Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(14): 5594-9, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23509296

RESUMEN

MicroRNA-9 (miR-9) is emerging as a critical regulator of organ development and neurogenesis. It is also deregulated in several types of solid tumors; however, its role in hematopoiesis and leukemogenesis is not yet known. Here we show that miR-9 is detected in hematopoietic stem cells and hematopoietic progenitor cells, and that its expression increases during hematopoietic differentiation. Ectopic expression of miR-9 strongly accelerates terminal myelopoiesis and promotes apoptosis in vitro and in vivo. Conversely, in hematopoietic progenitor cells, the inhibition of miR-9 with a miRNA sponge blocks myelopoiesis. Ecotropic viral integration site 1 (EVI1), required for normal embryogenesis, is considered an oncogene because its inappropriate up-regulation induces malignant transformation in solid and hematopoietic cancers. Here we show that EVI1 binds to the promoter of miR-9-3, leading to DNA hypermethylation of the promoter and repression of miR-9. Moreover, miR-9 expression reverses a myeloid differentiation block that is induced by EVI1. Our findings indicate that EVI1, when inappropriately expressed, delays or blocks myeloid differentiation at least in part by DNA hypermethylation and down-regulation of miR-9. It was reported that Forkhead box class O genes (FoxOs) inhibit myeloid differentiation and prevent differentiation of leukemia-initiating cells. Here we identify both FoxO1 and FoxO3 as direct targets of miR-9 in hematopoietic cells and find that up-regulation of FoxO3 inhibits miR-9-induced myelopoiesis. These results reveal a unique role of miR-9 in myelopoiesis and in the pathogenesis of EVI1-induced myeloid neoplasms and provide insights into the epigenetic regulation of miR9 in tumorigenesis.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/genética , Células Madre Hematopoyéticas/metabolismo , MicroARNs/metabolismo , Mielopoyesis/fisiología , Factores de Transcripción/metabolismo , Animales , Inmunoprecipitación de Cromatina , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN/genética , Citometría de Flujo , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Humanos , Proteína del Locus del Complejo MDS1 y EV11 , Ratones , Mielopoyesis/genética , Células 3T3 NIH , Proto-Oncogenes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
2.
Proc Natl Acad Sci U S A ; 110(29): 11994-9, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818607

RESUMEN

The ten-eleven translocation 1 (TET1) gene is the founding member of the TET family of enzymes (TET1/2/3) that convert 5-methylcytosine to 5-hydroxymethylcytosine. Although TET1 was first identified as a fusion partner of the mixed lineage leukemia (MLL) gene in acute myeloid leukemia carrying t(10,11), its definitive role in leukemia is unclear. In contrast to the frequent down-regulation (or loss-of-function mutations) and critical tumor-suppressor roles of the three TET genes observed in various types of cancers, here we show that TET1 is a direct target of MLL-fusion proteins and is significantly up-regulated in MLL-rearranged leukemia, leading to a global increase of 5-hydroxymethylcytosine level. Furthermore, our both in vitro and in vivo functional studies demonstrate that Tet1 plays an indispensable oncogenic role in the development of MLL-rearranged leukemia, through coordination with MLL-fusion proteins in regulating their critical cotargets, including homeobox A9 (Hoxa9)/myeloid ecotropic viral integration 1 (Meis1)/pre-B-cell leukemia homeobox 3 (Pbx3) genes. Collectively, our data delineate an MLL-fusion/Tet1/Hoxa9/Meis1/Pbx3 signaling axis in MLL-rearranged leukemia and highlight TET1 as a potential therapeutic target in treating this presently therapy-resistant disease.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Leucemia Mieloide Aguda/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , 5-Metilcitosina/análogos & derivados , Inmunoprecipitación de Cromatina , Cromatografía Liquida , Citosina/análogos & derivados , Citosina/metabolismo , Perfilación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/metabolismo , Humanos , Immunoblotting , Análisis por Micromatrices , Oxigenasas de Función Mixta , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/genética , Espectrometría de Masas en Tándem
3.
Proc Natl Acad Sci U S A ; 110(28): 11511-6, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23798388

RESUMEN

MicroRNAs (miRNAs), small noncoding RNAs that regulate target gene mRNAs, are known to contribute to pathogenesis of cancers. Acute myeloid leukemia (AML) is a group of heterogeneous hematopoietic malignancies with various chromosomal and/or molecular abnormalities. AML with chromosomal translocations involving the mixed lineage leukemia (MLL) gene are usually associated with poor survival. In the present study, through a large-scale, genomewide miRNA expression assay, we show that microRNA-9 (miR-9) is the most specifically up-regulated miRNA in MLL-rearranged AML compared with both normal control and non-MLL-rearranged AML. We demonstrate that miR-9 is a direct target of MLL fusion proteins and can be significantly up-regulated in expression by the latter in human and mouse hematopoietic stem/progenitor cells. Depletion of endogenous miR-9 expression by an appropriate antagomiR can significantly inhibit cell growth/viability and promote apoptosis in human MLL-rearranged AML cells, and the opposite is true when expression of miR-9 is forced. Blocking endogenous miR-9 function by anti-miRNA sponge can significantly inhibit, whereas forced expression of miR-9 can significantly promote, MLL fusion-induced immortalization/transformation of normal mouse bone marrow progenitor cells in vitro. Furthermore, forced expression of miR-9 can significantly promote MLL fusion-mediated leukemogenesis in vivo. In addition, a group of putative target genes of miR-9 exhibited a significant inverse correlation of expression with miR-9 in a series of leukemia sample sets, suggesting that they are potential targets of miR-9 in MLL-rearranged AML. Collectively, our data demonstrate that miR-9 is a critical oncomiR in MLL-rearranged AML and can serve as a potential therapeutic target to treat this dismal disease.


Asunto(s)
Leucemia Mieloide Aguda/genética , MicroARNs/fisiología , Proteína de la Leucemia Mieloide-Linfoide/genética , Apoptosis/genética , Supervivencia Celular/genética , Proteínas de Unión al ADN/fisiología , Humanos , Leucemia Mieloide Aguda/patología , Proteína del Locus del Complejo MDS1 y EV11 , MicroARNs/genética , Proto-Oncogenes/fisiología , Factores de Transcripción/fisiología
4.
Blood ; 121(8): 1422-31, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23264595

RESUMEN

Although PBX proteins are known to increase DNA-binding/transcriptional activity of HOX proteins through their direct binding, the functional importance of their interaction in leukemogenesis is unclear.We recently reported that overexpression of a 4-homeobox-gene signature (ie, PBX3/HOXA7/HOXA9/HOXA11) is an independent predictor of poor survival in patients with cytogenetically abnormal acute myeloid leukemia (CA-AML). Here we show that it is PBX3, but not PBX1 or PBX2, that is consistently coexpressed with HOXA9 in various subtypes of CA-AML, particularly MLL-rearranged AML, and thus appears as a potential pathologic cofactor of HOXA9 in CA-AML. We then show that depletion of endogenous Pbx3 expression by shRNA significantly inhibits MLL-fusion-mediated cell transformation, and coexpressed PBX3 exhibits a significantly synergistic effect with HOXA9 in promoting cell transformation in vitro and leukemogenesis in vivo. Furthermore, as a proof of concept, we show that a small peptide, namely HXR9, which was developed to specifically disrupt the interactions between HOX and PBX proteins, can selectively kill leukemic cells with overexpression of HOXA/PBX3 genes. Collectively, our data suggest that PBX3 is a critical cofactor of HOXA9 in leukemogenesis, and targeting their interaction is a feasible strategy to treat presently therapy resistant CA-AML (eg, MLL-rearranged leukemia) in which HOXA/PBX3 genes are overexpressed.


Asunto(s)
Regulación Leucémica de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Animales , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Línea Celular Transformada , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Reordenamiento Génico/genética , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/antagonistas & inhibidores , Humanos , Péptidos y Proteínas de Señalización Intercelular , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteína de la Leucemia Mieloide-Linfoide/genética , Péptidos/farmacología , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Ratas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Proc Natl Acad Sci U S A ; 109(47): 19397-402, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23132946

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous group of hematopoietic malignancies with variable response to treatment. AMLs bearing MLL (mixed lineage leukemia) rearrangements are associated with intermediate or poor survival. MicroRNAs (miRNAs), a class of small noncoding RNAs, have been postulated to be important gene expression regulators virtually in all biological processes, including leukemogenesis. Through a large-scale, genome-wide miRNA expression profiling assay of 85 human AML and 15 normal control samples, we show that among 48 miRNAs that are significantly differentially expressed between MLL- and non-MLL-rearranged AML samples, only one (miR-495) is expressed at a lower level in MLL-rearranged AML than in non-MLL-rearranged AML; meanwhile, miR-495 is also significantly down-regulated in MLL-rearranged AML samples compared with normal control samples. Through in vitro colony-forming/replating assays and in vivo bone marrow transplantation studies, we show that forced expression of miR-495 significantly inhibits MLL-fusion-mediated cell transformation in vitro and leukemogenesis in vivo. In human leukemic cells carrying MLL rearrangements, ectopic expression of miR-495 greatly inhibits cell viability and increases cell apoptosis. Furthermore, our studies demonstrate that PBX3 and MEIS1 are two direct target genes of miR-495, and forced expression of either of them can reverse the effects of miR-495 overexpression on inhibiting cell viability and promoting apoptosis of human MLL-rearranged leukemic cells. Thus, our data indicate that miR-495 likely functions as a tumor suppressor in AML with MLL rearrangements by targeting essential leukemia-related genes.


Asunto(s)
Regulación hacia Abajo/genética , Reordenamiento Génico/genética , Leucemia Mieloide Aguda/genética , MicroARNs/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Animales , Secuencia de Bases , Estudios de Casos y Controles , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Estudios de Asociación Genética , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia Mieloide Aguda/patología , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
6.
Blood ; 119(10): 2314-24, 2012 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-22251480

RESUMEN

Increased expression levels of miR-181 family members have been shown to be associated with favorable outcome in patients with cytogenetically normal acute myeloid leukemia. Here we show that increased expression of miR-181a and miR-181b is also significantly (P < .05; Cox regression) associated with favorable overall survival in cytogenetically abnormal AML (CA-AML) patients. We further show that up-regulation of a gene signature composed of 4 potential miR-181 targets (including HOXA7, HOXA9, HOXA11, and PBX3), associated with down-regulation of miR-181 family members, is an independent predictor of adverse overall survival on multivariable testing in analysis of 183 CA-AML patients. The independent prognostic impact of this 4-homeobox-gene signature was confirmed in a validation set of 271 CA-AML patients. Furthermore, our in vitro and in vivo studies indicated that ectopic expression of miR-181b significantly promoted apoptosis and inhibited viability/proliferation of leukemic cells and delayed leukemogenesis; such effects could be reversed by forced expression of PBX3. Thus, the up-regulation of the 4 homeobox genes resulting from the down-regulation of miR-181 family members probably contribute to the poor prognosis of patients with nonfavorable CA-AML. Restoring expression of miR-181b and/or targeting the HOXA/PBX3 pathways may provide new strategies to improve survival substantially.


Asunto(s)
Proteínas de Homeodominio/genética , Leucemia Mieloide/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas/genética , Enfermedad Aguda , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Humanos , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Leucemia Mieloide/patología , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Regulación hacia Arriba , Adulto Joven
7.
Nat Med ; 12(8): 945-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16892037

RESUMEN

The t(8;21)(q22;q22) translocation is one of the most common genetic abnormalities in acute myeloid leukemia (AML), identified in 15% of all cases of AML, including 40-50% of FAB M2 subtype and rare cases of M0, M1 and M4 subtypes. The most commonly known AML1-ETO fusion protein (full-length AML1-ETO) from this translocation has 752 amino acids and contains the N-terminal portion of RUNX1 (also known as AML1, CBFalpha2 or PEBP2alphaB), including its DNA binding domain, and almost the entire RUNX1T1 (also known as MTG8 or ETO) protein. Although alterations of gene expression and hematopoietic cell proliferation have been reported in the presence of AML1-ETO, its expression does not lead to the development of leukemia. Here, we report the identification of a previously unknown alternatively spliced isoform of the AML1-ETO transcript, AML1-ETO9a, that includes an extra exon, exon 9a, of the ETO gene. AML1-ETO9a encodes a C-terminally truncated AML1-ETO protein of 575 amino acids. Expression of AML1-ETO9a leads to rapid development of leukemia in a mouse retroviral transduction-transplantation model. More importantly, coexpression of AML1-ETO and AML1-ETO9a results in the substantially earlier onset of AML and blocks myeloid cell differentiation at a more immature stage. These results indicate that fusion proteins from alternatively spliced isoforms of a chromosomal translocation may work together to induce cancer development.


Asunto(s)
Empalme Alternativo , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/genética , Translocación Genética , Secuencia de Aminoácidos , Animales , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/análisis , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Exones , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Proteínas de Fusión Oncogénica/análisis , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteína 1 Compañera de Translocación de RUNX1 , Retroviridae/genética
8.
Proc Natl Acad Sci U S A ; 107(8): 3710-5, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20133587

RESUMEN

MicroRNA (miRNA)-17-92 cluster (miR-17-92), containing seven individual miRNAs, is frequently amplified and overexpressed in lymphomas and various solid tumors. We have found that it is also frequently amplified and the miRNAs are aberrantly overexpressed in mixed lineage leukemia (MLL)-rearranged acute leukemias. Furthermore, we show that MLL fusions exhibit a much stronger direct binding to the locus of this miRNA cluster than does wild-type MLL; these changes are associated with elevated levels of histone H3 acetylation and H3K4 trimethylation and an up-regulation of these miRNAs. We further observe that forced expression of this miRNA cluster increases proliferation and inhibits apoptosis of human cells. More importantly, we show that this miRNA cluster can significantly increase colony-forming capacity of normal mouse bone marrow progenitor cells alone and, particularly, in cooperation with MLL fusions. Finally, through combinatorial analysis of miRNA and mRNA arrays of mouse bone marrow progenitor cells transfected with this miRNA cluster and/or MLL fusion gene, we identified 363 potential miR-17-92 target genes that exhibited a significant inverse correlation of expression with the miRNAs. Remarkably, these potential target genes are significantly enriched (P < 0.01; >2-fold) in cell differentiation, hematopoiesis, cell cycle, and apoptosis. Taken together, our studies suggest that overexpression of miR-17-92 cluster in MLL-rearranged leukemias is likely attributed to both DNA copy number amplification and direct up-regulation by MLL fusions, and that the miRNAs in this cluster may play an essential role in the development of MLL-associated leukemias through inhibiting cell differentiation and apoptosis, while promoting cell proliferation, by regulating relevant target genes.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , MicroARNs/biosíntesis , Animales , Línea Celular Tumoral , Epigénesis Genética , Células HeLa , Humanos , Ratones , MicroARNs/genética , Familia de Multigenes
9.
Artículo en Inglés | MEDLINE | ID: mdl-19715438

RESUMEN

Although it was definitely not obvious at first, consistent chromosomal translocations are major contributors to cellular transformation in some leukemias, lymphomas, sarcomas, prostate cancer, and other benign and malignant neoplasms. In the 50 years since the discovery of the Ph chromosome, the elucidation of recurring abnormalities has been an ongoing challenge that has evolved as new technologies allowed an ever more accurate definition of the precise changes in DNA resulting from these abnormalities. As we enter a new era of understanding enriched by gene expression studies, we still know little about the changes in the level of critical proteins, which may be the ultimate effectors of the genetic/epigenetic abnormalities in cancer. Despite remarkable progress in identifying both obvious chromosome abnormalities and subtle changes in DNA such as mutations and small copy-number variations, the impact of this knowledge has been variable. The challenge for the future is to enhance our ability to translate these genetic changes into effective therapies for other malignant diseases.


Asunto(s)
Cromosomas Humanos , Leucemia/genética , Citogenética/historia , ADN/genética , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Leucemia/historia , Estados Unidos
10.
Blood Cells Mol Dis ; 49(2): 102-6, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22633751

RESUMEN

HOXA9 plays a critical role in both normal hematopoiesis and leukemogenesis, particularly in the development and maintenance of mixed lineage leukemia (MLL)-rearranged leukemia. Through reverse transcription-polymerase chain reaction (RT-PCR) analysis of HOXA9 transcripts in human leukemia and normal bone marrow samples, we identified a truncated isoform of HOXA9, namely HOXA9T, and found that both HOXA9T and canonical HOXA9 were highly expressed in leukemia cell lines bearing MLL rearrangements, relative to human normal bone marrow cells or other subtypes of leukemia cells. A frameshift in HOXA9T in exon I causes a premature stop codon upstream of the PBX-binding domain and the homeodomain, which leads to the generation of a non-homeodomain-containing protein. Unlike the canonical HOXA9, HOXA9T alone cannot transform normal bone marrow progenitor cells. Moreover, HOXA9T cannot cooperate with MEIS1 to transform cells, despite the presence of a MEIS1-binding domain. Remarkably, although the truncated isoforms of many proteins function as dominant-negative competitors or inhibitors of their full-length counterparts, this is not the case for HOXA9T; instead, HOXA9T synergized with HOXA9 in transforming mouse normal bone marrow progenitor cells through promoting self-renewal and proliferation of the cells. Collectively, our data indicate that both truncated and full-length forms of HOXA9 are highly expressed in human MLL-rearranged leukemia, and the truncated isoform of HOXA9 might also play an oncogenic role by cooperating with canonical HOXA9 in cell transformation and leukemogenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Leucemia/genética , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , Animales , Secuencia de Bases , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Codón sin Sentido , Exones , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patología , Ratones , Datos de Secuencia Molecular , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(20): 8278-83, 2009 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-19416867

RESUMEN

Studying gene expression at different hematopoietic stages provides insights for understanding the genetic basis of hematopoiesis. We analyzed gene expression in human CD34(+) hematopoietic cells that represent the stem-progenitor population (CD34(+) cells). We collected >459,000 transcript signatures from CD34(+) cells, including the de novo-generated 3' ESTs and the existing sequences of full-length cDNAs, ESTs, and serial analysis of gene expression (SAGE) tags, and performed an extensive annotation on this large set of CD34(+) transcript sequences. We determined the genes expressed in CD34(+) cells, verified the known genes and identified the new genes of different functional categories involved in hematopoiesis, dissected the alternative gene expression including alternative transcription initiation, splicing, and adenylation, identified the antisense and noncoding transcripts, determined the CD34(+) cell-specific gene expression signature, and developed the CD34(+) cell-transcription map in the human genome. Our study provides a current view on gene expression in human CD34(+) cells and reveals that early hematopoiesis is an orchestrated process with the involvement of over half of the human genes distributed in various functions. The data generated from our study provide a comprehensive and uniform resource for studying hematopoiesis and stem cell biology.


Asunto(s)
Antígenos CD34 , Perfilación de la Expresión Génica , Genes/fisiología , Células Madre Hematopoyéticas , Secuencia de Bases , ADN Complementario , Etiquetas de Secuencia Expresada , Marcadores Genéticos , Hematopoyesis/genética , Humanos , Datos de Secuencia Molecular
12.
J Exp Med ; 201(11): 1837-52, 2005 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-15939795

RESUMEN

Pre-B cells undergo apoptosis unless they are rescued by pre-B cell receptor-dependent survival signals. We previously showed that the BCR-ABL1 kinase that is expressed in pre-B lymphoblastic leukemia bypasses selection for pre-B cell receptor-dependent survival signals. Investigating possible interference of BCR-ABL1 with pre-B cell receptor signaling, we found that neither SYK nor SLP65 can be phosphorylated in response to pre-B cell receptor engagement. Instead, Bruton's tyrosine kinase (BTK) is constitutively phosphorylated by BCR-ABL1. Activated BTK is essential for survival signals that otherwise would arise from the pre-B cell receptor, including activation of PLCgamma1, autonomous Ca2+ signaling, STAT5-phosphorylation, and up-regulation of BCLX(L). Inhibition of BTK activity specifically induces apoptosis in BCR-ABL1+ leukemia cells to a similar extent as inhibition of BCR-ABL1 kinase activity itself. However, BCR-ABL1 cannot directly bind to full-length BTK. Instead, BCR-ABL1 induces the expression of a truncated splice variant of BTK that acts as a linker between the two kinases. As opposed to full-length BTK, truncated BTK lacks kinase activity yet can bind to BCR-ABL1 through its SRC-homology domain 3. Acting as a linker, truncated BTK enables BCR-ABL1-dependent activation of full-length BTK, which initiates downstream survival signals and mimics a constitutively active pre-B cell receptor.


Asunto(s)
Señalización del Calcio , Regulación Leucémica de la Expresión Génica , Glicoproteínas de Membrana/metabolismo , Imitación Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Adolescente , Adulto , Anciano , Línea Celular Tumoral , Supervivencia Celular , Niño , Preescolar , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Receptores de Células Precursoras de Linfocitos B , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos B
13.
Blood ; 113(14): 3314-22, 2009 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-19188669

RESUMEN

Chromosomal translocations involving the Mixed Lineage Leukemia (MLL) gene produce chimeric proteins that cause abnormal expression of a subset of HOX genes and leukemia development. Here, we show that MLL normally regulates expression of mir-196b, a hematopoietic microRNA located within the HoxA cluster, in a pattern similar to that of the surrounding 5' Hox genes, Hoxa9 and Hoxa10, during embryonic stem (ES) cell differentiation. Within the hematopoietic lineage, mir-196b is most abundant in short-term hematopoietic stem cells and is down-regulated in more differentiated hematopoietic cells. Leukemogenic MLL fusion proteins cause overexpression of mir-196b, while treatment of MLL-AF9 transformed bone marrow cells with mir-196-specific antagomir abrogates their replating potential in methylcellulose. This demonstrates that mir-196b function is necessary for MLL fusion-mediated immortalization. Furthermore, overexpression of mir-196b was found specifically in patients with MLL associated leukemias as determined from analysis of 55 primary leukemia samples. Overexpression of mir-196b in bone marrow progenitor cells leads to increased proliferative capacity and survival, as well as a partial block in differentiation. Our results suggest a mechanism whereby increased expression of mir-196b by MLL fusion proteins significantly contributes to leukemia development.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación de la Expresión Génica , MicroARNs/genética , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Animales , Secuencia de Bases , Diferenciación Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/patología , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Células Madre Embrionarias/fisiología , Regulación de la Expresión Génica/fisiología , N-Metiltransferasa de Histona-Lisina , Leucemia/etiología , Leucemia/genética , Leucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/fisiología , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/fisiología , Homología de Secuencia de Ácido Nucleico , Regulación hacia Arriba/fisiología
14.
Nat Med ; 8(7): 743-50, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12091906

RESUMEN

The t(8;21) is one of the most frequent chromosomal translocations associated with acute leukemia. This translocation creates a fusion protein consisting of the acute myeloid leukemia-1 transcription factor and the eight-twenty-one corepressor (AML1 ETO), which represses transcription through AML1 (RUNX1) DNA binding sites and immortalizes hematopoietic progenitor cells. We have identified the p14(ARF) tumor suppressor, a mediator of the p53 oncogene checkpoint, as a direct transcriptional target of AML1 ETO. AML1 ETO repressed the p14(ARF) promoter and reduced endogenous levels of p14(ARF) expression in multiple cell types. In contrast, AML1 stimulated p14(ARF) expression and induced phenotypes consistent with cellular senescence. Chromatin immunoprecipitation assays demonstrated that AML1 ETO was specifically bound to the p14(ARF) promoter. In acute myeloid leukemia samples containing the t(8;21), levels of p14(ARF) mRNA were markedly lower when compared with other acute myeloid leukemias lacking this translocation. Repression of p14(ARF) may explain why p53 is not mutated in t(8;21)-containing leukemias and suggests that p14(ARF) is an important tumor suppressor in a large number of human leukemias.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción/genética , Transcripción Genética/fisiología , Translocación Genética , Proteína p14ARF Supresora de Tumor/genética , Antígenos CD/análisis , Antígenos CD4/análisis , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Genes Reporteros , Células Madre Hematopoyéticas/patología , Humanos , Células K562 , Proteínas de Fusión Oncogénica/metabolismo , Plásmidos , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
15.
Proc Natl Acad Sci U S A ; 105(40): 15535-40, 2008 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-18832181

RESUMEN

MicroRNAs (miRNAs) are postulated to be important regulators in cancers. Here, we report a genome-wide miRNA expression analysis in 52 acute myeloid leukemia (AML) samples with common translocations, including t(8;21)/AML1(RUNX1)-ETO(RUNX1T1), inv(16)/CBFB-MYH11, t(15;17)/PML-RARA, and MLL rearrangements. Distinct miRNA expression patterns were observed for t(15;17), MLL rearrangements, and core-binding factor (CBF) AMLs including both t(8;21) and inv(16) samples. Expression signatures of a minimum of two (i.e., miR-126/126*), three (i.e., miR-224, miR-368, and miR-382), and seven (miR-17-5p and miR-20a, plus the aforementioned five) miRNAs could accurately discriminate CBF, t(15;17), and MLL-rearrangement AMLs, respectively, from each other. We further showed that the elevated expression of miR-126/126* in CBF AMLs was associated with promoter demethylation but not with amplification or mutation of the genomic locus. Our gain- and loss-of-function experiments showed that miR-126/126* inhibited apoptosis and increased the viability of AML cells and enhanced the colony-forming ability of mouse normal bone marrow progenitor cells alone and particularly, in cooperation with AML1-ETO, likely through targeting Polo-like kinase 2 (PLK2), a tumor suppressor. Our results demonstrate that specific alterations in miRNA expression distinguish AMLs with common translocations and imply that the deregulation of specific miRNAs may play a role in the development of leukemia with these associated genetic rearrangements.


Asunto(s)
Leucemia Mieloide Aguda/genética , MicroARNs/metabolismo , Translocación Genética , Animales , Apoptosis , Supervivencia Celular , Factores de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Humanos , Ratones , Mutación , Proteínas Serina-Treonina Quinasas/metabolismo
16.
J Exp Med ; 196(10): 1291-305, 2002 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-12438421

RESUMEN

To identify changes in the regulation of B cell receptor (BCR) signals during the development of human B cells, we generated genome-wide gene expression profiles using the serial analysis of gene expression (SAGE) technique for CD34(+) hematopoietic stem cells (HSCs), pre-B cells, naive, germinal center (GC), and memory B cells. Comparing these SAGE profiles, genes encoding positive regulators of BCR signaling were expressed at consistently lower levels in naive B cells than in all other B cell subsets. Conversely, a large group of inhibitory signaling molecules, mostly belonging to the immunoglobulin superfamily (IgSF), were specifically or predominantly expressed in naive B cells. The quantitative differences observed by SAGE were corroborated by semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry. In a functional assay, we show that down-regulation of inhibitory IgSF receptors and increased responsiveness to BCR stimulation in memory as compared with naive B cells at least partly results from interleukin (IL)-4 receptor signaling. Conversely, activation or impairment of the inhibitory IgSF receptor LIRB1 affected BCR-dependent Ca(2+) mobilization only in naive but not memory B cells. Thus, LIRB1 and IL-4 may represent components of two nonoverlapping gene expression programs in naive and memory B cells, respectively: in naive B cells, a large group of inhibitory IgSF receptors can elevate the BCR signaling threshold to prevent these cells from premature activation and clonal expansion before GC-dependent affinity maturation. In memory B cells, facilitated responsiveness upon reencounter of the immunizing antigen may result from amplification of BCR signals at virtually all levels of signal transduction.


Asunto(s)
Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Antígenos CD34/inmunología , Perfilación de la Expresión Génica , Humanos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología
17.
Proc Natl Acad Sci U S A ; 104(50): 19971-6, 2007 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-18056805

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer, whereas acute myeloid leukemia (AML) is the most common acute leukemia in adults. In general, ALL has a better prognosis than AML. To understand the distinct mechanisms in leukemogenesis between ALL and AML and to identify markers for diagnosis and treatment, we performed a large-scale genome-wide microRNA (miRNA, miR) expression profiling assay and identified 27 miRNAs that are differentially expressed between ALL and AML. Among them, miR-128a and -128b are significantly overexpressed, whereas let-7b and miR-223 are significantly down-regulated in ALL compared with AML. They are the most discriminatory miRNAs between ALL and AML. Using the expression signatures of a minimum of two of these miRNAs resulted in an accuracy rate of >95% in the diagnosis of ALL and AML. The differential expression patterns of these four miRNAs were validated further through large-scale real-time PCR on 98 acute leukemia samples covering most of the common cytogenetic subtypes, along with 10 normal control samples. Furthermore, we found that overexpression of miR-128 in ALL was at least partly associated with promoter hypomethylation and not with an amplification of its genomic locus. Taken together, we showed that expression signatures of as few as two miRNAs could accurately discriminate ALL from AML, and that epigenetic regulation might play an important role in the regulation of expression of miRNAs in acute leukemias.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Leucemia Mieloide Aguda/genética , MicroARNs/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Línea Celular Tumoral , ADN/genética , Epigénesis Genética/genética , Amplificación de Genes/genética , Perfilación de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/diagnóstico , Reacción en Cadena de la Polimerasa , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico
18.
Cancer Res ; 67(3): 992-1000, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17283131

RESUMEN

The tumor suppressor gene INK4b (p15) is silenced by CpG island hypermethylation in most acute myelogenous leukemias (AML), and this epigenetic phenomenon can be reversed by treatment with hypomethylating agents. Thus far, it was not investigated whether INK4b is hypermethylated in all cytogenetic subtypes of AML. A comparison of levels of INK4b methylation in AML with the three most common cytogenetic alterations, inv(16), t(8;21), and t(15;17), revealed a strikingly low level of methylation in all leukemias with inv(16) compared with the other types. Surprisingly, the expression level of INK4b in inv(16)+ AML samples was low and comparable with that of the other subtypes. An investigation into an alternative mechanism of INK4b silencing determined that the loss of INK4b expression was caused by inv(16)-encoded core binding factor beta-smooth muscle myosin heavy chain (CBFbeta-SMMHC). The silencing was manifested in an inability to activate the normal expression of INK4b RNA as shown in vitamin D3-treated U937 cells expressing CBFbeta-SMMHC. CBFbeta-SMMHC was shown to displace RUNX1 from a newly determined CBF site in the promoter of INK4b. Importantly, this study (a) establishes that the gene encoding the tumor suppressor p15(INK4b) is a target of CBFbeta-SMMHC, a finding relevant to the leukemogenesis process, and (b) indicates that, in patients with inv(16)-containing AML, reexpression from the INK4b locus in the leukemia would not be predicted to occur using hypomethylating drugs.


Asunto(s)
Inversión Cromosómica , Cromosomas Humanos Par 6 , Subunidad beta del Factor de Unión al Sitio Principal/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Leucemia Mieloide Aguda/genética , Cadenas Pesadas de Miosina/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/biosíntesis , Metilación de ADN , Silenciador del Gen , Humanos , Regiones Promotoras Genéticas , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Activación Transcripcional
19.
Trends Genet ; 21(4): 203-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15797613

RESUMEN

We suggest that small introns are favored in genes requiring a minimal response time ('nimble' genes), and that antisense genes might be prime candidates for such nimble genes. In this article, we show that antisense genes have significantly shorter introns than all other gene categories, which supports our proposal that the short introns of antisense genes might be functionally important and integrally related to their potential role in efficient gene regulation.


Asunto(s)
ADN sin Sentido/genética , Regulación de la Expresión Génica/genética , Genoma Humano , Intrones/genética , Humanos
20.
Trends Genet ; 21(6): 326-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15922830

RESUMEN

Is sense-antisense (SA) pairing of transcripts a common mode of gene regulation in the human genome? Although >20% of human genes might form SA pairs, the extent to which they are involved in antisense regulation is unknown. Simultaneous expression of paired sense and antisense genes is an essential step and an important indicator of antisense regulation. In this article, we demonstrate that human SA pairs tend to be co-expressed and/or inversely expressed more frequently than expected by chance. Moreover, co-expressed and inversely expressed SA pairs exhibit a striking pattern of evolutionary conservation. These findings suggest that antisense regulation is a common and important mechanism of gene regulation in the human genome.


Asunto(s)
Elementos sin Sentido (Genética) , Evolución Molecular , Regulación de la Expresión Génica , Genoma Humano , Transcripción Genética , Animales , Humanos , Ratones , ARN sin Sentido/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA