Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Viruses ; 12(11)2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33147869

RESUMO

Emerging and re-emerging arthritogenic alphaviruses, such as Chikungunya virus (CHIKV) and O'nyong nyong virus, cause acute and chronic crippling arthralgia associated with inflammatory immune responses. Approximately 50% of CHIKV-infected patients suffer from rheumatic manifestations that last 6 months to years. However, the physiological functions of individual immune signaling pathways in the pathogenesis of alphaviral arthritis remain poorly understood. Here, we report that a deficiency in CXCL10, which is a chemoattractant for monocytes/macrophages/T cells, led to the same viremia as wild-type animals, but fewer immune infiltrates and lower viral loads in footpads at the peak of arthritic disease (6-8 days post infection). Macrophages constituted the largest immune cell population in footpads following infection, and were significantly reduced in Cxcl10-/- mice. The viral RNA loads in neutrophils and macrophages were reduced in Cxcl10-/- compared to wild-type mice. In summary, our results demonstrate that CXCL10 signaling promotes the pathogenesis of alphaviral disease and suggest that CXCL10 may be a therapeutic target for mitigating alphaviral arthritis.


Assuntos
Infecções por Alphavirus/imunologia , Alphavirus/patogenicidade , Artrite Infecciosa/imunologia , Quimiocina CXCL10/imunologia , Transdução de Sinais/imunologia , Alphavirus/genética , Infecções por Alphavirus/fisiopatologia , Animais , Artrite Infecciosa/virologia , Quimiocina CXCL10/genética , Modelos Animais de Doenças , Feminino , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/virologia , Carga Viral , Viremia/imunologia
2.
J Gen Virol ; 100(1): 46-62, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30451651

RESUMO

Interferon (IFN) regulatory factors (IRFs) are important determinants of the innate response to infection. We evaluated the role(s) of combined and individual IRF deficiencies in the outcome of infection of C57BL/6 mice with Sindbis virus, an alphavirus that infects neurons and causes encephalomyelitis. The brain and spinal cord levels of Irf7, but not Irf3 mRNAs, were increased after infection. IRF3/5/7-/- and IRF3/7-/- mice died within 3-4 days with uncontrolled virus replication, similar to IFNα receptor-deficient mice, while all wild-type (WT) mice recovered. IRF3-/- and IRF7-/- mice had brain levels of IFNα that were lower, but brain and spinal cord levels of IFNß and IFN-stimulated gene mRNAs that were similar to or higher than WT mice without detectable serum IFN or increases in Ifna or Ifnb mRNAs in the lymph nodes, indicating that the differences in outcome were not due to deficiencies in the central nervous system (CNS) type I IFN response. IRF3-/- mice developed persistent neurological deficits and had more spinal cord inflammation and higher CNS levels of Il1b and Ifnγ mRNAs than WT mice, but all mice survived. IRF7-/- mice died 5-8 days after infection with rapidly progressive paralysis and differed from both WT and IRF3-/- mice in the induction of higher CNS levels of IFNß, tumour necrosis factor (TNF) α and Cxcl13 mRNA, delayed virus clearance and more extensive cell death. Therefore, fatal disease in IRF7-/- mice is likely due to immune-mediated neurotoxicity associated with failure to regulate the production of inflammatory cytokines such as TNFα in the CNS.


Assuntos
Infecções por Alphavirus/fisiopatologia , Encefalomielite/fisiopatologia , Interações Hospedeiro-Patógeno , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Sindbis virus/crescimento & desenvolvimento , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/deficiência , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medula Espinal/patologia , Análise de Sobrevida
3.
J Virol ; 90(8): 4150-4159, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26865723

RESUMO

UNLABELLED: The alphaviral6kgene region encodes the two structural proteins 6K protein and, due to a ribosomal frameshift event, the transframe protein (TF). Here, we characterized the role of the6kproteins in the arthritogenic alphavirus Ross River virus (RRV) in infected cells and in mice, using a novel6kin-frame deletion mutant. Comprehensive microscopic analysis revealed that the6kproteins were predominantly localized at the endoplasmic reticulum of RRV-infected cells. RRV virions that lack the6kproteins 6K and TF [RRV-(Δ6K)] were more vulnerable to changes in pH, and the corresponding virus had increased sensitivity to a higher temperature. While the6kdeletion did not reduce RRV particle production in BHK-21 cells, it affected virion release from the host cell. Subsequentin vivostudies demonstrated that RRV-(Δ6K) caused a milder disease than wild-type virus, with viral titers being reduced in infected mice. Immunization of mice with RRV-(Δ6K) resulted in a reduced viral load and accelerated viral elimination upon secondary infection with wild-type RRV or another alphavirus, chikungunya virus (CHIKV). Our results show that the6kproteins may contribute to alphaviral disease manifestations and suggest that manipulation of the6kgene may be a potential strategy to facilitate viral vaccine development. IMPORTANCE: Arthritogenic alphaviruses, such as chikungunya virus (CHIKV) and Ross River virus (RRV), cause epidemics of debilitating rheumatic disease in areas where they are endemic and can emerge in new regions worldwide. RRV is of considerable medical significance in Australia, where it is the leading cause of arboviral disease. The mechanisms by which alphaviruses persist and cause disease in the host are ill defined. This paper describes the phenotypic properties of an RRV6kdeletion mutant. The absence of the6kgene reduced virion release from infected cells and also reduced the severity of disease and viral titers in infected mice. Immunization with the mutant virus protected mice against viremia not only upon exposure to RRV but also upon challenge with CHIKV. These findings could lead to the development of safer and more immunogenic alphavirus vectors for vaccine delivery.


Assuntos
Infecções por Alphavirus/virologia , Ross River virus/genética , Ross River virus/imunologia , Proteínas Estruturais Virais/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/fisiopatologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Vírus Chikungunya/imunologia , Chlorocebus aethiops , Cricetinae , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Mutação , Fases de Leitura , Ross River virus/patogenicidade , Deleção de Sequência , Células Vero , Carga Viral , Proteínas Estruturais Virais/análise , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia , Replicação Viral
4.
J Virol ; 87(13): 7680-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23637415

RESUMO

Alphaviruses are small enveloped RNA viruses that include important emerging human pathogens, such as chikungunya virus (CHIKV). These viruses infect cells via a low-pH-triggered membrane fusion reaction, making this step a potential target for antiviral therapies. The E1 fusion protein inserts into the target membrane, trimerizes, and refolds to a hairpin-like conformation in which the combination of E1 domain III (DIII) and the stem region (DIII-stem) pack against a core trimer composed of E1 domains I and II (DI/II). Addition of exogenous DIII proteins from Semliki Forest virus (SFV) has been shown to inhibit E1 hairpin formation and SFV fusion and infection. Here we produced and characterized DIII and DI/II proteins from CHIKV and SFV. Unlike SFV DIII, both core trimer binding and fusion inhibition by CHIKV DIII required the stem region. CHIKV DIII-stem and SFV DIII-stem showed efficient cross-inhibition of SFV, Sindbis virus, and CHIKV infections. We developed a fluorescence anisotropy-based assay for the binding of SFV DIII-stem to the core trimer and used it to demonstrate the relatively high affinity of this interaction (Kd [dissociation constant], ∼85 nM) and the importance of the stem region. Together, our results support the conserved nature of the key contacts of DIII-stem in the alphavirus E1 homotrimer and describe a sensitive and quantitative in vitro assay for this step in fusion protein refolding.


Assuntos
Infecções por Alphavirus/fisiopatologia , Vírus Chikungunya/metabolismo , Vírus da Floresta de Semliki/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Ligação Viral , Animais , Linhagem Celular , Vírus Chikungunya/fisiologia , Cricetinae , Drosophila , Polarização de Fluorescência , Humanos , Lipossomos/metabolismo , Ligação Proteica , Vírus da Floresta de Semliki/fisiologia , Sindbis virus/fisiologia
5.
Arch Virol ; 158(6): 1159-72, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23334837

RESUMO

Successful infection with chikungunya virus (CHIKV) depends largely on the ability of this virus to manipulate cellular processes in its favour through specific interactions with several host factors. The knowledge of virus-host interactions is of particular value for understanding the interface through which therapeutic strategies could be applied. In the current study, the authors have employed a computational method to study the protein interactions between CHIKV and both its human host and its mosquito vector. In this structure-based study, 2028 human and 86 mosquito proteins were predicted to interact with those of CHIKV through 3918 and 112 unique interactions, respectively. This approach could predict 40 % of the experimentally confirmed CHIKV-host interactions along with several novel interactions, suggesting the involvement of CHIKV in intracellular cell signaling, programmed cell death, and transcriptional and translational regulation. The data corresponded to those obtained in earlier studies for HIV and dengue viruses using the same methodology. This study provides a conservative set of potential interactions that can be employed for future experimental studies with a view to understanding CHIKV biology.


Assuntos
Infecções por Alphavirus/virologia , Vírus Chikungunya/fisiologia , Interações Hospedeiro-Patógeno , Aedes/virologia , Infecções por Alphavirus/fisiopatologia , Animais , Apoptose/fisiologia , Autofagia/fisiologia , Drosophila melanogaster/virologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Inflamação/fisiopatologia , Inflamação/virologia , Biossíntese de Proteínas/fisiologia , Transcrição Gênica/fisiologia , Proteínas Virais/fisiologia , Proteínas Estruturais Virais/fisiologia
6.
J Virol ; 86(10): 5674-85, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438538

RESUMO

Autophagy is a cellular process that sequesters cargo in double-membraned vesicles termed autophagosomes and delivers this cargo to lysosomes to be degraded. It is enhanced during nutrient starvation to increase the rate of amino acid turnover. Diverse roles for autophagy have been reported for viral infections, including the assembly of viral replication complexes on autophagic membranes and protection of host cells from cell death. Here, we show that autophagosomes accumulate in Semliki Forest virus (SFV)-infected cells. Despite this, disruption of autophagy had no effect on the viral replication rate or formation of viral replication complexes. Also, viral proteins rarely colocalized with autophagosome markers, suggesting that SFV did not utilize autophagic membranes for its replication. Further, we found that SFV infection, unlike nutrient starvation, did not inactivate the constitutive negative regulator of autophagosome formation, mammalian target of rapamycin, suggesting that SFV-dependent accumulation of autophagosomes was not a result of enhanced autophagosome formation. In starved cells, addition of NH(4)Cl, an inhibitor of lysosomal acidification, caused a dramatic accumulation of starvation-induced autophagosomes, while in SFV-infected cells, NH(4)Cl did not further increase levels of autophagosomes. These results suggest that accumulation of autophagosomes in SFV-infected cells is due to an inhibition of autophagosome degradation rather than enhanced rates of autophagosome formation. Finally, we show that the accumulation of autophagosomes in SFV-infected cells is dependent on the expression of the viral glycoprotein spike complex.


Assuntos
Infecções por Alphavirus/fisiopatologia , Autofagia , Glicoproteínas/metabolismo , Fagossomos/metabolismo , Vírus da Floresta de Semliki/fisiologia , Proteínas Estruturais Virais/metabolismo , Infecções por Alphavirus/metabolismo , Infecções por Alphavirus/virologia , Animais , Linhagem Celular , Cricetinae , Glicoproteínas/genética , Humanos , Camundongos , Vírus da Floresta de Semliki/genética , Proteínas Estruturais Virais/genética
7.
Brain Behav Immun ; 26(4): 552-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22227623

RESUMO

The acute sickness response to infection is a conserved set of changes in physiology and behaviour, featuring fever, fatigue, musculo-skeletal pain, disturbed mood, and cognitive difficulties. The manifestations differ somewhat between individuals, including those infected with pathogens which do not have genetic variability--suggesting host determinants. Principal components analysis (PCA) was applied to acute phase, self-report symptom data from subjects in the Dubbo Infection Outcomes Study (n=296) to empirically derive indices of fatigue, pain, neurocognitive difficulties, and mood disturbance, as well as overall illness severity. Associations were sought with functional single nucleotide polymorphisms (SNPs) in the cytokine genes, interleukin (IL)-6, tumour necrosis factor (TNF)-α, interferon (IFN)-γ, and IL-10. The summed individual symptom indices correlated with overall severity and also with functional status. The relative contribution of individual symptom domains to the overall illness was stable over time within subjects, but varied between subjects with the same infection. The T allele of the IFN-γ +874 T/A SNP was associated with increased fatigue (p=0.0003; OR: 3.3). The C allele of the IL-10 -592 C/A SNP exerted a protective effect on neurocognitive difficulties (p=0.017; OR: 0.52); while the A allele for the IL-10 -592 SNP was associated with increased mood disturbance (p=0.044; OR: 1.83), as was the G allele of the IL-6 -174 G/C SNP (p=0.051; OR: 1.83). The acute sickness response has discrete symptom domains including fatigue, which have unique genetic associations. These data provide novel insights into the pathophysiology of fatigue states.


Assuntos
Citocinas/genética , Fadiga/genética , Infecções/genética , Infecções/fisiopatologia , Dor/genética , Índice de Gravidade de Doença , Adulto , Infecções por Alphavirus/genética , Infecções por Alphavirus/fisiopatologia , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/fisiopatologia , Fadiga/etiologia , Feminino , Estudos de Associação Genética , Genótipo , Humanos , Infecções/complicações , Masculino , Dor/etiologia , Polimorfismo de Nucleotídeo Único , Análise de Componente Principal , Febre Q/genética , Febre Q/fisiopatologia , Ross River virus
8.
Clin Infect Dis ; 47(11): 1418-25, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18937577

RESUMO

BACKGROUND: Functional polymorphisms in immune response genes are increasingly recognized as important contributors to the marked individual differences in susceptibility to and outcomes of infectious disease. The acute sickness response is a stereotypical set of illness manifestations mediated by the proinflammatory cytokines induced by many different pathogens. The genetic determinants of severity of the acute sickness response have not previously been explored. METHODS: We examined the impact of functional polymorphisms in cytokine genes with critical roles in the early immune response (tumor necrosis factor-alpha, interleukin-6, interleukin-10, and interferon-gamma) on the severity and duration of illness following acute infection with Epstein-Barr virus, Coxiella burnetii (the causative agent of Q fever), or Ross River virus. RESULTS: We found that the interferon-gamma +874T/A and the interleukin-10 -592C/A polymorphisms significantly affected illness severity, cytokine protein levels, and the duration of illness. These cytokine genotypes acted in synergy to potentiate their influence on disease outcomes. CONCLUSIONS: These findings suggest that genetically determined variations in the intensity of the inflammatory response underpin the severity of the acute sickness response and predict the recovery time across varied infections.


Assuntos
Infecções por Alphavirus/imunologia , Citocinas/genética , Citocinas/imunologia , Mononucleose Infecciosa/imunologia , Polimorfismo Genético , Febre Q/imunologia , Adolescente , Adulto , Idoso , Infecções por Alphavirus/patologia , Infecções por Alphavirus/fisiopatologia , Sequência de Bases , Coxiella burnetii/imunologia , Suscetibilidade a Doenças , Feminino , Herpesvirus Humano 4/imunologia , Humanos , Mononucleose Infecciosa/patologia , Mononucleose Infecciosa/fisiopatologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Febre Q/patologia , Febre Q/fisiopatologia , Ross River virus/imunologia
9.
PLoS Pathog ; 4(2): e29, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18282093

RESUMO

Chikungunya virus (CHIKV) is a re-emerging arbovirus responsible for a massive outbreak currently afflicting the Indian Ocean region and India. Infection from CHIKV typically induces a mild disease in humans, characterized by fever, myalgia, arthralgia, and rash. Cases of severe CHIKV infection involving the central nervous system (CNS) have recently been described in neonates as well as in adults with underlying conditions. The pathophysiology of CHIKV infection and the basis for disease severity are unknown. To address these critical issues, we have developed an animal model of CHIKV infection. We show here that whereas wild type (WT) adult mice are resistant to CHIKV infection, WT mouse neonates are susceptible and neonatal disease severity is age-dependent. Adult mice with a partially (IFN-alpha/betaR(+/-)) or totally (IFN-alpha/betaR(-/-)) abrogated type-I IFN pathway develop a mild or severe infection, respectively. In mice with a mild infection, after a burst of viral replication in the liver, CHIKV primarily targets muscle, joint, and skin fibroblasts, a cell and tissue tropism similar to that observed in biopsy samples of CHIKV-infected humans. In case of severe infections, CHIKV also disseminates to other tissues including the CNS, where it specifically targets the choroid plexuses and the leptomeninges. Together, these data indicate that CHIKV-associated symptoms match viral tissue and cell tropisms, and demonstrate that the fibroblast is a predominant target cell of CHIKV. These data also identify the neonatal phase and inefficient type-I IFN signaling as risk factors for severe CHIKV-associated disease. The development of a permissive small animal model will expedite the testing of future vaccines and therapeutic candidates.


Assuntos
Infecções por Alphavirus/metabolismo , Vírus Chikungunya/fisiologia , Modelos Animais de Doenças , Interferon Tipo I/metabolismo , Adulto , Fatores Etários , Infecções por Alphavirus/patologia , Infecções por Alphavirus/fisiopatologia , Animais , Animais Recém-Nascidos , Animais não Endogâmicos , Linhagem Celular Tumoral , Vírus Chikungunya/patogenicidade , Chlorocebus aethiops , Feminino , Humanos , Recém-Nascido , Interferon Tipo I/deficiência , Interferon Tipo I/genética , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Longevidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Células Vero , Carga Viral , Replicação Viral
10.
Pharmacol Ther ; 107(3): 329-42, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15923040

RESUMO

Ross River virus (RRV) is a mosquito-borne alphavirus indigenous to Australia and the Western Pacific region and is responsible for several thousand cases of human RRV disease (RRVD) per annum. The disease primarily involves polyarthritis/arthralgia, with many patients also presenting with rash, myalgia, fever, and/or lethargy. The symptoms can be debilitating at onset, but they usually resolve within 3-6 months. Recent insights into the RRV-host relationship, associated pathology, and molecular biology of infection have generated a number of potential avenues for improved treatment. Although vaccine development has been proposed, the small market size and potential for antibody-dependent enhancement (ADE) of disease make this approach unattractive. Recent insights into the molecular basis of RRV-ADE and the virus's ability to manipulate host inflammatory and immune responses create potential new opportunities for therapeutic invention. Such interventions should overcome virus-induced dysregulation of protective host responses to promote viral clearance and/or ameliorate inflammatory immunopathology.


Assuntos
Infecções por Alphavirus/fisiopatologia , Ross River virus/patogenicidade , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/imunologia , Animais , Formação de Anticorpos , Austrália/epidemiologia , Modelos Animais de Doenças , Humanos , Incidência , Inflamação , Macrófagos , Camundongos , Ilhas do Pacífico/epidemiologia , Linfócitos T , Carga Viral , Vacinas Virais
11.
Virology ; 337(2): 344-52, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15913697

RESUMO

Semliki Forest virus (SFV) membrane fusion is mediated by the viral E1 protein at acidic pH and regulated by the dimeric interaction of E1 with the E2 membrane protein. During low pH-triggered fusion, the E2/E1 heterodimer dissociates, freeing E1 to drive membrane fusion. E2 is synthesized as a precursor, p62, which is processed to mature E2 by the cellular protease furin. Both the dissociation of the p62/E1 dimer and the fusion reaction of p62 virus have a more acidic pH threshold than that of the mature E2 virus. We have previously isolated SFV mutations that allow virus growth in furin-deficient cells. Here we have used such pci mutations to compare the interactions of the p62/E1 and E2/E1 dimers. Our data suggest that there is an important p62/E1 dimer interaction site identified by an E2 R250G mutation and that this interaction is maintained after processing to the mature E2 protein.


Assuntos
Vírus da Floresta de Semliki/fisiologia , Proteínas do Envelope Viral/metabolismo , Infecções por Alphavirus/fisiopatologia , Animais , Linhagem Celular , Cricetinae , Dimerização , Eletroporação , Concentração de Íons de Hidrogênio , Rim , Cinética , Fusão de Membrana , Mutação , Peptídeo Hidrolases/metabolismo , RNA/genética , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/crescimento & desenvolvimento , Proteínas do Envelope Viral/genética
12.
Prog Mol Subcell Biol ; 36: 71-93, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15171608

RESUMO

Sindbis virus infects neurons of the brain and spinal cord leading to neuronal apoptosis and encephalitis in mice. During postnatal development, neurons of mice remain susceptible to infection but become refractory to SV-induced programmed cell death. Failure to undergo programmed cell death results in a persistent infection. However, some neurovirulent strains of Sindbis virus overcome the age-dependent protective function in neurons, leading to enhanced apoptotic cell death in the central nervous system and higher mortality rates. Sindbis virus infections can also cause hind-limb paralysis due to the death of infected spinal cord motor neurons. However, spinal cord neuron death in older mice appears to occur by mechanisms that differ from classical apoptosis observed in newborn mice based on the morphology of dying neurons at these two sites. Sindbis virus infections of mosquitoes and some mosquito cell lines, on the other hand, do not induce cell death but persistent infections, a phenomenon also observed occasionally in cultured mammalian cells as well as in brains of infected mice surviving lethal infections. Thus, both viral and cellular factors contribute to the varied outcomes of infection. The molecular mechanisms that govern the susceptibility or resistance of particular cell types to SV-induced cell death are not well understood. Furthermore, the cellular execution machinery that produces the characteristic morphological distinctions between brain and spinal cord (i.e. apoptotic versus non-apoptotic) remain to be discovered.


Assuntos
Infecções por Alphavirus/fisiopatologia , Apoptose , Encefalite Viral/fisiopatologia , Neurônios/fisiologia , Sindbis virus/fisiologia , Fatores Etários , Infecções por Alphavirus/virologia , Animais , Linhagem Celular , Encefalite Viral/virologia , Vetores Genéticos , Humanos , Neurônios/citologia , Neurônios/virologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Sindbis virus/genética , Sindbis virus/patogenicidade
13.
Metab Eng ; 5(4): 230-45, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14642351

RESUMO

The overexpression of bcl-2 and its homologues is a widely used strategy to inhibit apoptosis in mammalian cell culture systems. In this study, we have evaluated the Bcl-2 homologue, Bcl-x(L) and compared its effectiveness to a Bcl-x(L) mutant lacking most of the non-conserved unstructured loop domain, Bcl-x(L)Delta (deletion of amino acids 26 through 83). The cell line, Chinese hamster ovary (CHO), was genetically modified to express constitutively Bcl-x(L) or the Bcl-x(L) variant and subjected to model apoptotic insults including Sindbis virus (SV) infection, gradual serum withdrawal, and serum deprivation. When cells were engineered to overexpress Bcl-x(L)Delta, cell death due to the SV was inhibited, and Bcl-x(L)Delta provided comparable protection to the wild-type Bcl-x(L) even though expression levels were much lower for the mutant. Furthermore, the cells expressing Bcl-x(L)Delta continued to proliferate following infection while CHO-bcl-x(L) ceased proliferation immediately following infection. As a result, total production of a heterologous protein encoded on the SV was highest in cell lines expressing Bcl-x(L)Delta. Cells expressing the variant Bcl-x(L) also continued to proliferate and showed increased viable cell numbers following gradual serum withdrawal. In contrast, wild-type Bcl-x(L) expressing CHO cells were found to arrest growth but maintain viability following serum withdrawal. Interestingly, CHO cells expressing Bcl-x(L)Delta were also able to recover and return to rapid growth rates much faster than either the wild-type CHO-bcl-x(L) or CHO following the replenishment of fresh complete medium containing 10% FBS. Confocal imaging of yellow fluorescent protein (YFP) fused to the N terminus of Bcl-x(L) and Bcl-x(L)Delta indicated dense aggregates of the Bcl-x(L)Delta while the wild-type protein was distributed throughout the cell in a manner resembling transmembrane localization. As an alternative to complete removal of the loop domain, Bcl-x(L) variants were created in which aspartate residues containing potential caspase recognition sites within the loop domain of Bcl-x(L) were removed. Cell populations expressing various Bcl-x(L)-Asp mutants were exposed to an apoptotic spent medium stimulus, and the cells expressing these Bcl-x(L) variants provided increased viabilities as compared to cells containing wild-type Bcl-x(L) protein. These studies indicate that modification of anti-apoptotic genes can affect multiple cellular properties including response to apoptotic stimuli and cell growth. This knowledge can be valuable in the design of improved apoptosis inhibitors for biotechnology applications.


Assuntos
Infecções por Alphavirus/fisiopatologia , Apoptose , Técnicas de Cultura de Células/métodos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sindbis virus , Substituição de Aminoácidos , Animais , Células CHO , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Meios de Cultura Livres de Soro , Humanos , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/química , Proteínas Recombinantes/metabolismo , Estaurosporina/farmacologia , Relação Estrutura-Atividade , Distribuição Tecidual , Proteína bcl-X
14.
J Virol ; 76(22): 11254-64, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388685

RESUMO

Alphaviruses productively infect a variety of vertebrate and insect cell lines. In vertebrate cells, Sindbis virus redirects cellular processes to meet the needs of virus propagation. At the same time, cells respond to virus replication by downregulating virus growth and preventing dissemination of the infection. The balance between these two mechanisms determines the outcome of infection at the cellular and organismal levels. In this report, we demonstrate that a viral nonstructural protein, nsP2, is a significant regulator of Sindbis virus-host cell interactions. This protein not only is a component of the replicative enzyme complex required for replication and transcription of viral RNAs but also plays a role in suppressing the antiviral response in Sindbis virus-infected cells. nsP2 most likely acts by decreasing interferon (IFN) production and minimizing virus visibility. Infection of murine cells with Sindbis virus expressing a mutant nsP2 leads to higher levels of IFN secretion and the activation of 170 cellular genes that are induced by IFN and/or virus replication. Secreted IFN protects naive cells against Sindbis virus infection and also stops viral replication in productively infected cells. Mutations in nsP2 can also attenuate Sindbis virus cytopathogenicity. Such mutants can persist in mammalian cells with defects in the alpha/beta IFN (IFN-alpha/beta) system or when IFN activity is neutralized by anti-IFN-alpha/beta antibodies. These findings provide new insight into the alphavirus-host cell interaction and have implications for the development of improved alphavirus expression systems with better antigen-presenting potential.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/fisiopatologia , Cisteína Endopeptidases/metabolismo , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Sindbis virus/patogenicidade , Infecções por Alphavirus/virologia , Animais , Linhagem Celular , Cricetinae , Cisteína Endopeptidases/genética , Camundongos , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , RNA Viral/metabolismo , Sindbis virus/genética , Transcrição Gênica , Proteínas Virais/genética , Proteínas Virais/metabolismo , Virulência , Replicação Viral
15.
Virology ; 263(2): 339-48, 1999 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-10544107

RESUMO

The age-related acquisition of resistance to fatal Sindbis virus infection was examined using a molecularly cloned laboratory strain of the AR339 isolate designated TRSB. TRSB caused 100% mortality in mice up to 5 days of age. Resistance to fatal infection developed abruptly between 5 and 9 days of age. Lethal Sindbis virus infection of mice inoculated at 4 days of age was characterized by high levels of virus replication, induction of high levels of interferon-alpha/beta and TNF-alpha and severe thymic involution indicative of a systemic stress response. These changes correlated with predominantly noninflammatory lesions. In contrast, TRSB infection of older mice was characterized by survival, more limited virus replication, reduced cytokine induction, and the development of inflammatory responses leading to encephalitis, myositis, and myocarditis. Previous studies utilized infections of neonatal mice with TRSB and an attenuated mutant of TRSB to compare fatal and nonfatal Sindbis infection (Trgovcich et al., 1996. Virology 224, 73-83). The experiments reported here utilize mouse age at the time of infection to create conditions for examination of fatal and nonfatal TRSB infections. Both experiments suggest that fatal infection is associated with a shock-like syndrome and little or no inflammatory pathology, while survival is correlated with greatly reduced cytokine levels and inflammatory lesions.


Assuntos
Envelhecimento , Infecções por Alphavirus/fisiopatologia , Interferons/metabolismo , Sindbis virus/fisiologia , Estresse Fisiológico/patologia , Fator de Necrose Tumoral alfa/metabolismo , Infecções por Alphavirus/mortalidade , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Cricetinae , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Encefalite Viral/imunologia , Encefalite Viral/patologia , Encefalite Viral/virologia , Inflamação/patologia , Inflamação/virologia , Camundongos , Camundongos Endogâmicos , Sindbis virus/genética , Sindbis virus/imunologia , Sindbis virus/patogenicidade , Estresse Fisiológico/virologia , Taxa de Sobrevida , Timo/patologia , Timo/virologia , Virulência , Replicação Viral
16.
Nat Med ; 5(7): 832-5, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10395331

RESUMO

The Bax protein is widely known as a pro-apoptotic Bcl-2 family member that when overexpressed can trigger apoptosis in multiple cell types and is important for the developmental cell death of neurons. However, Bax was found here to be a potent inhibitor of neuronal cell death in mice infected with Sindbis virus. Newborn mice, which are highly susceptible to a fatal infection with neurotropic Sindbis virus, were significantly protected from neuronal apoptosis and fatal disease when infected with a recombinant Sindbis virus encoding Bax. Deletion of the N terminus of Bax, which mimics cleaved Bax, converted Bax into a pro-apoptotic factor in vivo. As mice mature during the first week after birth, they acquire resistance to a fatal Sindbis virus infection. However, Bax-deficient mice remained very sensitive to fatal disease compared with their control littermates, indicating that endogenous Bax functions as a survival factor and contributes to age-dependent resistance to Sindbis virus-induced mortality. The protective effects of Bax were reproduced in cultured hippocampal neurons but not in cultured dorsal root ganglia neurons. These findings indicate that cell-specific factors determine the anti-apoptotic versus pro-apoptotic function of Bax.


Assuntos
Infecções por Alphavirus/patologia , Apoptose/fisiologia , Encéfalo/patologia , Encéfalo/virologia , Neurônios/patologia , Neurônios/virologia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/fisiologia , Sindbis virus/patogenicidade , Infecções por Alphavirus/genética , Infecções por Alphavirus/fisiopatologia , Animais , Linhagem Celular , Células Cultivadas , Cricetinae , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Gânglios Espinais/virologia , Vetores Genéticos , Hipocampo/citologia , Hipocampo/fisiologia , Hipocampo/virologia , Humanos , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Sprague-Dawley , Sindbis virus/isolamento & purificação , Transfecção , Proteína X Associada a bcl-2
17.
Cell Death Differ ; 5(1): 50-9, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10200445

RESUMO

Multicellular organisms can employ a number of defences to combat viral replication, the most dramatic being implementation of a cell autonomous apoptotic process. The overall cost to the viability of an organism of losing infected cells by apoptosis may be small if the dying cells can be substituted. In contrast, suicide of irreplaceable cells such as highly specialised neurons may have a more dramatic, even fatal consequence. Previous in vitro approaches to understanding whether neurotropic viruses cause neurons to apoptose have utilised transformed cell lines. These are not in the appropriate state of differentiation to provide an accurate indication of events in vivo. We have chosen to characterise the ability of a model CNS disease-causing virus, Semliki Forest virus (SFV), to infect and trigger apoptosis in primary cultures of nerve growth factor (NGF)-dependent sensory neurons. These cells are known to die when deprived of NGF and constitute a useful indicator of apoptosis. We observe that infection causes cell death which bears the morphological hallmarks of apoptosis, this occurs even in the present of survival promoting NGF and is concomitant with new virus production. Using the TUNEL (transferase dUTP nick end labelling) technique we show that SFV-induced apoptosis involves DNA fragmentation and requires caspase (CED-3/ICE cysteine protease) activation, as does apoptosis induced by NGF-deprivation. Extensive areas of apoptosis, as defined using a combination of ultrastructural analysis and TUNEL occur in infected neonatal mouse brains. The novel evidence that infection of primary neurons with SFV induces apoptosis with activation of one or more caspases defines a system for the further anlaysis of apoptosis regulation in physiologically relevant neurons.


Assuntos
Infecções por Alphavirus/fisiopatologia , Apoptose/fisiologia , Fatores de Crescimento Neural/farmacologia , Neurônios/citologia , Neurônios/virologia , Vírus da Floresta de Semliki , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/enzimologia , Encéfalo/virologia , Inibidores de Caspase , Caspases/metabolismo , Núcleo Celular/enzimologia , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Inibidores de Cisteína Proteinase/farmacologia , Fragmentação do DNA , Encefalite Viral/enzimologia , Feminino , Marcação In Situ das Extremidades Cortadas , Camundongos , Microscopia Eletrônica , Neurônios/enzimologia , Oligopeptídeos/farmacologia , Gravidez , Inibidores de Serina Proteinase/farmacologia , Tosilina Clorometil Cetona/farmacologia
18.
Neuropathol Appl Neurobiol ; 23(3): 235-41, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9223133

RESUMO

Semliki Forest virus (SFV) infection of mice is used as a model to study pathogenic processes occurring in viral encephalitis and demyelinating disease. In this study, the long-term effects of infection by the avirulent M9 mutant of SPV on the central nervous system (CNS) of BALB/c and SJL mice were determined. The presence of infectious virus, viral RNA and cytokine mRNA in the brains of individual mice and the presence of lesions in the spinal cords of the same mice up to 360 days post-infection (d.p.i.) were analysed in order to detect any correlation between these parameters of pathogenesis. Infectious virus could not be detected beyond 7 d.p.i. for either mouse strain. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to detect the presence of the E2 and nsP1 regions of the virus genome and mRNA for interferon-gamma and tumour necrosis factor-alpha. Viral RNA could be detected up to 90 d.p.i. for both mouse strains. Cytokine mRNA could be detected up to 28 d.p.i. for BALB/c mice but up to 360 d.p.i. for SJL mice. Inflammatory lesions, which were associated with cytokine mRNA expression, were not detected in BALB/c mice beyond 28 d.p.i. but were detected in two SJL mice at 90 d.p.i. It is concluded that M9-SFV infection induces long-term prolonged expression of pro-inflammatory cytokines in the CNS of the majority of SJL (but not BALB/c) mice which is not associated with persistence of the virus genome. M9-SFV infection of SJL mice may be a relevant model for the pathogenesis of multiple sclerosis in man.


Assuntos
Infecções por Alphavirus/fisiopatologia , Vírus da Floresta de Semliki , Infecções por Alphavirus/patologia , Animais , Sistema Nervoso Central/virologia , Citocinas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Reação em Cadeia da Polimerase , RNA Viral/análise , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/isolamento & purificação , Medula Espinal/patologia , Fatores de Tempo , Transcrição Gênica
19.
Virology ; 227(1): 234-8, 1997 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-9007079

RESUMO

Neonatal mice were infected with virus derived from a molecular clone of a laboratory strain of Sindbis virus, TRSB. The resulting acute fatal infection was typified by few if any of the classic hallmarks of encephalitis, very high levels of interferon-alpha/beta (IFNalphabeta), and lesions in the thymus and hematopoietic tissues usually associated with a severe stress response. Infection with an attenuated mutant of TRSB, which harbors a single amino acid change in the E2 surface glycoprotein (TRSBr114), was characterized by encephalitis, reduced mortality, low levels of IFNalphabeta, and no thymic pathology (J. Trgovcich, J. F. Aronson, and R. E. Johnston, 1996, Virology 224, 73-83). Here we report that infection of neonatal mice with TRSB, but not TRSBr114, resulted in induction of high levels of tumor necrosis factor-alpha as well as high and sustained levels of adrenalcorticotropin-releasing hormone and corticosterone. This syndrome of potentially toxic cytokine and stress hormone induction correlates with lethal Sindbis virus infection and constitutes a previously unrecognized aspect of Sindbis virus pathogenesis in mice.


Assuntos
Infecções por Alphavirus/fisiopatologia , Sindbis virus , Estresse Fisiológico/virologia , Hormônio Adrenocorticotrópico/biossíntese , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/mortalidade , Infecções por Alphavirus/virologia , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Corticosterona/biossíntese , Camundongos , Mutação Puntual , Choque Séptico/imunologia , Choque Séptico/fisiopatologia , Choque Séptico/virologia , Sindbis virus/genética , Estresse Fisiológico/imunologia , Timo/patologia , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA