Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
6.
Mol Ther ; 31(2): 435-453, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36184851

RESUMEN

Treating osteoporosis and associated bone fractures remains challenging for drug development in part due to potential off-target side effects and the requirement for long-term treatment. Here, we identify recombinant adeno-associated virus (rAAV)-mediated gene therapy as a complementary approach to existing osteoporosis therapies, offering long-lasting targeting of multiple targets and/or previously undruggable intracellular non-enzymatic targets. Treatment with a bone-targeted rAAV carrying artificial microRNAs (miRNAs) silenced the expression of WNT antagonists, schnurri-3 (SHN3), and sclerostin (SOST), and enhanced WNT/ß-catenin signaling, osteoblast function, and bone formation. A single systemic administration of rAAVs effectively reversed bone loss in both postmenopausal and senile osteoporosis. Moreover, the healing of bone fracture and critical-sized bone defects was also markedly improved by systemic injection or transplantation of AAV-bound allograft bone to the osteotomy sites. Collectively, our data demonstrate the clinical potential of bone-specific gene silencers to treat skeletal disorders of low bone mass and impaired fracture repair.


Asunto(s)
Fracturas Óseas , Osteoporosis , Humanos , Proteínas Adaptadoras Transductoras de Señales/genética , Osteoporosis/genética , Osteoporosis/terapia , Fracturas Óseas/genética , Fracturas Óseas/terapia , Huesos , Terapia Genética
7.
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
8.
Nature ; 526(7571): 112-7, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26367794

RESUMEN

The extent to which low-frequency (minor allele frequency (MAF) between 1-5%) and rare (MAF ≤ 1%) variants contribute to complex traits and disease in the general population is mainly unknown. Bone mineral density (BMD) is highly heritable, a major predictor of osteoporotic fractures, and has been previously associated with common genetic variants, as well as rare, population-specific, coding variants. Here we identify novel non-coding genetic variants with large effects on BMD (ntotal = 53,236) and fracture (ntotal = 508,253) in individuals of European ancestry from the general population. Associations for BMD were derived from whole-genome sequencing (n = 2,882 from UK10K (ref. 10); a population-based genome sequencing consortium), whole-exome sequencing (n = 3,549), deep imputation of genotyped samples using a combined UK10K/1000 Genomes reference panel (n = 26,534), and de novo replication genotyping (n = 20,271). We identified a low-frequency non-coding variant near a novel locus, EN1, with an effect size fourfold larger than the mean of previously reported common variants for lumbar spine BMD (rs11692564(T), MAF = 1.6%, replication effect size = +0.20 s.d., Pmeta = 2 × 10(-14)), which was also associated with a decreased risk of fracture (odds ratio = 0.85; P = 2 × 10(-11); ncases = 98,742 and ncontrols = 409,511). Using an En1(cre/flox) mouse model, we observed that conditional loss of En1 results in low bone mass, probably as a consequence of high bone turnover. We also identified a novel low-frequency non-coding variant with large effects on BMD near WNT16 (rs148771817(T), MAF = 1.2%, replication effect size = +0.41 s.d., Pmeta = 1 × 10(-11)). In general, there was an excess of association signals arising from deleterious coding and conserved non-coding variants. These findings provide evidence that low-frequency non-coding variants have large effects on BMD and fracture, thereby providing rationale for whole-genome sequencing and improved imputation reference panels to study the genetic architecture of complex traits and disease in the general population.


Asunto(s)
Densidad Ósea/genética , Fracturas Óseas/genética , Genoma Humano/genética , Proteínas de Homeodominio/genética , Animales , Huesos/metabolismo , Modelos Animales de Enfermedad , Europa (Continente)/etnología , Exoma/genética , Femenino , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Genómica , Genotipo , Humanos , Ratones , Análisis de Secuencia de ADN , Población Blanca/genética , Proteínas Wnt/genética
9.
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
10.
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