Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 42(37): 2764-2775, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37573408

RESUMO

Leukaemia is caused by the clonal evolution of a cell that accumulates mutations/genomic rearrangements, allowing unrestrained cell growth. However, recent identification of leukaemic mutations in the blood cells of healthy individuals revealed that additional events are required to expand the mutated clones for overt leukaemia. Here, we assessed the functional consequences of deleting the Fanconi anaemia A (Fanca) gene, which encodes a DNA damage response protein, in Spi1 transgenic mice that develop preleukaemic syndrome. FANCA loss increases SPI1-associated disease penetrance and leukaemic progression without increasing the global mutation load of leukaemic clones. However, a high frequency of leukaemic FANCA-depleted cells display heterozygous activating mutations in known oncogenes, such as Kit or Nras, also identified but at low frequency in FANCA-WT mice with preleukaemic syndrome, indicating that FANCA counteracts the emergence of oncogene mutated leukaemic cells. A unique transcriptional signature is associated with the leukaemic status of FANCA-depleted cells, leading to activation of MDM4, NOTCH and Wnt/ß-catenin pathways. We show that NOTCH signalling improves the proliferation capacity of FANCA-deficient leukaemic cells. Collectively, our observations indicate that loss of the FANC pathway, known to control genetic instability, fosters the expansion of leukaemic cells carrying oncogenic mutations rather than mutation formation. FANCA loss may contribute to this leukaemogenic progression by reprogramming transcriptomic landscape of the cells.


Assuntos
Proteína do Grupo de Complementação A da Anemia de Fanconi , Leucemia , Animais , Camundongos , Heterozigoto , Leucemia/genética , Mutação , Oncogenes/genética , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética
2.
Bull Cancer ; 110(3): 331-335, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36775700

RESUMO

This article highlights the presentations from the 2021 scientific meeting of the Club Hematopoiesis and Oncogenesis. This annual meeting focuses on hematopoiesis and oncogenic mechanisms. Various topics were presented: expansion of hematopoietic stem cells with in vivo and ex vivo strategies, the role of the hematopoietic stem cell niches in aging and leukemic resistance, the crossroad between hematology and immunology, the importance of the metabolism in normal hematopoiesis and hematopoietic defects, solid tumors and oncogenesis, the noncoding genome, inflammation in monocyte differentiation and leukemia, and importantly, the recent advances in myeloid malignancies, lymphoid leukemia and lymphoma.


Assuntos
Leucemia , Linfoma , Humanos , Hematopoese/genética , Células-Tronco Hematopoéticas , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia
3.
Nucleic Acids Res ; 50(14): 7938-7958, 2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-35871293

RESUMO

Although originally described as transcriptional activator, SPI1/PU.1, a major player in haematopoiesis whose alterations are associated with haematological malignancies, has the ability to repress transcription. Here, we investigated the mechanisms underlying gene repression in the erythroid lineage, in which SPI1 exerts an oncogenic function by blocking differentiation. We show that SPI1 represses genes by binding active enhancers that are located in intergenic or gene body regions. HDAC1 acts as a cooperative mediator of SPI1-induced transcriptional repression by deacetylating SPI1-bound enhancers in a subset of genes, including those involved in erythroid differentiation. Enhancer deacetylation impacts on promoter acetylation, chromatin accessibility and RNA pol II occupancy. In addition to the activities of HDAC1, polycomb repressive complex 2 (PRC2) reinforces gene repression by depositing H3K27me3 at promoter sequences when SPI1 is located at enhancer sequences. Moreover, our study identified a synergistic relationship between PRC2 and HDAC1 complexes in mediating the transcriptional repression activity of SPI1, ultimately inducing synergistic adverse effects on leukaemic cell survival. Our results highlight the importance of the mechanism underlying transcriptional repression in leukemic cells, involving complex functional connections between SPI1 and the epigenetic regulators PRC2 and HDAC1.


Assuntos
Histona Desacetilase 1 , Leucemia Eritroblástica Aguda , Complexo Repressor Polycomb 2 , Proteínas Proto-Oncogênicas , Transativadores , Acetilação , Animais , Cromatina/genética , Histona Desacetilase 1/genética , Leucemia Eritroblástica Aguda/genética , Camundongos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Transativadores/genética
4.
BMC Bioinformatics ; 22(1): 407, 2021 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-34404353

RESUMO

BACKGROUND: Multiple studies rely on ChIP-seq experiments to assess the effect of gene modulation and drug treatments on protein binding and chromatin structure. However, most methods commonly used for the normalization of ChIP-seq binding intensity signals across conditions, e.g., the normalization to the same number of reads, either assume a constant signal-to-noise ratio across conditions or base the estimates of correction factors on genomic regions with intrinsically different signals between conditions. Inaccurate normalization of ChIP-seq signal may, in turn, lead to erroneous biological conclusions. RESULTS: We developed a new R package, CHIPIN, that allows normalizing ChIP-seq signals across different conditions/samples when spike-in information is not available, but gene expression data are at hand. Our normalization technique is based on the assumption that, on average, no differences in ChIP-seq signals should be observed in the regulatory regions of genes whose expression levels are constant across samples/conditions. In addition to normalizing ChIP-seq signals, CHIPIN provides as output a number of graphs and calculates statistics allowing the user to assess the efficiency of the normalization and qualify the specificity of the antibody used. In addition to ChIP-seq, CHIPIN can be used without restriction on open chromatin ATAC-seq or DNase hypersensitivity data. We validated the CHIPIN method on several ChIP-seq data sets and documented its superior performance in comparison to several commonly used normalization techniques. CONCLUSIONS: The CHIPIN method provides a new way for ChIP-seq signal normalization across conditions when spike-in experiments are not available. The method is implemented in a user-friendly R package available on GitHub: https://github.com/BoevaLab/CHIPIN.


Assuntos
Sequenciamento de Cromatina por Imunoprecipitação , Cromatina , Imunoprecipitação da Cromatina , Ligação Proteica , Análise de Sequência de DNA
5.
Cancer Discov ; 9(6): 796-811, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31018969

RESUMO

The ETS-domain transcription factors divide into subfamilies based on protein similarities, DNA-binding sequences, and interaction with cofactors. They are regulated by extracellular clues and contribute to cellular processes, including proliferation and transformation. ETS genes are targeted through genomic rearrangements in oncogenesis. The PU.1/SPI1 gene is inactivated by point mutations in human myeloid malignancies. We identified a recurrent somatic mutation (Q226E) in PU.1/SPI1 in Waldenström macroglobulinemia, a B-cell lymphoproliferative disorder. It affects the DNA-binding affinity of the protein and allows the mutant protein to more frequently bind and activate promoter regions with respect to wild-type protein. Mutant SPI1 binding at promoters activates gene sets typically promoted by other ETS factors, resulting in enhanced proliferation and decreased terminal B-cell differentiation in model cell lines and primary samples. In summary, we describe oncogenic subversion of transcription factor function through subtle alteration of DNA binding leading to cellular proliferation and differentiation arrest. SIGNIFICANCE: The demonstration that a somatic point mutation tips the balance of genome-binding pattern provides a mechanistic paradigm for how missense mutations in transcription factor genes may be oncogenic in human tumors.This article is highlighted in the In This Issue feature, p. 681.


Assuntos
Regulação da Expressão Gênica , Mutação de Sentido Incorreto , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Macroglobulinemia de Waldenstrom/genética , Macroglobulinemia de Waldenstrom/metabolismo , Animais , Azepinas/farmacologia , Linfócitos B/citologia , Linfócitos B/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Proliferação de Células , Humanos , Lenalidomida/farmacologia , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Motivos de Nucleotídeos , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Transativadores/genética , Fatores de Transcrição/metabolismo , Triazóis/farmacologia , Macroglobulinemia de Waldenstrom/diagnóstico
6.
Nucleic Acids Res ; 46(7): 3339-3350, 2018 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-29425303

RESUMO

The transcription factor PLZF (promyelocytic leukemia zinc finger protein) acts as an epigenetic regulator balancing self-renewal and differentiation of hematopoietic cells through binding to various chromatin-modifying factors. First described as a transcriptional repressor, PLZF is also associated with active transcription, although the molecular bases underlying the differences are unknown. Here, we reveal that in a hematopoietic cell line, PLZF is predominantly associated with transcribed genes. Additionally, we identify a new association between PLZF and the histone methyltransferase, EZH2 at the genomic level. We find that co-occupancy of PLZF and EZH2 on chromatin at PLZF target genes is not associated with SUZ12 or trimethylated lysine 27 of histone H3 (H3K27me3) but with the active histone mark H3K4me3 and active transcription. Removal of EZH2 leads to an increase of PLZF binding and increased gene expression. Our results suggest a new role of EZH2 in restricting PLZF positive transcriptional activity independently of its canonical PRC2 activity.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/genética , Complexo Repressor Polycomb 2/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Transcrição Gênica , Sítios de Ligação/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Autorrenovação Celular/genética , Cromatina/genética , Regulação da Expressão Gênica/genética , Células-Tronco Hematopoéticas/metabolismo , Histona Metiltransferases/genética , Histonas/genética , Humanos , Proteínas de Neoplasias , Ligação Proteica/genética , Fatores de Transcrição
7.
Haematologica ; 102(11): 1850-1860, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28912174

RESUMO

Transcriptional deregulation caused by epigenetic or genetic alterations is a major cause of leukemic transformation. The Spi1/PU.1 transcription factor is a key regulator of many steps of hematopoiesis, and limits self-renewal of hematopoietic stem cells. The deregulation of its expression or activity contributes to leukemia, in which Spi1 can be either an oncogene or a tumor suppressor. Herein we explored whether cellular senescence, an anti-tumoral pathway that restrains cell proliferation, is a mechanism by which Spi1 limits hematopoietic cell expansion, and thus prevents the development of leukemia. We show that Spi1 overexpression triggers cellular senescence both in primary fibroblasts and hematopoietic cells. Erythroid and myeloid lineages are both prone to Spi1-induced senescence. In hematopoietic cells, Spi1-induced senescence requires its DNA-binding activity and a functional p38MAPK14 pathway but is independent of a DNA-damage response. In contrast, in fibroblasts, Spi1-induced senescence is triggered by a DNA-damage response. Importantly, using our well-established Spi1 transgenic leukemia mouse model, we demonstrate that Spi1 overexpression also induces senescence in erythroid progenitors of the bone marrow in vivo before the onset of the pre-leukemic phase of erythroleukemia. Remarkably, the senescence response is lost during the progression of the disease and erythroid blasts do not display a higher expression of Dec1 and CDKN1A, two of the induced senescence markers in young animals. These results bring indirect evidence that leukemia develops from cells which have bypassed Spi1-induced senescence. Overall, our results reveal senescence as a Spi1-induced anti-proliferative mechanism that may be a safeguard against the development of acute myeloid leukemia.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Animais , Biomarcadores , Medula Óssea/metabolismo , Medula Óssea/patologia , Linhagem Celular , Proliferação de Células , Senescência Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Expressão Ectópica do Gene , Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Camundongos , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo
8.
Oncotarget ; 8(23): 37104-37114, 2017 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-28415748

RESUMO

Oncogenes trigger replicative stress that can lead to genetic instability, which participates in cancer progression. Thus, determining how cells cope with replicative stress can help our understanding of oncogenesis and lead to the identification of new antitumor treatment targets. We previously showed that constitutive overexpression of the oncogenic transcription factor Spi1/PU.1 leads to pre-leukemic cells that have a shortened S phase duration with an increased replication fork speed and increased mutability in the absence of DNA breaks. Here, we demonstrate that the S phase checkpoint protein CHK1 is maintained in a low phosphorylation state in Spi1/PU.1-overexpressing cells and provide evidence that this is not due to negative control of its primary kinase ATR. Notably, we found that the expression of the CHK1 phosphatase PP1α is increased in Spi1/PU.1-overexpressing cells. By exogenously modulating its activity, we demonstrate that PP1α is required to maintain CHK1 in a dephosphorylated state and, more importantly, that it is responsible for the accelerated replication fork progression in Spi1/PU.1-overexpressing cells. These results identify a novel pathway by which an oncogene influences replication in the absence of DNA damage.


Assuntos
Quinase 1 do Ponto de Checagem/metabolismo , Replicação do DNA , Proteína Fosfatase 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Ciclo Celular/genética , Células Cultivadas , Quinase 1 do Ponto de Checagem/genética , Regulação Leucêmica da Expressão Gênica , Humanos , Células K562 , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Camundongos Transgênicos , Fosforilação , Proteína Fosfatase 1/genética , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Transativadores/genética
9.
PLoS One ; 7(11): e49052, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23145067

RESUMO

Oncogenic mutations leading to persistent kinase activities are implicated in various human malignancies. Thereby, signaling pathway-targeted therapies are powerful customized treatment to eradicate cancer cells. In murine and human leukemia cells harboring mutations in Kit, we previously showed that distinct and independent pathways controlled resistance to apoptosis or cell cycle. A treatment with PI3Kinase inhibitors to reduce cell proliferation combined with inhibitors of Erk1/2 activity to promote apoptosis had synergistic effects allowing eradication of leukemia cell growth. We reported here that Bim(EL), a pro-apoptotic member of the Bcl2 family proteins, is the target of Erk1/2 signaling and that its down-regulation is responsible for the apoptosis resistance of murine and human leukemic cells. Downstream of Kit mutant, the tyrosine phosphatase Shp2 maintains Bim(EL) expression at a low level, through Erk/2 activation and proteosomal Bim(EL) degradation. This process is controlled by Shp2 independently of other signaling pathways activated downstream of oncogenic Kit, demonstrating that Shp2 is a key regulator of Bim expression in the context of an oncogenic signaling. The increase in Bim(EL) expression is associated to an increased apoptosis. Moreover, the depletion of Bim overcomes apoptosis associated with Erk1/2 inactivation in UO126-treated leukemic cells, thereby establishing the contribution of Bim to drug-induced apoptosis. These data provide a molecular rationale for using BH3 mimetics in combination with PI3K inhibitors to treat leukemia, especially in the case of an oncogenic signaling refractory to Tyrosine Kinase inhibitors.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Regulação para Baixo/genética , Leucemia/genética , Sistema de Sinalização das MAP Quinases/genética , Proteínas de Membrana/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral , Humanos , Leucemia/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
10.
Bioinformatics ; 28(19): 2517-9, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22829625

RESUMO

MOTIVATION: ChIP-seq consists of chromatin immunoprecipitation and deep sequencing of the extracted DNA fragments. It is the technique of choice for accurate characterization of the binding sites of transcription factors and other DNA-associated proteins. We present a web service, Nebula, which allows inexperienced users to perform a complete bioinformatics analysis of ChIP-seq data. RESULTS: Nebula was designed for both bioinformaticians and biologists. It is based on the Galaxy open source framework. Galaxy already includes a large number of functionalities for mapping reads and peak calling. We added the following to Galaxy: (i) peak calling with FindPeaks and a module for immunoprecipitation quality control, (ii) de novo motif discovery with ChIPMunk, (iii) calculation of the density and the cumulative distribution of peak locations relative to gene transcription start sites, (iv) annotation of peaks with genomic features and (v) annotation of genes with peak information. Nebula generates the graphs and the enrichment statistics at each step of the process. During Steps 3-5, Nebula optionally repeats the analysis on a control dataset and compares these results with those from the main dataset. Nebula can also incorporate gene expression (or gene modulation) data during these steps. In summary, Nebula is an innovative web service that provides an advanced ChIP-seq analysis pipeline providing ready-to-publish results. AVAILABILITY: Nebula is available at http://nebula.curie.fr/ SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Imunoprecipitação da Cromatina/métodos , Biologia Computacional/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Internet , Sítios de Ligação , Software
11.
Nucleic Acids Res ; 40(18): 8927-41, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22790984

RESUMO

Acute leukemias are characterized by deregulation of transcriptional networks that control the lineage specificity of gene expression. The aberrant overexpression of the Spi-1/PU.1 transcription factor leads to erythroleukemia. To determine how Spi-1 mechanistically influences the transcriptional program, we combined a ChIP-seq analysis with transcriptional profiling in cells from an erythroleukemic mouse model. We show that Spi-1 displays a selective DNA-binding that does not often cause transcriptional modulation. We report that Spi-1 controls transcriptional activation and repression partially through distinct Spi-1 recruitment to chromatin. We revealed several parameters impacting on Spi-1-mediated transcriptional activation. Gene activation is facilitated by Spi-1 occupancy close to transcriptional starting site of genes devoid of CGIs. Moreover, in those regions Spi-1 acts by binding to multiple motifs tightly clustered and with similar orientation. Finally, in contrast to the myeloid and lymphoid B cells in which Spi-1 exerts a physiological activity, in the erythroleukemic cells, lineage-specific cooperating factors do not play a prevalent role in Spi-1-mediated transcriptional activation. Thus, our work describes a new mechanism of gene activation through clustered site occupancy of Spi-1 particularly relevant in regard to the strong expression of Spi-1 in the erythroleukemic cells.


Assuntos
Leucemia Eritroblástica Aguda/genética , Proteínas Proto-Oncogênicas/metabolismo , Elementos Reguladores de Transcrição , Transativadores/metabolismo , Ativação Transcricional , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Ilhas de CpG , DNA/química , DNA/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genoma , Leucemia Eritroblástica Aguda/metabolismo , Camundongos , Camundongos Transgênicos , Motivos de Nucleotídeos , Análise de Sequência de DNA , Sítio de Iniciação de Transcrição
12.
Blood ; 119(18): 4228-41, 2012 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-22438255

RESUMO

Oncogenic mutations leading to persistent kinase activities are associated with malignancies. Therefore, deciphering the signaling networks downstream of these oncogenic stimuli remains a challenge to gather insights into targeted therapy. To elucidate the biochemical networks connecting the Kit mutant to leukemogenesis, in the present study, we performed a global profiling of tyrosine-phosphorylated proteins from mutant Kit-driven murine leukemia proerythroblasts and identified Shp2 and Stat5 as proximal effectors of Kit. Shp2 or Stat5 gene depletion by sh-RNA, combined with pharmacologic inhibition of PI3kinase or Mek/Erk activities, revealed 2 distinct and independent signaling pathways contributing to malignancy. We demonstrate that cell survival is driven by the Kit/Shp2/Ras/Mek/Erk1/2 pathway, whereas the G(1)/S transition during the cell cycle is accelerated by both the Kit/Stat5 and Kit/PI3K/Akt pathways. The combined use of the clinically relevant drugs NVP-BEZ235, which targets the cell cycle, and Obatoclax, which targets survival, demonstrated synergistic effects to inhibit leukemia cell growth. This synergy was confirmed with a human mast leukemia cell line (HMC-1.2) that expresses mutant Kit. The results of the present study using liquid chromatography/tandem mass spectrometry analysis have elucidated signaling networks downstream of an oncogenic kinase, providing a molecular rationale for pathway-targeted therapy to treat cancer cells refractory to tyrosine kinase inhibitors.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Adenilato Quinase/antagonistas & inibidores , Adenilato Quinase/fisiologia , Animais , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Imidazóis/farmacologia , Indóis , Leucemia de Mastócitos/patologia , Camundongos , Camundongos Nus , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Fosfotirosina/análise , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Pirróis/farmacologia , Quinolinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Fator de Transcrição STAT5/antagonistas & inibidores , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/fisiologia , Ensaio Tumoral de Célula-Tronco
13.
Cancer Res ; 70(17): 6757-66, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20660370

RESUMO

The multistage process of cancer formation is driven by the progressive acquisition of somatic mutations. Replication stress creates genomic instability in mammals. Using a well-defined multistep leukemia model driven by Spi-1/PU.1 overexpression in the mouse and Spi-1/PU.1-overexpressing human leukemic cells, we investigated the relationship between DNA replication and cancer progression. Here, using DNA molecular combing and flow cytometry methods, we show that Spi-1 increases the speed of replication by acting specifically on elongation rather than enhancing origin firing. This shortens the S-phase duration. Combining data from Spi-1 knockdown in murine cells with Spi-1 overexpression in human cells, we provide evidence that inappropriate Spi-1 expression is directly responsible for the replication alteration observed. Importantly, the acceleration of replication progression coincides with an increase in the frequency of genomic mutations without inducing DNA breakage. Thus, we propose that the hitherto unsuspected role for spi-1 oncogene in promoting replication elongation and genomic mutation promotes blastic progression during leukemic development.


Assuntos
Quebras de DNA , Replicação do DNA/genética , Leucemia/genética , Pré-Leucemia/genética , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Animais , Crise Blástica/genética , Diferenciação Celular/genética , DNA de Neoplasias/biossíntese , DNA de Neoplasias/genética , Regulação para Baixo , Eritroblastos/patologia , Eritroblastos/fisiologia , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Humanos , Leucemia/patologia , Camundongos , Camundongos Transgênicos , Pré-Leucemia/patologia , Proteínas Proto-Oncogênicas/biossíntese , RNA Interferente Pequeno/genética , Fase S/genética , Transativadores/biossíntese
14.
Mol Cell Biol ; 29(10): 2852-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19289502

RESUMO

Spi-1 and Fli-1 are ETS transcription factors recurrently deregulated in mouse erythroleukemia induced by Friend viruses. Since they share the same core DNA binding site, we investigated whether they may contribute to erythroleukemia by common mechanisms. Using inducible knockdown, we demonstrated that Fli-1 contributes to proliferation, survival, and differentiation arrest of erythroleukemic cells harboring an activated fli-1 locus. Similarly, we used inducible Fli-1 knockdown and either hexamethylenebisacetamide (HMBA)- or small interfering RNA-mediated Spi-1 knockdown to investigate their respective contributions in erythroleukemic cells harboring an activated spi-1 locus. In these cells, simple or double knockdown of both Spi-1 and Fli-1 additively contributed to induce proliferation arrest and differentiation. Transcriptome profiling revealed that virtually all transcripts affected by both Fli-1 knockdown and HMBA are affected in an additive manner. Among these additively downregulated transcripts, more than 20% encode proteins involved in ribosome biogenesis, and conserved ETS binding sites are present in their gene promoters. Through chromatin immunoprecipitation, we demonstrated the association of Spi-1 and Fli-1 on these promoters in Friend erythroleukemic cells. These data lead us to propose that the oncogenicity of Spi-1, Fli-1, and possibly other ETS transcription factors may involve their ability to stimulate ribosome biogenesis.


Assuntos
Vírus da Leucemia Murina de Friend/metabolismo , Leucemia Eritroblástica Aguda , Peptídeos/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Ribossomos/metabolismo , Células Tumorais Cultivadas/fisiologia , Animais , Apoptose/fisiologia , Proliferação de Células , Vírus da Leucemia Murina de Friend/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Peptídeos/genética , Fenótipo , Proteína Proto-Oncogênica c-fli-1/genética
15.
Blood ; 109(7): 3007-14, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17132716

RESUMO

Overexpression of the transcription factor Spi-1/PU.1 in mice leads to acute erythroleukemia characterized by a differentiation block at the proerythroblastic stage. In this study, we made use of a new cellular system allowing us to reach graded expression of Spi-1 in preleukemic cells to dissect mechanisms of Spi-1/ PU-1 in erythroleukemogenesis. This system is based on conditional production of 1 or 2 spi-1-interfering RNAs stably inserted into spi-1 transgenic proerythroblasts. We show that Spi-1 knock-down was sufficient to reinstate the erythroid differentiation program. This differentiation process was associated with an exit from the cell cycle. Evidence is provided that in the presence of erythropoietin (Epo), Spi-1 displays an antiapoptotic role that is independent of its function in blocking erythroid differentiation. Apoptosis inhibited by Spi-1 did not involve activation of the Fas/FasL signaling pathway nor a failure to activate Epo receptor (EpoR). Furthermore, we found that reducing the Spi-1 level yields to ERK dephosphorylation and increased phosphorylation of AKT and STAT5, suggesting that Spi-1 may affect major signaling pathways downstream of the EpoR in erythroid cells. These findings reveal 2 distinct roles for Spi-1 during erythroleukemogenesis: Spi-1 blocks the erythroid differentiation program and acts to impair apoptotic death in cooperation with an Epo signaling.


Assuntos
Eritropoetina/fisiologia , Leucemia Eritroblástica Aguda/etiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Animais , Apoptose/fisiologia , Sequência de Bases , Ciclo Celular/fisiologia , Diferenciação Celular , Eritroblastos/patologia , Eritroblastos/fisiologia , Eritropoese/fisiologia , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Leucemia Eritroblástica Aguda/fisiopatologia , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/genética , Receptores da Eritropoetina/fisiologia , Transdução de Sinais/fisiologia , Transativadores/antagonistas & inibidores , Transativadores/genética
16.
J Biol Chem ; 281(28): 19145-55, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16698794

RESUMO

The expression of the Spi-1/PU.1 transcription factor is tightly regulated as a function of the hematopoietic lineage. It is required for myeloid and B lymphoid differentiation. When overexpressed in mice, Spi-1 is associated with the emergence of transformed proerythroblasts unable to differentiate. In the course of a project undertaken to characterize the oncogenic function of Spi-1, we found that Spi-1 interacts with proteins of the spliceosome in Spi-1-transformed proerythroblasts and participates in alternative splice site selection. Because Spi-1 is a transcription factor, it could be hypothesized that these two functions are coordinated. Here, we have developed a system allowing the characterization of transcription and splicing from a single target. It is shown that Spi-1 is able to regulate alternative splicing of a pre-mRNA for a gene whose transcription it regulates. Using a combination of Spi-1 mutants and Spi-1-dependent promoters, we demonstrate that Spi-1 must bind and transactivate a given promoter to favor the use of the proximal 5' alternative site. This establishes that Spi-1 affects splicing decisions in a promoter binding-dependent manner. These results provide new insight into how Spi-1 may act in the blockage of differentiation by demonstrating that it can deregulate gene expression and also modify the nature of the products generated from target genes.


Assuntos
Processamento Alternativo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Mutação , Regiões Promotoras Genéticas , Núcleo Celular/metabolismo , Células HeLa , Humanos , Plasmídeos/metabolismo , Ligação Proteica , Splicing de RNA , RNA Mensageiro/metabolismo , Transcrição Gênica , Ativação Transcricional
17.
Oncogene ; 23(25): 4389-99, 2004 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15064749

RESUMO

The hematopoietic transcription factor Spi-1/PU.1 is an oncoprotein participating to the malignant transformation of proerythroblasts in the Friend erythroleukemia or in the erythroleukemic process developed in spi-1 transgenic mice. Overexpression of Spi-1 in proerythroblasts blocks their differentiation. We have shown that Spi-1 promotes the use of the proximal 5'-splice site during the E1A pre-mRNA splicing and interferes with the effect of TLS (Translocated in LipoSarcoma) in this splicing assay. TLS was identified from chromosomal translocations in human liposarcoma and acute myeloid leukemia. Here, we determine the function of Spi-1 domains in splicing and in the interference with TLS. In transient transfection assays in erythroid cells, we show that the DNA binding domain cooperates with the transactivation domain or the PEST region of Spi-1 to modify the function of TLS in splicing. Interestingly, the 27 C-terminal amino acids, which determine the DNA binding activity of Spi-1, are necessary for the splicing function of Spi-1 as well as for its ability to interfere with TLS. Finally, we demonstrate that in leukemic proerythroblasts overexpressing Spi-1, TLS has lost its splicing effect. Thus, we hypothesize that oncogenic pathways in proerythroblasts may involve the ability of Spi-1 to alter splicing.


Assuntos
Células Precursoras Eritroides/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Sítios de Splice de RNA/genética , Splicing de RNA/fisiologia , Proteína FUS de Ligação a RNA/fisiologia , Transativadores/fisiologia , Proteínas E1A de Adenovirus/genética , Animais , Sítios de Ligação , Transformação Celular Neoplásica , DNA/metabolismo , Genes Reporter , Leucemia Eritroblástica Aguda/genética , Camundongos , Células-Tronco Neoplásicas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína/fisiologia , Proteínas Proto-Oncogênicas/química , Precursores de RNA/metabolismo , Splicing de RNA/genética , RNA Neoplásico/metabolismo , Proteína FUS de Ligação a RNA/antagonistas & inibidores , Proteína FUS de Ligação a RNA/química , Relação Estrutura-Atividade , Transativadores/química , Ativação Transcricional , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...