Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 30(2): 589-604, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36624264

RESUMEN

Kinase signaling in the tiered activation of inflammasomes and associated pyroptosis is a prime therapeutic target for inflammatory diseases. While MAPKs subsume pivotal roles during inflammasome priming, specifically the MAP3K7/JNK1/NLRP3 licensing axis, their involvement in successive steps of inflammasome activation is poorly defined. Using live-cell MAPK biosensors to focus on the inflammasome triggering event allowed us to identify a subsequent process of biphasic JNK activation. We find that this biphasic post-trigger JNK signaling initially facilitates the mitochondrial reactive oxygen species generation needed to support core inflammasome formation, then supports the gasdermin-mediated cell permeation required for release of active IL-1ß from human macrophages. We further identify and characterize a xanthine oxidase-ROS activated MAP3K5/JNK2 substrate licensing complex as a novel regulator of the GSDMD mobilization which precedes pyroptosis. We show that inhibitors targeting this MAP3K5 cascade alleviate morbidity in mouse models of colitis and dampen both augmented IL-1ß release and cell permeation in monocytes derived from patients with gain-of-function inflammasomopathies.


Asunto(s)
Inflamasomas , Piroptosis , Animales , Humanos , Ratones , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/fisiología , Transducción de Señal
2.
Cell Rep ; 41(1): 111441, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36179680

RESUMEN

Biologically active small molecules can impart modulatory effects, in some cases providing extended long-term memory. In a screen of biologically active small molecules for regulators of tumor necrosis factor (TNF) induction, we identify several compounds with the ability to induce training effects on human macrophages. Rutaecarpine shows acute and long-term modulation, enhancing lipopolysaccharide (LPS)-induced pro-inflammatory cytokine secretion and relieving LPS tolerance in human macrophages. Rutaecarpine inhibits ß-glucan-induced H3K4Me3 marks at the promoters of several pro-inflammatory cytokines, highlighting the potential of this molecule to modulate chromosomal topology. Syk kinase inhibitor (SYKi IV), another screen hit, promotes an enhanced response to LPS similar to that previously reported for ß-glucan-induced training. Macrophages trained with SYKi IV show a high degree of resistance to influenza A, multiple variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and OC43 coronavirus infection, highlighting a potential application of this molecule and other SYKis as prophylactic treatments for viral susceptibility.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , beta-Glucanos , Citocinas , Humanos , Alcaloides Indólicos , Lipopolisacáridos , Macrófagos , Quinazolinonas , SARS-CoV-2 , Quinasa Syk , Factor de Necrosis Tumoral alfa
4.
Cell Rep ; 25(1): 95-106.e6, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30282041

RESUMEN

Activation of the TLR4 signaling pathway by lipopolysaccharide (LPS) leads to induction of both inflammatory and interferon-stimulated genes, but the mechanisms through which these coordinately activated transcriptional programs are balanced to promote an optimal innate immune response remain poorly understood. In a genome-wide small interfering RNA (siRNA) screen of the LPS-induced tumor necrosis factor α (TNF-α) response in macrophages, we identify the interferon-stimulated protein IFIT1 as a negative regulator of the inflammatory gene program. Transcriptional profiling further identifies a positive regulatory role for IFIT1 in type I interferon expression, implicating IFIT1 as a reciprocal modulator of LPS-induced gene classes. We demonstrate that these effects of IFIT1 are mediated through modulation of a Sin3A-HDAC2 transcriptional regulatory complex at LPS-induced gene loci. Beyond the well-studied role of cytosolic IFIT1 in restricting viral replication, our data demonstrate a function for nuclear IFIT1 in differential transcriptional regulation of separate branches of the LPS-induced gene program.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Macrófagos/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Transducción de Señal , Complejo Correpresor Histona Desacetilasa y Sin3 , Factor de Necrosis Tumoral alfa/inmunología , Células U937
5.
Front Microbiol ; 9: 788, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29740418

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus responsible for the development of Kaposi's sarcoma, primary effusion lymphoma (PEL), and Multicentric Castleman's disease in immunocompromised individuals. Despite the burden of these diseases there are few treatment options for afflicted individuals, due in part to our limited understanding of virus-host interactions. Tip60, a histone aceytltransferase (HAT) has been previously shown to interact with both the KSHV latency associated nuclear antigen protein (LANA), which is the main factor in maintaining the viral latent state, and ORF36, a viral kinase expressed in the lytic phase. We further investigated Tip60-virus interaction to ascertain Tip60's role in the viral life cycle and its potential as a target for future therapeutics. Through modulation of Tip60 expression in HEK293T cells harboring a plasmid containing the KSHV viral episome, Bac36, we found that Tip60 is vital for both lytic replication as well as efficient expression of latent genes. Interestingly, Tip60 small molecule inhibitors, MG149 and NU9056, similarly inhibited latent and lytic genes, and reduced virion production in wild-type KSHV+/EBV- PEL, BCBL-1 cells. Long-term treatment with these Tip60 inhibitors selectively decreased the viability of KSHV-infected B lymphoma cells compared to uninfected cells. From this study, we conclude that Tip60 is important for KSHV infection and its associated cancer development, and Tip60 is therefore a potential target for future antiviral and anticancer therapeutics.

6.
Cell Rep ; 16(1): 232-246, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27342126

RESUMEN

The flaviviruses dengue virus (DENV) and Zika virus (ZIKV) are severe health threats with rapidly expanding ranges. To identify the host cell dependencies of DENV and ZIKV, we completed orthologous functional genomic screens using RNAi and CRISPR/Cas9 approaches. The screens recovered the ZIKV entry factor AXL as well as multiple host factors involved in endocytosis (RAB5C and RABGEF), heparin sulfation (NDST1 and EXT1), and transmembrane protein processing and maturation, including the endoplasmic reticulum membrane complex (EMC). We find that both flaviviruses require the EMC for their early stages of infection. Together, these studies generate a high-confidence, systems-wide view of human-flavivirus interactions and provide insights into the role of the EMC in flavivirus replication.


Asunto(s)
Virus del Dengue/genética , Genómica/métodos , Virus Zika/genética , Sistemas CRISPR-Cas , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Pruebas Genéticas , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Membranas Intracelulares/metabolismo , Unión Proteica , Mapas de Interacción de Proteínas , Interferencia de ARN , Replicación Viral
7.
Sci Rep ; 5: 9559, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25831078

RESUMEN

Macrophages play a critical role in the innate immune response to pathogen infection, but few tools exist for systematic dissection of these responses using modern genome-wide perturbation methods. To develop an assay platform for high-throughput analysis of macrophage activation by pathogenic stimuli, we generated reporter systems in human and mouse macrophages with dynamic readouts for NF-κB and/or TNF-α responses. These reporter cells show responsiveness to a broad range of TLR ligands and to gram-negative bacterial infection. There are significant challenges to the use of RNAi in innate immune cells, including efficient small RNA delivery and non-specific immune responses to dsRNA. To permit the interrogation of the macrophage pathogen response pathways with RNAi, we employed the stably expressed reporter genes to develop efficient siRNA delivery protocols for maximal target gene silencing with minimal activation of the innate macrophage response to nucleic acids. We demonstrate the utility of these macrophage cell systems for siRNA screening of pathogen responses by targeting components of the human and mouse TLR pathways, and observe species-specific perturbation of signaling and cytokine responses. Our approach to reporter cell development and siRNA delivery optimization provides an experimental paradigm with significant potential for developing genetic screening platforms in mammalian cells.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Macrófagos/metabolismo , ARN Interferente Pequeño/genética , Animales , Línea Celular , Análisis por Conglomerados , Expresión Génica , Orden Génico , Técnicas de Transferencia de Gen , Genes Reporteros , Vectores Genéticos/genética , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Lentivirus/genética , Ligandos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Regiones Promotoras Genéticas , Interferencia de ARN , Reproducibilidad de los Resultados , Receptores Toll-Like/metabolismo
8.
Cell Rep ; 9(2): 752-66, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25373910

RESUMEN

RNAi screens have implicated hundreds of host proteins as HIV-1 dependency factors (HDFs). While informative, these early studies overlap poorly due to false positives and false negatives. To ameliorate these issues, we combined information from the existing HDF screens together with new screens performed with multiple orthologous RNAi reagents (MORR). In addition to being traditionally validated, the MORR screens and the historical HDF screens were quantitatively integrated by the adaptation of an established analysis program, RIGER, for the collective interpretation of each gene's phenotypic significance. False positives were addressed by the removal of poorly expressed candidates through gene expression filtering, as well as with GESS, which identifies off-target effects. This workflow produced a quantitatively integrated network of genes that modulate HIV-1 replication. We further investigated the roles of GOLGI49, SEC13, and COG in HIV-1 replication. Collectively, the MORR-RIGER method minimized the caveats of RNAi screening and improved our understanding of HIV-1-host cell interactions.


Asunto(s)
VIH-1/fisiología , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Huésped-Patógeno , Interferencia de ARN , Replicación Viral , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Algoritmos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN
9.
Cell Rep ; 5(4): 895-908, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24268777

RESUMEN

The IFITMs inhibit influenza A virus (IAV) replication in vitro and in vivo. Here, we establish that the antimycotic heptaen, amphotericin B (AmphoB), prevents IFITM3-mediated restriction of IAV, thereby increasing viral replication. Consistent with its neutralization of IFITM3, a clinical preparation of AmphoB, AmBisome, reduces the majority of interferon's protective effect against IAV in vitro. Mechanistic studies reveal that IFITM1 decreases host-membrane fluidity, suggesting both a possible mechanism for IFITM-mediated restriction and its negation by AmphoB. Notably, we reveal that mice treated with AmBisome succumbed to a normally mild IAV infection, similar to animals deficient in Ifitm3. Therefore, patients receiving antifungal therapy with clinical preparations of AmphoB may be functionally immunocompromised and thus more vulnerable to influenza, as well as other IFITM3-restricted viral infections.


Asunto(s)
Anfotericina B/efectos adversos , Antifúngicos/efectos adversos , Huésped Inmunocomprometido , Subtipo H1N1 del Virus de la Influenza A/inmunología , Proteínas de la Membrana/genética , Infecciones por Orthomyxoviridae/inmunología , Internalización del Virus/efectos de los fármacos , Acetilcolina/farmacología , Anfotericina B/administración & dosificación , Animales , Antibacterianos/farmacología , Antifúngicos/administración & dosificación , Antígenos de Diferenciación/metabolismo , Transporte Biológico/efectos de los fármacos , Células COS , Fusión Celular , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Chlorocebus aethiops , Células HeLa , Humanos , Gripe Humana/inmunología , Interferones/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nistatina/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Sodio/metabolismo , Tetraetilamonio/farmacología , Replicación Viral/efectos de los fármacos
10.
J Virol ; 87(14): 7837-52, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23658454

RESUMEN

The interferon-induced transmembrane protein 3 (IFITM3) gene is an interferon-stimulated gene that inhibits the replication of multiple pathogenic viruses in vitro and in vivo. IFITM3 is a member of a large protein superfamily, whose members share a functionally undefined area of high amino acid conservation, the CD225 domain. We performed mutational analyses of IFITM3 and identified multiple residues within the CD225 domain, consisting of the first intramembrane domain (intramembrane domain 1 [IM1]) and a conserved intracellular loop (CIL), that are required for restriction of both influenza A virus (IAV) and dengue virus (DENV) infection in vitro. Two phenylalanines within IM1 (F75 and F78) also mediate a physical association between IFITM proteins, and the loss of this interaction decreases IFITM3-mediated restriction. By extension, similar IM1-mediated associations may contribute to the functions of additional members of the CD225 domain family. IFITM3's distal N-terminal domain is also needed for full antiviral activity, including a tyrosine (Y20), whose alteration results in mislocalization of a portion of IFITM3 to the cell periphery and surface. Comparative analyses demonstrate that similar molecular determinants are needed for IFITM3's restriction of both IAV and DENV. However, a portion of the CIL including Y99 and R87 is preferentially needed for inhibition of the orthomyxovirus. Several IFITM3 proteins engineered with rare single-nucleotide polymorphisms demonstrated reduced expression or mislocalization, and these events were associated with enhanced viral replication in vitro, suggesting that possessing such alleles may impact an individual's risk for viral infection. On the basis of this and other data, we propose a model for IFITM3-mediated restriction.


Asunto(s)
Virus del Dengue/fisiología , Virus de la Influenza A/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Animales , Técnicas de Cultivo de Célula , Clonación Molecular , Secuencia Conservada/genética , Análisis Mutacional de ADN , ADN Complementario/genética , Perros , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Células de Riñón Canino Madin Darby , Espectrometría de Masas , Microscopía Confocal , Modelos Biológicos , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple/genética , Estructura Terciaria de Proteína/genética , Replicación Viral/genética
11.
Gastroenterology ; 144(7): 1438-49, 1449.e1-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23462180

RESUMEN

BACKGROUND & AIMS: Hepatitis C virus (HCV) infection is a leading cause of end-stage liver disease. Interferon-α (IFNα) is an important component of anti-HCV therapy; it up-regulates transcription of IFN-stimulated genes, many of which have been investigated for their antiviral effects. However, all of the genes required for the antiviral function of IFNα (IFN effector genes [IEGs]) are not known. IEGs include not only IFN-stimulated genes, but other nontranscriptionally induced genes that are required for the antiviral effect of IFNα. In contrast to candidate approaches based on analyses of messenger RNA (mRNA) expression, identification of IEGs requires a broad functional approach. METHODS: We performed an unbiased genome-wide small interfering RNA screen to identify IEGs that inhibit HCV. Huh7.5.1 hepatoma cells were transfected with small interfering RNAs incubated with IFNα and then infected with JFH1 HCV. Cells were stained using HCV core antibody, imaged, and analyzed to determine the percent infection. Candidate IEGs detected in the screen were validated and analyzed further. RESULTS: The screen identified 120 previously unreported IEGs. From these, we more fully evaluated the following: asparagine-linked glycosylation 10 homolog (yeast, α-1,2-glucosyltransferase); butyrylcholinesterase; dipeptidyl-peptidase 4 (CD26, adenosine deaminase complexing protein 2); glucokinase (hexokinase 4) regulator; guanylate cyclase 1, soluble, ß 3; MYST histone acetyltransferase 1; protein phosphatase 3 (formerly 2B), catalytic subunit, ß isoform; peroxisomal proliferator-activated receptor-γ-DBD-interacting protein 1; and solute carrier family 27 (fatty acid transporter), member 2; and demonstrated that they enabled IFNα-mediated suppression of HCV at multiple steps of its life cycle. Expression of these genes had more potent effects against flaviviridae because a subset was required for IFNα to suppress dengue virus but not influenza A virus. In addition, many of the host genes detected in this screen (92%) were not transcriptionally stimulated by IFNα; these genes represent a heretofore unknown class of non-IFN-stimulated gene IEGs. CONCLUSIONS: We performed a whole-genome loss-of-function screen to identify genes that mediate the effects of IFNα against human pathogenic viruses. We found that IFNα restricts HCV via actions of general and specific IEGs.


Asunto(s)
Antivirales/uso terapéutico , Hepacivirus/genética , Hepatitis C/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Replicación Viral/genética , Hepacivirus/efectos de los fármacos , Humanos , ARN Viral/genética , Replicación Viral/efectos de los fármacos
12.
Cell Rep ; 2(4): 807-16, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23041316

RESUMEN

HIV-1 depends on many host factors for propagation. Other host factors, however, antagonize HIV-1 and may have profound effects on viral activation. Curing HIV-1 requires the reduction of latent viral reservoirs that remain in the face of antiretroviral therapy. Using orthologous genetic screens, we identified bromodomain containing 4 (BRD4) as a negative regulator of HIV-1 replication. Antagonism of BRD4, via RNA interference or with a small molecule inhibitor, JQ1, both increased proviral transcriptional elongation and alleviated HIV-1 latency in cell-line models. In multiple instances, JQ1, when used in combination with the NF-κB activators Prostratin or PHA, enhanced the in vitro reactivation of latent HIV-1 in primary T cells. These data are consistent with a model wherein BRD4 competes with the virus for HIV-1 dependency factors (HDFs) and suggests that combinatorial therapies that activate HDFs and antagonize HIV-1 competitive factors may be useful for curing HIV-1 infection.


Asunto(s)
VIH-1/fisiología , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Azepinas/farmacología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proteínas de Ciclo Celular , Células Cultivadas , Expresión Génica , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ésteres del Forbol/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Triazoles/farmacología , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
13.
Nature ; 484(7395): 519-23, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22446628

RESUMEN

The 2009 H1N1 influenza pandemic showed the speed with which a novel respiratory virus can spread and the ability of a generally mild infection to induce severe morbidity and mortality in a subset of the population. Recent in vitro studies show that the interferon-inducible transmembrane (IFITM) protein family members potently restrict the replication of multiple pathogenic viruses. Both the magnitude and breadth of the IFITM proteins' in vitro effects suggest that they are critical for intrinsic resistance to such viruses, including influenza viruses. Using a knockout mouse model, we now test this hypothesis directly and find that IFITM3 is essential for defending the host against influenza A virus in vivo. Mice lacking Ifitm3 display fulminant viral pneumonia when challenged with a normally low-pathogenicity influenza virus, mirroring the destruction inflicted by the highly pathogenic 1918 'Spanish' influenza. Similar increased viral replication is seen in vitro, with protection rescued by the re-introduction of Ifitm3. To test the role of IFITM3 in human influenza virus infection, we assessed the IFITM3 alleles of individuals hospitalized with seasonal or pandemic influenza H1N1/09 viruses. We find that a statistically significant number of hospitalized subjects show enrichment for a minor IFITM3 allele (SNP rs12252-C) that alters a splice acceptor site, and functional assays show the minor CC genotype IFITM3 has reduced influenza virus restriction in vitro. Together these data reveal that the action of a single intrinsic immune effector, IFITM3, profoundly alters the course of influenza virus infection in mouse and humans.


Asunto(s)
Virus de la Influenza A/patogenicidad , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Proteínas de Unión al ARN/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Citocinas/inmunología , Inglaterra/epidemiología , Eliminación de Gen , Humanos , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/clasificación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza B/clasificación , Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/patogenicidad , Gripe Humana/complicaciones , Gripe Humana/epidemiología , Gripe Humana/mortalidad , Gripe Humana/virología , Leucocitos/inmunología , Pulmón/patología , Pulmón/virología , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/etiología , Neumonía Viral/patología , Neumonía Viral/prevención & control , Polimorfismo de Nucleótido Simple/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Escocia/epidemiología , Replicación Viral
14.
PLoS Pathog ; 7(10): e1002337, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22046135

RESUMEN

To replicate, viruses must gain access to the host cell's resources. Interferon (IFN) regulates the actions of a large complement of interferon effector genes (IEGs) that prevent viral replication. The interferon inducible transmembrane protein family members, IFITM1, 2 and 3, are IEGs required for inhibition of influenza A virus, dengue virus, and West Nile virus replication in vitro. Here we report that IFN prevents emergence of viral genomes from the endosomal pathway, and that IFITM3 is both necessary and sufficient for this function. Notably, viral pseudoparticles were inhibited from transferring their contents into the host cell cytosol by IFN, and IFITM3 was required and sufficient for this action. We further demonstrate that IFN expands Rab7 and LAMP1-containing structures, and that IFITM3 overexpression is sufficient for this phenotype. Moreover, IFITM3 partially resides in late endosomal and lysosomal structures, placing it in the path of invading viruses. Collectively our data are consistent with the prediction that viruses that fuse in the late endosomes or lysosomes are vulnerable to IFITM3's actions, while viruses that enter at the cell surface or in the early endosomes may avoid inhibition. Multiple viruses enter host cells through the late endocytic pathway, and many of these invaders are attenuated by IFN. Therefore these findings are likely to have significance for the intrinsic immune system's neutralization of a diverse array of threats.


Asunto(s)
Citosol/virología , Virus de la Influenza A/efectos de los fármacos , Gripe Humana/inmunología , Interferón gamma/farmacología , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Internalización del Virus/efectos de los fármacos , Animales , Pollos , Citosol/efectos de los fármacos , Citosol/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Interferón gamma/inmunología , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Replicación Viral
15.
Cell ; 139(7): 1243-54, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20064371

RESUMEN

Influenza viruses exploit host cell machinery to replicate, resulting in epidemics of respiratory illness. In turn, the host expresses antiviral restriction factors to defend against infection. To find host cell modifiers of influenza A H1N1 viral infection, we used a functional genomic screen and identified over 120 influenza A virus-dependency factors with roles in endosomal acidification, vesicular trafficking, mitochondrial metabolism, and RNA splicing. We discovered that the interferon-inducible transmembrane proteins IFITM1, 2, and 3 restrict an early step in influenza A viral replication. The IFITM proteins confer basal resistance to influenza A virus but are also inducible by interferons type I and II and are critical for interferon's virustatic actions. Further characterization revealed that the IFITM proteins inhibit the early replication of flaviviruses, including dengue virus and West Nile virus. Collectively this work identifies a family of antiviral restriction factors that mediate cellular innate immunity to at least three major human pathogens.


Asunto(s)
Infecciones por Flavivirus/inmunología , Gripe Humana/inmunología , Proteínas de la Membrana/inmunología , Animales , Antígenos de Diferenciación , Línea Celular Tumoral , Virus del Dengue/inmunología , Humanos , Inmunidad Innata , Virus de la Influenza A/inmunología , Interferones/inmunología , Ratones , Proteínas de Unión al ARN/inmunología , Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología
16.
J Virol ; 81(17): 8967-76, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17567691

RESUMEN

The Ebola virus (EBOV) genome encodes for several proteins that are necessary and sufficient for replication and transcription of the viral RNAs in vitro; NP, VP30, VP35, and L. VP30 acts in trans with an RNA secondary structure upstream of the first transcriptional start site to modulate transcription. Using a bioinformatics approach, we identified a region within the N terminus of VP30 with sequence features that typify intrinsically disordered regions and a putative RNA binding site. To experimentally assess the ability of VP30 to directly interact with the viral RNA, we purified recombinant EBOV VP30 to >90% homogeneity and assessed RNA binding by UV cross-linking and filter-binding assays. VP30 is a strongly acidophilic protein; RNA binding became stronger as pH was decreased. Zn(2+), but not Mg(2+), enhanced activity. Enhancement of transcription by VP30 requires a RNA stem-loop located within nucleotides 54 to 80 of the leader region. VP30 showed low binding affinity to the predicted stem-loop alone or to double-stranded RNA but showed a good binding affinity for the stem-loop when placed in the context of upstream and downstream sequences. To map the region responsible for interacting with RNA, we constructed, purified, and assayed a series of N-terminal deletion mutations of VP30 for RNA binding. The key amino acids supporting RNA binding activity map to residues 26 to 40, a region rich in arginine. Thus, we show for the first time the direct interaction of EBOV VP30 with RNA and the importance of the N-terminal region for binding RNA.


Asunto(s)
ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/genética , Coenzimas/farmacología , Biología Computacional , Ebolavirus/química , Ebolavirus/metabolismo , Concentración de Iones de Hidrógeno , Magnesio/farmacología , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Unión Proteica , ARN Bicatenario/metabolismo , ARN Viral/química , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/aislamiento & purificación , Eliminación de Secuencia , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/aislamiento & purificación , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación , Zinc/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA