Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
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.
Virus Res ; 153(2): 277-87, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20801176

RESUMO

Semliki Forest virus (SFV), an alphavirus, replicates in vertebrate host and mosquito vector cells. The virus-specific part of the replicase complex constitutes nonstructural proteins 1-4 (nsP1-nsP4) and is bound to cytoplasmic membranes by an amphipathic helix inside of nsP1 and through the palmitoylation of cysteine residues in nsP1. In mammalian cells, defects in these viral functions result in a nonviable phenotype or the emergence of second-site compensatory mutations that have a positive impact on SFV infection. In most cases, these second-site compensatory mutations were found to compensate for the defect caused by the absence of palmitoylation in mosquito cells (C6/36). In C6/36 cells, however, all palmitoylation-defective viruses had severely reduced synthesis of subgenomic RNA; at the same time, several of them had very efficient formation of defective interfering genomes. Analysis of C6/36 cells that individually expressed either wild type (wt) or palmitoylation-deficient nsP1 forms revealed that similar to mammalian cells, the wt nsP1 localized predominantly to the plasma membrane, whereas its mutant forms localized to the cytoplasm. In contrast to transfected mammalian cells, all forms of nsP1 induced the formation of filopodia-like structures on some, but not all, transfected mosquito cells. These findings indicate that the plasma membrane and associated host factors may have different roles in alphavirus replicase complex formation in mammalian and mosquito cells. In general, the lack of nsP1 palmitoylation had a less severe effect on the function of the replication complex in mammalian cells when compared with that in mosquito cells.


Assuntos
Proteínas Mutantes/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Membrana Celular/química , Cricetinae , Culicidae , Citoplasma/química , Lipoilação , Proteínas Mutantes/genética , Vírus da Floresta de Semliki/genética , Proteínas não Estruturais Virais/genética
11.
Immunobiology ; 214(6): 467-74, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19150742

RESUMO

Recombinant replicons of Semliki Forest virus (SFV) can be used to induce high-level, transient expression of heterologous proteins in vivo. We constructed infectious but replication-deficient SFV particles carrying recombinant RNA encoding the Brucella abortus translation initiation factor 3 (IF3). The recombinant SFV particles (SFV-IF3 particles) were then evaluated for their ability to induce immune responses and to protect BALB/c mice against a challenge with B. abortus 2308 following vaccination. Animals inoculated with SFV-IF3 developed IF3-specific IgM antibodies at day 14 post-immunization. In vitro stimulation of splenocytes from vaccinated mice with either recombinant IF3 (rIF3) or crude Brucella protein extracts resulted in a T-cell proliferative response and induction of interferon gamma secretion, but not interleukin-4. In addition, mice immunized with SFV-IF3 exhibited a significant level of resistance against challenge with the virulent B. abortus strain 2308 (P<0.01). These findings indicate that an SFV-based vector carrying RNA encoding Brucella IF3 has potential for use as a vaccine to induce protection against B. abortus infections.


Assuntos
Infecções por Alphavirus/imunologia , Fatores de Iniciação em Eucariotos/imunologia , Fator de Iniciação 3 em Procariotos/imunologia , Vírus da Floresta de Semliki/imunologia , Vacinação , Infecções por Alphavirus/prevenção & controle , Animais , Brucella abortus/genética , Fatores de Iniciação em Eucariotos/genética , Engenharia Genética , Imunidade Ativa/genética , Camundongos , Camundongos Endogâmicos BALB C , Fator de Iniciação 3 em Procariotos/genética , Recombinação Genética , Vírus da Floresta de Semliki/patogenicidade , Virulência
12.
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
13.
J Gen Virol ; 89(Pt 10): 2565-2568, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18796725

RESUMO

Semliki Forest virus (SFV) infection of the laboratory mouse provides a well-characterized tractable system to study the pathogenesis of virus encephalitis and virus induced demyelination. In microMT mice, which have no antibodies, infectious virus persisted in both the serum and the brain for several weeks, indicating that antibodies are required to eliminate infectious virus. In immunocompetent mice, virus infectivity in the brain was undetectable after the first week of infection, but virus RNA levels declined slowly. Following SFV infection, lesions of demyelination were present in the brains of both immunocompetent and microMT mice, indicating that antibodies are not required to generate lesions of demyelination.


Assuntos
Anticorpos Antivirais/imunologia , Encéfalo , Doenças Desmielinizantes/fisiopatologia , Encefalite Viral/imunologia , Encefalite Viral/fisiopatologia , Vírus da Floresta de Semliki/patogenicidade , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/fisiopatologia , Infecções por Alphavirus/virologia , Animais , Anticorpos Antivirais/sangue , Sangue/virologia , Encéfalo/imunologia , Encéfalo/virologia , Linhagem Celular , Doenças do Sistema Nervoso Central/imunologia , Doenças do Sistema Nervoso Central/fisiopatologia , Doenças do Sistema Nervoso Central/virologia , Cricetinae , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/virologia , Modelos Animais de Doenças , Encefalite Viral/virologia , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Floresta de Semliki/imunologia , Ensaio de Placa Viral
14.
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
15.
J Gen Virol ; 89(Pt 8): 1942-1944, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18632965

RESUMO

Previously, we have shown that mice defective in granule exocytosis and/or Fas.L/Fas-mediated cytolytic pathways are significantly more resistant to alphavirus, Semliki Forest virus (SFV), infection compared with wild-type mice. Here, we evaluated SFV replication in different tissues of mice defective in both cytolytic pathways (perf(-/-)xgld) relative to that in wild-type counterparts and found that viral replication in perf(-/-)xgld mice is remarkably restricted. Although the mechanism responsible for this observation is yet to be established, the lower virus titres found in these mice indicate that the role of cytolytic effector molecules in antiviral immunity needs to be re-evaluated.


Assuntos
Infecções por Alphavirus/imunologia , Proteína Ligante Fas/deficiência , Perforina/deficiência , Vírus da Floresta de Semliki/fisiologia , Vírus da Floresta de Semliki/patogenicidade , Replicação Viral , Infecções por Alphavirus/virologia , Animais , Proteína Ligante Fas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , Perforina/genética
16.
Virology ; 376(1): 242-51, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-18442838

RESUMO

Alphavirus vectors express high levels of recombinant proteins in mammalian cells, but their cytopathic nature makes this expression transient. In order to generate a Semliki Forest virus (SFV) noncytopathic vector we introduced mutations previously described to turn Sindbis virus noncytopathic into a conserved position in an SFV vector expressing LacZ. Interestingly, mutant P718T in replicase nsp2 subunit was able to replicate in only a small percentage of BHK cells, producing beta-gal-expressing colonies without selection. Puromycin N-acetyl-transferase (pac) gene was used to replace LacZ in this mutant allowing selection of an SFV noncytopathic replicon containing a second mutation in nsp2 nuclear localization signal (R649H). This latter mutation did not confer a noncytopathic phenotype by itself and did not alter nsp2 nuclear translocation. Replicase synthesis was diminished in the SFV double mutant, leading to genomic and subgenomic RNA levels that were 125-fold and 66-fold lower than in wild-type vector, respectively. Interestingly, this mutant expressed beta-gal levels similar to parental vector. By coexpressing pac and LacZ from independent subgenomic promoters this vector was able to generate stable cell lines maintaining high expression levels during at least 10 passages, indicating that it could be used as a powerful system for protein production in mammalian cells.


Assuntos
Efeito Citopatogênico Viral , Expressão Gênica , Vetores Genéticos , Biologia Molecular/métodos , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/patogenicidade , Virologia/métodos , Acetiltransferases/biossíntese , Acetiltransferases/genética , Substituição de Aminoácidos/genética , Animais , Fusão Gênica Artificial , Linhagem Celular , Cricetinae , Cisteína Endopeptidases/genética , Genes Reporter , Mutação de Sentido Incorreto , Sinais de Localização Nuclear/genética , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Inoculações Seriadas , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
17.
Virus Genes ; 36(2): 313-21, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18264748

RESUMO

An investigation of the role of the 5' untranslated region (UTR) of Semliki Forest virus (SFV) in determining pathogenicity in infected mice was carried out by constructing 5' UTR chimeras. Analysis of 5' UTR sequences showed nucleotide differences between virulent and avirulent strains at positions 21, 35 and 42. Reciprocal chimeras incorporating these changes were constructed from avirulent CA7 and rA7[74], and virulent SFV-4 virus, derived from infectious clones, and avirulent A7 and A7[74] plaque-purified stock virus. Survival rates and neuropathology in intranasally (i.n.) infected mice were analysed. While no statistically significant difference between rates of RNA synthesis was detected between strains in cell culture, an increase in survival of infected mice and a reduction in the severity of brain lesions was observed on substitution of the 5' UTR from a stock avirulent virus into an infectious clone where the remainder of the genome was derived from avirulent virus. However, substitution of a 5' UTR from an avirulent stock virus into an infectious clone where the remainder of the genome was from virulent virus did not affect virulence. These results and other studies suggest that control of virulence is polygenic, and that the SFV 5' UTR acts as a pathogenicity determinant in synergy with other determinants in the genome.


Assuntos
Regiões 5' não Traduzidas , Infecções por Alphavirus/virologia , Encefalopatias/virologia , Viroses do Sistema Nervoso Central/virologia , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/patogenicidade , Infecções por Alphavirus/mortalidade , Infecções por Alphavirus/patologia , Animais , Sequência de Bases , Encéfalo/patologia , Encéfalo/virologia , Encefalopatias/patologia , Linhagem Celular , Viroses do Sistema Nervoso Central/patologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Alinhamento de Sequência , Virulência/genética
18.
J Gen Virol ; 89(Pt 3): 676-686, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18272758

RESUMO

The cytotoxicity of Semliki Forest virus (SFV) infection is caused partly by the non-structural protein nsP2, an essential component of the SFV replicase complex. Due to the presence of a nuclear localization signal (NLS), nsP2 also localizes in the nucleus of infected cells. The present study analysed recombinant SFV replicons and genomes with various deletions or substitutions in the NLS, or with a proline-to-glycine mutation at position 718 of nsP2 (P718G). Deletion of one or two arginine residues from the NLS or substitution of two of the arginines with aspartic acid resulted in a virus with a temperature-sensitive phenotype, and substitution of all three arginines was lethal. Thus, most of the introduced mutations severely affected nsP2 functioning in viral replication; in addition, they inhibited the ability of SFV to induce translational shut-off and kill infected cells. SFV replicons with a P718G mutation or replacement of the NLS residues (648)RRR(650) with RDD were found to be the least cytotoxic. Corresponding replicons expressed non-structural proteins at normal levels, but had severely reduced genomic RNA synthesis and were virtually unable to replicate and transcribe co-electroporated helper RNA. The non-cytotoxic phenotype was maintained in SFV full-length genomes harbouring the corresponding mutations; however, during a single cycle of cell culture, these were converted to a cytotoxic phenotype, probably due to the accumulation of compensatory mutations.


Assuntos
Cisteína Endopeptidases/genética , Regulação Viral da Expressão Gênica , Mutação , Sinais de Localização Nuclear/genética , RNA Viral/biossíntese , Vírus da Floresta de Semliki/patogenicidade , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Cricetinae , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Efeito Citopatogênico Viral , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/metabolismo , Proteínas Virais/genética
19.
J Gen Virol ; 89(Pt 2): 467-473, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18198377

RESUMO

Blood-brain barrier (BBB) permeability was evaluated in mice and hamsters infected with West Nile virus (WNV, flavivirus) as compared to those infected with Semliki Forest (alphavirus) and Banzi (flavivirus) viruses. BBB permeability was determined by measurement of fluorescence in brain homogenates or cerebrospinal fluid (CSF) after intraperitoneal (i.p.) injection of sodium fluorescein, by macroscopic examination of brains after i.p. injection of Evans blue, or by measurement of total protein in CSF compared to serum. Lethal infection of BALB/c mice with Semliki Forest virus and Banzi virus caused the brain : serum fluorescence ratios to increase from a baseline of 2-4% to as high as 11 and 15%, respectively. Lethal infection of BALB/c mice with WNV did not increase BBB permeability. When C57BL/6 mice were used, BBB permeability was increased in some, but not all, of the WNV-infected animals. A procedure was developed to measure BBB permeability in live WNV-infected hamsters by comparing the fluorescence in the CSF, aspirated from the cisterna magnum, with the fluorescence in the serum. Despite a time-dependent tendency towards increased BBB permeability in some WNV-infected hamsters, the highest BBB permeability values did not correlate with mortality. These data indicated that a measurable increase in BBB permeability was not a primary determinant for lethality of WNV infection in rodents. The lack of a consistent increase in BBB permeability in WNV-infected rodents has implications for the understanding of viral entry, viral pathogenesis and accessibility of the CNS of rodents to drugs or effector molecules.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Infecções por Flavivirus/fisiopatologia , Febre do Nilo Ocidental/fisiopatologia , Vírus do Nilo Ocidental/patogenicidade , Infecções por Alphavirus/sangue , Infecções por Alphavirus/líquido cefalorraquidiano , Infecções por Alphavirus/fisiopatologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar , Cricetinae , Modelos Animais de Doenças , Infecções por Flavivirus/sangue , Infecções por Flavivirus/líquido cefalorraquidiano , Infecções por Flavivirus/mortalidade , Fluorescência , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Floresta de Semliki/imunologia , Vírus da Floresta de Semliki/patogenicidade , Coloração e Rotulagem , Febre do Nilo Ocidental/sangue , Febre do Nilo Ocidental/líquido cefalorraquidiano , Febre do Nilo Ocidental/mortalidade , Vírus do Nilo Ocidental/imunologia
20.
J Gen Virol ; 88(Pt 12): 3373-3384, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18024907

RESUMO

Semliki Forest virus (SFV) infection of the mouse provides a powerful model to study the pathogenesis of virus encephalitis. SFV and other alphavirus-based vector systems are increasingly used in biotechnology and medicine. This study analysed the strong susceptibility of this virus to type I interferon (IFN) responses. Following intraperitoneal infection of adult mice, SFV strain A7(74) was efficiently (100 %) neuroinvasive. In contrast, SFV4 was poorly (21 %) neuroinvasive. Upon entry into the brain, both viruses activated type I IFN responses. As determined by quantitative RT-PCR, activation of the IFN-alpha gene was proportional to virus RNA load. An intact type I IFN system was required for protection against both strains of SFV. IFN strongly curtailed virus spread in many cell types and in many tissues. In mice with an intact type I IFN system, infected cells were rarely observed and tissue tropism was difficult to determine. In the absence of a functional type I IFN system, the tropism and the potential for rapid and widespread infection of this virus was revealed. Virus infection was readily observed in the myocardium, endocardium, exocrine pancreas, adipose tissue, smooth muscle cells and in the brain in meningeal cells, ependymal cells and oligodendrocytes. In the brains of mice with and without type I IFN responses, virus infection of neurons remained rare and focal, indicating that the previously described restricted replication of SFV A7(74) in neurons is not mediated by type I IFN responses.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Interferon Tipo I/isolamento & purificação , Vírus da Floresta de Semliki/patogenicidade , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Genes/genética , Interferon Tipo I/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neurônios/virologia , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/isolamento & purificação , Carga Viral , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...