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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 44(6): 1455-69, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27332734

RESUMEN

Aedes aegypti mosquitoes are responsible for transmitting many medically important viruses such as those that cause Zika and dengue. The inoculation of viruses into mosquito bite sites is an important and common stage of all mosquito-borne virus infections. We show, using Semliki Forest virus and Bunyamwera virus, that these viruses use this inflammatory niche to aid their replication and dissemination in vivo. Mosquito bites were characterized by an edema that retained virus at the inoculation site and an inflammatory influx of neutrophils that coordinated a localized innate immune program that inadvertently facilitated virus infection by encouraging the entry and infection of virus-permissive myeloid cells. Neutrophil depletion and therapeutic blockade of inflammasome activity suppressed inflammation and abrogated the ability of the bite to promote infection. This study identifies facets of mosquito bite inflammation that are important determinants of the subsequent systemic course and clinical outcome of virus infection.


Asunto(s)
Infecciones por Arbovirus/inmunología , Virus Bunyamwera/fisiología , Inflamación/inmunología , Mordeduras y Picaduras de Insectos/inmunología , Neutrófilos/inmunología , Virus de los Bosques Semliki/fisiología , Replicación Viral , Animales , Movimiento Celular , Células Cultivadas , Culicidae/inmunología , Humanos , Inmunidad Innata , Inflamasomas/metabolismo , Inflamación/virología , Mordeduras y Picaduras de Insectos/virología , Ratones , Neutrófilos/virología
2.
Immunol Cell Biol ; 101(4): 333-344, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36702633

RESUMEN

Central nervous system virus infections are a major cause of morbidity and mortality worldwide and a significant global public health concern. As in many tissues, inflammation and immune responses in the brain, despite their protective roles, can also be harmful. Control of brain inflammation is important in many neurological diseases from encephalitis to multiple sclerosis and neurogenerative disease. The suppressors of cytokine signaling (SOCS) proteins are a key mechanism controlling inflammatory and immune responses across all tissues including the brain. Using a mouse model system, we demonstrate that lack of SOCS4 results in changes in the pathogenesis and clinical outcome of a neurotropic virus infection. Relative to wild-type mice, SOCS4-deficient mice showed accelerated clearance of virus from the brain, lower levels of persisting viral RNA in the brain, increased neuroinflammation and more severe neuropathology. We conclude that, in the mouse brain, SOCS4 is a vital regulator of antiviral immunity that mediates the critical balance between immunopathology and virus persistence.


Asunto(s)
Citocinas , Encefalitis , Proteínas Supresoras de la Señalización de Citocinas , Animales , Ratones , Citocinas/inmunología , Encefalitis/inmunología , Encefalitis/virología , Inmunidad , Virus de los Bosques Semliki , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
3.
J Virol ; 89(21): 11030-45, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311875

RESUMEN

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.


Asunto(s)
Neuronas/virología , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/metabolismo , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/patogenicidad , Proteínas no Estructurales Virales/genética , Sustitución de Aminoácidos/genética , Animales , Línea Celular , ADN Complementario/biosíntesis , Immunoblotting , Ratones , Microscopía Fluorescente , Procesamiento Proteico-Postraduccional/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus de los Bosques Semliki/metabolismo , Estadísticas no Paramétricas , Virulencia
4.
J Virol ; 89(15): 7536-49, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25972559

RESUMEN

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.


Asunto(s)
Infecciones por Alphavirus/virología , Barrera Hematoencefálica/virología , Encefalitis/virología , Virus de los Bosques Semliki/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Encéfalo/virología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Virus de los Bosques Semliki/química , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/patogenicidad , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Viremia/virología , Virulencia
5.
Nucleic Acids Res ; 42(14): 9436-46, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25053841

RESUMEN

Arboviruses are transmitted by distantly related arthropod vectors such as mosquitoes (class Insecta) and ticks (class Arachnida). RNA interference (RNAi) is the major antiviral mechanism in arthropods against arboviruses. Unlike in mosquitoes, tick antiviral RNAi is not understood, although this information is important to compare arbovirus/host interactions in different classes of arbovirus vectos. Using an Ixodes scapularis-derived cell line, key Argonaute proteins involved in RNAi and the response against tick-borne Langat virus (Flaviviridae) replication were identified and phylogenetic relationships characterized. Analysis of small RNAs in infected cells showed the production of virus-derived small interfering RNAs (viRNAs), which are key molecules of the antiviral RNAi response. Importantly, viRNAs were longer (22 nucleotides) than those from other arbovirus vectors and mapped at highest frequency to the termini of the viral genome, as opposed to mosquito-borne flaviviruses. Moreover, tick-borne flaviviruses expressed subgenomic flavivirus RNAs that interfere with tick RNAi. Our results characterize the antiviral RNAi response in tick cells including phylogenetic analysis of genes encoding antiviral proteins, and viral interference with this pathway. This shows important differences in antiviral RNAi between the two major classes of arbovirus vectors, and our data broadens our understanding of arthropod antiviral RNAi.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/genética , Ixodes/genética , Ixodes/virología , Interferencia de ARN , Animales , Proteínas Argonautas/fisiología , Línea Celular , ARN Interferente Pequeño/química , ARN Pequeño no Traducido/química , ARN Viral/química , Ribonucleasa III/fisiología
6.
J Virol ; 88(22): 13333-43, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25210177

RESUMEN

UNLABELLED: Chikungunya virus (CHIKV) is a reemerging mosquito-borne alphavirus that causes debilitating arthralgia in humans. Here we describe the development and testing of novel DNA replicon and protein CHIKV vaccine candidates and evaluate their abilities to induce antigen-specific immune responses against CHIKV. We also describe homologous and heterologous prime-boost immunization strategies using novel and previously developed CHIKV vaccine candidates. Immunogenicity and efficacy were studied in a mouse model of CHIKV infection and showed that the DNA replicon and protein antigen were potent vaccine candidates, particularly when used for priming and boosting, respectively. Several prime-boost immunization strategies eliciting unmatched humoral and cellular immune responses were identified. Further characterization by antibody epitope mapping revealed differences in the qualitative immune responses induced by the different vaccine candidates and immunization strategies. Most vaccine modalities resulted in complete protection against wild-type CHIKV infection; however, we did identify circumstances under which certain immunization regimens may lead to enhancement of inflammation upon challenge. These results should help guide the design of CHIKV vaccine studies and will form the basis for further preclinical and clinical evaluation of these vaccine candidates. IMPORTANCE: As of today, there is no licensed vaccine to prevent CHIKV infection. In considering potential new vaccine candidates, a vaccine that could raise long-term protective immunity after a single immunization would be preferable. While humoral immunity seems to be central for protection against CHIKV infection, we do not yet fully understand the correlates of protection. Therefore, in the absence of a functional vaccine, there is a need to evaluate a number of different candidates, assessing their merits when they are used either in a single immunization or in a homologous or heterologous prime-boost modality. Here we show that while single immunization with various vaccine candidates results in potent responses, combined approaches significantly enhance responses, suggesting that such approaches need to be considered in the further development of an efficacious CHIKV vaccine.


Asunto(s)
Fiebre Chikungunya/prevención & control , Virus Chikungunya/inmunología , Inmunización/métodos , Vacunas de ADN/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Fiebre Chikungunya/inmunología , Modelos Animales de Enfermedad , Femenino , Leucocitos Mononucleares/inmunología , Ratones Endogámicos C57BL , Análisis de Supervivencia , Vacunas de ADN/administración & dosificación , Vacunas Virales/administración & dosificación
7.
J Virol ; 88(5): 2858-66, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371047

RESUMEN

UNLABELLED: Chikungunya virus (CHIKV) is a reemerging mosquito-borne alphavirus that has caused severe epidemics in Africa and Asia and occasionally in Europe. As of today, there is no licensed vaccine available to prevent CHIKV infection. Here we describe the development and evaluation of novel CHIKV vaccine candidates that were attenuated by deleting a large part of the gene encoding nsP3 or the entire gene encoding 6K and were administered as viral particles or infectious genomes launched by DNA. The resulting attenuated mutants were genetically stable and elicited high magnitudes of binding and neutralizing antibodies as well as strong T cell responses after a single immunization in C57BL/6 mice. Subsequent challenge with a high dose of CHIKV demonstrated that the induced antibody responses protected the animals from viremia and joint swelling. The protective antibody response was long-lived, and a second homologous immunization further enhanced immune responses. In summary, this report demonstrates a straightforward means of constructing stable and efficient attenuated CHIKV vaccine candidates that can be administered either as viral particles or as infectious genomes launched by DNA. IMPORTANCE: Similar to other infectious diseases, the best means of preventing CHIKV infection would be by vaccination using an attenuated vaccine platform which preferably raises protective immunity after a single immunization. However, the attenuated CHIKV vaccine candidates developed to date rely on a small number of attenuating point mutations and are at risk of being unstable or even sensitive to reversion. We report here the construction and preclinical evaluation of novel CHIKV vaccine candidates that have been attenuated by introducing large deletions. The resulting mutants proved to be genetically stable, attenuated, highly immunogenic, and able to confer durable immunity after a single immunization. Moreover, these mutants can be administered either as viral particles or as DNA-launched infectious genomes, enabling evaluation of the most feasible vaccine modality for a certain setting. These CHIKV mutants could represent stable and efficient vaccine candidates against CHIKV.


Asunto(s)
Infecciones por Alphavirus/inmunología , Virus Chikungunya/inmunología , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Infecciones por Alphavirus/prevención & control , Infecciones por Alphavirus/virología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Fiebre Chikungunya , Virus Chikungunya/genética , Femenino , Orden Génico , Genoma Viral , Inmunidad Celular , Inmunización , Inmunización Secundaria , Ratones , Ratones Endogámicos C57BL , Mutación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Proteínas Virales/genética , Proteínas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
8.
PLoS Pathog ; 9(8): e1003514, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950709

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a neurotropic virus causing vesicular oral or genital skin lesions, meningitis and other diseases particularly harmful in immunocompromised individuals. To comprehensively investigate the complex interaction between HSV-1 and its host we combined two genome-scale screens for host factors (HFs) involved in virus replication. A yeast two-hybrid screen for protein interactions and a RNA interference (RNAi) screen with a druggable genome small interfering RNA (siRNA) library confirmed existing and identified novel HFs which functionally influence HSV-1 infection. Bioinformatic analyses found the 358 HFs were enriched for several pathways and multi-protein complexes. Of particular interest was the identification of Med23 as a strongly anti-viral component of the largely pro-viral Mediator complex, which links specific transcription factors to RNA polymerase II. The anti-viral effect of Med23 on HSV-1 replication was confirmed in gain-of-function gene overexpression experiments, and this inhibitory effect was specific to HSV-1, as a range of other viruses including Vaccinia virus and Semliki Forest virus were unaffected by Med23 depletion. We found Med23 significantly upregulated expression of the type III interferon family (IFN-λ) at the mRNA and protein level by directly interacting with the transcription factor IRF7. The synergistic effect of Med23 and IRF7 on IFN-λ induction suggests this is the major transcription factor for IFN-λ expression. Genotypic analysis of patients suffering recurrent orofacial HSV-1 outbreaks, previously shown to be deficient in IFN-λ secretion, found a significant correlation with a single nucleotide polymorphism in the IFN-λ3 (IL28b) promoter strongly linked to Hepatitis C disease and treatment outcome. This paper describes a link between Med23 and IFN-λ, provides evidence for the crucial role of IFN-λ in HSV-1 immune control, and highlights the power of integrative genome-scale approaches to identify HFs critical for disease progression and outcome.


Asunto(s)
Genoma Humano , Herpesvirus Humano 1/fisiología , Interleucinas/biosíntesis , Complejo Mediador/biosíntesis , Regulación hacia Arriba , Replicación Viral/fisiología , Eliminación de Gen , Células HeLa , Herpes Simple/genética , Herpes Simple/inmunología , Herpes Simple/metabolismo , Humanos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Interferones , Interleucinas/genética , Interleucinas/inmunología , Complejo Mediador/genética , Complejo Mediador/inmunología , Polimorfismo de Nucleótido Simple , ARN Polimerasa II/genética , ARN Polimerasa II/inmunología , ARN Polimerasa II/metabolismo
9.
PLoS Pathog ; 8(11): e1002977, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144608

RESUMEN

Several components of the mosquito immune system including the RNA interference (RNAi), JAK/STAT, Toll and IMD pathways have previously been implicated in controlling arbovirus infections. In contrast, the role of the phenoloxidase (PO) cascade in mosquito antiviral immunity is unknown. Here we show that conditioned medium from the Aedes albopictus-derived U4.4 cell line contains a functional PO cascade, which is activated by the bacterium Escherichia coli and the arbovirus Semliki Forest virus (SFV) (Togaviridae; Alphavirus). Production of recombinant SFV expressing the PO cascade inhibitor Egf1.0 blocked PO activity in U4.4 cell- conditioned medium, which resulted in enhanced spread of SFV. Infection of adult female Aedes aegypti by feeding mosquitoes a bloodmeal containing Egf1.0-expressing SFV increased virus replication and mosquito mortality. Collectively, these results suggest the PO cascade of mosquitoes plays an important role in immune defence against arboviruses.


Asunto(s)
Aedes , Infecciones por Alphavirus/inmunología , Inmunidad Innata , Proteínas de Insectos/inmunología , Monofenol Monooxigenasa/inmunología , Virus de los Bosques Semliki/fisiología , Replicación Viral/fisiología , Aedes/inmunología , Aedes/virología , Animales , Línea Celular , Cricetinae , Femenino
10.
Nat Commun ; 15(1): 2619, 2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38521764

RESUMEN

Immunity to infectious diseases is predominantly studied by measuring immune responses towards a single pathogen, although co-infections are common. In-depth mechanisms on how co-infections impact anti-viral immunity are lacking, but are highly relevant to treatment and prevention. We established a mouse model of co-infection with unrelated viruses, influenza A (IAV) and Semliki Forest virus (SFV), causing disease in different organ systems. SFV infection eight days before IAV infection results in prolonged IAV replication, elevated cytokine/chemokine levels and exacerbated lung pathology. This is associated with impaired lung IAV-specific CD8+ T cell responses, stemming from suboptimal CD8+ T cell activation and proliferation in draining lymph nodes, and dendritic cell paralysis. Prior SFV infection leads to increased blood brain barrier permeability and presence of IAV RNA in brain, associated with increased trafficking of IAV-specific CD8+ T cells and establishment of long-term tissue-resident memory. Relative to lung IAV-specific CD8+ T cells, brain memory IAV-specific CD8+ T cells have increased TCR repertoire diversity within immunodominant DbNP366+CD8+ and DbPA224+CD8+ responses, featuring suboptimal TCR clonotypes. Overall, our study demonstrates that infection with an unrelated neurotropic virus perturbs IAV-specific immune responses and exacerbates IAV disease. Our work provides key insights into therapy and vaccine regimens directed against unrelated pathogens.


Asunto(s)
Coinfección , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Virus , Ratones , Animales , Humanos , Gripe Humana/patología , Linfocitos T CD8-positivos , Coinfección/patología , Receptores de Antígenos de Linfocitos T , Pulmón/patología
11.
J Gen Virol ; 94(Pt 7): 1680-1689, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23559478

RESUMEN

The exogenous siRNA pathway is important in restricting arbovirus infection in mosquitoes. Less is known about the role of the PIWI-interacting RNA pathway, or piRNA pathway, in antiviral responses. Viral piRNA-like molecules have recently been described following infection of mosquitoes and derived cell lines with several arboviruses. The piRNA pathway has thus been suggested to function as an additional small RNA-mediated antiviral response to the known infection-induced siRNA response. Here we show that piRNA-like molecules are produced following infection with the naturally mosquito-borne Semliki Forest virus in mosquito cell lines. We show that knockdown of piRNA pathway proteins enhances the replication of this arbovirus and defines the contribution of piRNA pathway effectors, thus characterizing the antiviral properties of the piRNA pathway. In conclusion, arbovirus infection can trigger the piRNA pathway in mosquito cells, and knockdown of piRNA proteins enhances virus production.


Asunto(s)
Aedes/virología , Antivirales/metabolismo , Proteínas de Insectos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Virus de los Bosques Semliki/fisiología , Animales , Línea Celular , Proteínas de Insectos/genética , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/metabolismo , Replicación Viral
12.
Exp Appl Acarol ; 59(3): 319-38, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22773071

RESUMEN

Gene silencing by RNA interference (RNAi) is an important research tool in many areas of biology. To effectively harness the power of this technique in order to explore tick functional genomics and tick-microorganism interactions, optimised parameters for RNAi-mediated gene silencing in tick cells need to be established. Ten cell lines from four economically important ixodid tick genera (Amblyomma, Hyalomma, Ixodes and Rhipicephalus including the sub-species Boophilus) were used to examine key parameters including small interfering RNA (siRNA), double stranded RNA (dsRNA), transfection reagent and incubation time for silencing virus reporter and endogenous tick genes. Transfection reagents were essential for the uptake of siRNA whereas long dsRNA alone was taken up by most tick cell lines. Significant virus reporter protein knockdown was achieved using either siRNA or dsRNA in all the cell lines tested. Optimum conditions varied according to the cell line. Consistency between replicates and duration of incubation with dsRNA were addressed for two Ixodes scapularis cell lines; IDE8 supported more consistent and effective silencing of the endogenous gene subolesin than ISE6, and highly significant knockdown of the endogenous gene 2I1F6 in IDE8 cells was achieved within 48 h incubation with dsRNA. In summary, this study shows that gene silencing by RNAi in tick cell lines is generally more efficient with dsRNA than with siRNA but results vary between cell lines and optimal parameters need to be determined for each experimental system.


Asunto(s)
Técnicas de Silenciamiento del Gen , Ixodidae , Interferencia de ARN , Animales , Línea Celular , Genes Reporteros , Luciferasas de Renilla/genética , ARN Bicatenario , ARN Interferente Pequeño , Virus de los Bosques Semliki/genética
13.
J Virol ; 85(6): 2907-17, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21191029

RESUMEN

RNA interference (RNAi) is an important mosquito defense mechanism against arbovirus infection. In this paper we study the processes underlying antiviral RNAi in Aedes albopictus-derived U4.4 mosquito cells infected with Semliki Forest virus (SFV) (Togaviridae; Alphavirus). The production of virus-derived small interfering RNAs (viRNAs) from viral double-stranded RNA (dsRNA) is a key event in this host response. dsRNA could be formed by RNA replication intermediates, by secondary structures in RNA genomes or antigenomes, or by both. Which of these dsRNAs is the substrate for the generation of viRNAs is a fundamental question. Here we used deep sequencing of viRNAs and bioinformatic analysis of RNA secondary structures to gain insights into the characteristics and origins of viRNAs. An asymmetric distribution of SFV-derived viRNAs with notable areas of high-level viRNA production (hot spots) and no or a low frequency of viRNA production (cold spots) along the length of the viral genome with a slight bias toward the production of genome-derived viRNAs over antigenome-derived viRNAs was observed. Bioinformatic analysis suggests that hot spots of viRNA production are rarely but not generally associated with putative secondary structures in the SFV genome, suggesting that most viRNAs are derived from replicative dsRNA. A pattern of viRNAs almost identical to those of A. albopictus cells was observed for Aedes aegypti-derived Aag2 cells, suggesting common mechanisms that lead to viRNA production. Hot-spot viRNAs were found to be significantly less efficient at mediating antiviral RNAi than cold-spot viRNAs, pointing toward a nucleic acid-based viral decoy mechanism to evade the RNAi response.


Asunto(s)
Aedes/fisiología , Aedes/virología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Virus de los Bosques Semliki/crecimiento & desarrollo , Aedes/inmunología , Animales , Línea Celular , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/genética , ARN Viral/genética , ARN Viral/metabolismo , Virus de los Bosques Semliki/genética
14.
Viruses ; 14(11)2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36366574

RESUMEN

CNS viral infections are one of the major causes of morbidity and mortality worldwide and a significant global public health concern. Uncontrolled inflammation and immune responses in the brain, despite their protective roles, can also be harmful. The suppressor of cytokine signalling (SOCS) proteins is one of the key mechanisms controlling inflammatory and immune responses across all tissues including the brain. SOCS5 is highly expressed in the brain but there is little understanding of its role in the CNS. Using a mouse model of encephalitis, we demonstrate that lack of SOCS5 results in changes in the pathogenesis and clinical outcome of a neurotropic virus infection. Relative to wild-type mice, SOCS5-deficient mice had greater weight loss, dysregulated cytokine production and increased neuroinflammatory infiltrates composed predominantly of CD11b+ cells. We conclude that in the brain, SOCS5 is a vital regulator of anti-viral immunity that mediates the critical balance between immunopathology and virus persistence.


Asunto(s)
Infecciones por Alphavirus , Citocinas , Animales , Ratones , Citocinas/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/genética
15.
J Innate Immun ; : 1-17, 2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35738238

RESUMEN

Virus infections of the central nervous system (CNS) cause important diseases of humans and animals. As in other tissues, innate antiviral responses mediated by type I interferons (IFNs) are crucially important in controlling CNS virus infections. The maturity of neuronal populations is an established critical factor determining the outcome of CNS virus infection. Using primary cultures of mouse cortical neurons, we investigated the relationships between neuronal maturation, type I IFN responses, and the outcome of Semliki Forest virus infection. The virus replicated better, infected more cells, and produced higher titres of infectious viruses in immature neurons. Complete transcriptome analysis demonstrated that resting immature neurons have low transcriptional competence to mount antiviral responses. They had no detectable transcription of the genes Ddx58 and Ifih1, which encode key RNA virus cytoplasmic sensors RIG-I and MDA5, and very low expression of genes encoding key regulators of associated signalling pathways. Upon infection, immature neurons failed to mount an antiviral response as evidenced by their failure to produce chemokines, IFNs, and other cytokines. Treatment of immature neurons with exogenous IFNß prior to infection resulted in antiviral responses and lower levels of virus replication and infectious virus production. In contrast, resting mature neurons generated a robust antiviral response. This was augmented by pretreatment with IFNß. Infection of mature neurons derived from IFNAR-/- mice did not make an antiviral response and replicated virus to high levels.

16.
J Virol ; 84(14): 7369-77, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20427528

RESUMEN

The alphavirus Semliki Forest virus (SFV) and its derived vectors induce apoptosis in mammalian cells. Here, we show that apoptosis is associated with the loss of mitochondrial membrane potential followed by the activation of caspase-3, caspase-8, and caspase-9. Cell death can be partially suppressed by treatment with the pan-caspase inhibitor zVAD-fmk. To determine the role of SFV structural proteins in cell death, the temporal course of cell death was compared in cells infected with SFV and cells infected with SFV virus replicon particles (VRPs) lacking some or all of the virus structural genes. In the absence of virus structural proteins, cell death was delayed. The endoplasmic reticulum (ER) stress response, as determined by the splicing of X-box binding protein 1 (XBP1) transcripts and the activation of caspase-12, was activated in virus-infected cells but not in VRP (SFV lacking structural genes)-infected cells. The C/EBP-homologous protein (CHOP) was upregulated by both virus and VRP infections. The virus envelope proteins but not the virus capsid protein triggered ER stress. These results demonstrate that in NIH 3T3 cells, SFV envelope glycoproteins trigger the unfolded protein response of the ER and accelerate apoptotic cell death initiated by virus replicase activity.


Asunto(s)
Apoptosis/fisiología , Retículo Endoplásmico/metabolismo , Virus de los Bosques Semliki/metabolismo , Estrés Fisiológico , Clorometilcetonas de Aminoácidos/metabolismo , Animales , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Células 3T3 NIH , Virus de los Bosques Semliki/genética , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Respuesta de Proteína Desplegada/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo
18.
J Virol ; 83(11): 5735-48, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19297476

RESUMEN

In their vertebrate hosts, arboviruses such as Semliki Forest virus (SFV) (Togaviridae) generally counteract innate defenses and trigger cell death. In contrast, in mosquito cells, following an early phase of efficient virus production, a persistent infection with low levels of virus production is established. Whether arboviruses counteract RNA interference (RNAi), which provides an important antiviral defense system in mosquitoes, is an important question. Here we show that in Aedes albopictus-derived mosquito cells, SFV cannot prevent the establishment of an antiviral RNAi response or prevent the spread of protective antiviral double-stranded RNA/small interfering RNA (siRNA) from cell to cell, which can inhibit the replication of incoming virus. The expression of tombusvirus siRNA-binding protein p19 by SFV strongly enhanced virus spread between cultured cells rather than virus replication in initially infected cells. Our results indicate that the spread of the RNAi signal contributes to limiting virus dissemination.


Asunto(s)
Culicidae/virología , Interferencia de ARN , Virus de los Bosques Semliki/genética , Animales , Línea Celular , Cricetinae , Regulación Viral de la Expresión Génica , ARN Viral/genética , Replicación Viral
19.
Viruses ; 12(11)2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33167513

RESUMEN

The continuing emergence of arbovirus disease outbreaks around the world, despite the use of vector control strategies, warrants the development of new strategies to reduce arbovirus transmission. Superinfection exclusion, a phenomenon whereby a primary virus infection prevents the replication of a second closely related virus, has potential to control arbovirus disease emergence and outbreaks. This phenomenon has been observed for many years in plants, insects and mammalian cells. In this review, we discuss the significance of identifying novel vector control strategies, summarize studies exploring arbovirus superinfection exclusion and consider the potential for this phenomenon to be the basis for novel arbovirus control strategies.


Asunto(s)
Infecciones por Arbovirus/prevención & control , Arbovirus/fisiología , Culicidae/virología , Control de Mosquitos/métodos , Sobreinfección/virología , Animales , Infecciones por Arbovirus/transmisión , Humanos , Mosquitos Vectores/virología , Replicación Viral
20.
J Neurovirol ; 15(1): 57-70, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19115134

RESUMEN

Semliki Forest virus (SFV) provides an experimental model of acute virus encephalitis and virus-induced demyelinating disease. Two marker viruses expressing fluorescent proteins as part of the replicase or the structural open reading frame were used to evaluate virus replication in cells of the adult mouse brain. Both marker viruses established a high-titer infection in the adult mouse brain. As determined by location, morphology, and immunostaining with neural cell type-specific phenotypic markers, both viruses infected neurons and oligodendrocytes but not astrocytes. Determination of eGFP expression from either the replicase or the structural open-reading frame coupled with immunostaining for either the virus structural protein or the virus nonstructural protein-3 readily distinguished cells at early and late stages of infection. Neurons but not oligodendrocytes rapidly down-regulated virus replication. Rapid down-regulation of virus replication was also observed in mature but not immature primary cultures of rat hippocampal neurons. This study demonstrates for the first time that in vivo central nervous system (CNS) cells differ in their ability to suppress alphavirus replication.


Asunto(s)
Infecciones por Alphavirus/virología , Encéfalo/virología , Neuronas/virología , Oligodendroglía/virología , Virus de los Bosques Semliki/fisiología , Replicación Viral , Animales , Encéfalo/citología , Células Cultivadas , Regulación Viral de la Expresión Génica , Hipocampo/citología , Hipocampo/virología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Polimerasa Dependiente del ARN/genética , Ratas , Virus de los Bosques Semliki/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA