Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Emerg Infect Dis ; 30(4): 721-731, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38526136

RESUMEN

Genetically diverse simian arteriviruses (simarteriviruses) naturally infect geographically and phylogenetically diverse monkeys, and cross-species transmission and emergence are of considerable concern. Characterization of most simarteriviruses beyond sequence analysis has not been possible because the viruses fail to propagate in the laboratory. We attempted to isolate 4 simarteriviruses, Kibale red colobus virus 1, Pebjah virus, simian hemorrhagic fever virus, and Southwest baboon virus 1, by inoculating an immortalized grivet cell line (known to replicate simian hemorrhagic fever virus), primary macaque cells, macrophages derived from macaque induced pluripotent stem cells, and mice engrafted with macaque CD34+-enriched hematopoietic stem cells. The combined effort resulted in successful virus isolation; however, no single approach was successful for all 4 simarteriviruses. We describe several approaches that might be used to isolate additional simarteriviruses for phenotypic characterization. Our results will expedite laboratory studies of simarteriviruses to elucidate virus-host interactions, assess zoonotic risk, and develop medical countermeasures.


Asunto(s)
Arterivirus , Animales , Ratones , Arterivirus/genética , Macaca , Macrófagos , Línea Celular
2.
Emerg Infect Dis ; 27(2): 578-581, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33496231

RESUMEN

In the fall of 2019, a fatal encephalitis outbreak led to the deaths of >200 European hedgehogs (Erinaceus europaeus) in England. We used next-generation sequencing to identify a novel arterivirus with a genome coding sequence of only 43% similarity to existing GenBank arterivirus sequences.


Asunto(s)
Arterivirus , Encefalitis , Animales , Brotes de Enfermedades , Inglaterra/epidemiología , Erizos
3.
J Gen Virol ; 102(8)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34356005

RESUMEN

The family Arteriviridae comprises enveloped RNA viruses with a linear, positive-sense genome of approximately 12.7 to 15.7 kb. The spherical, pleomorphic virions have a median diameter of 50-74 nm and include eight to eleven viral proteins. Arteriviruses infect non-human mammals in a vector-independent manner. Infections are often persistent and can either be asymptomatic or produce overt disease. Some arteriviruses are important veterinary pathogens while others infect particular species of wild rodents or African non-human primates. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Arteriviridae, which is available at ictv.global/report/arteriviridae.


Asunto(s)
Arteriviridae/clasificación , Arteriviridae/genética , Filogenia , Animales , Arteriviridae/ultraestructura , Arterivirus/clasificación , Arterivirus/genética , Endocitosis , Genoma Viral , Primates , Infecciones por Virus ARN , Proteínas Virales/genética , Virión/clasificación , Virión/genética , Virión/ultraestructura , Acoplamiento Viral , Replicación Viral
4.
Fish Shellfish Immunol ; 98: 653-660, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31676431

RESUMEN

Trionyx sinensis Hemorrhagic Syndrome Virus (TSHSV) is the firstly discovered aquatic arterivirus inducing high mortality of Trionyx sinensis. So far, the lack of genomic resources has hindered further research on revealing the immunological characteristics of T. sinensis in response to TSHSV. In the present study, we performed a transcriptome analysis from the lungs of T. sinensis challenged by TSHSV using Illumina-based RNA-Seq. The validity of transcriptomic data was confirmed with the gradual increase of TSHSV RNA copies detected in lung. A total of 103079339 clean reads were generated, and 58374764 unique mapped reads were analyzed. Assembly of the sequence data allowed identifying 16383 unigenes consisting of 36 significant differentially expressed genes (DEGs). These DEGs were categorized into 30 GO-enriched bioprocesses and 9 KEGG pathways. The combinational analysis of GO-enriched bioprocesses and KEGG pathways demonstrated that TSHSV modulated several immune genes of T. sinensis related to various biological processes, including virus recognition (RIG-I/MDA-5), immune initiation (IFIT-1 and IFIT-5), endocytosis (CUBN, ENPP2 and LRP2) and steroid metabolism (FCNIL and STAR). In summary, the finding of this study revealed several immune pathways and candidated genes involved in the immune response of T. sinensis against TSHSV-infection. These results will provide helpful information to investigate molecular mechanism of T. sinensis in response to TSHSV.


Asunto(s)
Arteriviridae/fisiología , Pulmón/metabolismo , Infecciones por Virus ARN/veterinaria , Transcriptoma , Tortugas , Animales , Perfilación de la Expresión Génica/veterinaria , Pulmón/virología , Infecciones por Virus ARN/metabolismo , Infecciones por Virus ARN/virología , RNA-Seq/veterinaria , Proteínas de Reptiles/análisis
5.
J Virol ; 92(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29769343

RESUMEN

Glycoprotein 3 (GP3) of the arterivirus porcine reproductive and respiratory syndrome virus (PRRSV) consists of a cleaved signal peptide, a highly glycosylated domain, a short hydrophobic region, and an unglycosylated C-terminal domain. GP3 is supposed to form a complex with GP2 and GP4 in virus particles, but secretion of GP3 from cells has also been reported. We analyzed the membrane topology of GP3 from various PRRSV strains. A fraction of the protein is secreted from transfected cells, GP3 from PRRSV-1 strains to a greater extent than GP3 from PRRSV-2 strains. This secretion behavior is reversed after exchange of the variable C-terminal domain. A fluorescence protease protection assay shows that the C terminus of GP3, fused to green fluorescent protein (GFP), is resistant to proteolytic digestion in permeabilized cells. Furthermore, glycosylation sites inserted into the C-terminal part of GP3 are used. Both experiments indicate that the C terminus of GP3 is translocated into the lumen of the endoplasmic reticulum. Deletion of the conserved hydrophobic region greatly enhances secretion of GP3, and fusion of this domain to GFP promotes membrane anchorage. Bioinformatics suggests that the hydrophobic region forms an amphipathic helix. Accordingly, exchanging only a few amino acids in its hydrophilic face prevents secretion of GP3 and in its hydrophobic face enhances it. Exchanging the latter amino acids in the context of the viral genome did not affect release of virions, but released particles were not infectious. In sum, GP3 exhibits an unusual hairpin-like membrane topology that might explain why a fraction of the protein is secreted.IMPORTANCE PRRSV is the most important pathogen in the pork industry. It causes persistent infections that lead to reduced weight gain of piglets; highly pathogenic strains even kill 90% of an infected pig population. PRRSV cannot be eliminated from pig farms by vaccination due to the large amino acid variability between the existing strains, especially in the glycoproteins. Here, we analyzed basic structural features of GP3 from various PRRSV strains. We show that the protein exhibits an unusual hairpin-like membrane topology; membrane anchoring might occur via an amphipathic helix. This rather weak membrane anchor explains why a fraction of the protein is secreted from cells. Interestingly, PRRSV-1 strains secrete more GP3 than PRRSV-2. We speculate that secreted GP3 plays a role during PRRSV infection of pigs: it might serve as a decoy to distract antibodies away from virus particles.


Asunto(s)
Membrana Celular , Glicoproteínas , Fusión de Membrana , Virus del Síndrome Respiratorio y Reproductivo Porcino , Proteínas Virales , Sustitución de Aminoácidos , Animales , Células CHO , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Cricetulus , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilación , Células HEK293 , Humanos , Mutación Missense , Virus del Síndrome Respiratorio y Reproductivo Porcino/química , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Virus del Síndrome Respiratorio y Reproductivo Porcino/metabolismo , Dominios Proteicos , Estructura Secundaria de Proteína , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
6.
Virology ; 597: 110122, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38850896

RESUMEN

Glycoprotein 3 (GP3) serves as a structural protein in equine arteritis virus (EAV), forming a heterotrimeric complex that plays a pivotal role in virus tropism. In this study, we tested the membrane topology of GP3, both when expressed separately and during infection with recombinant tagged EAV GP3-HA. In our antibody accessibility experiment, we made a noteworthy discovery: GP3, when expressed separately, exhibits a dual topology. We introduced an additional N-glycosylation site, which was only partially used, providing further evidence for the dual topology of GP3. Intriguingly, this mutated GP3 was secreted into the medium, a result of the disruption of the ER retention motif RXR. The additional glycosylation site was not used when we examined the recombinant EAV virus with the same mutation. Despite the fact of higher expression levels of mutant GP3-HA, the protein was not secreted, and the recombinant mutant virus did not have growth delay compared to the EAV wild-type virus. This finding suggests that GP3 has a single type one membrane topology in virus infected cells, whereas the expression of GP3 in trans results in the dual topology of this protein. The RXR motif in the C-terminus is a co-factor of ER retention of the protein, but the main retention signal remains elusive.


Asunto(s)
Secuencias de Aminoácidos , Retículo Endoplásmico , Equartevirus , Equartevirus/genética , Equartevirus/metabolismo , Animales , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Arginina/metabolismo , Arginina/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/química , Glicosilación , Línea Celular , Caballos , Humanos
7.
Viruses ; 15(5)2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37243236

RESUMEN

As part of a continuous effort to investigate the viral communities associated with wild mammals at the human-animal interface in an Amazonian metropolitan region, this study describes the detection of a novel rodent-borne arterivirus. A sample containing pooled organs of Oecomys paricola was submitted to RNA sequencing, and four sequences taxonomically assigned as related to the Arteriviridae family were recovered, corresponding to an almost complete genome of nearly 13 kb summed. In the phylogenetic analysis with the standard domains used for taxa demarcation in the family, the tentatively named Oecomys arterivirus 1 (OAV-1) was placed within the clade of rodent- and porcine-associated viruses, corresponding to the Variarterivirinae subfamily. The divergence analysis, based on the same amino acid alignment, corroborated the hypothesis that the virus may represent a new genus within the subfamily. These findings contribute to the expansion of the current knowledge about the diversity, host and geographical range of the viral family. Arterivirids are non-human pathogens and are usually species-specific, but the susceptibility of cell lines derived from different organisms should be conducted to confirm these statements for this proposed new genus in an initial attempt to assess its spillover potential.


Asunto(s)
Arteriviridae , Arterivirus , Animales , Porcinos , Filogenia , Brasil , Arterivirus/genética , Mamíferos , Roedores
8.
Viruses ; 15(1)2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36680298

RESUMEN

The emergence of recombinant PRRSV strains has been observed for more than a decade. These recombinant viruses are characterized by a genome that contains genetic material from at least two different parental strains. Due to the advanced sequencing techniques and a growing number of data bank entries, the role of PRRSV recombinants has become increasingly important since they are sometimes associated with clinical outbreaks. Chimeric viruses observed more recently are products of PRRSV wild-type and vaccine strains. Here, we report on three PRRSV-1 isolates from geographically distant farms with differing clinical manifestations. A sequencing and recombination analysis revealed that these strains are crossovers between different wild-type strains and the same modified live virus vaccine strain. Interestingly, the recombination breakpoint of all analyzed isolates appears at the beginning of open reading frame 5 (ORF5). RNA structure predictions indicate a conserved stem loop in close proximity to the recombination hotspot, which is a plausible cause of a polymerase template switch during RNA replication. Further research into the mechanisms of the stem loop is needed to help understand the PRRSV recombination process and the role of MLVs as parental strains.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Animales , Porcinos , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Sistemas de Lectura Abierta , Recombinación Genética , Filogenia
9.
Viruses ; 14(9)2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36146698

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most relevant porcine pathogens worldwide. Active control of the disease relies on modified live virus vaccines (MLVs), as most inactivated vaccines provide very limited protection. Neutralizing antibodies occur late in infection; therefore, CD8+ T cells are considered important correlates of protection and are a frequent focus of investigation. Our aim was to identify viral peptides naturally bound by the class I major histocompatibility complex (MHC-I) and to confirm their ability to stimulate CD8+ T cells. For this purpose, we immunoprecipitated MHC-I/peptide complexes of PRRSV (strain AUT15-33) -infected cells (SLA-I Lr-Hp 35.0/24 mod) to isolate the viral epitopes and analyzed them with liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Furthermore, we employed these identified peptides to stimulate peripheral blood mononuclear cells (PBMCs) of previously PRRSV-infected pigs and measured the PRRSV-specific CD8+ T-cell response with an intracellular cytokine staining (ICS). Our data revealed that PRRSV non-structural proteins (NSPs), encoded in open reading frame 1a and 1b (ORF1), present the major source of MHC-I-presented peptides. Additionally, we show that our identified epitopes are able to trigger IFNγ responses in vitro. These findings are a basis for understanding the proteasomal degradation of PRRSV proteins, the cellular ability to display them via MHC-I, and their potential to restimulate CD8+ T cells.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Vacunas Virales , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Linfocitos T CD8-positivos , Cromatografía Liquida , Citocinas , Epítopos , Leucocitos Mononucleares , Complejo Mayor de Histocompatibilidad , Péptidos , Porcinos , Espectrometría de Masas en Tándem , Vacunas Atenuadas , Vacunas de Productos Inactivados
10.
Viruses ; 13(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34578423

RESUMEN

Recent years have witnessed the discovery of several new viruses belonging to the family Arteriviridae, expanding the known diversity and host range of this group of complex RNA viruses. Although the pathological relevance of these new viruses is not always clear, several well-studied members of the family Arteriviridae are known to be important animal pathogens. Here, we report the complete genome sequences of four new arterivirus variants, belonging to two putative novel species. These new arteriviruses were discovered in African rodents and were given the names Lopma virus and Praja virus. Their genomes follow the characteristic genome organization of all known arteriviruses, even though they are only distantly related to currently known rodent-borne arteriviruses. Phylogenetic analysis shows that Lopma virus clusters in the subfamily Variarterivirinae, while Praja virus clusters near members of the subfamily Heroarterivirinae: the yet undescribed forest pouched giant rat arterivirus and hedgehog arterivirus 1. A co-divergence analysis of rodent-borne arteriviruses confirms that they share similar phylogenetic patterns with their hosts, with only very few cases of host shifting events throughout their evolutionary history. Overall, the genomes described here and their unique clustering with other arteriviruses further illustrate the existence of multiple rodent-borne arterivirus lineages, expanding our knowledge of the evolutionary origin of these viruses.


Asunto(s)
Arteriviridae/genética , Genoma Viral , Infecciones por Virus ARN/veterinaria , Enfermedades de los Roedores/virología , Roedores/virología , África del Sur del Sahara , Animales , Arteriviridae/clasificación , Arteriviridae/aislamiento & purificación , Evolución Biológica , Secuenciación de Nucleótidos de Alto Rendimiento , Filogenia , Infecciones por Virus ARN/virología , Secuenciación Completa del Genoma
11.
Viruses ; 13(4)2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917085

RESUMEN

Simian hemorrhagic fever virus (SHFV) causes acute, lethal disease in macaques. We developed a single-plasmid cDNA-launch infectious clone of SHFV (rSHFV) and modified the clone to rescue an enhanced green fluorescent protein-expressing rSHFV-eGFP that can be used for rapid and quantitative detection of infection. SHFV has a narrow cell tropism in vitro, with only the grivet MA-104 cell line and a few other grivet cell lines being susceptible to virion entry and permissive to infection. Using rSHFV-eGFP, we demonstrate that one cricetid rodent cell line and three ape cell lines also fully support SHFV replication, whereas 55 human cell lines, 11 bat cell lines, and three rodent cells do not. Interestingly, some human and other mammalian cell lines apparently resistant to SHFV infection are permissive after transfection with the rSHFV-eGFP cDNA-launch plasmid. To further demonstrate the investigative potential of the infectious clone system, we introduced stop codons into eight viral open reading frames (ORFs). This approach suggested that at least one ORF, ORF 2b', is dispensable for SHFV in vitro replication. Our proof-of-principle experiments indicated that rSHFV-eGFP is a useful tool for illuminating the understudied molecular biology of SHFV.


Asunto(s)
Arterivirus/genética , ADN Complementario/genética , Proteínas Fluorescentes Verdes/genética , Sistemas de Lectura Abierta , ARN Viral/genética , Recombinación Genética , Replicación Viral/genética , Animales , Arterivirus/fisiología , Línea Celular , Quirópteros , Hominidae , Humanos , Plásmidos/genética , Prueba de Estudio Conceptual , Roedores
12.
Viruses ; 11(1)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650570

RESUMEN

Simian hemorrhagic fever virus (SHFV) causes a fulminant and typically lethal viral hemorrhagic fever (VHF) in macaques (Cercopithecinae: Macaca spp.) but causes subclinical infections in patas monkeys (Cercopithecinae: Erythrocebus patas). This difference in disease course offers a unique opportunity to compare host responses to infection by a VHF-causing virus in biologically similar susceptible and refractory animals. Patas and rhesus monkeys were inoculated side-by-side with SHFV. Unlike the severe disease observed in rhesus monkeys, patas monkeys developed a limited clinical disease characterized by changes in complete blood counts, serum chemistries, and development of lymphadenopathy. Viral RNA was measurable in circulating blood 2 days after exposure, and its duration varied by species. Infectious virus was detected in terminal tissues of both patas and rhesus monkeys. Varying degrees of overlap in changes in serum concentrations of interferon (IFN)-γ, monocyte chemoattractant protein (MCP)-1, and interleukin (IL)-6 were observed between patas and rhesus monkeys, suggesting the presence of common and species-specific cytokine responses to infection. Similarly, quantitative immunohistochemistry of livers from terminal monkeys and whole blood flow cytometry revealed varying degrees of overlap in changes in macrophages, natural killer cells, and T-cells. The unexpected degree of overlap in host response suggests that relatively small subsets of a host's response to infection may be responsible for driving hemorrhagic fever pathogenesis. Furthermore, comparative SHFV infection in patas and rhesus monkeys offers an experimental model to characterize host⁻response mechanisms associated with viral hemorrhagic fever and evaluate pan-viral hemorrhagic fever countermeasures.


Asunto(s)
Infecciones por Arterivirus/veterinaria , Arterivirus/patogenicidad , Fiebres Hemorrágicas Virales/veterinaria , Interacciones Huésped-Patógeno , Enfermedades de los Monos/inmunología , Animales , Anticuerpos Antivirales/sangre , Arterivirus/inmunología , Infecciones por Arterivirus/inmunología , Citocinas/sangre , Erythrocebus , Femenino , Fiebres Hemorrágicas Virales/inmunología , Macaca , Macrófagos/virología , Masculino , Enfermedades de los Monos/virología , ARN Viral , Replicación Viral
13.
Virology ; 517: 56-61, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29429554

RESUMEN

It has been established that reduced susceptibility to porcine reproductive and respiratory syndrome virus (PRRSV) has a genetic component. This genetic component may take the form of small non-coding RNAs (sncRNA), which are molecules that function as regulators of gene expression. Various sncRNAs have emerged as having an important role in the immune system in humans. The study uses transcriptomic read counts to profile the type and quantity of both well and lesser characterized sncRNAs, such as microRNAs and small nucleolar RNAs to identify and quantify the classes of sncRNA expressed in whole blood between healthy and highly pathogenic PRRSV-infected pigs. Our results returned evidence on nine classes of sncRNA, four of which were consistently statistically significantly different based on Fisher's Exact Test, that can be detected and possibly interrogated for their effect on host dysregulation during PRRSV infections.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , ARN Pequeño no Traducido/sangre , ARN Viral/genética , Porcinos/sangre , Animales , Regulación Viral de la Expresión Génica/fisiología , Síndrome Respiratorio y de la Reproducción Porcina/sangre
14.
Viruses ; 10(12)2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30544677

RESUMEN

Simarteriviruses (Arteriviridae: Simarterivirinae) are commonly found at high titers in the blood of African monkeys but do not cause overt disease in these hosts. In contrast, simarteriviruses cause severe disease in Asian macaques upon accidental or experimental transmission. Here, we sought to better understand the host-dependent drivers of simarterivirus pathogenesis by infecting olive baboons (n = 4) and rhesus monkeys (n = 4) with the simarterivirus Southwest baboon virus 1 (SWBV-1). Surprisingly, none of the animals in our study showed signs of disease following SWBV-1 inoculation. Three animals (two rhesus monkeys and one olive baboon) became infected and sustained high levels of SWBV-1 viremia for the duration of the study. The course of SWBV-1 infection was highly predictable: plasma viremia peaked between 1 × 107 and 1 × 108 vRNA copies/mL at 3⁻10 days post-inoculation, which was followed by a relative nadir and then establishment of a stable set-point between 1 × 106 and 1 × 107 vRNA copies/mL for the remainder of the study (56 days). We characterized cellular and antibody responses to SWBV-1 infection in these animals, demonstrating that macaques and baboons mount similar responses to SWBV-1 infection, yet these responses are ineffective at clearing SWBV-1 infection. SWBV-1 sequencing revealed the accumulation of non-synonymous mutations in a region of the genome that corresponds to an immunodominant epitope in the simarterivirus major envelope glycoprotein GP5, which likely contribute to viral persistence by enabling escape from host antibodies.


Asunto(s)
Arteriviridae/patogenicidad , Infecciones Asintomáticas , Macaca mulatta/virología , Papio/virología , Infecciones por Virus ARN/veterinaria , Animales , Anticuerpos Antivirales/sangre , Genoma Viral , Inmunidad Celular , Masculino , Mutación , Infecciones por Virus ARN/inmunología , Proteínas del Envoltorio Viral/inmunología , Carga Viral , Viremia , Replicación Viral
15.
Virology ; 517: 69-76, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29229370

RESUMEN

We reported previously that carbohydrate attachment to an overlapping glycosylation site adjacent to the signal peptide of GP3 from equine arteritis virus (EAV) prevents cleavage. Here we investigated whether this unusual processing scheme is a feature of GP3s of other Arteriviridae, which all contain a glycosylation site at a similar position. Expression of GP3 from type-1 and type-2 porcine reproductive and respiratory syndrome virus (PRRSV) and from lactate dehydrogenase-elevating virus (LDV) revealed that the first glycosylation site is used, but has no effect on signal peptide cleavage. Comparison of the SDS-PAGE mobility of deglycosylated GP3 from PRRSV and LDV with mutants having or not having a signal peptide showed that GP3´s signal peptide is cleaved. Swapping the signal peptides between GP3 of EAV and PRRSV revealed that the information for co-translational processing is not encoded in the signal peptide, but in the remaining part of GP3.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Cricetinae , Transducción de Señal , Proteínas del Envoltorio Viral/genética
16.
Virology ; 458-459: 136-50, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24928046

RESUMEN

Type I interferons (IFNs-α/ß) play a key role for the antiviral state of host, and the porcine arterivirus; porcine reproductive and respiratory syndrome virus (PRRSV), has been shown to down-regulate the production of IFNs during infection. Non-structural protein (nsp) 1 of PRRSV has been identified as a viral IFN antagonist, and the nsp1α subunit of nsp1 has been shown to degrade the CREB-binding protein (CBP) and to inhibit the formation of enhanceosome thus resulting in the suppression of IFN production. The study was expanded to other member viruses in the family Arteriviridae: equine arteritis virus (EAV), murine lactate dehydrogenase-elevating virus (LDV), and simian hemorrhagic fever virus (SHFV). While PRRSV-nsp1 and LDV-nsp1 were auto-cleaved to produce the nsp1α and nsp1ß subunits, EAV-nsp1 remained uncleaved. SHFV-nsp1 was initially predicted to be cleaved to generate three subunits (nsp1α, nsp1ß, and nsp1γ), but only two subunits were generated as SHFV-nsp1αß and SHFV-nsp1γ. The papain-like cysteine protease (PLP) 1α motif in nsp1α remained inactive for SHFV, and only the PLP1ß motif of nsp1ß was functional to generate SHFV-nsp1γ subunit. All subunits of arterivirus nsp1 were localized in the both nucleus and cytoplasm, but PRRSV-nsp1ß, LDV-nsp1ß, EAV-nsp1, and SHFV-nsp1γ were predominantly found in the nucleus. All subunits of arterivirus nsp1 contained the IFN suppressive activity and inhibited both interferon regulatory factor 3 (IRF3) and NF-κB mediated IFN promoter activities. Similar to PRRSV-nsp1α, CBP degradation was evident in cells expressing LDV-nsp1α and SHFV-nsp1γ, but no such degradation was observed for EAV-nsp1. Regardless of CBP degradation, all subunits of arterivirus nsp1 suppressed the IFN-sensitive response element (ISRE)-promoter activities. Our data show that the nsp1-mediated IFN modulation is a common strategy for all arteriviruses but their mechanism of action may differ from each other.


Asunto(s)
Arteriviridae/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Interferón Tipo I/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Arteriviridae/genética , Línea Celular , Clonación Molecular , Humanos , Proteínas no Estructurales Virales/genética , Replicación Viral
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda