Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
3.
Oncotarget ; 8(23): 37104-37114, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28415748

RESUMEN

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.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Replicación del ADN , Proteína Fosfatasa 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Ciclo Celular/genética , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Regulación Leucémica de la Expresión Génica , Humanos , Células K562 , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Ratones Transgénicos , Fosforilación , Proteína Fosfatasa 1/genética , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Transactivadores/genética
4.
PLoS One ; 7(11): e49052, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23145067

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/genética , Regulación hacia Abajo/genética , Leucemia/genética , Sistema de Señalización de MAP Quinasas/genética , Proteínas de la Membrana/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas/genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Humanos , Leucemia/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo
5.
Nucleic Acids Res ; 40(18): 8927-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22790984

RESUMEN

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.


Asunto(s)
Leucemia Eritroblástica Aguda/genética , Proteínas Proto-Oncogénicas/metabolismo , Elementos Reguladores de la Transcripción , Transactivadores/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Islas de CpG , ADN/química , ADN/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genoma , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Ratones Transgénicos , Motivos de Nucleótidos , Análisis de Secuencia de ADN , Sitio de Iniciación de la Transcripción
6.
Blood ; 119(18): 4228-41, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22438255

RESUMEN

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.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/fisiología , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Imidazoles/farmacología , Indoles , Leucemia de Mastocitos/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Fosfotirosina/análisis , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Pirroles/farmacología , Quinolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Factor de Transcripción STAT5/antagonistas & inhibidores , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/fisiología , Ensayo de Tumor de Célula Madre
7.
Cancer Res ; 70(17): 6757-66, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20660370

RESUMEN

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.


Asunto(s)
Roturas del ADN , Replicación del ADN/genética , Leucemia/genética , Preleucemia/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Crisis Blástica/genética , Diferenciación Celular/genética , ADN de Neoplasias/biosíntesis , ADN de Neoplasias/genética , Regulación hacia Abajo , Eritroblastos/patología , Eritroblastos/fisiología , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Inestabilidad Genómica , Humanos , Leucemia/patología , Ratones , Ratones Transgénicos , Preleucemia/patología , Proteínas Proto-Oncogénicas/biosíntesis , ARN Interferente Pequeño/genética , Fase S/genética , Transactivadores/biosíntesis
8.
PLoS One ; 4(5): e5721, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19492092

RESUMEN

Overexpression of the transcription factor Spi-1/PU.1 by transgenesis in mice induces a maturation arrest at the proerythroblastic stage of differentiation. We have previously isolated a panel of spi-1 transgenic erythroleukemic cell lines that proliferated in the presence of either erythropoietin (Epo) or stem cell factor (SCF). Using these cell lines, we observed that EpoR stimulation by Epo down-regulated expression of the SCF receptor Kit and induced expression of the Src kinase Lyn. Furthermore, enforced expression of Lyn in the cell lines increased cell proliferation in response to Epo, but reduced cell growth in response to SCF in accordance with Lyn ability to down-regulate Kit expression. Together, the data suggest that Epo-R/Lyn signaling pathway is essential for extinction of SCF signaling leading the proerythroblast to strict Epo dependency. These results highlight a new role for Lyn as an effector of EpoR in controlling Kit expression. They suggest that Lyn may play a central role in during erythroid differentiation at the switch between proliferation and maturation.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Eritroblastos/efectos de los fármacos , Eritroblastos/enzimología , Eritropoyetina/farmacología , Leucemia/enzimología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Familia-src Quinasas/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Citocinas/farmacología , Eritroblastos/citología , Ratones , Proteínas Mutantes/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Eritropoyetina/metabolismo , Factor de Transcripción STAT5/metabolismo , Transactivadores/metabolismo
9.
Mol Cell Biol ; 29(10): 2852-64, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19289502

RESUMEN

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.


Asunto(s)
Virus de la Leucemia Murina de Friend/metabolismo , Leucemia Eritroblástica Aguda , Péptidos/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Ribosomas/metabolismo , Células Tumorales Cultivadas/fisiología , Animales , Apoptosis/fisiología , Proliferación Celular , Virus de la Leucemia Murina de Friend/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Péptidos/genética , Fenotipo , Proteína Proto-Oncogénica c-fli-1/genética
10.
Retrovirology ; 5: 99, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18983647

RESUMEN

The Friend virus SFFV (Spleen Focus Forming Virus) provokes an acute erythroblastosis in susceptible strains of mice that progresses to overt erythroleukemia by a multi-step process. For virologists, the Friend virus-induced disease has provided deep insights into the host mechanisms influencing susceptibility to retroviral infection and viremia. These insights have contributed to the understanding of HIV and other human retroviral infections. For cell biologists and oncologists, this leukemia has been a powerful experimental model to identify critical oncogenes involved in a multi-stage process, to understand the contribution of host genes to cancer development, and to investigate the mechanisms leading to cell growth autonomy. This model also provided an example of oncogenic reversion since Friend tumor cells can reinitiate their erythroid differentiation program when exposed in vitro to some chemical inducers. This review highlights recent findings demonstrating that the leukemic progression depends on the cooperation of at least two oncogenic events, one interfering with differentiation and one conferring a proliferative advantage. The Friend model of leukemia progression recapitulates the two phases of human acute myeloid leukemia (AML). Coupling of insights from studies on the Friend erythroleukemia with knowledge on AML might allow a better understanding of the molecular mechanisms involved in the evolution of leukemia in mice and men.


Asunto(s)
Transformación Celular Neoplásica , Leucemia Eritroblástica Aguda/virología , Oncogenes , Virus Formadores de Foco en el Bazo/patogenicidad , Animales , Animales Recién Nacidos , Diferenciación Celular , Proliferación Celular , Humanos , Leucemia Eritroblástica Aguda/patología , Leucemia Mieloide Aguda/patología , Masculino , Ratones
11.
Blood ; 111(6): 3163-72, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18182570

RESUMEN

The model of erythroleukemia caused by Spi-1/PU.1 transgenesis in mice is a multistage disease. A preleukemic step is characterized by an acute proliferation of proerythroblasts due to the arrest of differentiation provoked by Spi-1/PU.1. Later on, a blastic crisis occurs associated with somatic oncogenic mutations in the stem cell factor (SCF) receptor kit. To gain insights into the mechanisms of the leukemic progression, we performed proteomic profiling analyses of proerythroblasts isolated at the 2 stages of the disease. Our results indicate that the level of ezrin, a membrane cytoskeletal crosslinker, is increased in the leukemic cells. We show that Kit oncogenic forms are responsible for ezrin phosphorylation and that phosphorylation rather than overexpression is essential in the leukemic proerythroblasts. Using expression of dominant-negative forms of ezrin, we show that phosphorylation of ezrin on residue Y353 participates in apoptosis resistance, whereas phosphorylation on residue Y145 promotes proliferation of the leukemic cells in vitro and in vivo. Another recurrent oncogenic form of tyrosine kinases (Flt3) most frequently involved in human myeloid leukemia was also able to phosphorylate ezrin. These findings point to a new role for ezrin as signaling player in the development of leukemia, being a downstream effector of oncogenic tyrosine kinases in leukemic blasts.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Línea Celular , Proliferación Celular , Proteínas del Citoesqueleto/genética , Eritroblastos/citología , Eritroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patología , Ratones , Ratones Transgénicos , Mutación/genética , Péptidos/genética , Péptidos/metabolismo , Fosfotirosina/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética
12.
Blood ; 109(7): 3007-14, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17132716

RESUMEN

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.


Asunto(s)
Eritropoyetina/fisiología , Leucemia Eritroblástica Aguda/etiología , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Animales , Apoptosis/fisiología , Secuencia de Bases , Ciclo Celular/fisiología , Diferenciación Celular , Eritroblastos/patología , Eritroblastos/fisiología , Eritropoyesis/fisiología , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patología , Leucemia Eritroblástica Aguda/fisiopatología , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , Receptores de Eritropoyetina/fisiología , Transducción de Señal/fisiología , Transactivadores/antagonistas & inhibidores , Transactivadores/genética
13.
Haematologica ; 91(12): 1644-52, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17145601

RESUMEN

The models of acute erythroleukemia caused in mice by the Friend retrovirus SFFV (spleen focus forming virus) and the Spi-1/PU.1 transgenesis provide considerable information to help to understand the molecular mechanisms underlying the multi-stage nature of leukemia. Leukemogenesis in these murine models is initiated from an acute hyperplasia of erythroid progenitor cells followed later on by a blastic crisis. This review highlights recent findings demonstrating the key roles of the co-operation of two mutations occurring during leukemic progression, a mutation interfering with differentiation and a mutation conferring a proliferative advantage to cells. Through their multi-step evolution, these mouse erythroleukemia models resemble the two phases of human acute myeloid leukemia (AML). The findings we discuss provide evidence for similar molecular mechanisms involved in the evolution of leukemia in mice and men.


Asunto(s)
Modelos Animales de Enfermedad , Leucemia Eritroblástica Aguda/genética , Leucemia Mieloide Aguda/genética , Animales , Virus de la Leucemia Murina de Friend/genética , Humanos , Leucemia Eritroblástica Aguda/patología , Leucemia Mieloide Aguda/patología , Ratones , Ratones Transgénicos
14.
J Biol Chem ; 281(28): 19145-55, 2006 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-16698794

RESUMEN

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.


Asunto(s)
Empalme Alternativo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Mutación , Regiones Promotoras Genéticas , Núcleo Celular/metabolismo , Células HeLa , Humanos , Plásmidos/metabolismo , Unión Proteica , Empalme del ARN , ARN Mensajero/metabolismo , Transcripción Genética , Activación Transcripcional
16.
J Mol Biol ; 355(1): 9-19, 2006 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-16298389

RESUMEN

The TAL-1/SCL gene encodes a basic helix-loop-helix (bHLH) transcription factor essential for primitive hematopoiesis and for adult erythroid and megakaryocytic development. Activated transcription of TAL-1 as a consequence of chromosomal rearrangements is associated with a high proportion of human T cell acute leukemias, showing that appropriate control of TAL-1 is crucial for the formation and subsequent fate of hematopoietic cells. Hence, the knowledge of the mechanisms, which govern the pattern of TAL-1 expression in hematopoiesis, is of great interest. We previously described a silencer in the 3'-untranslated region of human TAL-1, the activity of which is mediated through binding of a tissue-specific 40 kDa nuclear protein to a new DNA recognition motif, named tal-RE. Here, we show that tal-RE-binding activity, high in immature human hematopoietic progenitors is down regulated upon erythroid and megakaryocytic differentiation. This expression profile helped us to identify that PU.1/Spi-1 binds to the tal-RE sequences in vitro and occupies the TAL-1 silencer in vivo. By expressing a mutant protein containing only the ETS domain of PU.1 in human erythroleukemic HEL cells, we demonstrated that PU.1 mediates the transcriptional repression activity of the silencer. We found that ectopic PU.1 is not able to induce silencing activity in PU.1-negative Jurkat T cells, indicating that PU.1 activity, although necessary, is not sufficient to confer transcriptional repression activity to the TAL-1 silencer. Finally, we showed that the silencer is also active in TAL-1-negative myeloid HL60 cells that express PU.1 at high levels. In summary, our study shows that PU.1, in addition to its positive role in TAL-1 expression in early hematopoietic progenitors, may also act as a mediator of TAL-1 silencing in some hematopoietic lineages.


Asunto(s)
Silenciador del Gen , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Ubiquitina-Proteína Ligasas/genética , Línea Celular Tumoral , Regulación hacia Abajo , Hematopoyesis/genética , Humanos , Leucemia/patología , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Elementos de Respuesta , Transactivadores/metabolismo , Factores de Transcripción , Activación Transcripcional , Transfección
17.
Cancer Cell ; 8(6): 467-78, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16338660

RESUMEN

The erythroleukemia developed by spi-1/PU.1 transgenic mice is a multistage process characterized by an early arrest of the proerythroblast differentiation followed later on by malignant transformation. Herein, we report the presence of acquired mutations in the SCF receptor gene (Kit) in 86% of tumors isolated during the late stage of the disease. Kit mutations affect codon 814 or 818. Ectopic expression of Kit mutants in nonmalignant proerythroblasts confers erythropoietin independence and tumorigenicity to cells. Using PP1, PP2, and imatinib mesylate, we show that Kit mutants are responsible for the autonomous expansion of malignant cells via Erk1/2 and PI3K/Akt activations. These findings represent a proof of principle for oncogenic cooperativity between one proliferative and one differentiation blocking event for the development of an overt leukemia.


Asunto(s)
Leucemia Eritroblástica Aguda/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas/biosíntesis , Transactivadores/biosíntesis , Animales , Benzamidas , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mesilato de Imatinib , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Ratones Transgénicos , Mutación , Piperazinas/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-kit/efectos de los fármacos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Factores de Tiempo , Transactivadores/genética
18.
Blood ; 106(5): 1808-16, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15890687

RESUMEN

The erythroleukemia developed by spi-1/PU.1-transgenic mice is a model of multistage oncogenic process. Isolation of tumor cells representing discrete stages of leukemic progression enables the dissection of some of the critical events required for malignant transformation. To elucidate the molecular mechanisms of multistage leukemogenesis, we developed a microarray transcriptome analysis of nontumorigenic (HS1) and tumorigenic (HS2) proerythroblasts from spi-1-transgenic mice. The data show that transcriptional up-regulation of the sphingosine kinase gene (SPHK1) is a recurrent event associated with the tumorigenic phenotype of these transgenic proerythroblasts. SPHK1 is an enzyme of the metabolism of sphingolipids, which are essential in several biologic processes, including cell proliferation and apoptosis. HS1 erythroleukemic cells engineered to overexpress the SPHK1 protein exhibited growth proliferative advantage, increased clonogenicity, and resistance to apoptosis in reduced serum level by a mechanism involving activation of the extracellular signal-related kinases 1/2 (ERK1/2) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. In addition, SPHK1-overexpressing HS1 cells acquired tumorigenicity when engrafted in vivo. Finally, enforced expression of a dominant-negative mutant of SPHK1 in HS2 tumorigenic cells or treatment with a pharmacologic inhibitor reduced both cell growth and apoptosis resistance. Altogether, these data suggest that overexpression of the sphingosine kinase may represent an oncogenic event during the multistep progression of an erythroleukemia.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Regulación Enzimológica de la Expresión Génica , Leucemia Eritroblástica Aguda/metabolismo , Neoplasias Experimentales/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Transformación Celular Neoplásica/genética , Clonación Molecular , Progresión de la Enfermedad , Eritroblastos/citología , Eritroblastos/metabolismo , Perfilación de la Expresión Génica , Genes Dominantes , Leucemia Eritroblástica Aguda/genética , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Regulación hacia Arriba
19.
Neurosci Lett ; 379(3): 152-7, 2005 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-15843054

RESUMEN

Long-term synaptic change in the cortex and the hippocampus is believed to require the highly localized delivery and translation of mRNAs in the dendritic shafts and spines. The molecular interactions that underlie local signalling between synapses and mRNAs are still largely undefined. After purification from total brain extracts, the NMDA receptor is known to be associated with numerous proteins, including the multifunctional RNA-binding factor TLS (also called FUS). In non-neural tissue, TLS is a vital nuclear protein with roles in DNA repair, homologous recombination, transcriptional regulation and pre-mRNA processing. We have examined the distribution of TLS in hippocampal neurones, both in the adult brain and in mature primary cultures, using subcellular fractionation and immunofluorescence techniques. TLS immunoreactivity is largely excluded from the neuronal nucleus and is found in the cytosol and in somatodendritic particles. In some of these particles, TLS colocalizes with Sam68, a nuclear RNA-binding protein that we previously showed is incorporated into dendritic RNA granules. Some of the TLS clusters also colocalize with NMDA receptor clusters. Finally, TLS clusters are occasionally seen within spine heads. The apparent removal of TLS from the nucleus might result in specific patterns of mRNA transcription or splicing in hippocampal neurones. TLS may also contribute to steering, anchoring or regulating mRNAs at synaptic sites.


Asunto(s)
Núcleo Celular/metabolismo , Espinas Dendríticas/metabolismo , Hipocampo/citología , Neuronas/citología , Proteína FUS de Unión a ARN/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Western Blotting/métodos , Fraccionamiento Celular/métodos , Células Cultivadas , Proteínas de Unión al ADN , Embrión de Mamíferos , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo
20.
Mol Cell Biol ; 25(7): 2832-45, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15767686

RESUMEN

Regulation of the hematopoietic transcription factor PU.1 (Spi-1) plays a critical role in the development of white cells, and abnormal expression of PU.1 can lead to leukemia. We previously reported that the PU.1 promoter cannot induce expression of a reporter gene in vivo, and cell-type-specific expression of PU.1 in stable lines was conferred by a 3.4-kb DNA fragment including a DNase I hypersensitive site located 14 kb upstream of the transcription start site. Here we demonstrate that this kb -14 site confers lineage-specific reporter gene expression in vivo. This kb -14 upstream regulatory element contains two 300-bp regions which are highly conserved in five mammalian species. In Friend virus-induced erythroleukemia, the spleen focus-forming virus integrates into the PU.1 locus between these two conserved regions. DNA binding experiments demonstrated that PU.1 itself and Elf-1 bind to a highly conserved site within the proximal homologous region in vivo. A mutation of this site abolishing binding of PU.1 and Elf-1 led to a marked decrease in the ability of this upstream element to direct activity of reporter gene in myelomonocytic cell lines. These data suggest that a potential positive autoregulatory loop mediated through an upstream regulatory element is essential for proper PU.1 gene expression.


Asunto(s)
Regulación de la Expresión Génica/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Elementos de Respuesta/genética , Transactivadores/genética , Transactivadores/metabolismo , Animales , Linfocitos B/metabolismo , Secuencia de Bases , Sitios de Unión , Linaje de la Célula , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación/genética , Células Mieloides/metabolismo , Especificidad de Órganos , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...