Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 30(11): 1278-88, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27257215

RESUMEN

Acute myeloid leukemias (AMLs) with a rearrangement of the mixed-linage leukemia (MLL) gene are aggressive hematopoietic malignancies. Here, we explored the feasibility of using the H3K9- and H3K36-specific demethylases Jmjd2/Kdm4 as putative drug targets in MLL-AF9 translocated leukemia. Using Jmjd2a, Jmjd2b, and Jmjd2c conditional triple-knockout mice, we show that Jmjd2/Kdm4 activities are required for MLL-AF9 translocated AML in vivo and in vitro. We demonstrate that expression of the interleukin 3 receptor α (Il3ra also known as Cd123) subunit is dependent on Jmjd2/Kdm4 through a mechanism involving removal of H3K9me3 from the promoter of the Il3ra gene. Importantly, ectopic expression of Il3ra in Jmjd2/Kdm4 knockout cells alleviates the requirement of Jmjd2/Kdm4 for the survival of AML cells, showing that Il3ra is a critical downstream target of Jmjd2/Kdm4 in leukemia. These results suggest that the JMJD2/KDM4 proteins are promising drug targets for the treatment of AML.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Receptor de Interleucina-3/genética , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/fisiopatología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Metilación , Ratones , Ratones Noqueados , Unión Proteica , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico
2.
Blood ; 134(14): 1154-1158, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31434704

RESUMEN

KDM4/JMJD2 are H3K9- and H3K36-specific demethylases, which are considered promising therapeutic targets for the treatment of acute myeloid leukemia (AML) harboring MLL translocations. Here, we investigate the long-term effects of depleting KDM4 activity on normal hematopoiesis to probe potential side effects of continuous inhibition of these enzymes. Utilizing conditional Kdm4a/Kdm4b/Kdm4c triple-knockout mice, we show that KDM4 activity is required for hematopoietic stem cell (HSC) maintenance in vivo. The knockout of the KDM4 demethylases leads to accumulation of H3K9me3 on transcription start sites and the corresponding downregulation of expression of several genes in HSCs. We show that 2 of these genes, Taf1b and Nom1, are essential for the maintenance of hematopoietic cells. Taken together, our results show that the KDM4 demethylases are required for the expression of genes essential for the long-term maintenance of normal hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/citología , Histona Demetilasas/genética , Animales , Supervivencia Celular , Células Cultivadas , Regulación de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Histona Demetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Sitio de Iniciación de la Transcripción
3.
Blood ; 133(23): 2495-2506, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-30917958

RESUMEN

Recurrent inactivating mutations have been identified in the X-linked plant homeodomain finger protein 6 (PHF6) gene, encoding a chromatin-binding transcriptional regulator protein, in various hematological malignancies. However, the role of PHF6 in normal hematopoiesis and its tumor-suppressor function remain largely unknown. We herein generated mice carrying a floxed Phf6 allele and inactivated Phf6 in hematopoietic cells at various developmental stages. The Phf6 deletion in embryos augmented the capacity of hematopoietic stem cells (HSCs) to proliferate in cultures and reconstitute hematopoiesis in recipient mice. The Phf6 deletion in neonates and adults revealed that cycling HSCs readily acquired an advantage in competitive repopulation upon the Phf6 deletion, whereas dormant HSCs only did so after serial transplantations. Phf6-deficient HSCs maintained an enhanced repopulating capacity during serial transplantations; however, they did not induce any hematological malignancies. Mechanistically, Phf6 directly and indirectly activated downstream effectors in tumor necrosis factor α (TNFα) signaling. The Phf6 deletion repressed the expression of a set of genes associated with TNFα signaling, thereby conferring resistance against the TNFα-mediated growth inhibition on HSCs. Collectively, these results not only define Phf6 as a novel negative regulator of HSC self-renewal, implicating inactivating PHF6 mutations in the pathogenesis of hematological malignancies, but also indicate that a Phf6 deficiency alone is not sufficient to induce hematopoietic transformation.


Asunto(s)
Autorrenovación de las Células , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Represoras/metabolismo , Animales , Proliferación Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Curr Opin Hematol ; 27(4): 248-253, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32398456

RESUMEN

PURPOSE OF REVIEW: Even though an increasing amount of sequencing data on the leukemia genome has highlighted a tumor-suppressive function for plant homeodomain finger protein 6 (PHF6), its role in the hematopoietic system remained elusive until recently. The purpose of this review is to describe the role of PHF6 in normal hematopoiesis and leukemogenesis based on recent findings from knockout mouse models. RECENT FINDINGS: In a mouse model, the loss of Phf6 enhanced the bone marrow repopulating capacity of hematopoietic stem cells (HSCs) during serial transplantations without transforming hematopoietic cells, whereas donor mice, which lacked Phf6 expression in the hematopoietic system, did not show any apparent phenotypes in the steady-state. Mechanistically, Phf6 activates effectors in the tumor necrosis factor α (Tnfα) pathway. Therefore, a Phf6 deficiency attenuates the expression of the effectors and confers resistance against Tnfα-mediated growth inhibition to HSCs. Moreover, the loss of Phf6 promoted the development of leukemia induced by aberrant TLX3 expression or an active NOTCH mutation. SUMMARY: Phf6 restricts the self-renewal of HSCs by governing the Tnfα pathway. Phf6 fulfills a tumor-suppressive function, and its loss synergizes with leukemic lesions to promote the onset of hematological malignancies.


Asunto(s)
Carcinogénesis/metabolismo , Hematopoyesis , Leucemia/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Represoras/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia/genética , Leucemia/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Represoras/genética
5.
Blood ; 132(23): 2470-2483, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30228234

RESUMEN

BCOR, encoding BCL-6 corepressor (BCOR), is X-linked and targeted by somatic mutations in various hematological malignancies including myelodysplastic syndrome (MDS). We previously reported that mice lacking Bcor exon 4 (Bcor ΔE4/y ) in the hematopoietic compartment developed NOTCH-dependent acute T-cell lymphoblastic leukemia (T-ALL). Here, we analyzed mice lacking Bcor exons 9 and 10 (Bcor ΔE9-10/y ), which express a carboxyl-terminal truncated BCOR that fails to interact with core effector components of polycomb repressive complex 1.1. Bcor ΔE9-10/y mice developed lethal T-ALL in a similar manner to Bcor ΔE4/y mice, whereas Bcor ΔE9-10/y hematopoietic cells showed a growth advantage in the myeloid compartment that was further enhanced by the concurrent deletion of Tet2 Tet2 Δ/Δ Bcor ΔE9-10/y mice developed lethal MDS with progressive anemia and leukocytopenia, inefficient hematopoiesis, and the morphological dysplasia of blood cells. Tet2 Δ/Δ Bcor ΔE9-10/y MDS cells reproduced MDS or evolved into lethal MDS/myeloproliferative neoplasms in secondary recipients. Transcriptional profiling revealed the derepression of myeloid regulator genes of the Cebp family and Hoxa cluster genes in Bcor ΔE9-10/y progenitor cells and the activation of p53 target genes specifically in MDS erythroblasts where massive apoptosis occurred. Our results reveal a tumor suppressor function of BCOR in myeloid malignancies and highlight the impact of Bcor insufficiency on the initiation and progression of MDS.


Asunto(s)
Secuencia de Aminoácidos , Exones , Síndromes Mielodisplásicos , Proteínas Represoras , Eliminación de Secuencia , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Cell Microbiol ; 21(8): e13033, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31009148

RESUMEN

Vibrio cholerae produced-Cholix toxin (Cholix) is a cytotoxin that ADP-ribosylates eukaryotic elongation factor 2, inhibiting protein synthesis, and inducing apoptosis. Here, we identified prohibitin (PHB) 1 and 2 as novel Cholix-interacting membrane proteins in immortalised human hepatocytes and HepG2 cells by Cholix immunoprecipitation assays. The expression level of PHB1 was decreased by Cholix after a 12hr incubation. Cholix-induced poly (ADP-ribose) polymerase (PARP) cleavage was significantly enhanced in PHB (PHB1 or PHB2) knockdown cells. In contrast, transiently overexpressed PHB in hepatocytes attenuated Cholix-induced Bax/Bak conformational changes and PARP cleavage. In addition, Cholix-induced reactive oxygen species production and accumulation of fragmented mitochondria were enhanced in PHB-knockdown cells. Furthermore, Cholix induced activation of Rho-associated coiled coil-containing protein kinase 1 (ROCK1), which was enhanced in PHB-knockdown cells, followed by actin filament depolymerisation and accumulation of tubulin in the blebbing cells. Inhibition of ROCK1 by siRNA or its inhibitor suppressed Cholix-induced PARP cleavage and reactive oxygen species generation. Our findings identify PHB as a new protein that interacts with Cholix and is involved in Cholix-induced mitochondrial dysfunction and cytoskeletal rearrangement by ROCK1 activation during apoptosis.


Asunto(s)
Factores de Ribosilacion-ADP/química , Toxinas Bacterianas/química , Factor 2 Eucariótico de Iniciación/metabolismo , Interacciones Huésped-Patógeno/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Vibrio cholerae/genética , ADP-Ribosilación , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiología , Citoesqueleto de Actina/ultraestructura , Secuencia de Aminoácidos , Apoptosis/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular Transformada , Factor 2 Eucariótico de Iniciación/genética , Regulación de la Expresión Génica , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/microbiología , Hepatocitos/patología , Humanos , Mitocondrias/metabolismo , Mitocondrias/microbiología , Mitocondrias/ultraestructura , Prohibitinas , Unión Proteica , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/deficiencia , Transducción de Señal , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidad , Virulencia , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
7.
Blood ; 128(5): 638-49, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27301860

RESUMEN

Setdb1, also known as Eset, is a methyltransferase that catalyzes trimethylation of H3K9 (H3K9me3) and plays an essential role in the silencing of endogenous retroviral elements (ERVs) in the developing embryo and embryonic stem cells (ESCs). Its role in somatic stem cells, however, remains unclear because of the early death of Setdb1-deficient embryos. We demonstrate here that Setdb1 is the first H3K9 methyltransferase shown to be essential for the maintenance of hematopoietic stem and progenitor cells (HSPCs) in mice. The deletion of Setdb1 caused the rapid depletion of hematopoietic stem and progenitor cells (HSPCs), as well as leukemic stem cells. In contrast to ESCs, ERVs were largely repressed in Setdb1-deficient HSPCs. A list of nonhematopoietic genes was instead ectopically activated in HSPCs after reductions in H3K9me3 levels, including key gluconeogenic enzyme genes fructose-1,6-bisphosphatase 1 (Fbp1) and Fbp2 The ectopic activation of gluconeogenic enzymes antagonized glycolysis and impaired ATP production, resulting in a compromised repopulating capacity of HSPCs. Our results demonstrate that Setdb1 maintains HSPCs by restricting the ectopic activation of nonhematopoietic genes detrimental to their function and uncover that the gluconeogenic pathway is one of the critical targets of Setdb1 in HSPCs.


Asunto(s)
Regulación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Médula Ósea/patología , Retrovirus Endógenos/metabolismo , Molécula de Adhesión Celular Epitelial/metabolismo , Eliminación de Gen , Silenciador del Gen , Gluconeogénesis/genética , Homeostasis/genética , Leucemia/genética , Leucemia/patología , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
8.
Blood ; 123(12): 1870-82, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24501218

RESUMEN

Epigenetic regulatory mechanisms are implicated in the pathogenesis of acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL). Recent progress suggests that proteins involved in epigenetic control are amenable to drug intervention, but little is known about the cancer-specific dependency on epigenetic regulators for cell survival and proliferation. We used a mouse model of human AML induced by the MLL-AF9 fusion oncogene and an epigenetic short hairpin RNA (shRNA) library to screen for novel potential drug targets. As a counter-screen for general toxicity of shRNAs, we used normal mouse bone marrow cells. One of the best candidate drug targets identified in these screens was Jmjd1c. Depletion of Jmjd1c impairs growth and colony formation of mouse MLL-AF9 cells in vitro as well as establishment of leukemia after transplantation. Depletion of JMJD1C impairs expansion and colony formation of human leukemic cell lines, with the strongest effect observed in the MLL-rearranged ALL cell line SEM. In both mouse and human leukemic cells, the growth defect upon JMJD1C depletion appears to be primarily due to increased apoptosis, which implicates JMJD1C as a potential therapeutic target in leukemia.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/genética , Leucemia Mieloide Aguda/genética , Oxidorreductasas N-Desmetilantes/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Epigénesis Genética , Técnicas de Silenciamiento del Gen , Genes myb , Genes myc , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Leucemia Experimental/genética , Leucemia Experimental/patología , Leucemia Mieloide Aguda/patología , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Oxidorreductasas N-Desmetilantes/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Ensayo de Tumor de Célula Madre
9.
Blood ; 121(3): 447-58, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23169777

RESUMEN

To search for genes that promote hematopoietic development from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), we overexpressed several known hematopoietic regulator genes in hESC/iPSC-derived CD34(+)CD43(-) endothelial cells (ECs) enriched in hemogenic endothelium (HE). Among the genes tested, only Sox17, a gene encoding a transcription factor of the SOX family, promoted cell growth and supported expansion of CD34(+)CD43(+)CD45(-/low) cells expressing the HE marker VE-cadherin. SOX17 was expressed at high levels in CD34(+)CD43(-) ECs compared with low levels in CD34(+)CD43(+)CD45(-) pre-hematopoietic progenitor cells (pre-HPCs) and CD34(+)CD43(+)CD45(+) HPCs. Sox17-overexpressing cells formed semiadherent cell aggregates and generated few hematopoietic progenies. However, they retained hemogenic potential and gave rise to hematopoietic progenies on inactivation of Sox17. Global gene-expression analyses revealed that the CD34(+)CD43(+)CD45(-/low) cells expanded on overexpression of Sox17 are HE-like cells developmentally placed between ECs and pre-HPCs. Sox17 overexpression also reprogrammed both pre-HPCs and HPCs into HE-like cells. Genome-wide mapping of Sox17-binding sites revealed that Sox17 activates the transcription of key regulator genes for vasculogenesis, hematopoiesis, and erythrocyte differentiation directly. Depletion of SOX17 in CD34(+)CD43(-) ECs severely compromised their hemogenic activity. These findings suggest that SOX17 plays a key role in priming hemogenic potential in ECs, thereby regulating hematopoietic development from hESCs/iPSCs.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/fisiología , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/fisiología , Sangre Fetal/citología , Fibroblastos/citología , Hematopoyesis/genética , Humanos , Lentivirus/genética , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transducción Genética/métodos
10.
Blood ; 120(5): 1107-17, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22677129

RESUMEN

EZH2, a catalytic component of the polycomb repressive complex 2, trimethylates histone H3 at lysine 27 (H3K27) to repress the transcription of target genes. Although EZH2 is overexpressed in various cancers, including some hematologic malignancies, the role of EZH2 in acute myeloid leukemia (AML) has yet to be examined in vivo. In the present study, we transformed granulocyte macrophage progenitors from Cre-ERT;Ezh2(flox/flox) mice with the MLL-AF9 leukemic fusion gene to analyze the function of Ezh2 in AML. Deletion of Ezh2 in transformed granulocyte macrophage progenitors compromised growth severely in vitro and attenuated the progression of AML significantly in vivo. Ezh2-deficient leukemic cells developed into a chronic myelomonocytic leukemia-like disease with a lower frequency of leukemia-initiating cells compared with the control. Chromatin immunoprecipitation followed by sequencing revealed a significant reduction in the levels of trimethylation at H3K27 in Ezh2-deficient leukemic cells, not only at Cdkn2a, a known major target of Ezh2, but also at a cohort of genes relevant to the developmental and differentiation processes. Overexpression of Egr1, one of the derepressed genes in Ezh2-deficient leukemic cells, promoted the differentiation of AML cells profoundly. Our findings suggest that Ezh2 inhibits differentiation programs in leukemic stem cells, thereby augmenting their leukemogenic activity.


Asunto(s)
Diferenciación Celular/genética , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/fisiología , Leucemia Mieloide Aguda/genética , Factores de Transcripción/fisiología , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Proteína Potenciadora del Homólogo Zeste 2 , Eliminación de Gen , Células HEK293 , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Complejo Represivo Polycomb 2 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/genética
11.
Blood ; 118(25): 6553-61, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22042701

RESUMEN

Polycomb-group (PcG) proteins are essential regulators of hematopoietic stem cells (HSCs). In contrast to Bmi1, a component of Polycomb repressive complex 1 (PRC1), the role of PRC2 and its components in hematopoiesis remains elusive. Here we show that Ezh2, a core component of PRC2, is essential for fetal, but not adult, HSCs. Ezh2-deficient embryos died of anemia because of insufficient expansion of HSCs/progenitor cells and defective erythropoiesis in fetal liver. Deletion of Ezh2 in adult BM, however, did not significantly compromise hematopoiesis, except for lymphopoiesis. Of note, Ezh2-deficient fetal liver cells showed a drastic reduction in trimethylation of histone H3 at lysine 27 (H3K27me3) accompanied by derepression of a large cohort of genes, whereas on homing to BM, they acquired a high level of H3K27me3 and long-term repopulating capacity. Quantitative RT-PCR revealed that Ezh1, the gene encoding a backup enzyme, is highly expressed in HSCs/progenitor cells in BM compared with those in fetal liver, whereas Ezh2 is ubiquitously expressed. These findings suggest that Ezh1 complements Ezh2 in the BM, but not in the fetal liver, and reveal that the reinforcement of PcG-mediated gene silencing occurs during the transition from proliferative fetal HSCs to quiescent adult HSCs.


Asunto(s)
Células Madre Adultas/metabolismo , Células Madre Fetales/metabolismo , Células Madre Hematopoyéticas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Western Blotting , Médula Ósea/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Perfilación de la Expresión Génica , Hematopoyesis/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Hígado/citología , Hígado/embriología , Hígado/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Blood ; 118(9): 2443-53, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21753189

RESUMEN

The histone acetyltransferases (HATs) of the MYST family include TIP60, HBO1, MOZ/MORF, and MOF and function in multisubunit protein complexes. Bromodomain-containing protein 1 (BRD1), also known as BRPF2, has been considered a subunit of the MOZ/MORF H3 HAT complex based on analogy with BRPF1 and BRPF3. However, its physiologic function remains obscure. Here we show that BRD1 forms a novel HAT complex with HBO1 and regulates erythropoiesis. Brd1-deficient embryos showed severe anemia because of impaired fetal liver erythropoiesis. Biochemical analyses revealed that BRD1 bridges HBO1 and its activator protein, ING4. Genome-wide mapping in erythroblasts demonstrated that BRD1 and HBO1 largely colocalize in the genome and target key developmental regulator genes. Of note, levels of global acetylation of histone H3 at lysine 14 (H3K14) were profoundly decreased in Brd1-deficient erythroblasts and depletion of Hbo1 similarly affected H3K14 acetylation. Impaired erythropoiesis in the absence of Brd1 accompanied reduced expression of key erythroid regulator genes, including Gata1, and was partially restored by forced expression of Gata1. Our findings suggest that the Hbo1-Brd1 complex is the major H3K14 HAT required for transcriptional activation of erythroid developmental regulator genes.


Asunto(s)
Eritropoyesis , Histona Acetiltransferasas/fisiología , Hígado/embriología , Procesamiento Proteico-Postraduccional , Transactivadores/fisiología , Acetilación , Anemia/embriología , Anemia/genética , Animales , Proteínas Portadoras/fisiología , Daño del ADN , Replicación del ADN , Muerte Fetal/sangre , Muerte Fetal/etiología , Muerte Fetal/genética , Factor de Transcripción GATA1/metabolismo , Genes Letales , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Humanos , Células K562 , Hígado/fisiología , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos , Neoplasias/genética , Neoplasias/metabolismo , Mapeo de Interacción de Proteínas , ARN Interferente Pequeño/farmacología , Transactivadores/deficiencia , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/fisiología
13.
Int J Cancer ; 130(11): 2557-67, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21717453

RESUMEN

Recent advances in stem cell biology have identified tumor-initiating cells (TICs) in a variety of cancers including hepatocellular carcinoma (HCC). Polycomb group gene products such as BMI1 and EZH2 have been characterized as general self-renewal regulators in a wide range of normal stem cells and TICs. We previously reported that Ezh2 tightly regulates the self-renewal and differentiation of murine hepatic stem/progenitor cells. However, the role of EZH2 in tumor-initiating HCC cells remains unclear. In this study, we conducted loss-of-function assay of EZH2 using short-hairpin RNA and pharmacological inhibition of EZH2 by an S-adenosylhomocysteine hydrolase inhibitor, 3-deazaneplanocin A (DZNep). Both EZH2-knockdown and DZNep treatment impaired cell growth and anchorage-independent sphere formation of HCC cells in culture. Flow cytometric analyses revealed that the two approaches decreased the number of epithelial cell adhesion molecule (EpCAM)(+) tumor-initiating cells. Administration of 5-fluorouracil (5-FU) or DZNep suppressed the tumors by implanted HCC cells in non-obese diabetic/severe combined immunodeficient mice. Of note, however, DZNep but not 5-FU predominantly reduced the number of EpCAM(+) cells and diminished the self-renewal capability of these cells as judged by sphere formation assays. Our findings reveal that tumor-initiating HCC cells are highly dependent on EZH2 for their tumorigenic activity. Although further analyses of TICs from primary HCC would be necessary, pharmacological interference with EZH2 might be a promising therapeutic approach to targeting tumor-initiating HCC cells.


Asunto(s)
Adenosina/análogos & derivados , Adenosilhomocisteinasa/antagonistas & inhibidores , Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas de Unión al ADN/fisiología , Neoplasias Hepáticas/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Factores de Transcripción/fisiología , Adenosina/farmacología , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2 , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ratones , Complejo Represivo Polycomb 2
14.
Inflamm Regen ; 42(1): 6, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-35227325

RESUMEN

BACKGROUND: The expression of FZD5 distinguishes immature human mesenchymal stem/stromal cells (MSC) in cultures, and the function of FZD5 is crucial for maintaining the proliferation and multilineage differentiation capacity of human MSC. We herein investigated whether Fzd5 expression also marks undifferentiated MSC in animals. METHODS: We generated a transgenic mouse strain (Fzd5-CreERT-tFP635) that expresses CreERT and the fluorescent protein, TurboFP635 (tFP635), under the transcriptional control of the Fzd5 gene using the BAC transgenic technique, and identified cells expressing tFP635 by flow cytometry. We also conducted lineage tracing with this strain. RESULTS: In the bone marrow of transgenic mice, tFP635 was preferentially expressed in MSC, Leptin receptor-expressing MSC (LepR+MSCs), and some Pdgfrα+ Sca1+ MSC (PαS). Inducible lineage tracing using the Fzd5-CreERT-tFP635; CAG-CAT-EGFP strain at the adult stage showed that Fzd5-expressing cells and their descendants labeled with GFP were progressively dominant in LepR+MSC and PαS, and GFP+ cells persisted for 1 year after the activation of CreERT. Adipocyte progenitor cells (APCs), osteoblast progenitor cells (OPCs), and Cd51+ stromal cells were also labeled with GFP. CONCLUSIONS: Our transgenic mouse marks two different types of MSC, LepR+MSC and PαS.

15.
Br J Haematol ; 153(5): 589-98, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21492125

RESUMEN

Myeloproliferative neoplasms (MPN), a group of haematopoietic stem cell (HSC) disorders, are often accompanied by myelofibrosis. We previously identified the fusion of the ETV6 gene to the LYN gene (ETV6-LYN) in idiopathic myelofibrosis with ins(12;8)(p13;q11q21). The introduction of ETV6-LYN into HSCs resulted in fatal MPN with massive myelofibrosis in mice, implicating the rearranged LYN kinase in the pathogenesis of MPN with myelofibrosis. However, the signalling molecules directly downstream from and activated by ETV6-LYN remain unknown. In this study, we demonstrated that the direct activation of STAT5 by ETV6-LYN is crucial for the development of MPN. ETV6-LYN was constitutively active as a kinase through autophosphorylation. ETV6-LYN, but not its kinase-dead mutant, supported cytokine-free proliferation of haematopoietic cells. STAT5 was activated in a JAK2-independent manner in ETV6-LYN-expressing cells. ETV6-LYN interacted with STAT5 and directly activated STAT5 both in vitro and in vivo. Of note, ETV6-LYN did not support the formation of colonies by Stat5-deficient HSCs under cytokine-free conditions and the capacity of ETV6-LYN to induce MPN with myelofibrosis was profoundly attenuated in a Stat5-null background. These findings define STAT5 as a direct target of ETV6-LYN and unveil the LYN-STAT5 axis as a novel pathway to augment proliferative signals in MPN and leukaemia.


Asunto(s)
Trastornos Mieloproliferativos/metabolismo , Mielofibrosis Primaria/metabolismo , Proteínas Proto-Oncogénicas c-ets/fisiología , Proteínas Represoras/fisiología , Factor de Transcripción STAT5/metabolismo , Familia-src Quinasas/fisiología , Animales , Proliferación Celular , Células Cultivadas , Citocinas/fisiología , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Fusión Oncogénica/fisiología , Fosforilación/fisiología , Proteínas Recombinantes de Fusión , Transducción de Señal/fisiología , Proteína ETS de Variante de Translocación 6
16.
Hepatology ; 52(3): 1111-23, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20648475

RESUMEN

UNLABELLED: We previously reported that forced expression of Bmi1 (B lymphoma Moloney murine leukemia virus insertion region 1 homolog) in murine hepatic stem/progenitor cells purified from fetal liver enhances their self-renewal and drives cancer initiation. In the present study, we examined the contribution of the Ink4a/Arf tumor suppressor gene locus, one of the major targets of Bmi1, to stem cell expansion and cancer initiation. Bmi1(-/-) Delta-like protein (Dlk)(+) hepatic stem/progenitor cells showed de-repression of the Ink4a/Arf locus and displayed impaired growth activity. In contrast, Ink4a/Arf(-/-) Dlk(+) cells gave rise to considerably larger colonies containing a greater number of bipotent cells than wild-type Dlk(+) cells. Although Ink4a/Arf(-/-) Dlk(+) cells did not initiate tumors in recipient nonobese diabetic/severe combined immunodeficiency mice, enforced expression of Bmi1 in Ink4a/Arf(-/-) Dlk(+) cells further augmented their self-renewal capacity and resulted in tumor formation in vivo. Microarray analyses successfully identified five down-regulated genes as candidate downstream targets for Bmi1 in hepatic stem/progenitor cells. Of these genes, enforced expression of sex determining region Y-box 17 (Sox17) in Dlk(+) cells strongly suppressed colony propagation and tumor growth. CONCLUSION: These results indicate that repression of targets of Bmi1 other than the Ink4a/Arf locus plays a crucial role in the oncogenic transformation of hepatic stem/progenitor cells. Functional analyses of Bmi1 target genes would be of importance to elucidate the molecular machinery underlying hepatic stem cell system and explore therapeutic approaches for the eradication of liver cancer stem cells.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Madre Embrionarias/metabolismo , Hígado/embriología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Células Madre Embrionarias/citología , Proteínas HMGB/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Modelos Animales , Proteínas Nucleares/genética , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Factores de Transcripción SOXF/metabolismo
17.
J Hepatol ; 52(6): 854-63, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20395008

RESUMEN

BACKGROUND & AIMS: Polycomb group proteins initiate and maintain gene silencing through chromatin modifications and contribute to the maintenance of self-renewal in a variety of stem cells. Among polycomb repressive complexes (PRCs), PRC2 initiates gene silencing by methylating histone H3 lysine 27, and PRC1 maintains gene silencing through mono-ubiquitination of histone H2A lysine 119. We have previously shown that Bmi1, a core component of PRC1, tightly regulates the self-renewal of hepatic stem/progenitor cells. METHODS: In this study, we conducted lentivirus-mediated knockdown of Ezh2 to characterise the function of Ezh2, a major component of PRC2, in hepatic stem/progenitor cells. RESULTS: Loss of Ezh2 function in embryonic murine hepatic stem/progenitor cells severely impaired proliferation and self-renewal capability. This effect was more prominent than that of Bmi1-knockdown and was partially abrogated by the deletion of both Ink4a and Arf, major targets of PRC1 and PRC2. Importantly, Ezh2-knockdown but not Bmi1-knockdown promoted the differentiation and terminal maturation of hepatocytes, followed by the up-regulation of several transcriptional regulators of hepatocyte differentiation. CONCLUSIONS: Our findings indicate that Ezh2 plays an essential role in the maintenance of both the proliferative and self-renewal capacity of hepatic stem/progenitor cells and the full execution of their differentiation.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Hígado/citología , Células Madre/citología , Células Madre/fisiología , Factor 1 de Ribosilacion-ADP/genética , Animales , Conductos Biliares/citología , Diferenciación Celular/fisiología , División Celular/fisiología , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Hepatocitos/citología , Histonas/metabolismo , Homeostasis/fisiología , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Embarazo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
18.
Genes Cells ; 14(11): 1347-57, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19845771

RESUMEN

Faithful control of cell cycle checkpoint and DNA repair contributes to prevent the cells from chromosomal instability and tumorigenesis. Dnmt1-associated protein 1 (Dmap1), a component of the NuA4 histone acetyltransferase complex, was originally identified as an interacting molecule with DNMT1 which co-localizes with PCNA at DNA replication foci. However, its role in cellular functions remains largely unknown. Here we show that Dmap1 knockdown in mouse embryonic fibroblasts (MEFs) lead to spontaneous double-strand breaks (DSBs), resulting in growth arrest because of p53-dependent cell cycle checkpoint activation. Deletion of both Dmap1 and p53 in MEFs synergized towards enhanced generation of DSBs, chromosomal abnormalities and tumor development in mice. Notably, we found that, on DNA damage, Dmap1 was recruited to the damaged sites to form complexes with gamma-H2AX and replication factors, including Pcna. Depletion of Dmap1 in MEFs abrogated the stable accumulation of Pcna at the DNA damaged sites. Furthermore, the re-introduction of Dmap1 mutants with a reduced capacity to bind Pcna failed to ameliorate the impaired DNA repair in Dmap1-depleted cells. These findings indicate that Dmap1 is required to involve Pcna in DNA repair. Together, Dmap1 plays a crucial role in DNA repair, and is indispensable for the maintenance of chromosomal integrity.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Reparación del ADN , Genoma/genética , Animales , Proliferación Celular , Inestabilidad Cromosómica/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Fibroblastos/citología , Regulación de la Expresión Génica , Ratones , Antígeno Nuclear de Célula en Proliferación/metabolismo , Regulación hacia Arriba
19.
Cell Mol Life Sci ; 66(23): 3675-84, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19633813

RESUMEN

SoxB1 factors, which include Sox1, 2, and 3, share more than 90% amino acid identity in their DNA binding HMG box and participate in diverse developmental events. They are known to exert cell-type-specific functions in concert with other transcription factors on Sox factor-dependent regulatory enhancers. Due to the high degree of sequence similarity both within and outside the HMG box, SoxB1 members show almost identical biological activities. As a result, they exhibit strong functional redundancy in regions where SoxB1 members are coexpressed, such as neural stem/progenitor cells in the developing central nervous system.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción SOXB1/fisiología , Animales , Desarrollo Embrionario/genética , Células Madre Embrionarias/metabolismo , Oftalmopatías/genética , Humanos , Cristalino/citología , Cristalino/crecimiento & desarrollo , Cristalino/metabolismo , Ratones , Modelos Genéticos , Factores de Transcripción SOXB1/metabolismo
20.
Mol Cell Biol ; 26(17): 6557-70, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16914739

RESUMEN

Many genes have been identified that are specifically expressed in multiple types of stem cells in their undifferentiated state. It is generally assumed that at least some of these putative "stemness" genes are involved in maintaining properties that are common to all stem cells. We compared gene expression profiles between undifferentiated and differentiated embryonic stem cells (ESCs) using DNA microarrays. We identified several genes with much greater signal in undifferentiated ESCs than in their differentiated derivatives, among them the putative stemness gene encoding junctional adhesion molecule B (Jam-B gene). However, in spite of the specific expression in undifferentiated ESCs, Jam-B mutant ESCs had normal morphology and pluripotency. Furthermore, Jam-B homozygous mutant mice are fertile and have no overt developmental defects. Moreover, we found that neural and hematopoietic stem cells recovered from Jam-B mutant mice are not impaired in their ability to self-renew and differentiate. These results demonstrate that Jam-B is dispensable for normal mouse development and stem cell identity in embryonic, neural, and hematopoietic stem cells.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Embrión de Mamíferos/citología , Células Madre Hematopoyéticas/citología , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/metabolismo , Tejido Nervioso/citología , Animales , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Diferenciación Celular , Células Cultivadas , Cruzamientos Genéticos , Femenino , Regulación de la Expresión Génica , Marcación de Gen , Heterocigoto , Inmunoglobulinas/deficiencia , Inmunoglobulinas/genética , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Análisis por Micromatrices , Células Madre Multipotentes/citología , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Testículo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA