Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 150(3): 575-89, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863010

RESUMEN

The mechanism by which cells decide to skip mitosis to become polyploid is largely undefined. Here we used a high-content image-based screen to identify small-molecule probes that induce polyploidization of megakaryocytic leukemia cells and serve as perturbagens to help understand this process. Our study implicates five networks of kinases that regulate the switch to polyploidy. Moreover, we find that dimethylfasudil (diMF, H-1152P) selectively increased polyploidization, mature cell-surface marker expression, and apoptosis of malignant megakaryocytes. An integrated target identification approach employing proteomic and shRNA screening revealed that a major target of diMF is Aurora kinase A (AURKA). We further find that MLN8237 (Alisertib), a selective inhibitor of AURKA, induced polyploidization and expression of mature megakaryocyte markers in acute megakaryocytic leukemia (AMKL) blasts and displayed potent anti-AMKL activity in vivo. Our findings provide a rationale to support clinical trials of MLN8237 and other inducers of polyploidization and differentiation in AMKL.


Asunto(s)
Azepinas/farmacología , Descubrimiento de Drogas , Leucemia Megacarioblástica Aguda/tratamiento farmacológico , Megacariocitos/metabolismo , Poliploidía , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Aurora Quinasa A , Aurora Quinasas , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Leucemia Megacarioblástica Aguda/genética , Megacariocitos/citología , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas Asociadas a rho/metabolismo
2.
Nature ; 578(7795): 455-460, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32025035

RESUMEN

Xist represents a paradigm for the function of long non-coding RNA in epigenetic regulation, although how it mediates X-chromosome inactivation (XCI) remains largely unexplained. Several proteins that bind to Xist RNA have recently been identified, including the transcriptional repressor SPEN1-3, the loss of which has been associated with deficient XCI at multiple loci2-6. Here we show in mice that SPEN is a key orchestrator of XCI in vivo and we elucidate its mechanism of action. We show that SPEN is essential for initiating gene silencing on the X chromosome in preimplantation mouse embryos and in embryonic stem cells. SPEN is dispensable for maintenance of XCI in neural progenitors, although it significantly decreases the expression of genes that escape XCI. We show that SPEN is immediately recruited to the X chromosome upon the upregulation of Xist, and is targeted to enhancers and promoters of active genes. SPEN rapidly disengages from chromatin upon gene silencing, suggesting that active transcription is required to tether SPEN to chromatin. We define the SPOC domain as a major effector of the gene-silencing function of SPEN, and show that tethering SPOC to Xist RNA is sufficient to mediate gene silencing. We identify the protein partners of SPOC, including NCoR/SMRT, the m6A RNA methylation machinery, the NuRD complex, RNA polymerase II and factors involved in the regulation of transcription initiation and elongation. We propose that SPEN acts as a molecular integrator for the initiation of XCI, bridging Xist RNA with the transcription machinery-as well as with nucleosome remodellers and histone deacetylases-at active enhancers and promoters.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Silenciador del Gen , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Blastocisto/citología , Blastocisto/metabolismo , Cromatina/genética , Cromatina/metabolismo , Proteínas de Unión al ADN/química , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Histona Desacetilasas/metabolismo , Masculino , Metilación , Ratones , Regiones Promotoras Genéticas/genética , Dominios Proteicos , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/química
3.
Blood ; 141(18): 2245-2260, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36735909

RESUMEN

The NFIA-ETO2 fusion is the product of a t(1;16)(p31;q24) chromosomal translocation, so far, exclusively found in pediatric patients with pure erythroid leukemia (PEL). To address the role for the pathogenesis of the disease, we facilitated the expression of the NFIA-ETO2 fusion in murine erythroblasts (EBs). We observed that NFIA-ETO2 significantly increased proliferation and impaired erythroid differentiation of murine erythroleukemia cells and of primary fetal liver-derived EBs. However, NFIA-ETO2-expressing EBs acquired neither aberrant in vitro clonogenic activity nor disease-inducing potential upon transplantation into irradiated syngenic mice. In contrast, in the presence of 1 of the most prevalent erythroleukemia-associated mutations, TP53R248Q, expression of NFIA-ETO2 resulted in aberrant clonogenic activity and induced a fully penetrant transplantable PEL-like disease in mice. Molecular studies support that NFIA-ETO2 interferes with erythroid differentiation by preferentially binding and repressing erythroid genes that contain NFI binding sites and/or are decorated by ETO2, resulting in a activity shift from GATA- to ETS-motif-containing target genes. In contrast, TP53R248Q does not affect erythroid differentiation but provides self-renewal and survival potential, mostly via downregulation of known TP53 targets. Collectively, our work indicates that NFIA-ETO2 initiates PEL by suppressing gene expression programs of terminal erythroid differentiation and cooperates with TP53 mutation to induce erythroleukemia.


Asunto(s)
Leucemia Eritroblástica Aguda , Proteínas Represoras , Animales , Ratones , Proteínas Represoras/genética , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Diferenciación Celular/genética , Eritroblastos/metabolismo , Factores de Transcripción NFI/metabolismo
4.
Mol Cancer ; 21(1): 65, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246138

RESUMEN

BACKGROUND: Anaplastic large cell lymphoma positive for ALK (ALK+ ALCL) is a rare type of non-Hodgkin lymphoma. This lymphoma is caused by chromosomal translocations involving the anaplastic lymphoma kinase gene (ALK). In this study, we aimed to identify mechanisms of transformation and therapeutic targets by generating a model of ALK+ ALCL lymphomagenesis ab initio with the specific NPM-ALK fusion. METHODS: We performed CRISPR/Cas9-mediated genome editing of the NPM-ALK chromosomal translocation in primary human activated T lymphocytes. RESULTS: Both CD4+ and CD8+ NPM-ALK-edited T lymphocytes showed rapid and reproducible competitive advantage in culture and led to in vivo disease development with nodal and extra-nodal features. Murine tumors displayed the phenotypic diversity observed in ALK+ ALCL patients, including CD4+ and CD8+ lymphomas. Assessment of transcriptome data from models and patients revealed global activation of the WNT signaling pathway, including both canonical and non-canonical pathways, during ALK+ ALCL lymphomagenesis. Specifically, we found that the WNT signaling cell surface receptor ROR2 represented a robust and genuine marker of all ALK+ ALCL patient tumor samples. CONCLUSIONS: In this study, ab initio modeling of the ALK+ ALCL chromosomal translocation in mature T lymphocytes enabled the identification of new therapeutic targets. As ROR2 targeting approaches for other cancers are under development (including lung and ovarian tumors), our findings suggest that ALK+ ALCL cases with resistance to current therapies may also benefit from ROR2 targeting strategies.


Asunto(s)
Linfoma Anaplásico de Células Grandes , Quinasa de Linfoma Anaplásico/genética , Animales , Humanos , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/metabolismo , Linfoma Anaplásico de Células Grandes/patología , Ratones , Fenotipo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Translocación Genética
5.
Blood ; 136(6): 698-714, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32350520

RESUMEN

Acute erythroleukemia (AEL or acute myeloid leukemia [AML]-M6) is a rare but aggressive hematologic malignancy. Previous studies showed that AEL leukemic cells often carry complex karyotypes and mutations in known AML-associated oncogenes. To better define the underlying molecular mechanisms driving the erythroid phenotype, we studied a series of 33 AEL samples representing 3 genetic AEL subgroups including TP53-mutated, epigenetic regulator-mutated (eg, DNMT3A, TET2, or IDH2), and undefined cases with low mutational burden. We established an erythroid vs myeloid transcriptome-based space in which, independently of the molecular subgroup, the majority of the AEL samples exhibited a unique mapping different from both non-M6 AML and myelodysplastic syndrome samples. Notably, >25% of AEL patients, including in the genetically undefined subgroup, showed aberrant expression of key transcriptional regulators, including SKI, ERG, and ETO2. Ectopic expression of these factors in murine erythroid progenitors blocked in vitro erythroid differentiation and led to immortalization associated with decreased chromatin accessibility at GATA1-binding sites and functional interference with GATA1 activity. In vivo models showed development of lethal erythroid, mixed erythroid/myeloid, or other malignancies depending on the cell population in which AEL-associated alterations were expressed. Collectively, our data indicate that AEL is a molecularly heterogeneous disease with an erythroid identity that results in part from the aberrant activity of key erythroid transcription factors in hematopoietic stem or progenitor cells.


Asunto(s)
Leucemia Eritroblástica Aguda/genética , Proteínas de Neoplasias/fisiología , Factores de Transcripción/fisiología , Transcriptoma , Adulto , Animales , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Eritroblastos/metabolismo , Eritropoyesis/genética , Femenino , Factor de Transcripción GATA1/deficiencia , Factor de Transcripción GATA1/genética , Técnicas de Sustitución del Gen , Heterogeneidad Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Persona de Mediana Edad , Mutación , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , RNA-Seq , Quimera por Radiación , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Factores de Transcripción/genética , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/fisiología , Secuenciación del Exoma , Adulto Joven
8.
EMBO J ; 32(5): 645-55, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23353889

RESUMEN

TET proteins convert 5-methylcytosine to 5-hydroxymethylcytosine, an emerging dynamic epigenetic state of DNA that can influence transcription. Evidence has linked TET1 function to epigenetic repression complexes, yet mechanistic information, especially for the TET2 and TET3 proteins, remains limited. Here, we show a direct interaction of TET2 and TET3 with O-GlcNAc transferase (OGT). OGT does not appear to influence hmC activity, rather TET2 and TET3 promote OGT activity. TET2/3-OGT co-localize on chromatin at active promoters enriched for H3K4me3 and reduction of either TET2/3 or OGT activity results in a direct decrease in H3K4me3 and concomitant decreased transcription. Further, we show that Host Cell Factor 1 (HCF1), a component of the H3K4 methyltransferase SET1/COMPASS complex, is a specific GlcNAcylation target of TET2/3-OGT, and modification of HCF1 is important for the integrity of SET1/COMPASS. Additionally, we find both TET proteins and OGT activity promote binding of the SET1/COMPASS H3K4 methyltransferase, SETD1A, to chromatin. Finally, studies in Tet2 knockout mouse bone marrow tissue extend and support the data as decreases are observed of global GlcNAcylation and also of H3K4me3, notably at several key regulators of haematopoiesis. Together, our results unveil a step-wise model, involving TET-OGT interactions, promotion of GlcNAcylation, and influence on H3K4me3 via SET1/COMPASS, highlighting a novel means by which TETs may induce transcriptional activation.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transcripción Genética , 5-Metilcitosina/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Islas de CpG , Citosina/análogos & derivados , Citosina/metabolismo , Epigénesis Genética , Glicosilación , Histonas/metabolismo , Factor C1 de la Célula Huésped/metabolismo , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética
9.
Pediatr Hematol Oncol ; 34(8): 425-427, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29303660

RESUMEN

We report the outcome of 27 children with de novo acute megakaryoblastic leukemia (AMKL) (excluding Down syndrome) enrolled in the French multicenter prospective study ELAM02 (2005-2011). There was no difference in gender, initial leukocyte count, CNS involvement, and complete remission rate (88.9%), as compared to other acute myeloid leukemia (AML) subtypes. AMKL patients had a significantly poorer outcome (5-year overall survival 54% [CI 95% 33%-71%] than children with other AML subtypes (5-year overall survival 73% [CI 95% 68%-77%] p = 0.02). Gender, age, CNS leukemia, hyperleukocytosis, complete remission or cytogenetic subgroups were not significant prognostic factors of disease-free survival. AMKL (excluding Down syndrom) remains an AML subgroup with inferior outcome.


Asunto(s)
Leucemia Megacarioblástica Aguda/mortalidad , Niño , Preescolar , Supervivencia sin Enfermedad , Síndrome de Down , Femenino , Francia/epidemiología , Humanos , Lactante , Leucemia Megacarioblástica Aguda/sangre , Leucemia Megacarioblástica Aguda/terapia , Masculino , Estudios Prospectivos , Tasa de Supervivencia
10.
Blood ; 121(13): 2440-51, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23335373

RESUMEN

The transcription factor Ikaros regulates the development of hematopoietic cells. Ikaros-deficient animals fail to develop B cells and display a T-cell malignancy, which is correlated with altered Notch signaling. Recently, loss of Ikaros was associated with progression of myeloproliferative neoplasms to acute myeloid leukemia and increasing evidence shows that Ikaros is also critical for the regulation of myeloid development. Previous studies showed that Ikaros-deficient mice have increased megakaryopoiesis, but the molecular mechanism of this phenomenon remains unknown. Here, we show that Ikaros overexpression decreases NOTCH-induced megakaryocytic specification, and represses expression of several megakaryocytic genes including GATA-1 to block differentiation and terminal maturation. We also demonstrate that Ikaros expression is differentially regulated by GATA-2 and GATA-1 during megakaryocytic differentiation and reveal that the combined loss of Ikzf1 and Gata1 leads to synthetic lethality in vivo associated with prominent defects in erythroid cells and an expansion of megakaryocyte progenitors. Taken together, our observations demonstrate an important functional interplay between Ikaros, GATA factors, and the NOTCH signaling pathway in specification and homeostasis of the megakaryocyte lineage.


Asunto(s)
Factor de Transcripción GATA1/metabolismo , Factor de Transcripción Ikaros/fisiología , Receptores Notch/metabolismo , Trombopoyesis/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Megacariocitos/metabolismo , Megacariocitos/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Unión Proteica/genética , Unión Proteica/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
12.
Stem Cells ; 32(8): 2084-97, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24723429

RESUMEN

Ten-eleven-translocation 2 (TET2) belongs to the TET protein family that catalyzes the conversion of 5-methylcytosine into 5-hydroxymethylcytosine and plays a central role in normal and malignant adult hematopoiesis. Yet the role of TET2 in human hematopoietic development remains largely unknown. Here, we show that TET2 expression is low in human embryonic stem cell (ESC) lines and increases during hematopoietic differentiation. shRNA-mediated TET2 knockdown had no effect on the pluripotency of various ESCs. However, it skewed their differentiation into neuroectoderm at the expense of endoderm and mesoderm both in vitro and in vivo. These effects were rescued by reintroducing the targeted TET2 protein. Moreover, TET2-driven differentiation was dependent on NANOG transcriptional factor. Indeed, TET2 bound to NANOG promoter and in TET2-deficient cells the methylation of the NANOG promoter correlated with a decreased in NANOG expression. The altered differentiation resulting from TET2 knockdown in ESCs led to a decrease in both the number and the cloning capacities of hematopoietic progenitors. These defects were due to an increased apoptosis and an altered gene expression profile, including abnormal expression of neuronal genes. Intriguingly, when TET2 was knockdown in hematopoietic cells, it increased hematopoietic development. In conclusion, our work suggests that TET2 is involved in different stages of human embryonic development, including induction of the mesoderm and hematopoietic differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Proteínas Proto-Oncogénicas/metabolismo , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Dioxigenasas , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Proteína Homeótica Nanog , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Blood ; 120(24): 4846-9, 2012 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-23074272

RESUMEN

Although a role for oncogenic KIT in driving mast cell disease is clear, the mechanisms driving the multiple phenotypic and clinical manifestations of this disorder are not well elucidated. We now show, using a large cohort of mastocytosis patients, including an almost equal number of aggressive and nonaggressive cases of systemic mastocytosis, that in contrast to the oncogenic KITD816V, TET2 mutation statistically associates with aggressive forms of the disease. By infecting primary murine bone marrow-derived mast cells with KITD816V, we also observe a significant and competitive growth advantage for KITD816V in Tet2-nullizygous compared with wild-type cells. TET2-deficient cells display increased proliferation and can survive in the absence of cytokines. Taken together, these data demonstrate a oncogenic cooperation in mast cells and reveal TET2 mutation as a potential marker to diagnose and predict severe forms of mastocytosis.


Asunto(s)
Proteínas de Unión al ADN/genética , Mastocitos/metabolismo , Mastocitosis/genética , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas/genética , Adulto , Anciano , Anciano de 80 o más Años , Sustitución de Aminoácidos , Animales , Células de la Médula Ósea/metabolismo , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Células Cultivadas , Estudios de Cohortes , Dioxigenasas , Femenino , Humanos , Masculino , Mastocitos/patología , Mastocitosis/patología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Factores de Tiempo , Transfección
14.
Cancer Cell ; 10(1): 65-75, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843266

RESUMEN

Tyrosine kinases are aberrantly activated in numerous malignancies, including acute myeloid leukemia (AML). To identify tyrosine kinases activated in AML, we developed a screening strategy that rapidly identifies tyrosine-phosphorylated proteins using mass spectrometry. This allowed the identification of an activating mutation (A572V) in the JAK3 pseudokinase domain in the acute megakaryoblastic leukemia (AMKL) cell line CMK. Subsequent analysis identified two additional JAK3 alleles, V722I and P132T, in AMKL patients. JAK3(A572V), JAK3(V722I), and JAK3(P132T) each transform Ba/F3 cells to factor-independent growth, and JAK3(A572V) confers features of megakaryoblastic leukemia in a murine model. These findings illustrate the biological importance of gain-of-function JAK3 mutations in leukemogenesis and demonstrate the utility of proteomic approaches to identifying clinically relevant mutations.


Asunto(s)
Leucemia Experimental/genética , Leucemia Megacarioblástica Aguda/genética , Proteínas Tirosina Quinasas/genética , Alelos , Animales , Apoptosis/efectos de los fármacos , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Mesilato de Imatinib , Janus Quinasa 2 , Janus Quinasa 3 , Células K562 , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Megacarioblástica Aguda/metabolismo , Leucemia Megacarioblástica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , TYK2 Quinasa
15.
Hemasphere ; 8(6): e90, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38903535

RESUMEN

Transcriptional cofactors of the ETO family are recurrent fusion partners in acute leukemia. We characterized the ETO2 regulome by integrating transcriptomic and chromatin binding analyses in human erythroleukemia xenografts and controlled ETO2 depletion models. We demonstrate that beyond its well-established repressive activity, ETO2 directly activates transcription of MYB, among other genes. The ETO2-activated signature is associated with a poorer prognosis in erythroleukemia but also in other acute myeloid and lymphoid leukemia subtypes. Mechanistically, ETO2 colocalizes with EP300 and MYB at enhancers supporting the existence of an ETO2/MYB feedforward transcription activation loop (e.g., on MYB itself). Both small-molecule and PROTAC-mediated inhibition of EP300 acetyltransferases strongly reduced ETO2 protein, chromatin binding, and ETO2-activated transcripts. Taken together, our data show that ETO2 positively enforces a leukemia maintenance program that is mediated in part by the MYB transcription factor and that relies on acetyltransferase cofactors to stabilize ETO2 scaffolding activity.

16.
Biochim Biophys Acta ; 1825(2): 173-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22240200

RESUMEN

The TET family of proteins has been described a few years ago. Only recently, their roles in DNA modification, through the oxidation of methyl-cytosine, and in normal and malignant development, through the description of TET2 as a tumor suppressor have been documented. The conjunction of these findings has prompted large efforts to understand the biology of these novel entities. Here, we summarize the recent results implicating TET2 in hematological malignancies suggesting that further studies are required to fully understand the role of DNA methylation alterations during transformation.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes Supresores de Tumor , Neoplasias Hematológicas/genética , Proteínas Proto-Oncogénicas/genética , Animales , Metilación de ADN , Dioxigenasas , Modelos Animales de Enfermedad , Epigenómica , Humanos
17.
Blood ; 118(5): 1264-73, 2011 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-21653327

RESUMEN

The NOTCH signaling pathway is implicated in a broad range of developmental processes, including cell fate decisions. However, the molecular basis for its role at the different steps of stem cell lineage commitment is unclear. We recently identified the NOTCH signaling pathway as a positive regulator of megakaryocyte lineage specification during hematopoiesis, but the developmental pathways that allow hematopoietic stem cell differentiation into the erythro-megakaryocytic lineages remain controversial. Here, we investigated the role of downstream mediators of NOTCH during megakaryopoiesis and report crosstalk between the NOTCH and PI3K/AKT pathways. We demonstrate the inhibitory role of phosphatase with tensin homolog and Forkhead Box class O factors on megakaryopoiesis in vivo. Finally, our data annotate developmental mechanisms in the hematopoietic system that enable a decision to be made either at the hematopoietic stem cell or the committed progenitor level to commit to the megakaryocyte lineage, supporting the existence of 2 distinct developmental pathways.


Asunto(s)
Diferenciación Celular , Linaje de la Célula/fisiología , Megacariocitos/fisiología , Proteína Oncogénica v-akt/metabolismo , Receptores Notch/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula/genética , Células Cultivadas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/fisiología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/fisiología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/fisiología , Receptor Cross-Talk/fisiología , Receptores Notch/genética , Receptores Notch/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Trombopoyesis/genética
18.
Haematologica ; 98(11): 1748-52, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23872306

RESUMEN

STAT3 protein phosphorylation is a frequent event in various hematologic malignancies and solid tumors. Acquired STAT3 mutations have been recently identified in 40% of patients with T-cell large granular lymphocytic leukemia, a rare T-cell disorder. In this study, we investigated the mutational status of STAT3 in a large series of patients with lymphoid and myeloid diseases. STAT3 mutations were identified in 1.6% (4 of 258) of patients with T-cell neoplasms, in 2.5% (2 of 79) of patients with diffuse large B-cell lymphoma but in no other B-cell lymphoma patients (0 of 104) or patients with myeloid malignancies (0 of 96). Functional in vitro assays indicated that the STAT3Y640F mutation leads to a constitutive phosphorylation of the protein. STA21, a STAT3 small molecule inhibitor, inhibited the proliferation of two distinct STAT3 mutated cell lines. Using a mouse bone marrow transplantation assay, we observed that STAT3Y640F expression leads to the development of myeloproliferative neoplasms with expansion of either myeloid cells or megakaryocytes. Together, these data indicate that the STAT3Y640F mutation leads to constitutive activation of STAT3, induces malignant hematopoiesis in vivo, and may represent a novel therapeutic target in some lymphoid malignancies.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Modelos Animales de Enfermedad , Neoplasias Hematológicas/genética , Mutación/genética , Trastornos Mieloproliferativos/genética , Factor de Transcripción STAT3/genética , Animales , Neoplasias Hematológicas/diagnóstico , Humanos , Células K562 , Ratones , Ratones Endogámicos C57BL , Trastornos Mieloproliferativos/diagnóstico
19.
Cancer Cell ; 7(4): 387-97, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15837627

RESUMEN

Polycythemia vera (PV), essential thrombocythemia (ET), and myeloid metaplasia with myelofibrosis (MMM) are clonal disorders arising from hematopoietic progenitors. An internet-based protocol was used to collect clinical information and biological specimens from patients with these diseases. High-throughput DNA resequencing identified a recurrent somatic missense mutation JAK2V617F in granulocyte DNA samples of 121 of 164 PV patients, of which 41 had homozygous and 80 had heterozygous mutations. Molecular and cytogenetic analyses demonstrated that homozygous mutations were due to duplication of the mutant allele. JAK2V617F was also identified in granulocyte DNA samples from 37 of 115 ET and 16 of 46 MMM patients, but was not observed in 269 normal individuals. In vitro analysis demonstrated that JAK2V617F is a constitutively active tyrosine kinase.


Asunto(s)
Mutación Missense/genética , Policitemia Vera/genética , Mielofibrosis Primaria/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Trombocitemia Esencial/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Activación Enzimática/genética , Femenino , Genotipo , Granulocitos/metabolismo , Heterocigoto , Homocigoto , Humanos , Janus Quinasa 2 , Masculino , Persona de Mediana Edad , Mitosis/genética , Modelos Moleculares , Mucosa Bucal/metabolismo , Fosforilación , Mielofibrosis Primaria/complicaciones , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Recombinación Genética/genética , Transfección
20.
Blood Cancer Discov ; 4(1): 2-4, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36538046

RESUMEN

In this issue of Blood Cancer Discovery, Kodgule, Goldman, Monovichet al. cleverly analyzed the transcription regulatory elements to investigate why the second copy of ETV6 is often lost in ETV6::RUNX1-translocated in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). It turns out that ETV6 suppresses the enhancer activity of GGAA microsatellite repeats, preventing ERG from subverting them to activate aberrant oncogene transcription. See related article by Kodgule, Goldman, Monovich et al., p. 34 (5).


Asunto(s)
Linfoma de Burkitt , Neoplasias Hematológicas , Leucemia de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Humanos , Activación Transcripcional , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Regulador Transcripcional ERG/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA