Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Infect Dis ; 225(10): 1765-1772, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-33507266

RESUMEN

BACKGROUND: REGN3048 and REGN3051 are human monoclonal antibodies (mAb) targeting the spike glycoprotein on the Middle East respiratory syndrome coronavirus (MERS-CoV), which binds to the receptor dipeptidyl peptidase-4 (DPP4) and is necessary for infection of susceptible cells. METHODS: Preclinical study: REGN3048, REGN3051 and isotype immunoglobulin G (IgG) were administered to humanized DPP4 (huDPP4) mice 1 day prior to and 1 day after infection with MERS-CoV (Jordan strain). Virus titers and lung pathology were assessed. Phase 1 study: healthy adults received the combined mAb (n = 36) or placebo (n = 12) and followed for 121 days. Six dose levels were studied. Strict safety criteria were met prior to dose escalation. RESULTS: Preclinical study: REGN3048 plus REGN3051, prophylactically or therapeutically, was substantially more effective for reducing viral titer, lung inflammation, and pathology in huDPP4 mice compared with control antibodies and to each antibody monotherapy. Phase 1 study: REGN3048 plus REGN3051 was well tolerated with no dose-limiting adverse events, deaths, serious adverse events, or infusion reactions. Each mAb displayed pharmacokinetics expected of human IgG1 antibodies; it was not immunogenic. CONCLUSIONS: REGN3048 and REGN3051 in combination were well tolerated. The clinical and preclinical data support further development for the treatment or prophylaxis of MERS-CoV infection.


Asunto(s)
Infecciones por Coronavirus , Coronavirus del Síndrome Respiratorio de Oriente Medio , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Dipeptidil Peptidasa 4/metabolismo , Humanos , Inmunoglobulina G , Ratones , Glicoproteína de la Espiga del Coronavirus
2.
Curr Biol ; 31(2): 257-270.e5, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33157026

RESUMEN

Marburg virus (MARV) is among the most virulent pathogens of primates, including humans. Contributors to severe MARV disease include immune response suppression and inflammatory gene dysregulation ("cytokine storm"), leading to systemic damage and often death. Conversely, MARV causes little to no clinical disease in its reservoir host, the Egyptian rousette bat (ERB). Previous genomic and in vitro data suggest that a tolerant ERB immune response may underlie MARV avirulence, but no significant examination of this response in vivo yet exists. Here, using colony-bred ERBs inoculated with a bat isolate of MARV, we use species-specific antibodies and an immune gene probe array (NanoString) to temporally characterize the transcriptional host response at sites of MARV replication relevant to primate pathogenesis and immunity, including CD14+ monocytes/macrophages, critical immune response mediators, primary MARV targets, and skin at the inoculation site, where highest viral loads and initial engagement of antiviral defenses are expected. Our analysis shows that ERBs upregulate canonical antiviral genes typical of mammalian systems, such as ISG15, IFIT1, and OAS3, yet demonstrate a remarkable lack of significant induction of proinflammatory genes classically implicated in primate filoviral pathogenesis, including CCL8, FAS, and IL6. Together, these findings offer the first in vivo functional evidence for disease tolerance as an immunological mechanism by which the bat reservoir asymptomatically hosts MARV. More broadly, these data highlight factors determining disparate outcomes between reservoir and spillover hosts and defensive strategies likely utilized by bat hosts of other emerging pathogens, knowledge that may guide development of effective antiviral therapies.


Asunto(s)
Quirópteros/inmunología , Reservorios de Enfermedades/virología , Tolerancia Inmunológica/inmunología , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/inmunología , Animales , Infecciones Asintomáticas , Quirópteros/sangre , Quirópteros/genética , Quirópteros/virología , Femenino , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Tolerancia Inmunológica/genética , Masculino , Enfermedad del Virus de Marburg/virología , Monocitos/inmunología
3.
Front Immunol ; 11: 26, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117225

RESUMEN

In recent years, viruses similar to those that cause serious disease in humans and other mammals have been detected in apparently healthy bats. These include filoviruses, paramyxoviruses, and coronaviruses that cause severe diseases such as Ebola virus disease, Marburg haemorrhagic fever and severe acute respiratory syndrome (SARS) in humans. The evolution of flight in bats seem to have selected for a unique set of antiviral immune responses that control virus propagation, while limiting self-damaging inflammatory responses. Here, we summarize our current understanding of antiviral immune responses in bats and discuss their ability to co-exist with emerging viruses that cause serious disease in other mammals. We highlight how this knowledge may help us to predict viral spillovers into new hosts and discuss future directions for the field.


Asunto(s)
Quirópteros/inmunología , Quirópteros/virología , Virus ADN/inmunología , Adaptación al Huésped/inmunología , Sistema Inmunológico/virología , Virus ARN/inmunología , Inmunidad Adaptativa , Animales , Reservorios de Enfermedades/virología , Evolución Molecular , Inmunidad Innata , Interferones/metabolismo , Zoonosis Virales/inmunología , Zoonosis Virales/transmisión
4.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31550243

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) emerged in 2012 in Saudi Arabia and has caused over 2400 cases and more than 800 deaths. Epidemiological studies identified diabetes as the primary comorbidity associated with severe or lethal MERS-CoV infection. Understanding how diabetes affects MERS is important because of the global burden of diabetes and pandemic potential of MERS-CoV. We used a model in which mice were made susceptible to MERS-CoV by expressing human DPP4, and type 2 diabetes was induced by administering a high-fat diet. Upon infection with MERS-CoV, diabetic mice had a prolonged phase of severe disease and delayed recovery that was independent of virus titers. Histological analysis revealed that diabetic mice had delayed inflammation, which was then prolonged through 21 days after infection. Diabetic mice had fewer inflammatory monocyte/macrophages and CD4+ T cells, which correlated with lower levels of Ccl2 and Cxcl10 expression. Diabetic mice also had lower levels of Tnfa, Il6, Il12b, and Arg1 expression and higher levels of Il17a expression. These data suggest that the increased disease severity observed in individuals with MERS and comorbid type 2 diabetes is likely due to a dysregulated immune response, which results in more severe and prolonged lung pathology.


Asunto(s)
Infecciones por Coronavirus/inmunología , Diabetes Mellitus Tipo 2/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Quimiocina CCL2/análisis , Quimiocina CCL2/metabolismo , Quimiocina CXCL10/análisis , Quimiocina CXCL10/metabolismo , Comorbilidad , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Dieta Alta en Grasa/efectos adversos , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Coronavirus del Síndrome Respiratorio de Oriente Medio/aislamiento & purificación , Monocitos/inmunología , Monocitos/metabolismo , Factores de Riesgo , Índice de Severidad de la Enfermedad
5.
Viruses ; 11(1)2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30634396

RESUMEN

Bats are speculated to be reservoirs of several emerging viruses including coronaviruses (CoVs) that cause serious disease in humans and agricultural animals. These include CoVs that cause severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), porcine epidemic diarrhea (PED) and severe acute diarrhea syndrome (SADS). Bats that are naturally infected or experimentally infected do not demonstrate clinical signs of disease. These observations have allowed researchers to speculate that bats are the likely reservoirs or ancestral hosts for several CoVs. In this review, we follow the CoV outbreaks that are speculated to have originated in bats. We review studies that have allowed researchers to identify unique adaptation in bats that may allow them to harbor CoVs without severe disease. We speculate about future studies that are critical to identify how bats can harbor multiple strains of CoVs and factors that enable these viruses to "jump" from bats to other mammals. We hope that this review will enable readers to identify gaps in knowledge that currently exist and initiate a dialogue amongst bat researchers to share resources to overcome present limitations.


Asunto(s)
Quirópteros/virología , Coronavirus/fisiología , Brotes de Enfermedades , Reservorios de Enfermedades/veterinaria , Animales , Enfermedades Asintomáticas , Coronavirus/patogenicidad , Infecciones por Coronavirus , Reservorios de Enfermedades/virología , Evolución Molecular , Genoma Viral , Humanos , Filogenia , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Síndrome Respiratorio Agudo Grave , Porcinos
6.
Cell ; 173(5): 1098-1110.e18, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29706541

RESUMEN

Bats harbor many viruses asymptomatically, including several notorious for causing extreme virulence in humans. To identify differences between antiviral mechanisms in humans and bats, we sequenced, assembled, and analyzed the genome of Rousettus aegyptiacus, a natural reservoir of Marburg virus and the only known reservoir for any filovirus. We found an expanded and diversified KLRC/KLRD family of natural killer cell receptors, MHC class I genes, and type I interferons, which dramatically differ from their functional counterparts in other mammals. Such concerted evolution of key components of bat immunity is strongly suggestive of novel modes of antiviral defense. An evaluation of the theoretical function of these genes suggests that an inhibitory immune state may exist in bats. Based on our findings, we hypothesize that tolerance of viral infection, rather than enhanced potency of antiviral defenses, may be a key mechanism by which bats asymptomatically host viruses that are pathogenic in humans.


Asunto(s)
Quirópteros/genética , Genoma , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Quirópteros/clasificación , Quirópteros/inmunología , Mapeo Cromosómico , Reservorios de Enfermedades/virología , Egipto , Evolución Molecular , Variación Genética , Antígenos de Histocompatibilidad Clase I/clasificación , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Interferón Tipo I/clasificación , Interferón Tipo I/genética , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/patología , Marburgvirus/fisiología , Subfamília C de Receptores Similares a Lectina de Células NK/química , Subfamília C de Receptores Similares a Lectina de Células NK/clasificación , Subfamília C de Receptores Similares a Lectina de Células NK/genética , Subfamília D de Receptores Similares a Lectina de las Células NK/química , Subfamília D de Receptores Similares a Lectina de las Células NK/clasificación , Subfamília D de Receptores Similares a Lectina de las Células NK/genética , Filogenia , Alineación de Secuencia
7.
Sci Rep ; 7(1): 14756, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29116224

RESUMEN

Ebola virus disease (EVD) is a serious illness with mortality rates of 20-90% in various outbreaks. EVD is characterized by robust virus replication and strong host inflammatory response. Analyzing host immune responses has increasingly involved multimodal approaches including transcriptomics to profile gene expression. We studied cynomolgus macaques exposed to Ebola virus Makona via different routes with the intent of comparing RNA-Seq to a NanoString nCounter codeset targeting 769 non-human primate (NHP) genes. RNA-Seq analysis of serial blood samples showed different routes led to the same overall transcriptional response seen in previously reported EBOV-exposed NHP studies. Both platforms displayed a strong correlation in gene expression patterns, including a strong induction of innate immune response genes at early times post-exposure, and neutrophil-associated genes at later time points. A 41-gene classifier was tested in both platforms for ability to cluster samples by infection status. Both NanoString and RNA-Seq could be used to predict relative abundances of circulating immune cell populations that matched traditional hematology. This demonstrates the complementarity of RNA-Seq and NanoString. Moreover, the development of an NHP-specific NanoString codeset should augment studies of filoviruses and other high containment infectious diseases without the infrastructure requirements of RNA-Seq technology.


Asunto(s)
Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/genética , Transcriptoma , Animales , Modelos Animales de Enfermedad , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/inmunología , Humanos , Inmunidad Innata , Inmunidad Mucosa , Macaca fascicularis , Análisis de Secuencia de ARN , Transducción de Señal , Virulencia
8.
J Neuroimmunol ; 295-296: 60-7, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27235350

RESUMEN

Neuroadapted Sindbis virus infection of mice causes T cell-mediated fatal encephalomyelitis. In the absence of IL-10, pathogenic Th17 cells are increased and disease is accelerated. Lymphoid and myeloid cell contributions to IL-10 production were determined using VertX IL-10 transcriptional eGFP reporter mice. Effector and regulatory CD4(+) and CD8(+) T cells in the brain, but not the cervical lymph nodes, were the primary producers of IL-10. Th17 and Th1/Th17 cells were increased in mice that lacked T cell IL-10 production, although less than in the absence of IL-10. Morbidity and mortality were not affected suggesting an IL-10 threshold for disease exacerbation.


Asunto(s)
Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/patología , Encefalomielitis/inmunología , Encefalomielitis/patología , Interleucina-10/metabolismo , Células Th17/metabolismo , Alphavirus/patogenicidad , Infecciones por Alphavirus/genética , Animales , Antígenos CD/metabolismo , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Modelos Animales de Enfermedad , Encefalomielitis/genética , Encefalomielitis/virología , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estadísticas no Paramétricas , Células Th17/patología
9.
BMC Genomics ; 16: 1033, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26643810

RESUMEN

BACKGROUND: The Egyptian Rousette bat (Rousettus aegyptiacus), a common fruit bat species found throughout Africa and the Middle East, was recently identified as a natural reservoir host of Marburg virus. With Ebola virus, Marburg virus is a member of the family Filoviridae that causes severe hemorrhagic fever disease in humans and nonhuman primates, but results in little to no pathological consequences in bats. Understanding host-pathogen interactions within reservoir host species and how it differs from hosts that experience severe disease is an important aspect of evaluating viral pathogenesis and developing novel therapeutics and methods of prevention. RESULTS: Progress in studying bat reservoir host responses to virus infection is hampered by the lack of host-specific reagents required for immunological studies. In order to establish a basis for the design of reagents, we sequenced, assembled, and annotated the R. aegyptiacus transcriptome. We performed de novo transcriptome assembly using deep RNA sequencing data from 11 distinct tissues from one male and one female bat. We observed high similarity between this transcriptome and those available from other bat species. Gene expression analysis demonstrated clustering of expression profiles by tissue, where we also identified enrichment of tissue-specific gene ontology terms. In addition, we identified and experimentally validated the expression of novel coding transcripts that may be specific to this species. CONCLUSION: We comprehensively characterized the R. aegyptiacus transcriptome de novo. This transcriptome will be an important resource for understanding bat immunology, physiology, disease pathogenesis, and virus transmission.


Asunto(s)
Quirópteros/genética , Biología Computacional , Anotación de Secuencia Molecular , Transcriptoma , Animales , Análisis por Conglomerados , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Reproducibilidad de los Resultados
10.
PLoS One ; 10(10): e0141439, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26505998

RESUMEN

The apoptotic cascade is an orchestrated event, whose final stages are mediated by effector caspases. Regulatory binding proteins have been identified for caspases such as caspase-3, -7, -8, and -9. Many of these proteins belong to the inhibitor of apoptosis (IAP) family. By contrast, caspase-6 is not believed to be influenced by IAPs, and little is known about its regulation. We therefore performed a yeast-two-hybrid screen using a constitutively inactive form of caspase-6 for bait in order to identify novel regulators of caspase-6 activity. Sox11 was identified as a potential caspase-6 interacting protein. Sox11 was capable of dramatically reducing caspase-6 activity, as well as preventing caspase-6 self- cleavage. Several regions, including amino acids 117-214 and 362-395 within sox11 as well as a nuclear localization signal (NLS) all contributed to the reduction in caspase-6 activity. Furthermore, sox11 was also capable of decreasing other effector caspase activity but not initiator caspases -8 and -9. The ability of sox11 to reduce effector caspase activity was also reflected in its capacity to reduce cell death following toxic insult. Interestingly, other sox proteins also had the ability to reduce caspase-6 activity but to a lesser extent than sox11.


Asunto(s)
Apoptosis/genética , Caspasa 6/genética , Factores de Transcripción SOXC/genética , Proteínas Portadoras , Caspasa 6/biosíntesis , Caspasa 6/metabolismo , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , Neuronas/metabolismo , Señales de Localización Nuclear/biosíntesis , Señales de Localización Nuclear/genética , Mapas de Interacción de Proteínas/genética , Factores de Transcripción SOXC/biosíntesis , Factores de Transcripción SOXC/metabolismo , Factores de Transcripción
11.
J Virol ; 89(16): 8280-91, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26041298

RESUMEN

UNLABELLED: Susceptibility to alphavirus encephalomyelitis is dependent on a variety of factors, including the genetic background of the host. Neuroadapted Sindbis virus (NSV) causes uniformly fatal disease in adult C57BL/6 (B6) mice, but adult BALB/c (Bc) mice recover from infection. In B6 mice, fatal encephalomyelitis is immune mediated rather than a direct result of virus infection. To identify the immunological determinants of host susceptibility to fatal NSV-induced encephalomyelitis, we compared virus titers and immune responses in adult B6 and Bc mice infected intranasally with NSV. B6 mice had higher levels of virus replication, higher levels of type I interferon (IFN), and slower virus clearance than did Bc mice. B6 mice had more neuronal apoptosis, more severe neurologic disease, and higher mortality than Bc mice. B6 mice had more infiltration of inflammatory cells and higher levels of IL1b, IL-6, TNFa, Csf2, and CCL2 mRNAs and interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), IFN-γ, and C-C motif ligand 2 (CCL2) protein in brains than Bc mice. However, Bc mice had more brain antibody at day 7 and a higher percentage of CD4(+) T cells. CD4(+) T cells in the brains of Bc mice included fewer Th17 cells and more regulatory T cells (Tregs) producing IL-10 than B6 mice, accompanied by higher levels of Il2 and Cxcl10 mRNAs. In the absence of IL-10, resistant Bc mice became susceptible to fatal encephalomyelitis after NSV infection. These studies demonstrate the importance of the immune response and its regulation in determining host survival during alphavirus encephalomyelitis. IMPORTANCE: Mosquito-borne alphavirus infections are an important cause of encephalomyelitis in humans. The severity of disease is dependent both on the strain of the virus and on the age and genetic background of the host. A neurovirulent strain of Sindbis virus causes immune-mediated fatal encephalomyelitis in adult C57BL/6 mice but not in BALB/c mice. To determine the host-dependent immunological mechanisms underlying the differences in susceptibility between these two strains of mice, we compared their immune responses to infection. Resistance to fatal disease in BALB/c mice was associated with better antibody responses, more-rapid virus clearance, fewer Th17 cells, and more-potent regulatory T cell responses than occurred in susceptible C57BL/6 mice. In the absence of interleukin-10, a component of the regulatory immune response, resistant mice became susceptible to lethal disease. This study demonstrates the importance of the immune response and its regulation for host survival during alphavirus encephalomyelitis.


Asunto(s)
Alphavirus/patogenicidad , Susceptibilidad a Enfermedades/inmunología , Encefalomielitis/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Citocinas/genética , Encefalomielitis/virología , Expresión Génica , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Virulencia
12.
Proc Natl Acad Sci U S A ; 111(45): 16053-8, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25362048

RESUMEN

Mosquito-borne alphaviruses are important causes of epidemic encephalomyelitis. Neuronal cell death during fatal alphavirus encephalomyelitis is immune-mediated; however, the types of cells involved and their regulation have not been determined. We show that the virus-induced inflammatory response was accompanied by production of the regulatory cytokine IL-10, and in the absence of IL-10, paralytic disease occurred earlier and mice died faster. To determine the reason for accelerated disease in the absence of IL-10, immune responses in the CNS of IL-10(-/-) and wild-type (WT) mice were compared. There were no differences in the amounts of brain inflammation or peak virus replication; however, IL-10(-/-) animals had accelerated and increased infiltration of CD4(+)IL-17A(+) and CD4(+)IL-17A(+)IFNγ(+) cells compared with WT animals. Th17 cells infiltrating the brain demonstrated a pathogenic phenotype with the expression of the transcription factor, Tbet, and the production of granzyme B, IL-22, and GM-CSF, with greater production of GM-CSF in IL-10(-/-) mice. Therefore, in fatal alphavirus encephalomyelitis, pathogenic Th17 cells enter the CNS at the onset of neurologic disease and, in the absence of IL-10, appear earlier, develop into Th1/Th17 cells more often, and have greater production of GM-CSF. This study demonstrates a role for pathogenic Th17 cells in fatal viral encephalitis.


Asunto(s)
Infecciones por Alphavirus/inmunología , Encefalomielitis/inmunología , Interleucina-10/inmunología , Virus Sindbis/inmunología , Células Th17/inmunología , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/patología , Animales , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Encefalomielitis/genética , Encefalomielitis/patología , Encefalomielitis/virología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Granzimas/genética , Granzimas/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucinas/genética , Interleucinas/inmunología , Ratones , Ratones Noqueados , Células TH1/inmunología , Células TH1/patología , Células Th17/patología , Interleucina-22
13.
J Virol ; 87(15): 8511-23, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23720714

RESUMEN

Alphavirus dogma has long dictated the production of a discrete set of structural proteins during infection of a cell: capsid, pE2, 6K, and E1. However, bioinformatic analyses of alphavirus genomes (A. E. Firth, B. Y. Chung, M. N. Fleeton, and J. F. Atkins, Virol. J. 5:108, 2008) suggested that a ribosomal frameshifting event occurs during translation of the alphavirus structural polyprotein. Specifically, a frameshift event is suggested to occur during translation of the 6K gene, yielding production of a novel protein, termed transframe (TF), comprised of a C-terminal extension of the 6K protein in the -1 open reading frame (ORF). Here, we validate the findings of Firth and colleagues with respect to the production of the TF protein and begin to characterize the function of TF. Using a mass spectrometry-based approach, we identified TF in purified preparations of both Sindbis and Chikungunya virus particles. We next constructed a panel of Sindbis virus mutants with mutations which alter the production, size, or sequence of TF. We demonstrate that TF is not absolutely required in culture, although disrupting TF production leads to a decrease in virus particle release in both mammalian and insect cells. In a mouse neuropathogenesis model, mortality was <15% in animals infected with the TF mutants, whereas mortality was 95% in animals infected with the wild-type virus. Using a variety of additional assays, we demonstrate that TF retains ion-channel activity analogous to that of 6K and that lack of production of TF does not affect genome replication, particle infectivity, or envelope protein transit to the cell surface. The TF protein therefore represents a previously uncharacterized factor important for alphavirus assembly.


Asunto(s)
Virus Chikungunya/fisiología , Regulación Viral de la Expresión Génica , Virus Sindbis/fisiología , Proteínas Virales/biosíntesis , Ensamble de Virus , Infecciones por Alphavirus/mortalidad , Infecciones por Alphavirus/patología , Infecciones por Alphavirus/virología , Animales , Línea Celular , Virus Chikungunya/genética , Virus Chikungunya/patogenicidad , Modelos Animales de Enfermedad , Insectos , Ratones , Virus Sindbis/genética , Análisis de Supervivencia , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...