Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Sci Rep ; 11(1): 13158, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162911

RESUMEN

Post-transcriptional gene regulation, including that by RNA binding proteins (RBPs), has recently been described as an important mechanism in cancer. We had previously identified a set of RBPs that were highly dysregulated in B-cell acute lymphoblastic leukemia (B-ALL) with MLL translocations, which carry a poor prognosis. Here, we sought to functionally characterize these dysregulated RBP genes by performing a focused CRISPR dropout screen in B-ALL cell lines, finding dependencies on several genes including EIF3E, EPRS and USO1. Validating our findings, CRISPR/Cas9-mediated disruption of USO1 in MLL-translocated B-ALL cells reduced cell growth, promoted cell death, and altered the cell cycle. Transcriptomic analysis of USO1-deficient cells revealed alterations in pathways related to mTOR signaling, RNA metabolism, and targets of MYC. In addition, USO1-regulated genes from these experimental samples were significantly and concordantly correlated with USO1 expression in primary samples collected from B-ALL patients. Lastly, we found that loss of Uso1 inhibited colony formation of MLL-transformed in primary bone marrow cells from Cas9-EGFP mice. Together, our findings demonstrate an approach to performing focused sub-genomic CRISPR screens and highlight a putative RBP vulnerability in MLL-translocated B-ALL, thus identifying potential therapeutic targets in this disease.


Asunto(s)
Sistemas CRISPR-Cas , Proteínas de la Matriz de Golgi/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Neoplasias/fisiología , Proteínas de Fusión Oncogénica/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas de Transporte Vesicular/fisiología , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Regulación Leucémica de la Expresión Génica , Genes Reporteros , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Proteínas de la Matriz de Golgi/genética , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/genética , Procesamiento Postranscripcional del ARN , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Transgenes , Translocación Genética , Ensayo de Tumor de Célula Madre , Proteínas de Transporte Vesicular/genética
2.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34039707

RESUMEN

Specified intestinal epithelial cells reprogram and contribute to the regeneration and renewal of the epithelium upon injury. Mutations that deregulate such renewal processes may contribute to tumorigenesis. Using intestinal organoids, we show that concomitant activation of Notch signaling and ablation of p53 induce a highly proliferative and regenerative cell state, which is associated with increased levels of Yap and the histone methyltransferase Mll1. The induced signaling system orchestrates high proliferation, self-renewal, and niche-factor-independent growth, and elevates the trimethylation of histone 3 at lysine 4 (H3K4me3). We demonstrate that Yap and Mll1 are also elevated in patient-derived colorectal cancer (CRC) organoids and control growth and viability. Our data suggest that Notch activation and p53 ablation induce a signaling circuitry involving Yap and the epigenetic regulator Mll1, which locks cells in a proliferative and regenerative state that renders them susceptible for tumorigenesis.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Mutación , Organoides/metabolismo , Factores de Transcripción/metabolismo
3.
Biochim Biophys Acta Gene Regul Mech ; 1863(8): 194579, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32389825

RESUMEN

As demonstrated through early work in Drosophila, members of the MLL/SET family play essential roles during embryonic development through their participation in large protein complexes that are central to epigenetic regulation of gene expression. One of its members, MLL1, has additionally received a lot of attention as it is a potent oncogenic driver in different types of leukaemia when aberrantly fused to a large variety of partners as a result of chromosomal translocations. Its exclusive association with cancers of the haematopoietic system has prompted a large number of investigations into the role of MLL/SET proteins in haematopoiesis, a summary of which was attempted in this review. Interestingly, MLL-rearranged leukaemias are particularly prominent in infant and paediatric leukaemia, which commonly initiate in utero. This, together with the known function of MLL/SET proteins in embryonic development, has focussed research efforts in recent years on understanding the role of this protein family in developmental haematopoiesis and how this may be subverted by MLL oncofusions in infant leukaemia. A detailed understanding of these prenatal events is essential for the development of new treatments that improve the survival specifically of this very young patient group.


Asunto(s)
Hematopoyesis/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Animales , Niño , Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Epigénesis Genética , Hematopoyesis/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Lactante , Leucemia , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/metabolismo
4.
Blood ; 136(11): 1303-1316, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32458004

RESUMEN

Metabolic alterations in cancer represent convergent effects of oncogenic mutations. We hypothesized that a metabolism-restricted genetic screen, comparing normal primary mouse hematopoietic cells and their malignant counterparts in an ex vivo system mimicking the bone marrow microenvironment, would define distinctive vulnerabilities in acute myeloid leukemia (AML). Leukemic cells, but not their normal myeloid counterparts, depended on the aldehyde dehydrogenase 3a2 (Aldh3a2) enzyme that oxidizes long-chain aliphatic aldehydes to prevent cellular oxidative damage. Aldehydes are by-products of increased oxidative phosphorylation and nucleotide synthesis in cancer and are generated from lipid peroxides underlying the non-caspase-dependent form of cell death, ferroptosis. Leukemic cell dependence on Aldh3a2 was seen across multiple mouse and human myeloid leukemias. Aldh3a2 inhibition was synthetically lethal with glutathione peroxidase-4 (GPX4) inhibition; GPX4 inhibition is a known trigger of ferroptosis that by itself minimally affects AML cells. Inhibiting Aldh3a2 provides a therapeutic opportunity and a unique synthetic lethality to exploit the distinctive metabolic state of malignant cells.


Asunto(s)
Aldehído Oxidorreductasas/fisiología , Carbolinas/farmacología , Ciclohexilaminas/farmacología , Ferroptosis/efectos de los fármacos , Hematopoyesis/fisiología , Leucemia Mieloide Aguda/enzimología , Proteínas de Neoplasias/fisiología , Fenilendiaminas/farmacología , Aldehído Oxidorreductasas/genética , Aldehídos/farmacología , Animales , Línea Celular Tumoral , Citarabina/administración & dosificación , Doxorrubicina/administración & dosificación , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Peroxidación de Lípido , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Ácido Oléico/farmacología , Proteínas de Fusión Oncogénica/fisiología , Oxidación-Reducción , Estrés Oxidativo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , Fosfolípido Hidroperóxido Glutatión Peroxidasa/fisiología
5.
Cancer Cell ; 37(4): 599-617.e7, 2020 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-32243837

RESUMEN

Epigenetic modifiers frequently harbor loss-of-function mutations in lung cancer, but their tumor-suppressive roles are poorly characterized. Histone methyltransferase KMT2D (a COMPASS-like enzyme, also called MLL4) is among the most highly inactivated epigenetic modifiers in lung cancer. Here, we show that lung-specific loss of Kmt2d promotes lung tumorigenesis in mice and upregulates pro-tumorigenic programs, including glycolysis. Pharmacological inhibition of glycolysis preferentially impedes tumorigenicity of human lung cancer cells bearing KMT2D-inactivating mutations. Mechanistically, Kmt2d loss widely impairs epigenomic signals for super-enhancers/enhancers, including the super-enhancer for the circadian rhythm repressor Per2. Loss of Kmt2d decreases expression of PER2, which regulates multiple glycolytic genes. These findings indicate that KMT2D is a lung tumor suppressor and that KMT2D deficiency confers a therapeutic vulnerability to glycolytic inhibitors.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Proteínas de Unión al ADN/antagonistas & inhibidores , Desoxiglucosa/farmacología , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Glucólisis , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Neoplasias/antagonistas & inhibidores , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/metabolismo , Animales , Antimetabolitos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Science ; 368(6486): 48-53, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32241942

RESUMEN

Neural stem cells (NSCs) in the developing and postnatal brain have distinct positional identities that dictate the types of neurons they generate. Although morphogens initially establish NSC positional identity in the neural tube, it is unclear how such regional differences are maintained as the forebrain grows much larger and more anatomically complex. We found that the maintenance of NSC positional identity in the murine brain requires a mixed-lineage leukemia 1 (Mll1)-dependent epigenetic memory system. After establishment by sonic hedgehog, ventral NSC identity became independent of this morphogen. Even transient MLL1 inhibition caused a durable loss of ventral identity, resulting in the generation of neurons with the characteristics of dorsal NSCs in vivo. Thus, spatial information provided by morphogens can be transitioned to epigenetic mechanisms that maintain regionally distinct developmental programs in the forebrain.


Asunto(s)
Impresión Genómica , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/genética , Prosencéfalo/citología , Prosencéfalo/embriología , Factor Nuclear Tiroideo 1/genética , Animales , Proteínas Hedgehog/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Ratones , Ratones Mutantes , Proteína de la Leucemia Mieloide-Linfoide/genética , Células-Madre Neurales/citología , Transcriptoma
7.
Blood ; 136(1): 81-92, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32299104

RESUMEN

Through a clustered regularly insterspaced short palindromic repeats (CRISPR) screen to identify mitochondrial genes necessary for the growth of acute myeloid leukemia (AML) cells, we identified the mitochondrial outer membrane protein mitochondrial carrier homolog 2 (MTCH2). In AML, knockdown of MTCH2 decreased growth, reduced engraftment potential of stem cells, and induced differentiation. Inhibiting MTCH2 in AML cells increased nuclear pyruvate and pyruvate dehydrogenase (PDH), which induced histone acetylation and subsequently promoted the differentiation of AML cells. Thus, we have defined a new mechanism by which mitochondria and metabolism regulate AML stem cells and gene expression.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Proteínas de Neoplasias/fisiología , Acetilación , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Sangre Fetal/citología , Regulación Leucémica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Fusión Oncogénica/fisiología , Procesamiento Proteico-Postraduccional , Ácido Pirúvico/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
8.
Nat Genet ; 52(1): 95-105, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31844322

RESUMEN

Around implantation, the epiblast (Epi) transits from naïve to primed pluripotency, before giving rise to the three germ layers. How chromatin is reconfigured during this developmental window remains poorly understood. We performed a genome-wide investigation of chromatin landscapes during this period. We find that enhancers in ectoderm are already pre-accessible in embryonic day 6.5 (E6.5) Epi when cells enter a primed pluripotent state. Unexpectedly, strong trimethylation of histone H3 at lysine 4 (H3K4me3) emerges at developmental gene promoters in E6.5 Epi and positively correlates with H3K27me3, thus establishing bivalency. These genes also show enhanced spatial interactions. Both the strong bivalency and spatial clustering are virtually absent in preimplantation embryos and are markedly reduced in fate-committed lineages. Finally, we show that KMT2B is essential for establishing bivalent H3K4me3 at E6.5 but becomes partially dispensable later. Its deficiency leads to impaired activation of developmental genes and subsequent embryonic lethality. Thus, our data characterize lineage-specific chromatin reconfiguration and a unique chromatin state for primed pluripotency.


Asunto(s)
Cromatina/genética , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Epigenómica/métodos , Gastrulación , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Animales , Metilación de ADN , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Femenino , Histonas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Sci Rep ; 9(1): 20084, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882723

RESUMEN

Acute leukaemias express high levels of MYB which are required for the initiation and maintenance of the disease. Inhibition of MYB expression or activity has been shown to suppress MLL-fusion oncoprotein-induced acute myeloid leukaemias (AML), which are among the most aggressive forms of AML, and indeed MYB transcription has been reported to be regulated by the MLL-AF9 oncoprotein. This highlights the importance of understanding the mechanism of MYB transcriptional regulation in these leukaemias. Here we have demonstrated that the MLL-AF9 fusion protein regulates MYB transcription directly at the promoter region, in part by recruiting the transcriptional regulator kinase CDK9, and CDK9 inhibition effectively suppresses MYB expression as well as cell proliferation. However, MYB regulation by MLL-AF9 does not require H3K79 methylation mediated by the methyltransferase DOT1L, which has also been shown to be a key mediator of MLL-AF9 leukemogenicity. The identification of specific, essential and druggable transcriptional regulators may enable effective targeting of MYB expression, which in turn could potentially lead to new therapeutic approaches for acute myeloid leukaemia with MLL-AF9.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Genes myb , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Fusión Oncogénica/fisiología , Transcripción Genética , Animales , Humanos , Leucemia Mieloide Aguda/patología , Regiones Promotoras Genéticas
10.
J Biol Chem ; 294(51): 19667-19682, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31744881

RESUMEN

Estrogen and its cognate receptor, ERα, regulate cell proliferation, differentiation, and carcinogenesis in the endometrium by controlling gene transcription. ERα requires co-activators to mediate transcription via mechanisms that are largely uncharacterized. Herein, using growth-regulating estrogen receptor binding 1 (GREB1) as an ERα target gene in Ishikawa cells, we demonstrate that nuclear receptor co-activator 6 (NCOA6) is essential for estradiol (E2)/ERα-activated GREB1 transcription. We found that NCOA6 associates with the GREB1 promoter and enhancer in an E2-independent manner and that NCOA6 knockout reduces chromatin looping, enhancer-promoter interactions, and basal GREB1 expression in the absence of E2. In the presence of E2, ERα bound the GREB1 enhancer and also associated with its promoter, and p300, myeloid/lymphoid or mixed-lineage leukemia protein 4 (MLL4), and RNA polymerase II were recruited to the GREB1 enhancer and promoter. Consequently, the levels of the histone modifications H3K4me1/3, H3K9ac, and H3K27ac were significantly increased; enhancer and promoter regions were transcribed; and GREB1 mRNA was robustly transcribed. NCOA6 knockout reduced ERα recruitment and abolished all of the aforementioned E2-induced events, making GREB1 completely insensitive to E2 induction. We also found that GREB1-deficient Ishikawa cells are much more resistant to chemotherapy and that human endometrial cancers with low GREB1 expression predict poor overall survival. These results indicate that NCOA6 has an essential role in ERα-mediated transcription by increasing enhancer-promoter interactions through chromatin looping and by recruiting RNA polymerase II and the histone-code modifiers p300 and MLL4. Moreover, GREB1 loss may predict chemoresistance of endometrial cancer.


Asunto(s)
Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/fisiología , Estrógenos/farmacología , Proteínas de Neoplasias/fisiología , Coactivadores de Receptor Nuclear/fisiología , Regiones Promotoras Genéticas , Antineoplásicos/farmacología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Proteína p300 Asociada a E1A/fisiología , Receptor alfa de Estrógeno/genética , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/química , Humanos , Proteína de la Leucemia Mieloide-Linfoide/fisiología
11.
Endocrinology ; 160(8): 1926-1936, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31211356

RESUMEN

Menin is the protein mutated in patients with multiple endocrine neoplasia type 1 (MEN1) syndrome and their corresponding sporadic tumor counterparts. We have found that menin functions in promoting proper cell division. Here, we show that menin localizes to the mitotic spindle poles and the mitotic spindle during early mitosis and to the intercellular bridge microtubules during cytokinesis in HeLa cells. In our study, menin depletion led to defects in spindle assembly and chromosome congression during early mitosis, lagging chromosomes during anaphase, defective cytokinesis, multinucleated interphase cells, and cell death. In addition, pharmacological inhibition of the menin-MLL1 interaction also led to similar cell division defects. These results indicate that menin and the menin-MLL1 interaction are important for proper cell division. These results highlight a function for menin in cell division and aid our understanding of how mutation and misregulation of menin promotes tumorigenesis.


Asunto(s)
División Celular , Proteínas Proto-Oncogénicas/fisiología , Huso Acromático/fisiología , Células HCT116 , Células HeLa , N-Metiltransferasa de Histona-Lisina/fisiología , Humanos , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas Proto-Oncogénicas/genética
12.
BMJ Case Rep ; 12(2)2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30772833

RESUMEN

We describe an unusual case of pre-B lymphoblastic leukaemia presenting with a unilateral maxillary sinus mass in which biopsies of the primary mass and the bone marrow demonstrated conflicting immunophenotyping results. The extramedullary mass was consistent with a precursor B-cell malignancy, while the bone marrow was initially reported as a possible mature B-cell malignancy. The treatments for the two are fundamentally different, which necessitated a delay in the initiation of his chemotherapy until a clear diagnosis was made. Mixed lineage leukaemia gene rearrangement was confirmed by fluorescence in situ hybridisation in both the primary mass and bone marrow, which unified the diagnosis as pre-B acute lymphoblastic leukaemia given the common cytogenetic feature.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/fisiología , Neoplasias del Seno Maxilar/patología , Seno Maxilar/patología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Células Precursoras de Linfocitos B/patología , Protocolos de Quimioterapia Combinada Antineoplásica , Médula Ósea/patología , Preescolar , Citometría de Flujo , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Masculino , Neoplasias del Seno Maxilar/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Inducción de Remisión
13.
Int J Hematol ; 110(2): 170-178, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30706327

RESUMEN

Polycomb proteins function in the maintenance of gene silencing via post-translational modifications of histones and chromatin compaction. Genetic and biochemical studies have revealed that the repressive function of Polycomb repressive complexes (PRCs) in transcription is counteracted by the activating function of Trithorax-group complexes; this balance fine-tunes the expression of genes critical for development and tissue homeostasis. The function of PRCs is frequently dysregulated in various cancer cells due to altered expression or recurrent somatic mutations in PRC genes. The tumor suppressive functions of EZH2-containing PRC2 and a PRC2-related protein ASXL1 have been investigated extensively in the pathogenesis of hematological malignancies, including myeloproliferative neoplasms (MPN). BCOR, a component of non-canonical PRC1, suppresses various hematological malignancies including MPN. In this review, we focus on recent findings on the role of PRCs in the pathogenesis of MPN and the therapeutic impact of targeting the pathological functions of PRCs in MPN.


Asunto(s)
Transformación Celular Neoplásica/genética , Trastornos Mieloproliferativos/genética , Proteínas de Neoplasias/fisiología , Proteínas del Grupo Polycomb/fisiología , Proteínas de Ciclo Celular/fisiología , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/deficiencia , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Mutación con Ganancia de Función , Regulación Neoplásica de la Expresión Génica , Hematopoyesis , Código de Histonas , N-Metiltransferasa de Histona-Lisina/fisiología , Humanos , Terapia Molecular Dirigida , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Trastornos Mieloproliferativos/metabolismo , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Proteínas del Grupo Polycomb/deficiencia , Proteínas del Grupo Polycomb/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología
14.
Cell ; 175(1): 186-199.e19, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30220457

RESUMEN

Mutations or aberrant upregulation of EZH2 occur frequently in human cancers, yet clinical benefits of EZH2 inhibitor (EZH2i) remain unsatisfactory and limited to certain hematological malignancies. We profile global posttranslational histone modification changes across a large panel of cancer cell lines with various sensitivities to EZH2i. We report here oncogenic transcriptional reprogramming mediated by MLL1's interaction with the p300/CBP complex, which directs H3K27me loss to reciprocal H3K27ac gain and restricts EZH2i response. Concurrent inhibition of H3K27me and H3K27ac results in transcriptional repression and MAPK pathway dependency in cancer subsets. In preclinical models encompassing a broad spectrum of EZH2-aberrant solid tumors, a combination of EZH2 and BRD4 inhibitors, or a triple-combination including MAPK inhibition display robust efficacy with very tolerable toxicity. Our results suggest an attractive precision treatment strategy for EZH2-aberrant tumors on the basis of tumor-intrinsic MLL1 expression and concurrent inhibition of epigenetic crosstalk and feedback MAPK activation.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/genética , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Animales , Carcinogénesis/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Epigénesis Genética/genética , Epigenómica/métodos , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Código de Histonas/efectos de los fármacos , Código de Histonas/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/fisiología , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Complejo Represivo Polycomb 2/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Activación Transcripcional , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Factores de Transcripción p300-CBP/fisiología
15.
Nat Genet ; 50(10): 1452-1462, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30224650

RESUMEN

In embryonic stem cells (ESCs), developmental gene promoters are characterized by their bivalent chromatin state, with simultaneous modification by MLL2 and Polycomb complexes. Although essential for embryogenesis, bivalency is functionally not well understood. Here, we show that MLL2 plays a central role in ESC genome organization. We generate a catalog of bona fide bivalent genes in ESCs and demonstrate that loss of MLL2 leads to increased Polycomb occupancy. Consequently, promoters lose accessibility, long-range interactions are redistributed, and ESCs fail to differentiate. We pose that bivalency balances accessibility and long-range connectivity of promoters, allowing developmental gene expression to be properly modulated.


Asunto(s)
Diferenciación Celular/genética , Cromatina/genética , Cromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/fisiología , Células Madre Embrionarias de Ratones/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Regiones Promotoras Genéticas , Animales , Células Cultivadas , Cromatina/química , Ensamble y Desensamble de Cromatina/genética , Drosophila , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina/genética , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas del Grupo Polycomb/metabolismo , Unión Proteica/genética
16.
Cancer Discov ; 8(11): 1438-1457, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30139811

RESUMEN

Myelodysplastic syndromes (MDS) are heterogeneous hematopoietic disorders that are incurable with conventional therapy. Their incidence is increasing with global population aging. Although many genetic, epigenetic, splicing, and metabolic aberrations have been identified in patients with MDS, their clinical features are quite similar. Here, we show that hypoxia-independent activation of hypoxia-inducible factor 1α (HIF1A) signaling is both necessary and sufficient to induce dysplastic and cytopenic MDS phenotypes. The HIF1A transcriptional signature is generally activated in MDS patient bone marrow stem/progenitors. Major MDS-associated mutations (Dnmt3a, Tet2, Asxl1, Runx1, and Mll1) activate the HIF1A signature. Although inducible activation of HIF1A signaling in hematopoietic cells is sufficient to induce MDS phenotypes, both genetic and chemical inhibition of HIF1A signaling rescues MDS phenotypes in a mouse model of MDS. These findings reveal HIF1A as a central pathobiologic mediator of MDS and as an effective therapeutic target for a broad spectrum of patients with MDS.Significance: We showed that dysregulation of HIF1A signaling could generate the clinically relevant diversity of MDS phenotypes by functioning as a signaling funnel for MDS driver mutations. This could resolve the disconnection between genotypes and phenotypes and provide a new clue as to how a variety of driver mutations cause common MDS phenotypes. Cancer Discov; 8(11); 1438-57. ©2018 AACR. See related commentary by Chen and Steidl, p. 1355 This article is highlighted in the In This Issue feature, p. 1333.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Hipoxia/fisiopatología , Síndromes Mielodisplásicos/patología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Metaboloma , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo
17.
Biomed Res Int ; 2018: 6705842, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30003105

RESUMEN

The 11q23 of the mixed lineage leukemia 1 (MLL1) gene plays a crucial role in early embryonic development and hematopoiesis. The MLL-AF9 fusion gene, resulting from chromosomal translocation, often leads to acute myeloid leukemia with poor prognosis. Here, we generated a zebrafish model expressing the human MLL-AF9 fusion gene. Microinjection of human MLL-AF9 mRNA into zebrafish embryos resulted in enhanced hematopoiesis and the activation of downstream genes such as meis1 and hox cluster genes. Embryonic MLL-AF9 expression upregulated HSPC and myeloid lineage markers. Doxorubicin and MI-2 (a menin inhibitor) treatments significantly restored normal hematopoiesis in MLL-AF9-expressing animals. This study provides insight into the role of MLL-AF9 in zebrafish hematopoiesis and establishes a robust and efficient in vivo model for high-throughput drug screening.


Asunto(s)
Hematopoyesis/genética , Leucemia Mieloide Aguda/genética , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Fusión Oncogénica/fisiología , Translocación Genética , Animales , Modelos Animales de Enfermedad , N-Metiltransferasa de Histona-Lisina , Humanos , Pez Cebra
18.
Mol Cells ; 41(5): 444-453, 2018 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-29477140

RESUMEN

Aberrations in histone modifications are being studied in mixed-lineage leukemia (MLL)-AF9-driven acute myeloid leukemia (AML). In this study, we focused on the regulation of the differentiation of the MLL-AF9 type AML cell line THP-1. We observed that, upon phorbol 12-myristate 13-acetate (PMA) treatment, THP-1 cells differentiated into monocytes by down-regulating Aurora kinase A (AURKA), resulting in a reduction in H3S10 phosphorylation. We revealed that the AURKA inhibitor alisertib accelerates the expression of the H3K27 demethylase KDM6B, thereby dissociating AURKA and YY1 from the KDM6B promoter region. Using Flow cytometry, we found that alisertib induces THP-1 differentiation into monocytes. Furthermore, we found that treatment with the KDM6B inhibitor GSK-J4 perturbed the PMA-mediated differentiation of THP-1 cells. Thus, we discovered the mechanism of AURKA-KDM6B signaling that controls the differentiation of THP-1 cells, which has implications for biotherapy for leukemia.


Asunto(s)
Aurora Quinasa A/fisiología , Regulación Leucémica de la Expresión Génica , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Leucemia Monocítica Aguda/patología , Proteínas de Neoplasias/fisiología , Aurora Quinasa A/antagonistas & inhibidores , Azepinas/farmacología , Benzazepinas/farmacología , Diferenciación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Genes Reporteros , Células HEK293 , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Leucemia Monocítica Aguda/genética , Leucemia Monocítica Aguda/metabolismo , Monocitos/citología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/fisiología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Células THP-1 , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción YY1/metabolismo
19.
Cancer Cell ; 31(6): 755-770.e6, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28609655

RESUMEN

The MLL1 histone methyltransferase gene undergoes many distinct chromosomal rearrangements to yield poor-prognosis leukemia. The remaining wild-type allele is most commonly, but not always, retained. To what extent the wild-type allele contributes to leukemogenesis is unclear. Here we show, using rigorous, independent animal models, that endogenous MLL1 is dispensable for MLL-rearranged leukemia. Potential redundancy was addressed by co-deleting the closest paralog, Mll2. Surprisingly, Mll2 deletion alone had a significant impact on survival of MLL-AF9-transformed cells, and additional Mll1 loss further reduced viability and proliferation. We show that MLL1/MLL2 collaboration is not through redundancy, but regulation of distinct pathways. These findings highlight the relevance of MLL2 as a drug target in MLL-rearranged leukemia and suggest its broader significance in AML.


Asunto(s)
Proteínas de Unión al ADN/genética , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide Aguda/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Animales , Proliferación Celular/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/fisiología , Humanos , Ratones , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Proteínas Recombinantes de Fusión/genética
20.
Adv Exp Med Biol ; 978: 277-299, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28523552

RESUMEN

Huntington's disease (HD) is a genetic, fatal autosomal dominant neurodegenerative disorder typically occurring in midlife with symptoms ranging from chorea, to dementia, to personality disturbances (Philos Trans R Soc Lond Ser B Biol Sci 354:957-961, 1999). HD is inherited in a dominant fashion, and the underlying mutation in all cases is a CAG trinucleotide repeat expansion within exon 1 of the HD gene (Cell 72:971-983, 1993). The expanded CAG repeat, translated into a lengthened glutamine tract at the amino terminus of the huntingtin protein, affects its structural properties and functional activities. The effects are pleiotropic, as huntingtin is broadly expressed in different cellular compartments (i.e., cytosol, nucleus, mitochondria) as well as in all cell types of the body at all developmental stages, such that HD pathogenesis likely starts at conception and is a lifelong process (Front Neurosci 9:509, 2015). The rate-limiting mechanism(s) of neurodegeneration in HD still remains elusive: many different processes are commonly disrupted in HD cell lines and animal models, as well as in HD patient cells (Eur J Neurosci 27:2803-2820, 2008); however, epigenetic-chromatin deregulation, as determined by the analysis of DNA methylation, histone modifications, and noncoding RNAs, has now become a prevailing feature. Thus, the overarching goal of this chapter is to discuss the current status of the literature, reviewing how an aberrant epigenetic landscape can contribute to altered gene expression and neuronal dysfunction in HD.


Asunto(s)
Epigénesis Genética/genética , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Acetilación , Animales , Línea Celular , Ensamble y Desensamble de Cromatina/genética , Ensayos Clínicos como Asunto , Metilación de ADN/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Código de Histonas/genética , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/fisiología , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/fisiología , Homeostasis , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Metilación , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas del Grupo Polycomb/fisiología , Procesamiento Proteico-Postraduccional/genética , ARN no Traducido/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...