Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(1): 111949, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36640307

RESUMEN

Viruses must overcome the interferon-mediated antiviral response to replicate and propagate into their host. Rabies virus (RABV) phosphoprotein P is known to inhibit interferon induction. Here, using a global mass spectrometry approach, we show that RABV P binds to TBK1, a kinase located at the crossroads of many interferon induction pathways, resulting in innate immunity inhibition. Mutations of TBK1 phosphorylation sites abolish P binding. Importantly, we demonstrate that upon RABV infection or detection of dsRNA by innate immunity sensors, TBK1 and its adaptor proteins NAP1 and SINTBAD form dynamic cytoplasmic condensates that have liquid properties. These condensates can form larger aggregates having ring-like structures in which NAP1 and TBK1 exhibit locally restricted movement. P binding to TBK1 interferes with the formation of these structures. This work demonstrates that proteins of the signaling pathway leading to interferon induction transiently form liquid organelles that can be targeted by viruses.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Virus de la Rabia , Proteínas Serina-Treonina Quinasas/metabolismo , Inmunidad Innata , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interferones/metabolismo , Factor 3 Regulador del Interferón/metabolismo
2.
PLoS Pathog ; 12(10): e1005942, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27749929

RESUMEN

Stress granules (SGs) are membrane-less dynamic structures consisting of mRNA and protein aggregates that form rapidly in response to a wide range of environmental cellular stresses and viral infections. They act as storage sites for translationally silenced mRNAs under stress conditions. During viral infection, SG formation results in the modulation of innate antiviral immune responses, and several viruses have the ability to either promote or prevent SG assembly. Here, we show that rabies virus (RABV) induces SG formation in infected cells, as revealed by the detection of SG-marker proteins Ras GTPase-activating protein-binding protein 1 (G3BP1), T-cell intracellular antigen 1 (TIA-1) and poly(A)-binding protein (PABP) in the RNA granules formed during viral infection. As shown by live cell imaging, RABV-induced SGs are highly dynamic structures that increase in number, grow in size by fusion events, and undergo assembly/disassembly cycles. Some SGs localize in close proximity to cytoplasmic viral factories, known as Negri bodies (NBs). Three dimensional reconstructions reveal that both structures remain distinct even when they are in close contact. In addition, viral mRNAs synthesized in NBs accumulate in the SGs during viral infection, revealing material exchange between both compartments. Although RABV-induced SG formation is not affected in MEFs lacking TIA-1, TIA-1 depletion promotes viral translation which results in an increase of viral replication indicating that TIA-1 has an antiviral effect. Inhibition of PKR expression significantly prevents RABV-SG formation and favors viral replication by increasing viral translation. This is correlated with a drastic inhibition of IFN-B gene expression indicating that SGs likely mediate an antiviral response which is however not sufficient to fully counteract RABV infection.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Cuerpos de Inclusión Viral/virología , Virus de la Rabia , Rabia/virología , Replicación Viral/fisiología , Animales , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata , Hibridación Fluorescente in Situ , Microscopía Confocal , Rabia/inmunología , Virus de la Rabia/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
PLoS One ; 7(6): e38336, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22685561

RESUMEN

BACKGROUND: Induction of Type I Interferon (IFN) genes constitutes an essential step leading to innate immune responses during virus infection. Sendai virus (SeV) infection of B lymphoid Namalwa cells transiently induces the transcriptional expression of multiple IFN-A genes. Although transcriptional activation of IFN-A genes has been extensively studied, the mechanism responsible for the attenuation of their expression remains to be determined. PRINCIPAL FINDINGS: In this study, we demonstrate that virus infection of Namalwa cells induces transient recruitment of HDAC3 (histone deacetylase 3) to IFN-A promoters. Analysis of chromatin-protein association by Chip-QPCR demonstrated that recruitment of interferon regulatory factor (IRF)3 and IRF7, as well as TBP correlated with enhanced histone H3K9 and H3K14 acetylation, whereas recruitment of HDAC3 correlated with inhibition of histone H3K9/K14 acetylation, removal of IRF7 and TATA-binding protein (TBP) from IFN-A promoters and inhibition of virus-induced IFN-A gene transcription. Additionally, HDAC3 overexpression reduced, and HDAC3 depletion by siRNA enhanced IFN-A gene expression. Furthermore, activation of IRF7 enhanced histone H3K9/K14 acetylation and IFN-A gene expression, whereas activation of both IRF7 and IRF3 led to recruitment of HDAC3 to the IFN-A gene promoters, resulting in impaired histone H3K9 acetylation and attenuation of IFN-A gene transcription. CONCLUSION: Altogether these data indicate that reversal of histone H3K9/K14 acetylation by HDAC3 is required for attenuation of IFN-A gene transcription during viral infection.


Asunto(s)
Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Interferón-alfa/genética , Regiones Promotoras Genéticas/genética , Acetilación , Linfoma de Burkitt/genética , Linfoma de Burkitt/patología , Linfoma de Burkitt/virología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Células HEK293 , Histona Desacetilasas/genética , Histonas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Lisina/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sendai/fisiología , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo
4.
Nat Med ; 15(10): 1170-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19767732

RESUMEN

The therapeutic efficacy of anticancer chemotherapies may depend on dendritic cells (DCs), which present antigens from dying cancer cells to prime tumor-specific interferon-gamma (IFN-gamma)-producing T lymphocytes. Here we show that dying tumor cells release ATP, which then acts on P2X(7) purinergic receptors from DCs and triggers the NOD-like receptor family, pyrin domain containing-3 protein (NLRP3)-dependent caspase-1 activation complex ('inflammasome'), allowing for the secretion of interleukin-1beta (IL-1beta). The priming of IFN-gamma-producing CD8+ T cells by dying tumor cells fails in the absence of a functional IL-1 receptor 1 and in Nlpr3-deficient (Nlrp3(-/-)) or caspase-1-deficient (Casp-1(-/-)) mice unless exogenous IL-1beta is provided. Accordingly, anticancer chemotherapy turned out to be inefficient against tumors established in purinergic receptor P2rx7(-/-) or Nlrp3(-/-) or Casp1(-/-) hosts. Anthracycline-treated individuals with breast cancer carrying a loss-of-function allele of P2RX7 developed metastatic disease more rapidly than individuals bearing the normal allele. These results indicate that the NLRP3 inflammasome links the innate and adaptive immune responses against dying tumor cells.


Asunto(s)
Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interleucina-1beta/metabolismo , Animales , Caspasa 1/metabolismo , Células Dendríticas/citología , Inmunidad/inmunología , Inmunidad Innata , Inflamación/inmunología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Timoma/inmunología
5.
Cytokine Growth Factor Rev ; 20(4): 283-95, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19651532

RESUMEN

Immune recognition of virus-associated molecules by Toll-like receptors (TLRs) and/or RIG-I-like receptors (RLRs) triggers intracellular signaling cascades that converge on the activation of interferon regulatory factors - particularly IRF3 and IRF7, leading to the transcriptional induction of type 1 interferon genes. This review summarizes new data describing how these factors regulate the temporal and quantitative differences in the expression of the multigenic IFN-A family. The distinctive DNA-binding features of IRF3 and IRF7 affect the selectivity and affinity of these factors for IFN-A promoters; modification of the ratio of promoter-bound IRF3 and IRF7 during virus infection may influence both transcriptional activation and repression of IFN-A genes. This review also summarizes the structural differences between IFN-beta and different IFN-alpha subtypes, their interaction with their common receptor IFNAR, and their potency to elicit antiviral, antiproliferative and antitumoral responses. Taken together, this information enhances our understanding of the selective advantage of the multiplicity of IFN-alpha subtypes in the regulation of innate and adaptive immunity.


Asunto(s)
Perfilación de la Expresión Génica , Interferón-alfa/genética , Interferón-alfa/inmunología , Secuencia de Aminoácidos , Antivirales/inmunología , Antivirales/metabolismo , Secuencia de Bases , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
6.
Semin Immunol ; 21(4): 215-22, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19539500

RESUMEN

Viral and microbial pathogens contain specific motifs or pathogen-associated molecular patterns (PAMPs) that are recognized by cell surface- and endosome-associated Toll-like receptors (TLRs). RNA virus infection is also detected through TLR-independent mechanisms. Early viral replicative intermediates are detected by two recently characterized cystolic viral RNA receptors-RIG-I and MDA-5. Both are DExDH/box RNA helicases, and RIG-I specifically recognizes 5'-triphosphate containing viral RNA and transmits signals that induce type I interferon-mediated host immunity against virus infection. In this review, we will focus on RIG-I-like receptor (RLR) signal transduction and the regulatory mechanisms - ubiquitination, deubiquitination, ISGylation - underlying this important host response.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Infecciones por Virus ARN/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , ARN Helicasas DEAD-box/química , Humanos , ARN Viral/inmunología , Transducción de Señal , Ubiquitinación
7.
Mol Cell Biol ; 29(12): 3435-50, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19349300

RESUMEN

Differential expression of the human interferon A (IFN-A) gene cluster is modulated following paramyxovirus infection by the relative amounts of active interferon regulatory factor 3 (IRF-3) and IRF-7. IRF-3 expression activates predominantly IFN-A1 and IFN-B, while IRF-7 expression induces multiple IFN-A genes. IFN-A1 gene expression is dependent on three promoter proximal IRF elements (B, C, and D modules, located at positions -98 to -45 relative to the mRNA start site). IRF-3 binds the C module of IFN-A1, while other IFN-A gene promoters are responsive to the binding of IRF-7 to the B and D modules. Maximal expression of IFN-A1 is observed with complete occupancy of the three modules in the presence of IRF-7. Nucleotide substitutions in the C modules of other IFN-A genes disrupt IRF-3-mediated transcription, whereas a G/A substitution in the D modules enhances IRF7-mediated expression. IRF-3 exerts dual effects on IFN-A gene expression, as follows: a synergistic effect with IRF-7 on IFN-A1 expression and an inhibitory effect on other IFN-A gene promoters. Chromatin immunoprecipitation experiments reveal that transient binding of both IRF-3 and IRF-7, accompanied by CBP/p300 recruitment to the endogenous IFN-A gene promoters, is associated with transcriptional activation, whereas a biphasic recruitment of IRF-3 and CBP/p300 represses IFN-A gene expression. This regulatory mechanism contributes to differential expression of IFN-A genes and may be critical for alpha interferon production in different cell types by RIG-I-dependent signals, leading to innate antiviral immune responses.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Secuencia de Bases , Sitios de Unión , Línea Celular , ADN/genética , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/antagonistas & inhibidores , Factor 7 Regulador del Interferón/genética , Interferón Tipo I/biosíntesis , Datos de Secuencia Molecular , Familia de Multigenes , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Virus Sendai/inmunología , Virus Sendai/patogenicidad , Homología de Secuencia de Ácido Nucleico , Transducción de Señal , Activación Transcripcional , Factores de Transcripción p300-CBP/metabolismo
8.
Biochem Pharmacol ; 72(11): 1469-76, 2006 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-16846591

RESUMEN

Type I IFN (IFN-alpha/beta) have important biological functions ranging from immune cell development and activation, to tumor cell killing and most importantly inhibition of virus replication. Following viral infection or activation of Toll-like receptors (TLRs) via distinct ligands, IFN-alpha/beta are produced. Two members of the interferon regulatory factor (IRF) family - IRF-3 and IRF-7 - are the major modulators of IFN gene expression. Activation of IRF-3 and IRF-7 by TBK1/IKKvarepsilon mediated phosphorylation promotes IFN gene expression and potentiates the production of IFN responsive genes important to the development of an effective antiviral immune response. IFN treatment can augment anti-tumor properties and they are potentially key players in cancer therapy. For example, adoptive transfer of IFN-gamma-activated macrophages can mediate tumor cell killing via direct cell-cell contact, as well as release of soluble cytotoxic pro-inflammatory molecules. A recent study investigated whether IRF-3 and IRF-7 could mediate the acquisition of new anti-tumor effector functions in macrophages. Adenovirus mediated transduction of the active form of IRF-7 into primary macrophages resulted in the production of type I IFN, upregulation of target genes including TRAIL and increased tumoricidal activity of macrophages; in contrast, the active form of IRF-3 led to induction of cell death. These studies indicate that IRF-7 transduced macrophages may be an attractive candidate for in vivo adoptive therapy of cancer.


Asunto(s)
Antineoplásicos/inmunología , Citotoxicidad Inmunológica , Factor 3 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/inmunología , Interferón-alfa , Macrófagos/inmunología , Animales , Regulación Viral de la Expresión Génica , Humanos , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , Interferón-alfa/genética , Interferón-alfa/inmunología , Ratones , Transducción Genética
9.
J Biol Chem ; 281(8): 4856-66, 2006 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-16380379

RESUMEN

Virus-induced expression of interferon (IFN)-A genes is regulated by two members of the IFN regulatory factor (IRF) family, IRF-3 and IRF-7, which are activated by phosphorylation during viral infection by the IKK-related serine/threonine kinases TBK1 and IkappaB kinase epsilon (IKKepsilon). In this study, we demonstrate that three IRF-binding sites located in the virus-responsive element mediate the transcriptional activation of the IFN-A4 promoter by IRF-3. The precise arrangement of these IRF elements is required for synergistic activation of the IFN-A4 promoter following Newcastle disease virus infection or activation by TBK1 or IKKepsilon. The ordered assembly of IRF-3 multimers on the promoter also determines cooperative recruitment of IRF-3 and CREB-binding protein and differential virus-induced expression of IFN-A4 gene promoter compared with IFN-A11. Naturally occurring nucleotide substitutions disrupt two of the IRF elements in the IFN-A11 gene promoter, leading to a dramatic decrease in IRF-3 and CREB-binding protein recruitment and in IRF-3-dependent transcription. Transcription of the IFN-A4 promoter by IRF-7 is mediated by two IRF elements; promoter mutants that carry a reversed IRF element retain the ability to respond to IKKepsilon or TBK1 expression in the presence of IRF-7 but lose the capacity to respond to virus or kinase-induced IRF-3. Interestingly, IKKepsilon or TBK1 stimulates the IRF-7-mediated transcription of IFN-A11, although at a lesser extent compared with IFN-A4. Our data indicate that virus-induced expression of IFN-A genes is dictated by the organization of IRF elements within the IFN-A promoters and that the differential IFN-A gene expression, based on the IRF-3 responsiveness, is partially compensated in the presence of IRF-7 when both factors are activated by IKKepsilon or TBK1.


Asunto(s)
Regulación Viral de la Expresión Génica , Regulación de la Expresión Génica , Quinasa I-kappa B/metabolismo , Interferón-alfa/genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Inmunoprecipitación de Cromatina , Fibroblastos/metabolismo , Genes Reporteros , Células HeLa , Humanos , Immunoblotting , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Plásmidos/metabolismo , Unión Proteica , Transcripción Genética , Activación Transcripcional , Transfección
10.
J Virol ; 77(12): 7113-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12768031

RESUMEN

Two members of the signal transducer and activator of transcription family, STAT1 and STAT2, form, together with interferon regulatory factor 9 (IRF-9), the ISGF3 complex that activates the expression of the interferon-stimulated genes (ISG). The ISGF3 complex also participates in the virus-induced alpha/beta interferon (IFN-alpha/beta) gene amplification cascade by up-regulating IRF-7 gene expression. Here, we show that treatment of cells with trichostatin A (TSA), a deacetylase inhibitor, inhibits the virus-induced activation of IFN-alpha/beta promoters and dramatically reduces the ability of different ISG promoters to respond to IFN stimulation. Impairment of IFN-alpha/beta and ISG expression by TSA in infected cells is due to the blockage of interferon-stimulated ISGF3 complex formation, which leads to the abolition of IRF-7 gene expression. We also show that the TSA-dependent inhibition of ISGF3 is related to impaired nuclear accumulation of STAT2. Our data suggest that an acetylation/deacetylation mechanism participates in the regulation of cellular distribution and function of STAT2 in IFN-alpha/beta signaling.


Asunto(s)
Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Ácidos Hidroxámicos/farmacología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Factor 7 Regulador del Interferón , Factor 3 de Genes Estimulados por el Interferón , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón , Interferón-alfa/farmacología , Interferón beta/farmacología , Ratones , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/patogenicidad , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transactivadores/metabolismo , Factores de Transcripción/genética
11.
Autoimmunity ; 36(8): 447-55, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14984021

RESUMEN

Transcriptional regulation is a consequence of the combination of both activation and repression for establishing specific patterns of eukaryotic gene expression. The regulation of the expression of type I interferon (IFN-A and -B) multigene family is controlled primarily at the transcriptional level and has been widely studied as a model to understand the mechanisms of stable repression, transient expression and postinduction repression of genes. The positive and negative regulatory elements required for this on/off switch have been defined within a complex 5' upstream region of their transcription start site. The differential expression pattern of IFN-A genes is thought to involve both substitutions in the virus responsive element (VRE-A) and presence or absence of the distal negative regulatory element (DNRE) which is delimited upstream of the VRE-A. The interferon regulatory factors (IRF)-3 and -7 binding to the VRE-A and interacting as homodimers or heterodimers participate in the virus-induced transcriptional activation of IFN-A family. This data and the presence of homeodomain protein pituitary homeobox 1 (Pitx1) binding to the distal DNRE, negatively regulating the IRF-3 and IRF-7 activities and interacting physically with IRF-3 and IRF-7 contribute to our understanding of the complex differential transcriptional activation and repression of the IFN-A genes.


Asunto(s)
Interferón-alfa/genética , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor 3 Regulador del Interferón , Factor 7 Regulador del Interferón , Interferón-alfa/biosíntesis , Ratones , Datos de Secuencia Molecular , Factores de Transcripción Paired Box , Secuencias Reguladoras de Ácidos Nucleicos , Elementos de Respuesta , Factores de Transcripción/química , Factores de Transcripción/genética
12.
Biochimie ; 84(7): 643-54, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12453636

RESUMEN

Different members of the interferon regulatory factor (IRF) family are early activated by viral infection of eukaryotic cells. The IRFs participate in the virus-induced transcriptional regulation of different genes, including the multigenic interferon-A (IFN-A) family, members of which are involved in the establishment of an antiviral state, cell growth inhibition or apoptosis. This study presents the recent progress in the field of virus-induced transactivation and repression of IFN-A gene promoters. Data presented on the modular organization of IFN-A gene promoters and their transactivation dependent on IRF-3 and IRF-7 provide a new insight on the cooperativity mechanisms among the different IRF family members. Data on the transcriptional repression of virus-induced interferon-A promoters by the homeodomain protein Pitx1 contribute to our understanding of the complex differential transcriptional activation, repression and antirepression of the IFN-A genes.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Interferón-alfa/genética , Proteínas Virales/fisiología , Animales , Secuencia de Bases , Proteínas de Unión al ADN/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Factor 3 Regulador del Interferón , Factor 7 Regulador del Interferón , Interferón-alfa/biosíntesis , Ratones , Datos de Secuencia Molecular , Factores de Transcripción Paired Box , Regiones Promotoras Genéticas , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Activación Transcripcional
13.
J Mol Biol ; 316(5): 1009-22, 2002 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11884139

RESUMEN

Transcription of the murine interferon-A4 (IFN-A4) gene is mediated by a virus responsive element (VRE-A4) located in the promoter proximal [-120 to -43] region. VRE-A4 contains four DNA modules (A to D) which cooperate for maximal IFN-A4 activation following virus infection. The differential expression between the highly expressed IFN-A4 and the weakly inducible IFN-A11 gene promoters is essentially due to point mutations within the C and D modules of the virus-responsive element VRE-A11. We now demonstrate that in murine L929 and human 293 cells, transcription factors IRF-3 and IRF-7, which are potent activators of virus-induced type I IFN transcription, differentially affect IFN-A4 and IFN-A11 promoter activities. Using electrophoretic mobility shift assays and DNase I footprinting data, our studies demonstrate that the AB modules correspond to a preferential site for IRF-7, whereas the C module is preferentially recognized by IRF-3. Furthermore, transfection of reporter constructs driven by four copies of different GAAANN hexameric motifs found within VRE-A4 indicates that the NN residues of these hexameric sequences define the preferential binding sites for IRF-3 or IRF-7. Together, these experiments clarify the molecular basis for differential expression of IFN-A genes following virus infection by delineating the sequence requirements for IRF association with the virus responsive elements of the IFN-A genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/genética , Interferón-alfa/genética , Elementos de Respuesta/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Huella de ADN , Desoxirribonucleasa I/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Humanos , Factor 3 Regulador del Interferón , Factor 7 Regulador del Interferón , Ratones , Datos de Secuencia Molecular , Mutación/genética , Regiones Promotoras Genéticas/genética
14.
J Interferon Cytokine Res ; 22(1): 77-86, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11846978

RESUMEN

Maximal activation of murine infection-A4 (IFNA4) gene transcription following viral infection requires the presence of four cooperating DNA sequences (denoted A to D), which make up the virus responsive element VRE-A4. The B, C, and D modules, when tandemized, form binding sites for the virus-induced factor (VIF), a multiprotein complex that is detected early after viral infection in the nuclei of mouse L929 cells. We now demonstrate that IFN regulatory factor-3 (IRF-3) is a component of VIF and that VIF is different from the previously identified virus-activated complexes containing IRF-3 and coactivators of transcription, such as CREB binding protein (CBP) or p300. We also show that the C module is critical for both IRF-3-mediated and virus-induced transcription of the murine IFNA4 gene. Consistently, DNase I footprinting experiments and EMSA performed with increasing amounts of recombinant GST-IRF-3(DBD) fusion proteins demonstrate that cooperativity between the modules facilitate the binding of IRF-3 and recruitment of transcription coactivators on the IFNA4 promoter. These results indicate that VIF differentially recognizes the virus-responsive modules of VRE-A4 and further actualize our previous model concerning the differential expression of murine IFNA genes.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Interferón-alfa/genética , Virus de la Enfermedad de Newcastle/fisiología , Regiones Promotoras Genéticas , Elementos de Respuesta , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Proteína de Unión a CREB , Línea Celular , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Proteína p300 Asociada a E1A , Humanos , Factor 3 Regulador del Interferón , Interferón-alfa/metabolismo , Sustancias Macromoleculares , Ratones , Datos de Secuencia Molecular , Complejos Multiproteicos , Proteínas Nucleares/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...