Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Blood Adv ; 7(21): 6395-6410, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37224458

RESUMEN

Deregulated expression of lineage-affiliated transcription factors (TFs) is a major mechanism of oncogenesis. However, how the deregulation of nonlineage affiliated TF affects chromatin to initiate oncogenic transcriptional programs is not well-known. To address this, we studied the chromatin effects imposed by oncogenic MAF as the cancer-initiating driver in the plasma cell cancer multiple myeloma. We found that the ectopically expressed MAF endows myeloma plasma cells with migratory and proliferative transcriptional potential. This potential is regulated by the activation of enhancers and superenhancers, previously inactive in healthy B cells and plasma cells, and the cooperation of MAF with the plasma cell-defining TF IRF4. Forced ectopic MAF expression confirms the de novo ability of oncogenic MAF to convert transcriptionally inert chromatin to active chromatin with the features of superenhancers, leading to the activation of the MAF-specific oncogenic transcriptome and the acquisition of cancer-related cellular phenotypes such as CCR1-dependent cell migration. These findings establish oncogenic MAF as a pioneer transcription factor that can initiate as well as sustain oncogenic transcriptomes and cancer phenotypes. However, despite its pioneer function, myeloma cells remain MAF-dependent, thus validating oncogenic MAF as a therapeutic target that would be able to circumvent the challenges of subsequent genetic diversification driving disease relapse and drug resistance.


Asunto(s)
Mieloma Múltiple , Humanos , Mieloma Múltiple/tratamiento farmacológico , Regulación de la Expresión Génica , Células Plasmáticas/metabolismo , Linfocitos B/metabolismo , Cromatina
2.
Blood ; 139(13): 1939-1953, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35015835

RESUMEN

Understanding the biological and clinical impact of copy number aberrations (CNAs) on the development of precision therapies in cancer remains an unmet challenge. Genetic amplification of chromosome 1q (chr1q-amp) is a major CNA conferring an adverse prognosis in several types of cancer, including in the blood cancer multiple myeloma (MM). Although several genes across chromosome 1 (chr1q) portend high-risk MM disease, the underpinning molecular etiology remains elusive. Here, with reference to the 3-dimensional (3D) chromatin structure, we integrate multi-omics data sets from patients with MM with genetic variables to obtain an associated clinical risk map across chr1q and to identify 103 adverse prognosis genes in chr1q-amp MM. Prominent among these genes, the transcription factor PBX1 is ectopically expressed by genetic amplification and epigenetic activation of its own preserved 3D regulatory domain. By binding to reprogrammed superenhancers, PBX1 directly regulates critical oncogenic pathways and a FOXM1-dependent transcriptional program. Together, PBX1 and FOXM1 activate a proliferative gene signature that predicts adverse prognosis across multiple types of cancer. Notably, pharmacological disruption of the PBX1-FOXM1 axis with existing agents (thiostrepton) and a novel PBX1 small molecule inhibitor (T417) is selectively toxic against chr1q-amp myeloma and solid tumor cells. Overall, our systems medicine approach successfully identifies CNA-driven oncogenic circuitries, links them to clinical phenotypes, and proposes novel CNA-targeted therapy strategies in MM and other types of cancer.


Asunto(s)
Mieloma Múltiple , Cromosomas Humanos Par 1/metabolismo , Proteína Forkhead Box M1/genética , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Pronóstico , Análisis de Sistemas , Factores de Transcripción/genética
3.
J Immunol ; 207(12): 2976-2991, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34810221

RESUMEN

RUNX1 is a transcription factor that plays key roles in hematopoietic development and in hematopoiesis and lymphopoiesis. In this article, we report that RUNX1 regulates a gene expression program in naive mouse B cells that affects the dynamics of cell cycle entry in response to stimulation of the BCR. Conditional knockout of Runx1 in mouse resting B cells resulted in accelerated entry into S-phase after BCR engagement. Our results indicate that Runx1 regulates the cyclin D2 (Ccnd2) gene, the immediate early genes Fosl2, Atf3, and Egr2, and the Notch pathway gene Rbpj in mouse B cells, reducing the rate at which transcription of these genes increases after BCR stimulation. RUNX1 interacts with the chromatin remodeler SNF-2-related CREB-binding protein activator protein (SRCAP), recruiting it to promoter and enhancer regions of the Ccnd2 gene. BCR-mediated activation triggers switching between binding of RUNX1 and its paralog RUNX3 and between SRCAP and the switch/SNF remodeling complex member BRG1. Binding of BRG1 is increased at the Ccnd2 and Rbpj promoters in the Runx1 knockout cells after BCR stimulation. We also find that RUNX1 exerts positive or negative effects on a number of genes that affect the activation response of mouse resting B cells. These include Cd22 and Bank1, which act as negative regulators of the BCR, and the IFN receptor subunit gene Ifnar1 The hyperresponsiveness of the Runx1 knockout B cells to BCR stimulation and its role in regulating genes that are associated with immune regulation suggest that RUNX1 could be involved in regulating B cell tolerance.


Asunto(s)
Linfocitos B , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Animales , Linfocitos B/metabolismo , Ciclo Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Hematopoyesis , Ratones , Regiones Promotoras Genéticas
4.
Mol Biol Cell ; 32(22): ar40, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34613789

RESUMEN

Mesendoderm cells are key intermediate progenitors that form at the early primitive streak (PrS) and give rise to mesoderm and endoderm in the gastrulating embryo. We have identified an interaction between CNOT3 and the cell cycle kinase Aurora B that requires sequences in the NOT box domain of CNOT3 and regulates MAPK/ERK signaling during mesendoderm differentiation. Aurora B phosphorylates CNOT3 at two sites located close to a nuclear localization signal and promotes localization of CNOT3 to the nuclei of mouse embryonic stem cells (ESCs) and metastatic lung cancer cells. ESCs that have both sites mutated give rise to embryoid bodies that are largely devoid of mesoderm and endoderm and are composed mainly of cells with ectodermal characteristics. The mutant ESCs are also compromised in their ability to differentiate into mesendoderm in response to FGF2, BMP4, and Wnt3 due to reduced survival and proliferation of differentiating mesendoderm cells. We also show that the double mutation alters the balance of interaction of CNOT3 with Aurora B and with ERK and reduces phosphorylation of ERK in response to FGF2. Our results identify a potential adaptor function for CNOT3 that regulates the Ras/MEK/ERK pathway during embryogenesis.


Asunto(s)
Aurora Quinasa B/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Madre Embrionarias de Ratones/citología , Factores de Transcripción/metabolismo , Células A549 , Animales , Aurora Quinasa B/genética , Diferenciación Celular/fisiología , Supervivencia Celular , Células Cultivadas , Endodermo/citología , Endodermo/fisiología , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Humanos , Mesodermo/citología , Ratones , Células Madre Embrionarias de Ratones/fisiología , Mutación , Fosforilación , Factores de Transcripción/genética
5.
Oncotarget ; 7(40): 65923-65936, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27588474

RESUMEN

Ikaros and Foxp1 are transcription factors that play key roles in normal lymphopoiesis and lymphoid malignancies. We describe a novel physical and functional interaction between the proteins, which requires the central zinc finger domain of Ikaros. The Ikaros-Foxp1 interaction is abolished by deletion of this region, which corresponds to the IK6 isoform that is commonly associated with high-risk acute lymphoblastic leukemia (ALL). We also identify the Gpr132 gene, which encodes the orphan G protein-coupled receptor G2A, as a novel target for Foxp1. Increased expression of Foxp1 enhanced Gpr132 transcription and caused cell cycle changes, including G2 arrest. Co-expression of wild-type Ikaros, but not IK6, displaced Foxp1 binding from the Gpr132 gene, reversed the increase in Gpr132 expression and inhibited G2 arrest. Analysis of primary ALL samples revealed a significant increase in GPR132 expression in IKZF1-deleted BCR-ABL negative patients, suggesting that levels of wild-type Ikaros may influence the regulation of G2A in B-ALL. Our results reveal a novel effect of Ikaros haploinsufficiency on Foxp1 functioning, and identify G2A as a potential modulator of the cell cycle in Ikaros-deleted B-ALL.


Asunto(s)
Linfocitos B/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Factor de Transcripción Ikaros/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Represoras/metabolismo , Apoptosis , Linfocitos B/patología , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Proliferación Celular , Factores de Transcripción Forkhead/genética , Proteínas de Fusión bcr-abl/genética , Eliminación de Gen , Humanos , Factor de Transcripción Ikaros/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores Acoplados a Proteínas G/genética , Proteínas Represoras/genética , Células Tumorales Cultivadas
6.
Mol Biol Cell ; 25(6): 904-15, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24430871

RESUMEN

Methylated histones H3K9 and H3K27 are canonical epigenetic silencing modifications in metazoan organisms, but the relationship between the two modifications has not been well characterized. H3K9me3 coexists with H3K27me3 in pluripotent and differentiated cells. However, we find that the functioning of H3K9me3 is altered by H3S10 phosphorylation in differentiated postmitotic osteoblasts and cycling B cells. Deposition of H3K9me3/S10ph at silent genes is partially mediated by the mitogen- and stress-activated kinases (MSK1/2) and the Aurora B kinase. Acquisition of H3K9me3/S10ph during differentiation correlates with loss of paused S5 phosphorylated RNA polymerase II, which is present on Polycomb-regulated genes in embryonic stem cells. Reduction of the levels of H3K9me3/S10ph by kinase inhibition results in increased binding of RNAPIIS5ph and the H3K27 methyltransferase Ezh1 at silent promoters. Our results provide evidence of a novel developmentally regulated methyl-phospho switch that modulates Polycomb regulation in differentiated cells and stabilizes repressed states.


Asunto(s)
Linfocitos B/metabolismo , Epigénesis Genética , Histonas/genética , Osteoblastos/metabolismo , Proteínas del Grupo Polycomb/genética , ARN Polimerasa II/genética , Animales , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Linfocitos B/citología , Sitios de Unión , Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Activación de Linfocitos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Metilación , Ratones , Osteoblastos/citología , Fosforilación , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , ARN Polimerasa II/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal , Bazo/citología , Bazo/metabolismo
7.
Blood ; 116(18): 3445-55, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-20651073

RESUMEN

Apoptosis of short-lived plasma cells after a few days of intense immunoglobulin secretion is critical for maintaining a controlled humoral immune response. The mechanisms that regulate this process are poorly understood. Here we report that the key apoptotic caspases, caspase-3 and caspase-9, become resistant to activation by apoptotic stimuli when B cells differentiate into short-lived plasma cells. As a consequence, apoptosis of most short-lived plasma cells in vitro and in vivo is effector caspase-independent. We also show that a triaspartic acid repeat that normally prevents activation of caspase-3 becomes stabilized in short-lived plasma cells and myeloma cell lines. The block on caspase activation occurs before the accumulation of intracellular immunoglobulins and a progressive rise in secretory stress in the endoplasmic reticulum (ER). Plasma cells show increased susceptibility to ER stress-induced apoptosis and activate the ER-associated caspase-12, which is required specifically for nuclear apoptotic events. In nonlymphoid cells that cannot activate effector caspases, programmed cell death is delayed in response to ER stress. These observations suggest that the block on activation of key apoptotic caspases has evolved in short-lived plasma cells to prolong survival under conditions of ER stress resulting from high-level immunoglobulin secretion.


Asunto(s)
Apoptosis , Linfocitos B/citología , Caspasa 3/inmunología , Caspasa 9/inmunología , Retículo Endoplásmico/inmunología , Células Plasmáticas/citología , Animales , Linfocitos B/inmunología , Caspasa 12/inmunología , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Humanos , Linfoma/inmunología , Ratones , Células Plasmáticas/inmunología , Secuencias Repetitivas de Aminoácido
8.
EMBO J ; 26(22): 4657-69, 2007 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-17948062

RESUMEN

Combinatorial modifications of the core histones have the potential to fine-tune the epigenetic regulation of chromatin states. The Aurora B kinase is responsible for generating the double histone H3 modification tri-methylated K9/phosphorylated S10 (H3K9me3/S10ph), which has been implicated in chromosome condensation during mitosis. In this study, we have identified a novel role for Aurora B in epigenetic marking of silent chromatin during cell differentiation. We find that phosphorylation of H3 S10 by Aurora B generates high levels of the double H3K9me3/S10ph modification in differentiated postmitotic cells and also results in delocalisation of HP1beta away from heterochromatin in terminally differentiated plasma cells. Microarray analysis of the H3K9me3/S10ph modification shows a striking increase in the modification across repressed genes during differentiation of mesenchymal stem cells. Our results provide evidence that the Aurora B kinase has a role in marking silent chromatin independently of the cell cycle and suggest that targeting of Aurora B-mediated phosphorylation of H3 S10 to repressed genes could be a mechanism for epigenetic silencing of gene expression.


Asunto(s)
Silenciador del Gen , Heterocromatina/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Aurora Quinasa B , Aurora Quinasas , Linfocitos B/citología , Linfocitos B/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Epigénesis Genética , Histonas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Bazo/citología
9.
Immunity ; 26(3): 335-44, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17363301

RESUMEN

Ikaros DNA-binding proteins are critical for the development of lymphocytes and other hematopoietic lineages, but it remains unclear how they cooperate with other regulators of signaling and transcription to achieve ordered gene expression during development. Here, we show that Ikaros proteins regulate the pre-BCR component lambda5 in a stage-specific manner. In pre-BI cells, Ikaros modulated lambda5 expression in competition with the transcriptional activator EBF. This required Ikaros binding to the Igll1 (lambda5) promoter and was abolished either by mutation of the Ikaros DNA-binding domain or by deletion of a single Ikaros site from the Igll1 promoter. At the transition from the pre-BI to pre-BII stage, the expression of the Ikaros family member Aiolos was upregulated and required for the efficient silencing of Igll1. Aiolos expression was controlled by pre-BCR signals via the adaptor protein SLP-65. Thus, pre-BCR signaling regulates Aiolos and the silencing of Igll1 via a developmental-stage-specific feedback loop.


Asunto(s)
Linfocitos B/inmunología , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/fisiología , Cadenas Ligeras de Inmunoglobulina/genética , Glicoproteínas de Membrana/genética , Transactivadores/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Linfocitos B/química , Células Cultivadas , Proteínas de Unión al ADN/genética , Silenciador del Gen , Factor de Transcripción Ikaros/análisis , Factor de Transcripción Ikaros/genética , Inmunoglobulina de Cadenas Ligeras Subrogadas , Activación de Linfocitos/genética , Ratones , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína/genética , Transactivadores/fisiología
10.
Mol Immunol ; 42(11): 1283-92, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15950724

RESUMEN

The lambda5 and VpreB genes encode the components of the surrogate light-chain which forms part of the pre-B cell receptor and plays a key role in B cell development. In the mouse, the lambda5 and VpreB1 genes are closely linked and are co-regulated by a multi-component locus control region. To identify the sequences that regulate lambda5 and VpreB1 expression during B cell development, we have comprehensively mapped the DNaseI hypersensitive sites (HS) in the lambda5-VpreB1 functional domain. The active domain contains 12 HS that are distributed at high density across the 18.3 kb region that forms the lambda5 and VpreB1 functional unit. Analysis of a reporter gene driven by the VpreB1 promoter in transgenic mice identified a novel enhancer associated with two HS located upstream of lambda5. The lambda5-VpreB1 locus was also found to be closely linked to the ubiquitously expressed Topoisomerase-3beta (Topo3beta) gene. The VpreB1 and Topo3beta genes have entirely different expression patterns despite the fact that the two promoters are separated by a distance of only 1.5 kb.


Asunto(s)
Cadenas Ligeras de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Glicoproteínas de Membrana/genética , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Secuencia de Bases , Diferenciación Celular , Línea Celular , Mapeo Cromosómico , ADN Complementario/genética , Desoxirribonucleasa I , Elementos de Facilitación Genéticos , Genes Reguladores , Inmunoglobulina de Cadenas Ligeras Subrogadas , Ratones , Ratones Transgénicos , Plásmidos/genética , Receptores de Células Precursoras de Linfocitos B , Regiones Promotoras Genéticas , Receptores de Antígenos de Linfocitos B
11.
Semin Immunol ; 17(2): 121-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15737573

RESUMEN

The lambda5 and VpreB genes encode the components of the surrogate light-chain, which forms part of the pre-B cell receptor. In mouse, the lambda5 and VpreB1 genes of mouse are closely linked and coordinately regulated by a locus control region (LCR). Activation of the genes in pro-B cells depends on the combined effects of early B cell factor (EBF) and the E2A factors E12 and E47. Silencing of lambda5 expression in mature B cells occurs through the action of Ikaros on the gene promoter where it may compete for binding of EBF and initiate the formation of a silent chromatin structure.


Asunto(s)
Linfocitos B/inmunología , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Glicoproteínas de Membrana/genética , Factores de Transcripción/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Cadenas Ligeras de Inmunoglobulina , Inmunoglobulina de Cadenas Ligeras Subrogadas , Glicoproteínas de Membrana/fisiología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA