Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Vet Med Sci ; 78(1): 153-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26321298

RESUMO

The effect of environmental acidification on Ibaraki virus (IBAV) infection was tested using endosomal inhibitory chemicals and low pH treatment. Treatment of target cells with endosomal inhibitors significantly decreased the progeny virus production. IBAV outer capsid proteins, VP5 and VP2, were removed from virion when purified IBAV was exposed to low pH environment. Further experiment showed that the exposure to low pH buffer facilitated IBAV infection when the cellular endosomal pathway was impaired by bafilomycin A1. Results obtained in this study suggest that acidic environment is essential to initiate IBAV infection.


Assuntos
Orbivirus/fisiologia , Infecções por Reoviridae/fisiopatologia , Animais , Linhagem Celular , Cricetinae , Endossomos/efeitos dos fármacos , Endossomos/fisiologia , Endossomos/virologia , Concentração de Íons de Hidrogênio , Pulmão/citologia , Pulmão/virologia , Macrolídeos/farmacologia , Infecções por Reoviridae/virologia
2.
Arch Virol ; 160(12): 3001-10, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26350773

RESUMO

p17 is a nonstructural protein of avian reovirus (ARV) that induces autophagy in infected cells. In the present study, we investigated the effect of p17 and its nuclear localization signal (NLS) on autophagy and viral replication. When Vero cells and DF1 cells were transfected with mutant p17 in which lysine (K) at position 122 and arginine (R) at position 123 were mutated to alanine (A), the expression level of LC3 II decreased dramatically after transfection. The expression of the polypeptide encompassing the first 103 amino acids of p17, a region that did not contain the NLS, did not have a significant effect on autophagy. Moreover, when cells overexpressing mutant p17 were infected with the ARV GX2010/1 strain, the viral titer was significantly decreased compared with the expression of wild-type p17. In general, the NLS of p17 facilitates the induction of autophagy and is correlated with an increase in virus production.


Assuntos
Autofagia , Núcleo Celular/virologia , Orthoreovirus Aviário/fisiologia , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/veterinária , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Galinhas , Chlorocebus aethiops , Sinais de Localização Nuclear , Orthoreovirus Aviário/genética , Doenças das Aves Domésticas/fisiopatologia , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Células Vero , Proteínas não Estruturais Virais/genética
3.
Arch Virol ; 160(7): 1679-85, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25925704

RESUMO

Avian reovirus (ARV)-induced apoptosis contributes to the pathogenesis of reovirus in infected chickens. However, methods for effectively reducing ARV-triggered apoptosis remain to be explored. Here, we show that pretreatment with chloroquine (CQ) or E64d plus pepstatin A decreases ARV-mediated apoptosis in chicken DF-1 cells. By acting as autophagy inhibitors, CQ and E64d plus pepstatin A increase microtubule-associated protein 1 light chain 3-II (LC3II) accumulation in ARV-infected cells, which results in decreased ARV protein synthesis and virus yield and thereby contributes to the reduction of apoptosis. Furthermore, ARV-mediated apoptosis in the bursa, heart and intestines of chicken embryos is attenuated by CQ and E64d plus pepstatin A treatment. Importantly, treatment with these autophagy inhibitors increases the survival of infected chicken embryos. Together, our data suggest that pharmacological inhibition of autophagy might represent a novel strategy for reducing ARV-mediated apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Cloroquina/administração & dosagem , Leucina/análogos & derivados , Orthoreovirus Aviário/fisiologia , Pepstatinas/administração & dosagem , Doenças das Aves Domésticas/fisiopatologia , Infecções por Reoviridae/veterinária , Animais , Embrião de Galinha , Galinhas , Leucina/administração & dosagem , Orthoreovirus Aviário/efeitos dos fármacos , Doenças das Aves Domésticas/tratamento farmacológico , Doenças das Aves Domésticas/embriologia , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/tratamento farmacológico , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia
4.
Arch Virol ; 160(6): 1449-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25854689

RESUMO

Avian reovirus (ARV) causes viral arthritis, chronic respiratory diseases, retarded growth and malabsorption syndrome. It is well established that the ARV sigma-C protein induces apoptosis in host cells. However, the underlying molecular mechanism of this induction is still unclear. We report here the identification of eukaryotic elongation factor 1 alpha 1 (EEF1A1) as the interacting partner of σC. We found that σC-induced apoptosis in DF-1 cells could be completely abolished by knockdown of EEF1A1 by siRNA. Furthermore, knockdown of EEF1A1 markedly reduced ARV-induced apoptosis associated with decreased caspase-9 and -3 activation and cytochrome C release, leading to increased ARV growth in host cells. Thus, EEF1A1 plays a critical role in σC-induced apoptosis and inhibition of viral growth.


Assuntos
Apoptose , Proteínas do Capsídeo/fisiologia , Fator de Iniciação 1 em Eucariotos/fisiologia , Orthoreovirus Aviário/fisiologia , Infecções por Reoviridae/fisiopatologia , Animais , Apoptose/fisiologia , Western Blotting , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular , Embrião de Galinha/virologia , Imunofluorescência , Células HEK293/virologia , Humanos , Imunoprecipitação , Microscopia Confocal , Orthoreovirus Aviário/crescimento & desenvolvimento , Fator 1 de Elongação de Peptídeos/fisiologia , Infecções por Reoviridae/veterinária , Infecções por Reoviridae/virologia , Técnicas do Sistema de Duplo-Híbrido
5.
Apoptosis ; 20(4): 481-90, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25576194

RESUMO

In this study the mechanism of avian reovirus (ARV) S1133-induced pathogenesis was investigated, with a focus on the contribution of ER stress to apoptosis. Our results showed that upregulation of the ER stress response protein, as well as caspase-3 activation, occurred in ARV S1133-infected cultured cells and in SPF White Leghorn chicks organs. Upon infection, Bim was translocated specifically to the ER, but not mitochondria, in the middle to late infectious stages. In addition, ARV S1133 induced JNK phosphorylation and promoted JNK-Bim complex formation, which correlated with the Bim translocation and apoptosis induction that was observed at the same time point. Knockdown of BiP/GRP78 by siRNA and inhibition of BiP/GRP78 using EGCG both abolished the formation of the JNK-Bim complex, caspase-3 activation, and subsequent apoptosis induction by ARV S1133 efficiently. These results suggest that BiP/GRP78 played critical roles and works upstream of JNK-Bim in response to the ARV S1133-mediated apoptosis process.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Membrana/metabolismo , Orthoreovirus Aviário/fisiologia , Doenças das Aves Domésticas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/veterinária , Animais , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Caspase 3/metabolismo , Galinhas , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/genética , Proteínas de Membrana/genética , Orthoreovirus Aviário/genética , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/fisiopatologia , Doenças das Aves Domésticas/virologia , Transporte Proteico , Proteínas Proto-Oncogênicas/genética , Infecções por Reoviridae/genética , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Transdução de Sinais
6.
J Neurovirol ; 21(1): 1-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25604493

RESUMO

Apoptosis, or programmed cell death, is a fundamental and widespread cell biological process that is distinct from cell necrosis and can be induced by a wide variety of stimuli including viral infections. Apoptosis may occur via either the intrinsic or extrinsic pathways and confers several advantages to the virally infected host including the prevention of further viral propagation and the potential inhibition and resolution of inflammatory processes. Several viruses have been shown to have the capacity to induce apoptosis in susceptible cells including herpes simplex virus, Varicella-zoster virus, rabies virus, human immunodeficiency virus, and reovirus. Apoptosis has also been observed in human African trypanosomiasis which is an infection caused by a protozoan parasite. The mechanisms leading to apoptosis may differ depending on the type of infection. Apoptosis has been reported in several neurodegenerative diseases and also psychiatric disorders but the true clinical significance of such observations is not certain, and, though interesting, it is very difficult to ascribe causation in these conditions. The presence of inflammation in the central nervous system in any neurological condition, including those associated with a viral infection, is not necessarily an absolute marker of serious disease and the notion of 'good' versus 'bad' inflammation is considered to be valid in some circumstances. The precise relationship between viruses, apoptosis, and inflammation is viewed as a complex one requiring further investigation to unravel and understand its nature.


Assuntos
Apoptose , Sistema Nervoso Central/virologia , Neurônios/virologia , Sistema Nervoso Central/fisiopatologia , HIV/patogenicidade , HIV/fisiologia , Infecções por HIV/fisiopatologia , Infecções por HIV/virologia , Herpes Simples/fisiopatologia , Herpes Simples/virologia , Herpes Zoster/fisiopatologia , Herpes Zoster/virologia , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 3/patogenicidade , Herpesvirus Humano 3/fisiologia , Humanos , Inflamação/fisiopatologia , Inflamação/virologia , Neurônios/patologia , Orthoreovirus de Mamíferos/patogenicidade , Raiva/fisiopatologia , Raiva/virologia , Vírus da Raiva/patogenicidade , Vírus da Raiva/fisiologia , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Replicação Viral
7.
Virus Res ; 185: 77-81, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24680657

RESUMO

Grass carp hemorrhage is an acute contagious disease caused by grass carp reovirus (GCRV). The pathogenesis of GCRV and the relationship between GCRV and the host cells remain unclear. The aim of the present study was to investigate the relations among apoptosis, intracellular oxidative stress and virus replication in GCRV infected-cells. The results showed that GCRV induced activation of caspase proteases as early as 12 h, and reached maximum activities at 24 h or 48 h post-infection in a grass carp kidney cell line (CIK cells). Meanwhile, the levels of tumor necrosis factor (TNF-α) and interleukin-1ß (IL-1ß) also were increased in GCRV-infected CIK cells and showed a statistically significant difference from 24 h to 96 h post-infection. The infection of GCRV caused the destruction of entire monolayer and the death of host cells. Accompanied by the infection, a severe oxidative stress occurred, which led to extensive loss of antioxidants and formation of lipid peroxidation after 48 h post-infection. These data suggested that the apoptosis which was triggered at an early stage (12-24 h) in the viral infection cycle, might be independent of virus replication, while the oxidative stress induced by GCRV was mostly related to the virus replication.


Assuntos
Apoptose , Doenças dos Peixes/metabolismo , Infecções por Reoviridae/veterinária , Reoviridae/fisiologia , Animais , Carpas , Linhagem Celular , Doenças dos Peixes/genética , Doenças dos Peixes/fisiopatologia , Doenças dos Peixes/virologia , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Estresse Oxidativo , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Replicação Viral
8.
J Virol ; 87(24): 13397-408, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24089565

RESUMO

The mode and timing of virally induced cell death hold the potential of regulating viral yield, viral transmission, and the severity of virally induced disease. Orbiviruses such as the epizootic hemorrhagic disease virus (EHDV) are nonenveloped and cytolytic. To date, the death of cells infected with EHDV, the signal transduction pathways involved in this process, and the consequence of their inhibition have yet to be characterized. Here, we report that the Ibaraki strain of EHDV2 (EHDV2-IBA) induces apoptosis, autophagy, a decrease in cellular protein synthesis, the activation of c-Jun N-terminal kinase (JNK), and the phosphorylation of the JNK substrate c-Jun. The production of infectious virions decreased upon inhibition of apoptosis with the pan-caspase inhibitor Q-VD-OPH (quinolyl-valyl-O-methylaspartyl-[-2,6-difluorophenoxy]-methyl ketone), upon inhibition of autophagy with 3-methyladenine or via the knockout of the autophagy regulator Atg5, or upon treatment of infected cells with the JNK inhibitor SP600125 or the cyclin-dependent kinase (CDK) inhibitor roscovitine, which also inhibited c-Jun phosphorylation. Moreover, Q-VD-OPH, SP600125, and roscovitine partially reduced EHDV2-IBA-induced cell death, and roscovitine diminished the induction of autophagy by EHDV2-IBA. Taken together, our results imply that EHDV induces and benefits from the activation of signaling pathways involved in cell stress and death.


Assuntos
Apoptose , Autofagia , Doenças dos Bovinos/fisiopatologia , Vírus da Doença Hemorrágica Epizoótica/fisiologia , Infecções por Reoviridae/veterinária , Doenças dos Ovinos/fisiopatologia , Animais , Bovinos , Doenças dos Bovinos/genética , Doenças dos Bovinos/metabolismo , Doenças dos Bovinos/virologia , Linhagem Celular , Vírus da Doença Hemorrágica Epizoótica/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Ovinos , Doenças dos Ovinos/genética , Doenças dos Ovinos/metabolismo , Doenças dos Ovinos/virologia , Transdução de Sinais
9.
J Virol ; 87(23): 12967-79, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24067959

RESUMO

Reovirus nonstructural protein σ1s is implicated in cell cycle arrest at the G2/M boundary and induction of apoptosis. However, the contribution of σ1s to these effects in an otherwise isogenic viral background has not been defined. To evaluate the role of σ1s in cell cycle arrest and apoptosis, we used reverse genetics to generate a σ1s-null reovirus. Following infection with wild-type virus, we observed an increase in the percentage of cells in G2/M, whereas the proportion of cells in G2/M following infection with the σ1s-null mutant was unaffected. Similarly, we found that the wild-type virus induced substantially greater levels of apoptosis than the σ1s-null mutant. These data indicate that σ1s is required for both reovirus-induced cell cycle arrest and apoptosis. To define sequences in σ1s that mediate these effects, we engineered viruses encoding C-terminal σ1s truncations by introducing stop codons in the σ1s open reading frame. We also generated viruses in which charged residues near the σ1s amino terminus were replaced individually or as a cluster with nonpolar residues. Analysis of these mutants revealed that amino acids 1 to 59 and the amino-terminal basic cluster are required for induction of both cell cycle arrest and apoptosis. Remarkably, viruses that fail to induce cell cycle arrest and apoptosis also are attenuated in vivo. Thus, identical sequences in σ1s are required for reovirus-induced cell cycle arrest, apoptosis, and pathogenesis. Collectively, these findings provide evidence that the σ1s-mediated properties are genetically linked and suggest that these effects are mechanistically related.


Assuntos
Apoptose , Pontos de Checagem do Ciclo Celular , Orthoreovirus Mamífero 3/metabolismo , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Proteínas não Estruturais Virais/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular , Humanos , Orthoreovirus Mamífero 3/química , Orthoreovirus Mamífero 3/genética , Camundongos , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Replicação Viral
10.
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
11.
Arch Virol ; 156(11): 1917-29, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21779911

RESUMO

In this study, intracellular signaling in ARV S1133-mediated apoptosis was investigated. A microarray was used to examine the gene expression profiles of cells upon ARV S1133 infection and ARV-encoded pro-apoptotic protein σC overexpression. The analysis indicated that in the set of DNA-damage-responsive genes, DDIT-3 and GADD45α were both upregulated by viral infection and σC overexpression. Further investigation demonstrated that both treatments caused DNA breaks, which increased the expression and/or phosphorylation of DNA damage response proteins. ROS and lipid peroxidation levels were increased, and ARV S1133 and σC caused apoptosis mediated by DNA damage signaling. ROS scavenger NAC, caffeine and an ATM-specific inhibitor significantly reduced ARV S1133- and σC-induced DNA breaks, DDIT-3 and GADD45α expression, H2AX phosphorylation, and apoptosis. Overexpression of DDIT-3 and GADD45α enhanced the oxidative stress and apoptosis induced by ARV S1133 and σC. In conclusion, our results demonstrate the involvement of the DNA-damage-signaling pathway in ARV S1133- and σC-induced apoptosis.


Assuntos
Apoptose , Orthoreovirus Aviário/fisiologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/fisiopatologia , Infecções por Reoviridae/veterinária , Transdução de Sinais , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Células Cultivadas , Galinhas , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/genética , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Organismos Livres de Patógenos Específicos
12.
J Dairy Sci ; 93(6): 2486-95, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20494156

RESUMO

Epizootic hemorrhagic disease is caused by a Culicoides-borne Orbivirus. In cattle, the disease is characterized by reduced milk production and mortality. Recent outbreaks of epizootic hemorrhagic disease virus (EHDV) in North Africa, Israel, and Turkey increase the risk of its invasion into central and northern Europe. An outbreak of EHDV in Israel during the fall of 2006 enabled an assessment of the consequent production losses to the dairy cattle industry. Reduction in milk production and involuntary culling were modeled using a 4-yr database of monthly milk and mortality records from 48 affected and 63 unaffected herds. These indices were compared between periods of outbreak and no outbreak and assessed for various levels and exposure onset. Geospatial kriging interpolation of serological results from 127 herds was used to assess the total outbreak losses for the dairy cattle industry in Israel. Herds affected during the first, second, and third month of the outbreak (September-November) experienced an average loss of 207 (95% CI=154-261), 137 (63-211), and 52 (27-76) kg of milk/milking cow, respectively, during the outbreak period. An average excess mortality and involuntary culling of 1.47/100 cows was documented in herds affected in September. High correlation was observed between EHDV seroprevalence and milk loss; average milk loss for herds with seropositivity of 26 to 50, 51 to 75, and 76 to 100% was 84, 133, and 204 kg of milk/milking cow, respectively. A 1.42% (0.91-1.93%) increase in mortality was observed in herds with seroprevalence above 50%. Losses for the dairy cattle industry interpolated from these data were estimated at US$2,491,000 (US$1,591,000-3,391,000), an average loss of US$26.5/cow in the Israeli dairy cattle. This equals 0.55% of the average total value production of a dairy cow in Israel. This is the first study to estimate the production losses caused by EHDV or any bluetongue-like disease.


Assuntos
Doenças dos Bovinos/virologia , Vírus da Doença Hemorrágica Epizoótica , Infecções por Reoviridae/veterinária , Animais , Bovinos , Doenças dos Bovinos/economia , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/fisiopatologia , Custos e Análise de Custo , Indústria de Laticínios/economia , Surtos de Doenças/veterinária , Israel/epidemiologia , Lactação , Leite/metabolismo , Infecções por Reoviridae/economia , Infecções por Reoviridae/epidemiologia , Infecções por Reoviridae/fisiopatologia , Estudos Soroepidemiológicos , Fatores de Tempo
14.
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
15.
J Biol Chem ; 282(30): 21953-61, 2007 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-17540767

RESUMO

During viral infection, cells initiate antiviral responses to contain replication and inhibit virus spread. One protective mechanism involves activation of transcription factors interferon regulatory factor-3 (IRF-3) and NF-kappaB, resulting in secretion of the antiviral cytokine, interferon-beta. Another is induction of apoptosis, killing the host cell before virus disseminates. Mammalian reovirus induces both interferon-beta and apoptosis, raising the possibility that both pathways are initiated by a common cellular sensor. We show here that reovirus activates IRF-3 with kinetics that parallel the activation of NF-kappaB, a known mediator of reovirus-induced apoptosis. Activation of IRF-3 requires functional retinoic acid inducible gene-I and interferon-beta promoter stimulator-1, but these intracellular sensors are dispensable for activation of NF-kappaB. Interferon-beta promoter stimulator-1 and IRF-3 are required for efficient apoptosis following reovirus infection, suggesting a common mechanism of antiviral cytokine induction and activation of the cell death response.


Assuntos
Apoptose/fisiologia , RNA Helicases DEAD-box/genética , Fator Regulador 3 de Interferon/fisiologia , Interferon beta/genética , Infecções por Reoviridae/fisiopatologia , Animais , Linhagem Celular , Proteína DEAD-box 58 , Fibroblastos/fisiologia , Genes Reporter , Células HeLa , Humanos , Rim , Camundongos , Regiões Promotoras Genéticas , Receptores Imunológicos , Transfecção
16.
Emerg Infect Dis ; 11(11): 1673-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16318717

RESUMO

Coltiviruses are tickborne viruses of the genus Coltivirus. The type species, Colorado tick fever virus (from North America), has been isolated from patients with flulike syndromes, meningitis, encephalitis, and other severe complications. Another coltivirus, Eyach virus, has been isolated from ticks in France and Germany and incriminated in febrile illnesses and neurologic syndromes. Seadornaviruses are endemic in Southeast Asia, particularly Indonesia and China. The prototype virus of the genus, Banna virus (BAV), has been isolated from many mosquito species, humans with encephalitis, pigs, and cattle. Two other seadornaviruses, Kadipiro and Liao Ning, were isolated only from mosquitoes. The epidemiology of seadornaviruses remains poorly documented. Evidence suggests that BAV is responsible for encephalitis in humans. Infection with BAV may be underreported because it circulates in regions with a high incidence of Japanese encephalitis and could be misdiagnosed as this disease.


Assuntos
Coltivirus , Infecções por Reoviridae/epidemiologia , Reoviridae , Animais , Ásia/epidemiologia , Gatos , Coltivirus/classificação , Coltivirus/genética , Coltivirus/isolamento & purificação , Coltivirus/patogenicidade , Europa (Continente)/epidemiologia , Humanos , Camundongos , América do Norte/epidemiologia , Coelhos , Reoviridae/classificação , Reoviridae/genética , Reoviridae/isolamento & purificação , Reoviridae/patogenicidade , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia
17.
Apoptosis ; 10(3): 513-24, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15909114

RESUMO

Virus-induced activation of nuclear factor-kappa B (NF-kappaB) is required for Type 3 (T3) reovirus-induced apoptosis. We now show that NF-kappaB is also activated by the prototypic Type 1 reovirus strain Lang (T1L), which induces significantly less apoptosis than T3 viruses, indicating that NF-kappaB activation alone is not sufficient for apoptosis in reovirus-infected cells. A second phase of virus-induced NF-kappaB regulation, where NF-kappaB activation is inhibited at later times following infection with T3 Abney (T3A), is absent in T1L-infected cells. This suggests that inhibition of NF-kappaB activation at later times post infection also contributes to reovirus-induced apoptosis. Reovirus-induced inhibition of stimulus-induced activation of NF-kappaB is significantly associated with apoptosis following infection of HEK293 cells with reassortant reoviruses and is determined by the T3 S1 gene segment, which is also the primary determinant of reovirus-induced apoptosis. Inhibition of stimulus-induced activation of NF-kappaB also occurs following infection of primary cardiac myocytes with apoptotic (8B) but not non-apoptotic (T1L) reoviruses. Expression levels of the NF-kappaB-regulated cellular FLICE inhibitory protein (cFLIP) reflect NF-kappaB activation in reovirus-infected cells. Further, inhibition of NF-kappaB activity and cFLIP expression promote T1L-induced apoptosis. These results demonstrate that inhibition of stimulus-induced activation of NF-kappaB and the resulting decrease in cFLIP expression promote reovirus-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Orthoreovirus Mamífero 3/fisiologia , NF-kappa B/antagonistas & inibidores , Orthoreovirus de Mamíferos/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Miócitos Cardíacos/virologia , Vírus Reordenados/fisiologia , Infecções por Reoviridae/fisiopatologia
18.
Viral Immunol ; 18(1): 89-115, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15802955

RESUMO

Reoviruses have provided insight into the roles played by specific viral genes and the proteins they encode in virus-induced cell death and tissue injury. Apoptosis is a major mechanism of cell death induced by reoviruses. Reovirus-induced apoptosis involves both death-receptor and mitochondrial cell death pathways. Reovirus infection is associated with selective activation of mitogen activated protein kinase (MAPK) cascades including JNK/SAPK. Infection also perturbs transcription factor signaling resulting in the activation of c-Jun and initial activation followed by strain-specific inhibition of NF-kappaB. Infection results in changes in the expression of genes encoding proteins involved in cell cycle regulation, apoptosis, and DNA damage and repair processes. Apoptosis is a major mechanism of reovirus-induced injury to key target organs including the CNS and heart. Inhibition of apoptosis through the use of caspase or calpain inhibitors, minocycline, or in caspase 3(-/-) mice all reduce virus-associated tissue injury and enhance survival of infected animals. Reoviruses induce apoptotic cell death (oncolysis) in a wide variety of cancer cells and tumors. The capacity of reoviruses to grow efficiently in transformed cells is enhanced by the presence of an activated Ras signaling pathway likely through mechanisms involving inhibition of antiviral PKR signaling and activation of Ras/RalGEF/p38 pathways. The potential of reovirus-induced oncolysis in therapy of human cancers is currently being investigated in phase I/II clinical trials.


Assuntos
Apoptose , Morte Celular , Infecções por Reoviridae/fisiopatologia , Reoviridae/fisiologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Mitocondriais/metabolismo , Miocárdio/metabolismo , Orthoreovirus Aviário/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores Virais/metabolismo , Reoviridae/genética , Infecções por Reoviridae/virologia
19.
Proc Natl Acad Sci U S A ; 101(30): 11099-104, 2004 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-15263068

RESUMO

Reovirus is a benign human virus that was recently found to have oncolytic properties and is currently in clinical trials as a potential cancer therapy. We have previously demonstrated that activation of Ras signaling, a common event in cancer, renders cells susceptible to reovirus oncolysis. In this study, we investigate which elements downstream of Ras are important in reovirus infection. By using a panel of NIH 3T3 cells transformed with activated Ras mutated in the effector-binding domain, we found that only the RasV12G37 mutant, which was unable to signal to Raf or phosphatidylinositol 3-kinase but retained signaling capability to guanine nucleotide-exchange factors (GEFs) for the small G protein, Ral (known as RalGEFs), was permissive to reovirus. Expression of the activated mutant of the RalGEF, Rlf, also allowed reovirus replication. Specific inhibition of the Ral pathway by using dominant-negative RalA rendered normally permissive H-Ras cells (cells expressing activated Ras) resistant to reovirus. To further identify elements downstream of RalGEF that promote reovirus infection, we used chemical inhibitors of the downstream signaling elements p38 and JNK. We found that reovirus infection was blocked in the presence of the p38 inhibitor but not the JNK inhibitor. Together, these results implicate a Ras/RalGEF/p38 pathway in the regulation of reovirus replication and oncolysis.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Infecções por Reoviridae/fisiopatologia , Reoviridae/fisiologia , Fator ral de Troca do Nucleotídeo Guanina/metabolismo , Proteínas ras/metabolismo , Células 3T3 , Animais , DNA Viral/genética , GTP Fosfo-Hidrolases/metabolismo , Células L , Camundongos , Reoviridae/genética , Reoviridae/patogenicidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transfecção , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno
20.
J Clin Invest ; 111(12): 1823-33, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12813018

RESUMO

Infection of neonatal mice with some reovirus strains produces a disease similar to infantile biliary atresia, but previous attempts to correlate reovirus infection with this disease have yielded conflicting results. We used isogenic reovirus strains T3SA- and T3SA+, which differ solely in the capacity to bind sialic acid as a coreceptor, to define the role of sialic acid in reovirus encephalitis and biliary tract infection in mice. Growth in the intestine was equivalent for both strains following peroral inoculation. However, T3SA+ spread more rapidly from the intestine to distant sites and replicated to higher titers in spleen, liver, and brain. Strikingly, mice infected with T3SA+ but not T3SA- developed steatorrhea and bilirubinemia. Liver tissue from mice infected with T3SA+ demonstrated intense inflammation focused at intrahepatic bile ducts, pathology analogous to that found in biliary atresia in humans, and high levels of T3SA+ antigen in bile duct epithelial cells. T3SA+ bound 100-fold more efficiently than T3SA- to human cholangiocarcinoma cells. These observations suggest that the carbohydrate-binding specificity of a virus can dramatically alter disease in the host and highlight the need for epidemiologic studies focusing on infection by sialic acid-binding reovirus strains as a possible contributor to the pathogenesis of neonatal biliary atresia.


Assuntos
Atresia Biliar/etiologia , Orthoreovirus Mamífero 3/patogenicidade , Ácido N-Acetilneuramínico/fisiologia , Receptores Virais/fisiologia , Infecções por Reoviridae/complicações , Animais , Animais Recém-Nascidos , Antígenos Virais/metabolismo , Ductos Biliares/virologia , Atresia Biliar/fisiopatologia , Atresia Biliar/virologia , Linhagem Celular , Encefalite Viral/etiologia , Encefalite Viral/fisiopatologia , Encefalite Viral/virologia , Genótipo , Humanos , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/fisiologia , Camundongos , Fenótipo , Infecções por Reoviridae/fisiopatologia , Infecções por Reoviridae/virologia , Células Tumorais Cultivadas , Virulência/genética , Virulência/fisiologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...