Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(12): 1574-1583, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31745335

RESUMEN

Multiple type I interferons and interferon-γ (IFN-γ) are expressed under physiological conditions and are increased by stress and infections, and in autoinflammatory and autoimmune diseases. Interferons activate the Jak-STAT signaling pathway and induce overlapping patterns of expression, called 'interferon signatures', of canonical interferon-stimulated genes (ISGs) encoding molecules important for antiviral responses, antigen presentation, autoimmunity and inflammation. It has now become clear that interferons also induce an 'interferon epigenomic signature' by activating latent enhancers and 'bookmarking' chromatin, thus reprogramming cell responses to environmental cues. The interferon epigenomic signature affects ISGs and other gene sets, including canonical targets of the transcription factor NF-κB that encode inflammatory molecules, and is involved in the priming of immune cells, tolerance and the training of innate immune memory. Here we review the mechanisms through which interferon signatures and interferon epigenomic signatures are generated, as well as the expression and functional consequences of these signatures in homeostasis and autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis and systemic sclerosis.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Epigénesis Genética/inmunología , Inflamación/inmunología , Interferón Tipo I/metabolismo , Animales , Autoinmunidad , Homeostasis , Humanos , Tolerancia Inmunológica , Interferón Tipo I/genética , Quinasas Janus/metabolismo , Activación de Linfocitos , FN-kappa B/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Transcriptoma
2.
Nat Immunol ; 19(4): 407-419, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29483597

RESUMEN

Age-associated B cells (ABCs) are a subset of B cells dependent on the transcription factor T-bet that accumulate prematurely in autoimmune settings. The pathways that regulate ABCs in autoimmunity are largely unknown. SWAP-70 and DEF6 (also known as IBP or SLAT) are the only two members of the SWEF family, a unique family of Rho GTPase-regulatory proteins that control both cytoskeletal dynamics and the activity of the transcription factor IRF4. Notably, DEF6 is a newly identified human risk variant for systemic lupus erythematosus. Here we found that the lupus syndrome that developed in SWEF-deficient mice was accompanied by the accumulation of ABCs that produced autoantibodies after stimulation. ABCs from SWEF-deficient mice exhibited a distinctive transcriptome and a unique chromatin landscape characterized by enrichment for motifs bound by transcription factors of the IRF and AP-1 families and the transcription factor T-bet. Enhanced ABC formation in SWEF-deficient mice was controlled by the cytokine IL-21 and IRF5, whose variants are strongly associated with lupus. The lack of SWEF proteins led to dysregulated activity of IRF5 in response to stimulation with IL-21. These studies thus elucidate a previously unknown signaling pathway that controls ABCs in autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Subgrupos de Linfocitos B/inmunología , Factores Reguladores del Interferón/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Subgrupos de Linfocitos B/patología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/inmunología , Femenino , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/inmunología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/inmunología
3.
Nat Immunol ; 18(10): 1104-1116, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28825701

RESUMEN

Cross-regulation of Toll-like receptor (TLR) responses by cytokines is essential for effective host defense, avoidance of toxicity and homeostasis, but the underlying mechanisms are not well understood. Our comprehensive epigenomics approach to the analysis of human macrophages showed that the proinflammatory cytokines TNF and type I interferons induced transcriptional cascades that altered chromatin states to broadly reprogram responses induced by TLR4. TNF tolerized genes encoding inflammatory molecules to prevent toxicity while preserving the induction of genes encoding antiviral and metabolic molecules. Type I interferons potentiated the inflammatory function of TNF by priming chromatin to prevent the silencing of target genes of the transcription factor NF-κB that encode inflammatory molecules. The priming of chromatin enabled robust transcriptional responses to weak upstream signals. Similar chromatin regulation occurred in human diseases. Our findings reveal that signaling crosstalk between interferons and TNF is integrated at the level of chromatin to reprogram inflammatory responses, and identify previously unknown functions and mechanisms of action of these cytokines.


Asunto(s)
Epigénesis Genética , Inflamación/etiología , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Sitios de Unión , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Análisis por Conglomerados , Biología Computacional/métodos , Citocinas/genética , Citocinas/metabolismo , Epigenómica/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lipopolisacáridos/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Factores de Transcripción/metabolismo
4.
Immunity ; 51(2): 241-257.e9, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31303399

RESUMEN

Cytokine tumor necrosis factor (TNF)-mediated macrophage polarization is important for inflammatory disease pathogenesis, but the mechanisms regulating polarization are not clear. We performed transcriptomic and epigenomic analysis of the TNF response in primary human macrophages and revealed late-phase activation of SREBP2, the master regulator of cholesterol biosynthesis genes. TNF stimulation extended the genomic profile of SREBP2 occupancy to include binding to and activation of inflammatory and interferon response genes independently of its functions in sterol metabolism. Genetic ablation of SREBP function shifted the balance of macrophage polarization from an inflammatory to a reparative phenotype in peritonitis and skin wound healing models. Genetic ablation of SREBP activity in myeloid cells or topical pharmacological inhibition of SREBP improved skin wound healing under homeostatic and chronic inflammatory conditions. Our results identify a function and mechanism of action for SREBPs in augmenting TNF-induced macrophage activation and inflammation and open therapeutic avenues for promoting wound repair.


Asunto(s)
Inflamación/metabolismo , Macrófagos/inmunología , Peritonitis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Enfermedades de la Piel/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Epigenómica , Femenino , Humanos , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/genética , Transcriptoma , Cicatrización de Heridas
5.
Immunol Rev ; 323(1): 257-275, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38567833

RESUMEN

Training and priming of innate immune cells involve preconditioning by PAMPs, DAMPs, and/or cytokines that elicits stronger induction of inflammatory genes upon secondary challenge. Previous models distinguish training and priming based upon whether immune activation returns to baseline prior to secondary challenge. Tolerance is a protective mechanism whereby potent stimuli induce refractoriness to secondary challenge. Training and priming are important for innate memory responses that protect against infection, efficacy of vaccines, and maintaining innate immune cells in a state of readiness; tolerance prevents toxicity from excessive immune activation. Dysregulation of these processes can contribute to pathogenesis of autoimmune/inflammatory conditions, post-COVID-19 hyperinflammatory states, or sepsis-associated immunoparalysis. Training, priming, and tolerance regulate similar "signature" inflammatory genes such as TNF, IL6, and IL1B and utilize overlapping epigenetic mechanisms. We review how interferons (IFNs), best known for activating JAK-STAT signaling and interferon-stimulated genes, also play a key role in regulating training, priming, and tolerance via chromatin-mediated mechanisms. We present new data on how monocyte-to-macrophage differentiation modulates IFN-γ-mediated priming, affects regulation of AP-1 and CEBP activity, and attenuates superinduction of inflammatory genes. We present a "training-priming continuum" model that integrates IFN-mediated priming into current concepts about training and tolerance and proposes a central role for STAT1 and IRF1.


Asunto(s)
Epigénesis Genética , Células Madre Hematopoyéticas , Tolerancia Inmunológica , Interferones , Monocitos , Humanos , Monocitos/inmunología , Monocitos/metabolismo , Interferones/metabolismo , Animales , Células Madre Hematopoyéticas/metabolismo , Inmunidad Innata , Transducción de Señal , COVID-19/inmunología , SARS-CoV-2/inmunología , Diferenciación Celular , Memoria Inmunológica
6.
Nat Immunol ; 16(8): 838-849, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26147685

RESUMEN

Interferon-γ (IFN-γ) primes macrophages for enhanced microbial killing and inflammatory activation by Toll-like receptors (TLRs), but little is known about the regulation of cell metabolism or mRNA translation during this priming. We found that IFN-γ regulated the metabolism and mRNA translation of human macrophages by targeting the kinases mTORC1 and MNK, both of which converge on the selective regulator of translation initiation eIF4E. Physiological downregulation of mTORC1 by IFN-γ was associated with autophagy and translational suppression of repressors of inflammation such as HES1. Genome-wide ribosome profiling in TLR2-stimulated macrophages showed that IFN-γ selectively modulated the macrophage translatome to promote inflammation, further reprogram metabolic pathways and modulate protein synthesis. These results show that IFN-γ-mediated metabolic reprogramming and translational regulation are key components of classical inflammatory macrophage activation.


Asunto(s)
Interferón gamma/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Biosíntesis de Proteínas/inmunología , ARN Mensajero/inmunología , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Células Cultivadas , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/inmunología , Factor 4E Eucariótico de Iniciación/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Proteínas de Homeodominio/metabolismo , Humanos , Interferón gamma/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , MicroARNs/genética , Microscopía Fluorescente , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Factor de Transcripción HES-1
8.
Immunity ; 47(2): 235-250.e4, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813657

RESUMEN

Mechanisms by which interferon (IFN)-γ activates genes to promote macrophage activation are well studied, but little is known about mechanisms and functions of IFN-γ-mediated gene repression. We used an integrated transcriptomic and epigenomic approach to analyze chromatin accessibility, histone modifications, transcription-factor binding, and gene expression in IFN-γ-primed human macrophages. IFN-γ suppressed basal expression of genes corresponding to an "M2"-like homeostatic and reparative phenotype. IFN-γ repressed genes by suppressing the function of enhancers enriched for binding by transcription factor MAF. Mechanistically, IFN-γ disassembled a subset of enhancers by inducing coordinate suppression of binding by MAF, lineage-determining transcription factors, and chromatin accessibility. Genes associated with MAF-binding enhancers were suppressed in macrophages isolated from rheumatoid-arthritis patients, revealing a disease-associated signature of IFN-γ-mediated repression. These results identify enhancer inactivation and disassembly as a mechanism of IFN-γ-mediated gene repression and reveal that MAF regulates the macrophage enhancer landscape and is suppressed by IFN-γ to augment macrophage activation.


Asunto(s)
Artritis Reumatoide/inmunología , Ensamble y Desensamble de Cromatina , Interferón gamma/metabolismo , Macrófagos/inmunología , Proteínas Proto-Oncogénicas c-maf/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Citocinas/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Unión Proteica , Proteínas Proto-Oncogénicas c-maf/genética , Transcriptoma
10.
Immunity ; 47(1): 66-79.e5, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723554

RESUMEN

Hypoxia augments inflammatory responses and osteoclastogenesis by incompletely understood mechanisms. We identified COMMD1 as a cell-intrinsic negative regulator of osteoclastogenesis that is suppressed by hypoxia. In human macrophages, COMMD1 restrained induction of NF-κB signaling and a transcription factor E2F1-dependent metabolic pathway by the cytokine RANKL. Downregulation of COMMD1 protein expression by hypoxia augmented RANKL-induced expression of inflammatory and E2F1 target genes and downstream osteoclastogenesis. E2F1 targets included glycolysis and metabolic genes including CKB that enabled cells to meet metabolic demands in challenging environments, as well as inflammatory cytokine-driven target genes. Expression quantitative trait locus analysis linked increased COMMD1 expression with decreased bone erosion in rheumatoid arthritis. Myeloid deletion of Commd1 resulted in increased osteoclastogenesis in arthritis and inflammatory osteolysis models. These results identify COMMD1 and an E2F-metabolic pathway as key regulators of osteoclastogenic responses under pathological inflammatory conditions and provide a mechanism by which hypoxia augments inflammation and bone destruction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Artritis Reumatoide/inmunología , Macrófagos/inmunología , Osteogénesis/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/metabolismo , Femenino , Humanos , Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal
11.
Nat Immunol ; 13(7): 642-50, 2012 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-22610140

RESUMEN

Emerging concepts suggest that the functional phenotype of macrophages is regulated by transcription factors that define alternative activation states. We found that RBP-J, the main nuclear transducer of signaling via Notch receptors, augmented Toll-like receptor 4 (TLR4)-induced expression of key mediators of classically activated M1 macrophages and thus of innate immune responses to Listeria monocytogenes. Notch-RBP-J signaling controlled expression of the transcription factor IRF8 that induced downstream M1 macrophage-associated genes. RBP-J promoted the synthesis of IRF8 protein by selectively augmenting kinase IRAK2-dependent signaling via TLR4 to the kinase MNK1 and downstream translation-initiation control through eIF4E. Our results define a signaling network in which signaling via Notch-RBP-J and TLRs is integrated at the level of synthesis of IRF8 protein and identify a mechanism by which heterologous signaling pathways can regulate the TLR-induced inflammatory polarization of macrophages.


Asunto(s)
Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/inmunología , Inflamación/inmunología , Factores Reguladores del Interferón/inmunología , Macrófagos/inmunología , Receptores Notch/inmunología , Animales , Polaridad Celular/inmunología , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Factores Reguladores del Interferón/biosíntesis , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Listeriosis/inmunología , Activación de Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Factores de Transcripción/metabolismo
12.
Nat Immunol ; 12(7): 607-15, 2011 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-21602809

RESUMEN

Endotoxin tolerance, a key mechanism for suppressing excessive inflammatory cytokine production, is induced by prior exposure of macrophages to Toll-like receptor (TLR) ligands. Induction of cross-tolerance to endotoxin by endogenous cytokines has not been investigated. Here we show that prior exposure to tumor necrosis factor (TNF) induced a tolerant state in macrophages, with less cytokine production after challenge with lipopolysaccharide (LPS) and protection from LPS-induced death. TNF-induced cross-tolerization was mediated by suppression of LPS-induced signaling and chromatin remodeling. TNF-induced cross-tolerance was dependent on the kinase GSK3, which suppressed chromatin accessibility and promoted rapid termination of signaling via the transcription factor NF-κB by augmenting negative feedback by the signaling inhibitors A20 and IκBα. Our results demonstrate an unexpected homeostatic function for TNF and a GSK3-mediated mechanism for the prevention of prolonged and excessive inflammation.


Asunto(s)
Endotoxinas/inmunología , Glucógeno Sintasa Quinasa 3/inmunología , Macrófagos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/inmunología , Cisteína Endopeptidasas/inmunología , Cisteína Endopeptidasas/metabolismo , Citocinas/biosíntesis , Citocinas/inmunología , Femenino , Proteínas I-kappa B/inmunología , Proteínas I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , FN-kappa B/inmunología , FN-kappa B/metabolismo , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
13.
Immunity ; 41(1): 14-20, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25035950

RESUMEN

Description of macrophage activation is currently contentious and confusing. Like the biblical Tower of Babel, macrophage activation encompasses a panoply of descriptors used in different ways. The lack of consensus on how to define macrophage activation in experiments in vitro and in vivo impedes progress in multiple ways, including the fact that many researchers still consider there to be only two types of activated macrophages, often termed M1 and M2. Here, we describe a set of standards encompassing three principles-the source of macrophages, definition of the activators, and a consensus collection of markers to describe macrophage activation-with the goal of unifying experimental standards for diverse experimental scenarios. Collectively, we propose a common framework for macrophage-activation nomenclature.


Asunto(s)
Activación de Macrófagos/inmunología , Macrófagos/inmunología , Terminología como Asunto , Animales , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Guías como Asunto , Humanos , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Investigación
14.
Nature ; 542(7639): 110-114, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28150777

RESUMEN

CD4+ T cells are central mediators of autoimmune pathology; however, defining their key effector functions in specific autoimmune diseases remains challenging. Pathogenic CD4+ T cells within affected tissues may be identified by expression of markers of recent activation. Here we use mass cytometry to analyse activated T cells in joint tissue from patients with rheumatoid arthritis, a chronic immune-mediated arthritis that affects up to 1% of the population. This approach revealed a markedly expanded population of PD-1hiCXCR5-CD4+ T cells in synovium of patients with rheumatoid arthritis. However, these cells are not exhausted, despite high PD-1 expression. Rather, using multidimensional cytometry, transcriptomics, and functional assays, we define a population of PD-1hiCXCR5- 'peripheral helper' T (TPH) cells that express factors enabling B-cell help, including IL-21, CXCL13, ICOS, and MAF. Like PD-1hiCXCR5+ T follicular helper cells, TPH cells induce plasma cell differentiation in vitro through IL-21 secretion and SLAMF5 interaction (refs 3, 4). However, global transcriptomics highlight differences between TPH cells and T follicular helper cells, including altered expression of BCL6 and BLIMP1 and unique expression of chemokine receptors that direct migration to inflamed sites, such as CCR2, CX3CR1, and CCR5, in TPH cells. TPH cells appear to be uniquely poised to promote B-cell responses and antibody production within pathologically inflamed non-lymphoid tissues.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Linfocitos B/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Artritis Reumatoide/sangre , Linfocitos B/patología , Diferenciación Celular , Movimiento Celular , Quimiocina CXCL13/metabolismo , Perfilación de la Expresión Génica , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Interleucinas/metabolismo , Factores Activadores de Macrófagos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Receptores CXCR5/deficiencia , Receptores CXCR5/metabolismo , Receptores de Quimiocina/metabolismo , Proteínas Represoras/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Líquido Sinovial/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
15.
Immunity ; 39(1): 91-3, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23890067

RESUMEN

The tyrosine phosphatase PTPN22 regulates T cell receptor signaling. In this issue of Immunity, Wang et al. (2013) show that in myeloid cells PTPN22 potentiates TLR-induced type I interferon production and that autoimmunity-associated allele PTPN22W encodes a reduced-function variant.


Asunto(s)
Autoinmunidad/inmunología , Inmunidad/inmunología , Interferón Tipo I/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/inmunología , Receptores Toll-Like/inmunología , Animales , Humanos
16.
Immunity ; 39(3): 454-69, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-24012417

RESUMEN

Synergistic activation of inflammatory cytokine genes by interferon-γ (IFN-γ) and Toll-like receptor (TLR) signaling is important for innate immunity and inflammatory disease pathogenesis. Enhancement of TLR signaling, a previously proposed mechanism, is insufficient to explain strong synergistic activation of cytokine production in human macrophages. Rather, we found that IFN-γ induced sustained occupancy of transcription factors STAT1, IRF-1, and associated histone acetylation at promoters and enhancers at the TNF, IL6, and IL12B loci. This priming of chromatin did not activate transcription but greatly increased and prolonged recruitment of TLR4-induced transcription factors and RNA polymerase II to gene promoters and enhancers. Priming sensitized cytokine transcription to suppression by Jak inhibitors. Genome-wide analysis revealed pervasive priming of regulatory elements by IFN-γ and linked coordinate priming of promoters and enhancers with synergistic induction of transcription. Our results provide a synergy mechanism whereby IFN-γ creates a primed chromatin environment to augment TLR-induced gene transcription.


Asunto(s)
Ensamble y Desensamble de Cromatina , Citocinas/metabolismo , Interferón gamma/metabolismo , Receptores Toll-Like/metabolismo , Acetilación , Células Cultivadas , Activación Enzimática , Histonas/metabolismo , Humanos , Factor 1 Regulador del Interferón/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Interleucina-6/metabolismo , Quinasas Janus/antagonistas & inhibidores , Macrófagos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN Polimerasa II/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Transcripción Genética , Factores de Necrosis Tumoral/metabolismo
17.
Nat Immunol ; 10(4): 340-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19295630

RESUMEN

An important function of receptors that signal through immunoreceptor tyrosine-based activation motifs (ITAMs) is to regulate signaling by heterologous receptors. This review describes mechanisms by which ITAM-associated receptors modulate signaling by Toll-like receptors (TLRs), tumor necrosis factor receptor family members and cytokine receptors that use the Jak-STAT signaling pathway, and the biological importance of this signal transduction cross-talk. ITAM-mediated cross-regulation can either augment or dampen signaling by other receptors. Conversely, TLRs and cytokines modulate ITAM-mediated signaling, by means including activation of beta2 integrins that are coupled to the ITAM-containing adaptors DAP12 and FcRgamma. Integration of ITAM signaling into signaling networks through cross-talk with other signal transduction pathways results in tight regulation and fine tuning of cellular responses to various extracellular stimuli and contributes to induction of specific activation and differentiation pathways.


Asunto(s)
Secuencias de Aminoácidos , Receptores de Superficie Celular/fisiología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Antígenos CD18/inmunología , Antígenos CD18/metabolismo , Humanos , Región Variable de Inmunoglobulina/inmunología , Quinasas Janus/inmunología , Quinasas Janus/fisiología , Proteínas de la Membrana/inmunología , Receptor Cross-Talk/inmunología , Receptores de Superficie Celular/inmunología , Receptores de Citocinas/inmunología , Receptores de Citocinas/fisiología , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/fisiología , Receptores del Factor de Necrosis Tumoral/inmunología , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Transcripción STAT/inmunología , Factores de Transcripción STAT/fisiología , Receptores Toll-Like/inmunología , Receptores Toll-Like/fisiología
18.
Nat Immunol ; 9(4): 378-87, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18345002

RESUMEN

Rapid induction of inflammatory genes by tumor necrosis factor (TNF) has been well studied, but little is known about delayed and chronic TNF responses. Here we investigated the kinetics of primary macrophage responses to TNF and discovered that TNF initiates an interferon-beta-mediated autocrine loop that sustains expression of inflammatory genes and induces delayed expression of interferon-response genes such as those encoding the transcription factors STAT1 and IRF7, which enhance macrophage responses to stimulation of cytokines and Toll-like receptors. TNF-induced interferon-beta production depended on interferon-response factor 1, and downstream gene expression was mediated by synergy between small amounts of interferon-beta and canonical TNF-induced signals. Thus, TNF activates a 'feed-forward' loop that sustains inflammation but avoids the potential toxicity associated with the high interferon production induced by stimulation of Toll-like receptors.


Asunto(s)
Comunicación Autocrina/inmunología , Quimiocinas/biosíntesis , Quimiocinas/genética , Regulación de la Expresión Génica/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Factor de Transcripción STAT1/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Humanos , Mediadores de Inflamación/fisiología , Factor 1 Regulador del Interferón/fisiología , Interferón Tipo I/biosíntesis , Interferón Tipo I/fisiología , Interferón beta/biosíntesis , Interferón beta/genética , Interferón beta/fisiología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Factores de Tiempo
19.
Nat Immunol ; 9(2): 186-93, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18084294

RESUMEN

Immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors modulate the amplitude and nature of macrophage responses to Toll-like receptor and cytokine receptor stimulation. However, the molecular mechanisms enabling this receptor crosstalk are not known. Here we investigated the function of the calcium-dependent kinases CaMK and Pyk2 'downstream' of ITAM-associated receptors in the regulation of cytokine-induced activation of Jak kinases and STAT transcription factors. CaMK and Pyk2 relayed signals from integrins and the ITAM-containing adaptor DAP12 to augment interleukin 10- and interferon-alpha-induced Jak activation and STAT1-dependent gene expression. CaMK inhibition suppressed STAT1-mediated interferon-alpha signaling in a mouse model of systemic lupus erythematosus. Our results associate Pyk2 and Jak kinases with the linkage of signals emanating from cytokine and heterologous ITAM-dependent receptors.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Interferón Tipo I/farmacología , Quinasas Janus/metabolismo , Macrófagos/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Calcio/metabolismo , Células Cultivadas , Humanos , Macrófagos/inmunología , Proteínas de la Membrana , Ratones , Fosforilación , Receptores Inmunológicos/metabolismo , Transducción de Señal , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA