Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38875504

RESUMEN

Epidemiological studies report opposing influences of infection on childhood B cell acute lymphoblastic leukemia (B-ALL). Although infections in the first year of life appear to exert the largest impact on leukemia risk, the effect of early pathogen exposure on the fetal preleukemia cells (PLC) that lead to B-ALL has yet to be reported. Using cytomegalovirus as a model early-life infection, we show that virus exposure within one week of birth induces profound depletion of transplanted B-ALL cells in two mouse models and of in situ-generated PLC in Eu-ret mice. The age-dependent depletion of PLC results from an elevated STAT4-mediated cytokine response in neonates, with high levels of IL-12p40-driven IFN-g production inducing PLC death. Similar PLC depletion can be achieved in adult mice by impairing viral clearance. These findings provide mechanistic support for an inhibitory effect of early-life infection on B-ALL progression and could inform development of therapeutic or preventative approaches.

2.
Gene Ther ; 27(10-11): 525-534, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-32704085

RESUMEN

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have promising potential for opening new avenues in regenerative medicine. However, since the tumorigenic potential of undifferentiated pluripotent stem cells (PSCs) is a major safety concern for clinical transplantation, inducible Caspase-9 (iC9) is under consideration for use as a fail-safe system. Here, we used targeted gene editing to introduce the iC9 system into human iPSCs, and then interrogated the efficiency of inducible apoptosis with normal iPSCs as well as diseased iPSCs derived from patients with acute myeloid leukemia (AML-iPSCs). The iC9 system induced quick and efficient apoptosis to iPSCs in vitro. More importantly, complete eradication of malignant cells without AML recurrence was shown in disease mouse models by using AML-iPSCs. In parallel, it shed light on several limitations of the iC9 system usage. Our results suggest that careful use of the iC9 system will serve as an important countermeasure against posttransplantation adverse events in stem cell transplantation therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Animales , Apoptosis , Caspasa 9/genética , Caspasa 9/metabolismo , Diferenciación Celular , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células Madre Pluripotentes/metabolismo
3.
Haematologica ; 104(9): 1744-1755, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30792210

RESUMEN

The balance between self-renewal and differentiation is crucial to ensure the homeostasis of the hematopoietic system, and is a hallmark of hematopoietic stem cells. However, the underlying molecular pathways, including the role of micro-RNA, are not completely understood. To assess the contribution of micro-RNA, we performed micro-RNA profiling of hematopoietic stem cells and their immediate downstream progeny multi-potent progenitors from wild-type control and Pbx1-conditional knockout mice, whose stem cells display a profound self-renewal defect. Unsupervised hierarchical cluster analysis separated stem cells from multi-potent progenitors, suggesting that micro-RNA might regulate the first transition step in the adult hematopoietic development. Notably, Pbx1-deficient and wild-type cells clustered separately, linking micro-RNAs to self-renewal impairment. Differential expression analysis of micro-RNA in the physiological stem cell-to-multi-potent progenitor transition and in Pbx1-deficient stem cells compared to control stem cells revealed miR-127-3p as the most differentially expressed. Furthermore, miR-127-3p was strongly stem cell-specific, being quickly down-regulated upon differentiation and not re-expressed further downstream in the bone marrow hematopoietic hierarchy. Inhibition of miR-127-3p function in Lineage-negative cells, achieved through a lentiviral-sponge vector, led to severe stem cell depletion, as assessed with serial transplantation assays. miR-127-3p-sponged stem cells displayed accelerated differentiation, which was uncoupled from proliferation, accounting for the observed stem cell reduction. miR-127-3p overexpression in Lineage-negative cells did not alter stem cell pool size, but gave rise to lymphopenia, likely due to lack of miR-127-3p physiological downregulation beyond the stem cell stage. Thus, tight regulation of miR-127-3p is crucial to preserve the self-renewing stem cell pool and homeostasis of the hematopoietic system.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/citología , MicroARNs/fisiología , Animales , Linaje de la Célula/genética , Análisis por Conglomerados , Cruzamientos Genéticos , Perfilación de la Expresión Génica , Hematopoyesis , Homeostasis , Humanos , Células K562 , Lentivirus/genética , Ratones , Ratones Noqueados , Estrés Oxidativo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo
4.
Blood ; 126(14): 1683-94, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26311362

RESUMEN

Chromosomal rearrangements involving the mixed-lineage leukemia (MLL) gene occur in primary and treatment-related leukemias and confer a poor prognosis. Studies based primarily on mouse models have substantially advanced our understanding of MLL leukemia pathogenesis, but often use supraphysiological oncogene expression with uncertain implications for human leukemia. Genome editing using site-specific nucleases provides a powerful new technology for gene modification to potentially model human disease, however, this approach has not been used to re-create acute leukemia in human cells of origin comparable to disease observed in patients. We applied transcription activator-like effector nuclease-mediated genome editing to generate endogenous MLL-AF9 and MLL-ENL oncogenes through insertional mutagenesis in primary human hematopoietic stem and progenitor cells (HSPCs) derived from human umbilical cord blood. Engineered HSPCs displayed altered in vitro growth potentials and induced acute leukemias following transplantation in immunocompromised mice at a mean latency of 16 weeks. The leukemias displayed phenotypic and morphologic similarities with patient leukemia blasts including a subset with mixed phenotype, a distinctive feature seen in clinical disease. The leukemic blasts expressed an MLL-associated transcriptional program with elevated levels of crucial MLL target genes, displayed heightened sensitivity to DOT1L inhibition, and demonstrated increased oncogenic potential ex vivo and in secondary transplant assays. Thus, genome editing to create endogenous MLL oncogenes in primary human HSPCs faithfully models acute MLL-rearranged leukemia and provides an experimental platform for prospective studies of leukemia initiation and stem cell biology in a genetic subtype of poor prognosis leukemia.


Asunto(s)
Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Bifenotípica Aguda/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Animales , Antígenos CD34/inmunología , Separación Celular , Técnicas de Sustitución del Gen , Genoma Humano , Humanos , Ratones , Microscopía Confocal , Mutagénesis Sitio-Dirigida , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Transducción Genética , Transfección
5.
Cancer Sci ; 106(3): 227-36, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25529853

RESUMEN

Acute myeloid leukemia is a clonal malignant disorder derived from a small number of leukemic stem cells (LSCs). Rearrangements of the mixed lineage leukemia (MLL) gene are found in acute myeloid leukemia associated with poor prognosis. The upregulation of Hox genes is critical for LSC induction and maintenance, but is unlikely to support malignancy and the high LSC frequency observed in MLL leukemias. The present study shows that MLL fusion proteins interact with the transcription factor PU.1 to activate the transcription of CSF-1R, which is critical for LSC activity. Acute myeloid leukemia is cured by either deletion of PU.1 or ablation of cells expressing CSF-1R. Kinase inhibitors specific for CSF-1R prolong survival time. These findings indicate that PU.1-mediated upregulation of CSF-1R is a critical effector of MLL leukemogenesis.


Asunto(s)
Carcinogénesis/genética , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide Aguda/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Proto-Oncogénicas/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Transactivadores/genética , Animales , Regulación Leucémica de la Expresión Génica , Genes Homeobox , Leucemia Mieloide Aguda/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Neoplásicas , Compuestos de Fenilurea/farmacología , Pronóstico , Receptor de Factor Estimulante de Colonias de Macrófagos/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transducción de Señal , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Tiazoles/farmacología , Transcripción Genética , Activación Transcripcional , Regulación hacia Arriba
6.
J Cell Sci ; 126(Pt 14): 3181-91, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23660001

RESUMEN

The capacity of the hematopoietic system to promptly respond to peripheral demands relies on adequate pools of progenitors able to transiently proliferate and differentiate in a regulated manner. However, little is known about factors that may restrain progenitor maturation to maintain their reservoirs. Conditional knockout mice for the Pbx1 proto-oncogene have a significant reduction in lineage-restricted progenitors in addition to a profound defect in hematopoietic stem cell (HSC) self-renewal. Through analysis of purified progenitor proliferation, differentiation capacity and transcriptional profiling, we demonstrate that Pbx1 regulates the lineage-specific output of multipotent and oligopotent progenitors. In the absence of Pbx1 multipotent progenitor (MPP) and common myeloid progenitor (CMP) pools are reduced due to aberrantly rapid myeloid maturation. This is associated with premature expression of myeloid differentiation genes and decreased maintenance of proto-oncogene transcriptional pathways, including reduced expression of Meis1, a Pbx1 dimerization partner, and its subordinate transcriptional program. Conversely, Pbx1 maintains the lymphoid differentiation potential of lymphoid-primed MPPs (LMPPs) and common lymphoid progenitors (CLPs), whose reduction in the absence of Pbx1 is associated with a defect in lymphoid priming that is also present in CMPs, which persistently express lymphoid and HSC genes underlying a previously unappreciated lineage promiscuity that is maintained by Pbx1. These results demonstrate a role for Pbx1 in restraining myeloid maturation while maintaining lymphoid potential to appropriately regulate progenitor reservoirs.


Asunto(s)
Hematopoyesis , Proteínas de Homeodominio/metabolismo , Células Progenitoras Linfoides/fisiología , Células Progenitoras Mieloides/fisiología , Proteínas de Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Animales , Linaje de la Célula/genética , Supervivencia Celular/genética , Células Cultivadas , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Multimerización de Proteína , Factores de Transcripción/genética , Activación Transcripcional
7.
Genome Res ; 21(5): 798-810, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21451113

RESUMEN

Emerging evidence suggests that microRNAs (miRNAs), an abundant class of ∼22-nucleotide small regulatory RNAs, play key roles in controlling the post-transcriptional genetic programs in stem and progenitor cells. Here we systematically examined miRNA expression profiles in various adult tissue-specific stem cells and their differentiated counterparts. These analyses revealed miRNA programs that are common or unique to blood, muscle, and neural stem cell populations and miRNA signatures that mark the transitions from self-renewing and quiescent stem cells to proliferative and differentiating progenitor cells. Moreover, we identified a stem/progenitor transition miRNA (SPT-miRNA) signature that predicts the effects of genetic perturbations, such as loss of PTEN and the Rb family, AML1-ETO9a expression, and MLL-AF10 transformation, on self-renewal and proliferation potentials of mutant stem/progenitor cells. We showed that some of the SPT-miRNAs control the self-renewal of embryonic stem cells and the reconstitution potential of hematopoietic stem cells (HSCs). Finally, we demonstrated that SPT-miRNAs coordinately regulate genes that are known to play roles in controlling HSC self-renewal, such as Hoxb6 and Hoxa4. Together, these analyses reveal the miRNA programs that may control key processes in normal and aberrant stem and progenitor cells, setting the foundations for dissecting post-transcriptional regulatory networks in stem cells.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/genética , MicroARNs/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , MicroARNs/genética , Mutación , Mioblastos/citología , Mioblastos/metabolismo , Células-Madre Neurales , Especificidad de Órganos , Células Madre/citología
8.
Nat Cell Biol ; 9(10): 1208-15, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17891136

RESUMEN

Enzymes that mediate reversible epigenetic modifications have not only been recognized as key in regulating gene expression and oncogenesis, but also provide potential targets for molecular therapy. Although the methylation of arginine 3 of histone 4 (H4R3) by protein arginine methyltransferase 1 (PRMT1) is a critical modification for active chromatin and prevention of heterochromatin spread, there has been no direct evidence of any role of PRMTs in cancer. Here, we show that PRMT1 is an essential component of a novel Mixed Lineage Leukaemia (MLL) oncogenic transcriptional complex with both histone acetylation and H4R3 methylation activities, which also correlate with the expression of critical MLL downstream targets. Direct fusion of MLL with PRMT1 or Sam68, a bridging molecule in the complex for PRMT1 interaction, could enhance self-renewal of primary haematopoietic cells. Conversely, specific knockdown of PRMT1 or Sam68 expression suppressed MLL-mediated transformation. This study not only functionally dissects the oncogenic transcriptional machinery associated with an MLL fusion complex, but also uncovers--for the first time--an essential function of PRMTs in oncogenesis and reveals their potential as novel therapeutic targets in human cancer.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Acetilación , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Arginina/metabolismo , Línea Celular , Transformación Celular Neoplásica/genética , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Células HeLa , Histonas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Inmunoprecipitación , Metilación , Mutación , Regiones Promotoras Genéticas/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
9.
Cancer Cell ; 10(6): 456-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17157786

RESUMEN

Improved understanding of the molecular pathways that suppress the genesis and maintenance of cancer stem cells will facilitate development of rationally targeted therapies. PU.1 is a transcription factor that is required for normal myelomonocytic differentiation in hematopoiesis, and reduced PU.1 activity has been associated with myeloid leukemogenesis in man and in mouse models. A recent study by Steidl et al. demonstrates that Junb and Jun, two AP-1 transcription factors, are critical downstream effectors of the tumor suppressor activity of PU.1, and that reduced expression of Junb, in particular, may be a common feature of acute myeloid leukemogenesis.


Asunto(s)
Leucemia Mieloide Aguda/prevención & control , Células Madre Neoplásicas/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Humanos , Ratones
10.
Cancer Cell ; 10(4): 257-68, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17045204

RESUMEN

Using a mouse model of human acute myeloid leukemia (AML) induced by the MLL-AF9 oncogene, we demonstrate that colony-forming cells (CFCs) in the bone marrow and spleen of leukemic mice are also leukemia stem cells (LSCs). These self-renewing cells (1) are frequent, accounting for 25%-30% of myeloid lineage cells at late-stage disease; (2) generate a phenotypic, morphologic, and functional leukemia cell hierarchy; (3) express mature myeloid lineage-specific antigens; and (4) exhibit altered microenvironmental interactions by comparison with the oncogene-immortalized CFCs that initiated the disease. Therefore, the LSCs responsible for sustaining, expanding, and regenerating MLL-AF9 AML are downstream myeloid lineage cells, which have acquired an aberrant Hox-associated self-renewal program as well as other biologic features of hematopoietic stem cells.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Experimental/patología , Leucemia Mieloide Aguda/genética , Células Mieloides/patología , Proteínas de Fusión Oncogénica/metabolismo , Animales , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula , Línea Celular Transformada , Linaje de la Célula , Transformación Celular Neoplásica , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Leucemia Experimental/etiología , Leucemia Experimental/genética , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Retroviridae/genética , Bazo/patología , Transducción Genética , Trasplante Homólogo , Rayos X
11.
Nature ; 455(7217): 1205-9, 2008 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-18806775

RESUMEN

Glycogen synthase kinase 3 (GSK3) is a multifunctional serine/threonine kinase that participates in numerous signalling pathways involved in diverse physiological processes. Several of these pathways are implicated in disease pathogenesis, which has prompted efforts to develop GSK3-specific inhibitors for therapeutic applications. However, before now, there has been no strong rationale for targeting GSK3 in malignancies. Here we report pharmacological, physiological and genetic studies that demonstrate an oncogenic requirement for GSK3 in the maintenance of a specific subtype of poor prognosis human leukaemia, genetically defined by mutations of the MLL proto-oncogene. In contrast to its previously characterized roles in suppression of neoplasia-associated signalling pathways, GSK3 paradoxically supports MLL leukaemia cell proliferation and transformation by a mechanism that ultimately involves destabilization of the cyclin-dependent kinase inhibitor p27(Kip1). Inhibition of GSK3 in a preclinical murine model of MLL leukaemia provides promising evidence of efficacy and earmarks GSK3 as a candidate cancer drug target.


Asunto(s)
Transformación Celular Neoplásica , Glucógeno Sintasa Quinasa 3/metabolismo , Leucemia Linfoide/tratamiento farmacológico , Leucemia Linfoide/patología , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Animales , División Celular , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Modelos Animales de Enfermedad , Fase G1 , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/deficiencia , Glucógeno Sintasa Quinasa 3/genética , N-Metiltransferasa de Histona-Lisina , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoenzimas/metabolismo , Leucemia Linfoide/enzimología , Leucemia Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Células Progenitoras Mieloides/enzimología , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patología , Células Precursoras de Linfocitos B/enzimología , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/patología , Proto-Oncogenes Mas
13.
Blood Adv ; 8(11): 2846-2860, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38598725

RESUMEN

ABSTRACT: The t(1;19) translocation, encoding the oncogenic fusion protein E2A (TCF3)-PBX1, is involved in acute lymphoblastic leukemia (ALL) and associated with a pre-B-cell receptor (preBCR+) phenotype. Relapse in patients with E2A-PBX1+ ALL frequently occurs in the central nervous system (CNS). Therefore, there is a medical need for the identification of CNS active regimens for the treatment of E2A-PBX1+/preBCR+ ALL. Using unbiased short hairpin RNA (shRNA) library screening approaches, we identified Bruton tyrosine kinase (BTK) as a key gene involved in both proliferation and dasatinib sensitivity of E2A-PBX1+/preBCR+ ALL. Depletion of BTK by shRNAs resulted in decreased proliferation of dasatinib-treated E2A-PBX1+/preBCR+ cells compared with control-transduced cells. Moreover, the combination of dasatinib with BTK inhibitors (BTKi; ibrutinib, acalabrutinib, or zanubrutinib) significantly decreased E2A-PBX1+/preBCR+ human and murine cell proliferation, reduced phospholipase C gamma 2 (PLCG2) and BTK phosphorylation and total protein levels and increased disease-free survival of mice in secondary transplantation assays, particularly reducing CNS-leukemic infiltration. Hence, dasatinib with ibrutinib reduced pPLCG2 and pBTK in primary ALL patient samples, including E2A-PBX1+ ALLs. In summary, genetic depletion and pharmacological inhibition of BTK increase dasatinib effects in human and mouse with E2A-PBX1+/preBCR+ ALL across most of performed assays, with the combination of dasatinib and BTKi proving effective in reducing CNS infiltration of E2A-PBX1+/preBCR+ ALL cells in vivo.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa , Dasatinib , Inhibidores de Proteínas Quinasas , Dasatinib/uso terapéutico , Dasatinib/farmacología , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/metabolismo , Humanos , Animales , Ratones , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos
14.
J Cell Sci ; 124(Pt 13): 2208-19, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21670200

RESUMEN

The mixed lineage leukemia (MLL) proto-oncogenic protein is a histone-lysine N-methyltransferase that is produced by proteolytic cleavage and self-association of the respective functionally distinct subunits (MLL(N) and MLL(C)) to form a holocomplex involved in epigenetic transcriptional regulation. On the basis of studies in Drosophila it has been suggested that the separated subunits might also have distinct functions. In this study, we used a genetically engineered mouse line that lacked MLL(C) to show that the MLL(N)-MLL(C) holocomplex is responsible for MLL functions in various developmental processes. The stability of MLL(N) is dependent on its intramolecular interaction with MLL(C), which is mediated through the first and fourth plant homeodomain (PHD) fingers (PHD1 and PHD4) and the phenylalanine/tyrosine-rich (FYRN) domain of MLL(N). Free MLL(N) is destroyed by a mechanism that targets the FYRN domain, whereas free MLL(C) is exported to the cytoplasm and degraded by the proteasome. PHD1 is encoded by an alternatively spliced exon that is occasionally deleted in T-cell leukemia, and its absence produces an MLL mutant protein that is deficient for holocomplex formation. Therefore, this should be a loss-of-function mutant allele, suggesting that the known tumor suppression role of MLL may also apply to the T-cell lineage. Our data demonstrate that the dissociated MLL subunits are subjected to distinct degradation pathways and thus not likely to have separate functions unless the degradation mechanisms are inhibited.


Asunto(s)
Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Leucemia de Células T/genética , Ratones , Ratones Noqueados , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas del Grupo Polycomb , Procesamiento Proteico-Postraduccional , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
15.
J Cell Sci ; 124(Pt 10): 1681-90, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21511732

RESUMEN

The epidermal barrier varies over the body surface to accommodate regional environmental stresses. Regional skin barrier variation is produced by site-dependent epidermal differentiation from common keratinocyte precursors and often manifests as site-specific skin disease or irritation. There is strong evidence for body-site-dependent dermal programming of epidermal differentiation in which the epidermis responds by altering expression of key barrier proteins, but the underlying mechanisms have not been defined. The LCE multigene cluster encodes barrier proteins that are differentially expressed over the body surface, and perturbation of LCE cluster expression is linked to the common regional skin disease psoriasis. LCE subclusters comprise genes expressed variably in either external barrier-forming epithelia (e.g. skin) or in internal epithelia with less stringent barriers (e.g. tongue). We demonstrate here that a complex of TALE homeobox transcription factors PBX1, PBX2 and Pknox (homologues of Drosophila Extradenticle and Homothorax) preferentially regulate external rather than internal LCE gene expression, competitively binding with SP1 and SP3. Perturbation of TALE protein expression in stratified squamous epithelia in mice produces external but not internal barrier abnormalities. We conclude that epidermal barrier genes, such as the LCE multigene cluster, are regulated by TALE homeodomain transcription factors to produce regional epidermal barriers.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Fenómenos Fisiológicos de la Piel/genética , Animales , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas Ricas en Prolina del Estrato Córneo/genética , Células Epidérmicas , Epidermis/metabolismo , Humanos , Queratinocitos/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Piel/citología , Piel/metabolismo
17.
Nat Genet ; 30(4): 430-5, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11912494

RESUMEN

Pbx1 is a member of the TALE (three-amino acid loop extension) class of homeodomain transcription factors, which are components of hetero-oligomeric protein complexes thought to regulate developmental gene expression and to maintain differentiated cell states. In vitro studies have shown that Pbx1 regulates the activity of Ipf1 (also known as Pdx1), a ParaHox homeodomain transcription factor required for the development and function of the pancreas in mice and humans. To investigate in vivo roles of Pbx1 in pancreatic development and function, we examined pancreatic Pbx1 expression, and morphogenesis, cell differentiation and function in mice deficient for Pbx1. Pbx1-/- embryos had pancreatic hypoplasia and marked defects in exocrine and endocrine cell differentiation prior to death at embryonic day (E) 15 or E16. In these embryos, expression of Isl1 and Atoh5, essential regulators of pancreatic morphogenesis and differentiation, was severely reduced. Pbx1+/- adults had pancreatic islet malformations, impaired glucose tolerance and hypoinsulinemia. Thus, Pbx1 is essential for normal pancreatic development and function. Analysis of trans-heterozygous Pbx1+/- Ipf1+/- mice revealed in vivo genetic interactions between Pbx1 and Ipf1 that are essential for postnatal pancreatic function; these mice developed age-dependent overt diabetes mellitus, unlike Pbx1+/- or Ipf1+/- mice. Mutations affecting the Ipf1 protein may promote diabetes mellitus in mice and humans. This study suggests that perturbation of Pbx1 activity may also promote susceptibility to diabetes mellitus.


Asunto(s)
Proteínas de Unión al ADN/genética , Diabetes Mellitus/genética , Proteínas de Homeodominio , Páncreas/embriología , Páncreas/fisiología , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Bromodesoxiuridina/metabolismo , Diferenciación Celular , Proteínas de Unión al ADN/fisiología , Predisposición Genética a la Enfermedad , Genotipo , Inmunohistoquímica , Islotes Pancreáticos/citología , Ratones , Ratones Transgénicos , Microscopía Confocal , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/fisiología , Recombinación Genética , Factores de Tiempo
18.
Blood Adv ; 7(11): 2504-2519, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-36705973

RESUMEN

Acute myeloid leukemia (AML) with mixed-lineage leukemia (MLL) gene rearrangement (MLLr) comprises a cellular hierarchy in which a subpopulation of cells serves as functional leukemia stem cells (LSCs). They are maintained by a unique gene expression program and chromatin states, which are thought to reflect the actions of enhancers. Here, we delineate the active enhancer landscape and observe pervasive enhancer malfunction in LSCs. Reconstruction of regulatory networks revealed a master set of hematopoietic transcription factors. We show that EP300 is an essential transcriptional coregulator for maintaining LSC oncogenic potential because it controls essential gene expression through modulation of H3K27 acetylation and assessments of transcription factor dependencies. Moreover, the EP300 inhibitor A-485 affects LSC growth by targeting enhancer activity via histone acetyltransferase domain inhibition. Together, these data implicate a perturbed MLLr-specific enhancer accessibility landscape, suggesting the possibility for disruption of the LSC enhancer regulatory axis as a promising therapeutic strategy in AML.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Cromatina , Regulación de la Expresión Génica , Oncogenes , Células Madre/metabolismo
19.
Cancers (Basel) ; 15(17)2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37686604

RESUMEN

The multi-kinase inhibitor dasatinib has been implicated to be effective in pre-B-cell receptor (pre-BCR)-positive acute lymphoblastic leukemia (ALL) expressing the E2A-PBX1 fusion oncoprotein. The TGFß signaling pathway is involved in a wide variety of cellular processes, including embryonic development and cell homeostasis, and it can have dual roles in cancer: suppressing tumor growth at early stages and mediating tumor progression at later stages. In this study, we identified the upregulation of the TGFß signaling pathway in our previously generated human dasatinib-resistant pre-BCR+/E2A-PBX1+ ALL cells using global transcriptomic analysis. We confirm the upregulation of the TGFß pathway member SMAD3 at the transcriptional and translational levels in dasatinib-resistant pre-BCR+/E2A-PBX1+ ALL cells. Hence, dasatinib blocks, at least partially, TGFß-induced SMAD3 phosphorylation in several B-cell precursor (BCP) ALL cell lines as well as in dasatinib-resistant pre-BCR+/E2A-PBX1+ ALL cells. Activation of the TGFß signaling pathway by TGF-ß1 leads to growth inhibition by cell cycle arrest at the G0/G1 stage, increase in apoptosis and transcriptional changes of SMAD-targeted genes, e.g. c-MYC downregulation, in pre-BCR+/E2A-PBX1+ ALL cells. These results provide a better understanding about the role that the TGFß signaling pathway plays in leukemogenesis of BCP-ALL as well as in secondary drug resistance to dasatinib.

20.
Blood Adv ; 7(22): 7087-7099, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37824841

RESUMEN

Common infections have long been proposed to play a role in the development of pediatric B-cell acute lymphoblastic leukemia (B-ALL). However, epidemiologic studies report contradictory effects of infection exposure on subsequent B-ALL risk, and no specific pathogen has been definitively linked to the disease. A unifying mechanism to explain the divergent outcomes could inform disease prevention strategies. We previously reported that the pattern recognition receptor (PRR) ligand Poly(I:C) exerted effects on B-ALL cells that were distinct from those observed with other nucleic acid-based PRR ligands. Here, using multiple double-stranded RNA (dsRNA) moieties, we show that the overall outcome of exposure to Poly(I:C) reflects the balance of opposing responses induced by its ligation to endosomal and cytoplasmic receptors. This PRR response biology is shared between mouse and human B-ALL and can increase leukemia-initiating cell burden in vivo during the preleukemia phase of B-ALL, primarily through tumor necrosis factor α signaling. The age of the responding immune system further influences the impact of dsRNA exposure on B-ALL cells in both mouse and human settings. Overall, our study demonstrates that potentially proleukemic and antileukemic effects can each be generated by the stimulation of pathogen recognition pathways and indicates a mechanistic explanation for the contrasting epidemiologic associations reported for infection exposure and B-ALL.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras B , Transducción de Señal , Ratones , Humanos , Animales , Niño , Ligandos , ARN Bicatenario/farmacología , Linfocitos B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA