Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Genes Dev ; 27(13): 1511-25, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23824541

RESUMEN

Bunyaviruses are an emerging group of medically important viruses, many of which are transmitted from insects to mammals. To identify host factors that impact infection, we performed a genome-wide RNAi screen in Drosophila and identified 131 genes that impacted infection of the mosquito-transmitted bunyavirus Rift Valley fever virus (RVFV). Dcp2, the catalytic component of the mRNA decapping machinery, and two decapping activators, DDX6 and LSM7, were antiviral against disparate bunyaviruses in both insect cells and adult flies. Bunyaviruses 5' cap their mRNAs by "cap-snatching" the 5' ends of poorly defined host mRNAs. We found that RVFV cap-snatches the 5' ends of Dcp2 targeted mRNAs, including cell cycle-related genes. Loss of Dcp2 allows increased viral transcription without impacting viral mRNA stability, while ectopic expression of Dcp2 impedes viral transcription. Furthermore, arresting cells in late S/early G2 led to increased Dcp2 mRNA targets and increased RVFV replication. Therefore, RVFV competes for the Dcp2-accessible mRNA pool, which is dynamically regulated and can present a bottleneck for viral replication.


Asunto(s)
Drosophila melanogaster/genética , Drosophila melanogaster/virología , Genoma de los Insectos/genética , Orthobunyavirus/fisiología , Caperuzas de ARN/metabolismo , Factores de Transcripción , Replicación Viral/fisiología , Aedes/virología , Animales , Puntos de Control del Ciclo Celular , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Caperuzas de ARN/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Proc Natl Acad Sci U S A ; 114(14): 3720-3725, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28320935

RESUMEN

Viruses and their hosts can reach balanced states of evolution ensuring mutual survival, which makes it difficult to appreciate the underlying dynamics. To uncover hidden interactions, virus mutants that have lost defense genes may be used. Deletion of the gene that encodes serine protease inhibitor 1 (SPI-1) of rabbitpox virus and vaccinia virus, two closely related orthopoxviruses, prevents their efficient replication in human cells, whereas certain other mammalian cells remain fully permissive. Our high-throughput genome-wide siRNA screen identified host factors that prevent reproduction and spread of the mutant viruses in human cells. More than 20,000 genes were interrogated with individual siRNAs and those that prominently increased replication of the SPI-1 deletion mutant were subjected to a secondary screen. The top hits based on the combined data-replication factor C3 (RFC3), FAM111A, and interferon regulatory factor 2 (IRF2)-were confirmed by custom assays. The siRNAs to RFC1, RFC2, RFC4, and RFC5 mRNAs also enhanced spread of the mutant virus, strengthening the biological significance of the RFC complex as a host restriction factor for poxviruses. Whereas association with proliferating cell nuclear antigen and participation in processive genome replication are common features of FAM111A and RFC, IRF2 is a transcriptional regulator. Microarray analysis, quantitative RT-PCR, and immunoblotting revealed that IRF2 regulated the basal level expression of FAM111A, suggesting that the enhancing effect of depleting IRF2 on replication of the SPI-1 mutant was indirect. Thus, the viral SPI-1 protein and the host IRF2, FAM111A, and RFC complex likely form an interaction network that influences the ability of poxviruses to replicate in human cells.


Asunto(s)
Factor 2 Regulador del Interferón/metabolismo , Orthopoxvirus/fisiología , Receptores Virales/metabolismo , Proteína de Replicación C/metabolismo , Serpinas/genética , Células A549 , Humanos , Análisis por Micromatrices , Mutación , Orthopoxvirus/enzimología , Orthopoxvirus/genética , Infecciones por Poxviridae/metabolismo , Infecciones por Poxviridae/virología , Proteínas Virales/genética , Replicación Viral
3.
J Allergy Clin Immunol ; 143(3): 1176-1182.e5, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30217468

RESUMEN

BACKGROUND: Virus-associated febrile lower respiratory tract infections (fLRIs) during infancy have been identified as risk factors for persistent wheeze development. We hypothesized that variations in innate immune defense capacity during this period, as exemplified by production of type 1 and 3 interferons (T1/3IFNs), might be an underlying determinant of risk. OBJECTIVE: We sought to investigate relationships between postnatal development of innate interferon response capacity and susceptibility to early infections and persistent wheeze. METHODS: We studied a subset of subjects from a birth cohort at high risk for asthma/allergy and determined the capacity of cord blood cells (n = 151) to produce any of a panel of 17 T1/3IFNs in response to the viral mimic polyinosinic-polycytidylic acid using a sensitive PCR assay. We investigated relationships between neonatal interferon responses and lower respiratory tract infection history during infancy, wheezing history to 5 age years, and ensuing maturation of innate immune capacity by age 4 years (n = 160) and 10 years (n = 125). RESULTS: Although cohort subjects produced an average of 2.6 ± 0.3 of the 17 innate interferons tested at birth, 24% showed no T1/3IFN production. This nonproducer subgroup showed increased risk for infant fLRIs (odds ratio, 2.62; 95% CI, 1.14-6.06; P = .024) and persistent wheeze (odds ratio, 4.24; 95% CI, 1.60-11.24; P = .004) at age 5 years relative to those producing 1 or more T1/3IFNs, whereas risk for infant wheezy lower respiratory tract infections or "transient early wheeze" was unaffected. Moreover, infants who experienced fLRIs subsequently demonstrated accelerated development of T1/3IFN response capacity between 1 and 4 years of age. CONCLUSIONS: T1/3IFN response capacity appears strongly developmentally constrained at birth. Infants in whom this negative regulation is strongest manifest increased risk for severe respiratory tract infections during infancy and subsequent persistent wheeze.


Asunto(s)
Asma/inmunología , Interferones/inmunología , Ruidos Respiratorios/inmunología , Infecciones del Sistema Respiratorio/inmunología , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Leucocitos Mononucleares/inmunología , Masculino , Factores de Riesgo
4.
J Virol ; 92(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29343579

RESUMEN

Genome uncoating is essential for replication of most viruses. For poxviruses, the process is divided into two stages: removal of the envelope, allowing early gene expression, and breaching of the core wall, allowing DNA release, replication, and late gene expression. Subsequent studies showed that the host proteasome and the viral D5 protein, which has an essential role in DNA replication, are required for vaccinia virus (VACV) genome uncoating. In a search for additional VACV uncoating proteins, we noted a report that described a defect in DNA replication and late expression when the gene encoding a 68-kDa ankyrin repeat/F-box protein (68k-ank), associated with the cellular SCF (Skp1, cullin1, F-box-containing complex) ubiquitin ligase complex, was deleted from the attenuated modified vaccinia virus Ankara (MVA). Here we showed that the 68k-ank deletion mutant exhibited diminished genome uncoating, formation of DNA prereplication sites, and degradation of viral cores as well as an additional, independent defect in DNA synthesis. Deletion of the 68k-ank homolog of VACV strain WR, however, was without effect, suggesting the existence of compensating genes. By inserting VACV genes into an MVA 68k-ank deletion mutant, we discovered that M2, a member of the poxvirus immune evasion (PIE) domain superfamily and a regulator of NF-κB, and C5, a member of the BTB/Kelch superfamily associated with cullin-3-based ligase complexes, independently rescued the 68k-ank deletion phenotype. Thus, poxvirus uncoating and DNA replication are intertwined processes involving at least three viral proteins with mutually redundant functions in addition to D5.IMPORTANCE Poxviruses comprise a family of large DNA viruses that infect vertebrates and invertebrates and cause diseases of medical and zoological importance. Poxviruses, unlike most other DNA viruses, replicate in the cytoplasm, and their large genomes usually encode 200 or more proteins with diverse functions. About 90 genes may be essential for chordopoxvirus replication based either on their conservation or individual gene deletion studies. However, this number may underestimate the true number of essential functions because of redundancy. Here we show that any one of three seemingly unrelated and individually nonessential proteins is required for the incompletely understood processes of genome uncoating and DNA replication, an example of synthetic lethality. Thus, poxviruses appear to have a complex genetic interaction network that has not been fully appreciated and which will require multifactor deletion screens to assess.


Asunto(s)
Proteínas Cullin/inmunología , Replicación del ADN/inmunología , ADN Viral/inmunología , Genoma Viral/inmunología , Evasión Inmune , Proteínas Quinasas Asociadas a Fase-S/inmunología , Virus Vaccinia/inmunología , Proteínas Cullin/genética , ADN Viral/genética , Células HeLa , Humanos , Proteínas Quinasas Asociadas a Fase-S/genética , Virus Vaccinia/genética
5.
J Virol ; 92(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29769339

RESUMEN

Respiratory syncytial virus (RSV) infects small foci of respiratory epithelial cells via infected droplets. Infection induces expression of type I and III interferons (IFNs) and proinflammatory cytokines, the balance of which may restrict viral replication and affect disease severity. We explored this balance by infecting two respiratory epithelial cell lines with low doses of recombinant RSV expressing green fluorescent protein (rgRSV). A549 cells were highly permissive, whereas BEAS-2B cells restricted infection to individual cells or small foci. After infection, A549 cells expressed higher levels of IFN-ß-, IFN-λ-, and NF-κB-inducible proinflammatory cytokines. In contrast, BEAS-2B cells expressed higher levels of antiviral interferon-stimulated genes, pattern recognition receptors, and other signaling intermediaries constitutively and after infection. Transcriptome analysis revealed that constitutive expression of antiviral and proinflammatory genes predicted responses by each cell line. These two cell lines provide a model for elucidating critical mediators of local control of viral infection in respiratory epithelial cells.IMPORTANCE Airway epithelium is both the primary target of and the first defense against respiratory syncytial virus (RSV). Whether RSV replicates and spreads to adjacent epithelial cells depends on the quality of their innate immune responses. A549 and BEAS-2B are alveolar and bronchial epithelial cell lines, respectively, that are often used to study RSV infection. We show that A549 cells are permissive to RSV infection and express genes characteristic of a proinflammatory response. In contrast, BEAS-2B cells restrict infection and express genes characteristic of an antiviral response associated with expression of type I and III interferons. Transcriptome analysis of constitutive gene expression revealed patterns that may predict the response of each cell line to infection. This study suggests that restrictive and permissive cell lines may provide a model for identifying critical mediators of local control of infection and stresses the importance of the constitutive antiviral state for the response to viral challenge.


Asunto(s)
Citocinas/inmunología , Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Mucosa Respiratoria/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Células A549 , Células Epiteliales/virología , Humanos , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/patología
6.
Proc Natl Acad Sci U S A ; 112(25): 7797-802, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-26056282

RESUMEN

Viruses must gain entry into cells to establish infection. In general, viruses enter either at the plasma membrane or from intracellular endosomal compartments. Viruses that use endosomal pathways are dependent on the cellular factors that control this process; however, these genes have proven to be essential for endogenous cargo uptake, and thus are of limited value for therapeutic intervention. The identification of genes that are selectively required for viral uptake would make appealing drug targets, as their inhibition would block an early step in the life cycle of diverse viruses. At this time, we lack pan-antiviral therapeutics, in part because of our lack of knowledge of such cellular factors. RNAi screening has begun to reveal previously unknown genes that play roles in viral infection. We identified dRNASEK in two genome-wide RNAi screens performed in Drosophila cells against West Nile and Rift Valley Fever viruses. Here we found that ribonuclease kappa (RNASEK) is essential for the infection of human cells by divergent and unrelated positive- and negative-strand-enveloped viruses from the Flaviviridae, Togaviridae, Bunyaviridae, and Orthomyxoviridae families that all enter cells from endosomal compartments. In contrast, RNASEK was dispensable for viruses, including parainfluenza virus 5 and Coxsackie B virus, that enter at the plasma membrane. RNASEK is dispensable for attachment but is required for uptake of these acid-dependent viruses. Furthermore, this requirement appears specific, as general endocytic uptake of transferrin is unaffected in RNASEK-depleted cells. Therefore, RNASEK is a potential host cell Achilles' heel for viral infection.


Asunto(s)
Endocitosis , Fusión de Membrana , Ribonucleasas/metabolismo , Animales , Drosophila , Concentración de Iones de Hidrógeno , Virus de la Fiebre del Valle del Rift/fisiología , Virus del Nilo Occidental/fisiología
7.
Proc Natl Acad Sci U S A ; 112(22): E2920-9, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26038567

RESUMEN

The mosquito-transmitted bunyavirus, Rift Valley fever virus (RVFV), is a highly successful pathogen for which there are no vaccines or therapeutics. Translational arrest is a common antiviral strategy used by hosts. In response, RVFV inhibits two well-known antiviral pathways that attenuate translation during infection, PKR and type I IFN signaling. Despite this, translational arrest occurs during RVFV infection by unknown mechanisms. Here, we find that RVFV infection triggers the decay of core translation machinery mRNAs that possess a 5'-terminal oligopyrimidine (5'-TOP) motif in their 5'-UTR, including mRNAs encoding ribosomal proteins, which leads to a decrease in overall ribosomal protein levels. We find that the RNA decapping enzyme NUDT16 selectively degrades 5'-TOP mRNAs during RVFV infection and this decay is triggered in response to mTOR attenuation via the translational repressor 4EBP1/2 axis. Translational arrest of 5'-TOPs via 4EBP1/2 restricts RVFV replication, and this increased RNA decay results in the loss of visible RNA granules, including P bodies and stress granules. Because RVFV cap-snatches in RNA granules, the increased level of 5'-TOP mRNAs in this compartment leads to snatching of these targets, which are translationally suppressed during infection. Therefore, translation of RVFV mRNAs is compromised by multiple mechanisms during infection. Together, these data present a previously unknown mechanism for translational shutdown in response to viral infection and identify mTOR attenuation as a potential therapeutic avenue against bunyaviral infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fosfoproteínas/metabolismo , Biosíntesis de Proteínas/fisiología , Pirofosfatasas/metabolismo , Secuencia de Oligopirimidina en la Región 5' Terminal del ARN/fisiología , Fiebre del Valle del Rift/metabolismo , Virus de la Fiebre del Valle del Rift/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Ciclo Celular , Línea Celular , Cartilla de ADN/genética , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Modelos Lineales , Luciferasas , Secuencia de Oligopirimidina en la Región 5' Terminal del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Proc Natl Acad Sci U S A ; 111(37): E3890-9, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25197089

RESUMEN

In response to infection, the innate immune system rapidly activates an elaborate and tightly orchestrated gene expression program to induce critical antimicrobial genes. While many key players in this program have been identified in disparate biological systems, it is clear that there are additional uncharacterized mechanisms at play. Our previous studies revealed that a rapidly-induced antiviral gene expression program is active against disparate human arthropod-borne viruses in Drosophila. Moreover, one-half of this program is regulated at the level of transcriptional pausing. Here we found that Nup98, a virus-induced gene, was antiviral against a panel of viruses both in cells and adult flies since its depletion significantly enhanced viral infection. Mechanistically, we found that Nup98 promotes antiviral gene expression in Drosophila at the level of transcription. Expression profiling revealed that the virus-induced activation of 36 genes was abrogated upon loss of Nup98; and we found that a subset of these Nup98-dependent genes were antiviral. These Nup98-dependent virus-induced genes are Cdk9-dependent and translation-independent suggesting that these are rapidly induced primary response genes. Biochemically, we demonstrate that Nup98 is directly bound to the promoters of virus-induced genes, and that it promotes occupancy of the initiating form of RNA polymerase II at these promoters, which are rapidly induced on viral infection to restrict human arboviruses in insects.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/virología , Regulación de la Expresión Génica , Proteínas de Complejo Poro Nuclear/metabolismo , Infecciones por Virus ARN/genética , Infecciones por Virus ARN/virología , Virus ARN/fisiología , Envejecimiento/patología , Animales , Núcleo Celular/metabolismo , Genes de Insecto , Humanos , Poro Nuclear/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Transporte de Proteínas , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Virus Sindbis/fisiología
9.
Methods ; 91: 75-81, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26164699

RESUMEN

Systematic and comprehensive analysis of host cell proteins involved in virus infection has been difficult in large part due to the lack of robust unbiased methods for their identification. Recent technological breakthroughs allowing development of cell-based genetic screens have greatly facilitated our understanding of virus-host interactions. These include instrumentation for processing in microtiter plates (e.g., 384 well), coupled with sensitive readers and off-the-shelf analysis and informatics pipelines. Because viruses are a significant threat to human health, a better understanding of the cellular factors that impact infection would pave the way for the development of new therapeutics. Here we describe the development and implementation of a genome-wide siRNA screen against a virus using human cells.


Asunto(s)
Genes Virales , Interferencia de ARN , Virus/genética , Genoma Viral , Humanos , Análisis de Secuencia de ADN
10.
J Virol ; 88(6): 3103-13, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371060

RESUMEN

UNLABELLED: In a genome-wide small interfering RNA (siRNA) screen, we recently identified the interferon (IFN)-inducible protein 35 (IFI35; also known as IFP35) as a factor required for vesicular stomatitis virus (VSV) infection. Studies reported here were conducted to further understand the role and requirement of IFI35 in VSV infection. Consistent with the siRNA screening data, we found that depletion of IFI35 led to reduced VSV replication at the level of viral gene expression. Although no direct interaction of IFI35 with the viral replication machinery was observed, we found that IFI35 negatively regulated the host innate immune response and rescued poly(I·C)-induced inhibition of VSV replication. Promoter-driven reporter gene assays demonstrated that IFI35 overexpression suppressed the activation of IFN-ß and ISG56 promoters, whereas its depletion had the opposite effect. Further investigation revealed that IFI35 specifically interacted with retinoic acid-inducible gene I (RIG-I) and negatively regulated its activation through mechanisms that included (i) suppression of dephosphorylation (activation) of RIG-I and (ii) proteasome-mediated degradation of RIG-I via K48-linked ubiquitination. Overall, the results presented here suggest a novel role for IFI35 in negative regulation of RIG-I-mediated antiviral signaling, which will have implications for diseases associated with excessive immune signaling. IMPORTANCE: Mammalian cells employ a variety of mechanisms, including production of interferons (IFNs), to counteract invading pathogens. In this study, we identified a novel role for a cellular protein, IFN-inducible protein 35 (IFP35/IFI35), in negatively regulating the host IFN response during vesicular stomatitis virus (VSV) infection. Specifically, we found that IFI35 inhibited activation of the RNA sensor, the retinoic acid-inducible gene I (RIG-I), leading to inhibition of IFN production and thus resulting in better replication of VSV. The identification of a cellular factor that attenuates the IFN response will have implications toward understanding inflammatory diseases in humans that have been found to be associated with defects in the regulation of host IFN production, such as systemic lupus erythematosus and psoriasis.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , Regulación hacia Abajo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Unión Proteica , Receptores Inmunológicos , Estomatitis Vesicular/genética , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/genética
11.
J Virol ; 87(1): 372-83, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23077311

RESUMEN

Previous studies from our laboratory revealed that cellular poly(C) binding protein 2 (PCBP2) downregulates vesicular stomatitis virus (VSV) gene expression. We show here that VSV infection induces the formation of granular structures in the cytoplasm containing cellular RNA-binding proteins, including PCBP2, T-cell-restricted intracellular antigen 1 (TIA1), and TIA1-related protein (TIAR). Depletion of TIA1 via small interfering RNAs (siRNAs), but not depletion of TIAR, results in enhanced VSV growth and gene expression. The VSV-induced granules appear to be similar to the stress granules (SGs) generated in cells triggered by heat shock or oxidative stress but do not contain some of the bona fide SG markers, such as eukaryotic initiation factor 3 (eIF3) or eIF4A, or the processing body (PB) markers, such as mRNA-decapping enzyme 1A (DCP1a), and thus may not represent canonical SGs or PBs. Our results revealed that the VSV-induced granules, called SG-like structures here, contain the viral replicative proteins and RNAs. The formation and maintenance of the SG-like structures required viral replication and ongoing protein synthesis, but an intact cytoskeletal network was not necessary. These results suggest that cells respond to VSV infection by aggregating the antiviral proteins, such as PCBP2 and TIA1, to form SG-like structures. The functional significance of these SG-like structures in VSV-infected cells is currently under investigation.


Asunto(s)
Gránulos Citoplasmáticos/química , Proteínas de Unión a Poli(A)/análisis , Proteínas de Unión al ARN/análisis , Vesiculovirus/patogenicidad , Línea Celular , Silenciador del Gen , Humanos , Proteínas de Unión a Poli(A)/genética , ARN Viral/análisis , Proteínas de Unión al ARN/genética , Antígeno Intracelular 1 de las Células T , Vesiculovirus/crecimiento & desarrollo , Proteínas Virales/análisis
12.
Proc Natl Acad Sci U S A ; 108(47): 19036-41, 2011 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-22065774

RESUMEN

Negative-strand (NS) RNA viruses comprise many pathogens that cause serious diseases in humans and animals. Despite their clinical importance, little is known about the host factors required for their infection. Using vesicular stomatitis virus (VSV), a prototypic NS RNA virus in the family Rhabdoviridae, we conducted a human genome-wide siRNA screen and identified 72 host genes required for viral infection. Many of these identified genes were also required for infection by two other NS RNA viruses, the lymphocytic choriomeningitis virus of the Arenaviridae family and human parainfluenza virus type 3 of the Paramyxoviridae family. Genes affecting different stages of VSV infection, such as entry/uncoating, gene expression, and assembly/release, were identified. Depletion of the proteins of the coatomer complex I or its upstream effectors ARF1 or GBF1 led to detection of reduced levels of VSV RNA. Coatomer complex I was also required for infection of lymphocytic choriomeningitis virus and human parainfluenza virus type 3. These results highlight the evolutionarily conserved requirements for gene expression of diverse families of NS RNA viruses and demonstrate the involvement of host cell secretory pathway in the process.


Asunto(s)
Factores Celulares Derivados del Huésped/genética , Vías Secretoras/genética , Virus de la Estomatitis Vesicular Indiana/fisiología , Integración Viral/genética , Animales , Línea Celular , Perros , Electroforesis en Gel de Poliacrilamida , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/fisiología , Virus de la Parainfluenza 3 Humana/genética , Virus de la Parainfluenza 3 Humana/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus de la Estomatitis Vesicular Indiana/genética
13.
Cureus ; 16(5): e59702, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38841050

RESUMEN

We report an extremely rare case of long-standing (> six months) minimal pericardial effusion attributed to dermatomyositis. The patient was inadvertently administered antitubercular drug therapy for three months after which the patient developed significant weight loss, extreme anorexia, nausea, and vomiting refractory to conventional management. The key message in the manuscript is that even indolent dermatomyositis can present solely as an unexplained pericardial effusion in an individual.

14.
J Virol ; 85(18): 9459-71, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21752917

RESUMEN

Immunoprecipitation and subsequent mass spectrometry analysis of the cellular proteins from cells expressing the vesicular stomatitis virus (VSV) P protein identified the poly(C) binding protein 2 (PCBP2) as one of the P protein-interacting proteins. To investigate the role of PCBP2 in the viral life cycle, we examined the effects of depletion or overexpression of this protein on VSV growth. Small interfering RNA-mediated silencing of PCBP2 promoted VSV replication. Conversely, overexpression of PCBP2 in transfected cells suppressed VSV growth. Further studies revealed that PCBP2 negatively regulates overall viral mRNA accumulation and subsequent genome replication. Coimmunoprecipitation and immunofluorescence microscopic studies showed that PCBP2 interacts and colocalizes with VSV P protein in virus-infected cells. The P-PCBP2 interaction did not result in reduced levels of protein complex formation with the viral N and L proteins, nor did it induce degradation of the P protein. In addition, PCBP1, another member of the poly(C) binding protein family with homology to PCBP2, was also found to interact with the P protein and inhibit the viral mRNA synthesis at the level of primary transcription without affecting secondary transcription or genome replication. The inhibitory effects of PCBP1 on VSV replication were less pronounced than those of PCBP2. Overall, the results presented here suggest that cellular PCBP2 and PCBP1 antagonize VSV growth by affecting viral gene expression and highlight the importance of these two cellular proteins in restricting virus infections.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas/inmunología , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Interacciones Huésped-Patógeno , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/metabolismo , Vesiculovirus/fisiología , Replicación Viral , Línea Celular , Proteínas de Unión al ADN , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Inmunoprecipitación , Microscopía Confocal , Fosfoproteínas/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas de Unión al ARN/genética , Proteínas Estructurales Virales/metabolismo
15.
J Virol ; 84(9): 4826-31, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181705

RESUMEN

We show here that replication of defective interfering (DI) particle RNA in HEK293 cells stably expressing vesicular stomatitis virus (VSV) replication proteins potently activates interferon (IFN) and IFN signaling pathways through upregulation of IFN-beta promoter, IFN-stimulated response element (ISRE) promoter, and NF-kappaB promoter activities. Replication of DI particle RNA, not mere expression of the viral replication proteins, was found to be critical for induction of IFN and IFN signaling. The stable cells supporting replication of DI RNA described in this report will be useful in further examining the innate immune signaling pathways and the host cell functions in viral genome replication.


Asunto(s)
Virus Defectuosos/genética , Virus Defectuosos/inmunología , Interferones/biosíntesis , ARN Viral/metabolismo , Transducción de Señal , Vesiculovirus/genética , Vesiculovirus/inmunología , Línea Celular , Humanos , Interferón beta/biosíntesis , FN-kappa B/biosíntesis , Regiones Promotoras Genéticas , Activación Transcripcional , Regulación hacia Arriba
16.
J Virol ; 83(6): 2611-22, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19153240

RESUMEN

A recombinant vesicular stomatitis virus (VSV-PeGFP-M-MmRFP) encoding enhanced green fluorescent protein fused in frame with P (PeGFP) in place of P and a fusion matrix protein (monomeric red fluorescent protein fused in frame at the carboxy terminus of M [MmRFP]) at the G-L gene junction, in addition to wild-type (wt) M protein in its normal location, was recovered, but the MmRFP was not incorporated into the virions. Subsequently, we generated recombinant viruses (VSV-PeGFP-DeltaM-Mtc and VSV-DeltaM-Mtc) encoding M protein with a carboxy-terminal tetracysteine tag (Mtc) in place of the M protein. These recombinant viruses incorporated Mtc at levels similar to M in wt VSV, demonstrating recovery of infectious rhabdoviruses encoding and incorporating a tagged M protein. Virions released from cells infected with VSV-PeGFP-DeltaM-Mtc and labeled with the biarsenical red dye (ReAsH) were dually fluorescent, fluorescing green due to incorporation of PeGFP in the nucleocapsids and red due to incorporation of ReAsH-labeled Mtc in the viral envelope. Transport and subsequent association of M protein with the plasma membrane were shown to be independent of microtubules. Sequential labeling of VSV-DeltaM-Mtc-infected cells with the biarsenical dyes ReAsH and FlAsH (green) revealed that newly synthesized M protein reaches the plasma membrane in less than 30 min and continues to accumulate there for up to 2 1/2 hours. Using dually fluorescent VSV, we determined that following adsorption at the plasma membrane, the time taken by one-half of the virus particles to enter cells and to uncoat their nucleocapsids in the cytoplasm is approximately 28 min.


Asunto(s)
Arsénico/metabolismo , Coloración y Etiquetado/métodos , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas de la Matriz Viral/metabolismo , Internalización del Virus , Animales , Línea Celular , Membrana Celular/química , Color , Cricetinae , Fluorescencia , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Proteínas de la Matriz Viral/genética
17.
J Virol ; 83(11): 5525-34, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19321605

RESUMEN

The nucleocapsid protein (N) of vesicular stomatitis virus and other rhabdoviruses plays a central role in the assembly and template functions of the viral N-RNA complex. The crystal structure of the viral N-RNA complex suggests that the central region of the N protein interacts with the viral RNA. Sequence alignment of rhabdovirus N proteins revealed several highly conserved regions, one of which spanned residues 282 to 291 (GLSSKSPYSS) in the central region of the molecule. Alanine-scanning mutagenesis of this region suggested that replacement of the tyrosine residue at position 289 (Y289) with alanine resulted in an N-RNA template that is nonfunctional in viral genome replication and transcription. To establish the molecular basis of this defect, our further studies revealed that the Y289A mutant maintained its interaction with other N protein molecules but that its interactions with the P protein or with the viral RNA were defective. Replacement of Y289 with other aromatic, polar, or large amino acids indicated that the hydrophobic and aromatic nature of this position in the N protein is functionally important and that a larger aromatic residue is less favorable. Interestingly, we have observed that several single-amino-acid substitutions in this highly conserved region of the molecule rendered the nucleocapsid template nonfunctional in transcription without adversely affecting the replication functions. These results suggest that the structure of the N protein and the resulting N-RNA complex, in part, regulate the viral template functions in transcription and replication.


Asunto(s)
Proteínas de la Nucleocápside/metabolismo , Nucleocápside/metabolismo , Vesiculovirus/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Línea Celular , Secuencia Conservada , Cricetinae , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Nucleocápside/genética , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/genética , Fenotipo , Estructura Terciaria de Proteína , ARN Viral/genética , Alineación de Secuencia , Transcripción Genética/genética , Vesiculovirus/genética , Replicación Viral
18.
Res Vet Sci ; 86(1): 36-42, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18572211

RESUMEN

Gastroenteritis of viral origin has emerged as a major cause of morbidity and mortality in dogs during the last two decades. Amongst the viral etiologies responsible for gastroenteritis in dogs, canine parvovirus (CPV) is considered as the most pathogenic. The disease is characterized by hemorrhagic enteritis, bloody diarrhoea and myocarditis in young pups. The present study was carried out to examine alterations in oxidative stress indices in the erythrocytes from dogs suffering from gastroenteritis with or without canine parvoviral infection as confirmed by CPV-DNA amplification from faeces using specific primers for CPV-2 as well as CPV-2a and CPV-2b variants by polymerase chain reaction (PCR). The present investigation utilized clinical cases of dogs with signs of acute diarrhea (n=56), and 14 more apparently healthy dogs of similar age group. Erythrocytic oxidative stress indices such as lipid peroxides level and antioxidant enzymes like superoxide dismutase and catalase activity, and blood micro-mineral (iron, copper, cobalt and zinc) status were analyzed in each dog (n=70). The acute cases of gastroenteritis in dogs were associated with altered erythrocytic lipid peroxidation as evident by estimation of malonaldehyde (MDA) concentration. The activities of antioxidant enzymes catalase and superoxide dismutase, the first line of antioxidant defense against damaging effects of free radicals, were also altered. The alterations in oxidative stress indices were more pronounced in cases with involvement of canine parvovirus as compared to parvo-negative cases. Our results also revealed decreased blood zinc level in diarrhoea in dogs irrespective of involvement of canine parvovirus.


Asunto(s)
Enfermedades de los Perros/virología , Gastroenteritis/veterinaria , Estrés Oxidativo , Infecciones por Parvoviridae/veterinaria , Parvovirus Canino/aislamiento & purificación , Animales , Catalasa/sangre , ADN Viral/química , ADN Viral/genética , Enfermedades de los Perros/sangre , Perros , Eritrocitos/metabolismo , Eritrocitos/virología , Heces/virología , Gastroenteritis/sangre , Gastroenteritis/virología , Malondialdehído/sangre , Metales Pesados/sangre , Infecciones por Parvoviridae/sangre , Infecciones por Parvoviridae/virología , Parvovirus Canino/genética , Reacción en Cadena de la Polimerasa/veterinaria , Superóxido Dismutasa/sangre
19.
Front Immunol ; 10: 1019, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31156620

RESUMEN

Viral defense at mucosal sites depends on interferons (IFN) and IFN stimulated genes (ISGs), either of which may be constitutively expressed to maintain an "antiviral state" (AVS). However, the mechanisms that govern the AVS are poorly defined. Using a BEAS-2B respiratory epithelial cell line deficient in IRF1, we demonstrate higher susceptibility to infection with vesicular stomatitis virus (VSV) and influenza virus. IRF1-mediated restriction of VSV is IFN-independent, as blockade of types I and III IFNs and JAK-STAT signaling before infection did not affect VSV infection of either parent or IRF1 KO cells. Transcriptome analysis revealed that IRF1 regulates constitutive expression of ~300 genes, including antiviral ISGs: OAS2, BST2, and RNASEL and knockdown of any of these IRF1-dependent genes increased VSV infection. Additionally, IRF1 enhances rapid expression of IFNß and IFNλ after stimulation with poly I:C and also regulates ISG expression. Mechanistically, IRF1 enhances recruitment of BRD4 to promotor-enhancer regions of ISGs for rapid expression and maintains levels of histone H3K4me1 for optimal constitutive expression. Finally, IRF1 also regulates constitutive expression of TLR2 and TLR3 and promotes signaling through these pattern recognition receptors (PRR). These data reveal multiple roles for IRF1 toward effective anti-viral responses by maintaining IFN-independent constitutive expression of anti-viral ISGs and supporting early IFN-dependent responses to PRR stimulation.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/genética , Antígenos CD/genética , Endorribonucleasas/genética , Gripe Humana/inmunología , Factor 1 Regulador del Interferón/genética , Orthomyxoviridae/inmunología , Infecciones por Rhabdoviridae/inmunología , Vesiculovirus/inmunología , Células A549 , Células Epiteliales/metabolismo , Proteínas Ligadas a GPI/genética , Regulación de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Humanos , Gripe Humana/virología , Factor 1 Regulador del Interferón/metabolismo , Interferones/metabolismo , Mucosa Respiratoria/citología , Infecciones por Rhabdoviridae/virología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo , Transfección , Replicación Viral/genética , Replicación Viral/inmunología
20.
J Interferon Cytokine Res ; 39(5): 283-292, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30920934

RESUMEN

Type I interferons (IFNs) signal by forming a high affinity IFN-IFNAR2 dimer, which subsequently recruits IFNAR1 to form a ternary complex that initiates JAK/STAT signaling. Among the 12 IFNα subtypes, IFNα1 has a uniquely low affinity for IFNAR2 (<100 × of the other IFNα subtypes) and commensurately weak antiviral activity, suggesting an undefined function distinct from suppression of viral infections. Also unique in IFNα1 is substitution of a serine for phenylalanine at position 27, a contact point that stabilizes the IFNα:IFNAR2 hydrophobic interface. To determine whether IFNα1-S27 contributes to the low affinity for IFNAR2, we created an IFNα1 mutein, IFNα1-S27F, and compared it to wild-type IFNα1 and IFNα2. Substitution of phenylalanine for serine increased affinity for IFNAR2 ∼4-fold and commensurately enhanced activation of STAT1, STAT3, and STAT5, transcription of a subset of interferon stimulated genes, and restriction of vesicular stomatitis virus infection in vitro. Structural modeling suggests that S27 of IFNα1 disrupts the IFNα:IFNAR2 hydrophobic interface that is otherwise stabilized by F27 and that replacing S27 with phenylalanine partially restores the hydrophobic surface. Disruption of the hydrophobic IFNα:IFNAR2 interface by the unique S27 of IFN α1 contributes to its low affinity and weak antiviral activity.


Asunto(s)
Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Serina , Vesiculovirus/inmunología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Interferón-alfa/química , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Serina/genética , Serina/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA