Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Gen Virol ; 104(11)2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37976092

RESUMEN

Virus vectored vaccines are not available commercially for cattle even though compelling potential applications exist. Bovine papular stomatitis virus (BPSV), a highly prevalent parapoxvirus, causes self-limited oral lesions in cattle. Ability of virus to accommodate large amounts of foreign DNA, induce low level of antiviral immunity, and circulate and likely persist in cattle populations, make BPSV an attractive candidate viral vector. Here, recombinant BPSV were constructed expressing either Bovine herpesvirus 1 (BoHV-1) glycoprotein gD (BPSVgD), or gD and gB (BPSVgD/gB). Immunization of BPSV serologically-positive calves with BPSVgD or BPSVgD/gB induced BoHV-1 neutralization antibodies and provided protection for three of four animals following a high dose BoHV-1 challenge at day 70 pi. Results indicate BPSV suitability as a candidate virus vector for cattle vaccines.


Asunto(s)
Enfermedades de los Bovinos , Herpesvirus Bovino 1 , Parapoxvirus , Estomatitis , Vacunas , Vacunas Virales , Bovinos , Animales , Parapoxvirus/genética , Anticuerpos Antivirales , Herpesvirus Bovino 1/genética , Vacunas Virales/genética , Enfermedades de los Bovinos/prevención & control
2.
PLoS Pathog ; 17(10): e1009971, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34614034

RESUMEN

Viruses have evolved mechanisms to subvert critical cellular signaling pathways that regulate a wide range of cellular functions, including cell differentiation, proliferation and chemotaxis, and innate immune responses. Here, we describe a novel ORFV protein, ORFV113, that interacts with the G protein-coupled receptor Lysophosphatidic acid receptor 1 (LPA1). Consistent with its interaction with LPA1, ORFV113 enhances p38 kinase phosphorylation in ORFV infected cells in vitro and in vivo, and in cells transiently expressing ORFV113 or treated with soluble ORFV113. Infection of cells with virus lacking ORFV113 (OV-IA82Δ113) significantly decreased p38 phosphorylation and viral plaque size. Infection of cells with ORFV in the presence of a p38 kinase inhibitor markedly diminished ORFV replication, highlighting importance of p38 signaling during ORFV infection. ORFV113 enhancement of p38 activation was prevented in cells in which LPA1 expression was knocked down and in cells treated with LPA1 inhibitor. Infection of sheep with OV-IA82Δ113 led to a strikingly attenuated disease phenotype, indicating that ORFV113 is a major virulence determinant in the natural host. Notably, ORFV113 represents the first viral protein that modulates p38 signaling via interaction with LPA1 receptor.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Infecciones por Poxviridae/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Proteínas Virales/metabolismo , Animales , Parapoxvirus , Ovinos
3.
J Virol ; 95(15): e0036121, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980594

RESUMEN

Foot-and-mouth disease virus (FMDV) is the pathogen of foot-and-mouth disease (FMD), which is a highly contagious disease in cloven-hoofed animals. To survive in the host, FMDV has evolved multiple strategies to antagonize host innate immune responses. In this study, we showed that the leader protease (Lpro) of FMDV, a papain-like proteinase, promoted viral replication by evading the antiviral interferon response through counteracting the 2',5'-oligoadenylate synthetase (OAS)/RNase L system. Specifically, we observed that the titers of Lpro deletion virus were significantly lower than those of wild-type FMDV (FMDV-WT) in cultured cells. Our mechanistic studies demonstrated that Lpro interfered with the OAS/RNase L pathway by interacting with the N-terminal domain of swine RNase L (sRNase L). Remarkably, Lpro of FMDV exhibited species-specific binding to RNase L in that the interaction was observed only in swine cells, not human, monkey, or canine cells. Lastly, we presented evidence that by interacting with sRNase L, FMDV Lpro inhibited cellular apoptosis. Taken together, these results demonstrate a novel mechanism that Lpro utilizes to escape the OAS/RNase L-mediated antiviral defense pathway. IMPORTANCE FMDV is a picornavirus that causes a significant disease in agricultural animals. FMDV has developed diverse strategies to escape the host interferon response. Here, we show that Lpro of FMDV antagonizes the OAS/RNase L pathway, an important interferon effector pathway, by interacting with the N-terminal domain of sRNase L. Interestingly, such a virus-host interaction is species-specific because the interaction is detected only in swine cells, not in human, monkey, or canine cells. Furthermore, Lpro inhibits apoptosis through interacting with sRNase L. This study demonstrates a novel mechanism by which FMDV has evolved to inhibit host innate immune responses.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Endopeptidasas/metabolismo , Endorribonucleasas/metabolismo , Virus de la Fiebre Aftosa/inmunología , Evasión Inmune/inmunología , Inmunidad Innata/inmunología , Animales , Apoptosis/inmunología , Línea Celular , Cricetinae , Perros , Endopeptidasas/genética , Endopeptidasas/inmunología , Endorribonucleasas/genética , Fiebre Aftosa/inmunología , Fiebre Aftosa/virología , Células HEK293 , Haplorrinos , Humanos , Evasión Inmune/genética , Células de Riñón Canino Madin Darby , Dominios Proteicos , Porcinos
4.
Clin Infect Dis ; 70(8): 1768-1773, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-31620776

RESUMEN

Lyme disease, caused by some Borrelia burgdorferi sensu lato, is the most common tick-borne illness in the Northern Hemisphere and the number of cases, and geographic spread, continue to grow. Previously identified B. burgdorferi proteins, lipid immunogens, and live mutants lead the design of canonical vaccines aimed at disrupting infection in the host. Discovery of the mechanism of action of the first vaccine catalyzed the development of new strategies to control Lyme disease that bypassed direct vaccination of the human host. Thus, novel prevention concepts center on proteins produced by B. burgdorferi during tick transit and on tick proteins that mediate feeding and pathogen transmission. A burgeoning area of research is tick immunity as it can unlock mechanistic pathways that could be targeted for disruption. Studies that shed light on the mammalian immune pathways engaged during tick-transmitted B. burgdorferi infection would further development of vaccination strategies against Lyme disease.


Asunto(s)
Borrelia burgdorferi , Ixodes , Enfermedad de Lyme , Garrapatas , Vacunas , Animales , Humanos , Enfermedad de Lyme/prevención & control , Vacunación
5.
J Gen Virol ; 100(2): 259-265, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30628881

RESUMEN

African swine fever (ASF) is an emerging disease threat for the swine industry worldwide. No ASF vaccine is available, and progress is hindered by lack of knowledge concerning the extent of ASF virus (ASFV) strain diversity and the viral antigens conferring type-specific protective immunity in pigs. Previously, we demonstrated that ASFV serotype-specific proteins CD2v (EP402R) and/or C-type lectin (EP153R) are important for protection against homologous ASF infection. Here, we identified six discrete T-cell epitope regions present on CD2v and C-type lectin using IFN-γ ELISpot assay and PBMCs from ASF immune animals, indicating cellular reactivity to these proteins in the context of ASFV infection and protective immunity. Notably, three of the epitope regions map to previously described serotype-specific signature regions of these proteins. Improved understanding of ASFV protective antigens, relevant epitopes and their diversity in nature will facilitate ASFV subunit vaccine design and development.


Asunto(s)
Virus de la Fiebre Porcina Africana/inmunología , Epítopos de Linfocito T , Lectinas Tipo C/inmunología , Proteínas Virales/inmunología , Animales , Ensayo de Immunospot Ligado a Enzimas , Mapeo Epitopo , Interferón gamma/metabolismo , Porcinos
6.
PLoS Pathog ; 13(12): e1006779, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29244863

RESUMEN

Poxviruses have evolved multiple strategies to subvert signaling by Nuclear Factor κB (NF-κB), a crucial regulator of host innate immune responses. Here, we describe an orf virus (ORFV) virion-associated protein, ORFV119, which inhibits NF-κB signaling very early in infection (≤ 30 min post infection). ORFV119 NF-κB inhibitory activity was found unimpaired upon translation inhibition, suggesting that virion ORFV119 alone is responsible for early interference in signaling. A C-terminal LxCxE motif in ORFV119 enabled the protein to interact with the retinoblastoma protein (pRb) a multifunctional protein best known for its tumor suppressor activity. Notably, experiments using a recombinant virus containing an ORFV119 mutation which abrogates its interaction with pRb together with experiments performed in cells lacking or with reduced pRb levels indicate that ORFV119 mediated inhibition of NF-κB signaling is largely pRb dependent. ORFV119 was shown to inhibit IKK complex activation early in infection. Consistent with IKK inhibition, ORFV119 also interacted with TNF receptor associated factor 2 (TRAF2), an adaptor protein recruited to signaling complexes upstream of IKK in infected cells. ORFV119-TRAF2 interaction was enhanced in the presence of pRb, suggesting that ORFV119-pRb complex is required for efficient interaction with TRAF2. Additionally, transient expression of ORFV119 in uninfected cells was sufficient to inhibit TNFα-induced IKK activation and NF-κB signaling, indicating that no other viral proteins are required for the effect. Infection of sheep with ORFV lacking the ORFV119 gene led to attenuated disease phenotype, indicating that ORFV119 contributes to virulence in the natural host. ORFV119 represents the first poxviral protein to interfere with NF-κB signaling through interaction with pRb.


Asunto(s)
FN-kappa B/fisiología , Virus del Orf/fisiología , Virus del Orf/patogenicidad , Proteína de Retinoblastoma/fisiología , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Ectima Contagioso/etiología , Técnicas de Silenciamiento del Gen , Genes Virales , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Humanos , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Mutación , FN-kappa B/antagonistas & inhibidores , Virus del Orf/genética , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/genética , Ovinos , Transducción de Señal , Factor 2 Asociado a Receptor de TNF/metabolismo , Proteínas Virales/genética , Proteínas Virales/inmunología , Virulencia/genética , Virulencia/inmunología , Virulencia/fisiología
7.
PLoS Pathog ; 13(8): e1006561, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28787456

RESUMEN

Poxviruses have evolved unique proteins and mechanisms to counteract the nuclear factor κB (NF-κB) signaling pathway, which is an essential regulatory pathway of host innate immune responses. Here, we describe a NF-κB inhibitory virion protein of orf virus (ORFV), ORFV073, which functions very early in infected cells. Infection with ORFV073 gene deletion virus (OV-IA82Δ073) led to increased accumulation of NF-κB essential modulator (NEMO), marked phosphorylation of IκB kinase (IKK) subunits IKKα and IKKß, IκBα and NF-κB subunit p65 (NF-κB-p65), and to early nuclear translocation of NF-κB-p65 in virus-infected cells (≤ 30 min post infection). Expression of ORFV073 alone was sufficient to inhibit TNFα induced activation of the NF-κB signaling in uninfected cells. Consistent with observed inhibition of IKK complex activation, ORFV073 interacted with the regulatory subunit of the IKK complex NEMO. Infection of sheep with OV-IA82Δ073 led to virus attenuation, indicating that ORFV073 is a virulence determinant in the natural host. Notably, ORFV073 represents the first poxviral virion-associated NF-κB inhibitor described, highlighting the significance of viral inhibition of NF-κB signaling very early in infection.


Asunto(s)
Ectima Contagioso/virología , Evasión Inmune/fisiología , FN-kappa B/inmunología , Virus del Orf/patogenicidad , Virión/inmunología , Animales , Ectima Contagioso/inmunología , Células HeLa , Humanos , Inmunoprecipitación , Virus del Orf/inmunología , Virus del Orf/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Ovinos , Transducción de Señal/inmunología , Proteínas Virales/inmunología , Virulencia/fisiología
8.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2973-2986, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28768149

RESUMEN

A variable-length poly-T variant in intron 6 of the TOMM40 gene, rs10524523, is associated with risk and age-of-onset of sporadic (late-onset) Alzheimer's disease. In Caucasians, the three predominant alleles at this locus are Short (S), Long (L) or Very long (VL). On an APOE ε3/3 background, the S/VL and VL/VL genotypes are more protective than S/S. The '523 poly-T has regulatory properties, in that the VL poly-T results in higher expression than the S poly-T in luciferase expression systems. The aim of the current work was to identify effects on cellular bioenergetics of increased TOM40 protein expression. MitoTracker Green fluorescence and autophagic vesicle staining was the same in control and over-expressing cells, but TOM40 over-expression was associated with increased expression of TOM20, a preprotein receptor of the TOM complex, the mitochondrial chaperone HSPA9, and PDHE1a, and increased activities of the oxidative phosphorylation complexes I and IV and of the TCA member α-ketoglutaric acid dehydrogenase. Consistent with the complex I findings, respiration was more sensitive to inhibition by rotenone in control cells than in the TOM40 over-expressing cells. In the absence of inhibitors, total cellular ATP, the mitochondrial membrane potential, and respiration were elevated in the over-expressing cells. Spare respiratory capacity was greater in the TOM40 over-expressing cells than in the controls. TOM40 over-expression blocked Ab-elicited decreases in the mitochondrial membrane potential, cellular ATP levels, and cellular viability in the control cells. These data suggest elevated expression of TOM40 may be protective of mitochondrial function.


Asunto(s)
Enfermedad de Alzheimer , Regulación de la Expresión Génica , Potencial de la Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana , Mitocondrias , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Femenino , Sitios Genéticos , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Humanos , Complejo Cetoglutarato Deshidrogenasa/genética , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Proteínas de Transporte de Membrana/biosíntesis , Proteínas de Transporte de Membrana/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética
9.
Arch Virol ; 160(6): 1527-32, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25804193

RESUMEN

Bovine papular stomatitis virus (BPSV) infects cattle and, occupationally, humans. Prevalent subclinical infections, frequent reinfections, and virus persistence in healthy animals compound a poorly understood, but likely complex, scenario of BPSV perpetuation and transmission in nature. Here, we report the isolation of multiple BPSV strains coinfecting a single animal. Whole-genome analysis of isolated BPSV strains revealed genomic variability likely affecting virus virulence and infectivity. Further, incongruent phylogenetic relationships between viruses suggested genomic recombination. These results have significant implications for parapoxvirus infection biology and virus evolution in nature.


Asunto(s)
Enfermedades de los Bovinos/virología , Coinfección/veterinaria , Parapoxvirus/genética , Infecciones por Poxviridae/veterinaria , Animales , Secuencia de Bases , Bovinos/virología , Coinfección/virología , Datos de Secuencia Molecular , Sistemas de Lectura Abierta/genética , Filogenia , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/virología
10.
Arch Virol ; 160(1): 253-66, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25385177

RESUMEN

Orf virus (ORFV) is a typical member of the genus Parapoxvirus. The parapoxvirus genome consists of highly variable terminal regions and relatively conserved central regions with a high G + C content. In our previous study, a novel ORFV strain, NA1/11, was isolated from northeastern China. To fully characterize this strain, we sequenced the entire genome of NA1/11 and conducted a comparative analysis using multiple parapoxviruses. The genomic sequence of NA1/11 was found to consist of 137,080 nucleotides with a G + C content of 63.6 %, but it did not contain the terminal hairpin sequence. Alignment of ORFs from NA1/11 with NZ2, IA82 and SA00 revealed several highly variable ORFs, while the most evident ones are ORFs 001, 103, 109-110, 116 and 132. An odd phenomenon in the region of ORFs 118-120 is that the non-coding fragments are almost as long as the coding fragments. By comparative analysis of inverted terminal repeats, we identified one repeat motif and a long conserved fragment. By comparing the ITRs of SA00 with those of three other ORFVs, more clues were obtained about the correlation between ITR sequence and host adaption. Comparison of the NA1/11 genome with the sequences of other strains of ORFV revealed highly variable regions, thus providing new insights into the genetic diversity of ORFV.


Asunto(s)
Ectima Contagioso/virología , Virus del Orf/genética , Parapoxvirus/genética , Animales , China/epidemiología , Ectima Contagioso/epidemiología , Regulación Viral de la Expresión Génica , Genoma Viral , Datos de Secuencia Molecular , Virus del Orf/clasificación , Parapoxvirus/clasificación , Ovinos , Secuencias Repetidas Terminales , Proteínas Virales/genética , Proteínas Virales/metabolismo
12.
BMC Vet Res ; 8: 229, 2012 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-23174032

RESUMEN

BACKGROUND: Orf is a zoonotic and epitheliotrophic contagious disease that mainly affects sheep, goats, wild ruminants, and humans with a worldwide distribution. To date, there is little information on the characterization of ORFV strains that are endemic in Mainland China. In addition, the relationship between the severity of disease and the molecular profile of ORFV strains has not been fully elucidated. RESULTS: From the recent outbreak of a sheep herd in Nongan, northeast of China, the novel orf virus (ORFV) strain NA1/11 was successfully isolated. Western blot analysis indicated that the NA1/11 strain cross reacts with monoclonal antibody A3 and infected sheep ORFV antiserum. The purified virions revealed the typical ovoid shape when observed by atomic force microscopy. To determine the genetic characteristics of the NA1/11 strain, the sequences of ORFV011 (B2L), ORFV059 (F1L), ORFV109, ORFV110 and ORFv132 (VEGF) genes were amplified and compared with reference parapoxvirus strains. Non-metric multidimensional scaling (nMDS) was performed to analyze the nucleotide similarities between different ORFV strains. CONCLUSIONS: Phylogenetic analysis based on ORFV 011 nucleotide sequences showed that the NA1/11strain was closely related to Xinjiang and Gansu strains. ORFV110 and ORFV132 genes are highly variable. The results revealed that precise phylogenetic analysis might provide evidence for genetic variation and movement of circulating ORFV strains in Northeast China. In addition, nMDS analysis showed that geographic isolation and animal host are likely major factors resulting in genetic differences between ORFV strains.


Asunto(s)
Brotes de Enfermedades/veterinaria , Ectima Contagioso/virología , Virus del Orf/aislamiento & purificación , Zoonosis/virología , Animales , Secuencia de Bases , Western Blotting/veterinaria , China/epidemiología , ADN Viral/química , ADN Viral/genética , Ectima Contagioso/epidemiología , Microscopía de Fuerza Atómica/veterinaria , Microscopía Electrónica de Transmisión/veterinaria , Datos de Secuencia Molecular , Virus del Orf/genética , Virus del Orf/ultraestructura , Filogenia , Reacción en Cadena de la Polimerasa/veterinaria , Alineación de Secuencia , Análisis de Secuencia de ADN , Ovinos , Virión/ultraestructura , Zoonosis/epidemiología
13.
BMC Vet Res ; 7: 80, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-22192523

RESUMEN

BACKGROUND: Reporter genes are often used as a selectable marker for generation of recombinant viruses in order to investigate the mechanism of pathogenesis and to obtain candidate vaccine viruses. Routine selection of the recombinant parapoxvirus is time-consuming and labor intensive. Therefore, developing a novel method for selection is critical. RESULTS: In this study, we developed a rapid method to generate recombinant Orf viruses (ORFV) based on the enhanced green fluorescent protein (EGFP) reporter gene as a selectable marker. The coding sequence of EGFP gene was amplified from pEGFP-N1 vector and subcloned into the pZIPPY-neo/gus plasmid under the control of the early-late vaccinia virus (VACV) VV7.5 promoter and flanked by two multiple cloning sites (MCS) to generate a novel transfer vector pSPV-EGFP. Using the pSPV-EGFP, two recombination cassettes pSPV-113LF-EGFP-113RF and pSPV-116LF-EGFP-116RF were constructed by cloning the flanking regions of the ORFV113 and ORFV116 and inserted into two MCS flanking the EGFP gene. Using this novel system, two single gene deletion mutants OV-IA82Δ113 and OV-IA82Δ116 were successfully generated. CONCLUSIONS: This approach shortens the time needed to generate recombinant ORFVs (rORFVs). Thus, the pSPV-EGFP vector provides a direct, fast, and convenient way to manipulate the recombinant viruses, indicating that it is highly suited for its designed purpose.


Asunto(s)
Genes Reporteros/genética , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Virus del Orf/genética , Animales , Línea Celular , Clonación Molecular/métodos , Efecto Citopatogénico Viral/genética , Microscopía Fluorescente , Mutagénesis Insercional/métodos , Plásmidos/genética , Ovinos , Transfección/veterinaria
14.
Viruses ; 13(5)2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34065425

RESUMEN

African swine fever (ASF) is an acute viral hemorrhagic disease of domestic swine with mortality rates approaching 100%. Devastating ASF outbreaks and continuing epidemics starting in the Caucasus region and now in the Russian Federation, Europe, China, and other parts of Southeast Asia (2007 to date) highlight its significance. ASF strain Georgia-07 and its derivatives are now endemic in extensive regions of Europe and Asia and are "out of Africa" forever, a situation that poses a grave if not an existential threat to the swine industry worldwide. While our current concern is Georgia-07, other emerging ASFV strains will threaten for the indefinite future. Economic analysis indicates that an ASF outbreak in the U.S. would result in approximately $15 billion USD in losses, assuming the disease is rapidly controlled and the U.S. is able to reenter export markets within two years. ASF's potential to spread and become endemic in new regions, its rapid and efficient transmission among pigs, and the relative stability of the causative agent ASF virus (ASFV) in the environment all provide significant challenges for disease control. Effective and robust methods, including vaccines for ASF response and recovery, are needed immediately.


Asunto(s)
Virus de la Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/prevención & control , Fiebre Porcina Africana/transmisión , Brotes de Enfermedades/veterinaria , Epidemias/veterinaria , Vacunas Virales/inmunología , Fiebre Porcina Africana/inmunología , Animales , Epidemias/prevención & control , Especificidad del Huésped , Porcinos , Proteínas Virales/genética , Vacunas Virales/clasificación
15.
Viruses ; 13(8)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34452346

RESUMEN

African swine fever (ASF) is a hemorrhagic disease of swine characterized by massive lymphocyte depletion in lymphoid tissues due to the apoptosis of B and T cells, a process likely triggered by factors released or secreted by infected macrophages. ASFV CD2v (EP402R) has been implicated in viral virulence and immunomodulation in vitro; however, its actual function(s) remains unknown. We found that CD2v expression in swine PK15 cells induces NF-κB-dependent IFN-ß and ISGs transcription and an antiviral state. Similar results were observed for CD2v protein treated swine PBMCs and macrophages, the major ASFV target cell. Notably, treatment of swine PBMCs and macrophages with CD2v protein induced apoptosis. Immunoprecipitation and colocalization studies revealed that CD2v interacts with CD58, the natural host CD2 ligand. Additionally, CD58 knockdown in cells or treatment of cells with an NF-κB inhibitor significantly reduced CD2v-mediated NF-κB activation and IFN-ß induction. Further, antibodies directed against CD2v inhibited CD2v-induced NF-κB activation and IFN-ß transcription in cells. Overall, results indicate that ASFV CD2v activates NF-κB, which induces IFN signaling and apoptosis in swine lymphocytes/macrophages. We propose that CD2v released from infected macrophages may be a significant factor in lymphocyte apoptosis observed in lymphoid tissue during ASFV infection in pigs.


Asunto(s)
Virus de la Fiebre Porcina Africana/metabolismo , Fiebre Porcina Africana/genética , Fiebre Porcina Africana/fisiopatología , Interferón beta/genética , Leucocitos Mononucleares/citología , Proteínas Virales/metabolismo , Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/genética , Animales , Apoptosis , Interferón beta/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Macrófagos/inmunología , Macrófagos/virología , FN-kappa B/genética , FN-kappa B/inmunología , Porcinos , Proteínas Virales/genética
16.
J Virol ; 83(13): 6681-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19369337

RESUMEN

Foot-and-mouth disease virus (FMDV) produces one of the most infectious of all livestock diseases, causing extensive economic loss in areas of breakout. Like other viral pathogens, FMDV recruits proteins encoded by host cell genes to accomplish the entry, replication, and release of infectious viral particles. To identify such host-encoded proteins, we employed an antisense RNA strategy and a lentivirus-based library containing approximately 40,000 human expressed sequence tags (ESTs) to randomly inactivate chromosomal genes in a bovine kidney cell line (LF-BK) that is highly susceptible to FMDV infection and then isolated clones that survived multiple rounds of exposure to the virus. Here, we report the identification of ESTs whose expression in antisense orientation limited host cell killing by FMDV and restricted viral propagation. The role of one such EST, that of ectonucleoside triphosphate diphosphohydrolase 6 (NTPDase6; also known as CD39L2), a membrane-associated ectonucleoside triphosphate diphosphohydrolase that previously was not suspected of involvement in the propagation of viral pathogens and which we now show is required for normal synthesis of FMDV RNA and proteins, is described in this report.


Asunto(s)
Virus de la Fiebre Aftosa/patogenicidad , Fiebre Aftosa/genética , Pirofosfatasas/genética , ARN sin Sentido/genética , Animales , Bovinos , Línea Celular , Etiquetas de Secuencia Expresada , Biblioteca de Genes , Humanos , ARN Viral/metabolismo
17.
Genome Announc ; 6(26)2018 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-29954896

RESUMEN

Here, we report the complete genome sequencing of strains A/equine/Kostanay/9/2012(H3N8) and A/equine/LKZ/9/2012(H3N8) of the equine influenza virus belonging to Florida sublineage, clade 2. The strains were isolated in 2012 in the northern and southern regions of Kazakhstan, respectively.

18.
Front Microbiol ; 9: 1056, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29896166

RESUMEN

Apoptosis, a significant form of cell death, has a leading role in the host cell defense against virus infection. Viruses have evolved a series of strategies that block apoptosis during the early stage of viral infection to enhance viral replication, and induce apoptosis in the late stages to facilitate viral particle release from the cells. Here we show that orf virus (ORFV), the causative agent of orf, encodes an apoptosis-inducing protein ORFV119. ORFV119 targets the mitochondria in host cells, inhibits cell proliferation, and induces cell apoptosis. Protein array data indicated that ORFV119 could induce apoptosis via up-regulation of Smac, Bak, and Bax and down-regulation of anti-apoptotic proteins Bcl-2 and cIAP-2. Activation of caspase-9 and caspase-3, and consequent PARP cleavage, ultimately lead to apoptosis. ORFV119 could also directly activate caspase-8 and induce Bid, involved in the extrinsic pathway, to achieve cell death. Furthermore, sequence analysis and experiments with mutants of ORFV119 introduced revealed that ORFV119 contains a key N-terminal domain that is necessary and sufficient to direct the protein to the mitochondria. Together, we report, for the first time, the identification of the novel apoptosis-inducing protein ORFV119 encoded by a parapoxvirus. This provides an important reference for the study of pathogenesis, identification of immunomodulation mechanisms of ORFV, and may lead to new strategies for orf disease control.

19.
Gene ; 661: 60-67, 2018 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-29605601

RESUMEN

OBJECTIVE: Orf virus (ORFV) is the pathogen causing contagious pustular dermatitis in goats, sheep and herdsmen. Evidence has confirmed that ORFV can be used as a preventive and therapeutic immunomodulatory agent in several animal models. Our previous data demonstrated that ORFV024 is able to inhibit activation of the NF-κB signaling pathway and act as an important modulator for early immune responses against viral infection. However, the molecular mechanism by which ORFV024 exerting biological function remains unclear. In the present study, we explored and analyzed the function of host cellular proteins that interact with ORFV024. METHODS: The yeast two-hybrid (Y2H) assay was performed to screen proteins interacting with ORFV024 using a cDNA library derived from primary ovine fetal turbinate cells (OFTu). Two of the screened proteins were further confirmed by confocal microscopy, His-tag pull-down assay and CO-Immunoprecipitation (CO-IP) assay. In addition, the ORFV024 interaction network was constructed using the STRING database. RESULTS: In this study, 11 ovine cellular proteins were found to interact with ORFV024. In view of the importance of LAGE3 and IGFBP6 in the ORFV024 functional analysis, we further constructed LAGE3 and IGFBP6 interaction networks. The interactions between ORFV024 and LAGE3 or IGFBP6 were confirmed by confocal microscopy, LAGE3 was further confirmed in the His-tag pull-down assay and CO-IP assay. CONCLUSIONS: Our findings indicate that ORFV024 can interact with ovine cellular proteins LAGE3 and IGFBP6.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Virus del Orf , Proteínas Virales/metabolismo , Animales , Proteínas Portadoras/aislamiento & purificación , Células Cultivadas , Clonación Molecular , Feto/citología , Interacciones Huésped-Patógeno/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/aislamiento & purificación , Virus del Orf/metabolismo , Unión Proteica , Ovinos , Técnicas del Sistema de Dos Híbridos
20.
Front Microbiol ; 8: 160, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28179903

RESUMEN

[This corrects the article on p. 1389 in vol. 7, PMID: 27679610.].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA