Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Blood ; 127(19): 2322-6, 2016 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-26944543

RESUMEN

Inhibitor of DNA binding 1 (Id1) functions as an E protein inhibitor, and overexpression of Id1 is seen in acute myeloid leukemia (AML) patients. To define the effects of Id1 on leukemogenesis, we expressed MLL-AF9 in fetal liver (FL) cells or bone marrow (BM) cells isolated from wild-type, Id1(-/-), p21(-/-), or Id1(-/-)p21(-/-) mice, and transplanted them into syngeneic recipient mice. We found that although mice receiving MLL-AF9-transduced FL or BM cells develop AML, loss of Id1 significantly prolonged the median survival of mice receiving FL cells but accelerated leukemogenesis in recipients of BM cells. Deletion of Cdkn1a (p21), an Id1 target gene, can rescue the effect of Id1 loss in both models, suggesting that Cdkn1a is a critical target of Id1 in leukemogenesis. It has been suggested that the FL transplant model mimics human fetal-origin (infant) MLL fusion protein (FP)-driven leukemia, whereas the BM transplantation model resembles postnatal MLL leukemia; in fact, the analysis of clinical samples from patients with MLL-FP(+) leukemia showed that Id1 expression is elevated in the former and reduced in the latter type of MLL-FP(+) AML. Our findings suggest that Id1 could be a potential therapeutic target for infant MLL-AF9-driven leukemia.


Asunto(s)
Proteína 1 Inhibidora de la Diferenciación/metabolismo , Neoplasias Experimentales/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Animales , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
2.
Blood ; 126(5): 640-50, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26084673

RESUMEN

Transcriptional regulators are recurrently altered through translocations, deletions, or aberrant expression in acute myeloid leukemia (AML). Although critically important in leukemogenesis, the underlying pathogenetic mechanisms they trigger remain largely unknown. Here, we identified that Id1 (inhibitor of DNA binding 1) plays a pivotal role in acute myeloid leukemogenesis. Using genetically modified mice, we found that loss of Id1 inhibited t(8;21) leukemia initiation and progression in vivo by abrogating protein kinase B (AKT)1 activation, and that Id1 interacted with AKT1 through its C terminus. An Id1 inhibitor impaired the in vitro growth of AML cells and, when combined with an AKT inhibitor, triggered even greater apoptosis and growth inhibition, whereas normal hematopoietic stem/progenitor cells were largely spared. We then performed in vivo experiments and found that the Id1 inhibitor significantly prolonged the survival of t(8;21)(+) leukemic mice, whereas overexpression of activated AKT1 promoted leukemogenesis. Thus, our results establish Id1/Akt1 signaling as a potential therapeutic target in t(8;21) leukemia.


Asunto(s)
Proteína 1 Inhibidora de la Diferenciación/metabolismo , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis , Carcinogénesis , Línea Celular Tumoral , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Proteína 1 Inhibidora de la Diferenciación/deficiencia , Proteína 1 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/antagonistas & inhibidores , Proteínas Inhibidoras de la Diferenciación/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal , Translocación Genética
3.
Blood Cells Mol Dis ; 43(1): 30-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19386523

RESUMEN

In this report we review the current knowledge of the interaction of RUNX1(AML1) with serine/threonine kinases, lysine and arginine methyltransferases, lysine acetyltransferases, and histone deacetylases. We also discuss the effect of RUNX1-ETO fusion gene on DNA methylation. RUNX1 post-transcriptional modification can affect its role in influencing differentiation and self-renewal of hematopoietic cells. The goal of these studies is to develop targets for improved leukemia therapy.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Procesamiento Proteico-Postraduccional , Acetiltransferasas/metabolismo , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Metilación de ADN , Histona Desacetilasas/metabolismo , Humanos , Metiltransferasas/metabolismo
4.
Cancer Discov ; 9(12): 1686-1695, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31575540

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide with no clinically confirmed oncogenic driver. Although preclinical studies implicate the FGF19 receptor FGFR4 in hepatocarcinogenesis, the dependence of human cancer on FGFR4 has not been demonstrated. Fisogatinib (BLU-554) is a potent and selective inhibitor of FGFR4 and demonstrates clinical benefit and tumor regression in patients with HCC with aberrant FGF19 expression. Mutations were identified in the gatekeeper and hinge-1 residues in the kinase domain of FGFR4 upon disease progression in 2 patients treated with fisogatinib, which were confirmed to mediate resistance in vitro and in vivo. A gatekeeper-agnostic, pan-FGFR inhibitor decreased HCC xenograft growth in the presence of these mutations, demonstrating continued FGF19-FGFR4 pathway dependence. These results validate FGFR4 as an oncogenic driver and warrant further therapeutic targeting of this kinase in the clinic. SIGNIFICANCE: Our study is the first to demonstrate on-target FGFR4 kinase domain mutations as a mechanism of acquired clinical resistance to targeted therapy. This further establishes FGF19-FGFR4 pathway activation as an oncogenic driver. These findings support further investigation of fisogatinib in HCC and inform the profile of potential next-generation inhibitors.See related commentary by Subbiah and Pal, p. 1646.This article is highlighted in the In This Issue feature, p. 1631.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico por imagen , Resistencia a Antineoplásicos , Neoplasias Hepáticas/diagnóstico por imagen , Piranos/farmacología , Quinazolinas/farmacología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Anciano de 80 o más Años , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Modelos Moleculares , Mutación , Trasplante de Neoplasias , Dominios Proteicos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo
5.
Sci Rep ; 6: 22760, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26961797

RESUMEN

Multiple myeloma is a plasma cell neoplasm with an extremely variable clinical course. Animal models are needed to better understand its pathophysiology and for preclinical testing of potential therapeutic agents. Hematopoietic cells expressing the hypermorphic Rad50(s) allele show hematopoietic failure, which can be mitigated by the lack of a transcription factor, Mef/Elf4. However, we find that 70% of Mef(-/-)Rad50(s/s) mice die from multiple myeloma or other plasma cell neoplasms. These mice initially show an abnormal plasma cell proliferation and monoclonal protein production, and then develop anemia and a decreased bone mineral density. Tumor cells can be serially transplanted and according to array CGH and whole exome sequencing, the pathogenesis of plasma cell neoplasms in these mice is not linked to activation of a specific oncogene, or inactivation of a specific tumor suppressor. This model recapitulates the systemic manifestations of human plasma cell neoplasms, and implicates cooperativity between the Rad50(s) and Mef/Elf4 pathways in initiating myelomagenic mutations that promote plasma cell transformation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Proteínas de Unión al ADN/genética , Mieloma Múltiple/genética , Fenotipo , Factores de Transcripción/genética , Ácido Anhídrido Hidrolasas , Animales , Células Sanguíneas/metabolismo , Células Sanguíneas/patología , Células Sanguíneas/fisiología , Células Sanguíneas/trasplante , Densidad Ósea , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Exoma , Ratones , Ratones Endogámicos C57BL , Mieloma Múltiple/patología
6.
J Exp Med ; 213(1): 25-34, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26666262

RESUMEN

t(8;21) is one of the most frequent chromosomal abnormalities observed in acute myeloid leukemia (AML). However, expression of AML1-ETO is not sufficient to induce transformation in vivo. Consistent with this observation, patients with this translocation harbor additional genetic abnormalities, suggesting a requirement for cooperating mutations. To better define the genetic landscape in AML and distinguish driver from passenger mutations, we compared the mutational profiles of AML1-ETO-driven mouse models of leukemia with the mutational profiles of human AML patients. We identified TET2 and PTPN11 mutations in both mouse and human AML and then demonstrated the ability of Tet2 loss and PTPN11 D61Y to initiate leukemogenesis in concert with expression of AML1-ETO in vivo. This integrative genetic profiling approach allowed us to accurately predict cooperating events in t(8;21)(+) AML in a robust and unbiased manner, while also revealing functional convergence in mouse and human AML.


Asunto(s)
Alelos , Epistasis Genética , Genómica/métodos , Leucemia Mieloide Aguda/genética , Animales , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Regulación Leucémica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Mutación , Proteínas de Fusión Oncogénica/genética , Fenotipo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína 1 Compañera de Translocación de RUNX1 , Translocación Genética
7.
Cancer Cell ; 27(4): 502-15, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25873173

RESUMEN

Specific combinations of acute myeloid leukemia (AML) disease alleles, including FLT3 and TET2 mutations, confer distinct biologic features and adverse outcome. We generated mice with mutations in Tet2 and Flt3, which resulted in fully penetrant, lethal AML. Multipotent Tet2(-/-);Flt3(ITD) progenitors (LSK CD48(+)CD150(-)) propagate disease in secondary recipients and were refractory to standard AML chemotherapy and FLT3-targeted therapy. Flt3(ITD) mutations and Tet2 loss cooperatively remodeled DNA methylation and gene expression to an extent not seen with either mutant allele alone, including at the Gata2 locus. Re-expression of Gata2 induced differentiation in AML stem cells and attenuated leukemogenesis. TET2 and FLT3 mutations cooperatively induce AML, with a defined leukemia stem cell population characterized by site-specific changes in DNA methylation and gene expression.


Asunto(s)
Proteínas de Unión al ADN/genética , Epigénesis Genética , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas/genética , Tirosina Quinasa 3 Similar a fms/genética , Antineoplásicos/uso terapéutico , Diferenciación Celular/genética , Citarabina/uso terapéutico , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Doxorrubicina/uso terapéutico , Factor de Transcripción GATA2/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Haploinsuficiencia , Mutación , Proteínas Proto-Oncogénicas/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo
8.
Front Med ; 6(3): 248-62, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22875638

RESUMEN

The AML1-ETO fusion transcription factor is generated by the t(8;21) translocation, which is present in approximately 4%-12% of adult and 12%-30% of pediatric acute myeloid leukemia (AML) patients. Both human and mouse models of AML have demonstrated that AML1-ETO is insufficient for leukemogenesis in the absence of secondary events. In this review, we discuss the pathogenetic insights that have been gained from identifying the various events that can cooperate with AML1-ETO to induce AML in vivo. We also discuss potential therapeutic strategies for t(8;21) positive AML that involve targeting the fusion protein itself, the proteins that bind to it, or the genes that it regulates. Recently published studies suggest that a targeted therapy for t(8;21) positive AML is feasible and may be coming sometime soon.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Proteínas de Fusión Oncogénica/genética , Factores de Transcripción/genética , Translocación Genética , Animales , Biomarcadores de Tumor/análisis , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Modelos Animales de Enfermedad , Regulación Leucémica de la Expresión Génica , Genotipo , Humanos , Ratones , Ratones Transgénicos , Fenotipo , Proteína 1 Compañera de Translocación de RUNX1
9.
Cancer Cell ; 19(2): 283-94, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21316606

RESUMEN

The JAK2V617F constitutively activated tyrosine kinase is found in most patients with myeloproliferative neoplasms. While examining the interaction between JAK2 and PRMT5, an arginine methyltransferase originally identified as JAK-binding protein 1, we found that JAK2V617F (and JAK2K539L) bound PRMT5 more strongly than did wild-type JAK2. These oncogenic kinases also acquired the ability to phosphorylate PRMT5, greatly impairing its ability to methylate its histone substrates, and representing a specific gain-of-function that allows them to regulate chromatin modifications. We readily detected PRMT5 phosphorylation in JAK2V617F-positive patient samples, and when we knocked down PRMT5 in human CD34+ cells using shRNA, we observed increased colony formation and erythroid differentiation. These results indicate that phosphorylation of PRMT5 contributes to the mutant JAK2-induced myeloproliferative phenotype.


Asunto(s)
Regulación hacia Abajo , Janus Quinasa 2/metabolismo , Proteína Metiltransferasas/metabolismo , Línea Celular , Humanos , Janus Quinasa 2/genética , Mutación , Trastornos Mieloproliferativos , Fosforilación , Proteína-Arginina N-Metiltransferasas , Especificidad por Sustrato
10.
Science ; 333(6043): 765-9, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21764752

RESUMEN

The chromosomal translocations found in acute myelogenous leukemia (AML) generate oncogenic fusion transcription factors with aberrant transcriptional regulatory properties. Although therapeutic targeting of most leukemia fusion proteins remains elusive, the posttranslational modifications that control their function could be targetable. We found that AML1-ETO, the fusion protein generated by the t(8;21) translocation, is acetylated by the transcriptional coactivator p300 in leukemia cells isolated from t(8;21) AML patients, and that this acetylation is essential for its self-renewal-promoting effects in human cord blood CD34(+) cells and its leukemogenicity in mouse models. Inhibition of p300 abrogates the acetylation of AML1-ETO and impairs its ability to promote leukemic transformation. Thus, lysine acetyltransferases represent a potential therapeutic target in AML.


Asunto(s)
Transformación Celular Neoplásica , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda/metabolismo , Lisina/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Acetilación , Animales , Línea Celular , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Sangre Fetal/citología , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , Proteínas de Fusión Oncogénica/química , Preleucemia/metabolismo , Preleucemia/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteína 1 Compañera de Translocación de RUNX1 , Activación Transcripcional , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA