Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 17(5): e1009553, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33945523

RESUMO

The CBFB gene is frequently mutated in several types of solid tumors. Emerging evidence suggests that CBFB is a tumor suppressor in breast cancer. However, our understanding of the tumor suppressive function of CBFB remains incomplete. Here, we analyze genetic interactions between mutations of CBFB and other highly mutated genes in human breast cancer datasets and find that CBFB and TP53 mutations are mutually exclusive, suggesting a functional association between CBFB and p53. Integrated genomic studies reveal that TAp73 is a common transcriptional target of CBFB and p53. CBFB cooperates with p53 to maintain TAp73 expression, as either CBFB or p53 loss leads to TAp73 depletion. TAp73 re-expression abrogates the tumorigenic effect of CBFB deletion. Although TAp73 loss alone is insufficient for tumorigenesis, it enhances the tumorigenic effect of NOTCH3 overexpression, a downstream event of CBFB loss. Immunohistochemistry shows that p73 loss is coupled with higher proliferation in xenografts. Moreover, TAp73 loss-of-expression is a frequent event in human breast cancer tumors and cell lines. Together, our results significantly advance our understanding of the tumor suppressive functions of CBFB and reveal a mechanism underlying the communication between the two tumor suppressors CBFB and p53.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Subunidade beta de Fator de Ligação ao Core/genética , Regulação Neoplásica da Expressão Gênica , Proteína Tumoral p73/genética , Proteína Supressora de Tumor p53/genética , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/metabolismo , Feminino , Genes Supressores de Tumor , Humanos , Imuno-Histoquímica , Camundongos , Mutação , Receptor Notch3/genética , Receptor Notch3/metabolismo , Transcrição Gênica , Proteína Tumoral p73/deficiência , Proteína Tumoral p73/metabolismo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Dis Model Mech ; 12(6)2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31171577

RESUMO

Core binding factor ß (Cbfb) is a cofactor of the Runx family of transcription factors. Among these transcription factors, Runx1 is a prerequisite for anterior-specific palatal fusion. It was previously unclear, however, whether Cbfb served as a modulator or as an obligatory factor in the Runx signaling process that regulates palatogenesis. Here, we report that Cbfb is essential and indispensable in mouse anterior palatogenesis. Palatal fusion in Cbfb mutants is disrupted owing to failed disintegration of the fusing epithelium specifically at the anterior portion, as observed in Runx1 mutants. In these mutants, expression of TGFB3 is disrupted in the area of failed palatal fusion, in which phosphorylation of Stat3 is also affected. TGFB3 protein has been shown to rescue palatal fusion in vitro TGFB3 also activated Stat3 phosphorylation. Strikingly, the anterior cleft palate in Cbfb mutants is further rescued by pharmaceutical application of folic acid, which activates suppressed Stat3 phosphorylation and Tgfb3 expression in vitro With these findings, we provide the first evidence that Cbfb is a prerequisite for anterior palatogenesis and acts as an obligatory cofactor in the Runx1/Cbfb-Stat3-Tgfb3 signaling axis. Furthermore, the rescue of the mutant cleft palate using folic acid might highlight potential therapeutic targets aimed at Stat3 modification for the prevention and pharmaceutical intervention of cleft palate.


Assuntos
Fissura Palatina/tratamento farmacológico , Fissura Palatina/patologia , Subunidade beta de Fator de Ligação ao Core/deficiência , Ácido Fólico/uso terapêutico , Animais , Fissura Palatina/genética , Subunidade beta de Fator de Ligação ao Core/metabolismo , Epitélio/efeitos dos fármacos , Epitélio/patologia , Ácido Fólico/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Modelos Biológicos , Mutação/genética , Organogênese/efeitos dos fármacos , Palato/anormalidades , Palato/embriologia , Palato/patologia , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Crescimento Transformador beta3/metabolismo
3.
Nat Commun ; 10(1): 447, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30683858

RESUMO

Group 2 innate lymphoid cells (ILC2s) have tissue-resident competence and contribute to the pathogenesis of allergic diseases. However, the mechanisms regulating prolonged ILC2-mediated TH2 cytokine production under chronic inflammatory conditions are unclear. Here we show that, at homeostasis, Runx deficiency induces excessive ILC2 activation due to overly active GATA-3 functions. By contrast, during allergic inflammation, the absence of Runx impairs the ability of ILC2s to proliferate and produce effector TH2 cytokines and chemokines. Instead, functional deletion of Runx induces the expression of exhaustion markers, such as IL-10 and TIGIT, on ILC2s. Finally, these 'exhausted-like' ILC2s are unable to induce type 2 immune responses to repeated allergen exposures. Thus, Runx confers competence for sustained ILC2 activity at the mucosa, and contributes to allergic pathogenesis.


Assuntos
Asma/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/imunologia , Subunidade beta de Fator de Ligação ao Core/imunologia , Imunidade Inata , Pulmão/imunologia , Linfócitos/imunologia , Animais , Asma/induzido quimicamente , Asma/genética , Asma/patologia , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Modelos Animais de Doenças , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/imunologia , Regulação da Expressão Gênica/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/imunologia , Intestino Delgado/patologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Ativação Linfocitária , Linfócitos/classificação , Linfócitos/efeitos dos fármacos , Linfócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Papaína/administração & dosagem , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Transdução de Sinais , Baço/efeitos dos fármacos , Baço/imunologia , Baço/patologia
4.
Exp Hematol ; 68: 2-9, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30391350

RESUMO

The transcription factor RUNX1 is required in the embryo for formation of the adult hematopoietic system. Here, we describe the seminal findings that led to the discovery of RUNX1 and of its critical role in blood cell formation in the embryo from hemogenic endothelium (HE). We also present RNA-sequencing data demonstrating that HE cells in different anatomic sites, which produce hematopoietic progenitors with dissimilar differentiation potentials, are molecularly distinct. Hemogenic and non-HE cells in the yolk sac are more closely related to each other than either is to hemogenic or non-HE cells in the major arteries. Therefore, a major driver of the different lineage potentials of the committed erythro-myeloid progenitors that emerge in the yolk sac versus hematopoietic stem cells that originate in the major arteries is likely to be the distinct molecular properties of the HE cells from which they are derived. We used bioinformatics analyses to predict signaling pathways active in arterial HE, which include the functionally validated pathways Notch, Wnt, and Hedgehog. We also used a novel bioinformatics approach to assemble transcriptional regulatory networks and predict transcription factors that may be specifically involved in hematopoietic cell formation from arterial HE, which is the origin of the adult hematopoietic system.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Hemangioblastos/fisiologia , Hematopoese/fisiologia , Animais , Artérias/citologia , Artérias/embriologia , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/fisiologia , Proteínas de Drosophila/genética , Sangue Fetal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Leucemia Experimental/genética , Leucemia Experimental/virologia , Leucemia Mieloide Aguda/genética , Camundongos , Camundongos Knockout , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Transcrição Gênica , Saco Vitelino/citologia
5.
Exp Hematol ; 64: 84-96, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29733873

RESUMO

RUNX1 is frequently mutated in T-cell acute lymphoblastic leukemia (T-ALL). The spectrum of RUNX1 mutations has led to the notion that it acts as a tumor suppressor in this context; however, other studies have placed RUNX1, along with transcription factors TAL1 and NOTCH1, as core drivers of an oncogenic transcriptional program. To reconcile these divergent roles, we knocked down RUNX1 in human T-ALL cell lines and deleted Runx1 or Cbfb in primary mouse T-cell leukemias. RUNX1 depletion consistently resulted in reduced cell proliferation and increased apoptosis. RUNX1 upregulated variable sets of target genes in each cell line, but consistently included a core set of oncogenic effectors including insulin-like growth factor 1 receptor (IGF1R) and NRAS. Our results support the conclusion that RUNX1 has a net positive effect on cell growth in the context of established T-ALL.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Regulação Leucêmica da Expressão Gênica/genética , Proteínas de Neoplasias/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Animais , Divisão Celular , Linhagem Celular Tumoral , Tamanho Celular , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Deleção de Genes , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Leucemia Experimental/genética , Leucemia Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Interferência de RNA , RNA Interferente Pequeno/genética , Transcrição Gênica , Transcriptoma , Carga Tumoral
6.
J Exp Med ; 215(2): 595-610, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29343500

RESUMO

Multipotent hematopoietic progenitors must acquire thymus-homing capacity to initiate T lymphocyte development. Despite its importance, the transcriptional program underlying this process remains elusive. Cbfß forms transcription factor complexes with Runx proteins, and here we show that Cbfß2, encoded by an RNA splice variant of the Cbfb gene, is essential for extrathymic differentiation of T cell progenitors. Furthermore, Cbfß2 endows extrathymic progenitors with thymus-homing capacity by inducing expression of the principal thymus-homing receptor, Ccr9. This occurs via direct binding of Cbfß2 to cell type-specific enhancers, as is observed in Rorγt induction during differentiation of lymphoid tissue inducer cells by activation of an intronic enhancer. As in mice, an alternative splicing event in zebrafish generates a Cbfß2-specific mRNA, important for ccr9 expression. Thus, despite phylogenetically and ontogenetically variable sites of origin of T cell progenitors, their robust thymus-homing capacity is ensured by an evolutionarily conserved mechanism emerging from functional diversification of Runx transcription factor complexes by acquisition of a novel splice variant.


Assuntos
Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/imunologia , Células Precursoras de Linfócitos T/citologia , Células Precursoras de Linfócitos T/imunologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/imunologia , Processamento Alternativo , Animais , Diferenciação Celular , Linhagem da Célula , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/deficiência , Elementos Facilitadores Genéticos , Evolução Molecular , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Knockout , Camundongos Mutantes , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , RNA Mensageiro/genética , Receptores CCR/genética , Receptores CCR/imunologia , Especificidade da Espécie , Timo/citologia , Timo/embriologia , Timo/imunologia , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência
7.
Sci Immunol ; 2(18)2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29222089

RESUMO

Natural killer (NK) cells are innate lymphocytes that have features of adaptive immunity such as clonal expansion and generation of long-lived memory. Interleukin-12 (IL-12) signaling through its downstream transcription factor signal transducer and activator of transcription 4 (STAT4) is required for the generation of memory NK cells after expansion. We identify gene loci that are highly enriched for STAT4 binding using chromatin immunoprecipitation sequencing for STAT4 and the permissive histone mark H3K4me3 in activated NK cells. We found that promoter regions of Runx1 and Runx3 are targets of STAT4 and that STAT4 binding during NK cell activation induces epigenetic modifications of Runx gene loci resulting in increased expression. Furthermore, specific ablation of Runx1, Runx3, or their binding partner Cbfb in NK cells resulted in defective clonal expansion and memory formation during viral infection, with evidence for Runx1-mediated control of a cell cycle program. Thus, our study reveals a mechanism whereby STAT4-mediated epigenetic control of individual Runx transcription factors promotes the adaptive behavior of antiviral NK cells.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/metabolismo , Células Matadoras Naturais/imunologia , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 3 de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT4/metabolismo
8.
J Exp Med ; 214(10): 2933-2946, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28814567

RESUMO

The mouse Langerhans cell (LC) network is established through the differentiation of embryonic LC precursors. BMP7 and TGFß1 initiate cellular signaling that is essential for inducing LC differentiation and preserving LCs in a quiescent state, respectively. Here we show that loss of Cbfß2, one of two RNA splice variants of the Cbfb gene, results in long-term persistence of embryonic LC precursors after their developmental arrest at the transition into the EpCAM+ stage. This phenotype is caused by selective loss of BMP7-mediated signaling essential for LC differentiation, whereas TGFßR signaling is intact, maintaining cells in a quiescent state. Transgenic Cbfß2 expression at the neonatal stage, but not at the adult stage, restored differentiation from Cbfß2-deficient LC precursors. Loss of developmental potential in skin-residential precursor cells was accompanied by diminished BMP7-BMPR1A signaling. Collectively, our results reveal an essential requirement for the Cbfß2 variant in LC differentiation and provide novel insight into how the establishment and homeostasis of the LC network is regulated.


Assuntos
Subunidade beta de Fator de Ligação ao Core/deficiência , Células de Langerhans/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , Proteína Morfogenética Óssea 7/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Diferenciação Celular/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia
9.
Nat Immunol ; 16(11): 1124-33, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26414766

RESUMO

Subsets of innate lymphoid cells (ILCs) reside in the mucosa and regulate immune responses to external pathogens. While ILCs can be phenotypically classified into ILC1, ILC2 and ILC3 subsets, the transcriptional control of commitment to each ILC lineage is incompletely understood. Here we report that the transcription factor Runx3 was essential for the normal development of ILC1 and ILC3 cells but not of ILC2 cells. Runx3 controlled the survival of ILC1 cells but not of ILC3 cells. Runx3 was required for expression of the transcription factor RORγt and its downstream target, the transcription factor AHR, in ILC3 cells. The absence of Runx3 in ILCs exacerbated infection with Citrobacter rodentium. Therefore, our data establish Runx3 as a key transcription factor in the lineage-specific differentiation of ILC1 and ILC3 cells.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Imunidade Inata , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Animais , Antígenos Ly/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Citrobacter rodentium/imunologia , Citrobacter rodentium/patogenicidade , Subunidade alfa 3 de Fator de Ligação ao Core/deficiência , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/metabolismo , Infecções por Enterobacteriaceae/etiologia , Infecções por Enterobacteriaceae/imunologia , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Subpopulações de Linfócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/deficiência , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo
11.
J Bone Miner Res ; 29(7): 1564-1574, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24821091

RESUMO

Core binding factor beta (Cbfß) is essential for embryonic bone morphogenesis. Yet the mechanisms by which Cbfß regulates chondrocyte proliferation and differentiation as well as postnatal cartilage and bone formation remain unclear. Hence, using paired-related homeobox transcription factor 1-Cre (Prx1-Cre) mice, mesenchymal stem cell-specific Cbfß-deficient (Cbfß(f/f) Prx1-Cre) mice were generated to study the role of Cbfß in postnatal cartilage and bone development. These mutant mice survived to adulthood but exhibited severe sternum and limb malformations. Sternum ossification was largely delayed in the Cbfß(f/f) Prx1-Cre mice and the xiphoid process was noncalcified and enlarged. In newborn and 7-day-old Cbfß(f/f) Prx1-Cre mice, the resting zone was dramatically elongated, the proliferation zone and hypertrophic zone of the growth plates were drastically shortened and disorganized, and trabecular bone formation was reduced. Moreover, in 1-month-old Cbfß(f/f) Prx1-Cre mice, the growth plates were severely deformed and trabecular bone was almost absent. In addition, Cbfß deficiency impaired intramembranous bone formation both in vivo and in vitro. Interestingly, although the expression of Indian hedgehog (Ihh) was largely reduced, the expression of parathyroid hormone-related protein (PTHrP) receptor (PPR) was dramatically increased in the Cbfß(f/f) Prx1-Cre growth plate, indicating that that Cbfß deficiency disrupted the Ihh-PTHrP negative regulatory loop. Chromatin immunoprecipitation (ChIP) analysis and promoter luciferase assay demonstrated that the Runx/Cbfß complex binds putative Runx-binding sites of the Ihh promoter regions, and also the Runx/Cbfß complex directly upregulates Ihh expression at the transcriptional level. Consistently, the expressions of Ihh target genes, including CyclinD1, Ptc, and Pthlh, were downregulated in Cbfß-deficient chondrocytes. Taken together, our study reveals not only that Cbfß is essential for chondrocyte proliferation and differentiation for the growth and maintenance of the skeleton in postnatal mice, but also that it functions in upregulating Ihh expression to promoter chondrocyte proliferation and osteoblast differentiation, and inhibiting PPR expression to enhance chondrocyte differentiation.


Assuntos
Cartilagem/crescimento & desenvolvimento , Diferenciação Celular , Condrócitos/citologia , Subunidade beta de Fator de Ligação ao Core/metabolismo , Proteínas Hedgehog/genética , Osteogênese , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Regulação para Cima/genética , Animais , Animais Recém-Nascidos , Osso e Ossos/anormalidades , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Cartilagem/metabolismo , Proliferação de Células , Condrócitos/metabolismo , Subunidade beta de Fator de Ligação ao Core/deficiência , Ciclina D1/metabolismo , Nanismo/patologia , Extremidades/patologia , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/anormalidades , Lâmina de Crescimento/patologia , Proteínas Hedgehog/metabolismo , Integrases/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Fenótipo , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais , Crânio/metabolismo , Crânio/patologia , Fatores de Tempo
12.
J Cell Physiol ; 228(12): 2350-64, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23813439

RESUMO

Core binding factor (CBF) is a heterodimeric transcription factor containing one of three DNA-binding proteins of the Runt-related transcription factor family (RUNX1-3) and the non-DNA-binding protein, CBFß. RUNX1 and CBFß are the most common targets of chromosomal rearrangements in leukemia. CBF has been implicated in other cancer types; for example RUNX1 and RUNX2 are implicated in cancers of epithelial origin, including prostate, breast, and ovarian cancers. In these tumors, CBF is involved in maintaining the malignant phenotype and, when highly over-expressed, contributes to metastatic growth in bone. Herein, lentiviral delivery of CBFß-specific shRNAs was used to achieve a 95% reduction of CBFß in an ovarian cancer cell line. This drastic reduction in CBFß expression resulted in growth inhibition that was not associated with a cell cycle block or an increase in apoptosis. However, CBFß silencing resulted in increased autophagy and production of reactive oxygen species (ROS). Since sphingolipid and ceramide metabolism regulates non-apoptotic cell death, autophagy, and ROS production, fumonsin B1 (FB1), an inhibitor of ceramide synthase, was used to alter ceramide production in the CBFß-silenced cells. FB1 treatment inhibited the CBFß-dependent increase in autophagy and provided a modest increase in cell survival. To document alterations to sphingolipids in the CBFß-silenced cells, ceramide, and lactosylceramide levels were directly examined by mass spectrometry. Substantial increases in ceramide species and decreases in lactosylceramides were identified. Altogether, this report provides evidence that CBF transcriptional pathways control cellular survival, at least in part, through sphingolipid metabolism.


Assuntos
Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Proteínas de Ligação a DNA/genética , Esfingolipídeos/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptose/genética , Autofagia/genética , Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Lactosilceramidas/genética , Lactosilceramidas/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Esfingolipídeos/genética
13.
Nature ; 481(7381): 376-9, 2011 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-22190036

RESUMO

The human APOBEC3 cytidine deaminases are potent inhibitors of diverse retroviruses, including human immunodeficiency virus-1 (HIV-1). HIV-1 Vif forms an E3 ubiquitin ligase complex with cullin 5 (CUL5), elongin B and elongin C , which promotes the polyubiquitination and degradation of APOBEC3 substrates. Here we demonstrate in human T cells that core binding factor ß (CBF-ß) is a key regulator of the evasion of HIV-1 from the host defence mediated by APOBEC3. CBF-ß, the non-DNA-binding subunit of a heterodimeric transcription factor, regulates the folding and DNA-binding activity of partner RUNX family proteins, which have important roles in the development and differentiation of diverse cell types, including T lymphocytes. In our study, knockdown of endogenous CBF-ß blocked Vif-induced APOBEC3G polyubiquitination and degradation. CBF-ß was not required for the interaction between Vif and APOBEC3G, yet was essential for the assembly of the Vif-CUL5 E3-ubiquitin-ligase complex. CBF-ß proved to be a unique regulator of primate lentiviral Vif and not a general component of the CUL5 E3 ubiquitin ligase. We show that Vif and CBF-ß physically interact, and that the amino-terminal region of Vif is required for this interaction. Furthermore, interactions with Vif required regions in CBF-ß that are not involved in RUNX protein binding. Considering the importance of the interaction between Vif and CBF-ß, disrupting this interaction represents an attractive pharmacological intervention against HIV-1.


Assuntos
Diferenciação Celular , Subunidade beta de Fator de Ligação ao Core/metabolismo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Linfócitos T/citologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Desaminase APOBEC-3G , Linhagem Celular , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/química , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Proteínas Culina/metabolismo , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , HIV-1/genética , HIV-1/imunologia , Humanos , Imunoprecipitação , Modelos Moleculares , Ligação Proteica , Proteólise , Linfócitos T/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética
14.
J Immunol ; 186(3): 1450-7, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21178013

RESUMO

Hematopoietic lymphoid tissue inducer (LTi) cells are essential for the development of secondary lymphoid tissues including lymph nodes and Peyer's patches. Two transcription factors, the helix-loop-helix inhibitor Id2 and the retinoic acid-related orphan receptor γt (Rorγt), have been shown to be crucial for LTi cell development. However, it remains unclear how the specification of multipotent hematopoietic progenitor cells toward the LTi lineage is programmed. In this study, we report impaired lymphoid tissue organogenesis in mice in which the function of Runx1/Cbfß transcription factor complexes was attenuated by the loss of either the distal promoter-derived Runx1 or Cbfß2 variant protein. We found that LTi progenitors in fetal liver, defined previously as a lineage marker-negative α4ß7 integrin (α4ß7)(+) IL-7R α-chain (IL-7Rα)(+) population, can be subdivided into Rorγt-expressing IL-7Rα(high) cells and nonexpressing IL-7Rα(mid) cells. Whereas Id2 and Rorγt are required to direct α4ß7(+)IL-7Rα(mid) cells to become α4ß7(+)IL-7Rα(high) cells, Runx1/Cbfß2 complexes are necessary for the emergence of α4ß7(+)IL-7Rα(mid) cells. In addition, the loss of Cbfß2, but not P1-Runx1, resulted in an inefficient upregulation of Rorγt in residual α4ß7(+)IL-7Rα(+) LTi cells at anlagen. Our results thus revealed that Runx1/Cbfß2 complexes regulate the differentiation of LTi cells at two stages: an early specification of hematopoietic progenitors toward the LTi lineage and a subsequent activation of Rorγt expression at anlagen.


Assuntos
Diferenciação Celular/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Tecido Linfoide/imunologia , Animais , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Variação Genética/imunologia , Proteína 2 Inibidora de Diferenciação/biossíntese , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/fisiologia , Fígado/embriologia , Fígado/imunologia , Fígado/patologia , Tecido Linfoide/citologia , Tecido Linfoide/embriologia , Camundongos , Camundongos Mutantes , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/imunologia , Células-Tronco Multipotentes/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/fisiologia
15.
Blood ; 112(3): 480-92, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18390836

RESUMO

CBFbeta is the non-DNA binding subunit of the core binding factors (CBFs). Mice with reduced CBFbeta levels display profound, early defects in T-cell but not B-cell development. Here we show that CBFbeta is also required at very early stages of natural killer (NK)-cell development. We also demonstrate that T-cell development aborts during specification, as the expression of Gata3 and Tcf7, which encode key regulators of T lineage specification, is substantially reduced, as are functional thymic progenitors. Constitutively active Notch or IL-7 signaling cannot restore T-cell expansion or differentiation of CBFbeta insufficient cells, nor can overexpression of Runx1 or CBFbeta overcome a lack of Notch signaling. Therefore, the ability of the prethymic cell to respond appropriately to Notch is dependent on CBFbeta, and both signals converge to activate the T-cell developmental program.


Assuntos
Subunidade beta de Fator de Ligação ao Core/fisiologia , Células Matadoras Naturais/citologia , Linfopoese , Receptores Notch/fisiologia , Linfócitos T/citologia , Animais , Linhagem da Célula , Subunidade beta de Fator de Ligação ao Core/deficiência , Fator de Transcrição GATA3/deficiência , Fator 1-alfa Nuclear de Hepatócito , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Fator 1 de Transcrição de Linfócitos T/deficiência
16.
Blood ; 109(1): 11-21, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16940420

RESUMO

The family of core-binding factors includes the DNA-binding subunits Runx1-3 and their common non-DNA-binding partner CBFbeta. We examined the collective role of core-binding factors in hematopoiesis with a hypomorphic Cbfb allelic series. Reducing CBFbeta levels by 3- or 6-fold caused abnormalities in bone development, megakaryocytes, granulocytes, and T cells. T-cell development was very sensitive to an incremental reduction of CBFbeta levels: mature thymocytes were decreased in number upon a 3-fold reduction in CBFbeta levels, and were virtually absent when CBFbeta levels were 6-fold lower. Partially penetrant consecutive differentiation blocks were found among early T-lineage progenitors within the CD4- CD8- double-negative 1 and downstream double-negative 2 thymocyte subsets. Our data define a critical CBFbeta threshold for normal T-cell development, and situate an essential role for core-binding factors during the earliest stages of T-cell development.


Assuntos
Desenvolvimento Ósseo/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Granulócitos/patologia , Hematopoese/fisiologia , Megacariócitos/patologia , Subpopulações de Linfócitos T/patologia , Alelos , Animais , Linhagem da Célula , Ensaio de Unidades Formadoras de Colônias , Subunidades alfa de Fatores de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Fígado/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Quimera por Radiação , Baço/embriologia , Timo/embriologia
17.
Blood ; 109(8): 3432-40, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17185462

RESUMO

Recent studies suggest that the chromosome 16 inversion, associated with acute myeloid leukemia M4Eo, takes place in hematopoietic stem cells. If this is the case, it is of interest to know the effects of the resulting fusion gene, CBFB-MYH11, on other lineages. Here we studied T-cell development in mice expressing Cbfb-MYH11 and compared them with mice compound-heterozygous for a Cbfb null and a hypomorphic GFP knock-in allele (Cbfb(-/GFP)), which had severe Cbfb deficiency. We found a differentiation block at the DN1 stage of thymocyte development in Cbfb-MYH11 knock-in chimeras. In a conditional knock-in model in which Cbfb-MYH11 expression was activated by Lck-Cre, there was a 10-fold reduction in thymocyte numbers in adult thymus, resulting mainly from impaired survival of CD4+CD8+ thymocytes. Although Cbfb-MYH11 derepressed CD4 expression efficiently in reporter assays, such derepression was less pronounced in vivo. On the other hand, CD4 expression was derepressed and thymocyte development was blocked at DN1 and DN2 stages in E17.5 Cbfb(-/GFP) thymus, with a 20-fold reduction of total thymocyte numbers. Our data suggest that Cbfb-MYH11 suppressed Cbfb in several stages of T-cell development and provide a mechanism for CBFB-MYH11 association with myeloid but not lymphoid leukemia.


Assuntos
Inversão Cromossômica , Subunidade beta de Fator de Ligação ao Core/biossíntese , Leucemia Mieloide/metabolismo , Cadeias Pesadas de Miosina/biossíntese , Proteínas de Fusão Oncogênica/biossíntese , Linfócitos T/metabolismo , Timo/metabolismo , Animais , Antígenos CD4/biossíntese , Antígenos CD8/imunologia , Morte Celular/genética , Sobrevivência Celular/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Leucemia Linfoide/genética , Leucemia Linfoide/metabolismo , Leucemia Linfoide/patologia , Leucemia Mieloide/genética , Leucemia Mieloide/patologia , Camundongos , Camundongos Knockout , Cadeias Pesadas de Miosina/genética , Proteínas de Fusão Oncogênica/genética , Linfócitos T/patologia , Timo/patologia
18.
Am J Med Genet A ; 140(21): 2349-54, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17022082

RESUMO

The core binding factor beta gene (CBFB), essential to bone morphogenesis, is located at 16q22.1. Homozygous deficiency of CBFB leads to ossification defects in mice. CBFB forms a heterodimer with RUNX2 (CBFA1) during embryonic bone development. RUNX2 mutations lead to cleidocranial dysplasia in humans. We describe an infant boy with an interstitial deletion of 16q21q22, delayed skull ossification, cleft palate, and heart anomalies who had a difficult course in infancy but eventually improved and is healthy. He was found to have CBFB haploinsufficiency, but did not have mutations in RUNX2. We suggest that 16q21q22 deletion be considered when there are antenatal or postnatal findings of enlarged cranial sutures with or without cleft palate. The finding of CBFB haploinsufficiency in our case and the similarity of cranial ossification defects with a mouse model of CBFB deletion suggest a role for CBFB in cranial bone development in humans.


Assuntos
Anormalidades Múltiplas/genética , Deleção Cromossômica , Cromossomos Humanos Par 16/genética , Fissura Palatina/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Cardiopatias Congênitas/genética , Crânio/anormalidades , Animais , Transtornos de Alimentação na Infância/genética , Feminino , Humanos , Hibridização in Situ Fluorescente , Lactente , Masculino , Camundongos , Ossificação Heterotópica , Gravidez , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...