Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Curr Top Microbiol Immunol ; 309: 1-38, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16909895

RESUMO

Mammalian orthoreoviruses (reoviruses) serve as a tractable model system for studies of viral pathogenesis. Reoviruses infect virtually all mammals, but cause disease only in the very young. Prototype strains of the three reovirus serotypes differ in pathogenesis following infection of newborn mice. Reoviruses are nonenveloped, icosahedral particles that consist of ten segments of double-stranded RNA encapsidated within two protein shells, the inner core and outer capsid. High-resolution structures of individual components of the reovirus outer capsid and a single viral receptor have been solved and provide insight into the functions of these molecules in viral attachment, entry, and pathogenesis. Attachment of reovirus to target cells is mediated by the reovirus sigma1 protein, a filamentous trimer that projects from the outer capsid. Junctional adhesion molecule-A is a serotype-independent receptor for reovirus, and sialic acid is a coreceptor for serotype 3 strains. After binding to receptors on the cell surface, reovirus is internalized via receptor-mediated endocytosis. Internalization is followed by stepwise disassembly of the viral outer capsid in the endocytic compartment. Uncoating events, which require acidic pH and endocytic proteases, lead to removal of major outer-capsid protein sigma3, resulting in exposure of membrane-penetration mediator micro1 and a conformational change in attachment protein sigma1. After penetration of endosomes by uncoated particles, the transcriptionally active viral core is released into the cytoplasm, where replication proceeds. Despite major advances in defining reovirus attachment and entry mechanisms, many questions remain. Ongoing research is aimed at understanding serotype-dependent differences in reovirus tropism, viral cell-entry pathways, the individual and corporate roles of acidic pH and proteases in viral entry, and micro1 function in membrane penetration.


Assuntos
Orthoreovirus de Mamíferos/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação Microbiológicos , Proteínas do Capsídeo/química , Proteínas do Capsídeo/fisiologia , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/fisiologia , Endocitose , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulinas/química , Imunoglobulinas/fisiologia , Dados de Sequência Molecular , Ácido N-Acetilneuramínico/metabolismo , Peptídeo Hidrolases/fisiologia , Conformação Proteica , Receptores de Superfície Celular , Montagem de Vírus
2.
Histol Histopathol ; 20(3): 957-67, 2005 07.
Artigo em Inglês | MEDLINE | ID: mdl-15944946

RESUMO

Oxidative stress, primarily due to increased generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS), is a feature of many viral infections. ROS and RNS modulate the permissiveness of cells to viral replication, regulate host inflammatory and immune responses, and cause oxidative damage to both host tissue and progeny virus. The lipid-rich nervous system is particularly susceptible to lipid peroxidation, an autocatalytic process that damages lipid-containing structures and yields reactive by-products, which can covalently modify and damage cellular macromolecules. Oxidative injury is a component of acute encephalitis caused by herpes simplex virus type 1 and reovirus, neurodegenerative disease caused by human immunodeficiency virus and murine leukemia virus, and subacute sclerosing panencephalitis caused by measles virus. The extent to which oxidative damage plays a beneficial role for the host by limiting viral replication is largely unknown. An enhanced understanding of the role of oxidative damage in viral infections of the nervous system may lead to therapeutic strategies to reduce tissue damage during viral infection without impeding the host antiviral response.


Assuntos
Sistema Nervoso/virologia , Estresse Oxidativo/fisiologia , Viroses/fisiopatologia , Humanos , Modelos Biológicos , Sistema Nervoso/metabolismo , Sistema Nervoso/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Viroses/etiologia , Viroses/metabolismo
3.
J Virol ; 75(16): 7429-34, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11462015

RESUMO

Serotype 3 reoviruses inhibit cellular proliferation by inducing a G(2)/M phase cell cycle arrest. Reovirus-induced G(2)/M phase arrest requires the viral S1 gene-encoded sigma1s nonstructural protein. The G(2)-to-M transition represents a cell cycle checkpoint that is regulated by the kinase p34(cdc2). We now report that infection with serotype 3 reovirus strain Abney, but not serotype 1 reovirus strain Lang, is associated with inhibition and hyperphosphorylation of p34(cdc2). The sigma1s protein is necessary and sufficient for inhibitory phosphorylation of p34(cdc2), since a viral mutant lacking sigma1s fails to hyperphosphorylate p34(cdc2) and inducible expression of sigma1s is sufficient for p34(cdc2) hyperphosphorylation. These studies establish a mechanism by which reovirus can perturb cell cycle regulation.


Assuntos
Proteína Quinase CDC2/fisiologia , Ciclo Celular , Orthoreovirus Mamífero 3/fisiologia , Infecções por Reoviridae/virologia , Proteínas não Estruturais Virais/fisiologia , Animais , Linhagem Celular , Camundongos , Infecções por Reoviridae/patologia , Transdução de Sinais , Replicação Viral
4.
J Virol ; 75(14): 6517-26, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11413319

RESUMO

Reovirus mRNAs are efficiently translated within host cells despite the absence of 3' polyadenylated tails. The 3' nontranslated regions (3'NTRs) of reovirus mRNAs contain sequences that exhibit a high degree of gene-segment-specific conservation. To determine whether the 3'NTRs of reovirus mRNAs serve to facilitate efficient translation of viral transcripts, we used T7 RNA polymerase to express constructs engineered with full-length S4 gene cDNA or truncation mutants lacking sequences in the 3'NTR. Full-length and truncated s4 mRNAs were translated using rabbit reticulocyte lysates, and translation product sigma3 was quantitated by phosphorimager analysis. In comparison to full-length s4 mRNA, translation of the s4 mRNA lacking the 3'NTR resulted in a 20 to 50% decrease in sigma3 produced. Addition to translation reactions of an RNA oligonucleotide corresponding to the S4 3'NTR significantly enhanced translation of full-length s4 mRNA but had no effect on s4 mRNA lacking 3'NTR sequences. Translation of s4 mRNAs with smaller deletions within the 3'NTR identified a discrete region capable of translational enhancement and a second region capable of translational repression. Differences in translational efficiency of full-length and deletion-mutant mRNAs were independent of RNA stability. Protein complexes in reticulocyte lysates that specifically interact with the S4 3'NTR were identified by RNA mobility shift assays. RNA oligonucleotides lacking either enhancer or repressor sequences did not efficiently compete the binding of these complexes to full-length 3'NTR. These results indicate that the reovirus S4 gene 3'NTR contains a translational operator sequence that serves to regulate translational efficiency of the s4 mRNA. Moreover, these findings suggest that cellular proteins interact with reovirus 3'NTR sequences to regulate translation of the nonpolyadenylated reovirus mRNAs.


Assuntos
Regiões 3' não Traduzidas/metabolismo , Orthoreovirus Mamífero 3/metabolismo , Biossíntese de Proteínas , Regiões 3' não Traduzidas/genética , Animais , Linhagem Celular , RNA Polimerases Dirigidas por DNA , Deleção de Genes , Orthoreovirus Mamífero 3/genética , Mutação , Ligação Proteica , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética
5.
J Virol ; 75(14): 6625-34, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11413330

RESUMO

Reovirus virions are nonenveloped icosahedral particles consisting of two concentric protein shells, termed outer capsid and core. Outer-capsid protein sigma1 is the viral attachment protein and binds carbohydrate molecules on the surface of host cells. Monoclonal antibody (MAb) 4F2, which is specific for outer-capsid protein sigma3, blocks the binding of sigma1 protein to sialic acid and inhibits reovirus-induced hemagglutination (HA). To determine whether MAb 4F2 inhibits HA by altering sigma1-sigma3 interactions or by steric hindrance, we analyzed the effect of 4F2 immunoglobulin G (IgG) and Fab fragments (Fabs) on HA induced by reovirus strain type 3 Dearing (T3D). The concentration of 4F2 IgG sufficient to inhibit T3D-induced HA was 12.5 microg per ml, whereas that of Fabs was >200 microg per ml. Dynamic light scattering analysis showed that at the concentration of IgG sufficient to inhibit HA, virion-antibody complexes were monodispersed and not aggregated. The affinity of 4F2 Fabs for T3D virions was only threefold less than that of intact IgG, which suggests that differences in HA inhibition titer exhibited by 4F2 IgG and Fabs are not attributable to differences in the affinity of these molecules for T3D virions. We used cryoelectron microscopy and three-dimensional image analysis to visualize T3D virions alone and in complex with either IgG or Fabs of MAb 4F2. IgG and Fabs bind the same site at the distal portion of sigma3, and binding of IgG and Fabs induces identical conformational changes in outer-capsid proteins sigma3 and mu1. These results suggest that MAb 4F2 inhibits reovirus binding to sialic acid by steric hindrance and provide insight into the conformational flexibility of reovirus outer-capsid proteins.


Assuntos
Anticorpos Monoclonais/imunologia , Capsídeo/imunologia , Hemaglutininas Virais/imunologia , Reoviridae/imunologia , Animais , Anticorpos Antivirais/imunologia , Capsídeo/ultraestrutura , Linhagem Celular , Membrana Celular/química , Membrana Celular/virologia , Testes de Hemaglutinação , Hemaglutininas Virais/ultraestrutura , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Conformação Molecular , Testes de Neutralização , Relação Estrutura-Atividade
6.
J Virol ; 75(11): 5335-42, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333914

RESUMO

Mammalian reoviruses, prototype members of the Reoviridae family of nonenveloped double-stranded RNA viruses, use at least three proteins--sigma1, mu1, and sigma3--to enter host cells. sigma1, a major determinant of cell tropism, mediates viral attachment to cellular receptors. Studies of sigma1 functions in reovirus entry have been restricted by the lack of methodologies to produce infectious virions containing engineered mutations in viral proteins. To mitigate this problem, we produced virion-like particles by "recoating" genome-containing core particles that lacked sigma1, mu1, and sigma3 with recombinant forms of these proteins in vitro. Image reconstructions from cryoelectron micrographs of the recoated particles revealed that they closely resembled native virions in three-dimensional structure, including features attributable to sigma1. The recoated particles bound to and infected cultured cells in a sigma1-dependent manner and were approximately 1 million times as infectious as cores and 0.5 times as infectious as native virions. Experiments with recoated particles containing recombinant sigma1 from either of two different reovirus strains confirmed that differences in cell attachment and infectivity previously observed between those strains are determined by the sigma1 protein. Additional experiments showed that recoated particles containing sigma1 proteins with engineered mutations can be used to analyze the effects of such mutations on the roles of particle-bound sigma1 in infection. The results demonstrate a powerful new system for molecular genetic dissections of sigma1 with respect to its structure, assembly into particles, and roles in entry.


Assuntos
Proteínas do Capsídeo , Capsídeo/biossíntese , Reoviridae/patogenicidade , Proteínas Virais/genética , Replicação Viral , Baculoviridae , Capsídeo/genética , Capsídeo/ultraestrutura , Linhagem Celular , Microscopia Crioeletrônica , Células Eucarióticas/virologia , Testes de Hemaglutinação , Hemaglutininas Virais/biossíntese , Hemaglutininas Virais/genética , Técnicas In Vitro , Microscopia Eletrônica , Proteínas Recombinantes/biossíntese , Reoviridae/genética , Reoviridae/ultraestrutura , Proteínas Virais/biossíntese , Montagem de Vírus
7.
J Virol ; 75(9): 4029-39, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11287552

RESUMO

Reovirus induces apoptosis in cultured cells and in vivo. Genetic studies indicate that the efficiency with which reovirus strains induce apoptosis is determined by the viral S1 gene, which encodes attachment protein sigma1. However, the biochemical properties of sigma1 that influence apoptosis induction are unknown. To determine whether the capacity of sigma1 to bind cell surface sialic acid determines the magnitude of the apoptotic response, we used isogenic reovirus mutants that differ in the capacity to engage sialic acid. We found that T3SA+, a virus capable of binding sialic acid, induces high levels of apoptosis in both HeLa cells and L cells. In contrast, non-sialic-acid-binding strain T3SA- induces little or no apoptosis in these cell types. Differences in the capacity of T3SA- and T3SA+ to induce apoptosis are not due to differences in viral protein synthesis or production of viral progeny. Removal of cell surface sialic acid with neuraminidase abolishes the capacity of T3SA+ to induce apoptosis. Similarly, incubation of T3SA+ with sialyllactose, a trisaccharide comprised of lactose and sialic acid, blocks apoptosis. These findings demonstrate that reovirus binding to cell surface sialic acid is a critical requirement for the efficient induction of apoptosis and suggest that virus receptor utilization plays an important role in regulating cell death.


Assuntos
Apoptose , Proteínas do Capsídeo , Ácido N-Acetilneuramínico/metabolismo , Receptores Virais/metabolismo , Reoviridae/fisiologia , Proteínas Virais/metabolismo , Animais , Membrana Celular/virologia , Células HeLa , Humanos , Células L , Camundongos , NF-kappa B/metabolismo , Neuraminidase/metabolismo , Coelhos , Reoviridae/genética , Reoviridae/crescimento & desenvolvimento , Reoviridae/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética , Proteínas Virais/fisiologia
8.
J Virol ; 75(7): 3197-206, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11238846

RESUMO

Reovirus virions are internalized into cells by receptor-mediated endocytosis. Within the endocytic compartment, the viral outer capsid undergoes acid-dependent proteolysis leading to degradation of sigma3 protein and proteolytic cleavage of micro1/micro1C protein. E64 is a specific inhibitor of cysteine-containing proteases that blocks disassembly of reovirus virions. To identify domains in reovirus proteins that influence susceptibility to E64-mediated inhibition of disassembly, we selected variant viruses by serial passage of strain type 3 Dearing (T3D) in murine L929 cells treated with E64. E64-adapted variant viruses (D-EA viruses) produced 7- to 17-fold-greater yields than T3D did after infection of cells treated with 100 microM E64. Viral genes that segregate with growth of D-EA viruses in the presence of E64 were identified by using reassortant viruses isolated from independent crosses of E64-sensitive strain type 1 Lang and two prototype D-EA viruses. Growth of reassortant viruses in the presence of E64 segregated with the S4 gene, which encodes outer-capsid protein sigma3. Sequence analysis of S4 genes of three D-EA viruses isolated from independent passage series revealed a common tyrosine-to-histidine mutation at amino acid 354 in the deduced amino acid sequence of sigma3. Proteolysis of D-EA virions by endocytic protease cathepsin L occurred with faster kinetics than proteolysis of wild-type T3D virions. Treatment of D-EA virions, but not T3D virions, with cathepsin D resulted in proteolysis of sigma3, a property that also was found to segregate with the D-EA S4 gene. These results indicate that a region in sigma3 protein containing amino acid 354 influences susceptibility of sigma3 to proteolysis during reovirus disassembly.


Assuntos
Proteínas do Capsídeo , Capsídeo/fisiologia , Inibidores de Cisteína Proteinase/farmacologia , Endopeptidases , Leucina/análogos & derivados , Leucina/farmacologia , Proteínas de Ligação a RNA , Reoviridae/fisiologia , Adaptação Fisiológica , Animais , Capsídeo/química , Catepsina D/farmacologia , Catepsina L , Catepsinas/farmacologia , Cisteína Endopeptidases , Células L , Camundongos , Mutação , Reoviridae/efeitos dos fármacos , Reoviridae/genética , Relação Estrutura-Atividade , Montagem de Vírus
9.
Cell ; 104(3): 441-51, 2001 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-11239401

RESUMO

Virus attachment to cells plays an essential role in viral tropism and disease. Reovirus serotypes 1 and 3 differ in the capacity to target distinct cell types in the murine nervous system and in the efficiency to induce apoptosis. The binding of viral attachment protein sigma1 to unidentified receptors controls these phenotypes. We used expression cloning to identify junction adhesion molecule (JAM), an integral tight junction protein, as a reovirus receptor. JAM binds directly to sigma1 and permits reovirus infection of nonpermissive cells. Ligation of JAM is required for reovirus-induced activation of NF-kappaB and apoptosis. Thus, reovirus interaction with cell-surface receptors is a critical determinant of both cell-type specific tropism and virus-induced intracellular signaling events that culminate in cell death.


Assuntos
Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/fisiologia , Reoviridae/química , Animais , Apoptose , Células COS , Células CACO-2 , Morte Celular , Embrião de Galinha , Clonagem Molecular , DNA Complementar/metabolismo , Fibroblastos/metabolismo , Biblioteca Gênica , Células HeLa , Humanos , Moléculas de Adesão Juncional , Camundongos , Modelos Biológicos , NF-kappa B/metabolismo , Fenótipo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
10.
J Virol ; 75(3): 1459-75, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11152519

RESUMO

Progeny virions of mammalian reoviruses are assembled in the cytoplasm of infected cells at discrete sites termed viral inclusions. Studies of temperature-sensitive (ts) mutant viruses indicate that nonstructural protein sigmaNS and core protein mu2 are required for synthesis of double-stranded (ds) RNA, a process that occurs at sites of viral assembly. We used confocal immunofluorescence microscopy and ts mutant reoviruses to define the roles of sigmaNS and mu2 in viral inclusion formation. In cells infected with wild-type (wt) reovirus, sigmaNS and mu2 colocalize to large, perinuclear structures that correspond to viral inclusions. In cells infected at a nonpermissive temperature with sigmaNS-mutant virus tsE320, sigmaNS is distributed diffusely in the cytoplasm and mu2 is contained in small, punctate foci that do not resemble viral inclusions. In cells infected at a nonpermissive temperature with mu2-mutant virus tsH11.2, mu2 is distributed diffusely in the cytoplasm and the nucleus. However, sigmaNS localizes to discrete structures in the cytoplasm that contain other viral proteins and are morphologically indistinguishable from viral inclusions seen in cells infected with wt reovirus. Examination of cells infected with wt reovirus over a time course demonstrates that sigmaNS precedes mu2 in localization to viral inclusions. These findings suggest that viral RNA-protein complexes containing sigmaNS nucleate sites of viral replication to which other viral proteins, including mu2, are recruited to commence dsRNA synthesis.


Assuntos
Corpos de Inclusão Viral , Reoviridae/fisiologia , Proteínas não Estruturais Virais/fisiologia , Montagem de Vírus , Animais , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Core Viral/fisiologia
11.
J Biol Chem ; 276(3): 2200-11, 2001 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-11054410

RESUMO

Many serotype 3 reoviruses bind to two different host cell molecules, sialic acid and an unidentified protein, using discrete receptor-binding domains in viral attachment protein, final sigma1. To determine mechanisms by which these receptor-binding events cooperate to mediate cell attachment, we generated isogenic reovirus strains that differ in the capacity to bind sialic acid. Strain SA+, but not SA-, bound specifically to sialic acid on a biosensor chip with nanomolar avidity. SA+ displayed 5-fold higher avidity for HeLa cells when compared with SA-, although both strains recognized the same proteinaceous receptor. Increased avidity of SA+ binding was mediated by increased k(on). Neuraminidase treatment to remove cell-surface sialic acid decreased the k(on) of SA+ to that of SA-. Increased k(on) of SA+ enhanced an infectious attachment process, since SA+ was 50-100-fold more efficient than SA- at infecting HeLa cells in a kinetic fluorescent focus assay. Sialic acid binding was operant early during SA+ attachment, since the capacity of soluble sialyllactose to inhibit infection decreased rapidly during the first 20 min of adsorption. These results indicate that reovirus binding to sialic acid enhances virus infection through adhesion of virus to the cell surface where access to a proteinaceous receptor is thermodynamically favored.


Assuntos
Fusão de Membrana , Ácido N-Acetilneuramínico/metabolismo , Receptores Virais/metabolismo , Reoviridae/fisiologia , Animais , Técnicas Biossensoriais , Linhagem Celular , Humanos , Cinética , Camundongos , Ligação Proteica , Ressonância de Plasmônio de Superfície
13.
Virology ; 278(2): 309-21, 2000 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-11118355

RESUMO

The pathological consequences of herpes simplex virus type 1 (HSV-1) latency in the nervous system are not well understood. To determine whether acute and latent HSV-1 infections of the nervous system are associated with oxidative damage, mice were inoculated with HSV-1 by the corneal route, and the extent of viral infection and oxidative damage in trigeminal ganglia and brain was assessed at 7, 90, and 220 days after inoculation. Abundant HSV-1 protein expression in the nervous system was observed in neurons and non-neuronal cells at 7 days after inoculation, consistent with viral replication and spread through the trigeminal and olfactory systems. Acute HSV-1 infection was associated with focal, neuronal and non-neuronal 4-hydroxy-2-nonenal- and 8-hydroxyguanosine-specific immunoreactivity, indicating oxidative damage. Rare HSV-1 antigen-positive cells were observed at 90 and 220 days after inoculation; however, widespread HSV-1 latency-associated transcript expression was detected, consistent with latent HSV-1 infection in the nervous system. HSV-1 latency was detected predominantly in the trigeminal ganglia, brainstem, olfactory bulbs, and temporal cortex. Latent HSV-1 infection was associated with focal chronic inflammation and consistently detectable evidence of oxidative damage involving primarily neurons. These results indicate that both acute and latent HSV-1 infections in the murine nervous system are associated with oxidative damage.


Assuntos
Encéfalo/virologia , Herpes Simples/patologia , Herpesvirus Humano 1/fisiologia , Neurônios/virologia , Gânglio Trigeminal/virologia , Latência Viral , Animais , Apoptose , Encéfalo/patologia , Tronco Encefálico/patologia , Tronco Encefálico/virologia , Feminino , Herpes Simples/virologia , Marcação In Situ das Extremidades Cortadas , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/patologia , Bulbo Olfatório/patologia , Bulbo Olfatório/virologia , Lobo Temporal/patologia , Lobo Temporal/virologia , Gânglio Trigeminal/patologia , Ensaio de Placa Viral
14.
Virology ; 275(1): 125-32, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11017794

RESUMO

The rotavirus NSP4 protein is cytotoxic when transiently expressed in cells and is capable of inducing secretory diarrhea in neonatal mice. NSP4 consists of 175 amino acids, and sequences important for its toxic effects have been mapped to the carboxy-terminal half of the protein. In this report, we compared NSP4-encoding nucleotide sequences recovered from cell lines engineered to express NSP4 from human rotavirus strain Wa with NSP4 sequences recovered from cells persistently infected with either Wa or simian rotavirus strain SA11. In cells stably transfected with Wa NSP4, we found that proline(138) was changed to either serine or threonine. However, in cells persistently infected with SA11, we found that phenylalanine(33) was changed to leucine, and in cells persistently infected with Wa, no changes were observed in NSP4. Expression of Wa NSP4 in Caco-2 cells resulted in increased cell-doubling times and decreased cell viability in comparison to cells expressing NSP4-serine(138) or NSP4-threonine(138). This result suggests that sequence polymorphism at residue 138 in Wa NSP4 influences the cytotoxicity of the protein. Therefore, mutations in the carboxy-terminal half of NSP4 are selected when NSP4 is expressed in cells in the absence of other viral proteins, but not in the context of viral replication. These findings suggest that cytotoxic functions of NSP4 are not operant during natural rotavirus infection.


Assuntos
Glicoproteínas/genética , Glicoproteínas/metabolismo , Mutação/genética , Rotavirus/patogenicidade , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Substituição de Aminoácidos/genética , Animais , Células CACO-2 , Divisão Celular , Linhagem Celular , Sobrevivência Celular , Análise Mutacional de DNA , Expressão Gênica , Glicoproteínas/biossíntese , Glicoproteínas/química , Humanos , Fenótipo , Polimorfismo Genético/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Rotavirus/genética , Rotavirus/fisiologia , Toxinas Biológicas , Transfecção , Proteínas não Estruturais Virais/biossíntese , Proteínas não Estruturais Virais/química , Virulência
15.
J Virol ; 74(20): 9562-70, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11000227

RESUMO

Serotype-specific differences in the capacity of reovirus strains to inhibit proliferation of murine L929 cells correlate with the capacity to induce apoptosis. The prototype serotype 3 reovirus strains Abney (T3A) and Dearing (T3D) inhibit cellular proliferation and induce apoptosis to a greater extent than the prototype serotype 1 reovirus strain Lang (T1L). We now show that reovirus-induced inhibition of cellular proliferation results from a G(2)/M cell cycle arrest. Using T1L x T3D reassortant viruses, we found that strain-specific differences in the capacity to induce G(2)/M arrest, like the differences in the capacity to induce apoptosis, are determined by the viral S1 gene. The S1 gene is bicistronic, encoding the viral attachment protein sigma1 and the nonstructural protein sigma1s. A sigma1s-deficient reovirus strain, T3C84-MA, fails to induce G(2)/M arrest, yet retains the capacity to induce apoptosis, indicating that sigma1s is required for reovirus-induced G(2)/M arrest. Expression of sigma1s in C127 cells increases the percentage of cells in the G(2)/M phase of the cell cycle, supporting a role for this protein in reovirus-induced G(2)/M arrest. Inhibition of reovirus-induced apoptosis failed to prevent virus-induced G(2)/M arrest, indicating that G(2)/M arrest is not the result of apoptosis related DNA damage and suggests that these two processes occur through distinct pathways.


Assuntos
Apoptose , Proteínas do Capsídeo , Fase G2 , Mitose , Reoviridae/fisiologia , Proteínas Virais/fisiologia , Animais , Divisão Celular , Células HeLa , Humanos , Camundongos
16.
J Virol ; 74(18): 8472-9, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10954547

RESUMO

The reovirus attachment protein, sigma1, is responsible for strain-specific patterns of viral tropism in the murine central nervous system and receptor binding on cultured cells. The sigma1 protein consists of a fibrous tail domain proximal to the virion surface and a virion-distal globular head domain. To better understand mechanisms of reovirus attachment to cells, we conducted studies to identify the region of sigma1 that binds cell surface carbohydrate. Chimeric and truncated sigma1 proteins derived from prototype reovirus strains type 1 Lang (T1L) and type 3 Dearing (T3D) were expressed in insect cells by using a baculovirus vector. Assessment of expressed protein susceptibility to proteolytic cleavage, binding to anti-sigma1 antibodies, and oligomerization indicates that the chimeric and truncated sigma1 proteins are properly folded. To assess carbohydrate binding, recombinant sigma1 proteins were tested for the capacity to agglutinate mammalian erythrocytes and to bind sialic acid presented on glycophorin, the cell surface molecule bound by type 3 reovirus on human erythrocytes. Using a panel of two wild-type and ten chimeric and truncated sigma1 proteins, the sialic acid-binding domain of type 3 sigma1 was mapped to a region of sequence proposed to form the more amino terminal of two predicted beta-sheet structures in the tail. This unit corresponds to morphologic region T(iii) observed in computer-processed electron micrographs of sigma1 protein purified from virions. In contrast, the homologous region of T1L sigma1 sequence was not implicated in carbohydrate binding; rather, sequences in the distal portion of the tail known as the neck were required. Results of these studies demonstrate that a functional receptor-binding domain, which uses sialic acid as its ligand, is contained within morphologic region T(iii) of the type 3 sigma1 tail. Furthermore, our findings indicate that T1L and T3D sigma1 proteins contain different arrangements of receptor-binding domains.


Assuntos
Proteínas do Capsídeo , Metabolismo dos Carboidratos , Orthoreovirus Mamífero 3/metabolismo , Orthoreovirus/metabolismo , Receptores Virais/metabolismo , Proteínas Virais/metabolismo , Animais , Baculoviridae/genética , Linhagem Celular , Glicoforinas/química , Hemaglutinação , Humanos , Insetos/citologia , Orthoreovirus Mamífero 3/química , Ácido N-Acetilneuramínico/química , Orthoreovirus/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/química
18.
Virology ; 272(1): 16-26, 2000 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-10873745

RESUMO

Reovirus infection induces the formation of large cytoplasmic inclusions that serve as the major site of viral assembly. Reovirus strains type 3 Dearing (T3D) and type 1 Lang (T1L) differ in the rate of inclusion formation in L929 cells. The median time of inclusion formation is 18 h in cells infected with T3D and 39 h in cells infected with T1L. Using reassortant viruses that contain combinations of gene segments derived from T1L and T3D, we found that the M1 gene, which encodes the mu2 protein, is the primary determinant of the rate of inclusion formation. The S3 gene, which encodes the nonstructural protein sigmaNS, plays a secondary role in this process. The subcellular location of the mu2 protein was determined by confocal laser scanning microscopy using dual-fluorescence labeling of mu2 and the outer-capsid protein mu1/mu1C. In virus-infected cells, mu2 protein colocalized with other viral proteins in inclusions and was also distributed diffusely in the cytoplasm and nucleus. Expression of recombinant T1L and T3D mu2 proteins resulted in the formation of protein complexes resembling inclusions in both the cytoplasm and the nucleus with kinetics that reflected the strain of origin. The median time of mu2 protein complex formation was 22 h in cells transfected with the T3D M1 gene and 43 h in cells transfected with the T1L M1 gene. These findings suggest that the mu2 protein influences the rate of inclusion formation and contributes to inclusion morphogenesis. The requirement of mu2 protein in inclusion formation was tested by determining the subcellular localization of mu2 in cells infected with temperature-sensitive (ts) mutants that are defective in viral assembly. In contrast to infection with wild-type virus, mu2 did not colocalize with mu1/mu1C protein in subcellular structures that formed in cells infected at nonpermissive temperature with ts mutants tsH11.2, tsC447, and tsG453 with mutations in the M1, S2, and S4 genes, respectively. These results suggest that despite the role of the mu2 protein in controlling the rate of inclusion formation, this process is a concerted function of several reovirus proteins.


Assuntos
Proteínas do Capsídeo , Corpos de Inclusão Viral/metabolismo , Proteínas de Ligação a RNA , Reoviridae/classificação , Reoviridae/fisiologia , Proteínas Virais/metabolismo , Animais , Western Blotting , Capsídeo/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Citoplasma/metabolismo , Citoplasma/virologia , Fibroblastos/citologia , Fibroblastos/virologia , Imunofluorescência , Cinética , Células L , Camundongos , Mutação/genética , Ligação Proteica , Vírus Reordenados/genética , Vírus Reordenados/metabolismo , Vírus Reordenados/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reoviridae/genética , Reoviridae/metabolismo , Temperatura , Transfecção , Proteínas Virais/genética , Montagem de Vírus
19.
J Virol ; 74(7): 2981-9, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10708412

RESUMO

Reovirus infection induces apoptosis in cultured cells and in vivo. To identify host cell factors that mediate this response, we investigated whether reovirus infection alters the activation state of the transcription factor nuclear factor kappa B (NF-kappaB). As determined in electrophoretic mobility shift assays, reovirus infection of HeLa cells leads to nuclear translocation of NF-kappaB complexes containing Rel family members p50 and p65. Reovirus-induced activation of NF-kappaB DNA-binding activity correlated with the onset of NF-kappaB-directed transcription in reporter gene assays. Three independent lines of evidence indicate that this functional form of NF-kappaB is required for reovirus-induced apoptosis. First, treatment of reovirus-infected HeLa cells with a proteasome inhibitor prevents NF-kappaB activation following infection and substantially diminishes reovirus-induced apoptosis. Second, transient expression of a dominant-negative form of IkappaB that constitutively represses NF-kappaB activation significantly reduces levels of apoptosis triggered by reovirus infection. Third, mutant cell lines deficient for either the p50 or p65 subunits of NF-kappaB are resistant to reovirus-induced apoptosis compared with cells expressing an intact NF-kappaB signaling pathway. These findings indicate that NF-kappaB plays a significant role in the mechanism by which reovirus induces apoptosis in susceptible host cells.


Assuntos
Apoptose/fisiologia , NF-kappa B/metabolismo , Reoviridae/fisiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Cisteína Endopeptidases/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Células HeLa , Humanos , Camundongos , Complexos Multienzimáticos/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma , Replicação Viral
20.
J Neuropathol Exp Neurol ; 58(12): 1269-75, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10604752

RESUMO

Several lines of evidence point to inflammation and increased oxidant injury in brain regions of patients with Alzheimer disease (AD). Prostaglandin H synthase (PGHS) catalyzes the limiting step in prostaglandin synthesis and generates a potent oxidizing agent as by-product. One form of PGHS, PGHS-2, is induced by pro-inflammatory signals; thus leading to the 2-step hypothesis that pro-inflammatory signals in AD brain induce PGHS-2 that in turn contributes to brain oxidant injury. Here we have tested directly this 2-step hypothesis in a murine reovirus type 3 encephalitis model by measuring cerebral PGHS activity and quantifying oxidant injury. Our results showed a robust chronic inflammatory infiltrate and a 2-fold increase in PGHS activity in encephalitic mice compared with controls. Despite these changes, there was no significant increase in F2-isoprostanes or F4-neuroprostanes, accurate in vivo biomarkers of oxidant injury, and only minimal accumulation of protein adducts from the lipid peroxidation product 4-hydroxy-2-nonenal in the most intensely inflamed brain regions. These results challenge the proposal of others that pro-inflammatory induction of PGHS activity significantly contributes to oxidant injury in brain.


Assuntos
Encéfalo/metabolismo , Encefalite Viral/metabolismo , Encefalite Viral/virologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Infecções por Reoviridae , Aldeídos/metabolismo , Animais , Linhagem Celular , Encefalite Viral/enzimologia , Encefalite Viral/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Peróxidos Lipídicos/metabolismo , Camundongos , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA