Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 383
Filtrar
1.
mBio ; 15(6): e0073624, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38695564

RESUMEN

Sindbis virus (SINV) infection of mice provides a model system for studying the pathogenesis of alphaviruses that infect the central nervous system (CNS) to cause encephalomyelitis. While studies of human viral infections typically focus on accessible cells from the blood, this compartment is rarely evaluated in mice. To bridge this gap, single-cell RNA sequencing (scRNAseq) was combined with flow cytometry to characterize the transcriptional and phenotypic changes of peripheral blood mononuclear cells (PBMCs) from SINV-infected mice. Twenty-one clusters were identified by scRNAseq at 7 days after infection, with a unique cluster and overall increase in naive B cells for infected mice. Uninfected mice had fewer immature T cells and CCR9+ CD4 T cells and a unique immature T cell cluster. Gene expression was most altered in the Ki67+ CD8 T cell cluster, with chemotaxis and proliferation-related genes upregulated. Global analysis indicated metabolic changes in myeloid cells and increased expression of Ccl5 by NK cells. Phenotypes of PBMCs and cells infiltrating the CNS were analyzed by flow cytometry over 14 days after infection. In PBMCs, CD8 and Th1 CD4 T cells increased in representation, while B cells showed a transient decrease at day 5 in total, Ly6a+, and naive cells, and an increase in activated B cells. In the brain, CD8 T cells increased for the first 7 days, while Th1 CD4 T cells and naive and Ly6a+ B cells continued to accumulate for 14 days. Therefore, dynamic immune cell changes can be identified in the blood as well as the CNS during viral encephalomyelitis. IMPORTANCE: The outcome of viral encephalomyelitis is dependent on the host immune response, with clearance and resolution of infection mediated by the adaptive immune response. These processes are frequently studied in mouse models of infection, where infected tissues are examined to understand the mechanisms of clearance and recovery. However, studies of human infection typically focus on the analysis of cells from the blood, a compartment rarely examined in mice, rather than inaccessible tissue. To close this gap, we used single-cell RNA sequencing and flow cytometry to profile the transcriptomic and phenotypic changes of peripheral blood mononuclear cells (PBMCs) before and after central nervous system (CNS) infection in mice. Changes to T and B cell gene expression and cell composition occurred in PBMC and during entry into the CNS, with CCL5 being a differentially expressed chemokine. Therefore, dynamic changes occur in the blood as well as the CNS during the response of mice to virus infection, which will inform the analysis of human studies.


Asunto(s)
Infecciones por Alphavirus , Leucocitos Mononucleares , Animales , Ratones , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Infecciones por Alphavirus/virología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/genética , Virus Sindbis/genética , Virus Sindbis/inmunología , Ratones Endogámicos C57BL , Fenotipo , Femenino , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/virología , Encefalitis Viral/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Análisis de la Célula Individual
2.
J Virol ; 97(10): e0095923, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37772825

RESUMEN

IMPORTANCE: Viral encephalomyelitis outcome is dependent on host responses to neuronal infection. Interferon (IFN) is an important component of the innate response, and IFN regulatory factor (IRF) 7 is an inducible transcription factor for the synthesis of IFN-α. IRF7-deficient mice develop fatal paralysis after CNS infection with Sindbis virus, while wild-type mice recover. Irf7 -/- mice produce low levels of IFN-α but high levels of IFN-ß with induction of IFN-stimulated genes, so the reason for this difference is not understood. The current study shows that Irf7 -/- mice developed inflammation earlier but failed to clear virus from motor neuron-rich regions of the brainstem and spinal cord. Levels of IFN-γ and virus-specific antibody were comparable, indicating that IRF7 deficiency does not impair expression of these known viral clearance factors. Therefore, IRF7 is either necessary for the neuronal response to currently identified mediators of clearance or enables the production of additional antiviral factor(s) needed for clearance.


Asunto(s)
Infecciones por Alphavirus , Encefalomielitis , Factor 7 Regulador del Interferón , Virus Sindbis , Animales , Ratones , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Tronco Encefálico/virología , Encefalomielitis/inmunología , Encefalomielitis/virología , Inflamación/virología , Factor 7 Regulador del Interferón/deficiencia , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Interferón beta/inmunología , Interferón beta/metabolismo , Neuronas Motoras/virología , Virus Sindbis/inmunología , Médula Espinal/virología
3.
Viruses ; 14(2)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35215941

RESUMEN

Flavivirus outbreaks require fast and reliable diagnostics that can be easily adapted to newly emerging and re-emerging flaviviruses. Due to the serological cross-reactivity among flavivirus antibodies, neutralization tests (NT) are considered the gold standard for sero-diagnostics. Here, we first established wild-type single-round infectious virus replicon particles (VRPs) by packaging a yellow fever virus (YFV) replicon expressing Gaussia luciferase (Gluc) with YFV structural proteins in trans using a double subgenomic Sindbis virus (SINV) replicon. The latter expressed the YFV envelope proteins prME via the first SINV subgenomic promoter and the capsid protein via a second subgenomic SINV promoter. VRPs were produced upon co-electroporation of replicon and packaging RNA. Introduction of single restriction enzyme sites in the packaging construct flanking the prME sequence easily allowed to exchange the prME moiety resulting in chimeric VRPs that have the surface proteins of other flaviviruses including dengue virus 1--4, Zika virus, West Nile virus, and tick-borne encephalitis virus. Besides comparing the YF-VRP based NT assay to a YF reporter virus NT assay, we analyzed the neutralization efficiencies of different human anti-flavivirus sera or a monoclonal antibody against all established VRPs. The assays were performed in a 96-well high-throughput format setting with Gluc as readout in comparison to classical plaque reduction NTs indicating that the VRP-based NT assays are suitable for high-throughput analyses of neutralizing flavivirus antibodies.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Flavivirus/inmunología , Ensayos Analíticos de Alto Rendimiento/métodos , Reacciones Cruzadas , Flavivirus/clasificación , Flavivirus/genética , Flavivirus/fisiología , Genes Reporteros , Luciferasas/genética , Luciferasas/metabolismo , Pruebas de Neutralización , Replicón , Virus Sindbis/genética , Virus Sindbis/inmunología , Virus Sindbis/fisiología , Virión/genética , Virión/inmunología , Virión/fisiología , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/fisiología
4.
Cell Rep ; 37(13): 110150, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34965418

RESUMEN

Enteric pathogens overcome barrier immunity within the intestinal environment that includes the endogenous flora. The microbiota produces diverse ligands, and the full spectrum of microbial products that are sensed by the epithelium and prime protective immunity is unknown. Using Drosophila, we find that the gut presents a high barrier to infection, which is partially due to signals from the microbiota, as loss of the microbiota enhances oral viral infection. We report cyclic dinucleotide (CDN) feeding is sufficient to protect microbiota-deficient flies from enhanced oral infection, suggesting that bacterial-derived CDNs induce immunity. Mechanistically, we find CDN protection is dSTING- and dTBK1-dependent, leading to NF-kB-dependent gene expression. Furthermore, we identify the apical nucleoside transporter, CNT2, as required for oral CDN protection. Altogether, our studies define a role for bacterial products in priming immune defenses in the gut.


Asunto(s)
Infecciones por Alphavirus/inmunología , Antivirales/farmacología , Drosophila melanogaster/inmunología , Enterocitos/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Nucleótidos Cíclicos/administración & dosificación , Infecciones por Alphavirus/tratamiento farmacológico , Infecciones por Alphavirus/virología , Animales , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Enterocitos/efectos de los fármacos , Enterocitos/virología , Femenino , Inmunidad Innata , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Virus Sindbis/inmunología
5.
Front Immunol ; 12: 719077, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394127

RESUMEN

The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 is a major global public threat. Currently, a worldwide effort has been mounted to generate billions of effective SARS-CoV-2 vaccine doses to immunize the world's population at record speeds. However, there is still a demand for alternative effective vaccines that rapidly confer long-term protection and rely upon cost-effective, easily scaled-up manufacturing. Here, we present a Sindbis alphavirus vector (SV), transiently expressing the SARS-CoV-2 spike protein (SV.Spike), combined with the OX40 immunostimulatory antibody (αOX40) as a novel, highly effective vaccine approach. We show that SV.Spike plus αOX40 elicits long-lasting neutralizing antibodies and a vigorous T-cell response in mice. Protein binding, immunohistochemical, and cellular infection assays all show that vaccinated mice sera inhibits spike functions. Immunophenotyping, RNA Seq transcriptome profiles, and metabolic analysis indicate a reprogramming of T cells in vaccinated mice. Activated T cells were found to mobilize to lung tissue. Most importantly, SV.Spike plus αOX40 provided robust immune protection against infection with authentic coronavirus in transgenic mice expressing the human ACE2 receptor (hACE2-Tg). Finally, our immunization strategy induced strong effector memory response, potentiating protective immunity against re-exposure to SARS-CoV-2 spike protein. Our results show the potential of a new Sindbis virus-based vaccine platform to counteract waning immune response, which can be used as a new candidate to combat SARS-CoV-2. Given the T-cell responses elicited, our vaccine is likely to be effective against variants that are proving challenging, as well as serve as a platform to develop a broader spectrum pancoronavirus vaccine. Similarly, the vaccine approach is likely to be applicable to other pathogens.


Asunto(s)
Antígenos de Diferenciación/inmunología , Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2/inmunología , Virus Sindbis/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Cricetinae , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Virus Sindbis/genética , Linfocitos T/inmunología , Vacunación
6.
J Gen Virol ; 102(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33507144

RESUMEN

The zebrafish (Danio rerio) possesses evolutionarily conserved innate and adaptive immunity as a mammal and has recently become a popular vertebrate model to exploit infection and immunity. Antiviral RNA interference (RNAi) has been illuminated in various model organisms, including Arabidopsis thaliana, Drosophila melanogaster, Caenorhabditis elegans and mice. However, to date, there is no report on the antiviral RNAi pathway of zebrafish. Here, we have evaluated the possible use of zebrafish to study antiviral RNAi with Sindbis virus (SINV), vesicular stomatitis virus (VSV) and Nodamura virus (NoV). We find that SINVs and NoVs induce the production of virus-derived small interfering RNAs (vsiRNAs), the hallmark of antiviral RNAi, with a preference for a length of 22 nucleotides, after infection of larval zebrafish. Meanwhile, the suppressor of RNAi (VSR) protein, NoV B2, may affect the accumulation of the NoV in zebrafish. Furthermore, taking advantage of the fact that zebrafish argonaute-2 (Ago2) protein is naturally deficient in cleavage compared with that of mammals, we provide evidence that the slicing activity of human Ago2 can virtually inhibit the accumulation of RNA virus after being ectopically expressed in larval zebrafish. Thus, zebrafish may be a unique model organism to study the antiviral RNAi pathway.


Asunto(s)
Interferencia de ARN , Infecciones por Virus ARN/virología , Virus ARN/fisiología , ARN Interferente Pequeño/metabolismo , ARN Viral/metabolismo , Pez Cebra/virología , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Inmunidad Innata , Modelos Animales , Nodaviridae/inmunología , Nodaviridae/fisiología , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , Virus Sindbis/inmunología , Virus Sindbis/fisiología , Vesiculovirus/inmunología , Vesiculovirus/fisiología , Pez Cebra/genética , Pez Cebra/inmunología , Proteínas de Pez Cebra/metabolismo
7.
Cell ; 183(7): 1884-1900.e23, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33301709

RESUMEN

Eastern equine encephalitis virus (EEEV) is one of the most virulent viruses endemic to North America. No licensed vaccines or antiviral therapeutics are available to combat this infection, which has recently shown an increase in human cases. Here, we characterize human monoclonal antibodies (mAbs) isolated from a survivor of natural EEEV infection with potent (<20 pM) inhibitory activity of EEEV. Cryo-electron microscopy reconstructions of two highly neutralizing mAbs, EEEV-33 and EEEV-143, were solved in complex with chimeric Sindbis/EEEV virions to 7.2 Å and 8.3 Å, respectively. The mAbs recognize two distinct antigenic sites that are critical for inhibiting viral entry into cells. EEEV-33 and EEEV-143 protect against disease following stringent lethal aerosol challenge of mice with highly pathogenic EEEV. These studies provide insight into the molecular basis for the neutralizing human antibody response against EEEV and can facilitate development of vaccines and candidate antibody therapeutics.


Asunto(s)
Aerosoles/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Virus de la Encefalitis Equina del Este/inmunología , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/prevención & control , Adulto , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Microscopía por Crioelectrón , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/virología , Epítopos/química , Femenino , Glicoproteínas/inmunología , Humanos , Ratones , Modelos Moleculares , Mutagénesis/genética , Pruebas de Neutralización , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes/inmunología , Virus Sindbis/inmunología , Virión/inmunología , Virión/ultraestructura , Internalización del Virus
8.
Proc Natl Acad Sci U S A ; 117(46): 29035-29045, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33144502

RESUMEN

Alphaviruses are positive-sense, enveloped RNA viruses that are important causes of viral encephalomyelitis. Sindbis virus (SINV) is the prototype alphavirus and preferentially infects neurons in rodents to induce an encephalomyelitis similar to the human disease. Using a mouse model of SINV infection of the nervous system, many of the immune processes involved in recovery from viral encephalomyelitis have been identified. Antibody specific to the SINV E2 glycoprotein plays an important role in recovery and is sufficient for noncytolytic suppression of virus replication in vivo and in vitro. To investigate the mechanism of anti-E2 antibody-mediated viral suppression, a reverse-phase protein array was used to broadly survey cellular signaling pathway activation following antibody treatment of SINV-infected differentiated AP-7 neuronal cells. Anti-E2 antibody induced rapid transient NF-κB and later sustained Y705 STAT3 phosphorylation, outlining an intracellular signaling cascade activated by antiviral antibody. Because NF-κB target genes include the STAT3-activating IL-6 family cytokines, expression of these messenger RNAS (mRNAs) was assessed. Expression of leukemia inhibitory factor (LIF) cytokine mRNA, but not other IL-6 family member mRNAs, was up-regulated by anti-E2 antibody. LIF induced STAT3 Y705 phosphorylation in infected differentiated AP-7 cells but did not inhibit virus replication. However, anti-E2 antibody localized the LIF receptor to areas of E2 expression on the infected cell surface, and LIF enhanced the antiviral effects of antibody. These findings identify activation of the NF-κB/LIF/STAT3 signaling cascade as involved in inducing antibody-mediated viral suppression and highlight the importance of nonneutralizing antibody functions in viral clearance from neurons.


Asunto(s)
Factor Inhibidor de Leucemia/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Virus Sindbis/inmunología , Infecciones por Alphavirus/metabolismo , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C , Ratas , Proteínas del Envoltorio Viral , Replicación Viral
9.
Emerg Microbes Infect ; 9(1): 1580-1589, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32576094

RESUMEN

The RNA interference (RNAi) pathway directs an important antiviral immunity mechanism in plants and invertebrates. Recently, we and others have demonstrated that the antiviral RNAi response is also conserved in mammals, at least to five distinct RNA viruses, including Zika virus (ZIKV). ZIKV may preferentially infect neuronal progenitor cells (NPCs) in the developing foetal brain. Ex vivo ZIKV infection induces RNAi-mediated antiviral response in human NPCs, but not in the more differentiated NPCs or somatic cells. However, litter is known about the in vivo property or function of the virus-derived small-interfering RNAs (vsiRNAs) targeting ZIKV. Here we report a surprising observation: different from ex vivo observations, viral small RNAs (vsRNAs) targeting ZIKV were produced in vivo upon infection in both central neuron system (CNS) and muscle tissues. In addition, our findings demonstrate the production of canonical vsiRNAs in murine CNS upon antiviral RNAi activation by Sindbis virus (SINV), suggesting the possibility of antiviral immune strategy applied by mammals in the CNS.


Asunto(s)
Infecciones por Alphavirus/genética , Alphavirus/inmunología , Células-Madre Neurales/virología , ARN Interferente Pequeño/metabolismo , ARN Viral/inmunología , Alphavirus/genética , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Animales , Diferenciación Celular , Línea Celular , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Chlorocebus aethiops , Células HEK293 , Humanos , Ratones , Músculo Esquelético/inmunología , Músculo Esquelético/virología , Células-Madre Neurales/inmunología , ARN Viral/antagonistas & inhibidores , Virus Sindbis/genética , Virus Sindbis/inmunología , Células Vero , Replicación Viral , Virus Zika/genética , Virus Zika/inmunología
10.
Sci Rep ; 10(1): 5259, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32210257

RESUMEN

Sindbis virus (SINV) is an alphavirus that causes age-dependent encephalomyelitis in mice. Within 7-8 days after infection infectious virus is cleared from neurons through the antiviral effects of antibody and interferon-gamma (IFNγ), but RNA persists. To better understand changes in viral RNA associated with immune-mediated clearance we developed recombinant strains of SINV that have genomic and subgenomic viral RNAs tagged with the Broccoli RNA aptamer that binds and activates a conditional fluorophore for live cell imaging of RNA. Treatment of SINV-Broccoli-infected cells with antibody to the SINV E2 glycoprotein had cell type-specific effects. In BHK cells, antibody increased levels of intracellular viral RNA and changed the primary location of genomic RNA from the perinuclear region to the plasma membrane without improving cell viability. In undifferentiated and differentiated AP7 (dAP7) neuronal cells, antibody treatment decreased levels of viral RNA. Occasional dAP7 cells escaped antibody-mediated clearance by not expressing cell surface E2 or binding antibody to the plasma membrane. IFNγ decreased viral RNA levels only in dAP7 cells and synergized with antibody for RNA clearance and improved cell survival. Therefore, analysis of aptamer-tagged SINV RNAs identified cell type- and neuronal maturation-dependent responses to immune mediators of virus clearance.


Asunto(s)
Anticuerpos Antivirales/farmacología , Aptámeros de Nucleótidos/análisis , Fibroblastos/virología , Glicoproteínas/inmunología , Interferón gamma/farmacología , Neuronas/virología , ARN Viral/análisis , Virus Sindbis/genética , Análisis de la Célula Individual/métodos , Imagen de Lapso de Tiempo/métodos , Proteínas no Estructurales Virales/análisis , Proteínas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Diferenciación Celular , Línea Celular , Línea Celular Transformada , Cricetinae , Fibroblastos/ultraestructura , Proteínas Luminiscentes , Mesocricetus , Neuronas/ultraestructura , Neuronas Receptoras Olfatorias/ultraestructura , Neuronas Receptoras Olfatorias/virología , Ratas , Proteínas Recombinantes/análisis , Virus Sindbis/inmunología , Fracciones Subcelulares/química , Fracciones Subcelulares/ultraestructura , Proteína Fluorescente Roja
11.
Virology ; 542: 63-70, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32056669

RESUMEN

Sindbis virus (SINV) produces the small membrane protein TF from the 6K gene via a (-1) programmed ribosomal frameshifting. While several groups have shown that TF-deficient virus exhibits reduced virulence, the mechanism(s) by which this occurs remain unknown. Here, we demonstrate a role for TF in antagonizing the host interferon response. Using wild-type and type 1 interferon receptor-deficient mice and primary cells derived from these animals, we show that TF controls the induction of the host interferon response at early times during infection. Loss of TF production leads to elevated interferon and a concurrent reduction in viral loads with a loss of pathogenicity. Palmitoylation of TF has been shown to be important for particle assembly and morphology. We find that palmitoylation of TF also contributes to the ability of TF to antagonize host interferon responses as dysregulated palmitoylation of TF reduces virulence in a manner similar to loss of TF.


Asunto(s)
Interferón Tipo I/biosíntesis , Virus Sindbis/inmunología , Virus Sindbis/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Animales , Femenino , Genes Virales , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Interacciones Microbiota-Huesped/fisiología , Lipoilación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Virus Sindbis/genética , Proteínas Virales/genética , Virulencia/genética , Virulencia/inmunología , Virulencia/fisiología
12.
mBio ; 9(4)2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30131356

RESUMEN

Infection with Ross River virus (RRV) causes debilitating polyarthritis and arthralgia in individuals. Alphaviruses are highly sensitive to type I interferon (IFN). Mutations at the conserved P3 position of the cleavage site between nonstructural protein 1 (nsP1) and nsP2 (1/2 site) modulate type I IFN induction for both RRV and Sindbis virus (SINV). We constructed and characterized RRV-T48A534V, a mutant harboring an A534V substitution in the P1 position of the 1/2 site, and compared it to parental RRV-T48 and to RRV-T48A532V, SINVI538 and SINVT538 harboring different substitutions in the same region. A534V substitution resulted in impaired processing of RRV nonstructural polyprotein and in elevated production of replicase-generated pathogen-associated molecular pattern (PAMP) RNAs that induce expression of type I IFN. Both A532V and A534V substitutions affected synthesis of viral RNAs, though the effects of these closely located mutations were drastically different affecting mostly either the viral negative-strand RNA or genomic and subgenomic RNA levels, respectively. Synthesis of PAMP RNAs was also observed for SINV replicase, and it was increased by I538T substitution. In comparison to RRV-T48, RRV-T48A534V was attenuated in vitro and in vivo Interestingly, when type I IFN-deficient cells and type I IFN receptor-deficient mice were infected with RRV-T48 or RRV-T48A534V, differences between these viruses were no longer apparent. Compared to RRV-T48, RRV-T48A534V infection was associated with increased upregulation of type I IFN signaling proteins. We demonstrate novel mechanisms by which the A534V mutation affect viral nonstructural polyprotein processing that can impact PAMP RNA production, type I IFN induction/sensitivity, and disease.IMPORTANCE This study gives further insight into mechanisms of type I IFN modulation by the medically important alphaviruses Ross River virus (RRV) and Sindbis virus (SINV). By characterizing attenuated RRV mutants, the crucial role of amino acid residues in P1 and P3 positions (the first and third amino acid residues preceding the scissile bond) of the cleavage site between nsP1 and nsP2 regions was highlighted. The study uncovers a unique relationship between alphavirus nonstructural polyprotein processing, RNA replication, production of different types of pathogen-associated molecular pattern (PAMP) RNAs, type I IFN induction, and disease pathogenesis. This study also highlights the importance of the host innate immune response in RRV infections. The viral determinants of type I IFN modulation provide potential drug targets for clinical treatment of alphaviral disease and offer new approaches for rational attenuation of alphaviruses for construction of vaccine candidates.


Asunto(s)
Interferones/metabolismo , Proteínas Mutantes/inmunología , Mutación Missense , Poliproteínas/metabolismo , ARN Viral/inmunología , Virus del Río Ross/patogenicidad , Proteínas no Estructurales Virales/metabolismo , Infecciones por Alphavirus/patología , Infecciones por Alphavirus/virología , Animales , Antivirales/metabolismo , Modelos Animales de Enfermedad , Ratones , Proteínas Mutantes/genética , Poliproteínas/genética , ARN Viral/metabolismo , Virus del Río Ross/genética , Virus del Río Ross/inmunología , Virus Sindbis/genética , Virus Sindbis/inmunología , Virus Sindbis/patogenicidad , Proteínas no Estructurales Virales/genética , Virulencia
13.
J Virol ; 92(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29321331

RESUMEN

Sindbis virus (SINV) infection of neurons in the brain and spinal cord in mice provides a model system for investigating recovery from encephalomyelitis and antibody-mediated clearance of virus from the central nervous system (CNS). To determine the roles of IgM and IgG in recovery, we compared the responses of immunoglobulin-deficient activation-induced adenosine deaminase-deficient (AID-/-), secretory IgM-deficient (sIgM-/-), and AID-/- sIgM-/- double-knockout (DKO) mice with those of wild-type (WT) C57BL/6 mice for disease, clearance of infectious virus and viral RNA from brain and spinal cord, antibody responses, and B cell infiltration into the CNS. Because AID is essential for immunoglobulin class switch recombination and somatic hypermutation, AID-/- mice produce only germ line IgM, while sIgM-/- mice secrete IgG but no IgM and DKO mice produce no secreted immunoglobulin. After intracerebral infection with the TE strain of SINV, most mice recovered. Development of neurologic disease occurred slightly later in sIgM-/- mice, but disease severity, weight loss, and survival were similar between the groups. AID-/- mice produced high levels of SINV-specific IgM, while sIgM-/- mice produced no IgM and high levels of IgG2a compared to WT mice. All mice cleared infectious virus from the spinal cord, but DKO mice failed to clear infectious virus from brain and had higher levels of viral RNA in the CNS late after infection. The numbers of infected cells and the amount of cell death in brain were comparable. We conclude that antibody is required and that either germ line IgM or IgG is sufficient for clearance of virus from the CNS.IMPORTANCE Mosquito-borne alphaviruses that infect neurons can cause fatal encephalomyelitis. Recovery requires a mechanism for the immune system to clear virus from infected neurons without harming the infected cells. Antiviral antibody has previously been shown to be a noncytolytic means for alphavirus clearance. Antibody-secreting cells enter the nervous system after infection and produce antiviral IgM before IgG. Clinical studies of human viral encephalomyelitis suggest that prompt production of IgM is associated with recovery, but it was not known whether IgM is effective for clearance. Our studies used mice deficient in production of IgM, IgG, or both to characterize the antibody necessary for alphavirus clearance. All mice developed similar signs of neurologic disease and recovered from infection. Antibody was necessary for virus clearance from the brain, and either early germ line IgM or IgG was sufficient. These studies support the clinical observation that prompt production of antiviral antibody is a determinant of outcome.


Asunto(s)
Infecciones por Alphavirus/inmunología , Anticuerpos Antivirales/inmunología , Encéfalo/inmunología , Infecciones del Sistema Nervioso Central/inmunología , Inmunoglobulina M/inmunología , Virus Sindbis/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/patología , Animales , Anticuerpos Antivirales/genética , Encéfalo/patología , Encéfalo/virología , Línea Celular , Infecciones del Sistema Nervioso Central/genética , Infecciones del Sistema Nervioso Central/patología , Cricetinae , Citidina Desaminasa/deficiencia , Femenino , Inmunoglobulina M/genética , Ratones , Ratones Noqueados , Virus Sindbis/genética
14.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263262

RESUMEN

Alphaviruses are an important cause of mosquito-borne outbreaks of arthritis, rash, and encephalomyelitis. Previous studies in mice with a virulent strain (neuroadapted SINV [NSV]) of the alphavirus Sindbis virus (SINV) identified a role for Th17 cells and regulation by interleukin-10 (IL-10) in the pathogenesis of fatal encephalomyelitis (K. A. Kulcsar, V. K. Baxter, I. P. Greene, and D. E. Griffin, Proc Natl Acad Sci U S A 111:16053-16058, 2014, https://doi.org/10.1073/pnas.1418966111). To determine the role of virus virulence in generation of immune responses, we analyzed the modulatory effects of IL-10 on disease severity, virus clearance, and the CD4+ T cell response to infection with a recombinant strain of SINV of intermediate virulence (TE12). The absence of IL-10 during TE12 infection led to longer morbidity, more weight loss, higher mortality, and slower viral clearance than in wild-type mice. More severe disease and impaired virus clearance in IL-10-/- mice were associated with more Th1 cells, fewer Th2 cells, innate lymphoid type 2 cells, regulatory cells, and B cells, and delayed production of antiviral antibody in the central nervous system (CNS) without an effect on Th17 cells. Therefore, IL-10 deficiency led to more severe disease in TE12-infected mice by increasing Th1 cells and by hampering development of the local B cell responses necessary for rapid production of antiviral antibody and virus clearance from the CNS. In addition, the shift from Th17 to Th1 responses with decreased virus virulence indicates that the effects of IL-10 deficiency on immunopathologic responses in the CNS during alphavirus infection are influenced by virus strain.IMPORTANCE Alphaviruses cause mosquito-borne outbreaks of encephalomyelitis, but determinants of outcome are incompletely understood. We analyzed the effects of the anti-inflammatory cytokine IL-10 on disease severity and virus clearance after infection with an alphavirus strain of intermediate virulence. The absence of IL-10 led to longer illness, more weight loss, more death, and slower viral clearance than in mice that produced IL-10. IL-10 influenced development of disease-causing T cells and entry into the brain of B cells producing antiviral antibody. The Th1 pathogenic cell subtype that developed in IL-10-deficient mice infected with a less virulent virus was distinct from the Th17 subtype that developed in response to a more virulent virus, indicating a role for virus strain in determining the immune response. Slow production of antibody in the nervous system led to delayed virus clearance. Therefore, both the virus strain and the host response to infection are important determinants of outcome.


Asunto(s)
Infecciones por Alphavirus/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Encefalomielitis/inmunología , Interleucina-10/inmunología , Virus Sindbis/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/patología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/genética , Linfocitos B/patología , Linfocitos T CD4-Positivos/patología , Línea Celular , Cricetinae , Encefalomielitis/genética , Encefalomielitis/patología , Interleucina-10/genética , Ratones , Ratones Noqueados , Virus Sindbis/genética , Virus Sindbis/patogenicidad
15.
Cell Host Microbe ; 22(1): 48-60.e5, 2017 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-28704652

RESUMEN

Asymptomatic infections often proceed undetected, yet can still prime the host to be sensitive to secondary environmental stress. While the mechanisms underlying disease caused by asymptomatic infections are unknown, it is believed that productive pathogen replication is required. We report that the environmental stress of carbon dioxide (CO2) anesthesia converts an asymptomatic rhabdovirus infection in Drosophila to one that is lethal. This lethality results from a pool of infectious virus in glial cells and is regulated by the antiviral RNAi pathway of the host. CO2 sensitivity is caused by the fusogenic activity of the viral glycoprotein, which results in fusion of neurons and glia. Expression of the viral glycoprotein, but not a fusion defective mutant, is sufficient to cause CO2 sensitivity, which can occur even in the absence of productive viral replication. These findings highlight how viral proteins, independent of pathogen replication, may predispose hosts to life-threatening environmental stress.


Asunto(s)
Dióxido de Carbono/farmacología , Ambiente , Neuronas/virología , Estrés Fisiológico , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Virosis , Animales , Animales Modificados Genéticamente , Anopheles/inmunología , Anopheles/virología , Antivirales/farmacología , Conducta Animal , Drosophila melanogaster/inmunología , Drosophila melanogaster/virología , Glicoproteínas/metabolismo , Concentración de Iones de Hidrógeno , Inmunidad Innata , Neuroglía/inmunología , Neuroglía/virología , Neuronas/inmunología , Interferencia de ARN , Virus Sindbis/inmunología , Virus Sindbis/patogenicidad , Ensayo de Placa Viral , Proteínas Virales/metabolismo , Virosis/inmunología , Virosis/virología , Replicación Viral/efectos de los fármacos
16.
J Gen Virol ; 98(5): 992-1003, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28555544

RESUMEN

Viral RNA studies often rely on in situ hybridization and reverse transcriptase-PCR to provide snapshots of RNA dynamics in infected cells. To facilitate analysis of cellular RNAs, aptamers Spinach and Spinach2 that bind and activate the conditional fluorophore 3, 5-difluoro-4-hydroxybenzylidene imidazolinon have been developed. To determine the feasibility of applying this technology to viral RNA, we have used cDNA clones of the TE strain of Sindbis virus (SINV) to construct multiple viruses containing one or two copies of tRNA-scaffolded Spinach2 after a second subgenomic promoter, TEds-1Sp and TEds-2Sp within the 3'UTR, TE-1UTRSp, or after a second subgenomic promoter and in the 3'UTR, TEds-1Sp+1 UTRSp. TEds-1Sp+1 UTRSp gave the brightest signal and replicated well in cell culture, while TEds-2Sp was the dimmest and replicated poorly. Selection of baby hamster kidney cells infected with TEds-1Sp+1 UTRSp for improved signal intensity identified a virus with a stronger signal and point mutations in the tRNA scaffold. Imaging of SINV in BHK cells showed RNA to be concentrated in filopodia that contacted and transferred RNA to adjacent cells. The effect of treatment with anti-E2 antibody, which effects non-cytolytic clearance of SINV from neurons, on viral RNA was cell-type-dependent. In antibody-treated BHK cells, intracellular viral RNA increased and spread of infection continued. In undifferentiated and differentiated AP7 neuronal cells antibody treatment induced viral RNA clearance. Both viruses with two inserted aptamers were prone to deletion. These studies form the basis for further development of aptamer-labelled viral RNAs that will facilitate functional studies on the dynamics of infection and clearance.


Asunto(s)
Biología Molecular/métodos , ARN Viral/análisis , Virus Sindbis/inmunología , Virus Sindbis/fisiología , Coloración y Etiquetado/métodos , Virología/métodos , Replicación Viral , Animales , Aptámeros de Nucleótidos/análisis , Células Cultivadas , Cricetinae
17.
Proc Natl Acad Sci U S A ; 114(10): 2681-2686, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28213497

RESUMEN

The innate immune system senses RNA viruses by pattern recognition receptors (PRRs) and protects the host from virus infection. PRRs mediate the production of immune modulatory factors and direct the elimination of RNA viruses. Here, we show a unique PRR that mediates antiviral response. Tetrachlorodibenzo-p-dioxin (TCDD)-inducible poly(ADP ribose) polymerase (TIPARP), a Cysteine3 Histidine (CCCH)-type zinc finger-containing protein, binds to Sindbis virus (SINV) RNA via its zinc finger domain and recruits an exosome to induce viral RNA degradation. TIPARP typically localizes in the nucleus, but it accumulates in the cytoplasm after SINV infection, allowing targeting of cytoplasmic SINV RNA. Redistribution of TIPARP is induced by reactive oxygen species (ROS)-dependent oxidization of the nuclear pore that affects cytoplasmic-nuclear transport. BCL2-associated X protein (BAX) and BCL2 antagonist/killer 1 (BAK1), B-cell leukemia/lymphoma 2 (BCL2) family members, mediate mitochondrial damage to generate ROS after SINV infection. Thus, TIPARP is a viral RNA-sensing PRR that mediates antiviral responses triggered by BAX- and BAK1-dependent mitochondrial damage.


Asunto(s)
Inmunidad Innata/genética , Poli(ADP-Ribosa) Polimerasas/genética , Virus ARN/genética , Receptores de Reconocimiento de Patrones/genética , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/inmunología , Citoplasma/genética , Citoplasma/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Mitocondrias/genética , Mitocondrias/patología , Mitocondrias/virología , Proteínas de Transporte de Nucleósidos , Poli(ADP-Ribosa) Polimerasas/inmunología , Virus ARN/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Virus Sindbis/genética , Virus Sindbis/inmunología , Virus Sindbis/patogenicidad , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/inmunología , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/inmunología
18.
J Exp Med ; 213(13): 2931-2947, 2016 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-27899441

RESUMEN

The host responds to virus infection by activating type I interferon (IFN) signaling leading to expression of IFN-stimulated genes (ISGs). Dysregulation of the IFN response results in inflammatory diseases and chronic infections. In this study, we demonstrate that IFN regulatory factor 2 (IRF2), an ISG and a negative regulator of IFN signaling, influences alphavirus neuroinvasion and pathogenesis. A Sindbis virus strain that in wild-type (WT) mice only causes disease when injected into the brain leads to lethal encephalitis in Irf2-/- mice after peripheral inoculation. Irf2-/- mice fail to control virus replication and recruit immune infiltrates into the brain. Reduced B cells and virus-specific IgG are observed in the Irf2-/- mouse brains despite the presence of peripheral neutralizing antibodies, suggesting a defect in B cell trafficking to the central nervous system (CNS). B cell-deficient µMT mice are significantly more susceptible to viral infection, yet WT B cells and serum are unable to rescue the Irf2-/- mice. Collectively, our data demonstrate that proper localization of B cells and local production of antibodies in the CNS are required for protection. The work advances our understanding of host mechanisms that affect viral neuroinvasion and their contribution to immunity against CNS infections.


Asunto(s)
Infecciones por Alphavirus/inmunología , Linfocitos B/inmunología , Encefalopatías/inmunología , Movimiento Celular/inmunología , Factor 2 Regulador del Interferón/inmunología , Virus Sindbis/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/patología , Animales , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Linfocitos B/patología , Encefalopatías/genética , Encefalopatías/patología , Encefalopatías/virología , Movimiento Celular/genética , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Factor 2 Regulador del Interferón/genética , Ratones , Ratones Noqueados
19.
J Gen Virol ; 97(11): 2908-2925, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27667782

RESUMEN

Infection of mice with Sindbis virus (SINV) produces encephalomyelitis and provides a model for examination of the central nervous system (CNS) immune response to alphavirus infection. Clearance of infectious virus is accomplished through a cooperative effort between SINV-specific antibody and IFN-γ, but the regulatory interactions are poorly understood. To determine the effects of IFN-γ on clinical disease and the antiviral immune response, C57BL/6 mice lacking IFN-γ (Ifng-/-) or IFN-γ receptor (Ifngr1-/-) were studied in comparison to WT mice. Maximum production of Ifng mRNA and IFN-γ protein in the CNS of WT and Ifngr1-/- mice occurred 5-7 days after infection, with higher levels of IFN-γ in Ifngr1-/- mice. Onset of clinical disease was earlier in mice with impaired IFN-γ signalling, although Ifngr1-/- mice recovered more rapidly. Ifng-/- and Ifngr1-/- mice maintained body weight better than WT mice, associated with better food intake and lower brain levels of inflammatory cytokines. Clearance of infectious virus from the spinal cords was slower, and CNS, but not serum, levels of SINV-specific IgM, IgG2a and IgG2b were lower in Ifngr1-/- and Ifng-/- mice compared to WT mice. Decreased CNS antiviral antibody was associated with lower expression of mRNAs for B-cell attracting chemokines CXCL9, CXCL10 and CXCL13 and fewer B cells in the CNS. Therefore, IFN-γ signalling increases levels of CNS pro-inflammatory cytokines, leading to clinical disease, but synergistically clears virus with SINV-specific antibody at least in part by increasing chemokine production important for infiltration of antibody-secreting B cells into the CNS.


Asunto(s)
Infecciones por Alphavirus/inmunología , Anticuerpos Antivirales/inmunología , Encefalomielitis/inmunología , Interferón gamma/inmunología , Virus Sindbis/inmunología , Infecciones por Alphavirus/virología , Animales , Formación de Anticuerpos , Linfocitos B/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Citocinas/inmunología , Encefalomielitis/virología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus Sindbis/fisiología
20.
J Virol ; 90(22): 10247-10258, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27581990

RESUMEN

Viral infection induces production of type I interferons (IFNs), which stimulate the expression of a variety of antiviral factors to inhibit viral replication. To establish effective infection, viruses need to develop strategies to evade the immune responses. A neurovirulent Sindbis virus strain with neuroinvasive properties (SVNI) causes lethal encephalitis in mice, and its replication in cultured cells is inhibited by the zinc finger antiviral protein (ZAP), a host factor that specifically inhibits the replication of certain viruses by binding to the viral mRNAs, repressing the translation of target mRNA, and promoting the degradation of target mRNA. We report here that murine embryonic fibroblast cells from ZAP knockout mice supported more efficient SVNI replication than wild-type cells. SVNI infection of 10-day-old suckling mice led to reduced survival in the knockout mice. Unexpectedly, however, SVNI infection of 23-day-old weanling mice, whose immune system is more developed than that of the suckling mice, resulted in significantly improved survival in ZAP knockout mice. Further analyses revealed that in the weanling knockout mice, SVNI replicated more efficiently in lymphoid tissues at early times postinfection and induced higher levels of IFN production, which restricted viral spread to the central nervous system. Blocking IFN activity through the use of receptor-neutralizing antibodies rendered knockout mice more sensitive to SVNI infection than wild-type mice. These results uncover a mechanism by which SVNI exploits a host antiviral factor to evade innate immune surveillance. IMPORTANCE: Sindbis virus, a prototypic member of the Alphavirus genus, has been used to study the pathogenesis of acute viral encephalitis in mice for many years. How the virus evades immune surveillance to establish effective infection is largely unknown. ZAP is a host antiviral factor that potently inhibits Sindbis virus replication in cell culture. We show here that infection of ZAP knockout suckling mice with an SVNI led to faster disease progression. However, SVNI infection of weanling mice led to slower disease progression in knockout mice. Further analyses revealed that in weanling knockout mice, SVNI replicated more efficiently in lymphoid tissues at early times postinfection and induced higher levels of interferon production, which restricted viral spread to the central nervous system. These results uncover a mechanism by which SVNI exploits a host antiviral factor to evade innate immune surveillance and allow enhanced neuroinvasion.


Asunto(s)
Infecciones por Alphavirus/inmunología , Antivirales/inmunología , Virus Sindbis/inmunología , Infecciones por Alphavirus/virología , Animales , Línea Celular , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Cricetinae , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al ARN/inmunología , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...