Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
J Virol ; 97(5): e0058523, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37167564

RESUMEN

Structural metastability of viral capsids is pivotal for viruses to survive in harsh environments and to undergo timely conformational changes required for cell entry. Mammalian orthoreovirus (reovirus) is a model to study capsid metastability. Following initial disassembly of the reovirus particle mediated by proteases, a metastable intermediate called the infectious subvirion particle (ISVP) is generated. Using a σ1 monoreassortant virus, we recently showed that σ1 properties affect its encapsidation on particles and the metastability of ISVPs. How metastability is impacted by σ1 and whether the lower encapsidation level of σ1 is connected to this property is unknown. To define a correlation between encapsidation of σ1 and ISVP stability, we generated mutant viruses with single amino acid polymorphisms in σ1 or those that contain chimeric σ1 molecules composed of σ1 portions from type 1 and type 3 reovirus strains. We found that under most conditions where σ1 encapsidation on the particle was lower, ISVPs displayed lower stability. Characterization of mutant viruses selected for enhanced stability via a forward genetic approach also revealed that in some cases, σ1 properties influence stability without influencing σ1 encapsidation. These data indicate that σ1 can also influence ISVP stability independent of its level of incorporation. Together, our work reveals an underappreciated effect of the σ1 attachment protein on the properties of the reovirus capsid. IMPORTANCE Reovirus particles are comprised of eight proteins. Among them, the reovirus σ1 protein functions engages cellular receptors. σ1 also influences the stability of an entry intermediate called ISVP. Here, we sought to define the basis of the link between σ1 properties and stability of ISVPs. Using variety of mutant strains, we determined that when virus preparations contain particles with a high amount of encapsidated σ1, ISVP stability is higher. Additionally, we identified portions of σ1 that impact its encapsidation and consequently the stability of ISVPs. We also determined that in some cases, σ1 properties alter stability of ISVPs without affecting encapsidation. This work highlights that proteins of these complex particles are arranged in an intricate, interconnected manner such that changing the properties of these proteins has a profound impact on the remainder of the particle.


Asunto(s)
Orthoreovirus Mamífero 3 , Orthoreovirus de los Mamíferos , Internalización del Virus , Cápside/metabolismo , Línea Celular , Orthoreovirus de los Mamíferos/fisiología , Orthoreovirus Mamífero 3/fisiología
2.
J Virol ; 97(2): e0000923, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36744961

RESUMEN

Mammalian orthoreovirus serotype 3 Dearing is an oncolytic virus currently undergoing multiple clinical trials as a potential cancer therapy. Previous clinical trials have emphasized the importance of prescreening patients for prognostic markers to improve therapeutic success. However, only generic cancer markers such as epidermal growth factor receptor (EGFR), Hras, Kras, Nras, Braf, and p53 are currently utilized, with limited benefit in predicting therapeutic efficacy. This study aimed to investigate the role of p38 mitogen-activated protein kinase (MAPK) signaling during reovirus infection. Using a panel of specific p38 MAPK inhibitors and an inactive inhibitor analogue, p38 MAPK signaling was found to be essential for establishment of reovirus infection by enhancing reovirus endocytosis, facilitating efficient reovirus uncoating at the endo-lysosomal stage, and augmenting postuncoating replication steps. Using a broad panel of human breast cancer cell lines, susceptibility to reovirus infection corresponded with virus binding and uncoating efficiency, which strongly correlated with status of the p38ß isoform. Together, results suggest p38ß isoform as a potential prognostic marker for early stages of reovirus infection that are crucial to successful reovirus infection. IMPORTANCE The use of Pelareorep (mammalian orthoreovirus) as a therapy for metastatic breast cancer has shown promising results in recent clinical trials. However, the selection of prognostic markers to stratify patients has had limited success due to the fact that these markers are upstream receptors and signaling pathways that are present in a high percentage of cancers. This study demonstrates that the mechanism of action of p38 MAPK signaling plays a key role in establishment of reovirus infection at both early entry and late replication steps. Using a panel of breast cancer cell lines, we found that the expression levels of the MAPK11 (p38ß) isoform are a strong determinant of reovirus uncoating and infection establishment. Our findings suggest that selecting prognostic markers that target key steps in reovirus replication may improve patient stratification during oncolytic reovirus therapy.


Asunto(s)
Neoplasias de la Mama , Orthoreovirus Mamífero 3 , Infecciones por Reoviridae , Internalización del Virus , Proteínas Quinasas p38 Activadas por Mitógenos , Femenino , Humanos , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Orthoreovirus Mamífero 3/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Replicación Viral , Línea Celular Tumoral
3.
Environ Microbiol ; 23(1): 431-447, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33201573

RESUMEN

Gut microbiota could facilitate host to defense diseases, but fish-microbiota interactions during viral infection and the underlying mechanism are poorly understood. We examined interactions and responses of gut microbiota to grass carp reovirus (GCRV) infection in Ctenopharyngodon idellus, which is the most important aquaculture fish worldwide. We found that GCRV infection group with serious haemorrhagic symptoms (G7s) showed considerably different gut microbiota, especially with an abnormally high abundance of gram-negative anaerobic Cetobacterium somerae. It also showed the lowest (p < 0.05) alpha-diversity but with much higher ecological process of homogenizing dispersal (28.8%), confirming a dysbiosis of the gut microbiota after viral infection. Interestingly, signaling pathways of NOD-like receptors (NLRs), toll-like receptors (TLRs), and lipopolysaccharide (LPS) stimulation genes were significantly (q-value < 0.01) enriched in G7s, which also significantly (p < 0.01) correlated with the core gut microbial genera of Cetobacterium and Acinetobacter. The results suggested that an expansion of C. somerae initiated by GCRV could aggravate host inflammatory reactions through the LPS-related NLRs and TLRs pathways. This study advances our understanding of the interplay between fish immunity and gut microbiota challenged by viruses; it also sheds new insights for ecological defense of fish diseases with the help of gut microbiota.


Asunto(s)
Carpas/microbiología , Carpas/virología , Enfermedades de los Peces/virología , Microbioma Gastrointestinal , Orthoreovirus Mamífero 3/fisiología , Infecciones por Reoviridae/veterinaria , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Enfermedades de los Peces/microbiología , Fusobacterias , Interacciones Huésped-Patógeno , Orthoreovirus Mamífero 3/clasificación , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/aislamiento & purificación , Infecciones por Reoviridae/microbiología , Infecciones por Reoviridae/virología
4.
Nat Commun ; 11(1): 32, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31896744

RESUMEN

Many intracellular pathogens, such as mammalian reovirus, mimic extracellular matrix motifs to specifically interact with the host membrane. Whether and how cell-matrix interactions influence virus particle uptake is unknown, as it is usually studied from the dorsal side. Here we show that the forces exerted at the ventral side of adherent cells during reovirus uptake exceed the binding strength of biotin-neutravidin anchoring viruses to a biofunctionalized substrate. Analysis of virus dissociation kinetics using the Bell model revealed mean forces higher than 30 pN per virus, preferentially applied in the cell periphery where close matrix contacts form. Utilizing 100 nm-sized nanoparticles decorated with integrin adhesion motifs, we demonstrate that the uptake forces scale with the adhesion energy, while actin/myosin inhibitions strongly reduce the uptake frequency, but not uptake kinetics. We hypothesize that particle adhesion and the push by the substrate provide the main driving forces for uptake.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Orthoreovirus Mamífero 3/fisiología , Nanopartículas del Metal/química , Actinas/metabolismo , Animales , Avidina/química , Biotina/química , Cápside/química , Células Cultivadas , Fibroblastos/virología , Oro , Células HeLa , Humanos , Integrinas/metabolismo , Cinética , Orthoreovirus Mamífero 3/química , Orthoreovirus Mamífero 3/patogenicidad , Nanopartículas del Metal/virología , Modelos Teóricos , Miosinas/metabolismo , Ratas , Virión/patogenicidad , Virión/fisiología
5.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(5): 649-653, 2019 Sep.
Artículo en Chino | MEDLINE | ID: mdl-31762232

RESUMEN

OBJECTIVE: To investigate the molecular mechanism of apoptosis of HL60 cells induced by oncolytic virus Reovirus type 3 (Reo3). METHODS: HL60 cells were infected with Reo3 at different multiplicity of infection (MOI) with the uninfected HL60 cells as control group. After 48 h of infection, the activity of HL60 cells infected with virus at different MOI was detected by CCK8 method to investigate the influence of MOI to cell activity. Simultaneously, the apoptotic rate of HL60 cells was detected by flow cytometry, and the activation level of double-stranded RNA-dependent protein kinase (PKR) and the expression of apoptotic-related protein in HL60 cells were detected by Western blot. Before infection with Reo3 for 48 h, HL60 cells were treated with 2-aminopurine (2-AP), a specific inhibitor of PKR, for 24 h. Afterward, the apoptotic level and expression of apoptotic related proteins were detected. RESULTS: Activity of HL60 cells was obviously inhibited after infected with Reo3 with a MOI of 1 for 48 h. The cell survival rate was (24.333±3.396)% and the apoptotic rate was (29.96±2.06)%. Both rates were all higher than those in the control group (P < 0.05). Western blot results showed that the expression levels of PKR, p-PKR, Bax, Caspase3 and cleaved Caspase3 in HL60 cells infected with Reo3 were higher than those in the control group (P < 0.05), while the expression level of Bcl-2 was lower (P < 0.05). Compared with the group without inhibitor, the apoptotic rate of HL60 cells pretreated with 2-AP decreased (P < 0.05), the phosphorylation level of PKR and the expression level of apoptotic-related protein also decreased (P < 0.05). CONCLUSION: Oncolytic virus Reo3 could activate PKR in HL60 cells and thus induce apoptosis of HL60 cells.


Asunto(s)
Apoptosis , Orthoreovirus Mamífero 3/fisiología , eIF-2 Quinasa/metabolismo , 2-Aminopurina/farmacología , Caspasa 3/metabolismo , Citometría de Flujo , Células HL-60 , Humanos , Virus Oncolíticos/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
6.
mBio ; 10(4)2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31387911

RESUMEN

Blood-brain barrier (BBB) breakdown is a hallmark of many diseases of the central nervous system (CNS). Loss of BBB integrity in CNS diseases such as viral encephalitis results in the loss of nutrient/oxygen delivery, rapid infiltration of immune cells, and brain swelling that can exacerbate neuronal injury. Despite this, the cellular and molecular mechanisms that underlie BBB breakdown in viral encephalitis are incompletely understood. We undertook a comprehensive analysis of the cellular and molecular signaling events that induce BBB breakdown in an experimental model of virus-induced encephalitis in which neonatal mice are infected with reovirus (serotype 3 strain Abney). We show that BBB leakage during reovirus infection correlates with morphological changes in the vasculature, reductions in pericytes (BBB supporting cells), and disorganization of vascular junctions. Pathway analysis on RNA sequencing from brain endothelial cells identified the activation of interferon (IFN) signaling within the brain vasculature following reovirus infection. Our in vitro and in vivo studies show that type II IFN mediated by IFN-γ, a well known antiviral signal, is a major contributor to BBB leakage during reovirus infection. We show that IFN-γ reduces barrier properties in cultured brain endothelial cells through Rho kinase (ROCK)-mediated cytoskeletal contractions, resulting in junctional disorganization and cell-cell separations. In vivo neutralization of IFN-γ during reovirus infection significantly improved BBB integrity, pericyte coverage, attenuated vascular ROCK activity, and junctional disorganization. Our work supports a model in which IFN-γ acts directly on the brain endothelium to induce BBB breakdown through a mechanism involving ROCK-induced junctional disorganization.IMPORTANCE In an experimental viral encephalitis mouse model in which mice are infected with reovirus, we show that IFN-γ induces blood-brain barrier leakage. We show that IFN-γ promotes Rho kinase activity, resulting in actin cytoskeletal contractions in the brain endothelium that lead to vascular junctional disorganization and cell-cell separations. These studies now provide insight into a previously unknown mechanism for how blood-brain barrier breakdown occurs in viral encephalitis and implicates IFN-γ-Rho kinase activity as major contributor to this phenomenon. By identifying this mechanism of blood-brain barrier breakdown, we now provide potential therapeutic targets in treating patients with viral causes of encephalitis with the hope of limiting damage to the central nervous system.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalitis Viral/metabolismo , Interferón gamma/metabolismo , Orthoreovirus Mamífero 3/fisiología , Infecciones por Reoviridae/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Barrera Hematoencefálica/virología , Encéfalo/enzimología , Encéfalo/metabolismo , Encéfalo/virología , Modelos Animales de Enfermedad , Encefalitis Viral/genética , Encefalitis Viral/virología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Células Endoteliales/virología , Femenino , Humanos , Interferón gamma/genética , Masculino , Ratones , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/virología , Quinasas Asociadas a rho/genética
7.
J Virol ; 92(20)2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30068646

RESUMEN

Following attachment to host receptors via σ1, reovirus particles are endocytosed and disassembled to generate infectious subvirion particles (ISVPs). ISVPs undergo conformational changes to form ISVP*, releasing σ1 and membrane-targeting peptides from the viral µ1 protein. ISVP* formation is required for delivery of the viral core into the cytoplasm for replication. We characterized the properties of T3DF/T3DCS1, an S1 gene monoreassortant between two laboratory isolates of prototype reovirus strain T3D: T3DF and T3DC T3DF/T3DCS1 is poorly infectious. This deficiency is a consequence of inefficient encapsidation of S1-encoded σ1 on T3DF/T3DCS1 virions. Additionally, compared to T3DF, T3DF/T3DCS1 undergoes ISVP-to-ISVP* conversion more readily, revealing an unexpected role for σ1 in regulating ISVP* formation. The σ1 protein is held within turrets formed by the λ2 protein. To test if the altered properties of T3DF/T3DCS1 are due to a mismatch between σ1 and λ2 proteins from T3DF and T3DC, properties of T3DF/T3DCL2 and T3DF/T3DCS1L2, which express a T3DC-derived λ2, were compared. The presence of T3DC λ2 allowed more efficient σ1 incorporation, producing particles that exhibit T3DF-like infectivity. Compared to T3DF, T3DF/T3DCL2 prematurely converts to ISVP*, uncovering a role for λ2 in regulating ISVP* formation. Importantly, a virus with matching σ1 and λ2 displayed a more regulated conversion to ISVP* than either T3DF/T3DCS1 or T3DF/T3DCL2. In addition to identifying new regulators of ISVP* formation, our results highlight that protein mismatches produced by reassortment can alter virus assembly and thereby influence subsequent functions of the virus capsid.IMPORTANCE Cells coinfected with viruses that possess a multipartite or segmented genome reassort to produce progeny viruses that contain a combination of gene segments from each parent. Reassortment places new pairs of genes together, generating viruses in which mismatched proteins must function together. To test if such forced pairing of proteins that form the virus shell or capsid alters the function of the particle, we investigated properties of reovirus variants in which the σ1 attachment protein and the λ2 protein that anchors σ1 on the particle are mismatched. Our studies demonstrate that a σ1-λ2 mismatch produces particles with lower levels of encapsidated σ1, consequently decreasing virus attachment and infectivity. The mismatch between σ1 and λ2 also altered the capacity of the viral capsid to undergo conformational changes required for cell entry. These studies reveal new functions of reovirus capsid proteins and illuminate both predictable and novel implications of reassortment.


Asunto(s)
Cápside/fisiología , Orthoreovirus Mamífero 3/fisiología , Virus Reordenados/fisiología , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus , Internalización del Virus , Animales , Línea Celular , Endocitosis , Orthoreovirus Mamífero 3/genética , Ratones , Virus Reordenados/genética
8.
Gene Ther ; 25(5): 331-344, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30013187

RESUMEN

While the mammalian orthoreovirus type 3 dearing (reovirus T3D) infects many different tumour cells, various cell lines resist the induction of reovirus-mediated cell death. In an effort to increase the oncolytic potency, we introduced transgenes into the S1 segment of reovirus T3D. The adenovirus E4orf4 gene was selected as transgene since the encoded E4orf4 protein induces cell death in transformed cells. The induction of cell death by E4orf4 depends in part on its binding to phosphatase 2A (PP2A). In addition to the S1-E4orf4 reovirus, two other reoviruses were employed in our studies. The reovirus rS1-RFA encodes an E4orf4 double-mutant protein that cannot interact with PP2A and the rS1-iLOV virus encoding the fluorescent marker iLOV as a reporter. The replacement of the codons for the junction adhesion molecule-A (JAM-A) binding head domain of the truncated spike protein blocks the entry of these recombinant viruses via the reovirus receptor JAM-A. Instead these viruses rely on internalization via binding to sialic acids on the cell surface. This expands their tropism and allows infection of JAM-A-deficient tumour cells. Here we not only demonstrate the feasibility of this approach but also established that the cytolytic activity of these recombinant viruses is largely transgene independent.


Asunto(s)
Orthoreovirus Mamífero 3/fisiología , Proteínas Virales/fisiología , Tropismo Viral/genética , Línea Celular , Humanos , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/metabolismo , Infecciones por Reoviridae/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo
9.
Sci Rep ; 7(1): 17654, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29247249

RESUMEN

The mammalian orthoreovirus Type 3 Dearing has great potential as oncolytic agent in cancer therapy. One of the bottlenecks that hampers its antitumour efficacy in vivo is the limited tumour-cell infection and intratumoural distribution. This necessitates strategies to improve tumour penetration. In this study we employ the baculovirus Autographa californica multiple nucleopolyhedrovirus as a tool to expand the reovirus' tropism and to improve its spread in three-dimensional tumour-cell spheroids. We generated a recombinant baculovirus expressing the cellular receptor for reovirus, the Junction Adhesion Molecule-A, on its envelope. Combining these Junction Adhesion Molecule-A-expressing baculoviruses with reovirus particles leads to the formation of biviral complexes. Exposure of the reovirus-resistant glioblastoma cell line U-118 MG to the baculovirus-reovirus complexes results in efficient reovirus infection, high reovirus yields, and significant reovirus-induced cytopathic effects. As compared to the reovirus-only incubations, the biviral complexes demonstrated improved penetration and increased cell killing of three-dimensional U-118 MG tumour spheroids. Our data demonstrate that reovirus can be delivered with increased efficiency into two- and three-dimensional tumour-cell cultures via coupling the reovirus particles to baculovirus. The identification of baculovirus' capacity to penetrate into tumour tissue opens novel opportunities to improve cancer therapy by improved delivery of oncolytic viruses into tumours.


Asunto(s)
Glioma/virología , Orthoreovirus Mamífero 3/fisiología , Nucleopoliedrovirus/fisiología , Viroterapia Oncolítica , Infecciones por Reoviridae/inmunología , Animales , Línea Celular Tumoral , Efecto Citopatogénico Viral , Glioma/patología , Humanos , Moléculas de Adhesión de Unión/genética , Moléculas de Adhesión de Unión/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Esferoides Celulares/patología , Spodoptera , Carga Viral , Tropismo Viral
10.
PLoS One ; 12(9): e0184816, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28922411

RESUMEN

Oncolytic viruses (OV) have recently emerged as a promising therapeutic modality in cancer treatment. OV selectively infect and kill tumor cells, while sparing untransformed cells. The direct cytotoxic effects combined with the capacity to trigger an immune response make OV an appealing combination partner in the burgeoning field of cancer immunotherapy. One of the leading OV therapeutic candidates is the double-stranded RNA virus reovirus. In order to improve the oncolytic activity of reovirus and allow for systemic administration despite the prevalence of neutralizing antibodies, cytokine-induced killer (CIK) cells were explored as cell carriers for reovirus delivery. In this study, CIK cells were successfully loaded with reovirus ex vivo, and viral replication was limited in CIK cells. Confocal microscopy and flow cytometry demonstrated that CIK cells retained reovirus on the surface. Moreover, CIK cells could promote reovirus infection of tumor cells in the presence of neutralizing antibodies; meanwhile, cytotoxicity of CIK cells was increased after loading with reovirus. These findings support further investigation of reovirus and CIK combination for antitumor therapy.


Asunto(s)
Células Asesinas Inducidas por Citocinas/inmunología , Inmunidad Celular , Orthoreovirus Mamífero 3/fisiología , Neoplasias/inmunología , Neoplasias/terapia , Virus Oncolíticos/fisiología , Replicación Viral/inmunología , Animales , Línea Celular Tumoral , Humanos , Ratones , Neoplasias/patología
11.
J Mol Cell Cardiol ; 111: 102-113, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28822807

RESUMEN

Viral myocarditis is a leading cause of sudden death in young adults as the limited turnover of cardiac myocytes renders the heart particularly vulnerable to viral damage. Viruses induce an antiviral type I interferon (IFN-α/ß) response in essentially all cell types, providing an immediate innate protection. Cardiac myocytes express high basal levels of IFN-ß to help pre-arm them against viral infections, however the mechanism underlying this expression remains unclear. Using primary cultures of murine cardiac and skeletal muscle cells, we demonstrate here that the mitochondrial antiviral signaling (MAVS) pathway is spontaneously activated in unstimulated cardiac myocytes but not cardiac fibroblasts or skeletal muscle cells. Results suggest that MAVS association with the mitochondrial-associated ER membranes (MAM) is a determinant of high basal IFN-ß expression, and demonstrate that MAVS is essential for spontaneous high basal expression of IFN-ß in cardiac myocytes and the heart. Together, results provide the first mechanism for spontaneous high expression of the antiviral cytokine IFN-ß in a poorly replenished and essential cell type.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antivirales/metabolismo , Interferón beta/metabolismo , Miocitos Cardíacos/metabolismo , Transducción de Señal , Envejecimiento/metabolismo , Animales , Femenino , Fibroblastos/metabolismo , Fibroblastos/virología , Helicasa Inducida por Interferón IFIH1/metabolismo , Orthoreovirus Mamífero 3/fisiología , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/virología , Peroxisomas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo
12.
Virology ; 502: 133-143, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28043025

RESUMEN

Viral myocarditis is common and can progress to cardiac failure. Cardiac cell pro-inflammatory responses are critical for viral clearance, however sustained inflammatory responses contribute to cardiac damage. The transcription factor NF-κB regulates expression of many pro-inflammatory cytokines, but basal and induced activation of NF-κB in different cardiac cell types have not been compared. Here, we used primary cultures of cardiac myocytes and cardiac fibroblasts to identify cardiac cell type-specific events. We show that while viral infection readily stimulates activation of NF-κB in cardiac fibroblasts, cardiac myocytes are largely recalcitrant to activation of NF-κB. Moreover, we show that cardiac myocyte subpopulations differ in their NF-κB subcellular localization and identify the cis-Golgi as a cardiac myocyte-specific host compartment. Together, results indicate that NF-κB-dependent signaling in the heart is cardiac cell type-specific, likely reflecting mechanisms that have evolved to balance responses that can be either protective or damaging to the heart.


Asunto(s)
Corazón/virología , Orthoreovirus Mamífero 3/fisiología , FN-kappa B/genética , Infecciones por Reoviridae/metabolismo , Animales , Línea Celular , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/virología , Aparato de Golgi/metabolismo , Aparato de Golgi/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/virología , FN-kappa B/metabolismo , Especificidad de Órganos , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/virología , Transducción de Señal
13.
J Virol ; 90(23): 10951-10962, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27681135

RESUMEN

Proteins that form the reovirus outer capsid play an active role in the entry of reovirus into host cells. Among these, the σ1 protein mediates attachment of reovirus particles to host cells via interaction with cell surface glycans or the proteinaceous receptor junctional adhesion molecule A (JAM-A). The µ1 protein functions to penetrate the host cell membrane to allow delivery of the genome-containing viral core particle into the cytoplasm to initiate viral replication. We demonstrate that a reassortant virus that expresses the M2 gene-encoded µ1 protein derived from prototype strain T3D in an otherwise prototype T1L background (T1L/T3DM2) infects cells more efficiently than parental T1L. Unexpectedly, the enhancement in infectivity of T1L/T3DM2 is due to its capacity to attach to cells more efficiently. We present genetic data implicating the central region of µ1 in altering the cell attachment property of reovirus. Our data indicate that the T3D µ1-mediated enhancement in infectivity of T1L is dependent on the function of σ1 and requires the expression of JAM-A. We also demonstrate that T1L/T3DM2 utilizes JAM-A more efficiently than T1L. These studies revealed a previously unknown relationship between two nonadjacent reovirus outer capsid proteins, σ1 and µ1. IMPORTANCE: How reovirus attaches to host cells has been extensively characterized. Attachment of reovirus to host cells is mediated by the σ1 protein, and properties of σ1 influence the capacity of reovirus to target specific host tissues and produce disease. Here, we present new evidence indicating that the cell attachment properties of σ1 are influenced by the nature of µ1, a capsid protein that does not physically interact with σ1. These studies could explain the previously described role for µ1 in influencing reovirus pathogenesis. These studies are also of broader significance because they highlight an example of how genetic reassortment between virus strains could produce phenotypes that are distinct from those of either parent.


Asunto(s)
Proteínas de la Cápside/fisiología , Orthoreovirus Mamífero 3/fisiología , Orthoreovirus Mamífero 3/patogenicidad , Animales , Proteínas de la Cápside/genética , Moléculas de Adhesión Celular/fisiología , Línea Celular , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Orthoreovirus Mamífero 3/genética , Ratones , Orthoreovirus de los Mamíferos/genética , Orthoreovirus de los Mamíferos/patogenicidad , Orthoreovirus de los Mamíferos/fisiología , Receptores de Superficie Celular/fisiología , Receptores Virales/fisiología , Infecciones por Reoviridae/etiología , Infecciones por Reoviridae/virología , Virulencia/genética , Virulencia/fisiología , Acoplamiento Viral
14.
Oncotarget ; 7(30): 48517-48532, 2016 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-27384486

RESUMEN

Oncolytic viruses selectively target and replicate in cancer cells, providing us with a unique tool with which to target and kill tumour cells. These viruses come from a diverse range of viral families including reovirus type 3 Dearing (RT3D), a non-pathogenic human double-stranded RNA oncolytic virus, which has been shown to be an effective therapeutic agent, both as a mono-therapy and in combination with traditional chemotherapeutic drugs. This study investigated the interaction between RT3D and radiotherapy in melanoma cell lines with a BRAF mutant, Ras mutant or BRAF/Ras wild type genotype. The data indicates that RT3D combined with radiotherapy significantly increased cytotoxicity relative to either single agent, independent of genotype, both in vitro and in vivo. The mechanism of enhanced cytotoxicity was dependent on an increase in viral replication, mediated by CUG2 up-regulation and subsequent down-regulation of pPKR and p-eIF2α, leading to the activation of mitochondrial apoptotic signalling resulting in increased cell death.


Asunto(s)
Apoptosis/efectos de la radiación , Melanoma/terapia , Mitocondrias/metabolismo , Viroterapia Oncolítica/métodos , Transducción de Señal/efectos de la radiación , Replicación Viral , Animales , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/metabolismo , Terapia Combinada/métodos , Regulación hacia Abajo , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Orthoreovirus Mamífero 3/fisiología , Melanoma/genética , Ratones , Mitocondrias/efectos de la radiación , Mutación , Virus Oncolíticos/fisiología , Fosforilación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Regulación hacia Arriba , eIF-2 Quinasa/metabolismo
15.
Biologicals ; 43(6): 519-23, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26260690

RESUMEN

The efficacy of gaseous disinfection is critical for prevention and treatment of microbial contamination in biotechnological facilities. For an evaluation of gaseous disinfection efficacy, a down-scaled laboratory model was established, using currently available carrier tests and a custom-made dry fog box. A mixture of peroxyacetic acid and hydrogen peroxide (PAA/HP) was investigated as example, at concentrations between 0.4 and 2.9 mL/m(3) for up to 3 h for inactivation of a panel of lipid-enveloped and non-lipid-enveloped viruses. The influenza viruses were most sensitive to PAA/HP treatment and minute virus of mice was most resistant. Bovine viral diarrhea virus and reovirus III showed intermediate stability and similar inactivation kinetics. Use of the dry fog box circumvents dedicating an entire lab for the investigation, which renders the generation of data more cost-effective and allows for production of highly reproducible kinetic data.


Asunto(s)
Desinfectantes/farmacología , Gases , Peróxido de Hidrógeno/farmacología , Ácido Peracético/farmacología , Virología/instrumentación , Inactivación de Virus/efectos de los fármacos , Animales , Línea Celular , Virus de la Diarrea Viral Bovina/efectos de los fármacos , Virus de la Diarrea Viral Bovina/fisiología , Desinfección , Evaluación de Medicamentos , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/fisiología , Virus de la Influenza B/efectos de los fármacos , Virus de la Influenza B/fisiología , Orthoreovirus Mamífero 3/efectos de los fármacos , Orthoreovirus Mamífero 3/fisiología , Virus Diminuto del Ratón/efectos de los fármacos , Virus Diminuto del Ratón/fisiología , Factores de Tiempo , Carga Viral , Cultivo de Virus
16.
mBio ; 6(3): e00593-15, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25991685

RESUMEN

UNLABELLED: Since May 2013, outbreaks of porcine epidemic diarrhea have devastated the U.S. swine industry, causing immense economic losses. Two different swine enteric coronaviruses (porcine epidemic diarrhea virus and Delta coronavirus) have been isolated from the affected swine population. The disease has been reported from at least 32 states of the United States and other countries, including Mexico, Peru, Dominican Republic, Canada, Columbia, Ecuador, and Ukraine, with repeated outbreaks in previously infected herds. Here we report the isolation and characterization of a novel mammalian orthoreovirus 3 (MRV3) from diarrheic feces of piglets from these outbreaks in three states and ring-dried swine blood meal from multiple sources. MRV3 could not be isolated from healthy or pigs that had recovered from epidemic diarrhea from four states. Several MRV3 isolates were obtained from chloroform-extracted pig feces or blood meal in cell cultures or developing chicken embryos. Biological characterization of two representative isolates revealed trypsin resistance and thermostability at 90°C. NextGen sequencing of ultrapurified viruses indicated a strong homology of the S1 segment to mammalian and bat MRV3. Neonatal piglets experimentally infected with these viruses or a chloroform extract of swine blood meal developed severe diarrhea and acute gastroenteritis with 100% mortality within 3 days postinfection. Therefore, the novel porcine MRV3 may contribute to enteric disease along with other swine enteric viruses. The role of MRV3 in the current outbreaks of porcine epidemic diarrhea in the United States remains to be determined, but the pathogenic nature of the virus warrants further investigations on its epidemiology and prevalence. IMPORTANCE: Porcine orthoreoviruses causing diarrhea have been reported in China and Korea but not in the United States. We have isolated and characterized two pathogenic reassortant MRV3 isolates from swine fecal samples from porcine epidemic diarrhea outbreaks and ring-dried swine blood meal in the United States. These fecal and blood meal isolates or a chloroform extract of blood meal induced severe diarrhea and mortality in experimentally infected neonatal pigs. Genetic and phylogenetic analyses of two MRV3 isolates revealed that they are identical but differed significantly from nonpathogenic mammalian orthoreoviruses circulating in the United States. The present study provides a platform for immediate development of suitable vaccines and diagnostics to prevent and control porcine orthoreovirus diarrhea.


Asunto(s)
Sangre/virología , Diarrea/veterinaria , Heces/virología , Orthoreovirus Mamífero 3/clasificación , Orthoreovirus Mamífero 3/aislamiento & purificación , Enfermedades de los Porcinos/virología , Animales , Análisis por Conglomerados , Diarrea/virología , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/fisiología , Datos de Secuencia Molecular , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia , Porcinos , Estados Unidos , Cultivo de Virus
17.
Artículo en Inglés | MEDLINE | ID: mdl-25905045

RESUMEN

As obligate intracellular parasites, viruses are exclusively and intimately dependent upon their host cells for replication. During replication viruses induce profound changes within cells, including: induction of signaling pathways, morphological changes, and cell death. Many such cellular perturbations have been analyzed at the transcriptomic level by gene arrays and recent efforts have begun to analyze cellular proteomic responses. We recently described comparative stable isotopic (SILAC) analyses of reovirus, strain type 3 Dearing (T3D)-infected HeLa cells. For the present study we employed the complementary labeling strategy of iTRAQ (isobaric tags for relative and absolute quantitation) to examine HeLa cell changes induced by T3D, another reovirus strain, type 1 Lang, and UV-inactivated T3D (UV-T3D). Triplicate replicates of cytosolic and nuclear fractions identified a total of 2375 proteins, of which 50, 57, and 46 were significantly up-regulated, and 37, 26, and 44 were significantly down-regulated by T1L, T3D, and UV-T3D, respectively. Several pathways, most notably the Interferon signaling pathway and the EIF2 and ILK signaling pathways, were induced by virus infection. Western blots confirmed that cells were more strongly activated by live T3D as demonstrated by elevated levels of key proteins like STAT-1, ISG-15, IFIT-1, IFIT-3, and Mx1. This study expands our understanding of reovirus-induced host responses.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Interferones/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Orthoreovirus Mamífero 3/fisiología , Proteínas de Resistencia a Mixovirus/metabolismo , Infecciones por Reoviridae/metabolismo , Factor de Transcripción STAT1/metabolismo , Ubiquitinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/genética , Citocinas/genética , Células HeLa , Humanos , Interferones/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Orthoreovirus Mamífero 3/genética , Proteínas de Resistencia a Mixovirus/genética , Proteómica , Proteínas de Unión al ARN , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/microbiología , Factor de Transcripción STAT1/genética , Ubiquitinas/genética
18.
mBio ; 5(2): e00902-14, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24618253

RESUMEN

Flaviviruses, particularly Japanese encephalitis virus (JEV) and West Nile virus (WNV), are important causes of virus-induced central nervous system (CNS) disease in humans. We used microarray analysis to identify cellular genes that are differentially regulated following infection of the brain with JEV (P3) or WNV (New York 99). Gene expression data for these flaviviruses were compared to those obtained following infection of the brain with reovirus (type 3 Dearing), an unrelated neurotropic virus. We found that a large number of genes were up-regulated by all three viruses (using the criteria of a change of >2-fold and a P value of <0.001), including genes associated with interferon signaling, the immune system, inflammation, and cell death/survival signaling. In addition, genes associated with glutamate signaling were down-regulated in infections with all three viruses (criteria, a >2-fold change and a P value of <0.001). These genes may serve as broad-spectrum therapeutic targets for virus-induced CNS disease. A distinct set of genes were up-regulated following flavivirus infection but not following infection with reovirus. These genes were associated with tRNA charging and may serve as therapeutic targets for flavivirus-induced CNS disease. IMPORTANCE Viral infections of the central nervous system (CNS) are an important cause of morbidity and mortality. Treatment options for virus-induced CNS disease are limited, and for many clinically important neurotropic viruses, no specific therapy of proven benefit is currently available. We performed microarray analysis to identify genes that are differentially regulated in the brain following virus infection in order to identify pathways that might provide novel therapeutic targets for virus-induced CNS disease. Although several studies have described gene expression changes following virus infection of the brain, this report is the first to directly compare large-scale gene expression data from different viruses. We identified genes that are differentially regulated in infection of the brain with viruses from different families and those which appear to be specific to flavivirus infections.


Asunto(s)
Encéfalo/inmunología , Encéfalo/virología , Virus de la Encefalitis Japonesa (Subgrupo)/fisiología , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Virus del Nilo Occidental/fisiología , Aminoacil-ARNt Sintetasas/biosíntesis , Animales , Apoptosis , Virus de la Encefalitis Japonesa (Subgrupo)/inmunología , Ácido Glutámico , Interferones/biosíntesis , Orthoreovirus Mamífero 3/inmunología , Orthoreovirus Mamífero 3/fisiología , Ratones , Análisis por Micromatrices , Receptores de Interleucina-17/biosíntesis , Transducción de Señal , Regulación hacia Arriba , Virus del Nilo Occidental/inmunología
19.
J Virol ; 87(24): 13532-42, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24089575

RESUMEN

The reovirus outer capsid protein µ1 forms a lattice surrounding the viral core. In the native state, µ1 determines the environmental stability of the viral capsid. Additionally, during cell entry, µ1 undergoes structural rearrangements that facilitate delivery of the viral cores across the membrane. To determine how the capsid-stabilizing functions of µ1 impinge on the capacity of µ1 to undergo conformational changes required for cell entry, we characterized viruses with mutations engineered at charged residues within the µ1 loop formed by residues 72 to 96 (72-96 loop). This loop is proposed to stabilize the capsid by mediating interactions between neighboring µ1 trimers and between trimers and the core. We found that mutations at Glu89 (E89) within this loop produced viruses with compromised efficiency for completing their replication cycle. ISVPs of E89 mutants converted to ISVP*s more readily than those of wild-type viruses. The E89 mutants yielded revertants with second-site substitutions within regions that mediate interaction between µ1 trimers at a site distinct from the 72-96 loop. These viruses also contained changes in regions that control interactions within µ1 trimers. Viruses containing these second-site changes displayed restored plaque phenotypes and were capable of undergoing ISVP-to-ISVP* conversion in a regulated manner. These findings highlight regions of µ1 that stabilize the reovirus capsid and demonstrate that an enhanced propensity to form ISVP*s in an unregulated manner compromises viral fitness.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Orthoreovirus Mamífero 3/fisiología , Infecciones por Reoviridae/virología , Reoviridae/fisiología , Internalización del Virus , Animales , Cápside/química , Cápside/metabolismo , Proteínas de la Cápside/genética , Línea Celular , Cristalización , Orthoreovirus Mamífero 3/química , Orthoreovirus Mamífero 3/genética , Ratones , Mutación , Conformación Proteica , Reoviridae/genética
20.
J Virol ; 87(23): 12980-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24067960

RESUMEN

Apoptosis is a type of controlled cell death that is essential for development and tissue homeostasis. It also serves as a robust host response against infection by many viruses. The capacity of neurotropic viruses to induce apoptosis strongly correlates with virulence. However, the precise function of apoptosis in viral infection is not well understood. Reovirus is a neurotropic virus that induces apoptosis in a variety of cell types, including central nervous system neurons, leading to fatal encephalitis in newborn mice. To determine the effect of apoptosis on reovirus replication in the host, we generated two otherwise isogenic viruses that differ in a single amino acid in viral capsid protein µ1 that segregates with apoptotic capacity. Apoptosis-proficient and apoptosis-deficient viruses were compared for replication, dissemination, tropism, and tissue injury in newborn mice and for the capacity to spread to uninfected littermates. Our results indicate that apoptotic capacity enhances reovirus replication in the brain and consequent neurovirulence but reduces transmission efficiency. The replication advantage of the apoptosis-proficient strain is limited to the brain and correlates with enhanced infectivity of neurons. These studies reveal a new cell type-specific determinant of reovirus virulence.


Asunto(s)
Apoptosis , Orthoreovirus Mamífero 3/fisiología , Orthoreovirus Mamífero 3/patogenicidad , Infecciones por Reoviridae/fisiopatología , Infecciones por Reoviridae/virología , Replicación Viral , Animales , Animales Recién Nacidos , Encéfalo/citología , Encéfalo/virología , Femenino , Humanos , Masculino , Orthoreovirus Mamífero 3/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...