Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Front Cell Dev Biol ; 10: 920683, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36060812

RESUMEN

Osteoclasts are bone-resorbing cells that undergo extensive changes in morphology throughout their differentiation. Altered osteoclast differentiation and activity lead to changes in pathological bone resorption. The mammalian target of rapamycin (mTOR) is a kinase, and aberrant mTOR complex 1 (mTORC1) signaling is associated with altered bone homeostasis. The activation of mTORC1 is biphasically regulated during osteoclastogenesis; however, the mechanism behind mTORC1-mediated regulation of osteoclastogenesis and bone resorption is incompletely understood. Here, we found that MYC coordinates the dynamic regulation of mTORC1 activation during osteoclastogenesis. MYC-deficiency blocked the early activation of mTORC1 and also reversed the decreased activity of mTORC1 at the late stage of osteoclastogenesis. The suppression of mTORC1 activity by rapamycin in mature osteoclasts enhances bone resorption activity despite the indispensable role of high mTORC1 activation in osteoclast formation in both mouse and human cells. Mechanistically, MYC induces Growth arrest and DNA damage-inducible protein (GADD34) expression and suppresses mTORC1 activity at the late phase of osteoclastogenesis. Taken together, our findings identify a MYC-GADD34 axis as an upstream regulator of dynamic mTORC1 activation in osteoclastogenesis and highlight the interplay between MYC and mTORC1 pathways in determining osteoclast activity.

4.
Arthritis Rheumatol ; 74(9): 1544-1555, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35438841

RESUMEN

OBJECTIVE: Age-associated/autoimmune B cells (ABCs) are an emerging B cell subset with aberrant expansion in systemic lupus erythematosus. ABC generation and differentiation exhibit marked sexual dimorphism, and Toll-like receptor 7 (TLR-7) engagement is a key contributor to these sex differences. ABC generation is also controlled by interleukin-21 (IL-21) and its interplay with interferon-γ and IL-4. This study was undertaken to investigate whether IL-13 receptor α1 (IL-13Rα1), an X-linked receptor that transmits IL-4/IL-13 signals, regulates ABCs and lupus pathogenesis. METHODS: Mice lacking DEF-6 and switch-associated protein 70 (double-knockout [DKO]), which preferentially develop lupus in females, were crossed with IL-13Rα1-knockout mice. IL-13Rα1-knockout male mice were also crossed with Y chromosome autoimmune accelerator (Yaa) DKO mice, which overexpress TLR-7 and develop severe disease. ABCs were assessed using flow cytometry and RNA-Seq. Lupus pathogenesis was evaluated using serologic and histologic analyses. RESULTS: ABCs expressed higher levels of IL-13Rα1 than follicular B cells. The absence of IL-13Rα1 in either DKO female mice or Yaa DKO male mice decreased the accumulation of ABCs, the differentiation of ABCs into plasmablasts, and autoantibody production. Lack of IL-13Rα1 also prolonged survival and delayed the development of tissue inflammation. IL-13Rα1 deficiency diminished in vitro generation of ABCs, an effect that, surprisingly, could be observed in response to IL-21 alone. RNA-Seq revealed that ABCs lacking IL-13Rα1 down-regulated some histologic characteristics of B cells but up-regulated myeloid markers and proinflammatory mediators. CONCLUSION: Our findings indicate a novel role for IL-13Rα1 in controlling ABC generation and differentiation, suggesting that IL-13Rα1 contributes to these effects by regulating a subset of IL-21-mediated signaling events. These results also suggest that X-linked genes besides TLR7 participate in the regulation of ABCs in lupus.


Asunto(s)
Interleucina-13 , Lupus Eritematoso Sistémico , Receptores de Interleucina-13 , Animales , Femenino , Interleucina-13/metabolismo , Subunidad alfa1 del Receptor de Interleucina-13/genética , Interleucina-4 , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores de Interleucina-13/genética , Receptor Toll-Like 7
5.
Cell Rep ; 35(11): 109264, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34133930

RESUMEN

MYC activates different metabolic programs in a cell-type- and cell-status-dependent manner. However, the role of MYC in inflammatory macrophages has not yet been determined. Metabolic and molecular analyses reveal that MYC, but not hypoxia inducible factor 1 (HIF1), is involved in enhancing early glycolytic flux during inflammatory macrophage polarization. Ablation of MYC decreases lactate production by regulating lactate dehydrogenase (LDH) activity and causes increased inflammatory cytokines by regulating interferon regulatory factor 4 (IRF4) in response to lipopolysaccharide. Moreover, myeloid-specific deletion of MYC and pharmacological inhibition of the MYC/LDH axis enhance inflammation and the bacterial clearance in vivo. These results elucidate the potential role of the MYC/LDH/IRF4 axis in inflammatory macrophages by connecting early glycolysis with inflammatory responses and suggest that modulating early glycolytic flux mediated by the MYC/LDH axis can be used to open avenues for the therapeutic modulation of macrophage polarization to fight against bacterial infection.


Asunto(s)
Glucólisis , Inflamación/metabolismo , Inflamación/patología , Factores Reguladores del Interferón/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Bacterias/metabolismo , Citocinas/biosíntesis , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Ácido Láctico/metabolismo , Lipopolisacáridos , Masculino , Ratones Noqueados , Proteínas Proto-Oncogénicas c-myc/deficiencia
6.
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
7.
Nat Rev Rheumatol ; 15(6): 327-339, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31000790

RESUMEN

Rheumatic diseases have complex aetiologies that are not fully understood, which makes the study of pathogenic mechanisms in these diseases a challenge for researchers. Next-generation sequencing (NGS) and related omics technologies, such as transcriptomics, epigenomics and genomics, provide an unprecedented genome-wide view of gene expression, environmentally responsive epigenetic changes and genetic variation. The integrated application of NGS technologies to samples from carefully phenotyped clinical cohorts of patients has the potential to solve remaining mysteries in the pathogenesis of several rheumatic diseases, to identify new therapeutic targets and to underpin a precision medicine approach to the diagnosis and treatment of rheumatic diseases. This Review provides an overview of the NGS technologies available, showcases important advances in rheumatic disease research already powered by these technologies and highlights NGS approaches that hold particular promise for generating new insights and advancing the field.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Enfermedades Reumáticas/genética , Epigenómica , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Enfermedades Reumáticas/etiología , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
8.
Nat Commun ; 9(1): 4108, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30291236

RESUMEN

Targeting microRNAs recently shows significant therapeutic promise; however, such progress is underdeveloped in treatment of skeletal diseases with osteolysis, such as osteoporosis and rheumatoid arthritis (RA). Here, we identified miR-182 as a key osteoclastogenic regulator in bone homeostasis and diseases. Myeloid-specific deletion of miR-182 protects mice against excessive osteoclastogenesis and bone resorption in disease models of ovariectomy-induced osteoporosis and inflammatory arthritis. Pharmacological treatment of these diseases with miR-182 inhibitors completely suppresses pathologic bone erosion. Mechanistically, we identify protein kinase double-stranded RNA-dependent (PKR) as a new and essential miR-182 target that is a novel inhibitor of osteoclastogenesis via regulation of the endogenous interferon (IFN)-ß-mediated autocrine feedback loop. The expression levels of miR-182, PKR, and IFN-ß are altered in RA and are significantly correlated with the osteoclastogenic capacity of RA monocytes. Our findings reveal a previously unrecognized regulatory network mediated by miR-182-PKR-IFN-ß axis in osteoclastogenesis, and highlight the therapeutic implications of miR-182 inhibition in osteoprotection.


Asunto(s)
Resorción Ósea/prevención & control , Interferón beta/metabolismo , MicroARNs/metabolismo , Osteogénesis , eIF-2 Quinasa/metabolismo , Animales , Artritis Reumatoide/complicaciones , Artritis Reumatoide/metabolismo , Comunicación Autocrina , Resorción Ósea/etiología , Femenino , Homeostasis , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/antagonistas & inhibidores , Monocitos/fisiología
9.
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
10.
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
11.
PLoS One ; 12(7): e0179762, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28708839

RESUMEN

During rheumatoid arthritis (RA), Tumor Necrosis Factor (TNF) activates fibroblast-like synoviocytes (FLS) inducing in a temporal order a constellation of genes, which perpetuate synovial inflammation. Although the molecular mechanisms regulating TNF-induced transcription are well characterized, little is known about the impact of mRNA stability on gene expression and the impact of TNF on decay rates of mRNA transcripts in FLS. To address these issues we performed RNA sequencing and genome-wide analysis of the mRNA stabilome in RA FLS. We found that TNF induces a biphasic gene expression program: initially, the inducible transcriptome consists primarily of unstable transcripts but progressively switches and becomes dominated by very stable transcripts. This temporal switch is due to: a) TNF-induced prolonged stabilization of previously unstable transcripts that enables progressive transcript accumulation over days and b) sustained expression and late induction of very stable transcripts. TNF-induced mRNA stabilization in RA FLS occurs during the late phase of TNF response, is MAPK-dependent, and involves several genes with pathogenic potential such as IL6, CXCL1, CXCL3, CXCL8/IL8, CCL2, and PTGS2. These results provide the first insights into genome-wide regulation of mRNA stability in RA FLS and highlight the potential contribution of dynamic regulation of the mRNA stabilome by TNF to chronic synovitis.


Asunto(s)
Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Fibroblastos/citología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , ARN/química , ARN/aislamiento & purificación , ARN/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN , Sinoviocitos/citología , Sinoviocitos/efectos de los fármacos , Sinoviocitos/metabolismo
12.
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
13.
J Clin Invest ; 127(7): 2555-2568, 2017 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-28530645

RESUMEN

Osteoporosis is a metabolic bone disorder associated with compromised bone strength and an increased risk of fracture. Inhibition of the differentiation of bone-resorbing osteoclasts is an effective strategy for the treatment of osteoporosis. Prior work by our laboratory and others has shown that MYC promotes osteoclastogenesis in vitro, but the underlying mechanisms are not well understood. In addition, the in vivo importance of osteoclast-expressed MYC in physiological and pathological bone loss is not known. Here, we have demonstrated that deletion of Myc in osteoclasts increases bone mass and protects mice from ovariectomy-induced (OVX-induced) osteoporosis. Transcriptomic analysis revealed that MYC drives metabolic reprogramming during osteoclast differentiation and functions as a metabolic switch to an oxidative state. We identified a role for MYC action in the transcriptional induction of estrogen receptor-related receptor α (ERRα), a nuclear receptor that cooperates with the transcription factor nuclear factor of activated T cells, c1 (NFATc1) to drive osteoclastogenesis. Accordingly, pharmacological inhibition of ERRα attenuated OVX-induced bone loss in mice. Our findings highlight a MYC/ERRα pathway that contributes to physiological and pathological bone loss by integrating the MYC/ERRα axis to drive metabolic reprogramming during osteoclast differentiation.


Asunto(s)
Diferenciación Celular , Osteoclastos/metabolismo , Osteoporosis/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Osteoclastos/patología , Osteoporosis/genética , Osteoporosis/patología , Osteoporosis/terapia , Proteínas Proto-Oncogénicas c-myc/genética , Receptores de Estrógenos/genética , Transcriptoma , Receptor Relacionado con Estrógeno ERRalfa
14.
Methods Mol Biol ; 1507: 43-58, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27832531

RESUMEN

Chromatin immunoprecipitation followed by sequencing is an invaluable assay for identifying the genomic binding sites of transcription factors. However, transcription factors rarely bind chromatin alone but often bind together with other cofactors, forming protein complexes. Here, we describe a computational method that integrates multiple ChIP-seq and RNA-seq datasets to discover protein complexes and determine their role as activators or repressors. This chapter outlines a detailed computational pipeline for discovering and predicting binding partners from ChIP-seq data and inferring their role in regulating gene expression. This work aims at developing hypotheses about gene regulation via binding partners and deciphering the combinatorial nature of DNA-binding proteins.


Asunto(s)
Proteínas de Unión al ADN/aislamiento & purificación , Mapeo de Interacción de Proteínas , Línea Celular , Inmunoprecipitación de Cromatina , Simulación por Computador , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Complejos Multiproteicos/aislamiento & purificación , Análisis de Regresión , Análisis de Secuencia de ADN , Factores de Transcripción/aislamiento & purificación
15.
Cell Rep ; 16(12): 3121-3129, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27653678

RESUMEN

The mechanisms by which IFN-γ activates expression of interferon-stimulated genes that have inflammatory and host defense functions are well understood. In contrast, little is known about how IFN-γ represses gene expression. By using transcriptomic and epigenomic analysis, we found that stable repression of a small group of genes by IFN-γ is associated with recruitment of the histone methyltransferase EZH2 and deposition of the negative mark histone 3 lysine 27 trimethylation (H3K27me3) at their promoters. Repressed genes included MERTK, PPARG, and RANK, which have anti-inflammatory functions and promote osteoclast differentiation. Gene repression and H3K27me3 persisted after IFN-γ signaling was terminated, and these silenced genes were no longer responsive to glucocorticoids, IL-4, and M-CSF. These results identify cytokine-induced H3K27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. IFN-γ-induced macrophage activation is thus reinforced by a chromatin-based mechanism that blocks anti-inflammatory and opposing pathways.


Asunto(s)
Silenciador del Gen/fisiología , Histonas/metabolismo , Interferón gamma/biosíntesis , Activación de Macrófagos/genética , Macrófagos/inmunología , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histonas/genética , Humanos , Interferón gamma/genética , Lisina , Activación de Macrófagos/inmunología , Regiones Promotoras Genéticas/genética
16.
Nat Commun ; 7: 11275, 2016 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-27117588

RESUMEN

Spermatogonial stem and progenitor cells (SSCs) generate adult male gametes. During in vitro expansion, these unipotent murine cells spontaneously convert to multipotent adult spermatogonial-derived stem cells (MASCs). Here we investigate this conversion process through integrative transcriptomic and epigenomic analyses. We find in SSCs that promoters essential to maintenance and differentiation of embryonic stem cells (ESCs) are enriched with histone H3-lysine4 and -lysine 27 trimethylations. These bivalent modifications are maintained at most somatic promoters after conversion, bestowing MASCs an ESC-like promoter chromatin. At enhancers, the core pluripotency circuitry is activated partially in SSCs and completely in MASCs, concomitant with loss of germ cell-specific gene expression and initiation of embryonic-like programs. Furthermore, SSCs in vitro maintain the epigenomic characteristics of germ cells in vivo. Our observations suggest that SSCs encode innate plasticity through the epigenome and that both conversion of promoter chromatin states and activation of cell type-specific enhancers are prominent features of reprogramming.


Asunto(s)
Diferenciación Celular/genética , Plasticidad de la Célula/genética , Células Madre Embrionarias/metabolismo , Epigenómica/métodos , Células Madre Multipotentes/metabolismo , Espermatogonias/metabolismo , Animales , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Histonas/metabolismo , Lisina/metabolismo , Masculino , Metilación , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Espermatogénesis/genética , Espermatogonias/citología
17.
Nat Med ; 22(3): 298-305, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26855148

RESUMEN

An increasingly recognized resistance mechanism to androgen receptor (AR)-directed therapy in prostate cancer involves epithelial plasticity, in which tumor cells demonstrate low to absent AR expression and often have neuroendocrine features. The etiology and molecular basis for this 'alternative' treatment-resistant cell state remain incompletely understood. Here, by analyzing whole-exome sequencing data of metastatic biopsies from patients, we observed substantial genomic overlap between castration-resistant tumors that were histologically characterized as prostate adenocarcinomas (CRPC-Adeno) and neuroendocrine prostate cancer (CRPC-NE); analysis of biopsy samples from the same individuals over time points to a model most consistent with divergent clonal evolution. Genome-wide DNA methylation analysis revealed marked epigenetic differences between CRPC-NE tumors and CRPC-Adeno, and also designated samples of CRPC-Adeno with clinical features of AR independence as CRPC-NE, suggesting that epigenetic modifiers may play a role in the induction and/or maintenance of this treatment-resistant state. This study supports the emergence of an alternative, 'AR-indifferent' cell state through divergent clonal evolution as a mechanism of treatment resistance in advanced prostate cancer.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Óseas/genética , Evolución Clonal/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Tumores Neuroendocrinos/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/genética , Adenocarcinoma/secundario , Anciano , Anciano de 80 o más Años , Neoplasias Óseas/secundario , Metilación de ADN , Epigénesis Genética , Humanos , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/secundario , Estudios Prospectivos , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Retrospectivos
18.
Arthritis Rheumatol ; 68(6): 1454-66, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26816213

RESUMEN

OBJECTIVE: Treg cells need to acquire an effector phenotype to function in settings of inflammation. Whether effector Treg cells can limit disease severity in lupus is unknown. Interferon regulatory factor 4 (IRF-4) is an essential controller of effector Treg cells and regulates their ability to express interleukin-10 (IL-10). In non-Treg cells, IRF-4 activity is modulated by interactions with DEF-6 and its homolog switch-associated protein 70 (SWAP-70). Although mice lacking both DEF-6 and SWAP-70 (double-knockout [DKO] mice) develop lupus, they display normal survival, suggesting that in DKO mice, Treg cells can moderate disease development. The purpose of this study was to investigate whether Treg cells from DKO mice have an increased capacity to become effector Treg cells due to the ability of DEF-6 and SWAP-70 to restrain IRF-4 activity. METHODS: Treg cells were evaluated by fluorescence-activated cell sorting. The B lymphocyte-induced maturation protein 1 (BLIMP-1)/IL-10 axis was assessed by crossing DKO mice with BLIMP-1-YFP-10BiT dual-reporter mice. Deletion of IRF-4 in Treg cells from DKO mice was achieved by generating FoxP3(Cre) IRF-4(fl/fl) DKO mice. RESULTS: The concomitant absence of DEF-6 and SWAP-70 led to increased numbers of Treg cells, which acquired an effector phenotype in a cell-intrinsic manner. In addition, Treg cells from DKO mice exhibited enhanced expression of the BLIMP-1/IL-10 axis. Notably, DKO effector Treg cells survived and expanded as disease progressed. The accumulation of Treg cells from DKO mice was associated with the up-regulation of genes controlling autophagy. IRF-4 was required for the expansion and function of effector Treg cells from DKO mice. CONCLUSION: This study revealed the existence of mechanisms that, by acting on IRF-4, can fine-tune the function and survival of effector Treg cells in lupus. These findings suggest that the existence of a powerful effector Treg cell compartment that successfully survives in an unfavorable inflammatory environment could limit disease development.


Asunto(s)
Factores Reguladores del Interferón/fisiología , Lupus Eritematoso Sistémico/inmunología , Linfocitos T Reguladores/fisiología , Animales , Proteínas de Unión al ADN/biosíntesis , Femenino , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Masculino , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/biosíntesis , Proteínas Nucleares/biosíntesis , Linfocitos T Reguladores/metabolismo
19.
Science ; 350(6266): 1391-6, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26541605

RESUMEN

More than half of human colorectal cancers (CRCs) carry either KRAS or BRAF mutations and are often refractory to approved targeted therapies. We found that cultured human CRC cells harboring KRAS or BRAF mutations are selectively killed when exposed to high levels of vitamin C. This effect is due to increased uptake of the oxidized form of vitamin C, dehydroascorbate (DHA), via the GLUT1 glucose transporter. Increased DHA uptake causes oxidative stress as intracellular DHA is reduced to vitamin C, depleting glutathione. Thus, reactive oxygen species accumulate and inactivate glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Inhibition of GAPDH in highly glycolytic KRAS or BRAF mutant cells leads to an energetic crisis and cell death not seen in KRAS and BRAF wild-type cells. High-dose vitamin C impairs tumor growth in Apc/Kras(G12D) mutant mice. These results provide a mechanistic rationale for exploring the therapeutic use of vitamin C for CRCs with KRAS or BRAF mutations.


Asunto(s)
Ácido Ascórbico/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Ácido Ascórbico/administración & dosificación , Ácido Ascórbico/farmacología , Línea Celular Tumoral , Ácido Deshidroascórbico/metabolismo , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Ratones , Ratones Mutantes , Ratones Desnudos , Proteínas Proto-Oncogénicas p21(ras)/genética , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...