Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
EMBO J ; 43(11): 2233-2263, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38658796

RESUMEN

Type I interferons (IFN-I, including IFNß) and IFNγ produce overlapping, yet clearly distinct immunological activities. Recent data show that the distinctness of global transcriptional responses to the two IFN types is not apparent when comparing their immediate effects. By analyzing nascent transcripts induced by IFN-I or IFNγ over a period of 48 h, we now show that the distinctiveness of the transcriptomes emerges over time and is based on differential employment of the ISGF3 complex as well as of the second-tier transcription factor IRF1. The distinct transcriptional properties of ISGF3 and IRF1 correspond with a largely diverse nuclear protein interactome. Mechanistically, we describe the specific input of ISGF3 and IRF1 into enhancer activation and the regulation of chromatin accessibility at interferon-stimulated genes (ISG). We further report differences between the IFN types in altering RNA polymerase II pausing at ISG 5' ends. Our data provide insight how transcriptional regulators create immunological identities of IFN-I and IFNγ.


Asunto(s)
Regulación de la Expresión Génica , Factor 1 Regulador del Interferón , Interferón beta , Interferón gamma , Transducción de Señal , Interferón gamma/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Factor 1 Regulador del Interferón/genética , Interferón beta/metabolismo , Interferón beta/genética , Humanos , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor 3 de Genes Estimulados por el Interferón/genética , Animales , Ratones , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética
2.
Int J Mol Sci ; 24(24)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38139463

RESUMEN

In addition to the canonical ISGF3 and non-canonical STAT2/IRF9 complexes, evidence is emerging of the role of their unphosphorylated counterparts in IFN-dependent and -independent ISG transcription. To better understand the relation between ISGF3 and U-ISGF3 and STAT2/IRF9 and U-STAT2/IRF9 in IFN-I-stimulated transcriptional responses, we performed RNA-Seq and ChIP-Seq, in combination with phosphorylation inhibition and antiviral experiments. First, we identified a group of ISRE-containing ISGs that were commonly regulated in IFNα-treated WT and STAT1-KO cells. Thus, in 2fTGH and Huh7.5 WT cells, early and long-term IFNα-inducible transcription and antiviral activity relied on the DNA recruitment of the ISGF3 components STAT1, STAT2 and IRF9 in a phosphorylation- and time-dependent manner. Likewise, in ST2-U3C and Huh-STAT1KO cells lacking STAT1, delayed IFN responses correlated with DNA binding of phosphorylated STAT2/IRF9 but not U-STAT2/IRF9. In addition, comparative experiments in U3C (STAT1-KO) cells overexpressing all the ISGF3 components (ST1-ST2-IRF9-U3C) revealed U-ISGF3 (and possibly U-STAT2/IRF9) chromatin interactions to correlate with phosphorylation-independent ISG transcription and antiviral activity. Together, our data point to the dominant role of the canonical ISGF3 and non-canonical STAT2/IRF9, without a shift to U-ISGF3 or U-STAT2/IRF9, in the regulation of early and prolonged ISG expression and viral protection. At the same time, they suggest the threshold-dependent role of U-ISFG3, and potentially U-STAT2/IRF9, in the regulation of constitutive and possibly long-term IFNα-dependent responses.


Asunto(s)
Interferón Tipo I , Factor 3 de Genes Estimulados por el Interferón , Proteína 1 Similar al Receptor de Interleucina-1 , Factor de Transcripción STAT2 , Antivirales/farmacología , ADN/farmacología , Inmunoglobulinas/metabolismo , Interferón Tipo I/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Transducción de Señal , Factor de Transcripción STAT1/metabolismo , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor de Transcripción STAT2/metabolismo , Humanos
3.
Trends Cancer ; 9(1): 83-92, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36216730

RESUMEN

Acute exposure of cancer cells to high concentrations of type I interferon (IFN-I) drives growth arrest and apoptosis, whereas chronic exposure to low concentrations provides important prosurvival advantages. Tyrosine-phosphorylated IFN-stimulated gene (ISG) factor 3 (ISGF3) drives acute deleterious responses to IFN-I, whereas unphosphorylated (U-)ISGF3, lacking tyrosine phosphorylation, drives essential constitutive prosurvival mechanisms. Surprisingly, programmed cell death-ligand 1 (PD-L1), often expressed on the surfaces of tumor cells and well recognized for its importance in inactivating cytotoxic T cells, also has important cell-intrinsic protumor activities, including dampening acute responses to cytotoxic high levels of IFN-I and sustaining the expression of the low levels that benefit tumors. More thorough understanding of the newly recognized complex roles of IFN-I in cancer may lead to the identification of novel therapeutic strategies.


Asunto(s)
Interferones , Neoplasias , Humanos , Interferones/metabolismo , Factor 3 de Genes Estimulados por el Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Transducción de Señal , Tirosina , Neoplasias/tratamiento farmacológico , Neoplasias/genética
4.
Front Immunol ; 12: 651254, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897699

RESUMEN

Interferon ß (IFN-ß) signaling activates the transcription factor complex ISGF3 to induce gene expression programs critical for antiviral defense and host immune responses. It has also been observed that IFN-ß activates a second transcription factor complex, γ-activated factor (GAF), but the significance of this coordinated activation is unclear. We report that in murine lung epithelial cells (MLE12) high doses of IFN-ß indeed activate both ISGF3 and GAF, which bind to distinct genomic locations defined by their respective DNA sequence motifs. In contrast, low doses of IFN-ß preferentially activate ISGF3 but not GAF. Surprisingly, in MLE12 cells GAF binding does not induce nearby gene expression even when strongly bound to the promoter. Yet expression of interferon stimulated genes is enhanced when GAF and ISGF3 are both active compared to ISGF3 alone. We propose that GAF may function as a dose-sensitive amplifier of ISG expression to enhance antiviral immunity and establish pro-inflammatory states.


Asunto(s)
Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón beta/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Línea Celular , Secuenciación de Inmunoprecipitación de Cromatina , Relación Dosis-Respuesta Inmunológica , Células Epiteliales/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Multimerización de Proteína/inmunología , RNA-Seq
5.
J Transl Med ; 18(1): 452, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33256749

RESUMEN

BACKGROUND: Dysregulation of transcription and cytokine expression has been implicated in the pathogenesis of a variety inflammatory diseases. The resulting imbalance between inflammatory and resolving transcriptional programs can cause an overabundance of pro-inflammatory, classically activated macrophage type 1 (M1) and/or helper T cell type 1 (Th1) products, such as IFNγ, TNFα, IL1-ß, and IL12, that prevent immune switching to resolution and healing. The low molecular weight fraction of human serum albumin (LMWF5A) is a novel biologic drug that is currently under clinical investigation for the treatment of osteoarthritis and the hyper-inflammatory response associated with COVID-19. This study aims to elucidate transcriptional mechanisms of action involved with the ability of LMWF5A to reduce pro-inflammatory cytokine release. METHODS: ELISA arrays were used to identify cytokines and chemokines influenced by LMWF5A treatment of LPS-stimulated peripheral blood mononuclear cells (PBMC). The resulting profiles were analyzed by gene enrichment to gain mechanistic insight into the biologic processes and transcription factors (TFs) underlying the identified differentially expressed cytokines. DNA-binding ELISAs, luciferase reporter assays, and TNFα or IL-1ß relative potency were then employed to confirm the involvement of enriched pathways and TFs. RESULTS: LMWF5A was found to significantly inhibit a distinct set of pro-inflammatory cytokines (TNFα, IL-1ß, IL-12, CXCL9, CXCL10, and CXCL11) associated with pro-inflammatory M1/Th1 immune profiles. Gene enrichment analysis also suggests these cytokines are, in part, regulated by NF-κB and STAT transcription factors. Data from DNA-binding and reporter assays support this with LMWF5A inhibition of STAT1α DNA-binding activity as well as a reduction in overall NF-κB-driven luciferase expression. Experiments using antagonists specific for the immunomodulatory and NF-κB/STAT-repressing transcription factors, peroxisome proliferator-activated receptor (PPAR)γ and aryl hydrocarbon receptor (AhR), indicate these pathways are involved in the LMWF5A mechanisms of action by reducing LMWF5A drug potency as measured by TNFα and IL-1ß release. CONCLUSION: In this report, we provide evidence that LMWF5A reduces pro-inflammatory cytokine release by activating the immunoregulatory transcription factors PPARγ and AhR. In addition, our data indicate that LMWF5A suppresses NF-κB and STAT1α pro-inflammatory pathways. This suggests that LMWF5A acts through these mechanisms to decrease pro-inflammatory transcription factor activity and subsequent inflammatory cytokine production.


Asunto(s)
Citocinas/metabolismo , Inflamación/prevención & control , Leucocitos Mononucleares/efectos de los fármacos , Albúmina Sérica Humana/farmacología , Antiinflamatorios/farmacología , COVID-19/inmunología , COVID-19/patología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos , Activación de Linfocitos/efectos de los fármacos , Peso Molecular , FN-kappa B/metabolismo , Albúmina Sérica Humana/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Factores de Transcripción/metabolismo , Tratamiento Farmacológico de COVID-19
6.
Cytokine ; 126: 154870, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31629105

RESUMEN

Interferon stimulated genes (ISGs), a collection of genes important in the early innate immune response, are upregulated in response to stimulation by extracellular type I interferons. The regulation of ISGs has been extensively studied in cells exposed to significant interferon stimulation, but less is known about ISG regulation in homeostatic regimes in which extracellular interferon levels are low. Using a collection of pre-existing, publicly available microarray datasets, we investigated ISG regulation at homeostasis in CD4, pulmonary epithelial, fibroblast and macrophage cells. We used a linear regression model to predict ISG expression levels from regulator expression levels. Our results suggest significant regulation of ISG expression at homeostasis, both through the ISGF3 molecule and through IRF7 and IRF8 associated pathways. We find that roughly 50% of ISGs have expression levels significantly correlated with ISGF3 expression levels at homeostasis, supporting previous results suggesting that homeostatic IFN levels have broad functional consequences. We find that ISG expression levels varied in their correlation with ISGF3, with epithelial and macrophage cells showing more correlation than CD4 and fibroblast cells. Our analysis provides a novel approach for decomposing and quantifying ISG regulation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Inmunidad Innata , Interferón Tipo I/farmacología , Macrófagos/metabolismo , Animales , Bases de Datos de Proteínas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Homeostasis , Humanos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Factor 3 de Genes Estimulados por el Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Modelos Lineales , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
7.
Nat Commun ; 10(1): 2921, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31266943

RESUMEN

Cells maintain the balance between homeostasis and inflammation by adapting and integrating the activity of intracellular signaling cascades, including the JAK-STAT pathway. Our understanding of how a tailored switch from homeostasis to a strong receptor-dependent response is coordinated remains limited. Here, we use an integrated transcriptomic and proteomic approach to analyze transcription-factor binding, gene expression and in vivo proximity-dependent labelling of proteins in living cells under homeostatic and interferon (IFN)-induced conditions. We show that interferons (IFN) switch murine macrophages from resting-state to induced gene expression by alternating subunits of transcription factor ISGF3. Whereas preformed STAT2-IRF9 complexes control basal expression of IFN-induced genes (ISG), both type I IFN and IFN-γ cause promoter binding of a complete ISGF3 complex containing STAT1, STAT2 and IRF9. In contrast to the dogmatic view of ISGF3 formation in the cytoplasm, our results suggest a model wherein the assembly of the ISGF3 complex occurs on DNA.


Asunto(s)
Regulación de la Expresión Génica , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferones/metabolismo , Factor de Transcripción STAT2/metabolismo , Animales , Femenino , Humanos , Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Células RAW 264.7 , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/genética , Transcripción Genética
8.
J Immunol ; 201(7): 2070-2081, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30143585

RESUMEN

IL-27 is a cytokine exerting pleiotropic immunomodulatory effects on a broad spectrum of immune cells. Optimal IL-27 production downstream of TLR3/4 ligand stimulation relies on autocrine type I IFN signaling, defining a first and second phase in IL-27 production. This work shows that IL-1 receptor-associated kinase 1 (IRAK1) limits TLR3/4- and IFNAR-induced IL-27 production. At the mechanistic level, we identified IRAK1 as a novel regulator of STAT1, IRF1, and IRF9. We found hyperactivation of STAT1 together with increased nuclear levels of IRF1 and IRF9 in IRAK1-deficient murine macrophages compared with control cells following stimulation with LPS and poly(I:C). IRAK1-deficient human microglial cells showed higher basal levels of STAT1 and STAT2 compared with control cells. Blocking the kinase activity of TBK1/IKKε in IRAK1 knockdown human microglial cells reduced the high basal levels of STAT1/2, uncovering a TBK1/IKKε kinase-dependent mechanism controlling basal levels of STAT1/2. Stimulating IRAK1 knockdown human microglial cells with IFN-ß led to increased IL-27p28 expression compared with control cells. In IRAK1-deficient murine macrophages, increased IL-27 levels were detected by ELISA following IFN-ß stimulation compared with control macrophages together with increased nuclear levels of p-STAT1, IRF1, and IRF9. Treatment of wild-type and IRAK1-deficient murine macrophages with fludarabine similarly reduced TLR3/4-induced IL-27 cytokine levels. To our knowledge, this work represents the first report placing IRAK1 in the IFNAR pathway and identifies IRAK1 as an important regulator of STAT1, controlling IL-27 production downstream of TLR3/4 and IFNAR signaling pathways.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-27/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Animales , Células HEK293 , Humanos , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/genética , Ratones , Fosforilación , Receptor de Interferón alfa y beta/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo
9.
Front Immunol ; 9: 1135, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29892288

RESUMEN

Interferon (IFN)-I and IFN-II both induce IFN-stimulated gene (ISG) expression through Janus kinase (JAK)-dependent phosphorylation of signal transducer and activator of transcription (STAT) 1 and STAT2. STAT1 homodimers, known as γ-activated factor (GAF), activate transcription in response to all types of IFNs by direct binding to IFN-II activation site (γ-activated sequence)-containing genes. Association of interferon regulatory factor (IRF) 9 with STAT1-STAT2 heterodimers [known as interferon-stimulated gene factor 3 (ISGF3)] or with STAT2 homodimers (STAT2/IRF9) in response to IFN-I, redirects these complexes to a distinct group of target genes harboring the interferon-stimulated response element (ISRE). Similarly, IRF1 regulates expression of ISGs in response to IFN-I and IFN-II by directly binding the ISRE or IRF-responsive element. In addition, evidence is accumulating for an IFN-independent and -dependent role of unphosphorylated STAT1 and STAT2, with or without IRF9, and IRF1 in basal as well as long-term ISG expression. This review provides insight into the existence of an intracellular amplifier circuit regulating ISG expression and controlling long-term cellular responsiveness to IFN-I and IFN-II. The exact timely steps that take place during IFN-activated feedback regulation and the control of ISG transcription and long-term cellular responsiveness to IFN-I and IFN-II is currently not clear. Based on existing literature and our novel data, we predict the existence of a multifaceted intracellular amplifier circuit that depends on unphosphorylated and phosphorylated ISGF3 and GAF complexes and IRF1. In a combinatorial and timely fashion, these complexes mediate prolonged ISG expression and control cellular responsiveness to IFN-I and IFN-II. This proposed intracellular amplifier circuit also provides a molecular explanation for the existing overlap between IFN-I and IFN-II activated ISG expression.


Asunto(s)
Retroalimentación Fisiológica , Factores Reguladores del Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/genética , Interferones/metabolismo , Animales , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Mutación de Línea Germinal , Humanos , Factores Reguladores del Interferón/metabolismo , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Mutación , Unión Proteica , Factor de Transcripción STAT1/química , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/química , Factor de Transcripción STAT2/metabolismo
10.
Mediators Inflamm ; 2017: 8302636, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28473732

RESUMEN

Glial activation and subsequent release of neurotoxic proinflammatory factors are believed to play an important role in the pathogenesis of several neurological disorders including Parkinson's disease (PD). Inhibition of glial activation and inflammatory processes may represent a therapeutic target to alleviate neurodegeneration. Securinine, a major natural alkaloid product from the root of the plant Securinega suffruticosa, has been reported to have potent biological activity and is used in the treatment of neurological conditions such as amyotrophic lateral sclerosis, poliomyelitis, and multiple sclerosis. In this study, we explored the underlying mechanisms of neuroprotection elicited by securinine, particularly its anti-inflammatory effects in glial cells. Our results demonstrate that securinine significantly and dose-dependently suppressed the nitric oxide production in microglia and astrocytic cultures. In addition, securinine inhibited the activation of the inflammatory mediator NF-κB, as well as mitogen-activated protein kinases in lipopolysaccharide- (LPS-) stimulated BV2 cells. Additionally, securinine also inhibited interferon-γ- (IFN-γ-) induced nitric oxide levels and iNOS mRNA expression. Furthermore, conditioned media (CM) from securinine pretreated BV2 cells significantly reduced mesencephalic dopaminergic neurotoxicity compared with CM from LPS stimulated microglia. These findings suggest that securinine may be a potential candidate for the treatment of neurodegenerative diseases related to neuroinflammation.


Asunto(s)
Azepinas/uso terapéutico , Compuestos Heterocíclicos de Anillo en Puente/uso terapéutico , Lactonas/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Piperidinas/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Astrocitos/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Enfermedad de Parkinson/inmunología , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa
11.
Mar Biotechnol (NY) ; 19(3): 310-319, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28500614

RESUMEN

Virus infection often causes large amounts of mortality during teleost larvae stage. Strong induction of innate immunity to increase survival rates of teleost larvae has been less reported. In this study, we present a zebrafish IRF9-Stat2 fusion protein (zIRF9-S2C) as a strong innate immunity inducer and characterized induction of interferon-stimulated genes (ISGs) in zebrafish larvae. zIRF9-S2C could mimic IFN-stimulated gene factor 3 (ISGF3) complex to constitutively activate transcription of Mx promoter through IFN-stimulatory element (ISRE) sites. Mutation of two ISRE sites on Mx promoter reduced transactivation activities of Mx promoter induced by zIRF9-S2C. An electrophoretic mobility shift assay experiment shows that zIRF9-S2C could directly bind to two ISRE sites of Mx promoter. Induction of transactivation of Mx promoter by zIRF9-S2C shows significantly higher activity than by zebrafish IFN1 (zIFN1), IFNγ (zIFNγ), and Tetraodon IRF9-S2C (TnIRF9-S2C). zIRF9-S2C raises transcription of Mxa, Mxb, Mxc, Ifnφ1, Ifnφ2, and Ifnφ3 in zebrafish liver ((ZFL) cell line) cells and zebrafish larvae. Collectively, we suggest that IRF9-S2C could activate transcription of ISGs with species-specific recognition and could be an innate immunity inducer in teleost larvae.


Asunto(s)
Inmunidad Innata , Proteínas de Resistencia a Mixovirus/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Factor de Transcripción STAT2/genética , Pez Cebra/metabolismo , Animales , Células COS , Chlorocebus aethiops , Regulación de la Expresión Génica , Factor 3 de Genes Estimulados por el Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Larva/genética , Larva/inmunología , Larva/metabolismo , Proteínas de Resistencia a Mixovirus/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Transducción de Señal , Activación Transcripcional , Pez Cebra/genética , Pez Cebra/inmunología
12.
Sci Signal ; 10(476)2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-28442624

RESUMEN

Interferon (IFN)-stimulated genes (ISGs) are antiviral effectors that are induced by IFNs through the formation of a tripartite transcription factor ISGF3, which is composed of IRF9 and phosphorylated forms of STAT1 and STAT2. However, we found that IFN-independent ISG expression was detectable in immortalized cell lines, primary intestinal and liver organoids, and liver tissues. The constitutive expression of ISGs was mediated by the unphosphorylated ISGF3 (U-ISGF3) complex, consisting of IRF9 together with unphosphorylated STAT1 and STAT2. Under homeostatic conditions, STAT1, STAT2, and IRF9 were found in the nucleus. Analysis of a chromatin immunoprecipitation sequencing data set revealed that STAT1 specifically bound to the promoters of ISGs even in the absence of IFNs. Knockdown of STAT1, STAT2, or IRF9 by RNA interference led to the decreased expression of various ISGs in Huh7.5 human liver cells, which was confirmed in mouse embryonic fibroblasts (MEFs) from STAT1-/-, STAT2-/-, or IRF9-/- mice. Furthermore, decreased ISG expression was accompanied by increased replication of hepatitis C virus and hepatitis E virus. Conversely, simultaneous overexpression of all ISGF3 components, but not any single factor, induced the expression of ISGs and inhibited viral replication; however, no phosphorylated STAT1 and STAT2 were detected. A phosphorylation-deficient STAT1 mutant was comparable to the wild-type protein in mediating the IFN-independent expression of ISGs and antiviral activity, suggesting that ISGF3 works in a phosphorylation-independent manner. These data suggest that the U-ISGF3 complex is both necessary and sufficient for constitutive ISG expression and antiviral immunity under homeostatic conditions.


Asunto(s)
Hepatitis C/prevención & control , Hepatitis E/prevención & control , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Animales , Antivirales/farmacología , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/virología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación de la Expresión Génica/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Hepatitis C/metabolismo , Hepatitis C/virología , Hepatitis E/metabolismo , Hepatitis E/virología , Virus de la Hepatitis E/efectos de los fármacos , Humanos , Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Interferón-alfa/farmacología , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/efectos de los fármacos , Intestinos/virología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/virología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT2/genética , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
13.
J Biol Chem ; 292(14): 5845-5859, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28213522

RESUMEN

Type I interferons (IFN-I) are critical in antimicrobial and antitumor defense. Although IFN-I signal via the interferon-stimulated gene factor 3 (ISGF3) complex consisting of STAT1, STAT2, and IRF9, IFN-I can mediate significant biological effects via ISGF3-independent pathways. For example, the absence of STAT1, STAT2, or IRF9 exacerbates neurological disease in transgenic mice with CNS production of IFN-I. Here we determined the role of IFN-I-driven, ISGF3-independent signaling in regulating global gene expression in STAT1-, STAT2-, or IRF9-deficient murine mixed glial cell cultures (MGCs). Compared with WT, the expression of IFN-α-stimulated genes (ISGs) was reduced in number and magnitude in MGCs that lacked STAT1, STAT2, or IRF9. There were significantly fewer ISGs in the absence of STAT1 or STAT2 versus in the absence of IRF9. The majority of ISGs regulated in the STAT1-, STAT2-, or IRF9-deficient MGCs individually were shared with WT. However, only a minor number of ISGs were common to WT and STAT1-, STAT2-, and IRF9-deficient MGCs. Whereas signal pathway activation in response to IFN-α was rapid and transient in WT MGCs, this was delayed and prolonged and correlated with increased numbers of ISGs expressed at 12 h versus 4 h of IFN-α exposure in all three IFN-I signaling-deficient MGCs. In conclusion, 1) IFN-I can mediate ISG expression in MGCs via ISGF3-independent signaling pathways but with reduced efficiency, with delayed and prolonged kinetics, and is more dependent on STAT1 and STAT2 than IRF9; and 2) signaling pathways not involving STAT1, STAT2, or IRF9 play a minor role only in mediating ISG expression in MGCs.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón-alfa/farmacología , Neuroglía/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Factor 3 de Genes Estimulados por el Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Ratones , Ratones Noqueados , Neuroglía/citología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT2/genética
14.
EMBO J ; 36(2): 202-212, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-27852626

RESUMEN

The transcription factor ISGF3, comprised of IRF9 and tyrosine-phosphorylated STATs 1 and 2, transmits the signal from the type I interferon receptor to the genome. We have discovered a novel phosphorylation of STAT2 on T387 that negatively regulates this response. In most untreated cell types, the majority of STAT2 is phosphorylated on T387 constitutively. In response to interferon-ß, the T387A mutant of STAT2 is much more effective than wild-type STAT2 in mediating the expression of many interferon-stimulated genes, in protecting cells against virus infection, and in inhibiting cell growth. Interferon-ß-treated cells expressing wild-type STAT2 contain much less ISGF3 capable of binding to an interferon-stimulated response element than do cells expressing T387A STAT2. T387 lies in a cyclin-dependent kinase (CDK) consensus sequence, and CDK inhibitors decrease T387 phosphorylation. Using CDK inhibitors to reverse the constitutive inhibitory phosphorylation of T387 of U-STAT2 might enhance the efficacy of type I interferons in many different clinical settings.


Asunto(s)
Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón beta/metabolismo , Procesamiento Proteico-Postraduccional , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Línea Celular , Humanos , Modelos Biológicos , Fosforilación
15.
Cancer Prev Res (Phila) ; 9(7): 567-80, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27080594

RESUMEN

Although our most recent studies have identified Isorhapontigenin (ISO), a novel derivative of stilbene that isolated from a Chinese herb Gnetum cleistostachyum, for its inhibition of human bladder cancer growth, nothing is known whether ISO possesses an inhibitory effect on bladder cancer invasion. Thus, we addressed this important question in current study and discovered that ISO treatment could inhibit mouse-invasive bladder cancer development following bladder carcinogen N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) exposure in vivo We also found that ISO suppressed human bladder cancer cell invasion accompanied by upregulation of the forkhead box class O 1 (FOXO1) mRNA transcription in vitro Accordingly, FOXO1 was profoundly downregulated in human bladder cancer tissues and was negatively correlated with bladder cancer invasion. Forced expression of FOXO1 specifically suppressed high-grade human bladder cancer cell invasion, whereas knockdown of FOXO1 promoted noninvasive bladder cancer cells becoming invasive bladder cancer cells. Moreover, knockout of FOXO1 significantly increased bladder cancer cell invasion and abolished the ISO inhibition of invasion in human bladder cancer cells. Further studies showed that the inhibition of Signal transducer and activator of transcription 1 (STAT1) phosphorylation at Tyr701 was crucial for ISO upregulation of FOXO1 transcription. Furthermore, this study revealed that metalloproteinase-2 (MMP-2) was a FOXO1 downstream effector, which was also supported by data obtained from mouse model of ISO inhibition BBN-induced mouse-invasive bladder cancer formation. These findings not only provide a novel insight into the understanding of mechanism of bladder cancer's propensity to invasion, but also identify a new role and mechanisms underlying the natural compound ISO that specifically suppresses such bladder cancer invasion through targeting the STAT1-FOXO1-MMP-2 axis. Cancer Prev Res; 9(7); 567-80. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Invasividad Neoplásica/patología , Transducción de Señal/efectos de los fármacos , Estilbenos/farmacología , Neoplasias de la Vejiga Urinaria/patología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Humanos , Factor 3 de Genes Estimulados por el Interferón/efectos de los fármacos , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
16.
Virology ; 485: 414-21, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342467

RESUMEN

To subvert host antiviral immune responses, many viruses have evolved countermeasures to inhibit IFN signaling pathway. Porcine bocavirus (PBoV), a newly identified porcine parvovirus, has received attention because it shows clinically high co-infection prevalence with other pathogens in post-weaning multisystemic wasting syndrome (PWMS) and diarrheic piglets. In this study, we screened the structural and non-structural proteins encoded by PBoV and found that the non-structural protein NP1 significantly suppressed IFN-stimulated response element (ISRE) activity and subsequent IFN-stimulated gene (ISG) expression. However, NP1 affected neither the activation and translocation of STAT1/STAT2, nor the formation of the heterotrimeric transcription factor complex ISGF3 (STAT1/STAT2/IRF9). Detailed analysis demonstrated that PBoV NP1 blocked the ISGF3 DNA-binding activity by combining with the DNA-binding domain (DBD) of IRF9. In summary, these results indicate that PBoV NP1 interferes with type I IFN signaling pathway by blocking DNA binding of ISGF3 to attenuate innate immune responses.


Asunto(s)
Bocavirus/fisiología , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferones/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Proteínas no Estructurales Virales/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , ADN/metabolismo , Humanos , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/química , Modelos Biológicos , Fosforilación , Unión Proteica , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Porcinos
17.
Cell Rep ; 12(2): 300-12, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26146080

RESUMEN

The transcriptional response to infection with the bacterium Listeria monocytogenes (Lm) requires cooperative signals of the type I interferon (IFN-I)-stimulated JAK-STAT and proinflammatory NF-κB pathways. Using ChIP-seq analysis, we define genes induced in Lm-infected macrophages through synergistic transcriptional activation by NF-κB and the IFN-I-activated transcription factor ISGF3. Using the Nos2 and IL6 genes as prime examples of this group, we show that NF-κB functions to recruit enzymes that establish histone marks of transcriptionally active genes. In addition, NF-κB regulates transcriptional elongation by employing the mediator kinase module for the recruitment of the pTEFb complex. ISGF3 has a major role in associating the core mediator with the transcription start as a prerequisite for TFIID and RNA polymerase II (Pol II) binding. Our data suggest that the functional cooperation between two major antimicrobial pathways is based on promoter priming by NF-κB and the engagement of the core mediator for Pol II binding by ISGF3.


Asunto(s)
Complejo Mediador/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción STAT/metabolismo , Animales , Sitios de Unión , Histona Acetiltransferasas/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Factor 3 de Genes Estimulados por el Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Listeria monocytogenes/fisiología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Polimerasa II/metabolismo , Transducción de Señal , Factor de Transcripción TFIID/metabolismo , Activación Transcripcional
18.
PLoS One ; 9(2): e89878, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587093

RESUMEN

Hepatic stellate cells (HSCs), following transdifferentiation to myofibroblasts plays a key role in liver fibrosis. Therefore, attempts to attenuate this myofibroblastic phenotype would be a promising therapeutic approach. Interferon gamma (IFNγ) is a potent anti-fibrotic cytokine, but its pleiotropic receptor expression leading to severe adverse effects has limited its clinical application. Since, activated HSC express high-level of platelet derived growth factor beta receptor (PDGFßR), we investigated the potential of PDGFßR-specific targeting of IFNγ and its signaling peptide that lacks IFNγR binding site (mimetic IFNγ or mimIFNγ) in liver fibrosis. We prepared DNA constructs expressing IFNγ, mimIFNγ or BiPPB (PDGFßR-specific bicyclic peptide)-IFNγ, BiPPB-mimIFNγ fusion proteins. Both chimeric proteins alongwith IFNγ and mimIFNγ were produced in E.coli. The expressed proteins were purified and analyzed for PDGFßR-specific binding and in vitro effects. Subsequently, these recombinant proteins were investigated for the liver uptake (pSTAT1α signaling pathway), for anti-fibrotic effects and adverse effects (platelet counts) in CCl4-induced liver fibrogenesis in mice. The purified HSC-targeted IFNγ and mimIFNγ fusion proteins showed PDGFßR-specific binding and significantly reduced TGFß-induced collagen-I expression in human HSC (LX2 cells), while mouse IFNγ and mimIFNγ did not show any effect. Conversely, mouse IFNγ and BiPPB-IFNγ induced activation and dose-dependent nitric oxide release in mouse macrophages (express IFNγR while lack PDGFßR), which was not observed with mimIFNγ and BiPPB-mimIFNγ, due to the lack of IFNγR binding sites. In vivo, targeted BiPPB-IFNγ and BiPPB-mimIFNγ significantly activated intrahepatic IFNγ-signaling pathway compared to IFNγ and mimIFNγ suggesting increased liver accumulation. Furthermore, the targeted fusion proteins ameliorated liver fibrogenesis in mice by significantly reducing collagen and α-SMA expression and potentiating collagen degradation. IFNγ also induced reduction in fibrogenesis but showed significant decrease in platelet counts, which was restored with targeted proteins. These results suggest that these rationally designed proteins can be further developed as novel anti-fibrotic therapeutics.


Asunto(s)
Interferón gamma/metabolismo , Cirrosis Hepática/prevención & control , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Análisis de Varianza , Animales , Tetracloruro de Carbono/toxicidad , Células Cultivadas , Cartilla de ADN/genética , Escherichia coli , Técnica del Anticuerpo Fluorescente , Células Estrelladas Hepáticas/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón gamma/genética , Cirrosis Hepática/inducido químicamente , Ratones , Óxido Nítrico/metabolismo , Plásmidos/genética , Recuento de Plaquetas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/genética
19.
Arch Pharm Res ; 37(9): 1117-23, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24395532

RESUMEN

Rheum palmatum, Chinese traditional herb, exhibits a great variety of anti-cancer and anti-viruses properties. This study rates antiviral activity of R. palmatum extracts and its components against Japanese encephalitis virus (JEV) in vitro. Methanol extract of R. palmatum contained higher levels of aloe emodin, chrysophanol, rhein, emodin and physcion than water extract. Methanol extract (IC50 = 15.04 µg/ml) exhibited more potent inhibitory effects on JEV plaque reduction than water extract (IC50 = 51.41 µg/ml). Meanwhile, IC50 values determined by plaque reduction assay were 15.82 µg/ml for chrysophanol and 17.39 µg/ml for aloe-emodin, respectively. Virucidal activity of agents correlated with anti-JEV activity, while virucidal IC50 values were 7.58 µg/ml for methanol extract, 17.36 µg/ml for water extract, 0.75 µg/ml for chrysophanol and 0.46 µg/ml for aloe-emodin, respectively. In addition, 10 µg/ml of extract, chrysophanol or aloe emodin caused 90 % inhibition of JEV yields in cells and significantly activated gamma activated sequence-driven promoters. Hence, methanol extract of R. palmatum and chrysophanol with high therapeutic index might be useful for development of antiviral agents against JEV.


Asunto(s)
Antraquinonas/farmacología , Antivirales/farmacología , Medicamentos Herbarios Chinos/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Rheum/química , Replicación Viral/efectos de los fármacos , Animales , Antivirales/aislamiento & purificación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/aislamiento & purificación , Virus de la Encefalitis Japonesa (Especie)/crecimiento & desarrollo , Virus de la Encefalitis Japonesa (Especie)/fisiología , Etnofarmacología , Genes Reporteros/efectos de los fármacos , Concentración 50 Inhibidora , Factor 3 de Genes Estimulados por el Interferón/agonistas , Factor 3 de Genes Estimulados por el Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Luciferasas de Luciérnaga/química , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Mesocricetus , Metanol/química , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Solventes/química
20.
Nat Immunol ; 15(2): 168-76, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24413774

RESUMEN

STAT1 is an indispensable component of a heterotrimer (ISGF3) and a STAT1 homodimer (GAF) that function as transcription regulators in type 1 and type 2 interferon signaling, respectively. To investigate the importance of STAT1-cooperative DNA binding, we generated gene-targeted mice expressing cooperativity-deficient STAT1 with alanine substituted for Phe77. Neither ISGF3 nor GAF bound DNA cooperatively in the STAT1F77A mouse strain, but type 1 and type 2 interferon responses were affected differently. Type 2 interferon-mediated transcription and antibacterial immunity essentially disappeared owing to defective promoter recruitment of GAF. In contrast, STAT1 recruitment to ISGF3 binding sites and type 1 interferon-dependent responses, including antiviral protection, remained intact. We conclude that STAT1 cooperativity is essential for its biological activity and underlies the cellular responses to type 2, but not type 1 interferon.


Asunto(s)
Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Proteínas Mutantes/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Células Cultivadas , ADN/metabolismo , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Mutantes/genética , Unión Proteica/genética , Ingeniería de Proteínas , Factor de Transcripción STAT1/genética , Transducción de Señal/genética , Transgenes/genética , Virus de la Estomatitis Vesicular Indiana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...