Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Leukemia ; 32(4): 855-864, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28951561

RESUMEN

Acute myeloid leukemia (AML) is an aggressive and lethal blood cancer originating from rare populations of leukemia stem cells (LSCs). AML relapse after conventional chemotherapy is caused by a remaining population of drug-resistant LSCs. Selective targeting of the chemoresistant population is a promising strategy for preventing and treating AML relapse. Polycomb repressive complex 2 (PRC2) trimethylates histone H3 at lysine 27 to maintain the stemness of LSCs. Here, we show that quiescent LSCs expressed the highest levels of enhancer of zeste (EZH) 1 and EZH2, the PRC2 catalytic subunits, in the AML hierarchy, and that dual inactivation of EZH1/2 eradicated quiescent LSCs to cure AML. Genetic deletion of Ezh1/2 in a mouse AML model induced cell cycle progression of quiescent LSCs and differentiation to LSCs, eventually eradicating AML LSCs. Quiescent LSCs showed PRC2-mediated suppression of Cyclin D, and Cyclin D-overexpressing AML was more sensitive to chemotherapy. We have developed a novel EZH1/2 dual inhibitor with potent inhibitory activity against both EZH1/2. In AML mouse models and patient-derived xenograft models, the inhibitor reduced the number of LSCs, impaired leukemia progression, and prolonged survival. Taken together, these results show that dual inhibition of EZH1/2 is an effective strategy for eliminating AML LSCs.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Animales , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL
2.
Leukemia ; 31(10): 2200-2210, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28210005

RESUMEN

Rearrangements involving the NUP98 gene resulting in fusions to several partner genes occur in acute myeloid leukemia and myelodysplastic syndromes. This study demonstrates that the second FG repeat domain of the NUP98 moiety of the NUP98-HOXA9 fusion protein is important for its cell immortalization and leukemogenesis activities. We demonstrate that NUP98-HOXA9 interacts with mixed lineage leukemia (MLL) via this FG repeat domain and that, in the absence of MLL, NUP98-HOXA9-induced cell immortalization and leukemogenesis are severely inhibited. Molecular analyses indicate that MLL is important for the recruitment of NUP98-HOXA9 to the HOXA locus and for NUP98-HOXA9-induced HOXA gene expression. Our data indicate that MLL is crucial for NUP98-HOXA9 leukemia initiation.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación Leucémica de la Expresión Génica/genética , N-Metiltransferasa de Histona-Lisina/fisiología , Proteínas de Homeodominio/fisiología , Leucemia Experimental/genética , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Fusión Oncogénica/fisiología , Animales , Inmunoprecipitación de Cromatina , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Humanos , Leucemia Experimental/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteína de la Leucemia Mieloide-Linfoide/deficiencia , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Unión Proteica , Dominios Proteicos , Mapeo de Interacción de Proteínas , Quimera por Radiación , Transfección
3.
Leukemia ; 29(3): 576-85, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25234168

RESUMEN

In leukemogenesis, Notch signaling can be up and downregulated in a context-dependent manner. The transcription factor hairy and enhancer of split-1 (Hes1) is well-characterized as a downstream target of Notch signaling. Hes1 encodes a basic helix-loop-helix-type protein, and represses target gene expression. Here, we report that deletion of the Hes1 gene in mice promotes acute myeloid leukemia (AML) development induced by the MLL-AF9 fusion protein. We then found that Hes1 directly bound to the promoter region of the FMS-like tyrosine kinase 3 (FLT3) gene and downregulated the promoter activity. FLT3 was consequently upregulated in MLL-AF9-expressing immortalized and leukemia cells with a Hes1- or RBPJ-null background. MLL-AF9-expressing Hes1-null AML cells showed enhanced proliferation and ERK phosphorylation following FLT3 ligand stimulation. FLT3 inhibition efficiently abrogated proliferation of MLL-AF9-induced Hes1-null AML cells. Furthermore, an agonistic anti-Notch2 antibody induced apoptosis of MLL-AF9-induced AML cells in a Hes1-wild type but not a Hes1-null background. We also accessed two independent databases containing messenger RNA (mRNA) expression profiles and found that the expression level of FLT3 mRNA was negatively correlated with those of HES1 in patient AML samples. These observations demonstrate that Hes1 mediates tumor suppressive roles of Notch signaling in AML development, probably by downregulating FLT3 expression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/genética , Leucemia Mieloide Aguda/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Proliferación Celular , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/deficiencia , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Ratones , Ratones Transgénicos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Análisis de Supervivencia , Factor de Transcripción HES-1 , Tirosina Quinasa 3 Similar a fms/metabolismo
4.
Leukemia ; 22(2): 273-80, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17989716

RESUMEN

Promyelocytic leukemia (PML) is a nuclear protein that functions as a regulator of transcription, cell proliferation, apoptosis and myeloid cell differentiation. PML is subjected to post-translational modifications such as sumoylation and phosphorylation. However, the physiological significance of these modifications, especially for myeloid cell differentiation, remains unclear. In this report, we found that four serine residues in the PML C-terminal region are highly phosphorylated in a myeloid cell line. Wild-type PML accelerated G-CSF-induced granulocytic differentiation, but a phosphorylation-deficient PML mutant failed. PML interacted with C/EBP epsilon, a transcription factor essential for granulopoiesis, activated C/EBP epsilon-mediated transcription in concert with p300 and accelerated C/EBP epsilon-induced granulocytic differentiation. Phosphorylation of PML was required for stimulating C/EBP epsilon-dependent transcription and accelerating C/EBP epsilon-induced granulocytic differentiation. We also found that PML phosphorylation was required for stimulation of PU.1-dependent transcription and acceleration of PU.1-induced granulocytic differentiation. These results suggest that phosphorylation plays essential roles in the regulation of PML to accelerate granulocytic differentiation through multiple pathways.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Diferenciación Celular , Granulocitos/citología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Ratones , Fosforilación , Proteína de la Leucemia Promielocítica , Transcripción Genética
5.
Oncogene ; 26(51): 7231-9, 2007 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-17533375

RESUMEN

The AML1 transcription factor complex is the most frequent target of leukemia-associated chromosomal translocations. Homeodomain-interacting protein kinase 2 (HIPK2) is a part of the AML1 complex and activates AML1-mediated transcription. However, chromosomal translocations and mutations of HIPK2 have not been reported. In the current study, we screened mutations of the HIPK2 gene in 50 cases of acute myeloid leukemia (AML) and in 80 cases of myelodysplastic syndrome (MDS). Results indicated there were two missense mutations (R868W and N958I) in the speckle-retention signal (SRS) domain of HIPK2. Subcellular localization analyses indicated that the two mutants were largely localized to nuclear regions with conical or ring shapes, and were somewhat diffused in the nucleus, in contrast to the wild type, which were mainly localized in nuclear speckles. The mutations impaired the overlapping localization of AML1 and HIPK2. The mutants showed decreased activities and a dominant-negative function over wild-type protein in AML1- and p53-dependent transcription. These findings suggest that dysfunction of HIPK2 may play a role in the pathogenesis of leukemia.


Asunto(s)
Proteínas Portadoras/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Leucemia Mieloide Aguda/genética , Mutación Missense , Síndromes Mielodisplásicos/genética , Proteínas Serina-Treonina Quinasas/genética , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Fracciones Subcelulares/metabolismo
6.
Leukemia ; 20(6): 1002-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16628190

RESUMEN

Mutations of the GATA1 gene on chromosome X have been found in almost all cases of transient myeloproliferative disorder and acute megakaryoblastic leukemia (AMKL) accompanying Down syndrome (DS). Although most GATA1 mutations lead to the expression of GATA1s lacking the N-terminal activation domain, we recently found two novel GATA1 proteins with defects in another N-terminal region. It has been suggested that loss of the N-terminal portion of GATA1 might interfere with physiological interactions with the critical megakaryocytic transcription factor RUNX1, and this would imply that GATA1s is not able to interact properly with RUNX1. However, the interaction domain of GATA1 remains controversial. In this study, we show that GATA1 binds to RUNX1 through its zinc-finger domains, and that the C-finger is indispensable for synergy with RUNX1. All of the patient-specific GATA1 mutants interacted efficiently with RUNX1 and retained their ability to act synergistically with RUNX1 on the megakaryocytic GP1balpha promoter, whereas the levels of transcriptional activities were diverse among the mutants. Thus, our data indicate that physical interaction and synergy between GATA1 and RUNX1 are retained in DS-AMKL, although it is still possible that increased RUNX1 activity plays a role in the development of leukemia in DS.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Síndrome de Down/complicaciones , Síndrome de Down/genética , Factor de Transcripción GATA1/genética , Leucemia Megacarioblástica Aguda/complicaciones , Leucemia Megacarioblástica Aguda/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Aberraciones Cromosómicas , Humanos , Mutación , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Codorniz , Transcripción Genética
7.
EMBO J ; 20(24): 7184-96, 2001 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-11742995

RESUMEN

The AML1-CBF beta transcription factor complex is the most frequent target of specific chromosome translocations in human leukemia. The MOZ gene, which encodes a histone acetyltransferase (HAT), is also involved in some leukemia-associated translocations. We report here that MOZ is part of the AML1 complex and strongly stimulates AML1-mediated transcription. The stimulation of AML1-mediated transcription is independent of the inherent HAT activity of MOZ. Rather, a potent transactivation domain within MOZ appears to be essential for stimulation of AML1-mediated transcription. MOZ, as well as CBP and MOZ-CBP, can acetylate AML1 in vitro. The amount of AML1-MOZ complex increases during the differentiation of M1 myeloid cells into monocytes/macrophages, suggesting that the AML1-MOZ complex might play a role in cell differentiation. On the other hand, the MOZ-CBP fusion protein, which is created by the t(8;16) translocation associated with acute monocytic leukemia, inhibits AML1-mediated transcription and differentiation of M1 cells. These results suggest that MOZ-CBP might induce leukemia by antagonizing the function of the AML1 complex.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/fisiología , Activación Transcripcional/fisiología , Acetiltransferasas/química , Secuencia de Aminoácidos , Secuencia de Bases , Proteína de Unión a CREB , Diferenciación Celular , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Cartilla de ADN , Histona Acetiltransferasas , Humanos , Macrófagos/citología , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Recombinantes de Fusión/química , Homología de Secuencia de Aminoácido , Transactivadores/química
8.
J Virol ; 75(11): 5119-28, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11333893

RESUMEN

Epstein-Barr virus (EBV) nuclear antigen leader protein (EBNA-LP) is a phosphoprotein suggested to play important roles in EBV-induced immortalization of B cells. One of the potential functions of EBNA-LP is a cooperative induction with EBNA-2 of viral and cellular gene expression, including that of the genes for viral latent membrane protein 1 (LMP-1) and cellular cyclin D2. We report here that the phosphorylation of EBNA-LP by cellular kinase(s) is critical to its ability to cooperate with EBNA-2 in up-regulating the expression of LMP-1 in a B-lymphoma cell line. Our conclusion is based on the following observations. (i) Mass-spectrometric analysis of purified EBNA-LP and mutational analyses of EBNA-LP revealed that the serine residue at position 35 in the W2 repeat domain is the major phosphorylation site of EBNA-LP in vivo. (ii) Substitutions of this site in each W2 repeat domain with alanine markedly reduced the ability of the protein to induce LMP-1 expression in combination with EBNA-2 in Akata cells. (iii) Replacement at the major phosphorylation sites with glutamic acids restored the wild-type phenotype. It is well established that this substitution mimics constitutive phosphorylation. These results indicated that the coactivator function of EBNA-LP is regulated by phosphorylation.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas de la Matriz Viral/biosíntesis , Proteínas Virales/metabolismo , Alanina/metabolismo , Sustitución de Aminoácidos , Animales , Linfocitos B , Sitios de Unión , Linfoma de Burkitt , Células COS , Línea Celular , Línea Celular Transformada , Eliminación de Gen , Ácido Glutámico/metabolismo , Humanos , Espectrometría de Masas , Fosforilación , Fosfotransferasas/metabolismo , Transfección , Regulación hacia Arriba , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación
9.
Leukemia ; 15(1): 89-94, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11243405

RESUMEN

Histone acetyltransferase p300 functions as a transcriptional co-activator which interacts with a number of transcription factors. Monocytic leukemia zinc finger protein (MOZ) has histone acetyltransferase activity. We report the fusion of the MOZ gene to the p300 gene in acute myeloid leukemia with translocation t(8;22)(p11;q13). FISH and Southern blot analyses showed the rearrangement of the MOZ and p300 genes. We determined the genomic structure of the p300 and the MOZ genes and the breakpoints of the translocation. Analysis of fusion transcripts indicated that the zinc finger and acetyltransferase domains of MOZ are fused to a largely intact p300. These results suggest that MOZ-p300, which has two acetyltransferase domains, could be involved in leukemogenesis through aberrant regulation of histone acetylation.


Asunto(s)
Acetiltransferasas/genética , Proteínas de Ciclo Celular/genética , Cromosomas Humanos Par 22 , Cromosomas Humanos Par 8 , Leucemia Monocítica Aguda/genética , Translocación Genética , Histona Acetiltransferasas , Humanos , Leucemia Monocítica Aguda/patología , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica , Factores de Transcripción , Factores de Transcripción p300-CBP
10.
Virology ; 279(2): 401-13, 2001 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-11162796

RESUMEN

There is a growing body of evidence for the importance of the nuclear matrix in various nuclear events including gene expression and DNA replication. Epstein-Barr virus (EBV) nuclear antigen leader protein (EBNA-LP) is a nuclear matrix-associated protein that has been suggested to play an important role in EBV-induced transformation. To define the biological significance of the association of EBNA-LP with the nuclear matrix, we mapped the domain of EBNA-LP responsible for nuclear matrix association and investigated the functions of the EBNA-LP mutant mutagenized by substitution of alanines for the cluster of arginine residues in the mapped region. The results of the present study were as follows. (i) Transiently expressed EBNA-LP in COS-7 or BOSC23 cells was associated with the nuclear matrix, similarly to that in EBV-infected B cells. (ii) Mutational analysis of EBNA-LP revealed that a 10-amino acid segment of EBNA-LP is critical for nuclear matrix association of the protein. Interestingly, the identified region overlapped with the region CR2 of EBNA-LP conserved among a subset of primate gammaherpesviruses. The identified segment is referred to as EBNA-LP NMTS (nuclear matrix targeting signal). (iii) The EBNA-LP mutant with the arginine to alanine substitutions in NMTS was no longer localized not only to the nuclear matrix but also to the nucleus. (iv) The EBNA-LP mutant lacked its ability to coactivate EBNA-2-dependent transactivation. These results indicated that EBNA-LP needs to be localized in the nucleus and/or associated with the nuclear matrix through CR2 to elicit its function such as the coactivation of the EBNA-2-dependent transcriptional activation.


Asunto(s)
Núcleo Celular/metabolismo , Herpesvirus Humano 4/metabolismo , Matriz Nuclear/metabolismo , Proteínas Virales/metabolismo , Alanina/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Arginina/genética , Western Blotting , Células COS , Línea Celular , Transformación Celular Viral , Chlorocebus aethiops , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Técnica del Anticuerpo Fluorescente , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Activación Transcripcional , Proteínas Virales/análisis , Proteínas Virales/genética
11.
Blood ; 96(2): 655-63, 2000 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10887131

RESUMEN

The AML1-MTG8 fusion transcription factor generated by t(8;21) translocation is thought to dysregulate genes that are crucial for normal differentiation and proliferation of hematopoietic progenitors to cause acute myelogenous leukemia (AML). Although AML1-MTG8 has been shown to repress the transcription of AML1 targets, none of the known targets of AML1 are probably responsible for AML1-MTG8-mediated leukemogenesis. In this study, 24 genes under the downstream control of AML1-MTG8 were isolated by using a differential display technique. Analysis with deletion mutants of AML1-MTG8 demonstrated that the regulation of the majority of these genes requires the region of 51 residues (488-538) containing the Nervy homology region 2 (NHR2), through which AML1-MTG8 interacts with MTGR1. Among the 24 genes identified, 10 were considered to be genes under the control of AML1, because their expression was altered by AML1b or AML1a or both. However, the other 14 genes were not affected by either AML1b or AML1a, suggesting the possibility that AML1-MTG8 regulates a number of specific target genes that are not normally regulated by AML1. Furthermore, an up-regulated gene, TIS11b (ERF-1, cMG1), was highly expressed in t(8;21) leukemic cells, and the overexpression of TIS11b induced myeloid cell proliferation in response to granulocyte colony-stimulating factor. These results suggest that the high-level expression of TIS11b contributes to AML1-MTG8-mediated leukemogenesis. (Blood. 2000;96:655-663)


Asunto(s)
División Celular/genética , Proteínas de Unión al ADN , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/farmacología , Proteínas Inmediatas-Precoces , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/fisiología , Proteínas/genética , Factores de Transcripción/fisiología , Animales , Northern Blotting , Western Blotting , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Granulocitos/patología , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Proteína 1 Compañera de Translocación de RUNX1 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Translocación Genética , Tristetraprolina
12.
Blood ; 96(1): 288-96, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10891464

RESUMEN

The t(8;21) translocation is one of the most frequent chromosomal abnormalities associated with acute myeloid leukemia (AML). In this translocation, the AML1 (CBFA2/PEBP2aB) gene is disrupted and fused to the MTG8 (ETO) gene. The ectopic expression of the resulting AML1-MTG8 fusion gene product in L-G and 32Dcl3 murine myeloid precursor cells stimulates cell proliferation without inducing morphologic terminal differentiation into mature granulocytes in response to granulocyte-colony stimulating factor (G-CSF). This study found that the ectopic expression of AML1-MTG8 elevates the expression of the G-CSF receptor (G-CSFR). Analysis of the promoter region of the G-CSFR gene revealed that up-regulation of G-CSFR expression by AML1-MTG8 does not depend on the AML1-binding sequence, but on the C/EBP (CCAAT/enhancer binding protein) binding site. The results suggest that the overproduction of G-CSFR is at least partly mediated by C/EBPepsilon, whose expression is activated by AML1-MTG8. The ectopic expression of G-CSFR in L-G cells induced cell proliferation in response to G-CSF, but did not inhibit cell differentiation into mature neutrophils. Overexpression of C/EBPepsilon in L-G cells also stimulated G-CSF-dependent cell proliferation. High expression levels of G-CSFR were also found in the leukemic cells of AML patients with t(8;21). Therefore, G-CSF-dependent cell proliferation of myeloid precursor cells may be implicated in leukemogenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Leucemia Mieloide/genética , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Recombinantes/metabolismo , Transcripción Genética , Transfección , Translocación Genética
13.
J Biol Chem ; 275(35): 26856-63, 2000 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10856308

RESUMEN

Ca(2+)-sensitive tyrosine kinase Pyk2 was shown to be involved in angiotensin (Ang) II-mediated activation of extracellular signal-regulated kinase (ERK) via transactivation of epidermal growth factor receptor (EGF-R). In this study, we tested the involvement of Pyk2 and EGF-R in Ang II-induced activation of JNK and c-Jun in cardiac fibroblasts. Ang II markedly stimulated JNK activities, which were abolished by genistein and intracellular Ca(2+) chelators but partially by protein kinase C depletion. Inhibition of EGF-R did not affect Pyk2 and JNK activation by Ang II. Stable transfection with a dominant negative (DN) mutant for Pyk2 (PKM) completely blocked JNK activation by Ang II. DN mutants of Rac1 (DN-Rac1) and MEK kinase (DN-MEKK1) also abolished it, whereas those of Cdc42, RhoA, and Ha-Ras had no effect. Induction of c-Jun gene transcription by Ang II was abolished in PKM, DN-Rac1, and DN-MEKK1, in which Ang II-induced binding of ATF2/c-Jun heterodimer to the activator protein-1 sequence at -190 played a key role. These results suggest that 1) in cardiac fibroblasts activation of JNK and c-Jun by Ang II is initiated by Pyk2-dependent signalings but not by downstream signals of EGF-R or Ras, 2) Rac1 but not Cdc42 is required for JNK activation by Ang II upstream of MEKK1, and 3) ATF-2/c-Jun binding to the activator protein-1 sequence at -190 plays a key role for induction of c-Jun gene by Ang II.


Asunto(s)
Angiotensina II/farmacología , Señalización del Calcio/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Activación Enzimática , Receptores ErbB/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Quinasa 2 de Adhesión Focal , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos , Unión Proteica , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Wistar , Activación Transcripcional , Familia-src Quinasas/genética
14.
Gene ; 241(2): 287-95, 2000 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-10675041

RESUMEN

AML1-MTG8 fusion protein, which is produced from the rearranged gene formed between AML1 and MTG8 in myeloid leukemia with t(8;21) chromosomal translocation, plays an important role in the pathogenesis of leukemia. We previously showed that ectopically expressed AML1-MTG8 fusion protein is associated with an MTG8-like protein in the mouse myeloid precursor cell line L-G, and this association seemed to be required for AML1-MTG8 to stimulate proliferation. As a candidate cDNA for this MTG8-like protein, a 6.4 kb MTGR1 cDNA encoding human MTGR1b protein of 604 amino acids was isolated. Since this cDNA was shorter than the main mRNA (about 7.5 kb), the 5'-end of the MTGR1 cDNA was extended using Marathon Ready cDNA. When the newly obtained 5'-sequence was combined with the previous cDNA, the resultant MTGR1 cDNA (6995 bp), including exon 3 that the previous cDNA lacked, could encode MTGR1a protein of 575 amino acids. Transcripts of the MTGR1 gene were expressed ubiquitously in the human tissues and cell lines examined. PCR analyses of the cDNAs from human tissues showed the presence of various splicing variants with regard to the 5'-region including exons 1, 2 and 3. The MTGR1 gene consists of 14 exons and spans about 68 kb. The genomic structure of MTGR1 is highly similar to those of other MTG 8-family genes, MTG8 and MTG16. MTG16 was recently cloned from the translocation breakpoint of myeloid malignancies with t(16;21) chromosomal translocation.


Asunto(s)
Familia de Multigenes , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas , Proteínas Represoras/genética , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Northern Blotting , ADN Complementario/aislamiento & purificación , Proteínas de Unión al ADN/genética , Exones , Células HeLa , Humanos , Intrones , Datos de Secuencia Molecular , ARN Mensajero , Proteína 1 Compañera de Translocación de RUNX1 , Distribución Tisular , Factores de Transcripción/genética , Células Tumorales Cultivadas
16.
EMBO J ; 17(11): 2994-3004, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9606182

RESUMEN

The AML1 transcription factor and the transcriptional coactivators p300 and CBP are the targets of chromosome translocations associated with acute myeloid leukemia and myelodysplastic syndrome. In the t(8;21) translocation, the AML1 (CBFA2/PEBP2alphaB) gene becomes fused to the MTG8 (ETO) gene. We previously found that the terminal differentiation step leading to mature neutrophils in response to granulocyte colony-stimulating factor (G-CSF) was inhibited by the ectopic expression of the AML1-MTG8 fusion protein in L-G murine myeloid progenitor cells. We show here that overexpression of normal AML1 proteins reverses this inhibition and restores the competence to differentiate. Immunoprecipitation analysis shows that p300 and CREB-binding protein (CBP) interact with AML1. The C-terminal region of AML1 is responsible for the induction of cell differentiation and for the interaction with p300. Overexpression of p300 stimulates AML1-dependent transcription and the induction of cell differentiation. These results suggest that p300 plays critical roles in AML1-dependent transcription during the differentiation of myeloid cells. Thus, AML1 and its associated factors p300 and CBFbeta, all of which are targets of chromosomal rearrangements in human leukemia, function cooperatively in the differentiation of myeloid cells.


Asunto(s)
Acetiltransferasas/metabolismo , Acetiltransferasas/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas Proto-Oncogénicas , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Animales , Diferenciación Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Sinergismo Farmacológico , Regulación de la Expresión Génica , Inhibidores de Crecimiento/fisiología , Células Madre Hematopoyéticas , Histona Acetiltransferasas , Ratones , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Estructura Terciaria de Proteína , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Factores de Transcripción/genética , Células Tumorales Cultivadas , Factores de Transcripción p300-CBP
17.
Mol Cell Biol ; 18(2): 846-58, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9447981

RESUMEN

The AML1-CBFbeta transcription factor complex is essential for the definitive hematopoiesis of all lineages and is the most frequent target of chromosomal rearrangements in human leukemia. In the t(8;21) translocation associated with acute myeloid leukemia (AML), the AML1(CBFA2/PEBP2alphaB) gene is juxtaposed to the MTG8(ETO/CDR) gene. We show here that the resultant AML1-MTG8 gene product specifically and strongly interacts with an 85-kDa phosphoprotein. Molecular cloning of cDNA indicated that the AML1-MTG8-binding protein (MTGR1) is highly related to MTG8 and similar to Drosophila Nervy. Comparison of amino acid sequences among MTGR1, MTG8, and Nervy revealed four evolutionarily conserved regions (NHR1 to NHR4). Ectopic expression of AML1-MTG8 in L-G murine myeloid progenitor cells inhibits differentiation to mature neutrophils and induces cell proliferation in response to granulocyte colony-stimulating factor (G-CSF). Analysis with C-terminal deletion mutants of AML1-MTG8 indicated that the region of 51 residues (488 to 538), which contains NHR2, is essential for the induction of G-CSF-dependent cell proliferation. Immunoprecipitation analysis indicates that this region is required for AML1-MTG8 to form a stable complex with MTGR1. Overexpression of MTGR1 stimulates AML1-MTG8 to induce G-CSF-dependent proliferation of L-G cells and to interfere with AML1-dependent transcription. These results suggest that AML1-MTG8 could function as a complex with MTGR1 and that the complex might be important in promoting leukemogenesis.


Asunto(s)
Proteínas de Fusión Oncogénica , Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Línea Celular , Clonación Molecular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Sustancias Macromoleculares , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Fosfoproteínas/genética , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética
18.
Blood ; 90(12): 4699-704, 1997 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9389684

RESUMEN

p300, which was originally cloned as a nuclear binding target of the adenovirus E1A oncoprotein, forms a family with cyclic-AMP response element binding protein (CREB)-binding protein (CBP). p300/CBP are considered to be transcriptional coactivators that connect the basal transcriptional machinery to various DNA-binding transcriptional factors. p300/CBP are implicated in both cell differentiation and regulation of cell-cycle. We identify here that the p300 gene is fused to the MLL gene and that in-frame MLL-p300 fusion protein is generated in acute myeloid leukemia (AML) with t(11; 22)(q23; q13). These findings suggest that the basis for the leukemogenesis of t(11; 22)-AML is the inability of p300 to regulate cell-cycle and cell differentiation after fusion with MLL.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas Humanos Par 22 , Proteínas Nucleares/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proto-Oncogenes , Transactivadores , Factores de Transcripción/fisiología , Translocación Genética , Secuencia de Aminoácidos , Secuencia de Bases , Proteína de Unión a CREB , Preescolar , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina , Humanos , Masculino , Datos de Secuencia Molecular , Proteína de la Leucemia Mieloide-Linfoide
19.
EMBO J ; 14(14): 3496-509, 1995 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-7628451

RESUMEN

Transcription of the c-jun gene is up-regulated by either retinoic acid (RA) or adenovirus E1A during the differentiation of F9 cells. We show here that RA and E1A induce phosphorylation of the E1A-associated 300 kDa protein (p300) during the differentiation of F9 cells. The region of E1A that is required for interaction with cellular protein p300 overlaps with the region of E1A required for E1A to induce expression of the c-jun gene. Treatment of F9 cells with RA or infection of the cells by adenovirus led to a decrease in the electrophoretic mobility of p300. Phosphatase treatment of p300 from RA-treated or adenovirus-infected F9 cells reversed the changes in migration of p300, indicating that RA- and E1A-mediated changes in the mobility of p300 were due to phosphorylation. We also found factors, designated DRF1 and DRF2, that bound specifically to a sequence element that is necessary and sufficient for RA- and E1A-mediated up-regulation of the c-jun gene. The mobility of DRF complexes was changed by E1A or RA and the complexes were supershifted by addition of a polyclonal p300 antiserum. Moreover, overexpression of p300 resulted in an increase in the level of DRF1 complex. p300 fused to the DNA binding domain of the E2 protein of papilloma virus stimulated E2-dependent reporter activity in response to RA or E1A in F9 cells. Our results suggest that p300 is part of the DRF complexes, that it is differentially phosphorylated in undifferentiated versus differentiated cells and that it is likely involved in regulating transcription of the c-jun gene during F9 cell differentiation.


Asunto(s)
Proteínas E1A de Adenovirus/farmacología , Proteínas Nucleares/metabolismo , Transactivadores , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Anticuerpos Antivirales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Línea Celular , ADN , Proteína p300 Asociada a E1A , Regulación de la Expresión Génica , Genes jun , Datos de Secuencia Molecular , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/inmunología , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Especificidad de la Especie , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/inmunología , Transcripción Genética
20.
Biochim Biophys Acta ; 1266(2): 148-56, 1995 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-7742380

RESUMEN

The effects of the E1A protein of adenovirus-5 on the differentiation program of F9 teratocarcinoma cells were examined by the stable introduction of plasmids that expressed wild-type or mutated forms of E1A. Constitutive expression of plasmids for most of the mutant E1As induced loss of expression of the cell-surface antigen SSEA-1 and the enhanced expression of genes specific for the differentiated phenotype of F9 cells, such as genes for laminin B1, tissue-type plasminogen activator (tPA) and type IV collagen, as well as the altered cell morphology that is associated with the differentiated state. However, such changes were not observed in the case of genes for mutant proteins from which a conserved region (CR1) of E1A had been deleted. Furthermore, no significant induction of expression of the c-jun gene or transactivation of the c-jun-CAT reporter gene were observed when the sequence that encodes CR1 of E1A had been deleted. A palindromic sequence element (DRE) of the c-jun promoter was essential for the E1A-mediated up-regulation of the c-jun gene. These results imply that CR1 is required for activation of the c-jun gene and that it is implicated in the growth arrest, expression of parietal endoderm-specific functions and the orderly differentiation of F9 cells.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Secuencia Conservada , Proteínas E1A de Adenovirus/metabolismo , Secuencia de Aminoácidos , Diferenciación Celular/genética , Línea Celular , Células Clonales , Genes jun , Antígeno Lewis X/análisis , Mutación , Plásmidos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA