Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 208(4): 881-897, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35101891

RESUMEN

Diet plays an important role in lifestyle disorders associated with the disturbed immune system. During the study of methionine- and choline-deficient diet-induced nonalcoholic fatty liver disease, we observed a specific decrease in the plasmacytoid dendritic cell (pDC) fraction from murine spleens. While delineating the role for individual components, we identified that l-methionine supplementation correlates with representation of the pDC fraction. S-adenosylmethionine (SAM) is a key methyl donor, and we demonstrate that supplementation of methionine-deficient medium with SAM but not homocysteine reverses the defect in pDC development. l-Methionine has been implicated in maintenance of methylation status in the cell. Based on our observed effect of SAM and zebularine on DC subset development, we sought to clarify the role of DNA methylation in pDC biology. Whole-genome bisulfite sequencing analysis from the splenic DC subsets identified that pDCs display differentially hypermethylated regions in comparison with classical DC (cDC) subsets, whereas cDC1 and cDC2 exhibited comparable methylated regions, serving as a control in our study. We validated differentially methylated regions in the sorted pDC, CD8α+ cDC1, and CD4+ cDC2 subsets from spleens as well as FL-BMDC cultures. Upon analysis of genes linked with differentially methylated regions, we identified that differential DNA methylation is associated with the MAPK pathway such that its inhibition guides DC development toward the pDC subtype. Overall, our study identifies an important role for methionine in pDC biology.


Asunto(s)
Colina/metabolismo , Metilación de ADN , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Dieta , Metionina/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Biología Computacional/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Homeostasis , Inmunidad Innata , Inmunofenotipificación , Sistema de Señalización de MAP Quinasas , Metionina/deficiencia , Ratones , Mapeo de Interacción de Proteínas , Transcriptoma
2.
J Virol ; 95(21): e0040621, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34379515

RESUMEN

Interferon regulatory factor 8 (IRF8), a myeloid lineage transcription factor, emerges as an essential regulator for microglial activation. However, the precise role of IRF8 during Japanese encephalitis virus (JEV) infection in the brain remains elusive. Here, we report that JEV infection enhances IRF8 expression in the infected mouse brain. Comparative transcriptional profiling of whole-brain RNA analysis and validation by quantitative reverse transcription-PCR (qRT-PCR) reveals an impaired interferon gamma (IFN-γ) and related gene expression in Irf8 knockout (Irf8-/-)-infected mice. Further, Ifnγ knockout (Ifnγ-/-) mice exhibit a reduced level of Irf8. Both Ifnγ-/- and Irf8-/- mice exhibit significantly reduced levels of activated (CD11b+ CD45hi, CD11b+ CD45lo, Cd68, and CD86) and infiltrating immune cells (Ly6C+, CD4, and CD8) in the infected brain compared to those of wild-type (WT) mice. However, a higher level of granulocyte cell (Ly6G+) infiltration is evident in Irf8-/- mice as well as the increased concentration of tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), monocyte chemoattractant protein 1 (MCP1) levels in the brain. Interestingly, neither the Irf8-/- nor the Ifnγ-/- conferred protection against lethal JEV challenge to mice and exhibit augmentation in JEV replication in the brain. The gain of function of Irf8 by overexpressing functional IRF8 in an IRF8-deficient cell line attenuates viral replication and enhances IFN-γ production. Overall, we summarize that in the murine model of JEV encephalitis, IRF8 modulation affects JEV replication. We also show that lack of Irf8 affects immune cell abundance in circulation and the infected brain, leading to a reduction in IFN-γ level and increased viral load in the brain. IMPORTANCE Microglial cells, the resident macrophages in the brain, play a vital role in Japanese encephalitis virus (JEV) pathogenesis. The deregulated activity of microglia can be lethal for the brain. Therefore, it is crucial to understand the regulators that drive microglia phenotype changes and induce inflammation in the brain. Interferon regulatory factor 8 (IRF8) is a myeloid lineage transcription factor involved in microglial activation. However, the impact of IRF8 modulation on JEV replication remains elusive. Moreover, the pathways regulated by IRF8 to initiate and amplify pathological neuroinflammation are not well understood. Here, we demonstrated the effect of IRF8 modulation on JEV replication, microglial activation, and immune cells infiltration in the brain.


Asunto(s)
Encéfalo/virología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Factores Reguladores del Interferón/genética , Interferón gamma/inmunología , Replicación Viral/inmunología , Animales , Encéfalo/inmunología , Virus de la Encefalitis Japonesa (Especie)/fisiología , Femenino , Regulación de la Expresión Génica/inmunología , Factores Reguladores del Interferón/inmunología , Interferón gamma/genética , Masculino , Ratones , Ratones Noqueados , Microglía/inmunología , Microglía/fisiología , Microglía/virología , Transducción de Señal
3.
Cell Immunol ; 371: 104468, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34968772

RESUMEN

Dendritic cells (DCs) play central role in innate as well as adaptive immune responses regulated by diverse DC subtypes that vary in terms of surface markers, transcriptional profile and functional responses. Generation of DC diversity from progenitor stage is tightly regulated by complex molecular inter-play between transcription factors. We earlier demonstrated that Batf3 and Id2 expression have a synergistic effect on the Irf8 directed classical cDC1 development. In present study, Bi-molecular fluorescence complementation assay suggested that IRF8 interacts with BATF3, and ID2 may aid cDC1 development independently. Genome wide recruitment analysis of IRF8 and BATF3 from different DC subtypes led to identification of the overlapping regions of occupancy by these two transcription factors. Further analysis of overlapping peaks of IRF8 and BATF3 occupancy in promoter region within the cDC1 subtype specific transcriptional pattern identified a metabolically important Pfkfb3 gene. Among various immune cell types; splenic cDC1 subtype displayed enhanced expression of Pfkfb3. Analysis of Irf8-/-, Irf8R294C and Batf3DCKO DC confirmed direct regulation of Pfkfb3 enhanced expression specifically in cDC1 subtype. Further we show that inhibition of PFKFB3 enzymatic activity by a chemical agent PFK15 led to reduction in cDC1 subtype in both in vitro FLDC cultures as well as in vivo mouse spleens. Together, our study identified the direct regulation of cDC1 specific enhanced expression of Pfkfb3 in glycolysis and cDC1 biology.


Asunto(s)
Células Dendríticas/inmunología , Factores Reguladores del Interferón/metabolismo , Fosfofructoquinasa-2/biosíntesis , Proteínas Represoras/metabolismo , Animales , Línea Celular , Femenino , Regulación de la Expresión Génica/genética , Glucólisis/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Fosfofructoquinasa-2/antagonistas & inhibidores , Fosfofructoquinasa-2/genética , Regiones Promotoras Genéticas/genética , Piridinas/farmacología , Quinolinas/farmacología
4.
PLoS Comput Biol ; 17(9): e1009384, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34516563

RESUMEN

Apart from the canonical fingers, palm and thumb domains, the RNA dependent RNA polymerases (RdRp) from the viral order Nidovirales possess two additional domains. Of these, the function of the Nidovirus RdRp associated nucleotidyl transferase domain (NiRAN) remains unanswered. The elucidation of the 3D structure of RdRp from the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), provided the first ever insights into the domain organisation and possible functional characteristics of the NiRAN domain. Using in silico tools, we predict that the NiRAN domain assumes a kinase or phosphotransferase like fold and binds nucleoside triphosphates at its proposed active site. Additionally, using molecular docking we have predicted the binding of three widely used kinase inhibitors and five well characterized anti-microbial compounds at the NiRAN domain active site along with their drug-likeliness. For the first time ever, using basic biochemical tools, this study shows the presence of a kinase like activity exhibited by the SARS-CoV-2 RdRp. Interestingly, a well-known kinase inhibitor- Sorafenib showed a significant inhibition and dampened viral load in SARS-CoV-2 infected cells. In line with the current global COVID-19 pandemic urgency and the emergence of newer strains with significantly higher infectivity, this study provides a new anti-SARS-CoV-2 drug target and potential lead compounds for drug repurposing against SARS-CoV-2.


Asunto(s)
Antivirales/farmacología , ARN Polimerasa Dependiente de ARN de Coronavirus/antagonistas & inhibidores , Dominios Proteicos , SARS-CoV-2/efectos de los fármacos , Dominio Catalítico , Simulación por Computador , ARN Polimerasa Dependiente de ARN de Coronavirus/química , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , Humanos
5.
Mol Pharm ; 18(10): 3832-3842, 2021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34499836

RESUMEN

Use of tumor-associated antigens for cancer immunotherapy is limited due to their poor in vivo stability and low cellular uptake. Delivery of antigenic peptides using synthetic polymer-based nanostructures has been actively pursued but with limited success. Peptide-based nanostructures hold much promise as delivery vehicles due to their easy design and synthesis and inherent biocompatibility. Here, we report self-assembly of a dipeptide containing a non-natural amino acid, α,ß-dehydrophenylalanine (ΔF), into nanotubes, which efficiently entrapped a MAGE-3-derived peptide (M3). M3 entrapped in F-ΔF nanotubes was more stable to a nonspecific protease treatment and both F-ΔF and F-ΔF-M3 showed no cellular toxicity for four cancerous and noncancerous cell lines used. F-ΔF-M3 showed significantly higher cellular uptake in RAW 267.4 macrophage cells compared to M3 alone and also induced in vitro maturation of dendritic cells (DCs). Immunization of mice with F-ΔF-M3 selected a higher number of IFN-γ secreting CD8+ T cells and CD4+ T compared to M3 alone. On day 21, a tumor growth inhibition ratio (TGI, %) of 41% was observed in a murine melanoma model. These results indicate that F-ΔF nanotubes are highly biocompatible, efficiently delivered M3 to generate cytotoxic T lymphocytes responses, and able to protect M3 from degradation under in vivo conditions. The F-ΔF dipeptide-based nanotubes may be considered as a good platform for further development as delivery agents.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Sistema de Administración de Fármacos con Nanopartículas/administración & dosificación , Testículo/inmunología , Animales , Humanos , Inmunoterapia/métodos , Células MCF-7 , Masculino , Melanoma/inmunología , Melanoma/terapia , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Nanotubos de Péptidos , Trasplante de Neoplasias , Células RAW 264.7
6.
Cell Immunol ; 349: 104043, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32044112

RESUMEN

Type I Interferon (IFN) signaling plays a critical role in dendritic cell (DC) development and functions. Inhibition of hyper type I IFN signaling promotes cDC2 subtype development. Relb is essential to development of cDC2 subtype and here we analyzed its effect on type I IFN signaling in DCs. We show that Relb suppresses the homeostatic type I IFN signaling in cDC2 cultures. TLR stimulation of FL-DCs led to RelB induction coinciding with fall in IFN signatures; conforming with the observation Relb expression reduced TLR stimulated IFN induction along with decrease in ISGs. Towards understanding mechanism, we show that effects of RelB are mediated by increased levels of IκBα. We demonstrate that RelB dampened antiviral responses by lowering ISG levels and the defect in cDC2 development in RelB null mice can be rescued in Ifnar1-/- background. Overall, we propose a novel role of RelB as a negative regulator of the type I IFN signaling pathway; fine tuning development of cDC2 subtype.


Asunto(s)
Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Inhibidor NF-kappaB alfa/fisiología , Factor de Transcripción ReIB/fisiología , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Células Cultivadas , Cruzamientos Genéticos , Células Dendríticas/clasificación , Células Dendríticas/citología , Regulación de la Expresión Génica/inmunología , Ratones , Células 3T3 NIH , Virus de la Enfermedad de Newcastle/inmunología , Péptidos/farmacología , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/inmunología , Bazo/citología , Factor de Transcripción ReIB/deficiencia , Factor de Transcripción ReIB/genética , Carga Viral
7.
Immunity ; 31(6): 941-52, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-20064451

RESUMEN

Interleukin-21 (IL-21) is a pleiotropic cytokine that induces expression of transcription factor BLIMP1 (encoded by Prdm1), which regulates plasma cell differentiation and T cell homeostasis. We identified an IL-21 response element downstream of Prdm1 that binds the transcription factors STAT3 and IRF4, which are required for optimal Prdm1 expression. Genome-wide ChIP-Seq mapping of STAT3- and IRF4-binding sites showed that most regions with IL-21-induced STAT3 binding also bound IRF4 in vivo and furthermore revealed that the noncanonical TTCnnnTAA GAS motif critical in Prdm1 was broadly used for STAT3 binding. Comparing genome-wide expression array data to binding sites revealed that most IL-21-regulated genes were associated with combined STAT3-IRF4 sites rather than pure STAT3 sites. Correspondingly, ChIP-Seq analysis of Irf4(-/-) T cells showed greatly diminished STAT3 binding after IL-21 treatment, and Irf4(-/-) mice showed impaired IL-21-induced Tfh cell differentiation in vivo. These results reveal broad cooperative gene regulation by STAT3 and IRF4.


Asunto(s)
Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Interleucinas/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/genética , Animales , Linfocitos B/inmunología , Secuencia de Bases , Sitios de Unión , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Estudio de Asociación del Genoma Completo , Factores Reguladores del Interferón/genética , Intrones , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Factor de Transcripción STAT3/genética
8.
J Immunol ; 197(4): 1029-34, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27421479

RESUMEN

Dendritic cells (DCs) are a collection of different subtypes, each of which is characterized by specific surface markers, gene-expression patterns, and distinct functions. Members of the IFN regulatory factor family play critical roles in DC development and functions. Recently, Irf8 was shown to activate TGF-ß signaling, which led to exacerbated neuroinflammation in the experimental autoimmune encephalomyelitis mouse model. We analyzed the effect of Irf8 on TGF-ß/bone morphogenetic protein pathway-specific genes in DCs and identified Acvrl1, a type I TGF-ß superfamily receptor, as a gene strongly induced by Irf8 expression. Among various DC subtypes, Acvrl1 is differentially expressed in CD8α(+) DCs. ACVRL1 signaling augmented Irf8-directed classical CD8α(+) DC development. Irf8 expression is essential for plasmacytoid DC and CD8α(+) DC development, and this study demonstrates that ACVRL1 signaling plays a pivotal role whereby it suppresses plasmacytoid DC development while enhancing that of CD8α(+) DCs, thus contributing to DC diversity development.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Transducción de Señal/inmunología , Receptores de Activinas Tipo I/inmunología , Receptores de Activinas Tipo II , Animales , Antígenos CD8/inmunología , Células Dendríticas/metabolismo , Factores Reguladores del Interferón/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
9.
J Immunol ; 191(12): 5993-6001, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24227775

RESUMEN

Dendritic cells (DCs) are heterogeneous cell populations represented by different subtypes, each varying in terms of gene expression patterns and specific functions. Recent studies identified transcription factors essential for the development of different DC subtypes, yet molecular mechanisms for the developmental program and functions remain poorly understood. In this study, we developed and characterized a mouse DC progenitor-like cell line, designated DC9, from Irf8(-/-) bone marrow cells as a model for DC development and function. Expression of Irf8 in DC9 cells led to plasmacytoid DCs and CD8α(+) DC-like cells, with a concomitant increase in plasmacytoid DC- and CD8α(+) DC-specific gene transcripts and induction of type I IFNs and IL12p40 following TLR ligand stimulation. Irf8 expression in DC9 cells led to an increase in Id2 and Batf3 transcript levels, transcription factors shown to be important for the development of CD8α(+) DCs. We show that, without Irf8, expression of Id2 and Batf3 was not sufficient for directing classical CD8α(+) DC development. When coexpressed with Irf8, Batf3 and Id2 had a synergistic effect on classical CD8α(+) DC development. We demonstrate that Irf8 is upstream of Batf3 and Id2 in the classical CD8α(+) DC developmental program and define the hierarchical relationship of transcription factors important for classical CD8α(+) DC development.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Células Dendríticas/citología , Regulación de la Expresión Génica/inmunología , Proteína 2 Inhibidora de la Diferenciación/fisiología , Factores Reguladores del Interferón/fisiología , Proteínas Represoras/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/biosíntesis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Antígenos CD8/análisis , Diferenciación Celular/efectos de los fármacos , Línea Celular , Dendritas/ultraestructura , Células Dendríticas/química , Células Dendríticas/clasificación , Células Dendríticas/ultraestructura , Células Madre Hematopoyéticas/citología , Proteína 2 Inhibidora de la Diferenciación/biosíntesis , Proteína 2 Inhibidora de la Diferenciación/genética , Factores Reguladores del Interferón/biosíntesis , Factores Reguladores del Interferón/genética , Interferón-alfa/biosíntesis , Interferón-alfa/genética , Subunidad p40 de la Interleucina-12/biosíntesis , Subunidad p40 de la Interleucina-12/genética , Proteínas de la Membrana/farmacología , Ratones , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transducción Genética
10.
J Immunol ; 189(7): 3548-56, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22942423

RESUMEN

Macrophages, when activated by IFN-γ and TLR signaling, elicit innate immune responses. IFN regulatory factor 8 (IRF8) is a transcription factor that facilitates macrophage activation and innate immunity. We show that, in resting macrophages, some IRF8 is conjugated to small ubiquitin-like modifiers (SUMO) 2/3 through the lysine residue 310. SUMO3-conjugated IRF8 failed to induce IL12p40 and other IRF8 target genes, consistent with SUMO-mediated transcriptional repression reported for other transcription factors. SUMO3-conjugated IRF8 showed reduced mobility in live nuclei and bound poorly to the IL12p40 gene. However, macrophage activation caused a sharp reduction in the amount of SUMOylated IRF8. This reduction coincided with the induction of a deSUMOylating enzyme, sentrin-specific peptidase 1 (SENP1), in activated macrophages. In transfection analysis, SENP1 removed SUMO3 from IRF8 and enhanced expression of IL12p40 and other target genes. Conversely, SENP1 knockdown repressed IRF8 target gene expression. In parallel with IRF8 deSUMOylation, macrophage activation led to the induction of proteins active in the SUMO pathway and caused a global shift in nuclear protein SUMOylation patterns. Together, the IRF8 SUMO conjugation/deconjugation switch is part of a larger transition in SUMO modifications that takes place upon macrophage activation, serving as a mechanism to trigger innate immune responses.


Asunto(s)
Endopeptidasas/fisiología , Factores Reguladores del Interferón/metabolismo , Activación de Macrófagos/inmunología , Animales , Células Cultivadas , Cisteína Endopeptidasas , Células HEK293 , Humanos , Factores Reguladores del Interferón/fisiología , Subunidad p40 de la Interleucina-12/metabolismo , Lisina/metabolismo , Macrófagos/citología , Macrófagos/enzimología , Macrófagos/inmunología , Ratones , Células 3T3 NIH , Unión Proteica/inmunología , Proteínas Represoras/fisiología , Fase de Descanso del Ciclo Celular/inmunología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación/inmunología , Ubiquitinas/metabolismo
11.
Int J Biol Macromol ; 229: 515-528, 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36584781

RESUMEN

Given the paucity of antiviral treatments for monkeypox disease, caused by the Monkeypox virus (MPXV), there is a pressing need for the development/identification of new drugs to treat the infection. MPXV possesses a linear dsDNA genome that is replicated by a DNA replication complex of which DNA polymerase (DPol) forms an important component. Owing to the importance of DPol in the viral life cycle, identifying/designing small molecules abolishing its function could yield new antivirals. In this study, we first used the AlphaFold artificial intelligence program to model the 3D structure of the MPXV DPol; like the fold of DPol from other organisms, the MPXV DPol structure has the characteristic exonuclease, thumb, palm, and fingers sub-domains arrangement. Subsequently, we have identified several inhibitors through virtual screening of ZINC and antiviral libraries. Molecules with phenyl scaffold along with alanine-based and tetrazole-based molecules showed the best docking score of -8 to -10 kcal/mol. These molecules bind in the palm and fingers sub-domains interface region, which partially overlaps with the DNA binding path. The delineation of DPol/inhibitor interactions showed that majorly active site residues ASP549, ASP753, TYR550, ASN551, SER552, and ASN665 interact with the inhibitors. These compounds exhibit good Absorption, Distribution, Metabolism and Excretion properties.


Asunto(s)
Monkeypox virus , Mpox , Humanos , Monkeypox virus/genética , Inteligencia Artificial , Mpox/diagnóstico , ADN Polimerasa Dirigida por ADN , Antivirales/farmacología
12.
J Biol Chem ; 286(10): 7893-7904, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21216962

RESUMEN

Dendritic cells (DCs) are key mediators of immune function through robust and tightly regulated presentation of antigen in the context of the MHC Class II. MHC Class II expression is controlled by the transactivator CIITA. CIITA expression in conventional DCs is uniquely dependent on an uncharacterized myeloid cell-specific promoter, CIITApI. We now identify in vivo the promoter structure and factors regulating CIITApI. In immature DCs transcription requires binding of PU.1, IRF8, NFκB, and Sp1 to the promoter. PU.1 binds independently at one site and in a required heterodimer with IRF8 at a composite element. DCs from IRF8-null mice have an unoccupied CIITApI promoter that can be rescued by reconstitution with IRF8 in vitro. Furthermore, mutation of either PU.1 site or the IFR8 site inhibits transcriptional activation. In vivo footprinting and chromatin immunoprecipitation reveals that DC maturation induces complete disassociation of the bound activators paralleled by recruitment of PRDM1/Blimp-1 to the promoter. PRDM1 is a transcriptional repressor with essential roles in B cells, T cells, NK cells, and DCs. We show that PRDM1 co-repressors, G9a and HDAC2, are recruited to CIITApI, leading to a loss of histone acetylation and acquisition of histone H3K9 dimethylation and heterochromatin protein 1γ (HP1γ). PRDM1 binding also blocks IRF8-mediated activation dependent on the PU.1/IRF composite element. Together these findings reveal the mechanisms regulating CIITA and, thus, antigen presentation in DCs, demonstrating that PRDM1 and IRF8/PU.1 counter-regulate expression. The activity of PRDM1 in silencing all three cell type-specific CIITA promoters places it as a central regulator of antigen presentation.


Asunto(s)
Presentación de Antígeno/fisiología , Células Dendríticas/metabolismo , Factores Reguladores del Interferón/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Animales , Células Dendríticas/citología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad/inmunología , Antígenos de Histocompatibilidad/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/inmunología , Histona Desacetilasa 2/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/inmunología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/inmunología , Histonas/metabolismo , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Elementos de Respuesta/fisiología , Transactivadores/genética , Transactivadores/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Transcripción Genética/fisiología
13.
J Clin Invest ; 118(2): 545-59, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18188453

RESUMEN

The mechanisms underlying the susceptibility of individuals with caspase recruitment domain 15 (CARD15) mutations and corresponding abnormalities of nucleotide-binding oligomerization domain 2 (NOD2) protein to Crohn disease are still poorly understood. One possibility is based on previous studies showing that muramyl dipeptide (MDP) activation of NOD2 negatively regulates TLR2 responses and that absence of such regulation leads to heightened Th1 responses. We now report that administration of MDP protects mice from the development of experimental colitis by downregulating multiple TLR responses, not just TLR2. The basis of these in vivo findings was suggested by in vitro studies of DCs, in which we showed that prestimulation of cells with MDP reduces cytokine responses to multiple TLR ligands and this reduction is dependent on enhanced IFN regulatory factor 4 (IRF4) activity. Further studies of mouse models of colitis showed that this inhibitory role of IRF4 does in fact apply to MDP-mediated protection from colitis, since neither IRF4-deficient mice nor mice treated with siRNA specific for IRF4 were protected. These findings indicate that MDP activation of NOD2 regulates innate responses to intestinal microflora by downregulating multiple TLR responses and suggest that the absence of such regulation leads to increased susceptibility to Crohn disease.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Colitis/prevención & control , Enfermedad de Crohn/inmunología , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Colitis/inducido químicamente , Colitis/inmunología , Enfermedad de Crohn/tratamiento farmacológico , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Inmunidad Innata/efectos de los fármacos , Factores Reguladores del Interferón/antagonistas & inhibidores , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Ligandos , Ratones , Ratones Mutantes , Proteína Adaptadora de Señalización NOD2/genética , ARN Interferente Pequeño/farmacología , Receptores Toll-Like/antagonistas & inhibidores , Ácido Trinitrobencenosulfónico/toxicidad
14.
Front Immunol ; 12: 758190, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867997

RESUMEN

Plasmacytoid dendritic cells (pDCs) are the key producers of type I interferons (IFNs), thus playing a central role in initiating antiviral immune response. Besides robust type I IFN production, pDCs also act as antigen presenting cells post immunogenic stimulation. Transcription factor Irf8 is indispensable for the development of both pDC and cDC1 subset. However, the mechanism underlying the differential regulation by IRF8 in cDC1- and pDC-specific genomic architecture of developmental pathways still remains to be fully elucidated. Previous studies indicated that the Irf8R294C mutation specifically abrogates development of cDC1 without affecting that of pDC. In the present study using RNA-seq based approach, we have found that though the point mutation Irf8R294C did not affect pDC development, it led to defective type I IFN production, thus resulting in inefficient antiviral response. This observation unraveled the distinctive roles of IRF8 in these two subpopulations-regulating the development of cDC1 whereas modulating the functionality of pDCs without affecting development. We have reported here that Irf8R294C mutation also caused defect in production of ISGs as well as defective upregulation of costimulatory molecules in pDCs in response to NDV infection (or CpG stimulation). Through in vivo studies, we demonstrated that abrogation of type I IFN production was concomitant with reduced upregulation of costimulatory molecules in pDCs and increased NDV burden in IRF8R294C mice in comparison with wild type, indicating inefficient viral clearance. Further, we have also shown that Irf8R294C mutation abolished the activation of type I IFN promoter by IRF8, justifying the low level of type I IFN production. Taken together, our study signifies that the single point mutation in Irf8, Irf8R294C severely compromised type I IFN-mediated immune response by murine pDCs, thereby causing impairment in antiviral immunity.


Asunto(s)
Células Dendríticas/inmunología , Factores Reguladores del Interferón/genética , Interferón Tipo I/inmunología , Mutación Missense , Enfermedad de Newcastle/inmunología , Mutación Puntual , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Islas de CpG/inmunología , Células Dendríticas/metabolismo , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunidad Innata , Factores Reguladores del Interferón/inmunología , Interferón Tipo I/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Enfermedad de Newcastle , Osteosarcoma/patología , Transcriptoma
15.
Blood ; 112(10): 4028-38, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18799728

RESUMEN

PU.1, IKAROS, E2A, EBF, and PAX5 comprise a transcriptional network that orchestrates B-cell lineage specification, commitment, and differentiation. Here we identify interferon regulatory factor 8 (IRF8) as another component of this complex, and show that it also modulates lineage choice by hematopoietic stem cells (HSCs). IRF8 binds directly to an IRF8/Ets consensus sequence located in promoter regions of Sfpi1 and Ebf1, which encode PU.1 and EBF, respectively, and is associated with transcriptional repression of Sfpi1 and transcriptional activation of Ebf1. Bone marrows of IRF8 knockout mice (IRF8(-/-)) had significantly reduced numbers of pre-pro-B cells and increased numbers of myeloid cells. Although HSCs of IRF8(-/-) mice failed to differentiate to B220(+) B-lineage cells in vitro, the defect could be rescued by transfecting HSCs with wild-type but not with a signaling-deficient IRF8 mutant. In contrast, overexpression of IRF8 in HSC-differentiated progenitor cells resulted in growth inhibition and apoptosis. We also found that IRF8 was expressed at higher levels in pre-pro-B cells than more mature B cells in wild-type mice. Together, these results indicate that IRF8 modulates lineage choice by HSCs and is part of the transcriptional network governing B-cell lineage specification, commitment, and differentiation.


Asunto(s)
Linfocitos B/metabolismo , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Factores Reguladores del Interferón/metabolismo , Elementos de Respuesta/fisiología , Transducción de Señal/fisiología , Animales , Linfocitos B/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Madre Hematopoyéticas/química , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Factores Reguladores del Interferón/genética , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Noqueados , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Unión Proteica/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
16.
iScience ; 23(7): 101322, 2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-32688283

RESUMEN

Precise regulation of innate immunity is crucial for development of appropriate host immunity against microbial infections and maintenance of immune homeostasis. MicroRNAs are small non-coding RNAs, post-transcriptional regulator of multiple genes, and act as a rheostat for protein expression. Here, we identified microRNA-30e-5p induced by hepatitis B virus and other viruses that act as a master regulator for innate immunity. Moreover, pegylated interferons treatment of patients with HBV for viral reduction also reduces miRNA. Additionally, we have also shown the immuno-pathological effects of miR-30e in patients with systemic lupus erythematosus (SLE) and mouse model. Mechanistically, miR-30e targets multiple negative regulators of innate immune signaling and enhances immune responses. Furthermore, sequestering of miR-30e in patients with SLE and mouse model significantly reduces type-I interferon and pro-inflammatory cytokines. Collectively, our study demonstrates the novel role of miR-30e in innate immunity and its prognostic and therapeutic potential in infectious and autoimmune diseases.

17.
Mol Cell Biol ; 24(10): 4476-86, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15121865

RESUMEN

The elicitation of cellular antiviral activities is dependent on the rapid transcriptional activation of interferon (IFN) target genes. It is not clear how the interferon target promoters, which are organized into chromatin structures in cells, rapidly respond to interferon or viral stimulation. In this report, we show that alpha IFN (IFN-alpha) treatment of HeLa cells induced hundreds of genes. The induction of the majority of these genes was inhibited when one critical subunit of the chromatin-remodeling SWI/SNF-like BAF complexes, BAF47, was knocked down via RNA interference. Inhibition of BAF47 blocked the cellular response to viral infection and impaired cellular antiviral activity by inhibiting many IFN- and virus-inducible genes. We show that the BAF complex was required to mediate both the basal-level expression and the rapid induction of the antiviral genes. Further analyses indicated that the BAF complex primed some IFN target promoters by utilizing ATP-derived energy to maintain the chromatin in a constitutively open conformation, allowing faster and more potent induction after IFN-alpha treatment. We propose that constitutive binding of the BAF complex is an important mechanism for the IFN-inducible promoters to respond rapidly to IFN and virus stimulation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Antígenos de Diferenciación , Antivirales/metabolismo , Secuencia de Bases , Línea Celular , Proteínas Cromosómicas no Histona , ADN Helicasas , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Interferón Tipo I/farmacología , Proteínas de la Membrana/genética , Modelos Biológicos , Virus de la Enfermedad de Newcastle/patogenicidad , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Proteínas Recombinantes , Proteína SMARCB1 , Transducción de Señal , Factores de Transcripción/metabolismo
18.
Cell Death Dis ; 8(10): e3078, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28981114

RESUMEN

Cancer is a multifactorial disease and virus-mediated carcinogenesis is one of the crucial factors, which is poorly understood. Human cytomegalovirus (HCMV) is a herpesvirus and its components have been evidenced to be associated with cancer of different tissue origin. However, its role in cancer remains unknown. Here, we identified a conserved herpesviral tegument protein known as pUL48 of HCMV, encoding deubiquitinase enzyme, as having a key role in carcinogenesis. We show using deubiquitinase sufficient- and deficient-HCMV that HCMV deubiquitinase is a key in inducing enhanced cellular metabolic activity through upregulation of several anti-apoptotic genes and downregulation of several pro-apoptotic genes expression. Furthermore, HCMV deubiquitinase acquires pro-tumor functions by inhibiting PRR-mediated type I interferon via deubiquitination of TRAF6, TRAF3, IRAK1, IRF7 and STING. Taken together, our results suggest that HCMV infection may promote oncogenesis by inhibiting innate immunity of the host.


Asunto(s)
Carcinogénesis/genética , Enzimas Desubicuitinizantes/inmunología , Neoplasias/virología , Proteínas de la Matriz Viral/inmunología , Línea Celular Tumoral , Citomegalovirus/genética , Citomegalovirus/patogenicidad , Enzimas Desubicuitinizantes/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Neoplasias/genética , Neoplasias/inmunología , Activación Transcripcional , Proteínas de la Matriz Viral/genética , Replicación Viral/genética
19.
J Interferon Cytokine Res ; 26(12): 893-900, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17238832

RESUMEN

Interleukin-10 (IL-10) is an anti-inflammatory cytokine that modulates innate and adaptive immunity. IL-10 transcripts and the protein were induced in murine bone marrow-derived dendritic cells (BMDCs) after toll-like receptor (TLR) stimulation. IL-10 induction was TLR ligand selective, in that CpG DNA, imidazoquinolin, peptidoglycan, and zymosan but not lipopolysaccharide (LPS) and poly I:C led to IL-10 production. IL-10 induction was, however, completely absent in MyD88(/) DCs that lacked a TLR adaptor showing that IL-10 induction depends on TLR signaling. Kinetic analysis of IL-10 induction by CpG and imidazoquinolin revealed a prolonged lag phase prior to a measurable rise in transcript levels, which peaked at 12-24 h after stimulation. Stat3, implicated in IL-10 gene transcription, was also induced after TLR stimulation with the kinetics similar to those of IL-10 induction. Further, Stat3 was phosphorylated and bound to the IL-10 promoter in TLR-stimulated DCs. Supporting a link with IL-10 induction, STAT3 induction was absent in MyD88(/) DCs. These data suggest a two-step model where the initial TLR signaling induced proinflammatory cytokines, which then activated Stat3, leading to the induction of IL-10. TLR-stimulated IL-10 production may regulate DC maturation steps, thereby influencing the ensuing immune responses.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica , Interleucina-10/genética , Receptores Toll-Like/fisiología , Animales , Comunicación Autocrina , Imidazoles/farmacología , Inflamación/inmunología , Interleucina-10/análisis , Interleucina-10/metabolismo , Ligandos , Ratones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/fisiología , Oligodesoxirribonucleótidos/farmacología , Comunicación Paracrina , Peptidoglicano/farmacología , Poli I-C/farmacología , Regiones Promotoras Genéticas , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Receptores Toll-Like/agonistas , Transcripción Genética , Activación Transcripcional , Zimosan/farmacología
20.
Cell Res ; 16(2): 134-40, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16474425

RESUMEN

Dendritic cells (DC), although a minor population in hematopoietic cells, produce type I interferons (IFN) and other cytokines and are essential for innate immunity. They are also potent antigen presenters and regulate adaptive immunity. Among DC subtypes plasmacytoid DC (pDC) produce the highest amounts of type I IFN. In addition, pro- and anti-inflammatory cytokines such as IL-12 and IL-10 are induced in DC in response to Toll like receptor (TLR) signaling and upon viral infection. Proteins in the IRF family control many aspects of DC activity. IRF-8 and IRF-4 are essential for DC development. They differentially control the development of four DC subsets. IRF-8-/- mice are largely devoid of pDC and CD8alpha+ DC, while IRF-4-/- mice lack CD4+DC. IRF-8-/-, IRF4-/-, double knock-out mice have only few CD8á-CD4-DC that lack MHC II. IRF proteins also control type I IFN induction in DC. IRF-7, activated upon TLR signaling is required for IFN induction not only in pDC, but also in conventional DC (cDC) and non-DC cell types. IRF-3, although contributes to IFN induction in fibroblasts, is dispensable in IFN induction in DC. Our recent evidence reveals that type I IFN induction in DC is critically dependent on IRF-8, which acts in the feedback phase of IFN gene induction in DC. Type I IFN induction in pDC is mediated by MyD88 dependent signaling pathway, and differs from pathways employed in other cells, which mostly rely on TLR3 and RIG-I family proteins. Other pro-inflammatory cytokines are produced in an IRF-5 dependent manner. However, IRF-5 is not required for IFN induction, suggesting the presence of separate mechanisms for induction of type I IFN and other pro-inflammatory cytokines. IFN and other cytokines produced by activated DC in turn advance DC maturation and change the phenotype and function of DC. These processes are also likely to be governed by IRF family proteins.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/metabolismo , Animales , Interferón Tipo I/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA