Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Trends Genet ; 37(1): 46-58, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32977999

RESUMEN

The concept that type I interferons (IFN-I) are essential to antiviral immunity derives from studies on animal models and cell lines. Virtually all pathogenic viruses have evolved countermeasures to IFN-I restriction, and genetic loss of viral IFN-I antagonists leads to virus attenuation. But just how important is IFN-I to antiviral defence in humans? The recent discovery of genetic defects of IFN-I signalling illuminates this and other questions of IFN biology, including the role of the mucosa-restricted type III IFNs (IFN-III), informing our understanding of the place of the IFN system within the concerted antiviral response. Here we review monogenic lesions of IFN-I signalling pathways and summarise the organising principles which emerge.


Asunto(s)
Antivirales/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/antagonistas & inhibidores , Virus/inmunología , Animales , Antivirales/farmacología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Transducción de Señal , Virus/efectos de los fármacos
2.
J Gen Virol ; 104(11)2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37962188

RESUMEN

Parainfluenza virus type 5 (PIV5) can either have a persistent or a lytic phenotype in cultured cells. We have previously shown that the phenotype is determined by the phosphorylation status of the phosphoprotein (P). Single amino acid substitutions at critical residues, including a serine-to-phenylalanine substitution at position 157 on P, result in a switch between persistent and lytic phenotypes. Here, using PIV5 vectors expressing either mCherry or GFP with persistent or lytic phenotypes, we show that in co-infections the persistent phenotype is dominant. Thus, in contrast to the cell death observed with cells infected solely with the lytic variant, in co-infected cells persistence is immediately established and both lytic and persistent genotypes persist. Furthermore, 10-20 % of virus released from dually infected cells contains both genotypes, indicating that PIV5 particles can package more than one genome. Co-infected cells continue to maintain both genotypes/phenotypes during cell passage, as do individual colonies of cells derived from a culture of persistently infected cells. A refinement of our model on how the dynamics of virus selection may occur in vivo is presented.


Asunto(s)
Coinfección , Virus de la Parainfluenza 5 , Paramyxovirinae , Infecciones por Respirovirus , Humanos , Virus de la Parainfluenza 5/genética , Fenotipo
3.
J Immunol ; 205(1): 261-271, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32423918

RESUMEN

IFNs, produced during viral infections, induce the expression of hundreds of IFN-stimulated genes (ISGs). Some ISGs have specific antiviral activity, whereas others regulate the cellular response. Besides functioning as an antiviral effector, ISG15 is a negative regulator of IFN signaling, and inherited ISG15 deficiency leads to autoinflammatory IFNopathies, in which individuals exhibit elevated ISG expression in the absence of pathogenic infection. We have recapitulated these effects in cultured human A549-ISG15-/- cells and (using A549-UBA7-/- cells) confirmed that posttranslational modification by ISG15 (ISGylation) is not required for regulation of the type I IFN response. ISG15-deficient cells pretreated with IFN-α were resistant to paramyxovirus infection. We also showed that IFN-α treatment of ISG15-deficient cells led to significant inhibition of global protein synthesis, leading us to ask whether resistance was due to the direct antiviral activity of ISGs or whether cells were nonpermissive because of translation defects. We took advantage of the knowledge that IFN-induced protein with tetratricopeptide repeats 1 (IFIT1) is the principal antiviral ISG for parainfluenza virus 5. Knockdown of IFIT1 restored parainfluenza virus 5 infection in IFN-α-pretreated, ISG15-deficient cells, confirming that resistance was due to the direct antiviral activity of the IFN response. However, resistance could be induced if cells were pretreated with IFN-α for longer times, presumably because of inhibition of protein synthesis. These data show that the cause of virus resistance is 2-fold; ISG15 deficiency leads to the early overexpression of specific antiviral ISGs, but the later response is dominated by an unanticipated, ISG15-dependent loss of translational control.


Asunto(s)
Citocinas/deficiencia , Resistencia a la Enfermedad/genética , Interferón-alfa/metabolismo , Infecciones por Paramyxoviridae/inmunología , Transducción de Señal/inmunología , Ubiquitinas/deficiencia , Células A549 , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Chlorocebus aethiops , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Humanos , Virus de la Parainfluenza 2 Humana/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Virus de la Parainfluenza 5/inmunología , Infecciones por Paramyxoviridae/virología , Procesamiento Proteico-Postraduccional/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/genética , Enzimas Activadoras de Ubiquitina/genética , Células Vero
4.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32295916

RESUMEN

During the replication of parainfluenza virus 5 (PIV5), copyback defective virus genomes (DVGs) are erroneously produced and are packaged into "infectious" virus particles. Copyback DVGs are the primary inducers of innate intracellular responses, including the interferon (IFN) response. While DVGs can interfere with the replication of nondefective (ND) virus genomes and activate the IFN-induction cascade before ND PIV5 can block the production of IFN, we demonstrate that the converse is also true, i.e., high levels of ND virus can block the ability of DVGs to activate the IFN-induction cascade. By following the replication and amplification of DVGs in A549 cells that are deficient in a variety of innate intracellular antiviral responses, we show that DVGs induce an uncharacterized IFN-independent innate response(s) that limits their replication. High-throughput sequencing was used to characterize the molecular structure of copyback DVGs. While there appears to be no sequence-specific break or rejoining points for the generation of copyback DVGs, our findings suggest there are region, size, and/or structural preferences selected for during for their amplification.IMPORTANCE Copyback defective virus genomes (DVGs) are powerful inducers of innate immune responses both in vitro and in vivo They impact the outcome of natural infections, may help drive virus-host coevolution, and promote virus persistence. Due to their potent interfering and immunostimulatory properties, DVGs may also be used therapeutically as antivirals and vaccine adjuvants. However, little is known of the host cell restrictions which limit their amplification. We show here that the generation of copyback DVGs readily occurs during parainfluenza virus 5 (PIV5) replication, but that their subsequent amplification is restricted by the induction of innate intracellular responses. Molecular characterization of PIV5 copyback DVGs suggests that while there are no genome sequence-specific breaks or rejoin points for the generation of copyback DVGs, genome region, size, and structural preferences are selected for during their evolution and amplification.


Asunto(s)
Inmunidad Innata/inmunología , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/inmunología , Células A549 , Animales , Secuencia de Bases/genética , Línea Celular , Chlorocebus aethiops , Citoplasma , Virus Defectuosos/genética , Genoma Viral/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Interferones/genética , ARN Viral/genética , Células Vero , Virión/genética , Virosis/genética , Replicación Viral/genética
5.
PLoS Pathog ; 15(2): e1007561, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30742688

RESUMEN

Paramyxoviruses can establish persistent infections both in vitro and in vivo, some of which lead to chronic disease. However, little is known about the molecular events that contribute to the establishment of persistent infections by RNA viruses. Using parainfluenza virus type 5 (PIV5) as a model we show that phosphorylation of the P protein, which is a key component of the viral RNA polymerase complex, determines whether or not viral transcription and replication becomes repressed at late times after infection. If the virus becomes repressed, persistence is established, but if not, the infected cells die. We found that single amino acid changes at various positions within the P protein switched the infection phenotype from lytic to persistent. Lytic variants replicated to higher titres in mice than persistent variants and caused greater infiltration of immune cells into infected lungs but were cleared more rapidly. We propose that during the acute phases of viral infection in vivo, lytic variants of PIV5 will be selected but, as the adaptive immune response develops, variants in which viral replication can be repressed will be selected, leading to the establishment of prolonged, persistent infections. We suggest that similar selection processes may operate for other RNA viruses.


Asunto(s)
Infecciones por Paramyxoviridae/genética , Paramyxoviridae/genética , Fosfoproteínas/genética , Proteínas Virales/genética , Células A549 , Sustitución de Aminoácidos/genética , Animales , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/patogenicidad , Paramyxoviridae/patogenicidad , Infecciones por Paramyxoviridae/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Fosforilación , ARN Viral , Proteínas Virales/metabolismo , Proteínas Virales/fisiología , Replicación Viral
6.
J Virol ; 93(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31189700

RESUMEN

We have developed a high-throughput sequencing (HTS) workflow for investigating paramyxovirus transcription and replication. We show that sequencing of oligo(dT)-selected polyadenylated mRNAs, without considering the orientation of the RNAs from which they had been generated, cannot accurately be used to analyze the abundance of viral mRNAs because genomic RNA copurifies with the viral mRNAs. The best method is directional sequencing of infected cell RNA that has physically been depleted of ribosomal and mitochondrial RNA followed by bioinformatic steps to differentiate data originating from genomes from viral mRNAs and antigenomes. This approach has the advantage that the abundance of viral mRNA (and antigenomes) and genomes can be analyzed and quantified from the same data. We investigated the kinetics of viral transcription and replication during infection of A549 cells with parainfluenza virus type 2 (PIV2), PIV3, PIV5, or mumps virus and determined the abundances of individual viral mRNAs and readthrough mRNAs. We found that the mRNA abundance gradients differed significantly between all four viruses but that for each virus the pattern remained relatively stable throughout infection. We suggest that rapid degradation of non-poly(A) mRNAs may be primarily responsible for the shape of the mRNA abundance gradient in parainfluenza virus 3, whereas a combination of this factor and disengagement of RNA polymerase at intergenic sequences, particularly those at the NP:P and P:M gene boundaries, may be responsible in the other viruses.IMPORTANCE High-throughput sequencing (HTS) of virus-infected cells can be used to study in great detail the patterns of virus transcription and replication. For paramyxoviruses, and by analogy for all other negative-strand RNA viruses, we show that directional sequencing must be used to distinguish between genomic RNA and mRNA/antigenomic RNA because significant amounts of genomic RNA copurify with poly(A)-selected mRNA. We found that the best method is directional sequencing of total cell RNA, after the physical removal of rRNA (and mitochondrial RNA), because quantitative information on the abundance of both genomic RNA and mRNA/antigenomes can be simultaneously derived. Using this approach, we revealed new details of the kinetics of virus transcription and replication for parainfluenza virus (PIV) type 2, PIV3, PIV5, and mumps virus, as well as on the relative abundance of the individual viral mRNAs.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Infecciones por Paramyxoviridae/virología , Paramyxovirinae/fisiología , ARN Mensajero/genética , Secuenciación Completa del Genoma/métodos , Células A549 , Regulación Viral de la Expresión Génica , Tamaño del Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Paramyxovirinae/clasificación , Paramyxovirinae/patogenicidad , ARN Viral/genética , Especificidad de la Especie , Replicación Viral
7.
J Gen Virol ; 98(9): 2267-2273, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28869005

RESUMEN

NS1 proteins of influenza A and B viruses share limited sequence homology, yet both are potent manipulators of host cell processes, particularly interferon (IFN) induction. Although many cellular partners are reported for A/NS1, only a few (e.g. PKR and ISG15) have been identified for B/NS1. Here, affinity-purification and mass spectrometry were used to expand the known host interactome of B/NS1. We identified 22 human proteins as new putative targets for B/NS1, validating several, including DHX9, ILF3, YBX1 and HNRNPC. Consistent with two RNA-binding domains in B/NS1, many of the identified factors bind RNA and some interact with B/NS1 in an RNA-dependent manner. Functional characterization of several B/NS1 interactors identified SNRNP200 as a potential positive regulator of host IFN responses, while ILF3 exhibited dual roles in both IFN induction and influenza B virus replication. These data provide a resource for future investigations into the mechanisms underpinning host cell modulation by influenza B virus NS1.


Asunto(s)
Virus de la Influenza B/aislamiento & purificación , Gripe Humana/metabolismo , Proteínas no Estructurales Virales/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Humanos , Virus de la Influenza B/genética , Virus de la Influenza B/metabolismo , Gripe Humana/genética , Gripe Humana/virología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas del Factor Nuclear 90/genética , Proteínas del Factor Nuclear 90/metabolismo , Unión Proteica , Proteínas no Estructurales Virales/genética , Proteína 1 de Unión a la Caja Y/genética , Proteína 1 de Unión a la Caja Y/metabolismo
8.
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
9.
J Virol ; 90(5): 2616-27, 2015 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-26699638

RESUMEN

UNLABELLED: Oropouche virus (OROV) is a midge-borne human pathogen with a geographic distribution in South America. OROV was first isolated in 1955, and since then, it has been known to cause recurring outbreaks of a dengue-like illness in the Amazonian regions of Brazil. OROV, however, remains one of the most poorly understood emerging viral zoonoses. Here we describe the successful recovery of infectious OROV entirely from cDNA copies of its genome and generation of OROV mutant viruses lacking either the NSm or the NSs coding region. Characterization of the recombinant viruses carried out in vitro demonstrated that the NSs protein of OROV is an interferon (IFN) antagonist as in other NSs-encoding bunyaviruses. Additionally, we demonstrate the importance of the nine C-terminal amino acids of OROV NSs in IFN antagonistic activity. OROV was also found to be sensitive to IFN-α when cells were pretreated; however, the virus was still capable of replicating at doses as high as 10,000 U/ml of IFN-α, in contrast to the family prototype BUNV. We found that OROV lacking the NSm protein displayed characteristics similar to those of the wild-type virus, suggesting that the NSm protein is dispensable for virus replication in the mammalian and mosquito cell lines that were tested. IMPORTANCE: Oropouche virus (OROV) is a public health threat in Central and South America, where it causes periodic outbreaks of dengue-like illness. In Brazil, OROV is the second most frequent cause of arboviral febrile illness after dengue virus, and with the current rates of urban expansion, more cases of this emerging viral zoonosis could occur. To better understand the molecular biology of OROV, we have successfully rescued the virus along with mutants. We have established that the C terminus of the NSs protein is important in interferon antagonism and that the NSm protein is dispensable for virus replication in cell culture. The tools described in this paper are important in terms of understanding this important yet neglected human pathogen.


Asunto(s)
Interacciones Huésped-Patógeno , Orthobunyavirus/genética , Orthobunyavirus/fisiología , Proteínas no Estructurales Virales/deficiencia , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Animales , Línea Celular , Humanos , Interferones/antagonistas & inhibidores , Orthobunyavirus/inmunología , Genética Inversa
10.
J Virol ; 89(2): 1479-83, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392213

RESUMEN

The effect of abrogating the interferon (IFN) response on human cytomegalovirus (HCMV) replication was investigated using primary human cells engineered to block either the production of or the response to type I IFNs. In IFN-deficient cells, HCMV produced larger plaques and spread and replicated more rapidly than in parental cells. These cells demonstrate the vital role of IFNs in controlling HCMV replication and provide useful tools to investigate the IFN response to HCMV.


Asunto(s)
Citomegalovirus/inmunología , Citomegalovirus/fisiología , Interferón Tipo I/inmunología , Replicación Viral , Células Cultivadas , Humanos , Interferón Tipo I/deficiencia , Ensayo de Placa Viral
11.
Proc Natl Acad Sci U S A ; 110(8): 3053-8, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23391734

RESUMEN

Severe infectious disease in children may be a manifestation of primary immunodeficiency. These genetic disorders represent important experiments of nature with the capacity to elucidate nonredundant mechanisms of human immunity. We hypothesized that a primary defect of innate antiviral immunity was responsible for unusually severe viral illness in two siblings; the proband developed disseminated vaccine strain measles following routine immunization, whereas an infant brother died after a 2-d febrile illness from an unknown viral infection. Patient fibroblasts were indeed abnormally permissive for viral replication in vitro, associated with profound failure of type I IFN signaling and absence of STAT2 protein. Sequencing of genomic DNA and RNA revealed a homozygous mutation in intron 4 of STAT2 that prevented correct splicing in patient cells. Subsequently, other family members were identified with the same genetic lesion. Despite documented infection by known viral pathogens, some of which have been more severe than normal, surviving STAT2-deficient individuals have remained generally healthy, with no obvious defects in their adaptive immunity or developmental abnormalities. These findings imply that type I IFN signaling [through interferon-stimulated gene factor 3 (ISGF3)] is surprisingly not essential for host defense against the majority of common childhood viral infections.


Asunto(s)
Predisposición Genética a la Enfermedad , Factor de Transcripción STAT2/genética , Virosis/genética , Secuencia de Bases , Células Cultivadas , Preescolar , Cartilla de ADN , Femenino , Humanos , Interferón Tipo I/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virosis/metabolismo
12.
J Virol ; 88(8): 3942-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478437

RESUMEN

UNLABELLED: We have examined the requirements for virus transcription and replication and thus the roles of input and progeny genomes in the generation of interferon (IFN)-inducing pathogen-associated molecular patterns (PAMPs) by influenza A viruses using inhibitors of these processes. Using IFN regulatory factor 3 (IRF3) phosphorylation as a marker of activation of the IFN induction cascade that occurs upstream of the IFN-ß promoter, we demonstrate strong activation of the IFN induction cascade in A549 cells infected with a variety of influenza A viruses in the presence of cycloheximide or nucleoprotein (NP) small interfering RNA (siRNA), which inhibits viral protein synthesis and thus complementary ribonucleoprotein (cRNP) and progeny viral RNP (vRNP) synthesis. In contrast, activation of the IFN induction cascade by influenza viruses was very effectively abrogated by treatment with actinomycin D and other transcription inhibitors, which correlated with the inhibition of the synthesis of all viral RNA species. Furthermore, 5,6-dichloro-1-ß-d-ribofuranosyl-benzimidazole, an inhibitor that prevents viral RNA export from the nucleus, was also a potent inhibitor of IRF3 activation; thus, both viral RNA synthesis and nuclear export are required for IFN induction by influenza A viruses. While the exact nature of the viral PAMPs remains to be determined, our data suggest that in this experimental system the major influenza A virus PAMPs are distinct from those of incoming genomes or progeny vRNPs. IMPORTANCE: The host interferon system exerts an extremely potent antiviral response that efficiently restricts virus replication and spread; the interferon response can thus dictate the outcome of a virus infection, and it is therefore important to understand how viruses induce interferon. Both input and progeny genomes have been linked to interferon induction by influenza viruses. However, our experiments in tissue culture cells show that viral RNA synthesis and nuclear export are required to activate this response. Furthermore, the interferon induction cascade is activated under conditions in which the synthesis of progeny genomes is inhibited. Therefore, in tissue culture cells, input and progeny genomes are not the predominant inducers of interferon generated by influenza A viruses; the major viral interferon inducer(s) still remains to be identified.


Asunto(s)
Núcleo Celular/virología , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Gripe Humana/virología , Factor 3 Regulador del Interferón/genética , Interferón beta/metabolismo , ARN Viral/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Gripe Humana/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , ARN Viral/genética , Regulación hacia Arriba
13.
J Virol ; 88(7): 3826-36, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24453358

RESUMEN

UNLABELLED: The strain diversity of a rubulavirus, parainfluenza virus 5 (PIV5), was investigated by comparing 11 newly determined and 6 previously published genome sequences. These sequences represent 15 PIV5 strains, of which 6 were isolated from humans, 1 was from monkeys, 2 were from pigs, and 6 were from dogs. Strain diversity is remarkably low, regardless of host, year of isolation, or geographical origin; a total of 7.8% of nucleotides are variable, and the average pairwise difference between strains is 2.1%. Variation is distributed unevenly across the PIV5 genome, but no convincing evidence of selection for antibody-mediated evasion in hemagglutinin-neuraminidase was found. The finding that some canine and porcine, but not primate, strains are mutated in the SH gene, and do not produce SH, raised the possibility that dogs (or pigs) may not be the natural host of PIV5. The genetic stability of PIV5 was also demonstrated during serial passage of one strain (W3) in Vero cells at a high multiplicity of infection, under conditions of competition with large proportions of defective interfering genomes. A similar observation was made for a strain W3 mutant (PIV5VΔC) lacking V gene function, in which the dominant changes were related to pseudoreversion in this gene. The mutations detected in PIV5VΔC during pseudoreversion, and also those characterizing the SH gene in canine and porcine strains, predominantly involved U-to-C transitions. This suggests an important role for biased hypermutation via an adenosine deaminase, RNA-specific (ADAR)-like activity. IMPORTANCE: Here we report the sequence variation of 16 different isolates of parainfluenza virus 5 (PIV5) that were isolated from a number of species, including humans, monkeys, dogs, and pigs, over 4 decades. Surprisingly, strain diversity was remarkably low, regardless of host, year of isolation, or geographical origin. Variation was distributed unevenly across the PIV5 genome, but no convincing evidence of immune or host selection was found. This overall genome stability of PIV5 was also observed when the virus was grown in the laboratory, and the genome stayed remarkably constant even during the selection of virus mutants. Some of the canine isolates had lost their ability to encode one of the viral proteins, termed SH, suggesting that although PIV5 commonly infects dogs, dogs may not be the natural host for PIV5.


Asunto(s)
Variación Genética , Inestabilidad Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/aislamiento & purificación , Infecciones por Rubulavirus/veterinaria , Infecciones por Rubulavirus/virología , Animales , Humanos , Datos de Secuencia Molecular , Virus de la Parainfluenza 5/fisiología , Pase Seriado , Cultivo de Virus
14.
J Virol ; 88(9): 4632-46, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24574395

RESUMEN

UNLABELLED: Influenza A viruses counteract the cellular innate immune response at several steps, including blocking RIG I-dependent activation of interferon (IFN) transcription, interferon (IFN)-dependent upregulation of IFN-stimulated genes (ISGs), and the activity of various ISG products; the multifunctional NS1 protein is responsible for most of these activities. To determine the importance of other viral genes in the interplay between the virus and the host IFN response, we characterized populations and selected mutants of wild-type viruses selected by passage through non-IFN-responsive cells. We reasoned that, by allowing replication to occur in the absence of the selection pressure exerted by IFN, the virus could mutate at positions that would normally be restricted and could thus find new optimal sequence solutions. Deep sequencing of selected virus populations and individual virus mutants indicated that nonsynonymous mutations occurred at many phylogenetically conserved positions in nearly all virus genes. Most individual mutants selected for further characterization induced IFN and ISGs and were unable to counteract the effects of exogenous IFN, yet only one contained a mutation in NS1. The relevance of these mutations for the virus phenotype was verified by reverse genetics. Of note, several virus mutants expressing intact NS1 proteins exhibited alterations in the M1/M2 proteins and accumulated large amounts of deleted genomic RNAs but nonetheless replicated to high titers. This suggests that the overproduction of IFN inducers by these viruses can override NS1-mediated IFN modulation. Altogether, the results suggest that influenza viruses replicating in IFN-competent cells have tuned their complete genomes to evade the cellular innate immune system and that serial replication in non-IFN-responsive cells allows the virus to relax from these constraints and find a new genome consensus within its sequence space. IMPORTANCE: In natural virus infections, the production of interferons leads to an antiviral state in cells that effectively limits virus replication. The interferon response places considerable selection pressure on viruses, and they have evolved a variety of ways to evade it. Although the influenza virus NS1 protein is a powerful interferon antagonist, the contributions of other viral genes to interferon evasion have not been well characterized. Here, we examined the effects of alleviating the selection pressure exerted by interferon by serially passaging influenza viruses in cells unable to respond to interferon. Viruses that grew to high titers had mutations at many normally conserved positions in nearly all genes and were not restricted to the NS1 gene. Our results demonstrate that influenza viruses have fine-tuned their entire genomes to evade the interferon response, and by removing interferon-mediated constraints, viruses can mutate at genome positions normally restricted by the interferon response.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/inmunología , Interferones/genética , Interferones/metabolismo , Proteínas Virales/inmunología , Análisis Mutacional de ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Virus de la Influenza A/genética , Mutación , Genética Inversa , Selección Genética , Pase Seriado , Proteínas Virales/genética
15.
J Virol ; 87(23): 13053-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24049170

RESUMEN

The induction of an interferon-induced antiviral state is a powerful cellular response against viral infection that limits viral spread. Here, we show that a preexisting antiviral state inhibits the replication of influenza A viruses in human A549 cells by preventing transport of the viral genome to the nucleus and that the interferon-induced MxA protein is necessary but not sufficient for this process. This represents a previously unreported antiviral function of MxA against influenza A virus infection.


Asunto(s)
Citoplasma/virología , Genoma Viral , Subtipo H3N2 del Virus de la Influenza A/genética , Gripe Humana/metabolismo , Gripe Humana/virología , Interferón-alfa/metabolismo , Proteínas de Resistencia a Mixovirus/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Núcleo Celular/virología , Citoplasma/metabolismo , Regulación hacia Abajo , Interacciones Huésped-Patógeno , Humanos , Subtipo H3N2 del Virus de la Influenza A/fisiología , Gripe Humana/genética , Proteínas de Resistencia a Mixovirus/genética
16.
PLoS Pathog ; 7(2): e1001284, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21379343

RESUMEN

Cell-free HIV-1 virions are poor stimulators of type I interferon (IFN) production. We examined here how HIV-infected cells are recognized by plasmacytoid dendritic cells (pDCs) and by other cells. We show that infected lymphocytes are more potent inducers of IFN than virions. There are target cell-type differences in the recognition of infected lymphocytes. In primary pDCs and pDC-like cells, recognition occurs in large part through TLR7, as demonstrated by the use of inhibitors and by TLR7 silencing. Donor cells expressing replication-defective viruses, carrying mutated reverse transcriptase, integrase or nucleocapsid proteins induced IFN production by target cells as potently as wild-type virus. In contrast, Env-deleted or fusion defective HIV-1 mutants were less efficient, suggesting that in addition to TLR7, cytoplasmic cellular sensors may also mediate sensing of infected cells. Furthermore, in a model of TLR7-negative cells, we demonstrate that the IRF3 pathway, through a process requiring access of incoming viral material to the cytoplasm, allows sensing of HIV-infected lymphocytes. Therefore, detection of HIV-infected lymphocytes occurs through both endosomal and cytoplasmic pathways. Characterization of the mechanisms of innate recognition of HIV-infected cells allows a better understanding of the pathogenic and exacerbated immunologic events associated with HIV infection.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Linfocitos/metabolismo , Linfocitos/virología , Western Blotting , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virología , Glicoproteínas/genética , Glicoproteínas/metabolismo , VIH , Infecciones por VIH/metabolismo , Seropositividad para VIH , Células Madre Hematopoyéticas/metabolismo , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón-alfa/metabolismo , Linfocitos/inmunología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Virión/patogenicidad , Replicación Viral
17.
Proc Natl Acad Sci U S A ; 107(5): 1954-9, 2010 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-20133840

RESUMEN

Seasonal epidemics and periodic worldwide pandemics caused by influenza A viruses are of continuous concern. The viral nonstructural (NS1) protein is a multifunctional virulence factor that antagonizes several host innate immune defenses during infection. NS1 also directly stimulates class IA phosphoinositide 3-kinase (PI3K) signaling, an essential cell survival pathway commonly mutated in human cancers. Here, we present a 2.3-A resolution crystal structure of the NS1 effector domain in complex with the inter-SH2 (coiled-coil) domain of p85beta, a regulatory subunit of PI3K. Our data emphasize the remarkable isoform specificity of this interaction, and provide insights into the mechanism by which NS1 activates the PI3K (p85beta:p110) holoenzyme. A model of the NS1:PI3K heterotrimeric complex reveals that NS1 uses the coiled-coil as a structural tether to sterically prevent normal inhibitory contacts between the N-terminal SH2 domain of p85beta and the p110 catalytic subunit. Furthermore, in this model, NS1 makes extensive contacts with the C2/kinase domains of p110, and a small acidic alpha-helix of NS1 sits adjacent to the highly basic activation loop of the enzyme. During infection, a recombinant influenza A virus expressing NS1 with charge-disruption mutations in this acidic alpha-helix is unable to stimulate the production of phosphatidylinositol 3,4,5-trisphosphate or the phosphorylation of Akt. Despite this, the charge-disruption mutations in NS1 do not affect its ability to interact with the p85beta inter-SH2 domain in vitro. Overall, these data suggest that both direct binding of NS1 to p85beta (resulting in repositioning of the N-terminal SH2 domain) and possible NS1:p110 contacts contribute to PI3K activation.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/metabolismo , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Animales , Secuencia de Bases , Dominio Catalítico , Bovinos , Línea Celular , Cristalografía por Rayos X , Cartilla de ADN/genética , Perros , Activación Enzimática , Humanos , Técnicas In Vitro , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Modelos Moleculares , Complejos Multiproteicos , Mutagénesis Sitio-Dirigida , Dominios y Motivos de Interacción de Proteínas , Proteínas no Estructurales Virales/genética , Dominios Homologos src
18.
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
19.
Biosensors (Basel) ; 12(12)2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36551092

RESUMEN

Immunodiagnostics have been widely used in the detection of disease biomarkers. The conventional immunological tests in central laboratories require expensive equipment and, for non-specialists, the tests are technically demanding and time-consuming, which has prevented their use by the public. Thus, point-of-care tests (POCT), such as lateral flow immunoassays, are being, or have been, developed as more convenient and low-cost methods for immunodiagnostics. However, the sensitivity of such tests is often a concern. Here, a fluorescence-linked immunosorbent assay (FLISA) using organic light-emitting diodes (OLEDs) as excitation light sources was investigated as a way forward for the development of compact and sensitive POCTs. Phycoerythrin (PE) was selected as the fluorescent dye, and OLEDs were designed with different emission spectra. The leakage light of different OLEDs for exciting PE was then investigated to reduce the background noise and improve the sensitivity of the system. Finally, as proof-of-principle that OLED-based technology can be successfully further developed for POCT, antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in human serum was detected by OLED-FLISA.


Asunto(s)
COVID-19 , Inmunoadsorbentes , Humanos , SARS-CoV-2 , Fluorescencia , COVID-19/diagnóstico , Anticuerpos Antivirales
20.
Front Immunol ; 13: 888427, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36159783

RESUMEN

Purpose: Janus kinase-1 (JAK1) tyrosine kinase mediates signaling from multiple cytokine receptors, including interferon alpha/beta and gamma (IFN-α/ß and IFN-γ), which are important for viral and mycobacterial protection respectively. We previously reported autosomal recessive (AR) hypomorphic JAK1 mutations in a patient with recurrent atypical mycobacterial infections and relatively minor viral infections. This study tests the impact of partial JAK1 deficiency on cellular responses to IFNs and pathogen control. Methods: We investigated the role of partial JAK1 deficiency using patient cells and cell models generated with lentiviral vectors expressing shRNA. Results: Partial JAK1 deficiency impairs IFN-γ-dependent responses in multiple cell types including THP-1 macrophages, Epstein-Barr Virus (EBV)-transformed B cells and primary dermal fibroblasts. In THP-1 myeloid cells, partial JAK1 deficiency reduced phagosome acidification and apoptosis and resulted in defective control of mycobacterial infection with enhanced intracellular survival. Although both EBV-B cells and primary dermal fibroblasts with partial JAK1 deficiency demonstrate reduced IFN-α responses, control of viral infection was impaired only in patient EBV-B cells and surprisingly intact in patient primary dermal fibroblasts. Conclusion: Our data suggests that partial JAK1 deficiency predominantly affects susceptibility to mycobacterial infection through impact on the IFN-γ responsive pathway in myeloid cells. Susceptibility to viral infections as a result of reduced IFN-α responses is variable depending on cell type. Description of additional patients with inherited JAK1 deficiency will further clarify the spectrum of bacterial and viral susceptibility in this condition. Our results have broader relevance for anticipating infectious complications from the increasing use of selective JAK1 inhibitors.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Infecciones por Mycobacterium , Mycobacterium , Herpesvirus Humano 4/genética , Humanos , Interferón-alfa/farmacología , Interferón beta , Interferón gamma/genética , Janus Quinasa 1/genética , Mycobacterium/genética , Infecciones por Mycobacterium/genética , ARN Interferente Pequeño , Receptores de Citocinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA