de Bases , Marcadores Genéticos , Genoma Viral , Injeções Intramusculares , Dados de Sequência Molecular , Mutação , Suínos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/química , Vacinas Atenuadas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...