Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Blood ; 138(15): 1331-1344, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33971010

RESUMO

The mechanism underlying cell type-specific gene induction conferred by ubiquitous transcription factors as well as disruptions caused by their chimeric derivatives in leukemia is not well understood. Here, we investigate whether RNAs coordinate with transcription factors to drive myeloid gene transcription. In an integrated genome-wide approach surveying for gene loci exhibiting concurrent RNA and DNA interactions with the broadly expressed Runt-related transcription factor 1 (RUNX1), we identified the long noncoding RNA (lncRNA) originating from the upstream regulatory element of PU.1 (LOUP). This myeloid-specific and polyadenylated lncRNA induces myeloid differentiation and inhibits cell growth, acting as a transcriptional inducer of the myeloid master regulator PU.1. Mechanistically, LOUP recruits RUNX1 to both the PU.1 enhancer and the promoter, leading to the formation of an active chromatin loop. In t(8;21) acute myeloid leukemia (AML), wherein RUNX1 is fused to ETO, the resulting oncogenic fusion protein, RUNX1-ETO, limits chromatin accessibility at the LOUP locus, causing inhibition of LOUP and PU.1 expression. These findings highlight the important role of the interplay between cell-type-specific RNAs and transcription factors, as well as their oncogenic derivatives in modulating lineage-gene activation and raise the possibility that RNA regulators of transcription factors represent alternative targets for therapeutic development.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Leucemia Mieloide Aguda/genética , Proteínas de Fusão Oncogênica/genética , RNA Longo não Codificante/genética , Proteína 1 Parceira de Translocação de RUNX1/genética , Linhagem Celular Tumoral , Regulação Leucêmica da Expressão Gênica , Humanos , Ativação Transcricional
2.
Nature ; 503(7476): 371-6, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24107992

RESUMO

DNA methylation was first described almost a century ago; however, the rules governing its establishment and maintenance remain elusive. Here we present data demonstrating that active transcription regulates levels of genomic methylation. We identify a novel RNA arising from the CEBPA gene locus that is critical in regulating the local DNA methylation profile. This RNA binds to DNMT1 and prevents CEBPA gene locus methylation. Deep sequencing of transcripts associated with DNMT1 combined with genome-scale methylation and expression profiling extend the generality of this finding to numerous gene loci. Collectively, these results delineate the nature of DNMT1-RNA interactions and suggest strategies for gene-selective demethylation of therapeutic targets in human diseases.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , Regulação da Expressão Gênica/genética , RNA não Traduzido/metabolismo , Sequência de Bases , Linhagem Celular , DNA/genética , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , Perfilação da Expressão Gênica , Genoma Humano/genética , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA não Traduzido/genética , Proteínas de Ligação a RNA/metabolismo , Especificidade por Substrato , Transcrição Gênica/genética
3.
Blood ; 124(15): 2391-9, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25185713

RESUMO

Runx transcription factors contribute to hematopoiesis and are frequently implicated in hematologic malignancies. All three Runx isoforms are expressed at the earliest stages of hematopoiesis; however, their function in hematopoietic stem cells (HSCs) is not fully elucidated. Here, we show that Runx factors are essential in HSCs by driving the expression of the hematopoietic transcription factor PU.1. Mechanistically, by using a knockin mouse model in which all three Runx binding sites in the -14kb enhancer of PU.1 are disrupted, we observed failure to form chromosomal interactions between the PU.1 enhancer and its proximal promoter. Consequently, decreased PU.1 levels resulted in diminished long-term HSC function through HSC exhaustion, which could be rescued by reintroducing a PU.1 transgene. Similarly, in a mouse model of AML/ETO9a leukemia, disrupting the Runx binding sites resulted in decreased PU.1 levels. Leukemia onset was delayed, and limiting dilution transplantation experiments demonstrated functional loss of leukemia-initiating cells. This is surprising, because low PU.1 levels have been considered a hallmark of AML/ETO leukemia, as indicated in mouse models and as shown here in samples from leukemic patients. Our data demonstrate that Runx-dependent PU.1 chromatin interaction and transcription of PU.1 are essential for both normal and leukemia stem cells.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Animais , Pareamento de Bases/genética , Sítios de Ligação , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos Endogâmicos C57BL , Mutação/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Transdução de Sinais/genética , Transcrição Gênica
4.
EMBO J ; 30(19): 4059-70, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21873977

RESUMO

The transcription factor RUNX1 is essential to establish the haematopoietic gene expression programme; however, the mechanism of how it activates transcription of haematopoietic stem cell (HSC) genes is still elusive. Here, we obtained novel insights into RUNX1 function by studying regulation of the human CD34 gene, which is expressed in HSCs. Using transgenic mice carrying human CD34 PAC constructs, we identified a novel downstream regulatory element (DRE), which is bound by RUNX1 and is necessary for human CD34 expression in long-term (LT)-HSCs. Conditional deletion of Runx1 in mice harbouring human CD34 promoter-DRE constructs abrogates human CD34 expression. We demonstrate by chromosome conformation capture assays in LT-HSCs that the DRE physically interacts with the human CD34 promoter. Targeted mutagenesis of RUNX binding sites leads to perturbation of this interaction and decreased human CD34 expression in LT-HSCs. Overall, our in vivo data provide novel evidence about the role of RUNX1 in mediating interactions between distal and proximal elements of the HSC gene CD34.


Assuntos
Antígenos CD34/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Animais , Transplante de Medula Óssea , Cromatina/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Sangue Fetal/citologia , Genótipo , Células HL-60 , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Sequências Reguladoras de Ácido Nucleico/genética
5.
Blood ; 121(8): 1255-64, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23212524

RESUMO

Lineage-restricted cells can be reprogrammed to a pluripotent state known as induced pluripotent stem (iPS) cells through overexpression of 4 transcription factors. iPS cells are similar to human embryonic stem (hES) cells and have the same ability to generate all the cells of the human body, including blood cells. However, this process is extremely inefficient and to date has been unsuccessful at differentiating iPS into hematopoietic stem cells (HSCs). We hypothesized that iPS cells, injected into NOD.Cg-Prkdc(scid) Il2rg(tm1Wjl)/SzJ immunocompromised (NSG) mice could give rise to hematopoietic stem/progenitor cells (HSPCs) during teratoma formation. Here, we report a novel in vivo system in which human iPS cells differentiate within teratomas to derive functional myeloid and lymphoid cells. Similarly, HSPCs can be isolated from teratoma parenchyma and reconstitute a human immune system when transplanted into immunodeficient mice. Our data provide evidence that in vivo generation of patient customized cells is feasible, providing materials that could be useful for transplantation, human antibody generation, and drug screening applications.


Assuntos
Hematopoese/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Teratoma/patologia , Animais , Linfócitos B/citologia , Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Queratinócitos/fisiologia , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células Mieloides/citologia , Transplante de Neoplasias , Células Estromais/citologia , Células Estromais/fisiologia , Células Estromais/transplante , Linfócitos T/citologia , Teratoma/genética , Transplante Heterólogo , Células Tumorais Cultivadas
6.
Nat Genet ; 38(11): 1269-77, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17041602

RESUMO

Knockdown of the transcription factor PU.1 (encoded by Sfpi1) leads to acute myeloid leukemia (AML) in mice. We examined the transcriptome of preleukemic hematopoietic stem cells (HSCs) in which PU.1 was knocked down (referred to as 'PU.1-knockdown HSCs') to identify transcriptional changes preceding malignant transformation. Transcription factors c-Jun and JunB were among the top-downregulated targets. Restoration of c-Jun expression in preleukemic cells rescued the PU.1 knockdown-initiated myelomonocytic differentiation block. Lentiviral restoration of JunB at the leukemic stage led to loss of leukemic self-renewal capacity and prevented leukemia in NOD-SCID mice into which leukemic PU.1-knockdown cells were transplanted. Examination of human individuals with AML confirmed the correlation between PU.1 and JunB downregulation. These results delineate a transcriptional pattern that precedes leukemic transformation in PU.1-knockdown HSCs and demonstrate that decreased levels of c-Jun and JunB contribute to the development of PU.1 knockdown-induced AML by blocking differentiation and increasing self-renewal. Therefore, examination of disturbed gene expression in HSCs can identify genes whose dysregulation is essential for leukemic stem cell function and that are targets for therapeutic interventions.


Assuntos
Células-Tronco Hematopoéticas/patologia , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogênicas c-jun/fisiologia , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Animais , Diferenciação Celular/genética , Transformação Celular Neoplásica/genética , Regulação para Baixo , Granulócitos/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Monócitos/citologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Transdução Genética
7.
Development ; 137(13): 2147-56, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20530543

RESUMO

Coactivator-associated arginine methyltransferase I (CARM1; PRMT4) regulates gene expression by multiple mechanisms including methylation of histones and coactivation of steroid receptor transcription. Mice lacking CARM1 are small, fail to breathe and die shortly after birth, demonstrating the crucial role of CARM1 in development. In adults, CARM1 is overexpressed in human grade-III breast tumors and prostate adenocarcinomas, and knockdown of CARM1 inhibits proliferation of breast and prostate cancer cell lines. Based on these observations, we hypothesized that loss of CARM1 in mouse embryos would inhibit pulmonary cell proliferation, resulting in respiratory distress. By contrast, we report here that loss of CARM1 results in hyperproliferation of pulmonary epithelial cells during embryonic development. The lungs of newborn mice lacking CARM1 have substantially reduced airspace compared with their wild-type littermates. In the absence of CARM1, alveolar type II cells show increased proliferation. Electron microscopic analyses demonstrate that lungs from mice lacking CARM1 have immature alveolar type II cells and an absence of alveolar type I cells. Gene expression analysis reveals a dysregulation of cell cycle genes and markers of differentiation in the Carm1 knockout lung. Furthermore, there is an overlap in gene expression in the Carm1 knockout and the glucocorticoid receptor knockout lung, suggesting that hyperproliferation and lack of maturation of the alveolar cells are at least in part caused by attenuation of glucocorticoid-mediated signaling. These results demonstrate for the first time that CARM1 inhibits pulmonary cell proliferation and is required for proper differentiation of alveolar cells.


Assuntos
Células Epiteliais/metabolismo , Pulmão/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Proliferação de Células , Células Endoteliais/metabolismo , Glucocorticoides/metabolismo , Camundongos , Alvéolos Pulmonares/metabolismo , Transcrição Gênica
8.
Nat Commun ; 14(1): 99, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36609400

RESUMO

DNA methylation is a fundamental epigenetic modification regulating gene expression. Aberrant DNA methylation is the most common molecular lesion in cancer cells. However, medical intervention has been limited to the use of broadly acting, small molecule-based demethylating drugs with significant side-effects and toxicities. To allow for targeted DNA demethylation, we integrated two nucleic acid-based approaches: DNMT1 interacting RNA (DiR) and RNA aptamer strategy. By combining the RNA inherent capabilities of inhibiting DNMT1 with an aptamer platform, we generated a first-in-class DNMT1-targeted approach - aptaDiR. Molecular modelling of RNA-DNMT1 complexes coupled with biochemical and cellular assays enabled the identification and characterization of aptaDiR. This RNA bio-drug is able to block DNA methylation, impair cancer cell viability and inhibit tumour growth in vivo. Collectively, we present an innovative RNA-based approach to modulate DNMT1 activity in cancer or diseases characterized by aberrant DNA methylation and suggest the first alternative strategy to overcome the limitations of currently approved non-specific hypomethylating protocols, which will greatly improve clinical intervention on DNA methylation.


Assuntos
Metilação de DNA , RNA , RNA/genética , RNA/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Regulação Neoplásica da Expressão Gênica , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Epigênese Genética
9.
Blood ; 114(27): 5415-25, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19797526

RESUMO

Acute promyelocytic leukemia (APL) is characterized by a block in differentiation and accumulation of promyelocytes in the bone marrow and blood. The majority of APL patients harbor the t(15:17) translocation leading to expression of the fusion protein promyelocytic-retinoic acid receptor alpha. Treatment with retinoic acid leads to degradation of promyelocytic-retinoic acid receptor alpha protein and disappearance of leukemic cells; however, 30% of APL patients relapse after treatment. One potential mechanism for relapse is the persistence of cancer "stem" cells in hematopoietic organs after treatment. Using a novel sorting strategy we developed to isolate murine myeloid cells at distinct stages of differentiation, we identified a population of committed myeloid cells (CD34(+), c-kit(+), FcgammaRIII/II(+), Gr1(int)) that accumulates in the spleen and bone marrow in a murine model of APL. We observed that these cells are capable of efficiently generating leukemia in recipient mice, demonstrating that this population represents the APL cancer-initiating cell. These cells down-regulate the transcription factor CCAAT/enhancer binding protein alpha (C/EBPalpha) possibly through a methylation-dependent mechanism, indicating that C/EBPalpha deregulation contributes to transformation of APL cancer-initiating cells. Our findings provide further understanding of the biology of APL by demonstrating that a committed transformed progenitor can initiate and propagate the disease.


Assuntos
Células da Medula Óssea/patologia , Leucemia Promielocítica Aguda/patologia , Células Mieloides/patologia , Células-Tronco Neoplásicas/patologia , Animais , Antígenos CD34/metabolismo , Células da Medula Óssea/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Calgranulina A/genética , Calgranulina A/metabolismo , Linhagem Celular Tumoral , Metilação de DNA , Feminino , Citometria de Fluxo , Humanos , Estimativa de Kaplan-Meier , Leucemia Promielocítica Aguda/sangue , Leucemia Promielocítica Aguda/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células U937
10.
Cells ; 10(11)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34831209

RESUMO

Nutritional intake impacts the human epigenome by directing epigenetic pathways in normal cell development via as yet unknown molecular mechanisms. Consequently, imbalance in the nutritional intake is able to dysregulate the epigenetic profile and drive cells towards malignant transformation. Here we present a novel epigenetic effect of the essential nutrient, NAD. We demonstrate that impairment of DNMT1 enzymatic activity by NAD-promoted ADP-ribosylation leads to demethylation and transcriptional activation of the CEBPA gene, suggesting the existence of an unknown NAD-controlled region within the locus. In addition to the molecular events, NAD- treated cells exhibit significant morphological and phenotypical changes that correspond to myeloid differentiation. Collectively, these results delineate a novel role for NAD in cell differentiation, and indicate novel nutri-epigenetic strategies to regulate and control gene expression in human cells.


Assuntos
Diferenciação Celular , Metilação de DNA/genética , NAD/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Desmetilação do DNA/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Neoplasias/genética , Neoplasias/patologia , Fosforilação Oxidativa/efeitos dos fármacos , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos
11.
J Clin Invest ; 117(9): 2611-20, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17694175

RESUMO

Targeted disruption of a highly conserved distal enhancer reduces expression of the PU.1 transcription factor by 80% and leads to acute myeloid leukemia (AML) with frequent cytogenetic aberrations in mice. Here we identify a SNP within this element in humans that is more frequent in AML with a complex karyotype, leads to decreased enhancer activity, and reduces PU.1 expression in myeloid progenitors in a development-dependent manner. This SNP inhibits binding of the chromatin-remodeling transcriptional regulator special AT-rich sequence binding protein 1 (SATB1). Overexpression of SATB1 increased PU.1 expression, and siRNA inhibition of SATB1 downregulated PU.1 expression. Targeted disruption of the distal enhancer led to a loss of regulation of PU.1 by SATB1. Interestingly, disruption of SATB1 in mice led to a selective decrease of PU.1 RNA in specific progenitor types (granulocyte-macrophage and megakaryocyte-erythrocyte progenitors) and a similar effect was observed in AML samples harboring this SNP. Thus we have identified a SNP within a distal enhancer that is associated with a subtype of leukemia and exerts a deleterious effect through remote transcriptional dysregulation in specific progenitor subtypes.


Assuntos
Regulação Neoplásica da Expressão Gênica , Leucemia Mieloide Aguda/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Animais , Sequência de Bases , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Genoma Humano/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Retorno de Linfócitos/genética , Receptores de Retorno de Linfócitos/metabolismo , Células-Tronco/metabolismo , Transativadores/deficiência , Transativadores/metabolismo
12.
Mol Cell Biol ; 27(21): 7425-38, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17785440

RESUMO

The Ets family transcription factor PU.1 is crucial for the regulation of hematopoietic development. Pu.1 is activated in hematopoietic stem cells and is expressed in mast cells, B cells, granulocytes, and macrophages but is switched off in T cells. Many of the transcription factors regulating Pu.1 have been identified, but little is known about how they organize Pu.1 chromatin in development. We analyzed the Pu.1 promoter and the upstream regulatory element (URE) using in vivo footprinting and chromatin immunoprecipitation assays. In B cells, Pu.1 was bound by a set of transcription factors different from that in myeloid cells and adopted alternative chromatin architectures. In T cells, Pu.1 chromatin at the URE was open and the same transcription factor binding sites were occupied as in B cells. The transcription factor RUNX1 was bound to the URE in precursor cells, but binding was down-regulated in maturing cells. In PU.1 knockout precursor cells, the Ets factor Fli-1 compensated for the lack of PU.1, and both proteins could occupy a subset of Pu.1 cis elements in PU.1-expressing cells. In addition, we identified novel URE-derived noncoding transcripts subject to tissue-specific regulation. Our results provide important insights into how overlapping, but different, sets of transcription factors program tissue-specific chromatin structures in the hematopoietic system.


Assuntos
Cromatina/química , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Proteínas Proto-Oncogênicas/genética , RNA não Traduzido/genética , Transativadores/genética , Transcrição Gênica , Animais , Linfócitos B/enzimologia , Linfócitos B/metabolismo , Sequência de Bases , Diferenciação Celular , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Macrófagos/enzimologia , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Células Mieloides/citologia , Células Mieloides/metabolismo , Conformação de Ácido Nucleico , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Polimerase II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/enzimologia , Linfócitos T/metabolismo , Fatores de Transcrição/metabolismo
13.
Cell Rep ; 32(12): 108181, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32966797

RESUMO

Hemopexin (Hx) is a scavenger of labile heme. Herein, we present data defining the role of tumor stroma-expressed Hx in suppressing cancer progression. Labile heme and Hx levels are inversely correlated in the plasma of patients with prostate cancer (PCa). Further, low expression of Hx in PCa biopsies characterizes poorly differentiated tumors and correlates with earlier time to relapse. Significantly, heme promotes tumor growth and metastases in an orthotopic murine model of PCa, with the most aggressive phenotype detected in mice lacking Hx. Mechanistically, labile heme accumulates in the nucleus and modulates specific gene expression via interacting with guanine quadruplex (G4) DNA structures to promote PCa growth. We identify c-MYC as a heme:G4-regulated gene and a major player in heme-driven cancer progression. Collectively, these results reveal that sequestration of labile heme by Hx may block heme-driven tumor growth and metastases, suggesting a potential strategy to prevent and/or arrest cancer dissemination.


Assuntos
Heme/metabolismo , Hemopexina/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ciclo Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , DNA/genética , Progressão da Doença , Quadruplex G , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Metástase Neoplásica , Fenótipo , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/sangue , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Resultado do Tratamento , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
14.
EMBO J ; 5(9): 2341-2347, 1986 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16453705

RESUMO

We describe here the nucleotide sequences of several genomic and mRNA copies of the suffix, a short dispersed actively transcribed repeat located at the 3' ends of many different genes of Drosophila melanogaster. Only one strand of the suffix is transcribed. The patterns of suffix-containing mRNAs vary during development. The five randomly selected genomic copies of the suffix are 265 bp long and quite conservative in their sequence. The non-transcribed strand is terminated with oligo(A) preceded by AATAAA sequence. No repetitive flanking sequences can be detected. The three other genomic copies selected by hybridization with suffix-containing cDNA clones are less conservative, especially in the 5' part. In particular, they contain short insertions carrying a polyadenylation signal AATAAA at exactly the same position of the suffix. Comparison of genomic and cDNA clones shows that mRNAs are polyadenylated at the last nucleotide of these insertions. The cDNA clones include the same part of the suffix, from the 39th to 112th nucleotide. Thus, a segment of the suffix forms the last exon for both genes. In one case, the beginning of the last intron coincides with the beginning of suffix, creating a very unusual donor splicing site. We conclude that the suffix sequence is directly involved in the formation of the last splicing site and 3'-end maturation of mRNA, at least in the case of the two genes analysed.

15.
Nat Commun ; 7: 10968, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27005833

RESUMO

CCAAT/enhancer-binding protein alpha (C/EBPα) is an essential transcription factor for myeloid lineage commitment. Here we demonstrate that acetylation of C/EBPα at lysine residues K298 and K302, mediated at least in part by general control non-derepressible 5 (GCN5), impairs C/EBPα DNA-binding ability and modulates C/EBPα transcriptional activity. Acetylated C/EBPα is enriched in human myeloid leukaemia cell lines and acute myeloid leukaemia (AML) samples, and downregulated upon granulocyte-colony stimulating factor (G-CSF)- mediated granulocytic differentiation of 32Dcl3 cells. C/EBPα mutants that mimic acetylation failed to induce granulocytic differentiation in C/EBPα-dependent assays, in both cell lines and in primary hematopoietic cells. Our data uncover GCN5 as a negative regulator of C/EBPα and demonstrate the importance of C/EBPα acetylation in myeloid differentiation.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Regulação Neoplásica da Expressão Gênica , Granulócitos/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide/genética , Mielopoese/genética , Fatores de Transcrição de p300-CBP/genética , Acetilação , Diferenciação Celular/genética , Linhagem Celular Tumoral , Cromatografia Líquida , Ensaio de Desvio de Mobilidade Eletroforética , Fator Estimulador de Colônias de Granulócitos , Granulócitos/citologia , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Leucemia Mieloide/metabolismo , Leucemia Mieloide Aguda/metabolismo , Espectrometria de Massas , Fatores de Transcrição de p300-CBP/metabolismo
16.
Nat Commun ; 6: 7091, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25997600

RESUMO

Chronic myeloid leukaemia (CML) is a myeloproliferative disorder characterized by the genetic translocation t(9;22)(q34;q11.2) encoding for the BCR-ABL fusion oncogene. However, many molecular mechanisms of the disease progression still remain poorly understood. A growing body of evidence suggests that the epigenetic abnormalities are involved in tyrosine kinase resistance in CML, leading to leukaemic clone escape and disease propagation. Here we show that, by applying cellular reprogramming to primary CML cells, aberrant DNA methylation contributes to the disease evolution. Importantly, using a BCR-ABL inducible murine model, we demonstrate that a single oncogenic lesion triggers DNA methylation changes, which in turn act as a precipitating event in leukaemia progression.


Assuntos
Metilação de DNA , Genes abl , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Animais , Azacitidina , Diferenciação Celular , Técnicas de Reprogramação Celular , Humanos , Células K562 , Camundongos Transgênicos , Células U937
17.
Cancer Cell ; 24(5): 575-88, 2013 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-24183681

RESUMO

Mutation or epigenetic silencing of the transcription factor C/EBPα is observed in ∼10% of patients with acute myeloid leukemia (AML). In both cases, a common global gene expression profile is observed, but downstream targets relevant for leukemogenesis are not known. Here, we identify Sox4 as a direct target of C/EBPα whereby its expression is inversely correlated with C/EBPα activity. Downregulation of Sox4 abrogated increased self-renewal of leukemic cells and restored their differentiation. Gene expression profiles of leukemia-initiating cells (LICs) from both Sox4 overexpression and murine C/EBPα mutant AML models clustered together but differed from other types of AML. Our data demonstrate that Sox4 overexpression resulting from C/EBPα inactivation contributes to the development of leukemia with a distinct LIC phenotype.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , Fatores de Transcrição SOXC/genética , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Silenciamento de Genes , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Knockout , Mutação , Células Mieloides/metabolismo , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Oncogenes , Fatores de Transcrição SOXC/metabolismo , Transcriptoma
18.
J Clin Invest ; 122(12): 4490-504, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23160200

RESUMO

C/EBPs are a family of transcription factors that regulate growth control and differentiation of various tissues. We found that C/EBPγ is highly upregulated in a subset of acute myeloid leukemia (AML) samples characterized by C/EBPα hypermethylation/silencing. Similarly, C/EBPγ was upregulated in murine hematopoietic stem/progenitor cells lacking C/EBPα, as C/EBPα mediates C/EBPγ suppression. Studies in myeloid cells demonstrated that CEBPG overexpression blocked neutrophilic differentiation. Further, downregulation of Cebpg in murine Cebpa-deficient stem/progenitor cells or in human CEBPA-silenced AML samples restored granulocytic differentiation. In addition, treatment of these leukemias with demethylating agents restored the C/EBPα-C/EBPγ balance and upregulated the expression of myeloid differentiation markers. Our results indicate that C/EBPγ mediates the myeloid differentiation arrest induced by C/EBPα deficiency and that targeting the C/EBPα-C/EBPγ axis rescues neutrophilic differentiation in this unique subset of AMLs.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Diferenciação Celular , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/metabolismo , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Metilação de DNA , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Epigênese Genética , Genes Reporter , Fator Estimulador de Colônias de Granulócitos/fisiologia , Granulócitos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Neutrófilos/metabolismo , Neutrófilos/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Ligação Proteica , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Regulação para Cima
19.
Genes Dev ; 22(15): 2085-92, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18676813

RESUMO

The transcription factor PU.1 is an important regulator of hematopoiesis; precise expression levels are critical for normal hematopoietic development and suppression of leukemia. We show here that noncoding antisense RNAs are important modulators of proper dosages of PU.1. Antisense and sense RNAs are regulated by shared evolutionarily conserved cis-regulatory elements, and we can show that antisense RNAs inhibit PU.1 expression by modulating mRNA translation. We propose that such antisense RNAs will likely be important in the regulation of many genes and may be the reason for the large number of overlapping complementary transcripts with so far unknown function.


Assuntos
Expressão Gênica , Proteínas Proto-Oncogênicas/genética , RNA Antissenso/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Transativadores/genética , Animais , Linhagem Celular , Eletroporação , Granulócitos/citologia , Granulócitos/metabolismo , Células HL-60 , Humanos , Separação Imunomagnética , Células Jurkat , Macrófagos/metabolismo , Camundongos , Modelos Genéticos , Biossíntese de Proteínas , Interferência de RNA , RNA Antissenso/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Linfócitos T/metabolismo , Transcrição Gênica , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA