Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nucleic Acids Res ; 46(15): 7924-7937, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29796667

RESUMEN

To maintain genome integrity, segmented double-stranded RNA viruses of the Reoviridae family must accurately select and package a complete set of up to a dozen distinct genomic RNAs. It is thought that the high fidelity segmented genome assembly involves multiple sequence-specific RNA-RNA interactions between single-stranded RNA segment precursors. These are mediated by virus-encoded non-structural proteins with RNA chaperone-like activities, such as rotavirus (RV) NSP2 and avian reovirus σNS. Here, we compared the abilities of NSP2 and σNS to mediate sequence-specific interactions between RV genomic segment precursors. Despite their similar activities, NSP2 successfully promotes inter-segment association, while σNS fails to do so. To understand the mechanisms underlying such selectivity in promoting inter-molecular duplex formation, we compared RNA-binding and helix-unwinding activities of both proteins. We demonstrate that octameric NSP2 binds structured RNAs with high affinity, resulting in efficient intramolecular RNA helix disruption. Hexameric σNS oligomerizes into an octamer that binds two RNAs, yet it exhibits only limited RNA-unwinding activity compared to NSP2. Thus, the formation of intersegment RNA-RNA interactions is governed by both helix-unwinding capacity of the chaperones and stability of RNA structure. We propose that this protein-mediated RNA selection mechanism may underpin the high fidelity assembly of multi-segmented RNA genomes in Reoviridae.


Asunto(s)
Chaperonas Moleculares/metabolismo , Orthoreovirus Aviar/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencia de Bases , Genoma Viral/genética , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Conformación de Ácido Nucleico , Orthoreovirus Aviar/genética , Unión Proteica , Estructura Secundaria de Proteína , ARN Viral/química , ARN Viral/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
2.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29321312

RESUMEN

Avian reovirus (ARV) causes viral arthritis, chronic respiratory diseases, retarded growth, and malabsorption syndrome. The ARV p10 protein, a viroporin responsible for the induction of cell syncytium formation and apoptosis, is rapidly degraded in host cells. Our previous report demonstrated that cellular lysosome-associated membrane protein 1 (LAMP-1) interacted with p10 and was involved in its degradation. However, the molecular mechanism underlying LAMP-1-mediated p10 degradation remains elusive. We report here that the E3 ubiquitin ligase seven in absentia homolog 1 (Siah-1) is critical for p10 ubiquitylation. Our data show that Siah-1 ubiquitylated p10 and targeted it for proteasome degradation. Furthermore, the ubiquitylation of p10 by Siah-1 required the participation of LAMP-1 by forming a multicomponent complex. Thus, LAMP-1 promotes the proteasomal degradation of p10 via interacting with both p10 and the E3 ligase Siah-1. These data establish a novel host defense mechanism where LAMP-1 serves as a scaffold for both Siah-1 and p10 that allows the E3 ligase targeting p10 for ubiquitylation and degradation to suppress ARV infection.IMPORTANCE Avian reovirus (ARV) is an important poultry pathogen causing viral arthritis, chronic respiratory diseases, retarded growth, and malabsorption syndrome, leading to considerable economic losses to the poultry industry across the globe. The ARV p10 protein is a virulence factor responsible for the induction of cell syncytium formation and apoptosis and is rapidly degraded in host cells. We previously found that cellular lysosome-associated membrane protein 1 (LAMP-1) interacts with p10 and is involved in its degradation. Here we report that the E3 ubiquitin ligase seven in absentia homolog 1 (Siah-1) ubiquitylated p10 and targeted it for proteasomal degradation. Furthermore, the ubiquitylation of p10 by Siah-1 required the participation of LAMP-1 by forming a multicomponent complex. Thus, LAMP-1 serves as an adaptor to allow Siah-1 to target p10 for degradation, thereby suppressing ARV growth in host cells.


Asunto(s)
Proteínas Aviares/metabolismo , Fibroblastos/enzimología , Proteínas Nucleares/metabolismo , Orthoreovirus Aviar/metabolismo , Proteolisis , Infecciones por Reoviridae/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Animales , Proteínas Aviares/genética , Línea Celular Transformada , Embrión de Pollo , Fibroblastos/patología , Fibroblastos/virología , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Proteínas Nucleares/genética , Orthoreovirus Aviar/genética , Infecciones por Reoviridae/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética , Proteínas Virales/genética
3.
Arch Virol ; 161(4): 899-911, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26744063

RESUMEN

Avian reovirus (ARV) causes viral arthritis, chronic respiratory diseases, retarded growth and malabsorption syndrome. The ARV p10 protein, a viroporin responsible for the induction of cell syncytium formation and apoptosis, is rapidly degraded in host cells. However, the mechanism of p10 degradation and its relevance are still unclear. We report here the identification of cellular lysosome-associated membrane protein 1 (LAMP-1) as an interaction partner of p10 by yeast two-hybrid screening, immunoprecipitation and confocal microscopy assays. We found that rapid degradation of p10 was associated with ubiquitination. Importantly, ARV p10 degradation in host cells could be completely abolished by knockdown of LAMP-1 by siRNA, indicating that LAMP-1 is required for ARV p10 degradation in host cells. In contrast, overexpression of LAMP-1 facilitated p10 degradation. Furthermore, knockdown of LAMP-1 allowed p10 accumulation, enhancing p10-induced apoptosis and viral release. Thus, LAMP-1 plays a critical role in ARV p10 degradation associated with inhibition of apoptosis and viral release.


Asunto(s)
Apoptosis/fisiología , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Orthoreovirus Aviar/metabolismo , Proteínas Virales/metabolismo , Liberación del Virus/fisiología , Animales , Línea Celular , Embrión de Pollo , Cricetinae , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes , Humanos , Orthoreovirus Aviar/genética , Transporte de Proteínas , Proteínas Virales/genética
4.
Arch Virol ; 161(8): 2243-8, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27233800

RESUMEN

The present study was conducted to identify avian reovirus (ARV) proteins that can activate the phosphatidylinositol 3-kinase (PI3K)-dependent Akt pathway. Based on ARV protein amino acid sequence analysis, σA, σNS, µA, µB and µNS were identified as putative proteins capable of mediating PI3K/Akt pathway activation. The recombinant plasmids σA-pcAGEN, σNS-pcAGEN, µA-pcAGEN, µB-pcAGEN and µNS-pcAGEN were constructed and used to transfect Vero cells, and the expression levels of the corresponding genes were quantified by immunofluorescence and Western blot analysis. Phosphorylated Akt (P-Akt) levels in the transfected cells were measured by flow cytometry and Western blot analysis. The results showed that the σA, σNS, µA, µB and µNS genes were expressed in Vero cells. σA-expressing and σNS-expressing cells had higher P-Akt levels than negative control cells, pcAGEN-expressing cells and cells designed to express other proteins (i.e., µA, µB and µNS). Pre-treatment with the PI3K inhibitor LY294002 inhibited Akt phosphorylation in σA- and σNS-expressing cells. These results indicate that the σA and σNS proteins can activate the PI3K/Akt pathway.


Asunto(s)
Enfermedades de las Aves/enzimología , Enfermedades de las Aves/virología , Orthoreovirus Aviar/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/metabolismo , Infecciones por Reoviridae/enzimología , Infecciones por Reoviridae/veterinaria , Proteínas del Núcleo Viral/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Enfermedades de las Aves/genética , Chlorocebus aethiops , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Orthoreovirus Aviar/genética , Fosfatidilinositol 3-Quinasa/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas de Unión al ARN/genética , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/virología , Transducción de Señal , Células Vero , Proteínas del Núcleo Viral/genética , Proteínas Reguladoras y Accesorias Virales/genética
5.
J Biol Chem ; 288(5): 3571-84, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23233667

RESUMEN

Autophagy has been shown to facilitate replication or production of avian reovirus (ARV); nevertheless, how ARV induces autophagy remains largely unknown. Here, we demonstrate that the nonstructural protein p17 of ARV functions as an activator of autophagy. ARV-infected or p17-transfected cells present a fast and strong induction of autophagy, resulting in an increased level of autophagic proteins Beclin 1 and LC3-II. Although autophagy was suppressed by 3-methyladenine or shRNAs targeting autophagic proteins (Beclin 1, ATG7, and LC3) as well as by overexpression of Bcl-2, viral transcription, σC protein synthesis, and virus yield were all significantly reduced, suggesting a key role of autophagosomes in supporting ARV replication. Furthermore, we revealed for the first time that p17 positively regulates phosphatase and tensin deleted on chromosome 10 (PTEN), AMP-activated protein kinase (AMPK), and dsRNA dependent protein kinase RNA (PKR)/eIF2α signaling pathways, accompanied by down-regulation of Akt and mammalian target of rapamycin complex 1, thereby triggering autophagy. By using p53, PTEN, PKR, AMPK, and p17 short hairpin RNA (shRNA), activation of signaling pathways and LC3-II levels was significantly suppressed, suggesting that p17 triggers autophagy through activation of p53/PTEN, AMPK, and PKR signaling pathways. Furthermore, colocalization of LC3 with viral proteins (p17 and σC), p62 with LAMP2 and LC3 with Rab7 was observed under a fluorescence microscope. The expression level of p62 was increased at 18 h postinfection and then slightly decreased 24 h postinfection compared with mock infection and thapsigargin treatment. Furthermore, disruption of autophagosome-lysosome fusion by shRNAs targeting LAMP2 or Rab7a resulted in inhibition of viral protein synthesis and virus yield, suggesting that formation of autolysosome benefits virus replication. Taken together, our results suggest that ARV induces formation of autolysosome but does not induce complete autophagic flux.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Orthoreovirus Aviar/metabolismo , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Pollos , Activación Enzimática , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Orthoreovirus Aviar/crecimiento & desarrollo , Orthoreovirus Aviar/fisiología , Fosfohidrolasa PTEN/genética , Fagosomas/metabolismo , ARN Bicatenario/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/metabolismo , eIF-2 Quinasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Avian Dis ; 57(2): 220-4, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24689177

RESUMEN

Mycoplasma synoviae and avian reovirus (ARV) are associated with several disease syndromes in poultry and cause notable global economic losses in the poultry industry. Rapid and efficient diagnostics for these avian pathogens are important not only for disease control but also for prevention of clinical disease progression. However, current diagnostic methods used for surveillance of these diseases in poultry flocks are laborious and time-consuming, and they have low sensitivity. The multiplex PCR (mPCR) developed in this study has been proven to be both sensitive and specific for simultaneous M. synoviae and ARV detection and identification in clinical samples. To evaluate the mPCR assay, the diagnostic test was applied to different clinical samples from natural and experimental M. synoviae and ARV-infected poultry. Results were compared with serologic, single PCR, and immunofluorescence analyses. Tibiotarsal articulation could be the best target for simultaneous detection of M. synoviae and ARV infection. The detection limit by visualization of mPCR-amplified products was 100 pg for both pathogens. Overall, the mPCR developed and standardized in this research is a useful tool for diagnosis and screening and for surveillance and control of M. synoviae and ARV infection in poultry flocks.


Asunto(s)
Pollos , Reacción en Cadena de la Polimerasa Multiplex/métodos , Infecciones por Mycoplasma/veterinaria , Mycoplasma synoviae/genética , Orthoreovirus Aviar/genética , Enfermedades de las Aves de Corral/diagnóstico , Infecciones por Reoviridae/diagnóstico , Animales , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente/veterinaria , Reacción en Cadena de la Polimerasa Multiplex/veterinaria , Infecciones por Mycoplasma/diagnóstico , Infecciones por Mycoplasma/microbiología , Mycoplasma synoviae/aislamiento & purificación , Mycoplasma synoviae/metabolismo , Orthoreovirus Aviar/aislamiento & purificación , Orthoreovirus Aviar/metabolismo , Enfermedades de las Aves de Corral/microbiología , Infecciones por Reoviridae/microbiología , Sensibilidad y Especificidad
7.
Acta Virol ; 57(1): 69-74, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23530826

RESUMEN

This study investigated the anti-neoplastic potential of avian reovirus σC (sigma C) protein on Rous sarcoma virus-induced fibrosarcoma in chicken. The recombinant vector expressing σC protein was injected intra-tumorally into specific pathogen free chicken with fibro-sarcoma at the dose 100µg per bird, while control birds were mock-treated with 100µg of empty vector per bird. Recombinant σC protein induced apoptosis in tumors of treated birds resulting in progressive tumor regression, while similar changes were absent in tumors of mock-treated controls. The σC protein-induced apoptosis in tumors was quantified by flow cytometry and the mean level of apoptosis up to 66% was observed in treated tumors, whereas any significant level of apoptosis was absent in mock-treated controls.


Asunto(s)
Antineoplásicos/administración & dosificación , Proteínas de la Cápside/administración & dosificación , Pollos , Orthoreovirus Aviar/genética , Virus del Sarcoma de Rous/fisiología , Sarcoma Aviar/terapia , Animales , Apoptosis , Proteínas de la Cápside/genética , Embrión de Pollo , Femenino , Fibrosarcoma/terapia , Fibrosarcoma/virología , Terapia Genética , Vectores Genéticos , Humanos , Orthoreovirus Aviar/metabolismo , Proteínas Recombinantes , Sarcoma Aviar/virología , Organismos Libres de Patógenos Específicos
8.
Microbiol Spectr ; 11(3): e0000923, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37097149

RESUMEN

The specifics of cell receptor-modulated avian reovirus (ARV) entry remain unknown. By using a viral overlay protein-binding assay (VOPBA) and an in-gel digestion coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS), we determined that cell-surface annexin A2 (AnxA2) and adhesion G protein-coupled receptor Latrophilin-2 (ADGRL2) modulate ARV entry. Direct interaction between the ARV σC protein and AnxA2 and ADGRL2 in Vero and DF-1 cells was demonstrated in situ by proximity ligation assays. By using short hairpin RNAs (shRNAs) to silence the endogenous AnxA2 and ADGRL2 genes, ARV entry could be efficiently blocked. A significant decrease in virus yields and the intracellular specific signal for σC protein was observed in Vero cells preincubated with the specific AnxA2 and ADGRL2 monoclonal antibodies, indicating that AnxA2 and ADGRL2 are involved in modulating ARV entry. Furthermore, we found that cells pretreated with the AnxA2/S100A10 heterotetramer (A2t) inhibitor A2ti-1 suppressed ARV-mediated activation of Src and p38 mitogen-activated protein kinase (MAPK), demonstrating that Src and p38 MAPK serve as downstream molecules of cell-surface AnxA2 signaling. Our results reveal that suppression of cell-surface AnxA2 with the A2ti-1 inhibitor increased Csk-Cbp interaction, suggesting that ARV entry suppresses Cbp-mediated relocation of Csk to the membrane, thereby activating Src. Furthermore, reciprocal coimmunoprecipitation assays revealed that σC can interact with signaling molecules, lipid raft, and vimentin. The current study provides novel insights into cell-surface AnxA2- and ADGRL2-modulated cell entry of ARV which triggers Src and p38 MAPK signaling to enhance caveolin-1-, dynamin 2-, and lipid raft-dependent endocytosis. IMPORTANCE By analyzing results from VOPBA and LC-MS/MS, we have determined that cell-surface AnxA2 and ADGRL2 modulate ARV entry. After ARV binding to receptors, Src and p38 MAPK signaling were triggered and, in turn, increased the phosphorylation of caveolin-1 (Tyr14) and upregulated dynamin 2 expression to facilitate caveolin-1-mediated and dynamin 2-dependent endocytosis. In this work, we demonstrated that ARV triggers Src activation by impeding Cbp-mediated relocation of Csk to the membrane in the early stages of the life cycle. This work provides better insight into cell-surface AnxA2 and ADGRL2, which upregulate Src and p38MAPK signaling pathways to enhance ARV entry and productive infection.


Asunto(s)
Anexina A2 , Orthoreovirus Aviar , Animales , Chlorocebus aethiops , Caveolina 1/genética , Caveolina 1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Vero , Orthoreovirus Aviar/metabolismo , Internalización del Virus , Anexina A2/genética , Anexina A2/metabolismo , Dinamina II/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem , Endocitosis , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo
9.
J Biol Chem ; 286(35): 30780-30794, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21705803

RESUMEN

Very little is known about the mechanism of cell entry of avian reovirus (ARV). The aim of this study was to explore the mechanism of ARV entry and subsequent infection. Cholesterol mainly affected the early steps of the ARV life cycle, because the presence of cholesterol before and during viral adsorption greatly blocked ARV infectivity. Although we have demonstrated that ARV facilitating p38 MAPK is beneficial for virus replication, its mechanism remains unknown. Here, we show that ARV-induced phosphorylation of caveolin-1 (Tyr(14)), dynamin-2 expression, and Rac1 activation through activation of p38 MAPK and Src in the early stage of the virus life cycle is beneficial for virus entry and productive infection. The strong inhibition by dynasore, a specific inhibitor of dynamin-2, and depletion of endogenous caveolin-1 or dynamin-2 by siRNAs as well as the caveolin-1 colocalization study implicate caveolin-1-mediated and dynamin-2-dependent endocytosis as a significant avenue of ARV entry. By means of pharmacological inhibitors, dominant negative mutants, and siRNA of various cellular proteins and signaling molecules, phosphorylation of caveolin-1, dynamin-2 expression, and Rac1 activation were suppressed, suggesting that by orchestrating p38 MAPK, Src, and Rac1 signaling cascade in the target cells, ARV creates an appropriate intracellular environment facilitating virus entry and productive infection. Furthermore, disruption of microtubules, Rab5, or endosome acidification all inhibited ARV infection, suggesting that microtubules and small GTPase Rab5, which regulate transport to early endosome, are crucial for survival of ARV and that exposure of the virus to acidic pH is required for productive infection.


Asunto(s)
Caveolina 1/metabolismo , Dinamina II/metabolismo , Microtúbulos/metabolismo , Proteínas de Unión al GTP Monoméricas/química , Orthoreovirus Aviar/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Colesterol/metabolismo , Endocitosis , Activación Enzimática , Regulación de la Expresión Génica , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , ARN Interferente Pequeño/metabolismo , Células Vero
10.
Methods Mol Biol ; 2465: 27-40, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35118614

RESUMEN

Particulate material is more efficient in eliciting immune responses. Here we describe the production of micro- and nanospheres formed by protein muNS-Mi from avian reoviruses, loaded with foreign epitopes for their use as vaccines.


Asunto(s)
Orthoreovirus Aviar , Vacunas , Adyuvantes Inmunológicos , Epítopos/metabolismo , Orthoreovirus Aviar/metabolismo , Vacunas/metabolismo , Vacunas de Subunidad , Proteínas no Estructurales Virales/metabolismo
11.
Cells ; 10(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525607

RESUMEN

Avian reovirus p17 (ARV p17) is a non-structural protein known to activate autophagy, interfere with gene transcription and induce a significant tumor cell growth inhibition in vitro and in vivo. In this study, we show that ARV p17 is capable of exerting potent antiangiogenic properties. The viral protein significantly inhibited the physiological angiogenesis of human endothelial cells (ECs) by affecting migration, capillary-like structure and new vessel formation. ARV p17 was not only able to suppress the EC physiological angiogenesis but also rendered ECs insensitive to two different potent proangiogenic inducers, such as VEGF-A and FGF-2 in the three-dimensional (3D) Matrigel and spheroid assay. ARV p17 was found to exert its antiangiogenic activity by upregulating transcription and release of the well-known tumor suppressor molecule dipeptidyl peptidase 4 (DPP4). The ability of ARV p17 to impact on angiogenesis is completely new and highlights the "two compartments" activity of the viral protein that is expected to hamper the tumor parenchymal/stromal crosstalk. The complex antitumor activities of ARV p17 open the way to a new promising field of research aimed to develop new therapeutic approaches for treating tumor and cancer metastasis.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Neovascularización Patológica/metabolismo , Orthoreovirus Aviar/metabolismo , Proteínas no Estructurales Virales/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Pollos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microvasos/citología , Morfogénesis/efectos de los fármacos , Proteínas Recombinantes/farmacología , Transducción Genética , Regulación hacia Arriba/efectos de los fármacos
12.
J Virol ; 83(19): 10163-75, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19640987

RESUMEN

Avian reovirus sigmaA is a double-stranded RNA (dsRNA)-binding protein that has been shown to stabilize viral core particles and to protect the virus against the antiviral action of interferon. To continue with the characterization of this viral protein, we have investigated its intracellular distribution in avian cells. Most sigmaA accumulates into cytoplasmic viral factories of infected cells, and yet a significant fraction was detected in the nucleolus. The protein also localizes in the nucleolus of transfected cells, suggesting that nucleolar targeting is not facilitated by the viral infection or by viral factors. Assays performed in both intact cells and digitonin-permeabilized cells demonstrate that sigmaA is able to enter the nucleus via a nucleoporin-dependent nondiffusional mechanism that does not require added cytosolic factors or energy input. These results indicate that sigmaA by itself is able to penetrate into the nucleus using a process that is mechanistically different from the classical nuclear localization signal/importin pathway. On the other hand, two sigmaA arginines that are necessary for dsRNA binding are also required for nucleolar localization, suggesting that dsRNA-binding and nucleolar targeting are intimately linked properties of the viral protein.


Asunto(s)
Nucléolo Celular/metabolismo , Orthoreovirus Aviar/metabolismo , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Proteínas del Núcleo Viral/metabolismo , Proteínas del Núcleo Viral/fisiología , Animales , Línea Celular , Embrión de Pollo , Citoplasma/metabolismo , Citosol/metabolismo , Digitonina/farmacología , Carioferinas/metabolismo , Microscopía Fluorescente/métodos , Señales de Localización Nuclear/metabolismo , Proteínas Recombinantes/química , Fracciones Subcelulares/metabolismo
13.
Biochem Biophys Res Commun ; 384(3): 301-5, 2009 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-19406104

RESUMEN

Viral infection usually influences cellular protein synthesis either actively or passively via modification of various translation initiation factors. Here we demonstrated that infection with avian reovirus (ARV) interfered with cellular protein synthesis. This study demonstrated for the first time that ARV influenced the phosphorylation of translation initiation factors including eIF4E and eIF-4G. Interestingly, ARV also induced phosphorylation of eukaryotic translation elongation factor (eEF2) in a time- and dose-dependent manner. Inhibition of mTOR by rapamycin notably increased the level of phosphorylated eEF2 in infected cells. However, rapamycin did not show any negative effects on ARV replication, suggesting that phosphorylation of eEF2 in infected cells did not reduce ARV propagation. These results demonstrated for the first time that ARV promotes phosphorylation of eEF2 which in turn influenced host protein production not simply by modulating the function of translation initiation factors but also by regulating elongation factor eEF2.


Asunto(s)
Orthoreovirus Aviar/metabolismo , Factor 2 de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Infecciones por Reoviridae/metabolismo , Animales , Chlorocebus aethiops , Fosforilación , Células Vero
14.
J Proteomics ; 193: 162-172, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30339941

RESUMEN

Duck reovirus (DRV), a member of the genus Orthoreovirus in the family Reoviridae, was first isolated from Muscovy ducks. The disease associated with DRV causes great economic losses to the duck industry. However, the responses of duck (Cairna moschata) to the classical/novel DRV (C/NDRV) infections are largely unknown. To reveal the relationship of pathogenesis and immune response, the proteomes of duck spleen cells under the control and C/NDRV infections were compared. In total, 5986 proteins were identified, of which 5389 proteins were quantified. The different accumulated proteins (DAPs) under the C/NDRV infections showed displayed various biological functions and diverse subcellular localizations. The proteins related to the serine protease system were siginificantly changed, suggesting that the activated serine protease system may play an important role under the C/NDRV infections. Furthermore, the differences in the responses to the C/NRDV infections between the duck liver and spleen tissues were compared. Only a small number of common DAPs were identified in both liver and spleen tissues, suggesting diversified pattern involved in the responses to the C/NRDV infections. However, the changes in the proteins involved in the serine protease systems were similar in both liver and spleen cells. Our data may give a comprehensive resource for investigating the responses to C/NDRV infections in ducks. SIGNIFICANCE: A newly developed MS/MS-based method involving isotopomer labels and 'tandem mass' has been applied to protein accurate quantification in current years. However, no studies on the responses of duck (Cairna moschata) spleen tissue to the classical/novel DRV (C/NDRV) infections have been performed. As a continued study of our previous report on the responses of duck liver tissue to the C/NDRV infections, the current study further compared the differences in the responses to the C/NRDV infections between the duck liver and spleen tissues. Our results will provide an opportunity to reveal the relationship of pathogenesis and immune response and basic information on the pathogenicity of C/NDRV in ducks.


Asunto(s)
Anseriformes , Proteínas Aviares/metabolismo , Orthoreovirus Aviar/metabolismo , Infecciones por Reoviridae , Bazo , Animales , Anseriformes/metabolismo , Anseriformes/virología , Proteómica , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/veterinaria , Bazo/metabolismo , Bazo/virología
15.
Mater Sci Eng C Mater Biol Appl ; 105: 110052, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31546360

RESUMEN

Novel duck reovirus (NDRV) disease is a serious infectious disease for poultry, for which no effective therapy has been established. Therefore, development of novel antivirals against NDRV is urgently needed. In present study, we developed a complex wherein hypericin (HY), which shows broad-spectrum antiviral activity, was loaded onto graphene oxide (GO), which has a high drug-loading capacity and low cytotoxicity. The antiviral activity of the complex (GO/HY) was studied in DF-1 cells and in ducklings infected with the NDRV TH11 strain. GO/HY showed a dose-dependent inhibition of NDRV replication, which may be attributed to direct virus inactivation or inhibition of virus attachment. Western blotting and indirect immunofluorescence assay (IFA) showed markedly suppressed protein expression in GO/HY-treated NDRV-infected DF-1 cells. Moreover, GO/HY prolonged the survival time of the ducklings by reducing pathological lesions caused by the infection and inhibiting viral replication in the liver and lungs. These results suggest that GO/HY has antiviral activity against NDRV both in vitro and in vivo.


Asunto(s)
Portadores de Fármacos , Patos , Grafito , Orthoreovirus Aviar/metabolismo , Perileno/análogos & derivados , Enfermedades de las Aves de Corral , Infecciones por Reoviridae , Animales , Antracenos , Línea Celular , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Patos/metabolismo , Patos/virología , Grafito/química , Grafito/farmacología , Perileno/química , Perileno/farmacología , Enfermedades de las Aves de Corral/tratamiento farmacológico , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/patología , Enfermedades de las Aves de Corral/virología , Infecciones por Reoviridae/tratamiento farmacológico , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/patología , Infecciones por Reoviridae/veterinaria
16.
Mol Cells ; 26(4): 396-403, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18612239

RESUMEN

The first ORF of the ARV S1133 S1 segment encodes the nonstructural protein p10, which is responsible for the induction of cell syncytium formation. However, p10-dependent signaling during syncytium formation is fully unknown. Here, we show that dominant negative RhoA, Rho inhibitor C3 exoenzyme, ROCK/Rho-kinase inhibitor Y-27632 and Rac1 inhibitor NSC23766 inhibit p10-mediated cell fusion. p10 over-expression is concomitant with activation and membrane translocation of RhoA and Rac1, but not cdc42. RhoA and Rac1 downstream events, including JNK phosphorylation and transcription factor AP-1 and NF-kappaB activation, as well as MLC expression and phosphorylation are simultaneously activated by p10. p10 point mutant T13M possessed 20% fusion-inducing ability and four p10 fusion-deficient mutants V15M, V19M, C21S and L32A reduced or lost their ability to activate RhoA and Rac1 signaling. We conclude that p10-mediated syncytium formation proceeds by utilizing RhoA and Rac1-dependent signaling.


Asunto(s)
Células Gigantes/metabolismo , Orthoreovirus Aviar/metabolismo , Proteínas Virales/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Gigantes/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , FN-kappa B/metabolismo , Mutación Puntual/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Células Vero , Proteínas Virales/química , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
17.
Vet Microbiol ; 129(3-4): 252-61, 2008 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-18191923

RESUMEN

RNA interference was used to suppress protein expression of three S-class genome segments of avian reovirus (ARV). Viral progeny titer was successfully down-regulated by RNA interference. Suppression of S1 genome segment, which has three open reading frames, not only decreased the expression level of the structural protein sigmaC but also reduced cell fusion and the level of Ser(15)-phosphorylated p53 protein caused by the nonstructural proteins p10 and p17, respectively. Suppression of S2 or S4 genome segment by RNA interference could also reduce the expression level of sigmaA or sigmaNS. Interestingly, suppression of sigmaNS resulted in down regulation of the expression of other viral products. In terms of variability of different genes among viral strains and of the impact after their suppression, it seems that the viral products involved in construction of viroplasm or core particles, like sigmaNS, are considerable choices to efficiently inhibit ARV multiplication by RNA interference. Using a GFP reporter system, it was discovered that ARV could not inhibit activated RNA interference, suggesting that RNA interference may be used in the suppression of ARV infection.


Asunto(s)
Regulación Viral de la Expresión Génica , Orthoreovirus Aviar/genética , Interferencia de ARN , Proteínas Estructurales Virales/genética , Animales , Aves/virología , Chlorocebus aethiops , Regulación hacia Abajo , Genes Virales , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Orthoreovirus Aviar/metabolismo , Plásmidos , Transfección/veterinaria , Células Vero , Proteínas Virales/genética , Proteínas Estructurales Virales/biosíntesis
18.
PLoS One ; 11(9): e0162356, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27603133

RESUMEN

The p17 protein of avian reovirus (ARV) causes cell cycle retardation in a variety of cell lines; however, the underlying mechanism(s) by which p17 regulates the cell cycle remains largely unknown. We demonstrate for the first time that p17 interacts with CDK1 and vimentin as revealed by reciprocal co-immunoprecipitation and GST pull-down assays. Both in vitro and in vivo studies indicated that direct interaction of p17 and CDK1/vimentin was mapped within the amino terminus (aa 1-60) of p17 and central region (aa 27-118) of CDK1/vimentin. Furthermore, p17 was found to occupy the Plk1-binding site within the vimentin, thereby blocking Plk1 recruitment to CDK1-induced vimentin phosphorylation at Ser 56. Interaction of p17 to CDK1 or vimentin interferes with CDK1-catalyzed phosphorylation of vimentin at Ser 56 and subsequently vimentin phosphorylation at Ser 82 by Plk1. Furthermore, we have identified upstream signaling pathways and cellular factor(s) targeted by p17 and found that p17 regulates inhibitory phosphorylation of CDK1 and blocks vimentin phosphorylation at Ser 56 and Ser 82. The p17-mediated inactivation of CDK1 is dependent on several mechanisms, which include direct interaction with CDK1, p17-mediated suppression of Plk1 by activating the Tpr/p53 and ATM/Chk1/PP2A pathways, and p17-mediated cdc25C degradation via an ubiquitin- proteasome pathway. Additionally, depletion of p53 with a shRNA as well as inhibition of ATM and vimentin by inhibitors diminished virus yield while Tpr and CDK1 knockdown increased virus yield. Taken together, results demonstrate that p17 suppresses both CDK1 and Plk1functions, disrupts vimentin phosphorylation, causes G2/M cell cycle arrest and thus benefits virus replication.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular , Fase G2 , Orthoreovirus Aviar/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Vimentina/metabolismo , Proteínas Virales/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proliferación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Embrión de Pollo , Chlorocebus aethiops , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Inmunoprecipitación , Modelos Biológicos , Proteínas de Complejo Poro Nuclear/metabolismo , Fosforilación , Fosfoserina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Transducción de Señal , Transfección , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Regulación hacia Arriba , Células Vero , Proteínas Virales/química , Replicación Viral , Fosfatasas cdc25/metabolismo , Quinasa Tipo Polo 1
19.
J Mol Biol ; 341(2): 361-74, 2004 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-15276829

RESUMEN

We have recently shown that the avian reovirus non-structural protein microNS forms cytoplasmic inclusions in transfected cells and recruits sigmaNS to these structures. In the present study we further demonstrate that microNS mediates the association of the major core protein lambdaA, but not of sigmaA or sigmaC, with inclusions, indicating that the recruitment of viral proteins into avian reovirus factories has specificity. Thus, some proteins appear to be initially recruited to factories by association with microNS, whereas others are recruited subsequently through interaction with as-yet-unknown factors. We next used metabolic pulse-chase radiolabeling combined with cell fractionation and antibody immunoprecipitation to study the recruitment of newly synthesized viral polypeptides into viral factories and virus particles. The results of this combined approach revealed that avian reovirus morphogenesis is a complex and temporally controlled process that takes place exclusively within globular viral factories that are not microtubule-associated. Our findings further suggest that cores are assembled within the first 30 minutes after the synthesis of their polypeptide components, and that reovirion morphogenesis is completed over the next 30 minutes by the subsequent addition of outer capsid proteins.


Asunto(s)
Embrión de Pollo/virología , Cuerpos de Inclusión Viral/química , Orthoreovirus Aviar/metabolismo , Proteínas del Núcleo Viral/análisis , Proteínas Virales/análisis , Animales , Células Cultivadas , Cuerpos de Inclusión Viral/metabolismo , Datos de Secuencia Molecular , Morfogénesis , Orthoreovirus Aviar/genética , Orthoreovirus Aviar/crecimiento & desarrollo , Pruebas de Precipitina , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/metabolismo , Siphoviridae , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales
20.
Artículo en Inglés | MEDLINE | ID: mdl-16511119

RESUMEN

Avian reovirus fibre, a homotrimer of the sigmaC protein, is responsible for primary host-cell attachment. Using the protease trypsin, a C-terminal sigmaC fragment containing amino acids 156-326 has been generated which was subsequently purified and crystallized. Two different crystal forms were obtained, one grown in the absence of divalent cations and belonging to space group P6(3)22 (unit-cell parameters a = 75.6, c = 243.1 A) and one grown in the presence of either zinc or cadmium sulfate and belonging to space group P321 (unit-cell parameters a = 74.7, c = 74.5 A and a = 73.1, c = 69.9 A for the Zn(II)- and Cd(II)-grown crystals, respectively). The first crystal form diffracted synchrotron radiation to 3.0 A resolution and the second form to 2.2-2.3 A. Its closest related structure, the C-terminal fragment of mammalian reovirus fibre, has only 18% sequence identity and molecular-replacement attempts were unsuccessful. Therefore, a search is under way for suitable heavy-atom derivatives and attempts are being made to grow protein crystals containing selenomethionine instead of methionine.


Asunto(s)
Cápside/química , Orthoreovirus Aviar/metabolismo , Compuestos de Cadmio/química , Cationes , Cristalización , Cristalografía por Rayos X , ADN/química , Escherichia coli/metabolismo , Metionina/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Selenometionina/química , Sulfatos/química , Sulfato de Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA