Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 17568, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30514857

RESUMO

Long noncoding RNAs (lncRNAs) constitute a significant fraction of mammalian transcriptomes and they have emerged as intricate regulators of many biological processes. Their broad capacity to adopt diverse structures facilitates their involvement in the transcriptional, translational and signaling processes that are central to embryonic stem (ES) cell self-renewal and pluripotency. While lncRNAs have been implicated in ES cell maintenance, detailed analyses of those that show significant expression in ES cells is largely absent. Moreover, cooperative molecular relationships that facilitate lncRNA action are poorly understood. Cyrano is a developmentally important lncRNA, and in ES cells, it supports gene expression network maintenance, cell adhesion and cell survival. We have interrogated the interactome of Cyrano to identify protein partners and find that Cyrano is involved in multiple protein networks. We identify a developmentally important cell-signaling hub and find STAT3 as a candidate through which Cyrano can function to reinforce self-renewal of ES cells. Based on commonalities between ES cells and cancer cells, we postulate such functional interactions may support cell proliferation, cell identity and adhesion characteristics in rapidly proliferating cell types. The interactome data will therefore provide a resource for further investigations into interactions that regulate Cyrano or mediate its function.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Transcriptoma/genética , Animais , Adesão Celular/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Autorrenovação Celular/genética , Células-Tronco Embrionárias/citologia , Redes Reguladoras de Genes , Humanos , Camundongos
2.
J Clin Invest ; 128(10): 4727-4741, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30222134

RESUMO

Chronic inflammatory demyelinating polyneuropathy (CIDP) and Guillain-Barre syndrome (GBS) are inflammatory neuropathies that affect humans and are characterized by peripheral nerve myelin destruction and macrophage-containing immune infiltrates. In contrast to the traditional view that the peripheral nerve is simply the target of autoimmunity, we report here that peripheral nerve Schwann cells exacerbate the autoimmune process through extracellular matrix (ECM) protein induction. In a spontaneous autoimmune peripheral polyneuropathy (SAPP) mouse model of inflammatory neuropathy and CIDP nerve biopsies, the ECM protein periostin (POSTN) was upregulated in affected sciatic nerves and was primarily expressed by Schwann cells. Postn deficiency delayed the onset and reduced the extent of neuropathy, as well as decreased the number of macrophages infiltrating the sciatic nerve. In an in vitro assay, POSTN promoted macrophage chemotaxis in an integrin-AM (ITGAM) and ITGAV-dependent manner. The PNS-infiltrating macrophages in SAPP-affected nerves were pathogenic, since depletion of macrophages protected against the development of neuropathy. Our findings show that Schwann cells promote macrophage infiltration by upregulating Postn and suggest that POSTN is a novel target for the treatment of macrophage-associated inflammatory neuropathies.


Assuntos
Moléculas de Adesão Celular/imunologia , Macrófagos/imunologia , Células de Schwann/imunologia , Animais , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Moléculas de Adesão Celular/genética , Humanos , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/genética , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/patologia , Células de Schwann/patologia
3.
JCI Insight ; 2(18)2017 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-28931755

RESUMO

Blockade of immune checkpoint proteins (e.g., CTLA-4, PD-1) improves overall survival in advanced melanoma; however, therapeutic benefit is limited to only a subset of patients. Because checkpoint blockade acts by "removing the brakes" on effector T cells, the efficacy of checkpoint blockade may be constrained by the limited pool of melanoma-reactive T cells in the periphery. In the thymus, autoimmune regulator (Aire) promotes deletion of T cells reactive against self-antigens that are also expressed by tumors. Thus, while protecting against autoimmunity, Aire also limits the generation of melanoma-reactive T cells. Here, we show that Aire deficiency in mice expands the pool of CD4+ T cells capable of melanoma cell eradication and has additive effects with anti-CTLA-4 antibody in slowing melanoma tumor growth and increasing survival. Moreover, pharmacologic blockade of central T cell tolerance and peripheral checkpoint blockade in combination enhanced antimelanoma immunity in a synergistic manner. In melanoma patients treated with anti-CTLA-4 antibody, clinical response to therapy was associated with a human Aire polymorphism. Together, these findings suggest that Aire-mediated central tolerance constrains the efficacy of peripheral checkpoint inhibition and point to simultaneous blockade of Aire and checkpoint inhibitors as a novel strategy to enhance antimelanoma immunity.


Assuntos
Antígeno CTLA-4/antagonistas & inibidores , Tolerância Central , Melanoma/imunologia , Animais , Anticorpos Monoclonais/imunologia , Linfócitos T CD4-Positivos/imunologia , Citometria de Fluxo , Xenoenxertos , Humanos , Imunoterapia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Fatores de Transcrição/genética , Proteína AIRE
4.
BMC Bioinformatics ; 17: 144, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27009150

RESUMO

BACKGROUND: Correctly identifying genomic regions enriched with histone modifications and transcription factors is key to understanding their regulatory and developmental roles. Conceptually, these regions are divided into two categories, narrow peaks and broad domains, and different algorithms are used to identify each one. Datasets that span these two categories are often analyzed with a single program for peak calling combined with an ad hoc method for domains. RESULTS: We developed hiddenDomains, which identifies both peaks and domains, and compare it to the leading algorithms using H3K27me3, H3K36me3, GABP, ESR1 and FOXA ChIP-seq datasets. The output from the programs was compared to qPCR-validated enriched and depleted sites, predicted transcription factor binding sites, and highly-transcribed gene bodies. With every method, hiddenDomains, performed as well as, if not better than algorithms dedicated to a specific type of analysis. CONCLUSIONS: hiddenDomains performs as well as the best domain and peak calling algorithms, making it ideal for analyzing ChIP-seq datasets, especially those that contain a mixture of peaks and domains.


Assuntos
Algoritmos , Imunoprecipitação da Cromatina , Receptor alfa de Estrogênio/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Histonas/metabolismo , Humanos , Cadeias de Markov
5.
Immunity ; 43(4): 703-14, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26431949

RESUMO

Epigenetic changes, including histone methylation, control T cell differentiation and memory formation, though the enzymes that mediate these processes are not clear. We show that UTX, a histone H3 lysine 27 (H3K27) demethylase, supports T follicular helper (Tfh) cell responses that are essential for B cell antibody generation and the resolution of chronic viral infections. Mice with a T cell-specific UTX deletion had fewer Tfh cells, reduced germinal center responses, lacked virus-specific immunoglobulin G (IgG), and were unable to resolve chronic lymphocytic choriomeningitis virus infections. UTX-deficient T cells showed decreased expression of interleukin-6 receptor-α and other Tfh cell-related genes that were associated with increased H3K27 methylation. Additionally, Turner Syndrome subjects, who are predisposed to chronic ear infections, had reduced UTX expression in immune cells and decreased circulating CD4(+) CXCR5(+) T cell frequency. Thus, we identify a critical link between UTX in T cells and immunity to infection.


Assuntos
Histona Desmetilases/deficiência , Histona Desmetilases/fisiologia , Vírus da Coriomeningite Linfocítica/imunologia , Proteínas Nucleares/deficiência , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Viremia/imunologia , Animais , Anticorpos Antivirais/biossíntese , Diferenciação Celular , Feminino , Dosagem de Genes , Regulação da Expressão Gênica/imunologia , Predisposição Genética para Doença , Histonas/metabolismo , Humanos , Memória Imunológica , Subunidade alfa de Receptor de Interleucina-6/biossíntese , Subunidade alfa de Receptor de Interleucina-6/genética , Cooperação Linfocítica , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Metilação , Camundongos , Modelos Imunológicos , Otite Média/etiologia , Processamento de Proteína Pós-Traducional , Receptores CXCR5/análise , Especificidade da Espécie , Subpopulações de Linfócitos T/enzimologia , Subpopulações de Linfócitos T/virologia , Linfócitos T Auxiliares-Indutores/enzimologia , Linfócitos T Auxiliares-Indutores/virologia , Transcrição Gênica , Síndrome de Turner/complicações , Síndrome de Turner/enzimologia , Virulência , Inativação do Cromossomo X
6.
Genes Dev ; 29(13): 1377-92, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26159997

RESUMO

Histone H3.3 is a highly conserved histone H3 replacement variant in metazoans and has been implicated in many important biological processes, including cell differentiation and reprogramming. Germline and somatic mutations in H3.3 genomic incorporation pathway components or in H3.3 encoding genes have been associated with human congenital diseases and cancers, respectively. However, the role of H3.3 in mammalian development remains unclear. To address this question, we generated H3.3-null mouse models through classical genetic approaches. We found that H3.3 plays an essential role in mouse development. Complete depletion of H3.3 leads to developmental retardation and early embryonic lethality. At the cellular level, H3.3 loss triggers cell cycle suppression and cell death. Surprisingly, H3.3 depletion does not dramatically disrupt gene regulation in the developing embryo. Instead, H3.3 depletion causes dysfunction of heterochromatin structures at telomeres, centromeres, and pericentromeric regions of chromosomes, leading to mitotic defects. The resulting karyotypical abnormalities and DNA damage lead to p53 pathway activation. In summary, our results reveal that an important function of H3.3 is to support chromosomal heterochromatic structures, thus maintaining genome integrity during mammalian development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Instabilidade Genômica/genética , Crescimento e Desenvolvimento/genética , Histonas/metabolismo , Animais , Morte Celular/genética , Linhagem Celular , Proliferação de Células/genética , Células Cultivadas , Fertilidade/genética , Genes Letais/genética , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/genética , Camundongos , Mutação
7.
Nat Commun ; 6: 6118, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25625625

RESUMO

Ovarian clear-cell carcinoma (OCCC) is an aggressive form of ovarian cancer with high ARID1A mutation rates. Here we present a mutant mouse model of OCCC. We find that ARID1A inactivation is not sufficient for tumour formation, but requires concurrent activation of the phosphoinositide 3-kinase catalytic subunit, PIK3CA. Remarkably, the mice develop highly penetrant tumours with OCCC-like histopathology, culminating in haemorrhagic ascites and a median survival period of 7.5 weeks. Therapeutic treatment with the pan-PI3K inhibitor, BKM120, prolongs mouse survival by inhibiting the tumour cell growth. Cross-species gene expression comparisons support a role for IL-6 inflammatory cytokine signalling in OCCC pathogenesis. We further show that ARID1A and PIK3CA mutations cooperate to promote tumour growth through sustained IL-6 overproduction. Our findings establish an epistatic relationship between SWI/SNF chromatin remodelling and PI3K pathway mutations in OCCC and demonstrate that these pathways converge on pro-tumorigenic cytokine signalling. We propose that ARID1A protects against inflammation-driven tumorigenesis.


Assuntos
Adenocarcinoma de Células Claras/genética , Carcinogênese/genética , Citocinas/metabolismo , Proteínas de Ligação a DNA/genética , Inflamação/metabolismo , Mutação/genética , Proteínas Nucleares/genética , Neoplasias Ovarianas/genética , Fosfatidilinositol 3-Quinases/genética , Adenocarcinoma de Células Claras/tratamento farmacológico , Adenocarcinoma de Células Claras/patologia , Alelos , Animais , Carcinogênese/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Genes Supressores de Tumor , Haploinsuficiência/efeitos dos fármacos , Inflamação/patologia , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA