Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Virol ; 96(2): e0187921, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34757847

RESUMO

Although a broad range of viruses cause myocarditis, the mechanisms that underlie viral myocarditis are poorly understood. Here, we report that the M2 gene is a determinant of reovirus myocarditis. The M2 gene encodes outer capsid protein µ1, which mediates host membrane penetration during reovirus entry. We infected newborn C57BL/6 mice with reovirus strain type 1 Lang (T1L) or a reassortant reovirus in which the M2 gene from strain type 3 Dearing (T3D) was substituted into the T1L genetic background (T1L/T3DM2). T1L was nonlethal in wild-type mice, whereas more than 90% of mice succumbed to T1L/T3DM2 infection. T1L/T3DM2 produced higher viral loads than T1L at the site of inoculation. In secondary organs, T1L/T3DM2 was detected with more rapid kinetics and reached higher peak titers than T1L. We found that hearts from T1L/T3DM2-infected mice were grossly abnormal, with large lesions indicative of substantial inflammatory infiltrate. Lesions in T1L/T3DM2-infected mice contained necrotic cardiomyocytes with pyknotic debris, as well as extensive lymphocyte and histiocyte infiltration. In contrast, T1L induced the formation of small purulent lesions in a small subset of animals, consistent with T1L being mildly myocarditic. Finally, more activated caspase-3-positive cells were observed in hearts from animals infected with T1L/T3DM2 than T1L. Together, our findings indicate that substitution of the T3D M2 allele into an otherwise T1L genetic background is sufficient to change a nonlethal infection into a lethal infection. Our results further indicate that T3D M2 enhances T1L replication and dissemination in vivo, which potentiates the capacity of reovirus to cause myocarditis. IMPORTANCE Reovirus is a nonenveloped virus with a segmented double-stranded RNA genome that serves as a model for studying viral myocarditis. The mechanisms by which reovirus drives myocarditis development are not fully elucidated. We found that substituting the M2 gene from strain type 3 Dearing (T3D) into an otherwise type 1 Lang (T1L) genetic background (T1L/T3DM2) was sufficient to convert the nonlethal T1L strain into a lethal infection in neonatal C57BL/6 mice. T1L/T3DM2 disseminated more efficiently and reached higher maximum titers than T1L in all organs tested, including the heart. T1L is mildly myocarditic and induced small areas of cardiac inflammation in a subset of mice. In contrast, hearts from mice infected with T1L/T3DM2 contained extensive cardiac inflammatory infiltration and more activated caspase-3-positive cells, which is indicative of apoptosis. Together, our findings identify the reovirus M2 gene as a new determinant of reovirus-induced myocarditis.


Assuntos
Proteínas do Capsídeo/metabolismo , Orthoreovirus Mamífero 3/patogenicidade , Miocardite/virologia , Infecções por Reoviridae/virologia , Animais , Animais Recém-Nascidos , Proteínas do Capsídeo/genética , Inflamação , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/mortalidade , Miocardite/patologia , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/metabolismo , Orthoreovirus de Mamíferos/patogenicidade , Infecções por Reoviridae/mortalidade , Infecções por Reoviridae/patologia , Carga Viral , Virulência , Replicação Viral
2.
Nat Commun ; 11(1): 32, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31896744

RESUMO

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.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Orthoreovirus Mamífero 3/fisiologia , Nanopartículas Metálicas/química , Actinas/metabolismo , Animais , Avidina/química , Biotina/química , Capsídeo/química , Células Cultivadas , Fibroblastos/virologia , Ouro , Células HeLa , Humanos , Integrinas/metabolismo , Cinética , Orthoreovirus Mamífero 3/química , Orthoreovirus Mamífero 3/patogenicidade , Nanopartículas Metálicas/virologia , Modelos Teóricos , Miosinas/metabolismo , Ratos , Vírion/patogenicidade , Vírion/fisiologia
3.
Vet Microbiol ; 224: 23-30, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30269786

RESUMO

A novel U.S. strain of mammalian orthoreovirus type 3 (MRV3) isolated from diarrheic pigs in 2015 was reportedly highly pathogenic in pigs. In this study, we first developed an inactivated MRV3 vaccine and determined its protective efficacy against MRV3 infection in conventional neonatal piglets. A pathogenicity study was also conducted in gnotobiotic pigs to further assess the pathogenicity of MRV3. To evaluate if piglets could be protected against MRV3 infection after immunization of pregnant sows with an inactivated MRV3 vaccine, pregnant sows were vaccinated with 2 or 3 doses of the vaccine or with PBS buffer. Four-day-old piglets born to vaccinated and unvaccinated sows were subsequently challenged with MRV3. The results showed that piglets born from vaccinated sows had lower levels of fecal viral RNA shedding at 1, 3, and 4 days post-challenge, suggesting that the inactivated MRV3 vaccine can reduce MRV3 replication. Surprisingly, although the conventional piglets were infected, they did not develop severe enteric disease as reported previously. Therefore, in an effort to further definitively assess the pathogenicity of MRV3, we experimentally infected gnotobiotic pigs, a more sensitive model for pathogenicity study, with the wild-type MRV3 virus. The infected gnotobiotic piglets all survived and exhibited only very mild diarrhea in some pigs. Taken together, the results indicate that the novel strain of MRV3 recently isolated in the United States infected but caused only very mild diarrhea in pigs, and that maternal immunity acquired from sows vaccinated with an inactivated vaccine can reduce MRV3 replication in neonatal pigs.


Assuntos
Orthoreovirus Mamífero 3/patogenicidade , Infecções por Reoviridae/veterinária , Doenças dos Suínos/prevenção & controle , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/imunologia , Diarreia/veterinária , Diarreia/virologia , Fezes/virologia , Feminino , Vida Livre de Germes , Imunidade Materno-Adquirida/imunologia , Imunização/veterinária , Gravidez , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/prevenção & controle , Suínos , Doenças dos Suínos/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas Virais/administração & dosagem , Virulência
4.
J Virol ; 90(23): 10951-10962, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27681135

RESUMO

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.


Assuntos
Proteínas do Capsídeo/fisiologia , Orthoreovirus Mamífero 3/fisiologia , Orthoreovirus Mamífero 3/patogenicidade , Animais , Proteínas do Capsídeo/genética , Moléculas de Adesão Celular/fisiologia , Linhagem Celular , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Orthoreovirus Mamífero 3/genética , Camundongos , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/patogenicidade , Orthoreovirus de Mamíferos/fisiologia , Receptores de Superfície Celular/fisiologia , Receptores Virais/fisiologia , Infecções por Reoviridae/etiologia , Infecções por Reoviridae/virologia , Virulência/genética , Virulência/fisiologia , Ligação Viral
5.
PLoS One ; 11(9): e0161914, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27598998

RESUMO

Alternative splicing (AS) is a central mechanism of genetic regulation which modifies the sequence of RNA transcripts in higher eukaryotes. AS has been shown to increase both the variability and diversity of the cellular proteome by changing the composition of resulting proteins through differential choice of exons to be included in mature mRNAs. In the present study, alterations to the global RNA splicing landscape of cellular genes upon viral infection were investigated using mammalian reovirus as a model. Our study provides the first comprehensive portrait of global changes in the RNA splicing signatures that occur in eukaryotic cells following infection with a human virus. We identify 240 modified alternative splicing events upon infection which belong to transcripts frequently involved in the regulation of gene expression and RNA metabolism. Using mass spectrometry, we also confirm modifications to transcript-specific peptides resulting from AS in virus-infected cells. These findings provide additional insights into the complexity of virus-host interactions as these splice variants expand proteome diversity and function during viral infection.


Assuntos
Processamento Alternativo , Fibroblastos/metabolismo , Genoma , Interações Hospedeiro-Patógeno/genética , Orthoreovirus Mamífero 3/crescimento & desenvolvimento , RNA Mensageiro/genética , Sequência de Aminoácidos , Animais , Éxons , Fibroblastos/virologia , Ontologia Genética , Humanos , Orthoreovirus Mamífero 3/patogenicidade , Camundongos , Anotação de Sequência Molecular , Proteômica , RNA Mensageiro/metabolismo , Análise de Sequência de RNA
6.
J Virol ; 89(8): 4319-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25653434

RESUMO

UNLABELLED: Wild-type mammalian orthoreovirus serotype 3 Dearing (T3wt) is nonpathogenic in humans but preferentially infects and kills cancer cells in culture and demonstrates promising antitumor activity in vivo. Using forward genetics, we previously isolated two variants of reovirus, T3v1 and T3v2, with increased infectivity toward a panel of cancer cell lines and improved in vivo oncolysis in a murine melanoma model relative to that of T3wt. Our current study explored how mutations in T3v1 and T3v2 promote infectivity. Reovirions contain trimers of σ1, the reovirus cell attachment protein, at icosahedral capsid vertices. Quantitative Western blot analysis showed that purified T3v1 and T3v2 virions had ∼ 2- and 4-fold-lower levels of σ1 fiber than did T3wt virions. Importantly, using RNA interference to reduce σ1 levels during T3wt production, we were able to generate wild-type reovirus with reduced levels of σ1 per virion. As σ1 levels were reduced, virion infectivity increased by 2- to 5-fold per cell-bound particle, demonstrating a causal relationship between virion σ1 levels and the infectivity of incoming virions. During infection of tumorigenic L929 cells, T3wt, T3v1, and T3v2 uncoated the outer capsid proteins σ3 and µ1C at similar rates. However, having started with fewer σ1 molecules, a complete loss of σ1 was achieved sooner for T3v1 and T3v2. Distinct from intracellular uncoating, chymotrypsin digestion, as a mimic of natural enteric infection, resulted in more rapid σ3 and µ1C removal, unique disassembly intermediates, and a rapid loss of infectivity for T3v1 and T3v2 compared to T3wt. Optimal infectivity toward natural versus therapeutic niches may therefore require distinct reovirus structures and σ1 levels. IMPORTANCE: Wild-type reovirus is currently in clinical trials as a potential cancer therapy. Our molecular studies on variants of reovirus with enhanced oncolytic activity in vitro and in vivo now show that distinct reovirus structures promote adaptation toward cancer cells and away from conditions that mimic natural routes of infection. Specifically, we found that reovirus particles with fewer molecules of the cell attachment protein σ1 became more infectious toward transformed cells. Reduced σ1 levels conferred a benefit to incoming particles only, resulting in an earlier depletion of σ1 and a higher probability of establishing productive infection. Conversely, reovirus variants with fewer σ1 molecules showed reduced stability and infectivity and distinct disassembly when exposed to conditions that mimic natural intestinal proteolysis. These findings support a model where the mode of infection dictates the precise optimum of reovirus structure and provide a molecular rationale for considering alternative reovirus structures during oncolytic therapy.


Assuntos
Adaptação Biológica/genética , Proteínas do Capsídeo/metabolismo , Orthoreovirus Mamífero 3/genética , Vírus Oncolíticos/genética , Vírion/metabolismo , Internalização do Vírus , Animais , Western Blotting , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Quimotripsina , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Imunoprecipitação , Orthoreovirus Mamífero 3/patogenicidade , Camundongos , Mutação/genética , Vírus Oncolíticos/patogenicidade , Interferência de RNA
7.
Gene Ther ; 21(6): 609-17, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24739522

RESUMO

In the canonical pathway, infection of cells by the wild-type mammalian orthoreovirus Type 3 Dearing (T3D) is dependent on the interaction of the viral spike protein σ1 with the high-affinity cellular receptor junction adhesion molecule-A (JAM-A). We previously demonstrated that the human glioblastoma cell line U-118 MG does not express JAM-A and resists reovirus T3D infection in standard cell culture conditions (SCCC). Heterologous JAM-A expression sensitises U-118 MG cells to reovirus T3D. Here we studied reovirus infection in U-118 MG cells grown in spheroid cultures with the premise that cells in such cultures resemble cells in tumours more than those grown under standard adherent cell culture conditions on a plastic surface. Although the U-118 MG cells in spheroids do not express JAM-A, they are susceptible to reovirus T3D infection. We show that this can be attributed to factors secreted by cells in the spheroids. The concentration of active extracellular proteases cathepsin B and L in the medium of spheroid cultures was increased 19- and 24-fold, respectively, as compared with SCCC. These enzymes can convert the reovirus particles into a form that can infect the U-118 MG cells independent of JAM-A. Taken together, these data demonstrate that infection of tumour cells by wild-type reovirus T3D is not strictly dependent on the expression of JAM-A on the cell surface.


Assuntos
Moléculas de Adesão Celular/metabolismo , Glioblastoma/patologia , Glioblastoma/virologia , Orthoreovirus Mamífero 3/patogenicidade , Receptores de Superfície Celular/metabolismo , Esferoides Celulares/virologia , Catepsina B/metabolismo , Catepsina L/metabolismo , Humanos
8.
J Virol ; 87(23): 12980-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24067960

RESUMO

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.


Assuntos
Apoptose , Orthoreovirus Mamífero 3/fisiologia , Orthoreovirus Mamífero 3/patogenicidade , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Replicação Viral , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/virologia , Feminino , Humanos , Masculino , Orthoreovirus Mamífero 3/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Virais/genética , Proteínas Virais/metabolismo , Virulência
9.
J Virol ; 87(6): 3447-60, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23302889

RESUMO

Reovirus infection is a well-characterized experimental system for the study of viral pathogenesis and antiviral immunity within the central nervous system (CNS). We have previously shown that c-Jun N-terminal kinase (JNK) and the Fas death receptor each play a role in neuronal apoptosis occurring in reovirus-infected brains. Death-associated protein 6 (Daxx) is a cellular protein that mechanistically links Fas signaling to JNK signaling in several models of apoptosis. In the present study, we demonstrate that Daxx is upregulated in reovirus-infected brain tissue through a type I interferon-mediated mechanism. Daxx upregulation is limited to brain regions that undergo reovirus-induced apoptosis and occurs in the cytoplasm and nucleus of neurons. Cytoplasmic Daxx is present in Fas-expressing cells during reovirus encephalitis, suggesting a role for Daxx in Fas-mediated apoptosis following reovirus infection. Further, in vitro expression of a dominant negative form of Daxx (DN-Daxx), which binds to Fas but which does not transmit downstream signaling, inhibits apoptosis of reovirus-infected cells. In contrast, in vitro depletion of Daxx results in increased expression of caspase 3 and apoptosis, suggesting that Daxx plays an antiapoptotic role in the nucleus. Overall, these data imply a regulatory role for Daxx in reovirus-induced apoptosis, depending on its location in the nucleus or cytoplasm.


Assuntos
Apoptose , Proteínas de Transporte/biossíntese , Citoplasma/virologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Orthoreovirus Mamífero 3/patogenicidade , Neurônios/virologia , Proteínas Nucleares/biossíntese , Animais , Proteínas Correpressoras , Orthoreovirus Mamífero 3/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Chaperonas Moleculares , Regulação para Cima
10.
J Virol ; 86(24): 13164-73, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23035227

RESUMO

Mammalian reoviruses display serotype-specific patterns of tropism and disease in the murine central nervous system (CNS) attributable to polymorphisms in viral attachment protein σ1. While all reovirus serotypes use junctional adhesion molecule-A as a cellular receptor, they differ in their utilization of carbohydrate coreceptors. This observation raises the possibility that carbohydrate binding by σ1 influences reovirus pathology in the CNS. In this study, we sought to define the function of carbohydrate binding in reovirus neuropathogenesis. Newborn mice were inoculated intramuscularly with wild-type strain type 3 Dearing (T3D) and T3D-σ1R202W, a point mutant T3D derivative that does not bind sialic acid (SA). Infected mice were monitored for survival, and viral loads at the sites of primary and secondary replication were quantified. Fewer mice inoculated with the wild-type virus survived in comparison to those inoculated with the mutant virus. The wild-type virus also produced higher titers in the spinal cord and brain at late times postinoculation but lower titers in the liver in comparison to those produced by the mutant virus. In addition, the wild-type virus was more virulent and produced higher titers in the brain than the mutant following intracranial inoculation. These animal infectivity studies suggest that T3D-σ1R202W harbors a defect in neural growth. Concordantly, compared with the wild-type virus, the mutant virus displayed a decreased capacity to infect and replicate in primary cultures of cortical neurons, a property dependent on cell surface SA. These results suggest that SA binding enhances the kinetics of reovirus replication in neural tissues and highlight a functional role for sialylated glycans as reovirus coreceptors in the CNS.


Assuntos
Sistema Nervoso Central/virologia , Orthoreovirus Mamífero 3/patogenicidade , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/metabolismo , Receptores Virais/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Imuno-Histoquímica , Orthoreovirus Mamífero 3/isolamento & purificação , Orthoreovirus Mamífero 3/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Carga Viral , Virulência , Replicação Viral
11.
Virology ; 433(2): 489-97, 2012 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22999096

RESUMO

Human milk contains many bioactive components, including secretory IgA, oligosaccharides, and milk-associated proteins. We assessed the antiviral effects of several components of milk against mammalian reoviruses. We found that glucocerebroside (GCB) inhibited the infectivity of reovirus strain type 1 Lang (T1L), whereas gangliosides GD3 and GM3 and 3'-sialyllactose (3SL) inhibited the infectivity of reovirus strain type 3 Dearing (T3D). Agglutination of erythrocytes mediated by T1L and T3D was inhibited by GD3, GM3, and bovine lactoferrin. Additionally, α-sialic acid, 3SL, 6'-sialyllactose, sialic acid, human lactoferrin, osteopontin, and α-lactalbumin inhibited hemagglutination mediated by T3D. Using single-gene reassortant viruses, we found that serotype-specific differences segregate with the gene encoding the viral attachment protein. Furthermore, GD3, GM3, and 3SL inhibit T3D infectivity by blocking binding to host cells, whereas GCB inhibits T1L infectivity post-attachment. These results enhance an understanding of reovirus cell attachment and define a mechanism for the antimicrobial activity of human milk.


Assuntos
Proteínas do Capsídeo/imunologia , Orthoreovirus Mamífero 3/imunologia , Orthoreovirus Mamífero 3/patogenicidade , Leite Humano/imunologia , Orthoreovirus de Mamíferos/imunologia , Orthoreovirus de Mamíferos/patogenicidade , Polissacarídeos/imunologia , Animais , Proteínas do Capsídeo/genética , Bovinos , Feminino , Gangliosídeo G(M3)/imunologia , Gangliosídeos/imunologia , Genes Virais , Células HeLa , Testes de Inibição da Hemaglutinação , Interações Hospedeiro-Patógeno/imunologia , Humanos , Células L , Orthoreovirus Mamífero 3/classificação , Orthoreovirus Mamífero 3/genética , Camundongos , Leite Humano/virologia , Oligossacarídeos/imunologia , Orthoreovirus de Mamíferos/classificação , Orthoreovirus de Mamíferos/genética , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/prevenção & controle , Infecções por Reoviridae/virologia , Sorotipagem , Especificidade da Espécie , Ligação Viral
12.
J Virol ; 86(13): 7403-13, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22532697

RESUMO

Reovirus preferentially replicates in transformed cells and is being explored as a cancer therapy. Immunological and physical barriers to virotherapy inspired a quest for reovirus variants with enhanced oncolytic potency. Using a classical genetics approach, we isolated two reovirus variants (T3v1 and T3v2) with superior replication relative to wild-type reovirus serotype 3 Dearing (T3wt) on various human and mouse tumorigenic cell lines. Unique mutations in reovirus λ2 vertex protein and σ1 cell attachment protein were associated with the large plaque-forming phenotype of T3v1 and T3v2, respectively. Both T3v1 and T3v2 exhibited higher infectivity (i.e., a higher PFU-to-particle ratio) than T3wt. A detailed analysis of virus replication revealed that virus cell binding and uncoating were equivalent for variant and wild-type reoviruses. However, T3v1 and T3v2 were significantly more efficient than T3wt in initiating productive infection. Thus, when cells were infected with equivalent input virus particles, T3v1 and T3v2 produced significantly higher levels of early viral RNAs relative to T3wt. Subsequent steps of virus replication (viral RNA and protein synthesis, virus assembly, and cell death) were equivalent for all three viruses. In a syngeneic mouse model of melanoma, both T3v1 and T3v2 prolonged mouse survival compared to wild-type reovirus. Our studies reveal that oncolytic potency of reovirus can be improved through distinct mutations that increase the infectivity of reovirus particles.


Assuntos
Proteínas do Capsídeo/genética , Orthoreovirus Mamífero 3/patogenicidade , Mutação , Nucleotidiltransferases/genética , Vírus Oncolíticos/patogenicidade , Proteínas do Core Viral/genética , Fatores de Virulência/genética , Replicação Viral , Animais , Proteínas do Capsídeo/metabolismo , Modelos Animais de Doenças , Orthoreovirus Mamífero 3/genética , Melanoma/mortalidade , Melanoma/terapia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Nucleotidiltransferases/metabolismo , Vírus Oncolíticos/genética , Análise de Sequência de DNA , Análise de Sobrevida , Proteínas do Core Viral/metabolismo , Carga Viral , Ensaio de Placa Viral , Fatores de Virulência/metabolismo
13.
J Virol ; 85(22): 11781-90, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21917967

RESUMO

Nonstructural protein σ1s is a critical determinant of hematogenous dissemination by type 1 reoviruses, which reach the central nervous system (CNS) by a strictly blood-borne route. However, it is not known whether σ1s contributes to neuropathogenesis of type 3 reoviruses, which disseminate by both vascular and neural pathways. Using isogenic type 3 viruses that vary only in σ1s expression, we observed that mice survived at a higher frequency following hind-limb inoculation with σ1s-null virus than when inoculated with wild-type virus. This finding suggests that σ1s is essential for reovirus virulence when inoculated at a site that requires systemic spread to cause disease. Wild-type and σ1s-null viruses produced comparable titers in the spinal cord, suggesting that σ1s is dispensable for invasion of the CNS. Although the two viruses ultimately achieved similar peak titers in the brain, loads of wild-type virus were substantially greater than those of the σ1s-null mutant at early times after inoculation. In contrast, wild-type virus produced substantially higher titers than the σ1s-null virus in peripheral organs to which reovirus spreads via the blood, including the heart, intestine, liver, and spleen. Concordantly, viral titers in the blood were higher following infection with wild-type virus than following infection with the σ1s-null mutant. These results suggest that differences in viral brain titers at early time points postinfection are due to limited virus delivery to the brain by hematogenous pathways. Transection of the sciatic nerve prior to hind-limb inoculation diminished viral spread to the spinal cord. However, wild-type virus retained the capacity to disseminate to the brain following sciatic nerve transection, indicating that wild-type reovirus can spread to the brain by the blood. Together, these results indicate that σ1s is not required for reovirus spread by neural mechanisms. Instead, σ1s mediates hematogenous dissemination within the infected host, which is required for full reovirus neurovirulence.


Assuntos
Proteínas do Capsídeo/metabolismo , Orthoreovirus Mamífero 3/patogenicidade , Infecções por Reoviridae/virologia , Fatores de Virulência/metabolismo , Animais , Sangue/virologia , Encéfalo/virologia , Proteínas do Capsídeo/genética , Coração/virologia , Intestinos/virologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Medula Espinal/virologia , Baço/virologia , Análise de Sobrevida , Carga Viral , Fatores de Virulência/deficiência
14.
J Virol ; 85(1): 296-304, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980509

RESUMO

Mammalian orthoreoviruses induce apoptosis in vivo and in vitro; however, the specific mechanism by which apoptosis is induced is not fully understood. Recent studies have indicated that the reovirus outer capsid protein µ1 is the primary determinant of reovirus-induced apoptosis. Ectopically expressed µ1 induces apoptosis and localizes to intracellular membranes. Here we report that ectopic expression of µ1 activated both the extrinsic and intrinsic apoptotic pathways with activation of initiator caspases-8 and -9 and downstream effector caspase-3. Activation of both pathways was required for µ1-induced apoptosis, as specific inhibition of either caspase-8 or caspase-9 abolished downstream effector caspase-3 activation. Similar to reovirus infection, ectopic expression of µ1 caused release into the cytosol of cytochrome c and smac/DIABLO from the mitochondrial intermembrane space. Pancaspase inhibitors did not prevent cytochrome c release from cells expressing µ1, indicating that caspases were not required. Additionally, µ1- or reovirus-induced release of cytochrome c occurred efficiently in Bax(-/-)Bak(-/-) mouse embryonic fibroblasts (MEFs). Finally, we found that reovirus-induced apoptosis occurred in Bax(-/-)Bak(-/-) MEFs, indicating that reovirus-induced apoptosis occurs independently of the proapoptotic Bcl-2 family members Bax and Bak.


Assuntos
Apoptose/fisiologia , Proteínas do Capsídeo/metabolismo , Orthoreovirus Mamífero 3/patogenicidade , Orthoreovirus de Mamíferos/patogenicidade , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Células CHO , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/farmacologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspases/genética , Caspases/metabolismo , Linhagem Celular , Cricetinae , Cricetulus , Citocromos c/genética , Citocromos c/metabolismo , Citosol/metabolismo , Fibroblastos/virologia , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína X Associada a bcl-2/genética
15.
Oncol Rep ; 21(6): 1381-4, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19424613

RESUMO

The prognosis of pancreatic cancer with peritoneal dissemination has not improved. The aim of this study was to clarify whether oncolytic reovirus is effective against the peritoneal dissemination of pancreatic cancer in an immunocompetent animal model. The hamster pancreatic cancer cells HaP-T1 were inoculated into the peritoneal cavity of the hamster and reovirus (1x10(8) plaque-forming units) was administered into the peritoneal cavity on days 1, 3, 5 and 7 after HaP-T1 inoculations. The number and weight of the disseminated nodules in each group were recorded. Reovirus protein in the disseminated nodules was examined by immunohistochemical staining. The tumor volumes of peritoneal dissemination in the treatment group were significantly less than those in the control group (p<0.05). In addition, the amount of ascites was decreased in the treatment group in comparison to the control group. Immunohistochemical examination revealed that reovirus replication was seen only in the disseminated nodules but not in surrounding normal tissues. There were no serious side effects observed in this study. These data suggested that intraperitoneal administration of reovirus might be an effective form of oncolytic viral therapy for peritoneal dissemination of pancreatic cancer.


Assuntos
Orthoreovirus Mamífero 3/patogenicidade , Terapia Viral Oncolítica , Neoplasias Pancreáticas/terapia , Neoplasias Peritoneais/prevenção & controle , Animais , Ascite/patologia , Ascite/prevenção & controle , Ascite/virologia , Linhagem Celular Tumoral , Cricetinae , Imunocompetência , Injeções Intraperitoneais , Masculino , Mesocricetus , Neoplasias Experimentais , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/virologia , Neoplasias Peritoneais/secundário , Neoplasias Peritoneais/virologia , Fatores de Tempo
16.
J Virol ; 83(5): 2178-87, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19109390

RESUMO

The secreted cytokine alpha/beta interferon (IFN-alpha/beta) binds its receptor to activate the Jak-STAT signal transduction pathway, leading to formation of the heterotrimeric IFN-stimulated gene factor 3 (ISGF3) transcription complex for induction of IFN-stimulated genes (ISGs) and establishment of an antiviral state. Many viruses have evolved countermeasures to inhibit the IFN pathway, thereby subverting the innate antiviral response. Here, we demonstrate that the mildly myocarditic reovirus type 1 Lang (T1L), but not the nonmyocarditic reovirus type 3 Dearing, represses IFN induction of a subset of ISGs and that this repressor function segregates with the T1L M1 gene. Concordantly, the T1L M1 gene product, mu2, dramatically inhibits IFN-beta-induced reporter gene expression. Surprisingly, T1L infection does not degrade components of the ISGF3 complex or interfere with STAT1 or STAT2 nuclear translocation as has been observed for other viruses. Instead, infection with T1L or reassortant or recombinant viruses containing the T1L M1 gene results in accumulation of interferon regulatory factor 9 (IRF9) in the nucleus. This effect has not been previously described for any virus and suggests that mu2 modulates IRF9 interactions with STATs for both ISGF3 function and nuclear export. The M1 gene is a determinant of virus strain-specific differences in the IFN response, which are linked to virus strain-specific differences in induction of murine myocarditis. We find that virus-induced myocarditis is associated with repression of IFN function, providing new insights into the pathophysiology of this disease. Together, these data provide the first report of an increase in IRF9 nuclear accumulation associated with viral subversion of the IFN response and couple virus strain-specific differences in IFN antagonism to the pathogenesis of viral myocarditis.


Assuntos
Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Interferon beta/farmacologia , Orthoreovirus Mamífero 3/patogenicidade , Orthoreovirus de Mamíferos/patogenicidade , Proteínas Virais/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Fator Gênico 3 Estimulado por Interferon/metabolismo , Interferon-alfa/farmacologia , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/metabolismo , Camundongos , Miocardite/virologia , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/metabolismo , Vírus Reordenados/genética , Vírus Reordenados/metabolismo , Vírus Reordenados/patogenicidade , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Ativação Transcricional , Proteínas Virais/genética
17.
J Neuropathol Exp Neurol ; 67(3): 231-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18344914

RESUMO

Acute flaccid paralysis (AFP) describes the loss of motor function in 1 or more limbs commonly associated with viral infection and destruction of motor neurons in the anterior horns of the spinal cord. Therapy is limited, and the development of effective treatments is hampered by a lack of experimental models. Reovirus infection of neonatal mice provides a model for the study of CNS viral infection pathogenesis. Injection of the Reovirus serot Type 3 strains Abney (T3A) or Dearing (T3D) into the hindlimb of 1-day-old mice resulted in the development of AFP in more than 90% of infected mice. Acute flaccid paralysis began in the ipsilateral hindlimb at 8 to 10 days postinfection and progressed to paraplegia 24 hours later. Paralysis correlated with injury, neuron loss, and spread of viral antigen first to the ipsilateral and then to the contralateral anterior horns. As demonstrated by the activation of caspase 3 and its colocalization with viral antigen in the anterior horn and concomitant cleavage of poly-(adenosine diphosphate-ribose) polymerase, AFP was associated with apoptosis. Calpain activity and inducible nitric oxide synthase expression were both elevated in the spinal cords of paralyzed animals. This study represents the first detailed characterization of a novel and highly efficient experimental model of virus-induced AFP that will facilitate evaluation of therapeutic strategies targeting virus-induced paralysis.


Assuntos
Doença dos Neurônios Motores/virologia , Neurônios Motores/virologia , Paralisia/virologia , Infecções por Reoviridae/patologia , Doenças da Medula Espinal/virologia , Animais , Animais Recém-Nascidos , Antígenos Virais/análise , Antígenos Virais/metabolismo , Apoptose/fisiologia , Biomarcadores/análise , Biomarcadores/metabolismo , Calpaína/análise , Calpaína/metabolismo , Caspase 3/análise , Caspase 3/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Orthoreovirus Mamífero 3/patogenicidade , Orthoreovirus Mamífero 3/fisiologia , Camundongos , Doença dos Neurônios Motores/patologia , Doença dos Neurônios Motores/fisiopatologia , Neurônios Motores/patologia , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Degeneração Neural/virologia , Óxido Nítrico Sintase Tipo II/análise , Óxido Nítrico Sintase Tipo II/metabolismo , Paralisia/patologia , Paralisia/fisiopatologia , Poli(ADP-Ribose) Polimerases/análise , Poli(ADP-Ribose) Polimerases/metabolismo , Infecções por Reoviridae/fisiopatologia , Doenças da Medula Espinal/patologia , Doenças da Medula Espinal/fisiopatologia , Febre do Nilo Ocidental/patologia , Febre do Nilo Ocidental/fisiopatologia
18.
J Virol ; 82(1): 161-72, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17959662

RESUMO

Apoptosis plays an important role in the pathogenesis of reovirus encephalitis and myocarditis in infected animals. Differences in apoptosis efficiency displayed by reovirus strains are linked to the viral mu1-encoding M2 gene segment. Studies using pharmacologic inhibitors of reovirus replication demonstrate that apoptosis induction by reovirus requires viral disassembly in cellular endosomes but not RNA synthesis. Since the mu1 protein functions to pierce endosomal membranes during this temporal window, these findings point to an important role for mu1 in activating signaling pathways that lead to apoptosis. To understand mechanisms used by mu1 to induce apoptosis, a panel of mu1 mutant viruses generated by reverse genetics was analyzed for the capacities to penetrate host cell membranes, activate proapoptotic signaling pathways, evoke cell death, and produce encephalitis in newborn mice. We found that single amino acid changes within the delta region of mu1 reduce the efficiency of membrane penetration. These mutations also diminish the capacities of reovirus to activate proapoptotic transcription factors NF-kappaB and IRF-3 and elicit apoptosis. Additionally, we observed that following intracranial inoculation, an apoptosis-deficient mu1 mutant is less virulent in newborn mice in comparison to the wild-type virus. These results indicate a critical function for the membrane penetration activity of mu1 in evoking prodeath signaling pathways that regulate reovirus pathogenesis.


Assuntos
Apoptose , Proteínas do Capsídeo/fisiologia , Encefalite/virologia , Membranas Intracelulares/virologia , Orthoreovirus Mamífero 3/patogenicidade , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Humanos , Fator Regulador 3 de Interferon/metabolismo , Membranas Intracelulares/química , Camundongos , Mutagênese , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , NF-kappa B/metabolismo , Infecções por Reoviridae/virologia , Virulência
19.
J Virol ; 81(24): 13668-80, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17942530

RESUMO

Viral myocarditis is an important human disease, and reovirus-induced murine myocarditis provides an excellent model system for study. Cardiac myocytes, like neurons in the central nervous system, are not replenished, yet there is no cardiac protective equivalent to the blood-brain barrier. Thus, cardiac myocytes may have evolved a unique antiviral response relative to readily replenished cell types, such as cardiac fibroblasts. Our previous comparisons of these two cell types revealed a conundrum: reovirus T3D induces more beta-interferon (IFN-beta) mRNA in cardiac myocytes, yet there is a greater induction of IFN-stimulated genes (ISGs) in cardiac fibroblasts. Here, we investigated possible underlying molecular determinants. We found that greater basal expression of IFN-beta in cardiac myocytes results in greater basal activated nuclear STAT1 and STAT2 and greater basal ISG mRNA expression and provides greater basal antiviral protection relative to cardiac fibroblasts. Conversely, cardiac fibroblasts express greater basal IFN-alpha/beta receptor 1 (IFNAR1) and greater basal cytoplasmic Jak1, Tyk2, STAT2, and IRF9, leading to a greater increase in reovirus T3D- or IFN-induced nuclear activated STAT1 and STAT2 and greater induction of ISGs for a greater IFN-induced antiviral protection relative to cardiac myocytes. Our results suggest that high basal IFN-beta expression in cardiac myocytes prearms this vulnerable, nonreplenishable cell type, while high basal expression of IFNAR1 and latent Jak-STAT components in adjacent cardiac fibroblasts renders these cells more responsive to IFN and prevents them from inadvertently serving as a reservoir for viral replication and spread to cardiac myocytes. These studies provide the first indication of an integrated network of cell-type-specific innate immune components for organ protection.


Assuntos
Fibroblastos/imunologia , Janus Quinases/metabolismo , Miocardite/imunologia , Miocardite/virologia , Miócitos Cardíacos/imunologia , Receptor de Interferon alfa e beta/metabolismo , Fatores de Transcrição STAT/metabolismo , Animais , Células Cultivadas , Fibroblastos/virologia , Regulação da Expressão Gênica , Humanos , Interferon-alfa/genética , Interferon-alfa/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Janus Quinases/genética , Células L , Orthoreovirus Mamífero 3/patogenicidade , Camundongos , Camundongos Knockout , Miócitos Cardíacos/virologia , Orthoreovirus de Mamíferos/patogenicidade , Receptor de Interferon alfa e beta/genética , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/virologia , Fatores de Transcrição STAT/genética
20.
Virology ; 364(2): 301-16, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17451769

RESUMO

A low-copy component of mammalian reovirus particles is mu2, an 83-kDa protein encoded by the M1 viral genome segment and packaged within the viral core. Previous studies have identified mu2 as a nucleoside triphosphate phosphohydrolase (NTPase) as well as an RNA 5'-triphosphate phosphohydrolase (RTPase), putatively involved in reovirus RNA synthesis and/or 5'-capping. Other studies have identified mu2 as a microtubule-binding protein, which also associates with the viral factory matrix protein muNS and thereby anchors the factories to cellular microtubules during infections by most reovirus strains. To extend studies of mu2 functions during infection, we tested a small interfering RNA (siRNA) directed against the M1 plus-strand RNAs of reovirus strains Type 1 Lang (T1L) and Type 3 Dearing (T3D). The siRNA strongly suppressed mu2 expression by either strain and reduced infectious yields in a strain-dependent manner. This first strain difference was genetically mapped to the M1 genome segment and tentatively assigned to a single mu2 sequence polymorphism, Pro/Ser208, which also determines a T1L-T3D strain difference in microtubule association. The siRNA-based defect in mu2 expression was rescued by plasmids, containing silent mutations in the siRNA-targeted sequence, which encoded either T1L or T3D mu2, but the growth defect was rescued only by T1L mu2. This second strain difference was also mapped to Pro/Ser208, in that swapping this one residue between T1L and T3D mu2 reversed the rescue phenotypes. Thus, the T1L-T3D strain difference in mu2-microtubule association was correlated not only with the extent of reduction in infectious yields by the siRNA but also with the extent of rescue by plasmid-derived mu2. In addition, the rescue capacity of T1L mu2 was abrogated by nocodazole treatment, providing independent evidence for the importance of mu2-microtubule association in plasmid-based rescue. In two separate cases, the results revealed functional differences between virus- and plasmid-derived mu2. Ala substitutions within the NTP-binding motif of T1L mu2 also abrogated its rescue capacity, suggesting that the NTPase or RTPase activity of mu2 is additionally required for effective viral growth.


Assuntos
Orthoreovirus Mamífero 3/genética , Orthoreovirus de Mamíferos/genética , Proteínas do Core Viral/genética , Proteínas Virais/genética , Hidrolases Anidrido Ácido/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Inativação Gênica , Genes Virais , Teste de Complementação Genética , Orthoreovirus Mamífero 3/patogenicidade , Orthoreovirus Mamífero 3/fisiologia , Camundongos , Microtúbulos/metabolismo , Microtúbulos/virologia , Dados de Sequência Molecular , Nucleosídeo-Trifosfatase/metabolismo , Orthoreovirus de Mamíferos/patogenicidade , Orthoreovirus de Mamíferos/fisiologia , Plasmídeos/genética , RNA Interferente Pequeno/genética , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA