Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Blood ; 131(15): 1730-1742, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29453291

RESUMEN

Epigenetic regulators are recurrently mutated and aberrantly expressed in acute myeloid leukemia (AML). Targeted therapies designed to inhibit these chromatin-modifying enzymes, such as the histone demethylase lysine-specific demethylase 1 (LSD1) and the histone methyltransferase DOT1L, have been developed as novel treatment modalities for these often refractory diseases. A common feature of many of these targeted agents is their ability to induce myeloid differentiation, suggesting that multiple paths toward a myeloid gene expression program can be engaged to relieve the differentiation blockade that is uniformly seen in AML. We performed a comparative assessment of chromatin dynamics during the treatment of mixed lineage leukemia (MLL)-AF9-driven murine leukemias and MLL-rearranged patient-derived xenografts using 2 distinct but effective differentiation-inducing targeted epigenetic therapies, the LSD1 inhibitor GSK-LSD1 and the DOT1L inhibitor EPZ4777. Intriguingly, GSK-LSD1 treatment caused global gains in chromatin accessibility, whereas treatment with EPZ4777 caused global losses in accessibility. We captured PU.1 and C/EBPα motif signatures at LSD1 inhibitor-induced dynamic sites and chromatin immunoprecipitation coupled with high-throughput sequencing revealed co-occupancy of these myeloid transcription factors at these sites. Functionally, we confirmed that diminished expression of PU.1 or genetic deletion of C/EBPα in MLL-AF9 cells generates resistance of these leukemias to LSD1 inhibition. These findings reveal that pharmacologic inhibition of LSD1 represents a unique path to overcome the differentiation block in AML for therapeutic benefit.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Inhibidores Enzimáticos/farmacología , Histona Demetilasas/antagonistas & inhibidores , Leucemia Bifenotípica Aguda/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Leucemia Bifenotípica Aguda/genética , Leucemia Bifenotípica Aguda/metabolismo , Leucemia Bifenotípica Aguda/patología , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas Proto-Oncogénicas/genética , Elementos de Respuesta , Transactivadores/genética
2.
Nature ; 514(7523): 513-7, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25132549

RESUMEN

T-cell acute lymphoblastic leukaemia (T-ALL) is a haematological malignancy with a dismal overall prognosis, including a relapse rate of up to 25%, mainly because of the lack of non-cytotoxic targeted therapy options. Drugs that target the function of key epigenetic factors have been approved in the context of haematopoietic disorders, and mutations that affect chromatin modulators in a variety of leukaemias have recently been identified; however, 'epigenetic' drugs are not currently used for T-ALL treatment. Recently, we described that the polycomb repressive complex 2 (PRC2) has a tumour-suppressor role in T-ALL. Here we delineated the role of the histone 3 lysine 27 (H3K27) demethylases JMJD3 and UTX in T-ALL. We show that JMJD3 is essential for the initiation and maintenance of T-ALL, as it controls important oncogenic gene targets by modulating H3K27 methylation. By contrast, we found that UTX functions as a tumour suppressor and is frequently genetically inactivated in T-ALL. Moreover, we demonstrated that the small molecule inhibitor GSKJ4 (ref. 5) affects T-ALL growth, by targeting JMJD3 activity. These findings show that two proteins with a similar enzymatic function can have opposing roles in the context of the same disease, paving the way for treating haematopoietic malignancies with a new category of epigenetic inhibitors.


Asunto(s)
Histona Demetilasas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Animales , Benzazepinas/farmacología , Epigénesis Genética/efectos de los fármacos , Histona Demetilasas/genética , Histonas/química , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Lisina/metabolismo , Metilación/efectos de los fármacos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pirimidinas/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
3.
Cancer Cell ; 37(1): 71-84.e7, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31935373

RESUMEN

Cancer cells rely on altered metabolism to support abnormal proliferation. We performed a CRISPR/Cas9 functional genomic screen targeting metabolic enzymes and identified PDXK-an enzyme that produces pyridoxal phosphate (PLP) from vitamin B6-as an acute myeloid leukemia (AML)-selective dependency. PDXK kinase activity is required for PLP production and AML cell proliferation, and pharmacological blockade of the vitamin B6 pathway at both PDXK and PLP levels recapitulated PDXK disruption effects. PDXK disruption reduced intracellular concentrations of key metabolites needed for cell division. Furthermore, disruption of PLP-dependent enzymes ODC1 or GOT2 selectively inhibited AML cell proliferation and their downstream products partially rescued PDXK disruption induced proliferation blockage. Our work identifies the vitamin B6 pathway as a pharmacologically actionable dependency in AML.


Asunto(s)
Leucemia Mieloide Aguda/enzimología , Fosfotransferasas/metabolismo , Fosfato de Piridoxal/metabolismo , Vitamina B 6/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proliferación Celular , GTP Fosfohidrolasas/metabolismo , Regulación Leucémica de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Unión al GTP Monoméricas/metabolismo , Fosfotransferasas/genética , Fosfotransferasas (Aceptor de Grupo Alcohol) , Poliaminas/metabolismo , ARN Interferente Pequeño/metabolismo
4.
Cancer Discov ; 9(7): 962-979, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31068365

RESUMEN

Mutations in the TP53 tumor suppressor gene are common in many cancer types, including the acute myeloid leukemia (AML) subtype known as complex karyotype AML (CK-AML). Here, we identify a gain-of-function (GOF) Trp53 mutation that accelerates CK-AML initiation beyond p53 loss and, surprisingly, is required for disease maintenance. The Trp53R172H mutation (TP53R175H in humans) exhibits a neomorphic function by promoting aberrant self-renewal in leukemic cells, a phenotype that is present in hematopoietic stem and progenitor cells (HSPC) even prior to their transformation. We identify FOXH1 as a critical mediator of mutant p53 function that binds to and regulates stem cell-associated genes and transcriptional programs. Our results identify a context where mutant p53 acts as a bona fide oncogene that contributes to the pathogenesis of CK-AML and suggests a common biological theme for TP53 GOF in cancer. SIGNIFICANCE: Our study demonstrates how a GOF p53 mutant can hijack an embryonic transcription factor to promote aberrant self-renewal. In this context, mutant Trp53 functions as an oncogene to both initiate and sustain myeloid leukemia and suggests a potential convergent activity of mutant Trp53 across cancer types.This article is highlighted in the In This Issue feature, p. 813.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Mutación con Ganancia de Función , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Proteína p53 Supresora de Tumor/genética , Animales , Línea Celular Tumoral , Plasticidad de la Célula/genética , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Células Madre Neoplásicas/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Biotechnol ; 35(6): 577-582, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28459450

RESUMEN

Colorectal cancer (CRC) is a leading cause of death in the developed world, yet facile preclinical models that mimic the natural stages of CRC progression are lacking. Through the orthotopic engraftment of colon organoids we describe a broadly usable immunocompetent CRC model that recapitulates the entire adenoma-adenocarcinoma-metastasis axis in vivo. The engraftment procedure takes less than 5 minutes, shows efficient tumor engraftment in two-thirds of mice, and can be achieved using organoids derived from genetically engineered mouse models (GEMMs), wild-type organoids engineered ex vivo, or from patient-derived human CRC organoids. In this model, we describe the genotype and time-dependent progression of CRCs from adenocarcinoma (6 weeks), to local disseminated disease (11-12 weeks), and spontaneous metastasis (>20 weeks). Further, we use the system to show that loss of dysregulated Wnt signaling is critical for the progression of disseminated CRCs. Thus, our approach provides a fast and flexible means to produce tailored CRC mouse models for genetic studies and pre-clinical investigation.


Asunto(s)
Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Edición Génica/métodos , Genes Relacionados con las Neoplasias/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Trasplante de Órganos/métodos , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Femenino , Masculino , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia
6.
Nat Med ; 22(12): 1488-1495, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27841873

RESUMEN

Although the majority of patients with acute myeloid leukemia (AML) initially respond to chemotherapy, many of them subsequently relapse, and the mechanistic basis for AML persistence following chemotherapy has not been determined. Recurrent somatic mutations in DNA methyltransferase 3A (DNMT3A), most frequently at arginine 882 (DNMT3AR882), have been observed in AML and in individuals with clonal hematopoiesis in the absence of leukemic transformation. Patients with DNMT3AR882 AML have an inferior outcome when treated with standard-dose daunorubicin-based induction chemotherapy, suggesting that DNMT3AR882 cells persist and drive relapse. We found that Dnmt3a mutations induced hematopoietic stem cell expansion, cooperated with mutations in the FMS-like tyrosine kinase 3 gene (Flt3ITD) and the nucleophosmin gene (Npm1c) to induce AML in vivo, and promoted resistance to anthracycline chemotherapy. In patients with AML, the presence of DNMT3AR882 mutations predicts minimal residual disease, underscoring their role in AML chemoresistance. DNMT3AR882 cells showed impaired nucleosome eviction and chromatin remodeling in response to anthracycline treatment, which resulted from attenuated recruitment of histone chaperone SPT-16 following anthracycline exposure. This defect led to an inability to sense and repair DNA torsional stress, which resulted in increased mutagenesis. Our findings identify a crucial role for DNMT3AR882 mutations in driving AML chemoresistance and highlight the importance of chromatin remodeling in response to cytotoxic chemotherapy.


Asunto(s)
Antraciclinas/uso terapéutico , Ensamble y Desensamble de Cromatina/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Resistencia a Antineoplásicos/genética , Leucemia Mieloide Aguda/genética , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular , ADN Metiltransferasa 3A , Daunorrubicina/uso terapéutico , Células Madre Hematopoyéticas , Humanos , Immunoblotting , Inmunoprecipitación , Leucemia Mieloide Aguda/tratamiento farmacológico , Espectrometría de Masas , Ratones , Mutación , Proteínas Nucleares/genética , Nucleofosmina , Nucleosomas/metabolismo , Tirosina Quinasa 3 Similar a fms/genética
7.
Cancer Cell ; 23(3): 362-75, 2013 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-23518350

RESUMEN

The molecular mechanisms regulating leukemia-initiating cell (LIC) function are of important clinical significance. We use chronic myelogenous leukemia (CML) as a model of LIC-dependent malignancy and identify the interaction between the ubiquitin ligase Fbw7 and its substrate c-Myc as a regulator of LIC homeostasis. Deletion of Fbw7 leads to c-Myc overexpression, p53-dependent LIC-specific apoptosis, and the eventual inhibition of tumor progression. A decrease of either c-Myc protein levels or attenuation of the p53 response rescues LIC activity and disease progression. Further experiments showed that Fbw7 expression is required for survival and maintenance of human CML LIC. These studies identify a ubiquitin ligase:substrate pair regulating LIC activity, suggesting that targeting of the Fbw7:c-Myc axis is an attractive therapy target in refractory CML.


Asunto(s)
Proteínas F-Box/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Progresión de la Enfermedad , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Regulación Neoplásica de la Expresión Génica , Células Madre Hematopoyéticas , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
Cell Stem Cell ; 13(2): 190-204, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23791481

RESUMEN

Accumulating evidence suggests that Notch signaling is active at multiple points during hematopoiesis. Until recently, the majority of such studies focused on Notch signaling in lymphocyte differentiation and knowledge of individual Notch receptor roles has been limited due to a paucity of genetic tools available. In this manuscript we generate and describe animal models to identify and fate-map stem and progenitor cells expressing each Notch receptor, delineate Notch pathway activation, and perform in vivo gain- and loss-of-function studies dissecting Notch signaling in early hematopoiesis. These models provide comprehensive genetic maps of lineage-specific Notch receptor expression and activation in hematopoietic stem and progenitor cells. Moreover, they establish a previously unknown role for Notch signaling in the commitment of blood progenitors toward the erythrocytic lineage and link Notch signaling to optimal organismal response to stress erythropoiesis.


Asunto(s)
Hematopoyesis , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Transducción de Señal , Estrés Fisiológico , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , División Celular , Linaje de la Célula/genética , Células Eritroides/citología , Células Eritroides/metabolismo , Feto/citología , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Modelos Biológicos , Estrés Fisiológico/genética , Factor de Transcripción HES-1 , Transcripción Genética
9.
Cell Stem Cell ; 11(6): 783-98, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23103054

RESUMEN

Although transcriptional regulation of stem cell pluripotency and differentiation has been extensively studied, only a small number of studies have addressed the roles for posttranslational modifications in these processes. A key mechanism of posttranslational modification is ubiquitination by the ubiquitin-proteasome system (UPS). Here, using shotgun proteomics, we map the ubiquitinated protein landscape during embryonic stem cell (ESC) differentiation and induced pluripotency. Moreover, using UPS-targeted RNAi screens, we identify additional regulators of pluripotency and differentiation. We focus on two of these proteins, the deubiquitinating enzyme Psmd14 and the E3 ligase Fbxw7, and characterize their importance in ESC pluripotency and cellular reprogramming. This global characterization of the UPS as a key regulator of stem cell pluripotency opens the way for future studies that focus on specific UPS enzymes or ubiquitinated substrates.


Asunto(s)
Reprogramación Celular/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Semivida , Ratones , Estabilidad Proteica , Proteolisis , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/metabolismo , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA