Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Leukemia ; 31(1): 159-169, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27389055

RESUMEN

Granulocyte macrophage-colony-stimulating factor (GM-CSF) signaling regulates hematopoiesis and immune responses. CSF2RA, the gene encoding the α-subunit for GM-CSF, is significantly downregulated in t(8;21) (RUNX1-ETO or RE) leukemia patients, suggesting that it may serve as a tumor suppressor. We previously reported that GM-CSF signaling is inhibitory to RE leukemogenesis. Here we conducted gene expression profiling of primary RE hematopoietic stem/progenitor cells (HSPCs) treated with GM-CSF to elucidate the mechanisms mediating the negative effects of GM on RE leukemogenicity. We observed that GM treatment of RE HSPCs resulted in a unique gene expression profile that resembles primary human cells undergoing myelopoiesis, which was not observed in control HSPCs. Additionally, we discovered that GM-CSF signaling attenuates MYC-associated gene signatures in RE HSPCs. In agreement with this, a functional screen of a subset of GM-CSF-responsive genes demonstrated that a MYC inhibitor, MXI1 (Max interactor 1), reduced the leukemic potential of RE HSPCs and t(8;21) acute myeloid leukemia (AML) cells. Furthermore, MYC knockdown and treatment with the BET (bromodomain and extra terminal domain) inhibitor JQ1 reduced the leukemic potential of t(8;21) cell lines. Altogether, we discovered a novel molecular mechanism mediating the GM-CSF-induced reduction in leukemic potential of RE cells, and our findings support MYC inhibition as an effective strategy for reducing the leukemogenicity of t(8;21) AML.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/etiología , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ratones , Mielopoyesis/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteína 1 Compañera de Translocación de RUNX1 , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/fisiología
2.
Leukemia ; 31(6): 1434-1442, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27885273

RESUMEN

Obesity is becoming more prevalent worldwide and is a major risk factor for cancer development. Acute myeloid leukemia (AML), the most common acute leukemia in adults, remains a frequently fatal disease. Here we investigated the molecular mechanisms by which obesity favors AML growth and uncovered the fatty acid-binding protein 4 (FABP4) and DNA methyltransferase 1 (DNMT1) regulatory axis that mediates aggressive AML in obesity. We showed that leukemia burden was much higher in high-fat diet-induced obese mice, which had higher levels of FABP4 and interleukin (IL)-6 in the sera. Upregulation of environmental and cellular FABP4 accelerated AML cell growth in both a cell-autonomous and cell-non-autonomous manner. Genetic disruption of FABP4 in AML cells or in mice blocked cell proliferation in vitro and induced leukemia regression in vivo. Mechanistic investigations showed that FABP4 upregulation increased IL-6 expression and signal transducer and activator of transcription factor 3 phosphorylation leading to DNMT1 overexpression and further silencing of the p15INK4B tumor-suppressor gene in AML cells. Conversely, FABP4 ablation reduced DNMT1-dependent DNA methylation and restored p15INK4B expression, thus conferring substantial protection against AML growth. Our findings reveal the FABP4/DNMT1 axis in the control of AML cell fate in obesity and suggest that interference with the FABP4/DNMT1 axis might be a new strategy to treat leukemia.


Asunto(s)
Metilación de ADN , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/patología , Obesidad/complicaciones , Animales , Apoptosis , Proliferación Celular , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Dieta Alta en Grasa/efectos adversos , Proteínas de Unión a Ácidos Grasos/genética , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/inducido químicamente , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Tumorales Cultivadas
4.
Leukemia ; 29(8): 1730-40, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25727291

RESUMEN

The mechanisms by which AML1/ETO (A/E) fusion protein induces leukemogenesis in acute myeloid leukemia (AML) without mutagenic events remain elusive. Here we show that interactions between A/E and hypoxia-inducible factor 1α (HIF1α) are sufficient to prime leukemia cells for subsequent aggressive growth. In agreement with this, HIF1α is highly expressed in A/E-positive AML patients and strongly predicts inferior outcomes, regardless of gene mutations. Co-expression of A/E and HIF1α in leukemia cells causes a higher cell proliferation rate in vitro and more serious leukemic status in mice. Mechanistically, A/E and HIF1α form a positive regulatory circuit and cooperate to transactivate DNMT3a gene leading to DNA hypermethylation. Pharmacological or genetic interventions in the A/E-HIF1α loop results in DNA hypomethylation, a re-expression of hypermethylated tumor-suppressor p15(INK4b) and the blockage of leukemia growth. Thus high HIF1α expression serves as a reliable marker, which identifies patients with a poor prognosis in an otherwise prognostically favorable AML group and represents an innovative therapeutic target in high-risk A/E-driven leukemia.


Asunto(s)
Transformación Celular Neoplásica/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Animales , Apoptosis , Western Blotting , Proliferación Celular , Inmunoprecipitación de Cromatina , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , ADN Metiltransferasa 3A , Citometría de Flujo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Técnicas para Inmunoenzimas , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Proteína 1 Compañera de Translocación de RUNX1 , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Leukemia ; 27(12): 2272-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23812420

RESUMEN

Oncogenic mutations in components of the JAK/STAT pathway, including those in cytokine receptors and JAKs, lead to increased activity of downstream signaling and are frequently found in leukemia and other hematological disorders. Thus, small-molecule inhibitors of this pathway have been the focus of targeted therapy in these hematological diseases. We previously showed that t(8;21) fusion protein acute myeloid leukemia (AML)1-ETO and its alternatively spliced variant AML1-ETO9a (AE9a) enhance the JAK/STAT pathway via downregulation of CD45, a negative regulator of this pathway. To investigate the therapeutic potential of targeting JAK/STAT in t(8;21) leukemia, we examined the effects of a JAK2-selective inhibitor TG101209 and a JAK1/2-selective inhibitor INCB18424 on t(8;21) leukemia cells. TG101209 and INCB18424 inhibited proliferation and promoted apoptosis of these cells. Furthermore, TG101209 treatment in AE9a leukemia mice reduced tumor burden and significantly prolonged survival. TG101209 also significantly impaired the leukemia-initiating potential of AE9a leukemia cells in secondary recipient mice. These results demonstrate the potential therapeutic efficacy of JAK inhibitors in treating t(8;21) AML.


Asunto(s)
Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Quinasas Janus/antagonistas & inhibidores , Leucemia/genética , Inhibidores de Proteínas Quinasas/farmacología , Translocación Genética , Animales , Apoptosis , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Citometría de Flujo , Humanos , Leucemia/patología , Leucemia/prevención & control , Ratones , Ratones Endogámicos C57BL
6.
Leukemia ; 23(4): 739-45, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19151769

RESUMEN

AML1/RUNX1 is a critical transcription factor in hematopoietic cell differentiation and proliferation. From the AML1 gene, at least three isoforms, AML1a, AML1b and AML1c, are produced through alternative splicing. AML1a interferes with the function of AML1b/1c, which are often called AML1. In this study, we found a higher expression level of AML1a in acute lymphoblastic leukemia and acute myeloid leukemia (AML)-M2 patients in comparison to the controls. Additionally, AML1a represses transcription of promoter of macrophage colony-stimulating factor receptor mediated by AML1b, indicating that AML1a antagonized the effect of AML1b. To investigate the role of AML1a in hematopoiesis and leukemogenesis in vivo, murine bone marrow mononuclear cells were transduced with AML1a and then transplanted into lethally irradiated mice, which developed lymphoblastic leukemia after transplantation. Taken together, these results indicate that overexpression of AML1a may be an important contributing factor to leukemogenesis.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación Neoplásica de la Expresión Génica , Leucemia/genética , Enfermedad Aguda , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Estudios de Casos y Controles , Hematopoyesis , Humanos , Leucemia/etiología , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/genética , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Transcripción Genética , Transducción Genética
7.
Leukemia ; 21(9): 2010-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17657222

RESUMEN

The 8;21 translocation is a common chromosomal abnormality in acute myeloid leukemia (AML). We recently identified a naturally occurring leukemogenic splice variant, AML1-ETO9a (acute myeloid leukemia-1 transcription factor and the eight-twenty-one corepressor-9a), of t(8;21). To understand the leukemic potential of AML1-ETO9a, we performed microarray analysis with the murine multipotential hematopoietic FDCP-mix A4 cell line. We identified changes in expression of various genes including CD44. CD44 is a type I transmembrane protein and functions as the major cellular adhesion molecule for hyaluronic acid, a component of the extracellular matrix. CD44 is expressed in most human cell types and is implicated in myeloid leukemia pathogenesis. We show that the presence of AML1-ETO9a significantly increased the expression of CD44 at both RNA and protein levels. Furthermore, the CD44 promoter is bound by AML1-ETO9a and AML1-ETO at the chromatin level. In addition, in the AML1-ETO9a leukemia mouse model CD44 is regulated in a cell context-dependent manner. Thus, our observations suggest that AML1-ETO and its splice variant AML1-ETO9a are able to regulate the expression of the CD44 gene, linking the 8;21 translocation to the regulation of a cell adhesion molecule that is involved in the growth and maintenance of the AML blast/stem cells.


Asunto(s)
Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Receptores de Hialuranos/genética , Leucemia Mieloide/genética , Translocación Genética , Enfermedad Aguda , Empalme Alternativo , Animales , Diferenciación Celular , División Celular , Supervivencia Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación Leucémica de la Expresión Génica , Humanos , Receptores de Hialuranos/metabolismo , Células K562 , Leucemia Mieloide/patología , Ratones , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1
8.
Proc Natl Acad Sci U S A ; 98(18): 10398-403, 2001 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-11526243

RESUMEN

The t(8;21) is one of the most frequent chromosomal abnormalities associated with acute myeloid leukemia (AML). The translocation, which involves the AML1 gene on chromosome 21 and the ETO gene on chromosome 8, generates an AML1-ETO fusion transcription factor. To examine the effect of the AML1-ETO fusion protein on leukemogenesis, we made transgenic mice in which expression of AML1-ETO is under the control of the human MRP8 promoter (hMRP8-AML1-ETO). AML1-ETO is specifically expressed in myeloid cells, including common myeloid progenitors of hMRP8-AML1-ETO transgenic mice. The transgenic mice were healthy during their life spans, suggesting that AML1-ETO alone is not sufficient for leukemogenesis. However, after treatment of newborn hMRP8-AML1-ETO transgenic mice and their wild-type littermates with a strong DNA-alkylating mutagen, N-ethyl-N-nitrosourea, 55% of transgenic mice developed AML and the other 45% of transgenic mice and all of the wild-type littermates developed acute T lymphoblastic leukemia. Our results provide direct evidence that AML1-ETO is critical for causing myeloid leukemia, but one or more additional mutations are required for leukemogenesis. The hMRP8-AML1-ETO-transgenic mice provide an excellent model that can be used to isolate additional genetic events and to further understand the molecular pathogenesis of AML1-ETO-related leukemia.


Asunto(s)
Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/genética , Mutación , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción/genética , Animales , Antígenos de Diferenciación/genética , Secuencia de Bases , Proteínas de Unión al Calcio/genética , Calgranulina A , Carcinógenos/toxicidad , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Cartilla de ADN/genética , Etilnitrosourea/toxicidad , Expresión Génica , Hematopoyesis/genética , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteína 1 Compañera de Translocación de RUNX1 , Translocación Genética
9.
Mol Cell Biol ; 21(16): 5577-90, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11463839

RESUMEN

The fusion gene AML1-ETO is the product of t(8;21)(q22;q22), one of the most common chromosomal translocations associated with acute myeloid leukemia. To investigate the impact of AML1-ETO on hematopoiesis, tetracycline-inducible AML1-ETO-expressing cell lines were generated using myeloid cells. AML1-ETO is tightly and strongly induced upon tetracycline withdrawal. The proliferation of AML1-ETO(+) cells was markedly reduced, and most of the cells eventually underwent apoptosis. RNase protection assays revealed that the amount of Bcl-2 mRNA was decreased after AML1-ETO induction. Enforced expression of Bcl-2 was able to significantly delay, but not completely overcome, AML1-ETO-induced apoptosis. Prior to the onset of apoptosis, we also studied the ability of AML1-ETO to modulate differentiation. AML1-ETO expression altered granulocytic differentiation of U937T-A/E cells. More significantly, this change of differentiation was associated with the down-regulation of CCAAT/enhancer binding protein alpha (C/EBPalpha), a key regulator of granulocytic differentiation. These observations suggest a dichotomy in the functions of AML1-ETO: (i) reduction of granulocytic differentiation correlated with decreased expression of C/EBPalpha and (ii) growth arrest leading to apoptosis with decreased expression of CDK4, c-myc, and Bcl-2. We predict that the preleukemic AML1-ETO(+) cells must overcome AML1-ETO-induced growth arrest and apoptosis prior to fulfilling their leukemogenic potential.


Asunto(s)
Apoptosis/fisiología , Hematopoyesis/fisiología , Proteínas de Fusión Oncogénica/fisiología , Factores de Transcripción/fisiología , Diferenciación Celular/genética , División Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Regulación de la Expresión Génica , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Leucemia Mieloide/fisiopatología , Proteína 1 Compañera de Translocación de RUNX1 , Translocación Genética , Células Tumorales Cultivadas
10.
Nat Med ; 7(4): 444-51, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11283671

RESUMEN

The transcription factor CCAAT/enhancer binding protein alpha, or C/EBPalpha, encoded by the CEBPA gene, is crucial for the differentiation of granulocytes. Conditional expression of C/EBPalpha triggers neutrophilic differentiation, and Cebpa knockout mice exhibit an early block in maturation. Dominant-negative mutations of CEBPA have been found in some patients with acute myeloid leukemia (AML), but not in AML with the t(8;21) translocation which gives rise to the fusion gene RUNX1-CBF2T1 (also known as AML1-ETO) encoding the AML1-ETO fusion protein. RUNX1-CBF2T1 positive-AML blasts had eight-fold lower CEBPA RNA levels and undetectable C/EBPalpha protein levels compared with other subgroups of AML patients. Conditional expression of RUNX1-CBF2T1 in U937 cells downregulated CEBPA mRNA, protein and DNA binding activity. AML1-ETO appears to suppress C/EBPalpha expression indirectly by inhibiting positive autoregulation of the CEBPA promoter. Conditional expression of C/EBPalpha in AML1-ETO-positive Kasumi-1 cells results in neutrophilic differentiation. We suggest that restoring C/EBPalpha expression will have therapeutic implications in RUNX1-CBF2T1-positive leukemias.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/genética , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/fisiología , Proteínas Proto-Oncogénicas , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Diferenciación Celular/genética , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Factores de Transcripción/genética , Translocación Genética
12.
Blood ; 97(7): 2168-70, 2001 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11264187

RESUMEN

AML1 is a transcription factor that is essential for normal hematopoietic development. It is the most frequent target for translocations in acute leukemia. Recently, fluorescence in situ hybridization was used to identify a novel syndrome of radiation-associated secondary acute myelogenous leukemia that had AML1 translocations. Using polymerase chain reaction, the AML1 fusion transcript was isolated from the patient who had a t(19;21) radiation-associated leukemia. The AML1 gene is fused out of frame to chromosome 19 sequences, resulting in a truncated AML protein bearing the DNA binding domain but not the transcriptional activation domain. This fusion AML1 protein functions as an inhibitor of the normal AML1 protein. (Blood. 2001;97:2168-2170)


Asunto(s)
Cromosomas Humanos Par 19/ultraestructura , Cromosomas Humanos Par 21/ultraestructura , Proteínas de Unión al ADN/genética , Leucemia Mieloide/genética , Leucemia Inducida por Radiación/genética , Proteínas de Neoplasias , Proteínas Proto-Oncogénicas , Proteínas Recombinantes de Fusión/genética , Factores de Transcripción/genética , Activación Transcripcional/genética , Translocación Genética/genética , Enfermedad Aguda , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Cromosomas Humanos Par 19/genética , Cromosomas Humanos Par 21/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal , ADN Complementario/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Humanos , Hibridación in Situ , Leucemia Mieloide/etiología , Datos de Secuencia Molecular , Proteínas de Fusión Oncogénica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacología , Eliminación de Secuencia , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química
13.
Blood ; 96(6): 2108-15, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10979955

RESUMEN

As reported previously, AML1-ETO knock-in mice were generated to investigate the role of AML1-ETO in leukemogenesis and to mimic the progression of t(8;21) leukemia. These knock-in mice died in midgestation because of hemorrhaging in the central nervous system and a block of definitive hematopoiesis during embryogenesis. Therefore, they are not a good model system for the development of acute myeloid leukemia. Therefore, mice were generated in which the expression of AML1-ETO is under the control of a tetracycline-inducible system. Multiple lines of transgenic mice have been produced with the AML1-ETO complementary DNA controlled by a tetracycline-responsive element. In the absence of the antibiotic tetracycline, AML1-ETO is strongly expressed in the bone marrow of AML1-ETO and tet-controlled transcriptional activator double-positive transgenic mice. Furthermore, the addition of tetracycline reduces AML1-ETO expression in double-positive mice to nondetectable levels. Throughout the normal murine lifespan of 24 months, mice expressing AML1-ETO have not developed leukemia. In spite of this, abnormal maturation and proliferation of progenitor cells have been observed from these animals. These results demonstrate that AML1-ETO has a very restricted capacity to transform cells. Either the introduction of additional genetic changes or the expression of AML1-ETO at a particular stage of hematopoietic cell differentiation will be necessary to develop a model for studying the pathogenesis of t(8;21).


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide/etiología , Leucemia Mieloide/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas , Factores de Transcripción/genética , Enfermedad Aguda , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Ratones , Ratones Transgénicos , Proteína 1 Compañera de Translocación de RUNX1
14.
Genomics ; 65(1): 44-52, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10777664

RESUMEN

The ubiquitin-specific proteases (UBP) are a family of enzymes that cleave ubiquitin from ubiquitinated protein substrates. We have recently cloned UBP43, a novel member of this family from AML1-ETO knock-in mice. To analyze the role of UBP43 in hematopoiesis and leukemogenesis, we have cloned a full-length human UBP43 cDNA by screening a human monocytic cDNA library as well as by 5'- and 3'-rapid amplification of cDNA ends analyses. This cDNA encodes a polypeptide of 372 amino acids with all of the structural motifs of a deubiquitinating enzyme. The human UBP43 mRNA is strongly expressed in human liver and thymus. Transfection analysis has demonstrated that UBP43 is a nuclear protein. Interestingly, the gene encoding human UBP43 maps to chromosome 22q11.2. This region, known as DiGeorge syndrome critical region, contains a minimal area of 2 Mb and is consistently deleted in DiGeorge syndrome and related disorders. The syndrome is marked by thymic aplasia or hypoplasia, parathyroid hypoplasia, or congenital cardiac abnormalities. Taken together, our results broaden the understanding of a new human ubiquitin-specific protease, UBP43, and suggest that this gene may also be related to DiGeorge syndrome.


Asunto(s)
Endopeptidasas/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Cromosomas Humanos Par 22/genética , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Endopeptidasas/metabolismo , Exones , Expresión Génica , Genes/genética , Células HL-60 , Células HeLa , Humanos , Hibridación Fluorescente in Situ , Intrones , Células Jurkat , Células K562 , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Distribución Tisular , Células Tumorales Cultivadas , Ubiquitina Tiolesterasa , Proteasas Ubiquitina-Específicas , Ubiquitinas/genética , Ubiquitinas/metabolismo , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
15.
J Biol Chem ; 274(35): 24671-6, 1999 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-10455134

RESUMEN

AML1 plays a critical role during hematopoiesis and chromosomal translocations involving AML1 are commonly associated with different forms of leukemia, including pre-B acute lymphoblastic leukemia. To understand the function of AML1 during B cell differentiation, we analyzed regulatory regions of B cell-specific genes for potential AML1-binding sites and have identified a putative AML1-binding site in the promoter of the B cell-specific tyrosine kinase gene, blk. Gel mobility shift assays and transient transfection assays demonstrate that AML1 binds specifically to this site in the blk promoter and this binding site is important for blk promoter activity. Furthermore, in vitro binding analysis revealed that the AML1 runt DNA-binding domain physically interacts with the paired DNA-binding domain of BSAP, a B cell-specific transcription factor. BSAP has been shown previously to be important for B cell-specific regulation of the blk gene. Physical interaction of AML1 with BSAP correlates with functional cooperativity in transfection studies where AML1 and BSAP synergistically activate blk promoter transcription by more than 50-fold. These results demonstrate physical and functional interactions between AML1 and BSAP and suggest that AML1 is an important factor for regulating a critical B cell-specific gene, blk.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Linfocitos B/metabolismo , Proteínas Portadoras/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Mitocondriales , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas , Factores de Transcripción/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Secuencia de Bases , Sitios de Unión , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Ratones , Datos de Secuencia Molecular , Mutación , Factor de Transcripción PAX5 , Regiones Promotoras Genéticas , Unión Proteica , Activación Transcripcional
16.
Mol Cell Biol ; 19(4): 3029-38, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10082570

RESUMEN

Using PCR-coupled subtractive screening-representational difference analysis, we have cloned a novel gene from AML1-ETO knockin mice. This gene is highly expressed in the yolk sac and fetal liver of the knockin mice. Nucleotide sequence analysis indicates that its cDNA contains an 1,107-bp open reading frame encoding a 368-amino-acid polypeptide. Further protein sequence and protein translation analysis shows that it belongs to a family of ubiquitin-specific proteases (UBP), and its molecular mass is 43 kDa. Therefore, we have named this gene UBP43. Like other ubiquitin proteases, the UBP43 protein has deubiquitinating enzyme activity. Protein ubiquitination has been implicated in many important cellular events. In wild-type adult mice, UBP43 is highly expressed in the thymus and in peritoneal macrophages. Among nine different murine hematopoietic cell lines analyzed, UBP43 expression is detectable only in cell lines related to the monocytic lineage. Furthermore, its expression is regulated during cytokine-induced monocytic cell differentiation. We have investigated its function in the hematopoietic myeloid cell line M1. UBP43 was introduced into M1 cells by retroviral gene transfer, and several high-expressing UBP43 clones were obtained for further study. Morphologic and cell surface marker examination of UBP43/M1 cells reveals that overexpression of UBP43 blocks cytokine-induced terminal differentiation of monocytic cells. These data suggest that UBP43 plays an important role in hematopoiesis by modulating either the ubiquitin-dependent proteolytic pathway or the ubiquitination state of another regulatory factor(s) during myeloid cell differentiation.


Asunto(s)
Proteínas de Unión al ADN , Endopeptidasas/genética , Hematopoyesis/genética , Leucemia Experimental/etiología , Proteínas de Fusión Oncogénica , Proteínas Proto-Oncogénicas , Factores de Transcripción/genética , Ubiquitinas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular , Clonación Molecular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Endopeptidasas/metabolismo , Células Madre Hematopoyéticas/citología , Leucemia Experimental/genética , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/genética , Macrófagos/citología , Ratones , Ratones Noqueados , Ratones Mutantes , Datos de Secuencia Molecular , Monocitos/citología , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Recombinantes de Fusión/genética , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Ubiquitina Tiolesterasa
17.
Methods ; 17(3): 231-7, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10080908

RESUMEN

Acute myeloid leukemia (AML) is still associated with a mortality of 60 to 80%. AML is characterized by a block in myeloid differentiation. The transcription factors PU.1 and C/EBPalpha are responsible for normal myeloid differentiation from stem cells to monocytes or granulocytes. In particular, PU.1 induces expression of the macrophage colony-stimulating factor (M-CSF) receptor and the development of monocytes, whereas C/EBPalpha increases the expression of the granulocyte colony-stimulating factor (G-CSF) receptor and leads to mature granulocytes. In AML, chromosomal aberrations result in oncoproteins such as AML1/ETO, PML/RARalpha, or activated Ras, which can deregulate genes important for normal myelopoiesis. Thus, AML1/ETO can bind to the transcription factor C/EBPalpha, inhibit C/EBPalpha-dependent transcription, and block granulocytic differentiation. However, AML1/ETO can also synergize with the transcription factor AML1 to enhance the activity of the M-CSF receptor promoter. On the other hand, the PML/RARalpha fusion protein causes transcriptional repression by recruiting the nuclear corepressor (N-CoR) histone deacetylase complex to the DNA, which results in decreased histone acetylation and a repressive chromatin organization. Here we describe methods to investigate whether and how signaling agonists induce myeloid differentiation and how oncoproteins might cause AML by modulating the activity of transcription factors that are pivotal for normal myeloid development.


Asunto(s)
Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucopoyesis/genética , Proteínas Oncogénicas/análisis , Factores de Transcripción/metabolismo , Enfermedad Aguda , Bioquímica/métodos , Western Blotting , Diferenciación Celular , Humanos , Monocitos/metabolismo , Neutrófilos/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Mapeo Peptídico/métodos , Transducción de Señal , Factores de Transcripción/análisis , Factores de Transcripción/genética , Transfección/métodos
18.
J Biol Chem ; 274(8): 4939-46, 1999 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-9988737

RESUMEN

The ETS domain transcription factor PU.1 is necessary for the development of monocytes and regulates, in particular, the expression of the monocyte-specific macrophage colony-stimulating factor (M-CSF) receptor, which is critical for monocytic cell survival, proliferation, and differentiation. The bZIP transcription factor c-Jun, which is part of the AP-1 transcription factor complex, is also important for monocytic differentiation, but the monocyte-specific M-CSF receptor promoter has no AP-1 consensus binding sites. We asked the question of whether c-Jun could promote the induction of the M-CSF receptor by collaborating with PU.1. We demonstrate that c-Jun enhances the ability of PU.1 to transactivate the M-CSF receptor promoter as well as a minimal thymidine kinase promoter containing only PU.1 DNA binding sites. c-Jun does not directly bind to the M-CSF receptor promoter but associates via its basic domain with the ETS domain of PU.1. Consistent with our observation that AP-1 binding does not contribute to c-Jun coactivation is the observation that the activation of PU.1 by c-Jun is blocked by overexpression of c-Fos. Phosphorylation of c-Jun by c-Jun NH2-terminal kinase on Ser-63 and -73 does not alter the ability of c-Jun to enhance PU.1 transactivation. Activated Ras enhances the transcriptional activity of PU.1 by up-regulating c-Jun expression without changing the phosphorylation pattern of PU.1. The activation of PU.1 by Ras is blocked by a mutant c-Jun protein lacking the basic domain. The expression of this mutant form of c-Jun also completely blocks 12-O-tetradecanoylphorbol-13-acetate-induced M-CSF receptor promoter activity during monocytic differentiation. We propose therefore that c-Jun acts as a c-Jun NH2-terminal kinase-independent coactivator of PU.1, resulting in M-CSF receptor expression and development of the monocytic lineage.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Línea Celular , ADN/metabolismo , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Haplorrinos , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Monocitos/citología , Monocitos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Acetato de Tetradecanoilforbol/farmacología , Timidina Quinasa/genética , Activación Transcripcional , Células Tumorales Cultivadas , Regulación hacia Arriba
19.
J Immunol ; 162(1): 503-9, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9886426

RESUMEN

CD14 is a major receptor for the bacterial endotoxin LPS. Since CD14 is specifically and highly expressed on the surface of monocytic cells, it has been used as a monocyte/macrophage differentiation marker. To identify elements that are critical for the direction of the tissue-specific expression of CD14, an 80-kb genomic DNA fragment containing the coding region of the CD14 gene, as well as a considerable amount of both upstream and downstream sequence, was used to generate transgenic mice. The analysis of mice from six different founder lines demonstrated that this genomic DNA fragment was sufficient to direct human CD14 gene expression in a monocyte-specific manner among hematopoietic cells. Furthermore, the data lead us to a new finding that CD14 is highly expressed in the human liver, a primary organ involved in the acute phase response. These transgenic mice provide a useful model to analyze the biological function of human CD14.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Receptores de Lipopolisacáridos/biosíntesis , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/metabolismo , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Animales , Clonación Molecular , Exones , Humanos , Intrones , Receptores de Lipopolisacáridos/química , Hígado/inmunología , Hígado/metabolismo , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Receptores Inmunológicos/química , Secuencias Reguladoras de Ácidos Nucleicos/inmunología , Transgenes/inmunología
20.
Mol Cell Biol ; 18(7): 3915-25, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9632776

RESUMEN

Control elements of many genes are regulated by multiple activators working in concert to confer the maximal level of expression, but the mechanism of such synergy is not completely understood. The promoter of the human macrophage colony-stimulating factor (M-CSF) receptor presents an excellent model with which we can study synergistic, tissue-specific activation for two reasons. First, myeloid-specific expression of the M-CSF receptor is regulated transcriptionally by three factors which are crucial for normal hematopoiesis: PU.1, AML1, and C/EBPalpha. Second, these proteins interact in such a way as to demonstrate at least two examples of synergistic activation. We have shown that AML1 and C/EBPalpha activate the M-CSF receptor promoter in a synergistic manner. As we report here, AML1 also synergizes, and interacts physically, with PU. 1. Detailed analysis of the physical and functional interaction of AML1 with PU.1 and C/EBPalpha has revealed that the proteins contact one another through their DNA-binding domains and that AML1 exhibits cooperative DNA binding with C/EBPalpha but not with PU.1. This difference in DNA-binding abilities may explain, in part, the differences observed in synergistic activation. Furthermore, the activation domains of all three factors are required for synergistic activation, and the region of AML1 required for synergy with PU.1 is distinct from that required for synergy with C/EBPalpha. These observations present the possibility that synergistic activation is mediated by secondary proteins contacted through the activation domains of AML1, C/EBPalpha, and PU.1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT , Células COS , Línea Celular , Chlorocebus aethiops , Subunidad alfa 2 del Factor de Unión al Sitio Principal , ADN/metabolismo , Células HeLa , Humanos , Mutagénesis , Fosforilación , Regiones Promotoras Genéticas , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA