Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
Arch Virol ; 166(4): 1015-1033, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33582855

RESUMO

Multiple sclerosis (MS) is a common inflammatory demyelinating disease of the central nervous system. Although the etiology of MS is unknown, genetics and environmental factors, such as infections, play a role. Viral infections of mice have been used as model systems to study this demyelinating disease of humans. Three viruses that have long been studied in this capacity are Theiler's murine encephalomyelitis virus, mouse hepatitis virus, and Semliki Forest virus. This review describes the viruses themselves, the infection process, the disease caused by infection and its accompanying pathology, and the model systems and their usefulness in studying MS.


Assuntos
Modelos Animais de Doenças , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Infecções por Vírus de RNA/patologia , Infecções por Vírus de RNA/virologia , Animais , Sistema Nervoso Central/patologia , Sistema Nervoso Central/fisiologia , Sistema Nervoso Central/virologia , Humanos , Camundongos , Esclerose Múltipla/imunologia , Esclerose Múltipla/fisiopatologia , Vírus da Hepatite Murina/patogenicidade , Vírus da Hepatite Murina/fisiologia , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/fisiopatologia , Vírus da Floresta de Semliki/patogenicidade , Vírus da Floresta de Semliki/fisiologia , Theilovirus/patogenicidade , Theilovirus/fisiologia
2.
PLoS Pathog ; 14(1): e1006815, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29370307

RESUMO

Introduced transinfections of the inherited bacteria Wolbachia can inhibit transmission of viruses by Aedes mosquitoes, and in Ae. aegypti are now being deployed for dengue control in a number of countries. Only three Wolbachia strains from the large number that exist in nature have to date been introduced and characterized in this species. Here novel Ae. aegypti transinfections were generated using the wAlbA and wAu strains. In its native Ae. albopictus, wAlbA is maintained at lower density than the co-infecting wAlbB, but following transfer to Ae. aegypti the relative strain density was reversed, illustrating the strain-specific nature of Wolbachia-host co-adaptation in determining density. The wAu strain also reached high densities in Ae. aegypti, and provided highly efficient transmission blocking of dengue and Zika viruses. Both wAu and wAlbA were less susceptible than wMel to density reduction/incomplete maternal transmission resulting from elevated larval rearing temperatures. Although wAu does not induce cytoplasmic incompatibility (CI), it was stably combined with a CI-inducing strain as a superinfection, and this would facilitate its spread into wild populations. Wolbachia wAu provides a very promising new option for arbovirus control, particularly for deployment in hot tropical climates.


Assuntos
Aedes/microbiologia , Aedes/virologia , Antibiose/fisiologia , Vírus da Dengue , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Wolbachia/fisiologia , Infecções por Alphavirus/microbiologia , Infecções por Alphavirus/transmissão , Infecções por Alphavirus/virologia , Animais , Vírus da Dengue/patogenicidade , Feminino , Transmissão Vertical de Doenças Infecciosas/veterinária , Padrões de Herança , Masculino , Controle Biológico de Vetores , Vírus da Floresta de Semliki/patogenicidade , Vírus da Floresta de Semliki/fisiologia , Carga Viral
3.
Cell Mol Life Sci ; 73(20): 3897-916, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27117550

RESUMO

Alphavirus budding is driven by interactions between nucleocapsids assembled in the cytoplasm and envelope proteins present at the plasma membrane. So far, the expression of capsid and envelope proteins in infected cells has been considered an absolute requirement for alphavirus budding and propagation. In the present study, we show that Semliki Forest virus and Sindbis virus lacking the capsid gene can propagate in mammalian and insect cells. This propagation is mediated by the release of infectious microvesicles (iMVs), which are pleomorphic and have a larger size and density than wild-type virus. iMVs, which contain viral RNA inside and viral envelope proteins on their surface, are released at the plasma membrane and infect cells using the endocytic pathway in a similar way to wild-type virus. iMVs are not pathogenic in immunocompetent mice when injected intravenously, but can infect different organs like lungs and heart. Finally, we also show that alphavirus genomes without capsid can mediate the propagation of heterologous genes, making these vectors potentially interesting for gene therapy or vaccination studies. The minimalist infectious system described in this study shows that a self-replicating RNA able to express membrane proteins with binding and fusion properties is able to propagate, providing some insights into virus evolution.


Assuntos
Alphavirus/patogenicidade , Capsídeo/metabolismo , Membrana Celular/virologia , Micropartículas Derivadas de Células/virologia , Alphavirus/genética , Animais , Fusão Celular , Linhagem Celular , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/ultraestrutura , Feminino , Genoma Viral , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Testes de Neutralização , RNA Viral/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Transfecção , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo
4.
J Virol ; 90(5): 2418-33, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26676771

RESUMO

UNLABELLED: Alphaviruses represent a diverse set of arboviruses, many of which are important pathogens. Chikungunya virus (CHIKV), an arthritis-inducing alphavirus, is the cause of a massive ongoing outbreak in the Caribbean and South America. In contrast to CHIKV, other related alphaviruses, such as Venezuelan equine encephalitis virus (VEEV) and Semliki Forest virus (SFV), can cause encephalitic disease. E2, the receptor binding protein, has been implicated as a determinant in cell tropism, host range, pathogenicity, and immunogenicity. Previous reports also have demonstrated that E2 contains residues important for host range expansions and monoclonal antibody binding; however, little is known about what role each protein domain (e.g., A, B, and C) of E2 plays on these factors. Therefore, we constructed chimeric cDNA clones between CHIKV and VEEV or SFV to probe the effect of each domain on pathogenicity in vitro and in vivo. CHIKV chimeras containing each of the domains of the E2 (ΔDomA, ΔDomB, and ΔDomC) from SFV, but not VEEV, were successfully rescued. Interestingly, while all chimeric viruses were attenuated compared to CHIKV in mice, ΔDomB virus showed similar rates of infection and dissemination in Aedes aegypti mosquitoes, suggesting differing roles for the E2 protein in different hosts. In contrast to CHIKV; ΔDomB, and to a lesser extent ΔDomA, caused neuron degeneration and demyelination in mice infected intracranially, suggesting a shift toward a phenotype similar to SFV. Thus, chimeric CHIKV/SFV provide insights on the role the alphavirus E2 protein plays on pathogenesis. IMPORTANCE: Chikungunya virus (CHIKV) has caused large outbreaks of acute and chronic arthritis throughout Africa and Southeast Asia and has now become a massive public health threat in the Americas, causing an estimated 1.2 million human cases in just over a year. No approved vaccines or antivirals exist for human use against CHIKV or any other alphavirus. Despite the threat, little is known about the role the receptor binding protein (E2) plays on disease outcome in an infected host. To study this, our laboratory generated chimeric CHIKV containing corresponding regions of the Semliki Forest virus (SFV) E2 (domains A, B, and C) substituted into the CHIKV genome. Our results demonstrate that each domain of E2 likely plays a critical, but dissimilar role in the viral life cycle. Our experiments show that manipulation of E2 domains can be useful for studies on viral pathogenesis and potentially the production of vaccines and/or antivirals.


Assuntos
Infecções por Alphavirus/patologia , Vírus Chikungunya/patogenicidade , Vírus da Encefalite Equina Venezuelana/patogenicidade , Vírus da Floresta de Semliki/patogenicidade , Proteínas do Envelope Viral/metabolismo , Aedes/virologia , Infecções por Alphavirus/virologia , Animais , Encéfalo/patologia , Vírus Chikungunya/genética , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/genética , Feminino , Masculino , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Vírus da Floresta de Semliki/genética , Proteínas do Envelope Viral/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
5.
J Virol ; 89(21): 11030-45, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26311875

RESUMO

UNLABELLED: The A7(74) strain of Semliki Forest virus (SFV; genus Alphavirus) is avirulent in adult mice, while the L10 strain is virulent in mice of all ages. It has been previously demonstrated that this phenotypic difference is associated with nonstructural protein 3 (nsP3). Consensus clones of L10 (designated SFV6) and A7(74) (designated A774wt) were used to construct a panel of recombinant viruses. The insertion of nsP3 from A774wt into the SFV6 backbone had a minor effect on the virulence of the resulting recombinant virus. Conversely, insertion of nsP3 from SFV6 into the A774wt backbone or replacement of A774wt nsP3 with two copies of nsP3 from SFV6 resulted in virulent viruses. Unexpectedly, duplication of nsP3-encoding sequences also resulted in elevated levels of nsP4, revealing that nsP3 is involved in the stabilization of nsP4. Interestingly, replacement of nsP3 of SFV6 with that of A774wt resulted in a virulent virus; the virulence of this recombinant was strongly reduced by functionally coupled substitutions for amino acid residues 534 (P4 position of the cleavage site between nsP1 and nsP2) and 1052 (S4 subsite residue of nsP2 protease) in the nonstructural polyprotein. Pulse-chase experiments revealed that A774wt and avirulent recombinant virus were characterized by increased processing speed of the cleavage site between nsP1 and nsP2. A His534-to-Arg substitution specifically activated this cleavage, while a Val1052-to-Glu substitution compensated for this effect by reducing the basal protease activity of nsP2. These findings provide a link between nonstructural polyprotein processing and the virulence of SFV. IMPORTANCE: SFV infection of mice provides a well-characterized model to study viral encephalitis. SFV also serves as a model for studies of alphavirus molecular biology and host-pathogen interactions. Thus far, the genetic basis of different properties of SFV strains has been studied using molecular clones, which often contain mistakes originating from standard cDNA synthesis and cloning procedures. Here, for the first time, consensus clones of SFV strains were used to map virulence determinants. Existing data on the importance of nsP3 for virulent phenotypes were confirmed, another determinant of neurovirulence and its molecular basis was characterized, and a novel function of nsP3 was identified. These findings provide links between the molecular biology of SFV and its biological properties and significantly increase our understanding of the basis of alphavirus-induced pathology. In addition, the usefulness of consensus clones as tools for studies of alphaviruses was demonstrated.


Assuntos
Neurônios/virologia , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/metabolismo , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/patogenicidade , Proteínas não Estruturais Virais/genética , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , DNA Complementar/biossíntese , Immunoblotting , Camundongos , Microscopia de Fluorescência , Processamento de Proteína Pós-Traducional/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus da Floresta de Semliki/metabolismo , Estatísticas não Paramétricas , Virulência
6.
J Virol ; 89(15): 7536-49, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25972559

RESUMO

UNLABELLED: Semliki Forest virus (SFV) provides a well-characterized model system to study the pathogenesis of virus encephalitis. Several studies have used virus derived from the molecular clone SFV4. SFV4 virus does not have the same phenotype as the closely related L10 or the prototype virus from which its molecular clone was derived. In mice, L10 generates a high-titer plasma viremia, is efficiently neuroinvasive, and produces a fatal panencephalitis, whereas low-dose SFV4 produces a low-titer viremia, rarely enters the brain, and generally is avirulent. To determine the genetic differences responsible, the consensus sequence of L10 was determined and compared to that of SFV4. Of the 12 nucleotide differences, six were nonsynonymous; these were engineered into a new molecular clone, termed SFV6. The derived virus, SFV6, generated a high-titer viremia and was efficiently neuroinvasive and virulent. The phenotypic difference mapped to a single amino acid residue at position 162 in the E2 envelope glycoprotein (lysine in SFV4, glutamic acid in SFV6). Analysis of the L10 virus showed it contained different plaque phenotypes which differed in virulence. A lysine at E2 247 conferred a small-plaque avirulent phenotype and glutamic acid a large-plaque virulent phenotype. Viruses with a positively charged lysine at E2 162 or 247 were more reliant on glycosaminoglycans (GAGs) to enter cells and were selected for by passage in BHK-21 cells. Interestingly, viruses with the greatest reliance on binding to GAGs replicated to higher titers in the brain and more efficiently crossed an in vitro blood-brain barrier (BBB). IMPORTANCE: Virus encephalitis is a major disease, and alphaviruses, as highlighted by the recent epidemic of chikungunya virus (CHIKV), are medically important pathogens. In addition, alphaviruses provide well-studied experimental systems with extensive literature, many tools, and easy genetic modification. In this study, we elucidate the genetic basis for the difference in phenotype between SFV4 and the virus stocks from which it was derived and correct this by engineering a new molecular clone. We then use this clone in one comprehensive study to demonstrate that positively charged amino acid residues on the surface of the E2 glycoprotein, mediated by binding to GAGs, determine selective advantage and plaque size in BHK-21 cells, level of viremia in mice, ability to cross an artificial BBB, efficiency of replication in the brain, and virulence. Together with studies on Sindbis virus (SINV), this study provides an important advance in understanding alphavirus, and probably other virus, encephalitis.


Assuntos
Infecções por Alphavirus/virologia , Barreira Hematoencefálica/virologia , Encefalite/virologia , Vírus da Floresta de Semliki/metabolismo , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Encéfalo/virologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Vírus da Floresta de Semliki/química , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/patogenicidade , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Viremia/virologia , Virulência
7.
J Virol ; 87(1): 335-44, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23077310

RESUMO

Artificial target sequences for tissue-specific miRNAs have recently been introduced as a new means for altering the tissue tropism of viral replication. This approach can be used to improve the safety of oncolytic viruses for cancer virotherapy by restricting their replication in unwanted tissues, such as the liver. Semliki Forest virus (SFV) is a positive-strand RNA virus and, similar to the related alphaviruses, like Sindbis virus, has potential as a gene therapy vector and an oncolytic virotherapy agent, but this potential is limited by the neurovirulence of these alphaviruses. Here, we have generated a replicative SFV4 carrying six tandem targets for the neuron-specific miR124 between the viral nonstructural protein 3 and 4 (nsp3 and nsp4) genes. When administered intraperitoneally into adult BALB/c mice, SFV4-miRT124 displayed an attenuated spread into the central nervous system (CNS) and greatly increased survival. Peripheral replication was not affected, indicating neuron-specific attenuation. Moreover, a strong protective SFV immunity was elicited in these animals. Intracranial infection of adult mice with SFV4-miRT124 showed greatly reduced infection of neurons in the brain but led to the infection of oligodendrocytes in the corpus callosum. Taken together, our data show that miR124-mediated attenuation of neurovirulence is a feasible and promising strategy for generating safer oncolytic alphavirus virotherapy agents.


Assuntos
MicroRNAs/metabolismo , RNA Viral/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Tropismo Viral , Replicação Viral , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Encéfalo/virologia , Linhagem Celular , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , RNA Viral/genética , Vírus da Floresta de Semliki/fisiologia , Análise de Sobrevida
8.
J Virol ; 86(13): 7180-91, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22514352

RESUMO

The Old World alphaviruses are emerging human pathogens with an ability to cause widespread epidemics. The latest epidemic of Chikungunya virus, from 2005 to 2007, affected over 40 countries in Africa, Asia, and Europe. The Old World alphaviruses are highly cytopathic and known to evade the cellular antiviral response by inducing global inhibition of transcription in vertebrate cells. This function was shown to be mediated by their nonstructural nsP2 protein; however, the detailed mechanism of this phenomenon has remained unknown. Here, we report that nsP2 proteins of Sindbis, Semliki Forest, and Chikungunya viruses inhibit cellular transcription by inducing rapid degradation of Rpb1, a catalytic subunit of the RNAPII complex. This degradation of Rpb1 is independent of the nsP2-associated protease activity, but, instead, it proceeds through nsP2-mediated Rpb1 ubiquitination. This function of nsP2 depends on the integrity of the helicase and S-adenosylmethionine (SAM)-dependent methyltransferase-like domains, and point mutations in either of these domains abolish Rpb1 degradation. We go on to show that complete degradation of Rpb1 in alphavirus-infected cells occurs within 6 h postinfection, before other previously described virus-induced changes in cell physiology, such as apoptosis, autophagy, and inhibition of STAT1 phosphorylation, are detected. Since Rpb1 is a subunit that catalyzes the polymerase reaction during RNA transcription, degradation of Rpb1 plays an indispensable role in blocking the activation of cellular genes and downregulating cellular antiviral response. This indicates that the nsP2-induced degradation of Rpb1 is a critical mechanism utilized by the Old World alphaviruses to subvert the cellular antiviral response.


Assuntos
Vírus Chikungunya/enzimologia , Cisteína Endopeptidases/metabolismo , Evasão da Resposta Imune , Proteólise , RNA Polimerase II/antagonistas & inibidores , Vírus da Floresta de Semliki/enzimologia , Sindbis virus/enzimologia , Animais , Domínio Catalítico , Linhagem Celular , Vírus Chikungunya/patogenicidade , Cricetinae , Imunidade Inata , Camundongos , RNA Polimerase II/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Sindbis virus/patogenicidade , Ubiquitinação
9.
J Virol ; 85(13): 6334-42, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21543498

RESUMO

The alphavirus Semliki Forest virus (SFV) infects cells through a low-pH-dependent membrane fusion reaction mediated by the virus fusion protein E1. Acidic pH initiates a series of E1 conformational changes that culminate in membrane fusion and include dissociation of the E1/E2 heterodimer, insertion of the E1 fusion loop into the target membrane, and refolding of E1 to a stable trimeric hairpin conformation. A highly conserved histidine (H3) on the E1 protein was previously shown to promote low-pH-dependent E1 refolding. An SFV mutant with an alanine substitution at this position (H3A) has a lower pH threshold and reduced efficiency of virus fusion and E1 trimer formation than wild-type SFV. Here we addressed the mechanism by which H3 promotes E1 refolding and membrane fusion. We identified E1 mutations that rescue the H3A defect. These revertants implicated a network of interactions that connect the domain I-domain III (DI-DIII) linker region with the E1 core trimer, including H3. In support of the importance of these interactions, mutation of residues in the network resulted in more acidic pH thresholds and reduced efficiencies of membrane fusion. In vitro studies of truncated E1 proteins demonstrated that the DI-DIII linker was required for production of a stable E1 core trimer on target membranes. Together, our results suggest a critical and previously unidentified role for the DI-DIII linker region during the low-pH-dependent refolding of E1 that drives membrane fusion.


Assuntos
Fusão de Membrana , Glicoproteínas de Membrana/química , Vírus da Floresta de Semliki/patogenicidade , Proteínas do Envelope Viral/química , Proteínas Virais de Fusão/química , Alphavirus/patogenicidade , Alphavirus/fisiologia , Animais , Cricetinae , Histidina/química , Concentração de Íons de Hidrogênio , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutação , Conformação Proteica , Dobramento de Proteína , Multimerização Proteica , Estrutura Terciária de Proteína , Vírus da Floresta de Semliki/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
10.
J Virol ; 82(18): 9245-53, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18632857

RESUMO

The class II fusion proteins of the alphaviruses and flaviviruses mediate virus infection by driving the fusion of the virus membrane with that of the cell. These fusion proteins are triggered by low pH, and their structures are strikingly similar in both the prefusion dimer and the postfusion homotrimer conformations. Here we have compared cholesterol interactions during membrane fusion by these two groups of viruses. Using cholesterol-depleted insect cells, we showed that fusion and infection by the alphaviruses Semliki Forest virus (SFV) and Sindbis virus were strongly promoted by cholesterol, with similar sterol dependence in laboratory and field isolates and in viruses passaged in tissue culture. The E1 fusion protein from SFV bound cholesterol, as detected by labeling with photocholesterol and by cholesterol extraction studies. In contrast, fusion and infection by numerous strains of the flavivirus dengue virus (DV) and by yellow fever virus 17D were cholesterol independent, and the DV fusion protein did not show significant cholesterol binding. SFV E1 is the first virus fusion protein demonstrated to directly bind cholesterol. Taken together, our results reveal important functional differences conferred by the cholesterol-binding properties of class II fusion proteins.


Assuntos
Alphavirus/patogenicidade , Colesterol/metabolismo , Flavivirus/patogenicidade , Fusão de Membrana/fisiologia , Proteínas Virais de Fusão/metabolismo , Alphavirus/genética , Alphavirus/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Cricetinae , Culicidae , Vírus da Dengue/metabolismo , Vírus da Dengue/patogenicidade , Flavivirus/genética , Flavivirus/metabolismo , Mutação , Vírus da Floresta de Semliki/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Sindbis virus/metabolismo , Sindbis virus/patogenicidade , Proteínas Virais de Fusão/genética , Vírus da Febre Amarela/metabolismo , Vírus da Febre Amarela/patogenicidade
11.
J Cell Biol ; 134(4): 863-72, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8769412

RESUMO

Semliki Forest virus (SFV) infects cells by an acid-dependent membrane fusion reaction catalyzed by the virus spike protein, a complex containing E1 and E2 transmembrane subunits. E1 carries the putative virus fusion peptide, and mutations in this domain of the spike protein were previously shown to shift the pH threshold of cell-cell fusion (G91A), or block cell-cell fusion (G91D). We have used an SFV infectious clone to characterize virus particles containing these mutations. In keeping with the previous spike protein results, G91A virus showed limited secondary infection and an acid-shifted fusion threshold, while G91D virus was noninfectious and inactive in both cell-cell and virus-liposome fusion assays. During the low pH- induced SFV fusion reaction, the E1 subunit exposes new epitopes for monoclonal antibody (mAb) binding and forms an SDS-resistant homotrimer, the virus associates hydrophobically with the target membrane, and fusion of the virus and target membranes occurs. After low pH treatment, G91A spike proteins were shown to bind conformation-specific mAbs, associate with target liposome membranes, and form the E1 homotrimer. However, both G91A membrane association and homotrimer formation had an acid-shifted pH threshold and reduced efficiency compared to wt virus. In contrast, studies of the fusion-defective G91D mutant showed that the virus efficiently reacted with low pH as assayed by mAb binding and liposome association, but was essentially inactive in homotrimer formation. These results suggest that the G91D mutant is noninfectious due to a block in a late step in membrane fusion, separate from the initial reaction to low pH and interaction with the target membrane, and involving the lack of efficient formation of the E1 homotrimer.


Assuntos
Fusão Celular , Fusão de Membrana , Mutação , Vírus da Floresta de Semliki/fisiologia , Vírus da Floresta de Semliki/patogenicidade , Proteínas do Envelope Viral/metabolismo , Animais , Anticorpos Monoclonais , Anticorpos Antivirais , Capsídeo/química , Linhagem Celular , Cricetinae , Concentração de Íons de Hidrogênio , Rim , Lipossomos , Conformação Proteica , Vírus da Floresta de Semliki/genética , Tripsina , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
12.
Science ; 156(3783): 1760-1, 1967 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-5611040

RESUMO

Chick cells incubated at 1 degrees C with interferon fail to develop antiviral activity, but this activity appears subsequent to a 7-hour incubation at 37 degrees C after removal of interferon by repeated washings. Treatment with actinomycin D blocks the development of the latter activity. Cells incubated with interferon at 1 degrees C for up to 1 hour and then washed and incubated for 2 hours at 37 degrees C develop a degree of antiviral activity proportional to the concentration of interferon at initial incubation; at any concentration, the antiviral activity increased with the duration of initial incubation at 1 degrees C, but a maximal response was reached at 10 or 20 minutes. Treatment with trypsin after incubation with interferon at 1 degrees C inhibited development of antiviral activity. Interferon is rapidly bound to a superficial cell site, and this binding is necessary for development of antiviral activity in chick cells.


Assuntos
Antivirais , Sítios de Ligação , Interferons/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Animais , Embrião de Galinha/citologia , Efeito Citopatogênico Viral , Dactinomicina/farmacologia , Depressão Química , Fibroblastos , Tripsina
13.
Expert Rev Mol Med ; 10: e33, 2008 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19000329

RESUMO

Alphavirus vectors are high-level, transient expression vectors for therapeutic and prophylactic use. These positive-stranded RNA vectors, derived from Semliki Forest virus, Sindbis virus and Venezuelan equine encephalitis virus, multiply and are expressed in the cytoplasm of most vertebrate cells, including human cells. Part of the genome encoding the structural protein genes, which is amplified during a normal infection, is replaced by a transgene. Three types of vector have been developed: virus-like particles, layered DNA-RNA vectors and replication-competent vectors. Virus-like particles contain replicon RNA that is defective since it contains a cloned gene in place of the structural protein genes, and thus are able to undergo only one cycle of expression. They are produced by transfection of vector RNA, and helper RNAs encoding the structural proteins. Layered DNA-RNA vectors express the Semliki Forest virus replicon from a cDNA copy via a cytomegalovirus promoter. Replication-competent vectors contain a transgene in addition to the structural protein genes. Alphavirus vectors are used for three main applications: vaccine construction, therapy of central nervous system disease, and cancer therapy.


Assuntos
Vetores Genéticos , Vírus da Floresta de Semliki/genética , Animais , Doenças do Sistema Nervoso Central/tratamento farmacológico , Genoma Viral , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/prevenção & controle , Vírus da Floresta de Semliki/patogenicidade , Vacinas Sintéticas/administração & dosagem , Replicação Viral
14.
J Virol ; 81(24): 13631-9, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913808

RESUMO

The budding reactions of a number of enveloped viruses use the cellular machinery involved in the formation of the luminal vesicles of endosomal multivesicular bodies (MVB). Budding of these viruses is dependent on the presence of specific late-domain motifs in membrane-associated viral proteins. Such budding reactions usually involve ubiquitin and are blocked by expression of an ATPase-deficient form of VPS4, a cellular AAA+ ATPase believed to be required late in the MVB pathway for the disassembly/release of the MVB machinery. Here we examined the role of the MVB pathway in the budding of the late-domain-containing rhabdovirus vesicular stomatitis virus (VSV) and the alphavirus Semliki Forest virus (SFV). We tested early and late steps in the MVB pathway by depleting ubiquitin with the proteasome inhibitor MG-132 and by using cell lines inducibly expressing VPS4A or VPS4B protein. As previously shown, VSV budding was strongly dependent on ubiquitin. In contrast to the findings of previous studies with VPS4A, expression of ATPase-deficient mutants of either VPS4A or VPS4B inhibited VSV budding. Inhibition by VPS4 required the presence of the PPPY late domain on the VSV matrix protein and resulted in the accumulation of nonreleased VSV particles at the plasma membrane. In contrast, SFV budding was independent of both ubiquitin and the activity of VPS4, perhaps reflecting the important role of the highly organized envelope protein lattice during alphavirus budding.


Assuntos
Adenosina Trifosfatases/metabolismo , Endossomos/ultraestrutura , Vírus da Floresta de Semliki/crescimento & desenvolvimento , Ubiquitina/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Vesiculovirus/crescimento & desenvolvimento , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Alphavirus/genética , Alphavirus/crescimento & desenvolvimento , Alphavirus/metabolismo , Alphavirus/patogenicidade , Animais , Linhagem Celular/ultraestrutura , Linhagem Celular/virologia , Cricetinae , Complexos Endossomais de Distribuição Requeridos para Transporte , Endossomos/metabolismo , Humanos , Leupeptinas/farmacologia , Microscopia Eletrônica , Rhabdoviridae/genética , Rhabdoviridae/crescimento & desenvolvimento , Rhabdoviridae/metabolismo , Rhabdoviridae/patogenicidade , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Ubiquitina/efeitos dos fármacos , Ubiquitina/genética , ATPases Vacuolares Próton-Translocadoras , Proteínas de Transporte Vesicular/genética , Vesiculovirus/genética , Vesiculovirus/metabolismo , Vesiculovirus/patogenicidade , Montagem de Vírus
15.
Parasite ; 13(3): 251-5, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17007218

RESUMO

Environmental, technological and societal factors continue to have a dramatic effect on infectious diseases worldwide and are considered to be facilitating the emergence of several infectious diseases at a time. Co-infection with different species of viral and malaria infections are currently emerging problems of dual infection in the developing as well as developed countries. Understanding of interactions between the host, malaria and virus infection is of current concern and we have initiated studies to delineate the mechanisms involved during the progression of Semliki forest virus (SFV) and Plasmodium yoelii (P. yoelii) infection in mice. Enhanced virus multiplication and up-regulation of cytokine mRNA level in P. yoelii and SFV co-infected mice were observed on day 4 post-infection compared to respective controls. Collectively, our observations indicate that malaria infection may influence virus multiplication, pathogenesis and up-regulation of cytokine mRNA during co-infection in mice.


Assuntos
Infecções por Alphavirus/complicações , Citocinas/biossíntese , Malária/complicações , Plasmodium yoelii/patogenicidade , Vírus da Floresta de Semliki/patogenicidade , Infecções por Alphavirus/imunologia , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Linhagem Celular , Citocinas/genética , Modelos Animais de Doenças , Suscetibilidade a Doenças , Eritrócitos/parasitologia , Malária/imunologia , Camundongos , RNA Mensageiro/análise , Vírus da Floresta de Semliki/fisiologia , Replicação Viral
16.
J Neuroimmunol ; 125(1-2): 15-22, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11960636

RESUMO

Experimental infection of mice with avirulent Semliki Forest virus (SFV) has been used as a model of demyelinating disease in humans. A number of studies have shown that T cells may be important for mediating demyelination, but the role of T cells is still, unclear. Here, we show that neuronal necrosis, but not demyelination, was more severe in interleukin (IL)-12-defective mice compared with wild-type mice and this correlated with higher virus titers in the brain. In contrast, the severity of demyelination and neuronal depletion was reduced in IL-4-defective mice and this correlated with reduced brain virus titers and enhanced SFV-specific IFN-gamma production. The findings indicate that type 1 T cells play a role in the control of SFV replication but not directly in SFV-induced pathology in the CNS.


Assuntos
Infecções por Alphavirus/imunologia , Interleucina-12/genética , Interleucina-4/genética , Vírus da Floresta de Semliki/crescimento & desenvolvimento , Células Th1/imunologia , Infecções por Alphavirus/patologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/virologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Modelos Animais de Doenças , Interleucina-12/imunologia , Interleucina-4/imunologia , Metaloproteinase 9 da Matriz/análise , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Floresta de Semliki/patogenicidade , Organismos Livres de Patógenos Específicos , Carga Viral , Virulência , Replicação Viral/imunologia
17.
J Neurosci Methods ; 137(1): 111-21, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15196833

RESUMO

Establishing efficient gene transfer and expression in post-mitotic neurons is important in understanding the genetic basis of neural circuits with cellular complexity. This study evaluates the properties of exogenous green fluorescent protein (GFP) expression mediated by the Semliki forest virus (SFV) and adenovirus (Ad) vectors in dissociated and slice cultures of the mouse cerebellum. Infection with SFV-GFP resulted in early-onset and high-level GFP expression in about 90% of Purkinje cells and in about 40% of granule cells in dissociated cultures at 1 day after infection. Two days after infection, GFP-positive cells showed signs of SFV-derived cytotoxicity. Ad-GFP infected almost all astrocytes and granule cells in dissociated cultures, and showed a steady increase in GFP fluorescence with a plateau at around 2 days post-infection. Ad vector-mediated GFP expression lasted for several weeks with no significant cell damage. In the slice cultures, both viral vectors mainly infected astroglial cells, but also showed a similar cell preference as that in dissociated cultures. These data indicate that the use of different viral vectors and infection conditions offers a powerful means of expressing exogenous genes in cerebellar cultures with different cell-type specificity and timing and duration of expression.


Assuntos
Adenoviridae/genética , Córtex Cerebelar/virologia , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Neurônios/virologia , Vírus da Floresta de Semliki/genética , Adenoviridae/patogenicidade , Animais , Linhagem Celular , Córtex Cerebelar/citologia , Córtex Cerebelar/metabolismo , Cricetinae , Feto , Regulação Viral da Expressão Gênica/genética , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Vias Neurais/citologia , Vias Neurais/metabolismo , Vias Neurais/virologia , Neurônios/citologia , Neurônios/metabolismo , Técnicas de Cultura de Órgãos/métodos , Células de Purkinje/citologia , Células de Purkinje/metabolismo , Células de Purkinje/virologia , Reprodutibilidade dos Testes , Vírus da Floresta de Semliki/patogenicidade , Especificidade da Espécie , Fatores de Tempo
18.
J Virol Methods ; 8(4): 349-51, 1984 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-6206084

RESUMO

Virus infectivity in samples of culture medium or suspensions of animal tissue which are required for interferon assay can be rapidly and conveniently inactivated by overnight incubation with beta-propiolactone (BPL). As BPL hydrolyses spontaneously samples can be assayed with no further treatment. BPL does not affect the interfering activity of alpha, beta or gamma mouse interferons.


Assuntos
Interferons/análise , Lactonas/farmacologia , Propiolactona/farmacologia , Vírus/efeitos dos fármacos , Animais , Encéfalo/microbiologia , Células Cultivadas , Camundongos , Vírus da Floresta de Semliki/efeitos dos fármacos , Vírus da Floresta de Semliki/patogenicidade , Virulência/efeitos dos fármacos , Vírus/patogenicidade
19.
Mol Biotechnol ; 5(1): 33-8, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8853014

RESUMO

The Semliki Forest virus (SFV) expression vector consists of a plasmid based on the SFV infectious clone. Foreign genes may be inserted into the structural coding region, transcribed as RNA, and expressed in cell culture after transfection. RNA containing inserted sequences may be packaged into virions using a helper systems. This allows efficient infection and expression without chemical transfection, but only one round of multiplication is possible. The biosafety of the system has been increased by the introduction of multiple mutations, specifying a maturation defect, into the helper. Potential vaccines can be constructed by insertion of genes coding for antigenic proteins into the vector. Following insertion of the influenza virus nucleoprotein (NP) into the SFV vector, immunity was induced following injection of packaged or naked RNA into mice. The SFV vector is a "suicide" expression vector that has great potential for the construction of vaccines for both human and veterinary use.


Assuntos
Genoma Viral , Vírus da Floresta de Semliki/genética , Vacinas Virais , Animais , Camundongos , Plasmídeos , Vírus da Floresta de Semliki/imunologia , Vírus da Floresta de Semliki/patogenicidade , Virulência/genética
20.
J Neurol Sci ; 85(1): 87-96, 1988 May.
Artigo em Inglês | MEDLINE | ID: mdl-3385430

RESUMO

Electron microscopical (EM) studies were carried out on the retinas of 2-3-(baby), 12-, 14- and 21-28-day-old (adult) mice infected with avirulent (A774) Semliki Forest virus (SFV). Virions (mature virus), spherules and advanced stages of virus replication, cytopathic vacuoles type II (CPV II), were seen in the retinal neurons of baby mice after intracerebral (i.c.) or intraperitoneal (i.p.) infection. Some virions and spherules were also seen in the retinas of 12- and 14-day-old mice. Virions and advanced stages of virus replication were not seen in adult mice despite high virus titres. Some neurons of the inner nuclear layer and some ganglion cells showed reduced basophilia and appeared pale and occasionally some dense clumps of fine granules (DC) were seen in the neurones of the inner nuclear layer in these mice. A few small spherules were seen in the extracellular spaces. Some infiltrating cells were seen in the retinas in all ages of mice. We suggest that SFV causes retinopathy in baby mice and the neurophysiological changes reported in adult mice may be contributed to by virus replication in the retinal neurones and the presence of infiltrating cells in the retina.


Assuntos
Retina/microbiologia , Vírus da Floresta de Semliki/fisiologia , Replicação Viral , Animais , Feminino , Masculino , Camundongos , Microscopia Eletrônica , Retina/ultraestrutura , Vírus da Floresta de Semliki/patogenicidade , Vírus da Floresta de Semliki/ultraestrutura , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA