Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 35(11): 109264, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34133930

RESUMO

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.


Assuntos
Glicólise , Inflamação/metabolismo , Inflamação/patologia , Fatores Reguladores de Interferon/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Bactérias/metabolismo , Citocinas/biossíntese , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imunidade Inata , Mediadores da Inflamação/metabolismo , Ácido Láctico/metabolismo , Lipopolissacarídeos , Masculino , Camundongos Knockout , Proteínas Proto-Oncogênicas c-myc/deficiência
2.
Immunity ; 51(2): 241-257.e9, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31303399

RESUMO

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.


Assuntos
Inflamação/metabolismo , Macrófagos/imunologia , Peritonite/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Dermatopatias/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Epigenômica , Feminino , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , RNA Interferente Pequeno/genética , Receptores Acoplados a Proteínas G/genética , Transcriptoma , Cicatrização
3.
Nat Immunol ; 18(10): 1104-1116, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28825701

RESUMO

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.


Assuntos
Epigênese Genética , Inflamação/etiologia , Inflamação/metabolismo , Interferon Tipo I/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Sítios de Ligação , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Análise por Conglomerados , Biologia Computacional/métodos , Citocinas/genética , Citocinas/metabolismo , Epigenômica/métodos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lipopolissacarídeos/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Motivos de Nucleotídeos , Regiões Promotoras Genéticas , Ligação Proteica , Transporte Proteico , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Fatores de Transcrição/metabolismo
4.
Immunity ; 47(2): 235-250.e4, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813657

RESUMO

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.


Assuntos
Artrite Reumatoide/imunologia , Montagem e Desmontagem da Cromatina , Interferon gama/metabolismo , Macrófagos/imunologia , Proteínas Proto-Oncogênicas c-maf/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Citocinas/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Histonas/metabolismo , Humanos , Ligação Proteica , Proteínas Proto-Oncogênicas c-maf/genética , Transcriptoma
5.
PLoS One ; 12(7): e0179762, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28708839

RESUMO

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.


Assuntos
Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Fibroblastos/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , RNA/química , RNA/isolamento & purificação , RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Sinoviócitos/citologia , Sinoviócitos/efeitos dos fármacos , Sinoviócitos/metabolismo
6.
Immunity ; 47(1): 66-79.e5, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28723554

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Artrite Reumatoide/imunologia , Macrófagos/imunologia , Osteogênese/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Fator de Transcrição E2F1/metabolismo , Feminino , Humanos , Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais
7.
J Clin Invest ; 127(7): 2555-2568, 2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28530645

RESUMO

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.


Assuntos
Diferenciação Celular , Osteoclastos/metabolismo , Osteoporose/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Osteoclastos/patologia , Osteoporose/genética , Osteoporose/patologia , Osteoporose/terapia , Proteínas Proto-Oncogênicas c-myc/genética , Receptores de Estrogênio/genética , Transcriptoma , Receptor ERRalfa Relacionado ao Estrogênio
8.
Cell Rep ; 16(12): 3121-3129, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27653678

RESUMO

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.


Assuntos
Inativação Gênica/fisiologia , Histonas/metabolismo , Interferon gama/biossíntese , Ativação de Macrófagos/genética , Macrófagos/imunologia , Metilação de DNA , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histonas/genética , Humanos , Interferon gama/genética , Lisina , Ativação de Macrófagos/imunologia , Regiões Promotoras Genéticas/genética
9.
Nat Med ; 22(3): 298-305, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26855148

RESUMO

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.


Assuntos
Adenocarcinoma/genética , Neoplasias Ósseas/genética , Evolução Clonal/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Tumores Neuroendócrinos/genética , Neoplasias de Próstata Resistentes à Castração/genética , Receptores Androgênicos/genética , Adenocarcinoma/secundário , Idoso , Idoso de 80 Anos ou mais , Neoplasias Ósseas/secundário , Metilação de DNA , Epigênese Genética , Humanos , Neoplasias Hepáticas/secundário , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/secundário , Estudos Prospectivos , Neoplasias de Próstata Resistentes à Castração/patologia , Estudos Retrospectivos
10.
Science ; 350(6266): 1391-6, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26541605

RESUMO

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.


Assuntos
Ácido Ascórbico/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/farmacologia , Linhagem Celular Tumoral , Ácido Desidroascórbico/metabolismo , Feminino , Transportador de Glucose Tipo 1/metabolismo , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Camundongos , Camundongos Mutantes , Camundongos Nus , Proteínas Proto-Oncogênicas p21(ras)/genética , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nat Immunol ; 16(8): 838-849, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26147685

RESUMO

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.


Assuntos
Interferon gama/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Biossíntese de Proteínas/imunologia , RNA Mensageiro/imunologia , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Células Cultivadas , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/imunologia , Fator de Iniciação 4E em Eucariotos/metabolismo , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Proteínas de Homeodomínio/metabolismo , Humanos , Interferon gama/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , MicroRNAs/genética , Microscopia de Fluorescência , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Complexos Multiproteicos/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia , Serina-Treonina Quinases TOR/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Receptor 2 Toll-Like/metabolismo , Fatores de Transcrição HES-1
12.
Nat Commun ; 5: 5418, 2014 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25391636

RESUMO

Emerging evidence suggests that RANKL-induced changes in chromatin state are important for osteoclastogenesis, but these epigenetic mechanisms are not well understood and have not been therapeutically targeted. In this study, we find that the small molecule I-BET151 that targets bromo and extra-terminal (BET) proteins that 'read' chromatin states by binding to acetylated histones strongly suppresses osteoclastogenesis. I-BET151 suppresses pathologic bone loss in TNF-induced inflammatory osteolysis, inflammatory arthritis and post-ovariectomy models. Transcriptome analysis identifies a MYC-NFAT axis important for osteoclastogenesis. Mechanistically, I-BET151 inhibits expression of the master osteoclast regulator NFATC1 by suppressing expression and recruitment of its newly identified upstream regulator MYC. MYC is elevated in rheumatoid arthritis macrophages and its induction by RANKL is important for osteoclastogenesis and TNF-induced bone resorption. These findings highlight the importance of an I-BET151-inhibited MYC-NFAT axis in osteoclastogenesis, and suggest targeting epigenetic chromatin regulators holds promise for treatment of inflammatory and oestrogen deficiency-mediated pathologic bone resorption.


Assuntos
Reabsorção Óssea/fisiopatologia , Epigênese Genética/fisiologia , Inflamação/fisiopatologia , Osteoclastos/fisiologia , Osteogênese/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/fisiologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoporose/fisiopatologia , Ovariectomia , Ligante RANK/fisiologia
13.
Nat Commun ; 5: 5383, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25415230

RESUMO

The androgen receptor (AR) plays a central role in establishing an oncogenic cascade that drives prostate cancer progression. Some prostate cancers escape androgen dependence and are often associated with an aggressive phenotype. The oestrogen receptor alpha (ERα) is expressed in prostate cancers, independent of AR status. However, the role of ERα remains elusive. Using a combination of chromatin immunoprecipitation (ChIP) and RNA-sequencing data, we identified an ERα-specific non-coding transcriptome signature. Among putatively ERα-regulated intergenic long non-coding RNAs (lncRNAs), we identified nuclear enriched abundant transcript 1 (NEAT1) as the most significantly overexpressed lncRNA in prostate cancer. Analysis of two large clinical cohorts also revealed that NEAT1 expression is associated with prostate cancer progression. Prostate cancer cells expressing high levels of NEAT1 were recalcitrant to androgen or AR antagonists. Finally, we provide evidence that NEAT1 drives oncogenic growth by altering the epigenetic landscape of target gene promoters to favour transcription.


Assuntos
Adenocarcinoma/genética , Epigênese Genética , Receptor alfa de Estrogênio/genética , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Progressão da Doença , Receptor alfa de Estrogênio/metabolismo , Humanos , Masculino , Neoplasias da Próstata/metabolismo , RNA Longo não Codificante/metabolismo , Análise de Sequência de RNA
14.
J Clin Invest ; 124(11): 5057-73, 2014 11.
Artigo em Inglês | MEDLINE | ID: mdl-25329696

RESUMO

Osteoclastogenesis requires activation of RANK signaling as well as costimulatory signals from immunoreceptor tyrosine-based activation motif-containing (ITAM-containing) receptors/adaptors, predominantly tyrosine kinase-binding proteins DAP12 and FcRγ, in osteoclast precursors. It is not well understood how costimulatory signals are regulated and integrated with RANK signaling. Here, we found that osteopetrotic bone phenotypes in mice lacking DAP12 or DAP12 and FcRγ are mediated by the transcription factor RBP-J, as deletion of Rbpj in these mice substantially rescued the defects of bone remodeling. Using a TNF-α-induced model of inflammatory bone resorption, we determined that RBP-J deficiency enables TNF-α to induce osteoclast formation and bone resorption in DAP12-deficient animals. Thus, RBP-J imposes a requirement for ITAM-mediated costimulation of RANKL or TNF-α-induced osteoclastogenesis. Mechanistically, RBP-J suppressed induction of key osteoclastogenic factors NFATc1, BLIMP1, and c-FOS by inhibiting ITAM-mediated expression and function of PLCγ2 and activation of downstream calcium-CaMKK/PYK2 signaling. Moreover, RBP-J suppressed Plcg2 expression and downstream calcium oscillations indirectly by a TGF-ß/PLCγ2/calcium axis. Together, our findings indicate that RBP-J suppresses ITAM-mediated costimulation, thereby limiting crosstalk between ITAM and RANK/TNFR signaling and allowing fine tuning of osteoclastogenesis during bone homeostasis and under inflammatory conditions. Furthermore, these data suggest that environmental cues that regulate RBP-J expression/function potentially modulate the requirement for costimulatory signaling for osteoclast differentiation and bone remodeling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/fisiologia , Osteoclastos/fisiologia , Receptores de IgG/metabolismo , Animais , Reabsorção Óssea , Sinalização do Cálcio , Diferenciação Celular , Células Cultivadas , Camundongos Knockout , Fatores de Transcrição NFATC/metabolismo , Fosfolipase C gama/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/fisiologia
15.
PLoS Genet ; 10(9): e1004566, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25188243

RESUMO

Overexpression of the histone methyltransferase MMSET in t(4;14)+ multiple myeloma patients is believed to be the driving factor in the pathogenesis of this subtype of myeloma. MMSET catalyzes dimethylation of lysine 36 on histone H3 (H3K36me2), and its overexpression causes a global increase in H3K36me2, redistributing this mark in a broad, elevated level across the genome. Here, we demonstrate that an increased level of MMSET also induces a global reduction of lysine 27 trimethylation on histone H3 (H3K27me3). Despite the net decrease in H3K27 methylation, specific genomic loci exhibit enhanced recruitment of the EZH2 histone methyltransferase and become hypermethylated on this residue. These effects likely contribute to the myeloma phenotype since MMSET-overexpressing cells displayed increased sensitivity to EZH2 inhibition. Furthermore, we demonstrate that such MMSET-mediated epigenetic changes require a number of functional domains within the protein, including PHD domains that mediate MMSET recruitment to chromatin. In vivo, targeting of MMSET by an inducible shRNA reversed histone methylation changes and led to regression of established tumors in athymic mice. Together, our work elucidates previously unrecognized interplay between MMSET and EZH2 in myeloma oncogenesis and identifies domains to be considered when designing inhibitors of MMSET function.


Assuntos
Metilação de DNA/genética , Epigênese Genética/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Mieloma Múltiplo/genética , Complexo Repressor Polycomb 2/metabolismo , Ligação Proteica/genética , Animais , Linhagem Celular , Transformação Celular Neoplásica/genética , Cromatina/genética , Feminino , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Lisina/genética , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/metabolismo , Complexo Repressor Polycomb 2/genética , RNA Interferente Pequeno/genética
16.
J Immunol ; 193(5): 2373-83, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25057003

RESUMO

Mesenchymal stromal cells have emerged as powerful modulators of the immune system. In this study, we explored how the human macrophage response to TNF is regulated by human synovial fibroblasts, the representative stromal cell type in the synovial lining of joints that become activated during inflammatory arthritis. We found that synovial fibroblasts strongly suppressed TNF-mediated induction of an IFN-ß autocrine loop and downstream expression of IFN-stimulated genes (ISGs), including chemokines CXCL9 and CXCL10 that are characteristic of classical macrophage activation. TNF induced the production of soluble synovial fibroblast factors that suppressed the macrophage production of IFN-ß, and cooperated with TNF to limit the responsiveness of macrophages to IFN-ß by suppressing activation of Jak-STAT signaling. Genome-wide transcriptome analysis showed that cocultured synovial fibroblasts modulate the expression of approximately one third of TNF-regulated genes in macrophages, including genes in pathways important for macrophage survival and polarization toward an alternatively activated phenotype. Pathway analysis revealed that gene expression programs regulated by synovial fibroblasts in our coculture system were also regulated in rheumatoid arthritis synovial macrophages, suggesting that these fibroblast-mediated changes may contribute to rheumatoid arthritis pathogenesis. This work furthers our understanding of the interplay between innate immune and stromal cells during an inflammatory response, one that is particularly relevant to inflammatory arthritis. Our findings also identify modulation of macrophage phenotype as a new function for synovial fibroblasts that may prove to be a contributing factor in arthritis pathogenesis.


Assuntos
Artrite Reumatoide/imunologia , Fibroblastos/imunologia , Macrófagos/imunologia , Transdução de Sinais/imunologia , Membrana Sinovial/imunologia , Fator de Necrose Tumoral alfa/imunologia , Artrite Reumatoide/patologia , Comunicação Autócrina/imunologia , Células Cultivadas , Quimiocina CXCL10/imunologia , Quimiocina CXCL9/imunologia , Técnicas de Cocultura , Feminino , Fibroblastos/patologia , Estudo de Associação Genômica Ampla , Humanos , Interferon beta/imunologia , Janus Quinases/imunologia , Macrófagos/patologia , Masculino , Fatores de Transcrição STAT/imunologia , Membrana Sinovial/patologia , Transcrição Gênica/imunologia
17.
Immunity ; 39(3): 454-69, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-24012417

RESUMO

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.


Assuntos
Montagem e Desmontagem da Cromatina , Citocinas/metabolismo , Interferon gama/metabolismo , Receptores Toll-Like/metabolismo , Acetilação , Células Cultivadas , Ativação Enzimática , Histonas/metabolismo , Humanos , Fator Regulador 1 de Interferon/metabolismo , Subunidade p40 da Interleucina-12/metabolismo , Interleucina-6/metabolismo , Janus Quinases/antagonistas & inibidores , Macrófagos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Polimerase II/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Transcrição Gênica , Fatores de Necrose Tumoral/metabolismo
18.
Cell Rep ; 4(3): 578-88, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23911289

RESUMO

The BCL6 transcriptional repressor is required for the development of germinal center (GC) B cells and diffuse large B cell lymphomas (DLBCLs). Although BCL6 can recruit multiple corepressors, its transcriptional repression mechanism of action in normal and malignant B cells is unknown. We find that in B cells, BCL6 mostly functions through two independent mechanisms that are collectively essential to GC formation and DLBCL, both mediated through its N-terminal BTB domain. These are (1) the formation of a unique ternary BCOR-SMRT complex at promoters, with each corepressor binding to symmetrical sites on BCL6 homodimers linked to specific epigenetic chromatin features, and (2) the "toggling" of active enhancers to a poised but not erased conformation through SMRT-dependent H3K27 deacetylation, which is mediated by HDAC3 and opposed by p300 histone acetyltransferase. Dynamic toggling of enhancers provides a basis for B cells to undergo rapid transcriptional and phenotypic changes in response to signaling or environmental cues.


Assuntos
Linfócitos B/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Animais , Linfócitos B/metabolismo , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/metabolismo , Camundongos , Modelos Moleculares , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-bcl-6 , Transdução de Sinais
19.
Neoplasia ; 15(4): 373-83, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23555183

RESUMO

Although prostate cancer (PCa) is the second leading cause of cancer death among men worldwide, not all men diagnosed with PCa will die from the disease. A critical challenge, therefore, is to distinguish indolent PCa from more advanced forms to guide appropriate treatment decisions. We used Enhanced Reduced Representation Bisulfite Sequencing, a genome-wide high-coverage single-base resolution DNA methylation method to profile seven localized PCa samples, seven matched benign prostate tissues, and six aggressive castration-resistant prostate cancer (CRPC) samples. We integrated these data with RNA-seq and whole-genome DNA-seq data to comprehensively characterize the PCa methylome, detect changes associated with disease progression, and identify novel candidate prognostic biomarkers. Our analyses revealed the correlation of cytosine guanine dinucleotide island (CGI)-specific hypermethylation with disease severity and association of certain breakpoints (deletion, tandem duplications, and interchromosomal translocations) with DNA methylation. Furthermore, integrative analysis of methylation and single-nucleotide polymorphisms (SNPs) uncovered widespread allele-specific methylation (ASM) for the first time in PCa. We found that most DNA methylation changes occurred in the context of ASM, suggesting that variations in tumor epigenetic landscape of individuals are partly mediated by genetic differences, which may affect PCa disease progression. We further selected a panel of 13 CGIs demonstrating increased DNA methylation with disease progression and validated this panel in an independent cohort of 20 benign prostate tissues, 16 PCa, and 8 aggressive CRPCs. These results warrant clinical evaluation in larger cohorts to help distinguish indolent PCa from advanced disease.


Assuntos
Adenocarcinoma/genética , Biomarcadores Tumorais/genética , Epigênese Genética , Neoplasias da Próstata/genética , Adenocarcinoma/patologia , Idoso , Pontos de Quebra do Cromossomo , Ilhas de CpG , Metilação de DNA , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Diagnóstico Molecular , Mutação , Gradação de Tumores , Polimorfismo de Nucleotídeo Único , Prognóstico , Neoplasias da Próstata/patologia , Análise de Sequência de DNA
20.
Nature ; 495(7439): 98-102, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23389444

RESUMO

Adult stem cells reside in specialized niches where they receive environmental cues to maintain tissue homeostasis. In mammals, the stem cell niche within hair follicles is home to epithelial hair follicle stem cells and melanocyte stem cells, which sustain cyclical bouts of hair regeneration and pigmentation. To generate pigmented hairs, synchrony is achieved such that upon initiation of a new hair cycle, stem cells of each type activate lineage commitment. Dissecting the inter-stem-cell crosstalk governing this intricate coordination has been difficult, because mutations affecting one lineage often affect the other. Here we identify transcription factor NFIB as an unanticipated coordinator of stem cell behaviour. Hair follicle stem-cell-specific conditional targeting of Nfib in mice uncouples stem cell synchrony. Remarkably, this happens not by perturbing hair cycle and follicle architecture, but rather by promoting melanocyte stem cell proliferation and differentiation. The early production of melanin is restricted to melanocyte stem cells at the niche base. Melanocyte stem cells more distant from the dermal papilla are unscathed, thereby preventing hair greying typical of melanocyte stem cell differentiation mutants. Furthermore, we pinpoint KIT-ligand as a dermal papilla signal promoting melanocyte stem cell differentiation. Additionally, through chromatin-immunoprecipitation with high-throughput-sequencing and transcriptional profiling, we identify endothelin 2 (Edn2) as an NFIB target aberrantly activated in NFIB-deficient hair follicle stem cells. Ectopically induced Edn2 recapitulates NFIB-deficient phenotypes in wild-type mice. Conversely, endothelin receptor antagonists and/or KIT blocking antibodies prevent precocious melanocyte stem cell differentiation in the NFIB-deficient niche. Our findings reveal how melanocyte and hair follicle stem cell behaviours maintain reliance upon cooperative factors within the niche, and how this can be uncoupled in injury, stress and disease states.


Assuntos
Folículo Piloso/citologia , Melanócitos/citologia , Fatores de Transcrição NFI/metabolismo , Nicho de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Imunoprecipitação da Cromatina , Endotelina-2/genética , Endotelina-2/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Cabelo/citologia , Cabelo/crescimento & desenvolvimento , Cor de Cabelo , Folículo Piloso/metabolismo , Melanócitos/metabolismo , Camundongos , Fatores de Transcrição NFI/deficiência , Fatores de Transcrição NFI/genética , Análise de Sequência , Fator de Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA