Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 18(9): e1010316, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36103568

RESUMEN

The evolutionarily successful poxviruses possess effective and diverse strategies to circumvent or overcome host defense mechanisms. Poxviruses encode many immunoregulatory proteins to evade host immunity to establish a productive infection and have unique means of inhibiting DNA sensing-dependent type 1 interferon (IFN-I) responses, a necessity given their dsDNA genome and exclusively cytoplasmic life cycle. We found that the key DNA sensing inhibition by poxvirus infection was dominant during the early stage of poxvirus infection before DNA replication. In an effort to identify the poxvirus gene products which subdue the antiviral proinflammatory responses (e.g., IFN-I response), we investigated the function of one early gene that is the known host range determinant from the highly conserved poxvirus host range C7L superfamily, myxoma virus (MYXV) M062. Host range factors are unique features of poxviruses that determine the species and cell type tropism. Almost all sequenced mammalian poxviruses retain at least one homologue of the poxvirus host range C7L superfamily. In MYXV, a rabbit-specific poxvirus, the dominant and broad-spectrum host range determinant of the C7L superfamily is the M062R gene. The M062R gene product is essential for MYXV infection in almost all cells tested from different mammalian species and specifically inhibits the function of host Sterile α Motif Domain-containing 9 (SAMD9), as M062R-null (ΔM062R) MYXV causes abortive infection in a SAMD9-dependent manner. In this study we investigated the immunostimulatory property of the ΔM062R. We found that the replication-defective ΔM062R activated host DNA sensing pathway during infection in a cGAS-dependent fashion and that knocking down SAMD9 expression attenuated proinflammatory responses. Moreover, transcriptomic analyses showed a unique feature of the host gene expression landscape that is different from the dsDNA alone-stimulated inflammatory state. This study establishes a link between the anti-neoplastic function of SAMD9 and the regulation of innate immune responses.


Asunto(s)
Interferón Tipo I , Myxoma virus , Infecciones por Poxviridae , Poxviridae , Animales , Especificidad del Huésped/genética , Humanos , Interferón Tipo I/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/metabolismo , Mamíferos , Monocitos/metabolismo , Myxoma virus/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Poxviridae/genética , Poxviridae/metabolismo , Infecciones por Poxviridae/genética , Conejos , Transcriptoma , Virus Vaccinia/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
Gene ; 801: 145850, 2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34274484

RESUMEN

This study explored the transcriptome of lamb testis cells infected with sheeppox virus (SPPV) wild strain (WS) and vaccine strain (VS) at an immediate-early time. Most of the differentially expressed genes (DEGs) and differentially expressed highly connected (DEHC) gene network were found to be involved in SPPV-VS infection compared to SPPV-WS. Further, the signaling pathways were mostly involved in SPPV-VS infection than SPPV-WS. SPPV modulates the expression of several important host proteins such as CD40, FAS, ITGß1, ITGα1, Pak1, Pak2, CD14, ILK leading to viral attachment and entry; immune-related DEGs such as MAPK, JNK, ERK, NFKB, IKB, PI3K, STAT which provide optimal cellular condition for early viral protein expression; and FOXO3, ATF, CDKNA1, TCF, SRF, BDNF which help in inducing apoptosis and MPTP, BAD and Tp53 inhibits apoptosis or cell death at the immediate-early time. The results captured the specific genes and enabled to understand distinct pathogenic mechanisms employed by VS and WS of SPPV.


Asunto(s)
Capripoxvirus , Genes Inmediatos-Precoces , Interacciones Huésped-Patógeno/genética , Infecciones por Poxviridae/genética , Enfermedades de las Ovejas/genética , Animales , Capripoxvirus/patogenicidad , Células Cultivadas , Expresión Génica , Masculino , Infecciones por Poxviridae/veterinaria , Mapas de Interacción de Proteínas/genética , Ovinos , Enfermedades de las Ovejas/virología
4.
PLoS Pathog ; 17(1): e1009183, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33444388

RESUMEN

The antiviral protein kinase R (PKR) is an important host restriction factor, which poxviruses must overcome to productively infect host cells. To inhibit PKR, many poxviruses encode a pseudosubstrate mimic of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2), designated K3 in vaccinia virus. Although the interaction between PKR and eIF2α is highly conserved, some K3 orthologs from host-restricted poxviruses were previously shown to inhibit PKR in a species-specific manner. To better define this host range function, we compared the sensitivity of PKR from 17 mammals to inhibition by K3 orthologs from closely related orthopoxviruses, a genus with a generally broader host range. The K3 orthologs showed species-specific inhibition of PKR and exhibited three distinct inhibition profiles. In some cases, PKR from closely related species showed dramatic differences in their sensitivity to K3 orthologs. Vaccinia virus expressing the camelpox virus K3 ortholog replicated more than three orders of magnitude better in human and sheep cells than a virus expressing vaccinia virus K3, but both viruses replicated comparably well in cow cells. Strikingly, in site-directed mutagenesis experiments between the variola virus and camelpox virus K3 orthologs, we found that different amino acid combinations were necessary to mediate improved or diminished inhibition of PKR derived from different host species. Because there is likely a limited number of possible variations in PKR that affect K3-interactions but still maintain PKR/eIF2α interactions, it is possible that by chance PKR from some potential new hosts may be susceptible to K3-mediated inhibition from a virus it has never previously encountered. We conclude that neither the sensitivity of host proteins to virus inhibition nor the effectiveness of viral immune antagonists can be inferred from their phylogenetic relatedness but must be experimentally determined.


Asunto(s)
Antivirales/antagonistas & inhibidores , Especificidad del Huésped , Orthopoxvirus/clasificación , Orthopoxvirus/fisiología , Infecciones por Poxviridae/virología , Replicación Viral , eIF-2 Quinasa/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Antivirales/metabolismo , Células HeLa , Humanos , Fosforilación , Filogenia , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/metabolismo , Homología de Secuencia , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
5.
Front Immunol ; 11: 567348, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33154747

RESUMEN

We conducted a large genome-wide association study (GWAS) of the immune responses to primary smallpox vaccination in a combined cohort of 1,653 subjects. We did not observe any polymorphisms associated with standard vaccine response outcomes (e.g., neutralizing antibody, T cell ELISPOT response, or T cell cytokine production); however, we did identify a cluster of SNPs on chromosome 5 (5q31.2) that were significantly associated (p-value: 1.3 x 10-12 - 1.5x10-36) with IFNα response to in vitro poxvirus stimulation. Examination of these SNPs led to the functional testing of rs1131769, a non-synonymous SNP in TMEM173 causing an Arg-to-His change at position 232 in the STING protein-a major regulator of innate immune responses to viral infections. Our findings demonstrate differences in the ability of the two STING variants to phosphorylate the downstream intermediates TBK1 and IRF3 in response to multiple STING ligands. Further downstream in the STING pathway, we observed significantly reduced expression of type I IFNs (including IFNα) and IFN-response genes in cells carrying the H232 variant. Subsequent molecular modeling of both alleles predicted altered ligand binding characteristics between the two variants, providing a potential mechanism underlying differences in inter-individual responses to poxvirus infection. Our data indicate that possession of the H232 variant may impair STING-mediated innate immunity to poxviruses. These results clarify prior studies evaluating functional effects of genetic variants in TMEM173 and provide novel data regarding genetic control of poxvirus immunity.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/inmunología , Poxviridae/inmunología , Alelos , Susceptibilidad a Enfermedades , Efecto Fundador , Expresión Génica , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Inmunidad Innata/genética , Fenómenos Inmunogenéticos , Ligandos , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Fosforilación , Infecciones por Poxviridae/virología , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
6.
Front Immunol ; 11: 2154, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013908

RESUMEN

The salmon gill poxvirus (SGPV) is a large DNA virus that infects gill epithelial cells in Atlantic salmon and is associated with acute high mortality disease outbreaks in aquaculture. The pathological effects of SGPV infection include gill epithelial apoptosis in the acute phase of the disease and hyperplasia of gill epithelial cells in surviving fish, causing damage to the gill respiratory surface. In this study, we sampled gills from Atlantic salmon presmolts during a natural outbreak of SGPV disease (SGPVD). Samples covered the early phase of infection, the acute mortality phase, the resolving phase of the disease and control fish from the same group and facility. Mortality, the presence and level of SGPV and gill epithelial apoptosis were clearly associated. The gene expression pattern in the acute phase of SGPVD was in tune with the pathological findings and revealed novel transcript-based disease biomarkers, including pro-apoptotic and proliferative genes, along with changes in expression of ion channels and mucins. The innate antiviral response was strongly upregulated in infected gills and chemokine expression was altered. The regenerating phase did not reveal adaptive immune activity within the study period, but several immune effector genes involved in mucosal protection were downregulated into the late phase, indicating that SGPV infection could compromise mucosal defense. These data provide novel insight into the infection mechanisms and host interaction of SGPV.


Asunto(s)
Enfermedades de los Peces/inmunología , Branquias/metabolismo , Infecciones por Poxviridae/inmunología , Poxviridae/fisiología , Salmo salar , Animales , Apoptosis/genética , Biomarcadores/metabolismo , Proliferación Celular/genética , Brotes de Enfermedades , Enfermedades de los Peces/epidemiología , Enfermedades de los Peces/genética , Proteínas de Peces/genética , Branquias/patología , Branquias/virología , Inmunidad Mucosa , Terapia de Inmunosupresión , Canales Iónicos/genética , Mucinas/genética , Noruega/epidemiología , Infecciones por Poxviridae/epidemiología , Infecciones por Poxviridae/genética , Transcriptoma
7.
Infect Genet Evol ; 85: 104472, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32711078

RESUMEN

Sheeppox and goatpox are highly contagious viral diseases of small ruminants causing severe economic losses to the livestock farmers. The disease is enzootic in Asia including India, Middle East and African countries. In the present study, a total of 28 isolates from twenty five sheeppox and goatpox disease outbreaks were phylogenetically analyzed based on P32 gene/protein along with homology modeling and docking using heparan sulfate and UDP-glucose. Three distinct lineage-specific clusters as per their host origin were recorded. Multiple sequence analysis of P32 gene revealed that genetically similar sheeppox virus (SPPV) and goatpox virus (GTPV) strains are circulating in India. Phylogenetically, Lumpy skin disease (LSDV) and SPPV had a closer genetic relationship than GTPV. Comparative sequence alignment indicated conservation of various motifs such as glycosaminoglycan (GAG), chemokine like motif (CX3C) and Asp-Glu-any other residue-Asp (D/ExD), as well as viral specific signature residues in SPPV and GTPV isolates. Structurally, P32 protein of SPPV and GTPV with mixed α helices and ß sheets resembled with crystal structure of homologue vaccinia virus H3L protein. Docking studies in P32 protein of SPPV and GTPV revealed conserved binding pattern with heparan sulfate which is involved in the virus attachment and varied glycosyltransferase fold with UDP-glucose. These findings may help in development of suitable vaccines/diagnostics and therapeutics against capripoxviruses.


Asunto(s)
Capripoxvirus/clasificación , Capripoxvirus/genética , Enfermedades de las Cabras/virología , Infecciones por Poxviridae/genética , Enfermedades de las Ovejas/virología , Proteínas del Envoltorio Viral/genética , Animales , Cabras/virología , India , Filogenia , Mapeo de Interacción de Proteínas , Análisis de Secuencia de ADN , Ovinos/virología
8.
Viruses ; 12(7)2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629851

RESUMEN

Although orthopoxviruses (OPXV) are known to encode a majority of the genes required for replication in host cells, genome-wide genetic screens have revealed that several host pathways are indispensable for OPXV infection. Through a haploid genetic screen, we previously identified several host genes required for monkeypox virus (MPXV) infection, including the individual genes that form the conserved oligomeric Golgi (COG) complex. The COG complex is an eight-protein (COG1-COG8) vesicle tethering complex important for regulating membrane trafficking, glycosylation enzymes, and maintaining Golgi structure. In this study, we investigated the role of the COG complex in OPXV infection using cell lines with individual COG gene knockout (KO) mutations. COG KO cells infected with MPXV and vaccinia virus (VACV) produced small plaques and a lower virus yield compared to wild type (WT) cells. In cells where the KO phenotype was reversed using a rescue plasmid, the size of virus plaques increased demonstrating a direct link between the decrease in viral spread and the KO of COG genes. KO cells infected with VACV displayed lower levels of viral fusion and entry compared to WT suggesting that the COG complex is important for early events in OPXV infection. Additionally, fewer actin tails were observed in VACV-infected KO cells compared to WT. Since COG complex proteins are required for cellular trafficking of glycosylated membrane proteins, the disruption of this process due to lack of individual COG complex proteins may potentially impair the virus-cell interactions required for viral entry and egress. These data validate that the COG complex previously identified in our genetic screens plays a role in OPXV infection.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Orthopoxvirus/fisiología , Infecciones por Poxviridae/metabolismo , Infecciones por Poxviridae/virología , Internalización del Virus , Proteínas Adaptadoras del Transporte Vesicular/genética , Glicosilación , Aparato de Golgi , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Mutación , Orthopoxvirus/genética , Infecciones por Poxviridae/genética
9.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30728252

RESUMEN

Myxoma virus (MYXV) has been evolving in a novel host species-European rabbits-in Australia since 1950. Previous studies of viruses sampled from 1950 to 1999 revealed a remarkably clock-like evolutionary process across all Australian lineages of MYXV. Through an analysis of 49 newly generated MYXV genome sequences isolated in Australia between 2008 and 2017, we show that MYXV evolution in Australia can be characterized by three lineages, one of which exhibited a greatly elevated rate of evolutionary change and a dramatic breakdown of temporal structure. Phylogenetic analysis revealed that this apparently punctuated evolutionary event occurred between 1996 and 2012. The branch leading to the rapidly evolving lineage contained a relatively high number of nonsynonymous substitutions, and viruses in this lineage reversed a mutation found in the progenitor standard laboratory strain (SLS) and all previous sequences that disrupts the reading frame of the M005L/R gene. Analysis of genes encoding proteins involved in DNA synthesis or RNA transcription did not reveal any mutations likely to cause rapid evolution. Although there was some evidence for recombination across the MYXV phylogeny, this was not associated with the increase in the evolutionary rate. The period from 1996 to 2012 saw significant declines in wild rabbit numbers, due to the introduction of rabbit hemorrhagic disease and prolonged drought in southeastern Australia, followed by the partial recovery of populations. It is therefore possible that a rapidly changing environment for virus transmission changed the selection pressures faced by MYXV, altering the course and pace of virus evolution.IMPORTANCE The coevolution of myxoma virus (MYXV) and European rabbits in Australia is one of the most important natural experiments in evolutionary biology, providing insights into virus adaptation to new hosts and the evolution of virulence. Previous studies of MYXV evolution have also shown that the virus evolves both relatively rapidly and in a strongly clock-like manner. Using newly acquired MYXV genome sequences from Australia, we show that the virus has experienced a dramatic change in evolutionary behavior over the last 20 years, with a breakdown in clock-like structure, the appearance of a rapidly evolving virus lineage, and the accumulation of multiple nonsynonymous and indel mutations. We suggest that this punctuated evolutionary event may reflect a change in selection pressures as rabbit numbers declined following the introduction of rabbit hemorrhagic disease virus and drought in the geographic regions inhabited by rabbits.


Asunto(s)
Evolución Molecular , Genes Virales , Myxoma virus/genética , Sistemas de Lectura Abierta , Filogenia , Infecciones por Poxviridae , Animales , Australia , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/veterinaria , Conejos , Factores de Tiempo , Proteínas Virales/genética , Secuenciación Completa del Genoma
10.
Wiley Interdiscip Rev RNA ; 10(2): e1515, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30381906

RESUMEN

Poxviruses are an unusual family of large double-stranded (ds) DNA viruses that exhibit an incredible degree of self-sufficiency and complexity in their replication and immune evasion strategies. Indeed, amongst their approximately 200 open reading frames (ORFs), poxviruses encode approximately 100 immunomodulatory proteins to counter host responses along with complete DNA synthesis, transcription, mRNA processing and cytoplasmic redox systems that enable them to replicate exclusively in the cytoplasm of infected cells. However, like all other viruses poxviruses do not encode ribosomes and therefore remain completely dependent on gaining access to the host translational machinery in order to synthesize viral proteins. Early studies of these intriguing viruses helped discover the mRNA cap and polyadenylated (polyA) tail that we now know to be present on most eukaryotic messages and which play fundamental roles in mRNA translation, while more recent studies have begun to reveal the remarkable lengths poxviruses go to in order to control both host and viral protein synthesis. Here, we discuss some of the central strategies used by poxviruses and the broader battle that ensues with the host cell to control the translation system, the outcome of which ultimately dictates the fate of infection. This article is categorized under: Translation > Translation Regulation.


Asunto(s)
Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/metabolismo , Poxviridae/genética , Poxviridae/metabolismo , Biosíntesis de Proteínas , Proteínas Virales/genética , Proteínas Virales/metabolismo
11.
Cell Rep ; 23(5): 1249-1258, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29719242

RESUMEN

Post-translational modification of ribosomal subunit proteins (RPs) is emerging as an important means of regulating gene expression. Recently, regulatory ubiquitination of small RPs RPS10 and RPS20 by the ubiquitin ligase ZNF598 was found to function in ribosome sensing and stalling on internally polyadenylated mRNAs during ribosome quality control (RQC). Here, we reveal that ZNF598 and RPS10 negatively regulate interferon-stimulated gene (ISG) expression in primary cells, depletion of which induced ISG expression and a broad antiviral state. However, cell lines lacking interferon responses revealed that ZNF598 E3 ligase activity and ubiquitination of RPS20, but not RPS10, were specifically required for poxvirus replication and synthesis of poxvirus proteins whose encoding mRNAs contain unusual 5' poly(A) leaders. Our findings reveal distinct functions for ZNF598 and its downstream RPS targets, one that negatively regulates ISG expression and infection by a range of viruses while the other is positively exploited by poxviruses.


Asunto(s)
Proteínas Portadoras/inmunología , Regulación Viral de la Expresión Génica/inmunología , Infecciones por Poxviridae/inmunología , Poxviridae/inmunología , Biosíntesis de Proteínas/inmunología , Proteínas Virales/inmunología , Proteínas Portadoras/genética , Células HCT116 , Células HEK293 , Humanos , Interferones/genética , Interferones/inmunología , Poxviridae/genética , Infecciones por Poxviridae/genética , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/inmunología , Proteínas Virales/genética
12.
Sci Rep ; 8(1): 5623, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29618766

RESUMEN

Crocodilepox virus is a large dsDNA virus belonging to the genus Crocodylidpoxvirus, which infects a wide range of host species in the order Crocodylia worldwide. Here, we present genome sequences for a novel saltwater crocodilepox virus, with two subtypes (SwCRV-1 and -2), isolated from the Australian saltwater crocodile. Affected belly skins of juvenile saltwater crocodiles were used to sequence complete viral genomes, and perform electron microscopic analysis that visualized immature and mature virions. Analysis of the SwCRV genomes showed a high degree of sequence similarity to CRV (84.53% and 83.70%, respectively), with the novel SwCRV-1 and -2 complete genome sequences missing 5 and 6 genes respectively when compared to CRV, but containing 45 and 44 predicted unique genes. Similar to CRV, SwCRV also lacks the genes involved in virulence and host range, however, considering the presence of numerous hypothetical and or unique genes in the SwCRV genomes, it is completely reasonable that the genes encoding these functions are present but not recognized. Phylogenetic analysis suggested a monophyletic relationship between SwCRV and CRV, however, SwCRV is quite distinct from other chordopoxvirus genomes. These are the first SwCRV complete genome sequences isolated from saltwater crocodile skin lesions.


Asunto(s)
Caimanes y Cocodrilos/virología , Chordopoxvirinae/genética , Genoma Viral , Genómica/métodos , Infecciones por Poxviridae/genética , Enfermedades de la Piel/genética , Animales , Australia , Chordopoxvirinae/clasificación , Filogenia , Infecciones por Poxviridae/virología , Análisis de Secuencia de ADN , Enfermedades de la Piel/virología , Virulencia
13.
PLoS Pathog ; 14(2): e1006884, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29447249

RESUMEN

Host restriction factors constitute a formidable barrier for viral replication to which many viruses have evolved counter-measures. Human SAMD9, a tumor suppressor and a restriction factor for poxviruses in cell lines, is antagonized by two classes of poxvirus proteins, represented by vaccinia virus (VACV) K1 and C7. A paralog of SAMD9, SAMD9L, is also encoded by some mammals, while only one of two paralogs is retained by others. Here, we show that SAMD9L functions similarly to SAMD9 as a restriction factor and that the two paralogs form a critical host barrier that poxviruses must overcome to establish infection. In mice, which naturally lack SAMD9, overcoming SAMD9L restriction with viral inhibitors is essential for poxvirus replication and pathogenesis. While a VACV deleted of both K1 and C7 (vK1L-C7L-) was restricted by mouse cells and highly attenuated in mice, its replication and virulence were completely restored in SAMD9L-/- mice. In humans, both SAMD9 and SAMD9L are poxvirus restriction factors, although the latter requires interferon induction in many cell types. While knockout of SAMD9 with Crispr-Cas9 was sufficient for abolishing the restriction for vK1L-C7L- in many human cells, knockout of both paralogs was required for abolishing the restriction in interferon-treated cells. Both paralogs are antagonized by VACV K1, C7 and C7 homologs from diverse mammalian poxviruses, but mouse SAMD9L is resistant to the C7 homolog encoded by a group of poxviruses with a narrow host range in ruminants, indicating that host species-specific difference in SAMD9/SAMD9L genes serves as a barrier for cross-species poxvirus transmission.


Asunto(s)
Especificidad del Huésped/genética , Infecciones por Poxviridae/genética , Poxviridae/genética , Poxviridae/patogenicidad , Proteínas/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mamíferos , Ratones , Ratones Noqueados , Células 3T3 NIH , Infecciones por Poxviridae/transmisión , Infecciones por Poxviridae/virología , Proteínas/genética , Homología de Secuencia , Proteínas Supresoras de Tumor/genética , Virus Vaccinia/genética , Virus Vaccinia/patogenicidad , Células Vero
14.
Vaccine ; 35(38): 5131-5139, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28818566

RESUMEN

Infection with human cytomegalovirus (HCMV) can cause severe complications in newborns and immunocompromised patients, and a prophylactic or therapeutic vaccine against HCMV is not available. Here, we generated a HCMV vaccine candidate fulfilling the regulatory requirements for GMP-compliant production and clinical testing. A novel synthetic fusion gene consisting of the coding sequences of HCMV pp65 and IE1 having a deleted nuclear localization sequence and STAT2 binding domain was introduced into the genome of the attenuated vaccinia virus strain MVA. This recombinant MVA, MVA-syn65_IE1, allowed for the production of a stable ∼120kDa syn65_IE1 fusion protein upon tissue culture infection. MVA-syn65_IE1 infected CD40-activated B cells activated and expanded pp65- and IE1-specific T cells derived from HCMV-seropositive donors to at least equal levels as control recombinant MVA expressing single genes for pp65 or IE1. Additionally, we show that MVA-syn65_IE1 induced HCMV pp65- and IE1-epitope specific T cells in HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-knockout mice. Thus, MVA-syn65_IE1 represents a promising vaccine candidate against HCMV and constitutes a basis for the generation of a multivalent vaccine targeting relevant pathogens in immunocompromised patients.


Asunto(s)
Antígenos Virales/inmunología , Citomegalovirus/inmunología , Animales , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Vectores Genéticos/genética , Herpesviridae/genética , Herpesviridae/inmunología , Humanos , Ratones , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
15.
PLoS Pathog ; 13(8): e1006602, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28854224

RESUMEN

The poly(A) leader at the 5'-untranslated region (5'-UTR) is an unusually striking feature of all poxvirus mRNAs transcribed after viral DNA replication (post-replicative mRNAs). These poly(A) leaders are non-templated and of heterogeneous lengths; and their function during poxvirus infection remains a long-standing question. Here, we discovered that a 5'-poly(A) leader conferred a selective translational advantage to mRNA in poxvirus-infected cells. A constitutive and uninterrupted 5'-poly(A) leader with 12 residues was optimal. Because the most frequent lengths of the 5'-poly(A) leaders are 8-12 residues, the result suggests that the poly(A) leader has been evolutionarily optimized to boost poxvirus protein production. A 5'-poly(A) leader also could increase protein production in the bacteriophage T7 promoter-based expression system of vaccinia virus, the prototypic member of poxviruses. Interestingly, although vaccinia virus post-replicative mRNAs do have 5'- methylated guanosine caps and can use cap-dependent translation, in vaccinia virus-infected cells, mRNA with a 5'-poly(A) leader could also be efficiently translated in cells with impaired cap-dependent translation. However, the translation was not mediated through an internal ribosome entry site (IRES). These results point to a fundamental mechanism poxvirus uses to efficiently translate its post-replicative mRNAs.


Asunto(s)
Biosíntesis de Proteínas/genética , ARN Mensajero/genética , ARN Viral/genética , Virus Vaccinia/genética , Replicación Viral/genética , Regiones no Traducidas 5'/genética , Western Blotting , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Reacción en Cadena de la Polimerasa , Infecciones por Poxviridae/genética , Caperuzas de ARN/genética
16.
PLoS One ; 11(12): e0168690, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28005936

RESUMEN

Avipoxvirus (APV) is a fairly common virus affecting birds that causes morbidity and mortality in wild and captive birds. We studied the prevalence of pox-like lesions and genetic diversity of APV in house sparrows (Passer domesticus) in natural, agricultural and urban areas in southern Spain in 2013 and 2014 and in central Spain for 8 months (2012-2013). Overall, 3.2% of 2,341 house sparrows visually examined in southern Spain had cutaneous lesions consistent with avian pox. A similar prevalence (3%) was found in 338 birds from central Spain. Prevalence was higher in hatch-year birds than in adults. We did not detect any clear spatial or temporal patterns of APV distribution. Molecular analyses of poxvirus-like lesions revealed that 63% of the samples were positive. Molecular and phylogenetic analyses of 29 DNA sequences from the fpv167 gene, detected two strains belonging to the canarypox clade (subclades B1 and B2) previously found in Spain. One of them appears predominant in Iberia and North Africa and shares 70% similarity to fowlpox and canarypox virus. This APV strain has been identified in a limited number of species in the Iberian Peninsula, Morocco and Hungary. The second one has a global distribution and has been found in numerous wild bird species around the world. To our knowledge, this represents the largest study of avian poxvirus disease in the broadly distributed house sparrow and strongly supports the findings that Avipox prevalence in this species in South and central Spain is moderate and the genetic diversity low.


Asunto(s)
Avipoxvirus/genética , Enfermedades de las Aves/epidemiología , Enfermedades de las Aves/genética , Variación Genética/genética , Infecciones por Poxviridae/veterinaria , Gorriones/genética , Animales , Anticuerpos Antivirales/sangre , Enfermedades de las Aves/patología , Filogenia , Infecciones por Poxviridae/epidemiología , Infecciones por Poxviridae/genética , Prevalencia , España/epidemiología , Gorriones/virología , Proteínas Virales/genética
17.
Virus Res ; 216: 1-15, 2016 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-26519757

RESUMEN

High-throughput genomics technologies are currently being used to study a wide variety of viral infections, providing insight into which cellular genes and pathways are regulated after infection, and how these changes are related, or not, to efficient elimination of the pathogen. This article will focus on how gene expression studies of infections with non-replicative poxviruses currently used as vaccine vectors provide a global perspective of the molecular events associated with the viral infection in human cells. These high-throughput genomics approaches have the potential to lead to the identification of specific new properties of the viral vector or novel cellular targets that may aid in the development of more effective pox-derived vaccines and antivirals.


Asunto(s)
Interacciones Huésped-Patógeno , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/virología , Poxviridae/genética , Replicación Viral , Animales , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Poxviridae/fisiología , Infecciones por Poxviridae/metabolismo
18.
Mol Immunol ; 67(2 Pt B): 357-68, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26190308

RESUMEN

Interleukin-26 (IL-26) is a member of the IL-10 family of cytokines. Though conserved across vertebrates, the IL-26 gene is functionally inactivated in a few mammals like rat, mouse and horse. We report here the identification, isolation and cloning of the cDNA of IL-26 from the dromedary camel. The camel cDNA contains a 516 bp open reading frame encoding a 171 amino acid precursor protein, including a 21 amino acid signal peptide. Sequence analysis revealed high similarity with other mammalian IL-26 homologs and the conservation of IL-10 cytokine family domain structure including key amino acid residues. We also report the identification and cloning of four novel transcript variants produced by alternative splicing at the Exon 3-Exon 4 regions of the gene. Three of the alternative splice variants had premature termination codons and are predicted to code for truncated proteins. The transcript variant 4 (Tv4) having an insertion of an extra 120 bp nucleotides in the ORF was predicted to encode a full length protein product with 40 extra amino acid residues. The mRNA transcripts of all the variants were identified in lymph node, where as fewer variants were observed in other tissues like blood, liver and kidney. The expression of Tv2 and Tv3 were found to be up regulated in mitogen induced camel peripheral blood mononuclear cells. IL-26-Tv2 expression was also induced in camel fibroblast cells infected with Camel pox virus in-vitro. The identification of the transcript variants of IL-26 from the dromedary camel is the first report of alternative splicing for IL-26 in a species in which the gene has not been inactivated.


Asunto(s)
Empalme Alternativo/genética , Camelus/genética , Interleucinas/genética , Empalme Alternativo/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Clonación Molecular , ADN Complementario/genética , Exones/genética , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Perfilación de la Expresión Génica , Interleucinas/metabolismo , Intrones/genética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Mitógenos/farmacología , Datos de Secuencia Molecular , Orthopoxvirus , Ovario/patología , Filogenia , Infecciones por Poxviridae/genética , Infecciones por Poxviridae/patología , Infecciones por Poxviridae/virología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Homología Estructural de Proteína
19.
J Virol ; 89(18): 9348-67, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26136578

RESUMEN

UNLABELLED: Poxviruses are large DNA viruses of vertebrates and insects causing disease in many animal species, including reptiles, birds, and mammals. Although poxvirus-like particles were detected in diseased farmed koi carp, ayu, and Atlantic salmon, their genetic relationships to poxviruses were not established. Here, we provide the first genome sequence of a fish poxvirus, which was isolated from farmed Atlantic salmon. In the present study, we used quantitative PCR and immunohistochemistry to determine aspects of salmon gill poxvirus disease, which are described here. The gill was the main target organ where immature and mature poxvirus particles were detected. The particles were detected in detaching, apoptotic respiratory epithelial cells preceding clinical disease in the form of lethargy, respiratory distress, and mortality. In moribund salmon, blocking of gas exchange would likely be caused by the adherence of respiratory lamellae and epithelial proliferation obstructing respiratory surfaces. The virus was not found in healthy salmon or in control fish with gill disease without apoptotic cells, although transmission remains to be demonstrated. PCR of archival tissue confirmed virus infection in 14 cases with gill apoptosis in Norway starting from 1995. Phylogenomic analyses showed that the fish poxvirus is the deepest available branch of chordopoxviruses. The virus genome encompasses most key chordopoxvirus genes that are required for genome replication and expression, although the gene order is substantially different from that in other chordopoxviruses. Nevertheless, many highly conserved chordopoxvirus genes involved in viral membrane biogenesis or virus-host interactions are missing. Instead, the salmon poxvirus carries numerous genes encoding unknown proteins, many of which have low sequence complexity and contain simple repeats suggestive of intrinsic disorder or distinct protein structures. IMPORTANCE: Aquaculture is an increasingly important global source of high-quality food. To sustain the growth in aquaculture, disease control in fish farming is essential. Moreover, the spread of disease from farmed fish to wildlife is a concern. Serious poxviral diseases are emerging in aquaculture, but very little is known about the viruses and the diseases that they cause. There is a possibility that viruses with enhanced virulence may spread to new species, as has occurred with the myxoma poxvirus in rabbits. Provision of the first fish poxvirus genome sequence and specific diagnostics for the salmon gill poxvirus in Atlantic salmon may help curb this disease and provide comparative knowledge. Furthermore, because salmon gill poxvirus represents the deepest branch of chordopoxvirus so far discovered, the genome analysis provided substantial insight into the evolution of different functional modules in this important group of viruses.


Asunto(s)
Carpas/virología , Chordopoxvirinae/genética , Enfermedades de los Peces/virología , Branquias/virología , Filogenia , Infecciones por Poxviridae/genética , Salmo salar/virología , Animales , Chordopoxvirinae/metabolismo , Enfermedades de los Peces/genética , Enfermedades de los Peces/metabolismo , Branquias/metabolismo , Infecciones por Poxviridae/metabolismo , Conejos
20.
Virus Res ; 208: 180-8, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26113305

RESUMEN

Interferons (IFNs) play a critical role as a first line of defence against viral infection. Activation of the Janus kinase/signal transducer and activation of transcription (JAK/STAT) pathway by IFNs leads to the production of IFN stimulated genes (ISGs) that block viral replication. The Parapoxvirus, Orf virus (ORFV) induces acute pustular skin lesions of sheep and goats and is transmissible to man. The virus replicates in keratinocytes that are the immune sentinels of skin. We investigated whether or not ORFV could block the expression of ISGs. The human gene GBP1 is stimulated exclusively by type II IFN while MxA is stimulated exclusively in response to type I IFNs. We found that GBP1 and MxA were strongly inhibited in ORFV infected HeLa cells stimulated with IFN-γ or IFN-α respectively. Furthermore we showed that ORFV inhibition of ISG expression was not affected by cells pretreated with adenosine N1-oxide (ANO), a molecule that inhibits poxvirus mRNA translation. This suggested that new viral gene synthesis was not required and that a virion structural protein was involved. We next investigated whether ORFV infection affected STAT1 phosphorylation in IFN-γ or IFN-α treated HeLa cells. We found that ORFV reduced the levels of phosphorylated STAT1 in a dose-dependent manner and was specific for Tyr701 but not Ser727. Treatment of cells with sodium vanadate suggested that a tyrosine phosphatase was responsible for dephosphorylating STAT1-p. ORFV encodes a factor, ORFV057, with homology to the vaccinia virus structural protein VH1 that impairs the JAK/STAT pathway by dephosphorylating STAT1. Our findings show that ORFV has the capability to block ISG expression and modulate the JAK/STAT signalling pathway.


Asunto(s)
Interferones/metabolismo , Quinasas Janus/metabolismo , Infecciones por Poxviridae/genética , Factor de Transcripción STAT1/metabolismo , Línea Celular , Proteínas de Unión al GTP/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Quinasas Janus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Virus del Orf/genética , Virus del Orf/metabolismo , Fosforilación , Infecciones por Poxviridae/metabolismo , Factor de Transcripción STAT1/genética , Transducción de Señal , Proteínas Virales/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...