Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nature ; 619(7969): 338-347, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380775

RESUMEN

Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.


Asunto(s)
Aves , Interacciones Microbiota-Huesped , Virus de la Influenza A , Gripe Aviar , Gripe Humana , Zoonosis Virales , Animales , Humanos , Aves/virología , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/transmisión , Gripe Aviar/virología , Gripe Humana/prevención & control , Gripe Humana/transmisión , Gripe Humana/virología , Primates , Sistema Respiratorio/metabolismo , Sistema Respiratorio/virología , Medición de Riesgo , Zoonosis Virales/prevención & control , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Replicación Viral
2.
mBio ; 14(3): e0010123, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37097030

RESUMEN

Infected hosts possess two alternative strategies to protect themselves against the negative impact of virus infections: resistance, used to abrogate virus replication, and disease tolerance, used to avoid tissue damage without controlling viral burden. The principles governing pathogen resistance are well understood, while less is known about those involved in disease tolerance. Here, we studied bluetongue virus (BTV), the cause of bluetongue disease of ruminants, as a model system to investigate the mechanisms of virus-host interactions correlating with disease tolerance. BTV induces clinical disease mainly in sheep, while cattle are considered reservoirs of infection, rarely exhibiting clinical symptoms despite sustained viremia. Using primary cells from multiple donors, we show that BTV consistently reaches higher titers in ovine cells than cells from cattle. The variable replication kinetics of BTV in sheep and cow cells were mostly abolished by abrogating the cell type I interferon (IFN) response. We identified restriction factors blocking BTV replication, but both the sheep and cow orthologues of these antiviral genes possess anti-BTV properties. Importantly, we demonstrate that BTV induces a faster host cell protein synthesis shutoff in primary sheep cells than cow cells, which results in an earlier downregulation of antiviral proteins. Moreover, by using RNA sequencing (RNA-seq), we also show a more pronounced expression of interferon-stimulated genes (ISGs) in BTV-infected cow cells than sheep cells. Our data provide a new perspective on how the type I IFN response in reservoir species can have overall positive effects on both virus and host evolution. IMPORTANCE The host immune response usually aims to inhibit virus replication in order to avoid cell damage and disease. In some cases, however, the infected host avoids the deleterious effects of infection despite high levels of viral replication. This strategy is known as disease tolerance, and it is used by animal reservoirs of some zoonotic viruses. Here, using a virus of ruminants (bluetongue virus [BTV]) as an experimental system, we dissected virus-host interactions in cells collected from species that are susceptible (sheep) or tolerant (cow) to disease. We show that (i) virus modulation of the host antiviral type I interferon (IFN) responses, (ii) viral replication kinetics, and (iii) virus-induced cell damage differ in tolerant and susceptible BTV-infected cells. Understanding the complex virus-host interactions in disease tolerance can allow us to disentangle the critical balance between protective and damaging host immune responses.


Asunto(s)
Lengua Azul , Interferón Tipo I , Femenino , Ovinos , Animales , Bovinos , Interferón Tipo I/genética , Lengua Azul/metabolismo , Viremia , Antivirales
3.
Am J Trop Med Hyg ; 106(2): 643-647, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34814103

RESUMEN

Visceral leishmaniasis (VL) is a tropical disease endemic to Brazil. The clinical manifestations of the infection range from asymptomatic to severe. In VL, changes in lipid metabolism, such as hypocholesterolemia and hypertriglyceridemia, occur that are believed to be related to its progression and severity. This study investigated the associations between serum levels of cholesterol, triglycerides, and lipoproteins (high-density lipoprotein, low-density lipoprotein, and very low-density lipoprotein) with clinical and hematological parameters that predict severity in a case series of 83 VL patients. Severely ill patients had higher mean serum triglyceride levels than non-severely ill patients. There was a significant positive correlation between disease severity score and serum triglyceride levels, very low-density lipoprotein, international normalized ratio for prothrombin time test, total bilirubin, and age. An inverse correlation was detected between the disease severity score and mean platelet and neutrophil counts. Hypertriglyceridemia can be a prognostic indicator of severity in patients diagnosed with VL.


Asunto(s)
Hipertrigliceridemia/complicaciones , Leishmaniasis Visceral/sangre , Leishmaniasis Visceral/fisiopatología , Índice de Severidad de la Enfermedad , Adolescente , Adulto , Brasil , Niño , Preescolar , Colesterol/sangre , Estudios Transversales , Femenino , Humanos , Lactante , Recién Nacido , Metabolismo de los Lípidos , Masculino , Triglicéridos/sangre , Adulto Joven
4.
World Neurosurg ; 145: 89-97, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32916360

RESUMEN

Language localization has been an evolving concept over the past 150 years, with the emergence of several important yet conflicting ideologies. The classical theory, starting from the phrenologic work of Gall to the identification of specific regions of language function by Broca, Wernicke, and others, proposed that discrete subcomponents of language were organized into separate anatomic structural regions. The holism theory was postulated in an attempt to disclose that language function was instead attributed to a larger region of the cortex, in which cerebral regions may have the capability of assuming the function of damaged areas. However, this theory was largely abandoned in favor of discrete structural localizationist viewpoints. The subsequent cortical stimulatory work of Penfield led to the development of maps of localization, assigning an eloquent designation to specific regions. The expanding knowledge of cortical and subcortical anatomy allowed for the development of anatomically and functionally integrative language models. In particular, the dual stream model revisited the concept of regional interconnectivity and expanded the concept of eloquence. Advancements in cortical-subcortical stimulation, neurophysiologic monitoring, magnetic resonance diffusion tensor imaging/functional magnetic resonance imaging, awake neurosurgical technique, and knowledge gained by white matter tract anatomy and the Human Connectome Project, shed new light on the dynamic interconnectivity of the cerebrum. New studies are progressively opening doors to this paradigm, showing the dynamic and interdependent nature of language function. In this review, the evolution of language toward the evolving paradigm of dynamic language function and interconnectivity and its impact on shaping the neurosurgical paradigm are outlined.


Asunto(s)
Encéfalo/fisiología , Lenguaje/historia , Encéfalo/anatomía & histología , Mapeo Encefálico , Historia del Siglo XIX , Historia del Siglo XX , Humanos , Imagen por Resonancia Magnética , Neurocirugia/historia , Neurocirugia/tendencias
5.
Cir Cir ; 88(4): 453-460, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32567594

RESUMEN

INTRODUCTION: Olfactory neuroblastoma (ONB) is a malignant neoplasm that arises from the upper nasal vault. OBJECTIVE: We present a retrospective case series and clinical analysis of 12 ONB cases. MATERIALS AND METHODS: Patients with ONB treated at Mexico´s National Cancer Institute between 2011 and 2018. RESULTS: The Kadish proportion of B, C, and D stage was 16%, 58%, or 25%, respectively. Hyams Grade 1, 2, or 3 was 25%, 50%, and 25%, respectively. The most common surgical approach was the craniofacial in 5 cases (42%), followed by the transfacial in 4 cases (33%), and the endonasal endoscopic approach in 3 cases (25%). Gross total resection was achieved in 8 patients (67%). Five patients (42%) underwent a second operation due to recurrent/progressive disease. The surgical complication rate was 8.3%. Progression-free survival was 41 months and the mean overall survival was 63.6 months. CONCLUSIONS: Surgical resection followed by radiotherapy, and chemotherapy for metastatic and recurrent disease provides the best outcome in terms of survival and recurrence. To the best of our knowledge, this is the first series of cases reported in Mexico.


ANTECEDENTES: El neuroblastoma olfatorio es una neoplasia maligna que se origina en la bóveda nasal superior. OBJETIVO: Presentar una serie de casos y un análisis clínico retrospectivo. MÉTODO: Pacientes con neuroblastoma olfatorio tratados en el Instituto Nacional de Cancerología, de México, entre 2011 y 2018. RESULTADOS: La proporción de Kadish en las etapas B, C y D fue del 16, el 58 y el 25%, respectivamente. Los grados 1, 2 y 3 de Hyams fueron el 25, el 50 y el 25%, respectivamente. El abordaje quirúrgico más frecuente fue el craneofacial, en cinco casos (42%), seguido del transfacial en cuatro (33%) y del abordaje endoscópico endonasal en tres (25%). La resección total macroscópica se logró en ocho pacientes (67%). Cinco pacientes (42%) se sometieron a una segunda operación debido a enfermedad recurrente o progresiva. La tasa de complicaciones quirúrgicas fue del 8,3%. La sobrevida libre de progresión fue de 41 meses y la supervivencia media global fue de 63,6 meses. CONCLUSIONES: La resección quirúrgica seguida de radioterapia y quimioterapia para la enfermedad metastásica y recurrente proporciona el mejor resultado en términos de supervivencia y recurrencia. Hasta donde sabemos, esta es la primera serie de casos reportados en México.


Asunto(s)
Estesioneuroblastoma Olfatorio/terapia , Cavidad Nasal , Recurrencia Local de Neoplasia/terapia , Neoplasias Nasales/terapia , Academias e Institutos , Antineoplásicos/uso terapéutico , Quimioterapia Adyuvante , Cisplatino/uso terapéutico , Estesioneuroblastoma Olfatorio/diagnóstico por imagen , Estesioneuroblastoma Olfatorio/mortalidad , Estesioneuroblastoma Olfatorio/patología , Femenino , Humanos , Masculino , México , Persona de Mediana Edad , Cavidad Nasal/patología , Cavidad Nasal/cirugía , Recurrencia Local de Neoplasia/mortalidad , Neoplasias Nasales/mortalidad , Neoplasias Nasales/patología , Supervivencia sin Progresión , Dosificación Radioterapéutica , Radioterapia Adyuvante , Reoperación , Estudios Retrospectivos , Resultado del Tratamiento
6.
J Virol ; 93(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31375575

RESUMEN

Vesicular stomatitis Indiana virus (VSIV), formerly known as vesicular stomatitis virus (VSV) Indiana (VSVIND), is a model virus that is exceptionally sensitive to the inhibitory action of interferons (IFNs). Interferons induce an antiviral state by stimulating the expression of hundreds of interferon-stimulated genes (ISGs). These ISGs can constrain viral replication, limit tissue tropism, reduce pathogenicity, and inhibit viral transmission. Since VSIV is used as a backbone for multiple oncolytic and vaccine strategies, understanding how ISGs restrict VSIV not only helps in understanding VSIV-induced pathogenesis but also helps us evaluate and understand the safety and efficacy of VSIV-based therapies. Thus, there is a need to identify and characterize the ISGs that possess anti-VSIV activity. Using arrayed ISG expression screening, we identified TRIM69 as an ISG that potently inhibits VSIV. This inhibition was highly specific as multiple viruses, including influenza A virus, HIV-1, Rift Valley fever virus, and dengue virus, were unaffected by TRIM69. Indeed, just one amino acid substitution in VSIV can govern sensitivity/resistance to TRIM69. Furthermore, TRIM69 is highly divergent in human populations and exhibits signatures of positive selection that are consistent with this gene playing a key role in antiviral immunity. We propose that TRIM69 is an IFN-induced inhibitor of VSIV and speculate that TRIM69 could be important in limiting VSIV pathogenesis and might influence the specificity and/or efficacy of vesiculovirus-based therapies.IMPORTANCE Vesicular stomatitis Indiana virus (VSIV) is a veterinary pathogen that is also used as a backbone for many oncolytic and vaccine strategies. In natural and therapeutic settings, viral infections like VSIV are sensed by the host, and as a result the host cells make proteins that can protect them from viruses. In the case of VSIV, these antiviral proteins constrain viral replication and protect most healthy tissues from virus infection. In order to understand how VSIV causes disease and how healthy tissues are protected from VSIV-based therapies, it is crucial that we identify the proteins that inhibit VSIV. Here, we show that TRIM69 is an antiviral defense that can potently and specifically block VSIV infection.


Asunto(s)
Interacciones Huésped-Patógeno , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral , Alelos , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Virus del Dengue/fisiología , Resistencia a la Enfermedad , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferones/metabolismo , Interferones/farmacología , Familia de Multigenes , Fosforilación , Transducción de Señal , Proteínas de Motivos Tripartitos/química , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Estomatitis Vesicular/genética , Estomatitis Vesicular/inmunología
7.
PLoS Biol ; 15(12): e2004086, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29253856

RESUMEN

The host innate immune response mediated by type I interferon (IFN) and the resulting up-regulation of hundreds of interferon-stimulated genes (ISGs) provide an immediate barrier to virus infection. Studies of the type I 'interferome' have mainly been carried out at a single species level, often lacking the power necessary to understand key evolutionary features of this pathway. Here, using a single experimental platform, we determined the properties of the interferomes of multiple vertebrate species and developed a webserver to mine the dataset. This approach revealed a conserved 'core' of 62 ISGs, including genes not previously associated with IFN, underscoring the ancestral functions associated with this antiviral host response. We show that gene expansion contributes to the evolution of the IFN system and that interferomes are shaped by lineage-specific pressures. Consequently, each mammal possesses a unique repertoire of ISGs, including genes common to all mammals and others unique to their specific species or phylogenetic lineages. An analysis of genes commonly down-regulated by IFN suggests that epigenetic regulation of transcription is a fundamental aspect of the IFN response. Our study provides a resource for the scientific community highlighting key paradigms of the type I IFN response.


Asunto(s)
Inmunidad Innata , Factores Reguladores del Interferón/fisiología , Interferón Tipo I/fisiología , Mamíferos/inmunología , Animales , Minería de Datos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/metabolismo , Especificidad de la Especie , Virosis/inmunología
8.
Virology ; 509: 121-130, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28628828

RESUMEN

Orthobunyaviruses include several recently emerging viruses of significant medical and veterinary importance. There is currently very limited understanding on what determines the host species range of these pathogens. In this study we discovered that BST-2/tetherin restricts orthobunyavirus replication in a host-specific manner. We show that viruses with human tropism (Oropouche virus and La Crosse virus) are restricted by sheep BST-2 but not by the human orthologue, while viruses with ruminant tropism (Schmallenberg virus and others) are restricted by human BST-2 but not by the sheep orthologue. We also show that BST-2 blocks orthobunyaviruses replication by reducing the amount of envelope glycoprotein into viral particles egressing from infected cells. This is the first study identifying a restriction factor that correlates with species susceptibility to orthobunyavirus infection. This work provides insight to help us dissect the adaptive changes that bunyaviruses require to cross the species barrier and emerge into new species.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Especificidad del Huésped , Interacciones Huésped-Patógeno , Orthobunyavirus/inmunología , Orthobunyavirus/fisiología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Animales , Línea Celular , Proteínas Ligadas a GPI/metabolismo , Humanos , Ovinos , Liberación del Virus
9.
J Virol ; 91(1)2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27795408

RESUMEN

Schmallenberg virus (SBV) was discovered in Germany in late 2011 and then spread rapidly to many European countries. SBV is an orthobunyavirus that causes abortion and congenital abnormalities in ruminants. A virus-encoded nonstructural protein, termed NSs, is a major virulence factor of SBV, and it is known to promote the degradation of Rpb1, a subunit of the RNA polymerase II (Pol II) complex, and therefore hampers global cellular transcription. In this study, we found that NSs is mainly localized in the nucleus of infected cells and specifically appears to target the nucleolus through a nucleolar localization signal (NoLS) localized between residues 33 and 51 of the protein. NSs colocalizes with nucleolar markers such as B23 (nucleophosmin) and fibrillarin. We observed that in SBV-infected cells, B23 undergoes a nucleolus-to-nucleoplasm redistribution, evocative of virus-induced nucleolar disruption. In contrast, the nucleolar pattern of B23 was unchanged upon infection with an SBV recombinant mutant with NSs lacking the NoLS motif (SBVΔNoLS). Interestingly, unlike wild-type SBV, the inhibitory activity of SBVΔNoLS toward RNA Pol II transcription is impaired. Overall, our results suggest that a putative link exists between NSs-induced nucleolar disruption and its inhibitory function on cellular transcription, which consequently precludes the cellular antiviral response and/or induces cell death. IMPORTANCE: Schmallenberg virus (SBV) is an emerging arbovirus of ruminants that spread in Europe between 2011 and 2013. SBV induces fetal abnormalities during gestation, with the central nervous system being one of the most affected organs. The virus-encoded NSs protein acts as a virulence factor by impairing host cell transcription. Here, we show that NSs contains a nucleolar localization signal (NoLS) and induces disorganization of the nucleolus. The NoLS motif in the SBV NSs is absolutely necessary for virus-induced inhibition of cellular transcription. To our knowledge, this is the first report of nucleolar functions for NSs within the Bunyaviridae family.


Asunto(s)
Nucléolo Celular/virología , Células Ependimogliales/virología , Interacciones Huésped-Patógeno , Orthobunyavirus/patogenicidad , ARN Polimerasa II/química , Proteínas no Estructurales Virales/química , Animales , Línea Celular Transformada , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestructura , Plexo Coroideo/citología , Plexo Coroideo/metabolismo , Plexo Coroideo/virología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Células Ependimogliales/metabolismo , Células Ependimogliales/ultraestructura , Regulación de la Expresión Génica , Células HeLa , Humanos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Orthobunyavirus/genética , Orthobunyavirus/metabolismo , Señales de Clasificación de Proteína , Transporte de Proteínas , Proteolisis , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Ovinos , Transducción de Señal , Transcripción Genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
10.
J Virol ; 90(11): 5427-39, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27009961

RESUMEN

UNLABELLED: Bluetongue virus (BTV) is the causative agent of bluetongue, a major infectious disease of ruminants with serious consequences to both animal health and the economy. The clinical outcome of BTV infection is highly variable and dependent on a variety of factors related to both the virus and the host. In this study, we show that the BTV nonstructural protein NS4 favors viral replication in sheep, the animal species most affected by bluetongue. In addition, NS4 confers a replication advantage on the virus in interferon (IFN)-competent primary sheep endothelial cells and immortalized cell lines. We determined that in cells infected with an NS4 deletion mutant (BTV8ΔNS4), there is increased synthesis of type I IFN compared to cells infected with wild-type BTV-8. In addition, using RNA sequencing (RNA-seq), we show that NS4 modulates the host IFN response and downregulates mRNA levels of type I IFN and interferon-stimulated genes. Moreover, using reporter assays and protein synthesis assays, we show that NS4 downregulates the activities of a variety of promoters, such as the cytomegalovirus immediate-early promoter, the IFN-ß promoter, and a promoter containing interferon-stimulated response elements (ISRE). We also show that the NS4 inhibitory activity on gene expression is related to its nucleolar localization. Furthermore, NS4 does not affect mRNA splicing or cellular translation. The data obtained in this study strongly suggest that BTV NS4 is an IFN antagonist and a key determinant of viral virulence. IMPORTANCE: Bluetongue is one of the main infectious diseases of ruminants and is caused by bluetongue virus (BTV), an arthropod-borne virus transmitted from infected to susceptible animals by Culicoides biting midges. Bluetongue has a variable clinical outcome that can be related to both virus and host factors. It is therefore critical to understand the interplay between BTV and the host immune responses. In this study, we show that a nonstructural protein of BTV (NS4) is critical to counteract the innate immune response of the host. Infection of cells with a BTV mutant lacking NS4 results in increased synthesis of IFN-ß and upregulation of interferon-stimulated genes. In addition, we show that NS4 is a virulence factor for BTV by favoring viral replication in sheep, the animal species most susceptible to bluetongue.


Asunto(s)
Virus de la Lengua Azul/química , Virus de la Lengua Azul/patogenicidad , Lengua Azul/virología , Interferón Tipo I/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/inmunología , Línea Celular , Células Endoteliales/virología , Inmunidad Innata , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Interferón beta/genética , Regiones Promotoras Genéticas , Eliminación de Secuencia , Ovinos , Virulencia , Factores de Virulencia/química , Factores de Virulencia/aislamiento & purificación , Replicación Viral
11.
J Virol ; 90(11): 5440-5450, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26984728

RESUMEN

UNLABELLED: Serial passage of viruses in cell culture has been traditionally used to attenuate virulence and identify determinants of viral pathogenesis. In a previous study, we found that a strain of Schmallenberg virus (SBV) serially passaged in tissue culture (termed SBVp32) unexpectedly displayed increased pathogenicity in suckling mice compared to wild-type SBV. In this study, we mapped the determinants of SBVp32 virulence to the viral genome M segment. SBVp32 virulence is associated with the capacity of this virus to reach high titers in the brains of experimentally infected suckling mice. We also found that the Gc glycoprotein, encoded by the M segment of SBVp32, facilitates host cell protein shutoff in vitro Interestingly, while the M segment of SBVp32 is a virulence factor, we found that the S segment of the same virus confers by itself an attenuated phenotype to wild-type SBV, as it has lost the ability to block the innate immune system of the host. Single mutations present in the Gc glycoprotein of SBVp32 are sufficient to compensate for both the attenuated phenotype of the SBVp32 S segment and the attenuated phenotype of NSs deletion mutants. Our data also indicate that the SBVp32 M segment does not act as an interferon (IFN) antagonist. Therefore, SBV mutants can retain pathogenicity even when they are unable to fully control the production of IFN by infected cells. Overall, this study suggests that the viral glycoprotein of orthobunyaviruses can compensate, at least in part, for the function of NSs. In addition, we also provide evidence that the induction of total cellular protein shutoff by SBV is determined by multiple viral proteins, while the ability to control the production of IFN maps to the NSs protein. IMPORTANCE: The identification of viral determinants of pathogenesis is key to the development of prophylactic and intervention measures. In this study, we found that the bunyavirus Gc glycoprotein is a virulence factor. Importantly, we show that mutations in the Gc glycoprotein can restore the pathogenicity of attenuated mutants resulting from deletions or mutations in the nonstructural protein NSs. Our findings highlight the fact that careful consideration should be taken when designing live attenuated vaccines based on deletions of nonstructural proteins since single mutations in the viral glycoproteins appear to revert attenuated mutants to virulent phenotypes.


Asunto(s)
Infecciones por Bunyaviridae/virología , Glicoproteínas/genética , Mutación , Orthobunyavirus/patogenicidad , Biosíntesis de Proteínas , Proteínas no Estructurales Virales/genética , Proteínas Virales/metabolismo , Animales , Encéfalo/virología , Línea Celular , Genoma Viral , Glicoproteínas/química , Glicoproteínas/metabolismo , Interacciones Huésped-Patógeno , Interferones/antagonistas & inhibidores , Interferones/genética , Ratones , Orthobunyavirus/química , Orthobunyavirus/genética , Orthobunyavirus/metabolismo , Eliminación de Secuencia , Carga Viral , Proteínas Virales/genética , Virión , Factores de Virulencia
12.
Infect Genet Evol ; 40: 47-53, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26921797

RESUMEN

Capim and Enseada viruses are members of the genus Orthobunyavirus isolated from mosquitoes and mammals in Brazil. Despite seroprevalence studies indicating human infections in Latin America, these viruses remain relatively unknown and unstudied. In order to better understand the genetic and evolutionary relationships among orthobunyaviruses, we sequenced the three genomic segments of Capim and Enseada orthobunyaviruses. Based on phylogenetic analysis, we demonstrated that these viruses depicted two new distinct clades, one represented by Enseada and another composed of Capim virus. In general, the genome organization and genetic traits of these viruses are similar to other orthobunyaviruses however, the open reading frame (ORF) of the putative nonstructural NSs protein of Enseada orthobunyavirus precedes the nucleocapsid ORF. Overall, our study provides details on the molecular characteristics of the prototype species of two groups within the Orthobunyavirus genus, revealing novel features into the genetic diversity and evolution of this genus.


Asunto(s)
Culicidae/virología , Mamíferos/virología , Orthobunyavirus/clasificación , Análisis de Secuencia de ARN/métodos , Animales , Brasil , Genoma Viral , Humanos , Proteínas de la Nucleocápside/genética , Sistemas de Lectura Abierta , Orthobunyavirus/genética , Filogenia , Proteínas no Estructurales Virales/genética
13.
J Gen Virol ; 96(11): 3280-3293, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26290332

RESUMEN

Viruses have often evolved overlapping reading frames in order to maximize their coding capacity. Until recently, the segmented dsRNA genome of viruses of the Orbivirus genus was thought to be monocistronic, but the identification of the bluetongue virus (BTV) NS4 protein changed this assumption. A small ORF in segment 10, overlapping the NS3 ORF in the +1 position, is maintained in more than 300 strains of the 27 different BTV serotypes and in more than 200 strains of the phylogenetically related African horse sickness virus (AHSV). In BTV, this ORF (named S10-ORF2 in this study) encodes a putative protein 50-59 residues in length and appears to be under strong positive selection. HA- or GFP-tagged versions of S10-ORF2 expressed from transfected plasmids localized within the nucleoli of transfected cells, unless a putative nucleolar localization signal was mutated. S10-ORF2 inhibited gene expression, but not RNA translation, in transient transfection reporter assays. In both mammalian and insect cells, BTV S10-ORF2 deletion mutants (BTV8ΔS10-ORF2) displayed similar replication kinetics to wt virus. In vivo, S10-ORF2 deletion mutants were pathogenic in mouse models of disease. Although further evidence is required for S10-ORF2 expression during infection, the data presented provide an initial characterization of this ORF.


Asunto(s)
Virus de la Lengua Azul/genética , Lengua Azul/virología , Genoma Viral , Sistemas de Lectura Abierta , Proteínas Virales/genética , Animales , Virus de la Lengua Azul/clasificación , Virus de la Lengua Azul/metabolismo , Línea Celular , Ratones , Filogenia , Proteínas Virales/metabolismo
14.
J Virol ; 89(1): 535-44, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339764

RESUMEN

UNLABELLED: Bone marrow stromal cell antigen 2 (BST2) is a cellular restriction factor with a broad antiviral activity. In sheep, the BST2 gene is duplicated into two paralogs termed oBST2A and oBST2B. oBST2A impedes viral exit of the Jaagsiekte sheep retroviruses (JSRV), most probably by retaining virions at the cell membrane, similar to the "tethering" mechanism exerted by human BST2. In this study, we provide evidence that unlike oBST2A, oBST2B is limited to the Golgi apparatus and disrupts JSRV envelope (Env) trafficking by sequestering it. In turn, oBST2B leads to a reduction in Env incorporation into viral particles, which ultimately results in the release of virions that are less infectious. Furthermore, the activity of oBST2B does not seem to be restricted to retroviruses, as it also acts on vesicular stomatitis virus glycoproteins. Therefore, we suggest that oBST2B exerts antiviral activity using a mechanism distinct from the classical tethering restriction observed for oBST2A. IMPORTANCE: BST2 is a powerful cellular restriction factor against a wide range of enveloped viruses. Sheep possess two paralogs of the BST2 gene called oBST2A and oBST2B. JSRV, the causative agent of a transmissible lung cancer of sheep, is known to be restricted by oBST2A. In this study, we show that unlike oBST2A, oBST2B impairs the normal cellular trafficking of JSRV envelope glycoproteins by sequestering them within the Golgi apparatus. We also show that oBST2B decreases the incorporation of envelope glycoprotein into JSRV viral particles, which in turn reduces virion infectivity. In conclusion, oBST2B exerts a novel antiviral activity that is distinct from those of BST2 proteins of other species.


Asunto(s)
Retrovirus Ovino Jaagsiekte/inmunología , Retrovirus Ovino Jaagsiekte/fisiología , Glicoproteínas de Membrana/inmunología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Virión/metabolismo , Ensamble de Virus , Animales , Aparato de Golgi/metabolismo , Transporte de Proteínas , Ovinos
15.
Viruses ; 6(12): 4926-45, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25502326

RESUMEN

Sheep betaretroviruses represent a fascinating model to study the complex evolutionary interplay between host and pathogen in natural settings. In infected sheep, the exogenous and pathogenic Jaagsiekte sheep retrovirus (JSRV) coexists with a variety of highly related endogenous JSRVs, referred to as enJSRVs. During evolution, some of them were co-opted by the host as they fulfilled important biological functions, including placental development and protection against related exogenous retroviruses. In particular, two enJSRV loci, enJS56A1 and enJSRV-20, were positively selected during sheep domestication due to their ability to interfere with the replication of related competent retroviruses. Interestingly, viruses escaping these transdominant enJSRVs have recently emerged, probably less than 200 years ago. Overall, these findings suggest that in sheep the process of endogenization is still ongoing and, therefore, the evolutionary interplay between endogenous and exogenous sheep betaretroviruses and their host has not yet reached an equilibrium.


Asunto(s)
Evolución Biológica , Retrovirus Endógenos/genética , Retrovirus Ovino Jaagsiekte/genética , Enfermedades de las Ovejas/virología , Animales , Retrovirus Endógenos/fisiología , Retrovirus Ovino Jaagsiekte/fisiología , Ovinos , Enfermedades de las Ovejas/genética , Oveja Doméstica/genética , Oveja Doméstica/virología
16.
J Gen Virol ; 95(Pt 8): 1640-1646, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24828331

RESUMEN

Bunyaviruses have evolved a variety of strategies to counteract the antiviral defence systems of mammalian cells. Here we show that the NSs protein of Schmallenberg virus (SBV) induces the degradation of the RPB1 subunit of RNA polymerase II and consequently inhibits global cellular protein synthesis and the antiviral response. In addition, we show that the SBV NSs protein enhances apoptosis in vitro and possibly in vivo, suggesting that this protein could be involved in SBV pathogenesis in different ways.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Orthobunyavirus/fisiología , ARN Polimerasa II/metabolismo , Proteínas no Estructurales Virales/metabolismo , Humanos , Orthobunyavirus/inmunología , Proteolisis
17.
PLoS One ; 8(8): e70814, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967111

RESUMEN

Understanding the genetic, antigenic and structural changes that occur during HIV-1 infection in response to pre-existing immunity will facilitate current efforts to develop an HIV-1 vaccine. Much is known about HIV-1 variation at the population level but little with regard to specific changes occurring in the envelope glycoprotein within a host in response to immune pressure elicited by antibodies. The aim of this study was to track and map specific early genetic changes occurring in the viral envelope gene following vaccination using a highly controlled viral challenge setting in the SHIV macaque model. We generated 449 full-length env sequences from vaccinees, and 63 from the virus inoculum. Analysis revealed a different pattern in the distribution and frequency of mutations in the regions of the envelope gene targeted by the vaccine as well as different patterns of diversification between animals in the naïve control group and vaccinees. Given the high stringency of the model it is remarkable that we were able to identify genetic changes associated with the vaccination. This work provides insight into the characterization of breakthrough viral populations in less than fully efficacious vaccines and illustrates the value of HIV-1 Env SHIV challenge model in macaques to unravel the mechanisms driving HIV-1 envelope genetic diversity in the presence of vaccine induced-responses.


Asunto(s)
Genoma Viral/genética , Impresión Genómica/inmunología , VIH-1/genética , VIH-1/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunación , Análisis de Varianza , Animales , Evolución Molecular , Macaca mulatta , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética
18.
PLoS One ; 8(5): e62939, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23667545

RESUMEN

Schmallenberg virus (SBV) is a recently discovered Bunyavirus associated mainly with abortions, stillbirths and malformations of the skeletal and central nervous system (CNS) in newborn ruminants. In this study, a detailed immunophenotyping of the inflammatory cells of the CNS of affected animals was carried out in order to increase our understanding of SBV pathogenesis. A total of 82 SBV-polymerase chain reaction (PCR) positive neonatal ruminants (46 sheep lambs, 34 calves and 2 goat kids) were investigated for the presence of inflammation in the brain and spinal cord. The study focused on 15 out of 82 animals (18.3%) showing inflammation in the CNS. All 15 neonates displayed lymphohistiocytic meningoencephalomyelitis affecting most frequently the mesencephalon and the parietal and temporal lobes. The majority of infiltrating cells were CD3-positive T cells, followed by CD79α-positive B cells and CD68-positive microglia/macrophages. Malformations like por- and hydranencephaly, frequently found in the temporal lobe, showed associated demyelination and axonal loss. SBV antigen was detected in 37 out of 82 (45.1%) neonatal brains by immunohistochemistry. In particular, SBV antigen was found in 93.3% (14 out of 15 ruminants) and 32.8% (22 out of 67 ruminants) of animals with and without encephalitis, respectively. Highest amounts of virus-protein expression levels were found in the temporal lobe. Our findings suggest that: (i) different brain regions display differential susceptibility to SBV infection; (ii) inflammatory cells in the CNS are found only in a minority of virus infected animals; (iii) malformations occur in association with and without inflammation in the CNS; and (iv) viral antigen is strongly associated with the presence of inflammation in naturally infected animals. Further studies are required to explore the cell tropism and pathogenesis of SBV infection in ruminants.


Asunto(s)
Encéfalo/virología , Infecciones por Bunyaviridae/inmunología , Inmunofenotipificación , Orthobunyavirus/fisiología , Rumiantes/virología , Médula Espinal/virología , Animales , Astrocitos/inmunología , Astrocitos/patología , Axones/inmunología , Axones/patología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Infecciones por Bunyaviridae/patología , Inflamación/inmunología , Inflamación/patología , Vaina de Mielina/metabolismo , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología
19.
PLoS One ; 8(1): e54871, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23372784

RESUMEN

Following long-term infection with virus derived from the pathogenic GL8 molecular clone of feline immunodeficiency virus (FIV), a range of viral variants emerged with distinct modes of interaction with the viral receptors CD134 and CXCR4, and sensitivities to neutralizing antibodies. In order to assess whether this viral diversity would be maintained following subsequent transmission, a synthetic quasispecies was reconstituted comprising molecular clones bearing envs from six viral variants and its replicative capacity compared in vivo with a clonal preparation of the parent virus. Infection with either clonal (Group 1) or diverse (Group 2) challenge viruses, resulted in a reduction in CD4+ lymphocytes and an increase in CD8+ lymphocytes. Proviral loads were similar in both study groups, peaking by 10 weeks post-infection, a higher plateau (set-point) being achieved and maintained in study Group 1. Marked differences in the ability of individual viral variants to replicate were noted in Group 2; those most similar to GL8 achieved higher viral loads while variants such as the chimaeras bearing the B14 and B28 Envs grew less well. The defective replication of these variants was not due to suppression by the humoral immune response as virus neutralising antibodies were not elicited within the study period. Similarly, although potent cellular immune responses were detected against determinants in Env, no qualitative differences were revealed between animals infected with either the clonal or the diverse inocula. However, in vitro studies indicated that the reduced replicative capacity of variants B14 and B28 in vivo was associated with altered interactions between the viruses and the viral receptor and co-receptor. The data suggest that viral variants with GL8-like characteristics have an early, replicative advantage and should provide the focus for future vaccine development.


Asunto(s)
Virus de la Inmunodeficiencia Felina/fisiología , Infecciones por Lentivirus/virología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Gatos , Línea Celular , Mapeo Epitopo , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Productos del Gen env/química , Productos del Gen env/inmunología , Humanos , Inmunidad Celular/inmunología , Virus de la Inmunodeficiencia Felina/patogenicidad , Infecciones por Lentivirus/inmunología , Provirus , Receptores CXCR4/antagonistas & inhibidores , Carga Viral , Replicación Viral
20.
PLoS Pathog ; 9(1): e1003133, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23326235

RESUMEN

Schmallenberg virus (SBV) is an emerging orthobunyavirus of ruminants associated with outbreaks of congenital malformations in aborted and stillborn animals. Since its discovery in November 2011, SBV has spread very rapidly to many European countries. Here, we developed molecular and serological tools, and an experimental in vivo model as a platform to study SBV pathogenesis, tropism and virus-host cell interactions. Using a synthetic biology approach, we developed a reverse genetics system for the rapid rescue and genetic manipulation of SBV. We showed that SBV has a wide tropism in cell culture and "synthetic" SBV replicates in vitro as efficiently as wild type virus. We developed an experimental mouse model to study SBV infection and showed that this virus replicates abundantly in neurons where it causes cerebral malacia and vacuolation of the cerebral cortex. These virus-induced acute lesions are useful in understanding the progression from vacuolation to porencephaly and extensive tissue destruction, often observed in aborted lambs and calves in naturally occurring Schmallenberg cases. Indeed, we detected high levels of SBV antigens in the neurons of the gray matter of brain and spinal cord of naturally affected lambs and calves, suggesting that muscular hypoplasia observed in SBV-infected lambs is mostly secondary to central nervous system damage. Finally, we investigated the molecular determinants of SBV virulence. Interestingly, we found a biological SBV clone that after passage in cell culture displays increased virulence in mice. We also found that a SBV deletion mutant of the non-structural NSs protein (SBVΔNSs) is less virulent in mice than wild type SBV. Attenuation of SBV virulence depends on the inability of SBVΔNSs to block IFN synthesis in virus infected cells. In conclusion, this work provides a useful experimental framework to study the biology and pathogenesis of SBV.


Asunto(s)
Infecciones por Bunyaviridae/virología , Corteza Cerebral/virología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Orthobunyavirus/patogenicidad , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Infecciones por Bunyaviridae/inmunología , Infecciones por Bunyaviridae/mortalidad , Infecciones por Bunyaviridae/patología , Bovinos , Línea Celular , Enfermedades Cerebelosas/inmunología , Enfermedades Cerebelosas/patología , Enfermedades Cerebelosas/virología , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Endotelio Vascular/virología , Ratones , Datos de Secuencia Molecular , Neuronas/inmunología , Neuronas/patología , Neuronas/virología , Orthobunyavirus/genética , Orthobunyavirus/aislamiento & purificación , Eliminación de Secuencia , Ovinos , Médula Espinal/inmunología , Médula Espinal/patología , Médula Espinal/virología , Tasa de Supervivencia , Vacuolas , Tropismo Viral , Virulencia , Cultivo de Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...