Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 71(1): 103-116.e7, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-30008316

RESUMEN

The control of cell fate is an epigenetic process initiated by transcription factors (TFs) that recognize DNA motifs and recruit activator complexes and transcriptional machineries to chromatin. Lineage specificity is thought to be provided solely by TF-motif pairing, while the recruited activators are passive. Here, we show that INTS13, a subunit of the Integrator complex, operates as monocytic/macrophagic differentiation factor. Integrator is a general activator of transcription at coding genes and is required for eRNA maturation. Here, we show that INTS13 functions as an independent sub-module and targets enhancers through Early Growth Response (EGR1/2) TFs and their co-factor NAB2. INTS13 binds poised monocytic enhancers eliciting chromatin looping and activation. Independent depletion of INTS13, EGR1, or NAB2 impairs monocytic differentiation of cell lines and primary human progenitors. Our data demonstrate that Integrator is not functionally homogeneous and has TF-specific regulatory potential, revealing a new enhancer regulatory axis that controls myeloid differentiation.


Asunto(s)
Diferenciación Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Elementos de Facilitación Genéticos , Monocitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Proteínas Represoras/metabolismo , Línea Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Humanos , Células Progenitoras Mieloides/citología , Proteínas Represoras/genética
2.
Mol Cell ; 66(2): 247-257.e5, 2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28410996

RESUMEN

Recruitment of transcription factors (TFs) to repressed genes in euchromatin is essential to activate new transcriptional programs during cell differentiation. However, recruitment of all TFs, including pioneer factors, is impeded by condensed H3K27me3-containing chromatin. Single-cell and gene-specific analyses revealed that, during the first hours of induction of differentiation of mammalian embryonic stem cells (ESCs), accumulation of the repressive histone mark H3K27me3 is delayed after DNA replication, indicative of a decondensed chromatin structure in all regions of the replicating genome. This delay provides a critical "window of opportunity" for recruitment of lineage-specific TFs to DNA. Increasing the levels of post-replicative H3K27me3 or preventing S phase entry inhibited recruitment of new TFs to DNA and significantly blocked cell differentiation. These findings suggest that recruitment of lineage-specifying TFs occurs soon after replication and is facilitated by a decondensed chromatin structure. This insight may explain the developmental plasticity of stem cells and facilitate their exploitation for therapeutic purposes.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Replicación del ADN , ADN/biosíntesis , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Sitios de Unión , Plasticidad de la Célula , Cromatina/química , ADN/química , ADN/genética , Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas/metabolismo , Histonas/química , Humanos , Metilación , Ratones , Proteínas Nucleares/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Relación Estructura-Actividad , Factores de Tiempo , Factores de Transcripción/genética
3.
Blood ; 135(18): 1560-1573, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32040545

RESUMEN

Expression of the cell cycle regulatory gene CDK6 is required for Philadelphia-positive (Ph+) acute lymphoblastic leukemia (ALL) cell growth, whereas expression of the closely related CDK4 protein is dispensable. Moreover, CDK6 silencing is more effective than treatment with the dual CDK4/6 inhibitor palbociclib in suppressing Ph+ ALL in mice, suggesting that the growth-promoting effects of CDK6 are, in part, kinase-independent in Ph+ ALL. Accordingly, we developed CDK4/6-targeted proteolysis-targeting chimeras (PROTACs) that inhibit CDK6 enzymatic activity in vitro, promote the rapid and preferential degradation of CDK6 over CDK4 in Ph+ ALL cells, and markedly suppress S-phase cells concomitant with inhibition of CDK6-regulated phospho-RB and FOXM1 expression. No such effects were observed in CD34+ normal hematopoietic progenitors, although CDK6 was efficiently degraded. Treatment with the CDK6-degrading PROTAC YX-2-107 markedly suppressed leukemia burden in mice injected with de novo or tyrosine kinase inhibitor-resistant primary Ph+ ALL cells, and this effect was comparable or superior to that of the CDK4/6 enzymatic inhibitor palbociclib. These studies provide "proof of principle" that targeting CDK6 with PROTACs that inhibit its enzymatic activity and promote its degradation represents an effective strategy to exploit the "CDK6 dependence" of Ph+ ALL and, perhaps, of other hematologic malignancies. Moreover, they suggest that treatment of Ph+ ALL with CDK6-selective PROTACs would spare a high proportion of normal hematopoietic progenitors, preventing the neutropenia induced by treatment with dual CDK4/6 inhibitors.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes de Fusión/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Perfilación de la Expresión Génica , Genes cdc , Humanos , Ratones , Estructura Molecular , Fosforilación , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/uso terapéutico , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Haematologica ; 104(1): 82-92, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30076175

RESUMEN

MicroRNAs, non-coding regulators of gene expression, are likely to function as important downstream effectors of many transcription factors including MYB. Optimal levels of MYB are required for transformation/maintenance of BCR-ABL-expressing cells. We investigated whether MYB silencing modulates microRNA expression in Philadelphia-positive (Ph+) leukemia cells and if MYB-regulated microRNAs are important for the "MYB addiction" of these cells. Thirty-five microRNAs were modulated by MYB silencing in lymphoid and erythromyeloid chronic myeloid leukemia-blast crisis BV173 and K562 cells; 15 of these were concordantly modulated in both lines. We focused on the miR-17-92 cluster because of its oncogenic role in tumors and found that: i) it is a direct MYB target; ii) it partially rescued the impaired proliferation and enhanced apoptosis of MYB-silenced BV173 cells. Moreover, we identified FRZB, a Wnt/ß-catenin pathway inhibitor, as a novel target of the miR-17-92 cluster. High expression of MYB in blast cells from 2 Ph+leukemia patients correlated positively with the miR-17-92 cluster and inversely with FRZB. This expression pattern was also observed in a microarray dataset of 122 Ph+acute lymphoblastic leukemias. In vivo experiments in NOD scid gamma mice injected with BV173 cells confirmed that FRZB functions as a Wnt/ß-catenin inhibitor even as they failed to demonstrate that this pathway is important for BV173-dependent leukemogenesis. These studies illustrate the global effects of MYB expression on the microRNAs profile of Ph+cells and supports the concept that the "MYB addiction" of these cells is, in part, caused by modulation of microRNA-regulated pathways affecting cell proliferation and survival.


Asunto(s)
Crisis Blástica/metabolismo , Regulación Leucémica de la Expresión Génica , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , MicroARNs/biosíntesis , Familia de Multigenes , Proteínas Proto-Oncogénicas c-myb/biosíntesis , ARN Neoplásico/biosíntesis , Activación Transcripcional , Animales , Crisis Blástica/tratamiento farmacológico , Crisis Blástica/genética , Crisis Blástica/patología , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , Proteínas Proto-Oncogénicas c-myb/genética , ARN Neoplásico/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Genet ; 30(1): 48-58, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11753385

RESUMEN

The arrest of differentiation is a feature of both chronic myelogenous leukemia cells in myeloid blast crisis and myeloid precursors that ectopically express the p210BCR-ABL oncoprotein; however, its underlying mechanisms remain poorly understood. Here we show that expression of BCR-ABL in myeloid precursor cells leads to transcriptional suppression of the granulocyte colony-stimulating factor receptor G-CSF-R (encoded by CSF3R), possibly through down-modulation of C/EBPalpha-the principal regulator of granulocytic differentiation. Expression of C/EBPalpha protein is barely detectable in primary marrow cells taken from individuals affected with chronic myeloid leukemia in blast crisis. In contrast, CEBPA RNA is clearly present. Ectopic expression of C/EBPalpha induces granulocytic differentiation of myeloid precursor cells expressing BCR-ABL. Expression of C/EBPalpha is suppressed at the translational level by interaction of the poly(rC)-binding protein hnRNP E2 with CEBPA mRNA, and ectopic expression of hnRNP E2 in myeloid precursor cells down-regulates both C/EBPalpha and G-CSF-R and leads to rapid cell death on treatment with G-CSF (encoded by CSF3). Our results indicate that BCR-ABL regulates the expression of C/EBPalpha by inducing hnRNP E2-which inhibits the translation of CEBPA mRNA.


Asunto(s)
Apoptosis/fisiología , Proteína alfa Potenciadora de Unión a CCAAT/biosíntesis , Proteínas de Unión al ADN , Proteínas de Fusión bcr-abl/fisiología , Regulación de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas , Proteínas de Unión al ARN/fisiología , Receptores de Factor Estimulante de Colonias de Granulocito/biosíntesis , Factores de Transcripción , Animales , Apoptosis/genética , Benzamidas , Crisis Blástica/metabolismo , Crisis Blástica/patología , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT , Proteínas Portadoras/metabolismo , Células Cultivadas/metabolismo , Regulación hacia Abajo , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Células Mieloides/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/metabolismo , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/farmacología , Piperazinas/farmacología , Biosíntesis de Proteínas , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Pirimidinas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/aislamiento & purificación , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Transcripción Genética , Transfección
6.
Leuk Res ; 129: 107075, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37079999

RESUMEN

PURPOSE: Despite advances in the treatment of B-cell acute lymphoblastic leukemia (B-ALL), outcomes for relapsed/refractory (R/R) disease remain poor. Preclinical studies suggest that the combination of the CDK4/6 inhibitor palbociclib and dexamethasone may be effective in targeting leukemic cell growth. We conducted a phase I study of escalating doses of palbociclib in combination with dexamethasone in adults with R/R B-ALL. METHODS: Cycle 1 consisted of single agent palbociclib given for 7 days and continued for 28 additional days in combination with dexamethasone 20 mg daily. Palbociclib dosing began at 100 mg daily. Patients with a response were eligible for maintenance consisting of 1 week of palbociclib plus dexamethasone (20 mg daily × 2 days, 16 mg daily × 2 days, 12 mg daily × 2 days, 6 mg daily × 1 day), followed by 3 weeks of palbociclib alone. Safety, efficacy, and the expression of phospho-RB and c-MYB/BCL-2 were measured. CONCLUSIONS: Seven patients were treated on study before it was closed early due to slow accrual. No dose limiting toxicities were identified. One patient had a complete response with incomplete hematologic recovery, suggesting possible efficacy of the treatment. Reduction in CD34+ cells, p-RB, c-MYB, and BCL-2 expression also suggested on-target therapy effects.


Asunto(s)
Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Adulto , Humanos , Linfoma de Células B/tratamiento farmacológico , Piridinas/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Dexametasona , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos
7.
J Cell Biochem ; 113(5): 1704-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22212957

RESUMEN

The CCAAT/enhancer binding protein α (C/EBPα) is a transcription factor required for differentiation of myeloid progenitors. In acute myeloid leukemia (AML) cells expressing the constitutively active FLT3-ITD receptor tyrosine kinase, MAP kinase-dependent phosphorylation of serine 21 (S21) inhibits the ability of C/EBPα to induce granulocytic differentiation. To assess whether this post-translational modification also modulates the activity of C/EBPα in BCR/ABL-expressing cells, we tested the biological effects of wild-type and mutant C/EBPα mimicking phosphorylated or non-phosphorylatable serine 21 (S21D and S21A, respectively) in K562 cells ectopically expressing tamoxifen-regulated C/EBPα-ER chimeric proteins. We show here that S21D C/EBPα-ER induced terminal granulocytic differentiation of K562 cells almost as well as wild-type C/EBPα-ER, while S21A C/EBPα-ER was less efficient. Furthermore, wild-type C/EBPα suppressed the proliferation and colony formation of K562 cells vigorously, while S21D and S21A C/EBPα mutants had more modest anti-proliferative effects. Both mutants were less effective than wild-type C/EBPα in suppressing endogenous E2F-dependent transactivation and bound less E2F-2 and/or E2F-3 proteins in anti-C/EBPα immunoprecipitates. Together, these findings suggest that mutation of S21 more than its phosphorylation inhibits the anti-proliferative effects of C/EBPα due to reduced interaction with or impaired regulation of the activity of E2F proteins. By contrast, phosphorylation of serine 21 appears to have a modest role in modulating the differentiation-inducing effects of C/EBPα in K562 cells.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/química , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Sustitución de Aminoácidos , Secuencia de Bases , Benzamidas/farmacología , Proteína alfa Potenciadora de Unión a CCAAT/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Cartilla de ADN/genética , Factores de Transcripción E2F/metabolismo , Granulocitos/metabolismo , Granulocitos/patología , Humanos , Células K562 , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutagénesis Sitio-Dirigida , Fosforilación , Regiones Promotoras Genéticas , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Serina/química , Ensayo de Tumor de Célula Madre
8.
Cancer Cell ; 3(2): 145-60, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12620409

RESUMEN

In a BCR/ABL-expressing myeloid precursor cell line, p53 levels were markedly downmodulated. Expression of MDM2, the negative regulator of p53, was upregulated in a tyrosine kinase-dependent manner in growth factor-independent BCR/ABL-expressing cells, and in accelerated phase and blast crisis CML samples. Increased MDM2 expression was associated with enhanced mdm2 mRNA translation, which required the interaction of the La antigen with mdm2 5' UTR. Expression of MDM2 correlated with that of La and was suppressed by La siRNAs and by a dominant negative La mutant, which also enhanced the susceptibility to drug-induced apoptosis of BCR/ABL-transformed cells. By contrast, La overexpression led to increased MDM2 levels and enhanced resistance to apoptosis. Thus, La-dependent activation of mdm2 translation might represent an important molecular mechanism involved in BCR/ABL leukemogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas de Fusión bcr-abl/fisiología , Proteínas Nucleares , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/metabolismo , Ribonucleoproteínas/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autoantígenos , Northern Blotting , Western Blotting , Resistencia a Antineoplásicos , Proteína Adaptadora GRB2 , Sustancias de Crecimiento/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Biosíntesis de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleoproteínas/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Antígeno SS-B
9.
Exp Cell Res ; 317(4): 488-95, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21111732

RESUMEN

MicroRNA145 (miR145), a tumor suppressor miR, has been reported to inhibit growth of human cancer cells, to induce differentiation and to cause apoptosis, all conditions that result in growth arrest. In order to clarify the functional effects of miR145, we have investigated its expression in diverse conditions and different cell lines. Our results show that miR145 levels definitely increase in differentiating cells and also in growth-arrested cells, even in the absence of differentiation. Increased expression during differentiation sometimes occurs as a late event, suggesting that miR145 could be required either early or late during the differentiation process.


Asunto(s)
Diferenciación Celular/genética , Inhibidores de Crecimiento/análisis , MicroARNs/análisis , Regulación hacia Arriba , Apoptosis , Butiratos/farmacología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Ciclo Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Inhibidores de Crecimiento/farmacología , Humanos , Cloruro de Litio/farmacología , MicroARNs/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc , Tretinoina/farmacología , Regulación hacia Arriba/efectos de los fármacos
10.
Cancer Res ; 82(3): 458-471, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34903608

RESUMEN

Despite treatment with intensive chemotherapy, acute myelogenous leukemia (AML) remains an aggressive malignancy with a dismal outcome in most patients. We found that AML cells exhibit an unusually rapid accumulation of the repressive histone mark H3K27me3 on nascent DNA. In cell lines, primary cells and xenograft mouse models, inhibition of the H3K27 histone methyltransferase EZH2 to decondense the H3K27me3-marked chromatin of AML cells enhanced chromatin accessibility and chemotherapy-induced DNA damage, apoptosis, and leukemia suppression. These effects were further promoted when chromatin decondensation of AML cells was induced upon S-phase entry after release from a transient G1 arrest mediated by CDK4/6 inhibition. In the p53-null KG-1 and THP-1 AML cell lines, EZH2 inhibitor and doxorubicin cotreatment induced transcriptional reprogramming that was, in part, dependent on derepression of H3K27me3-marked gene promoters and led to increased expression of cell death-promoting and growth-inhibitory genes.In conclusion, decondensing H3K27me3-marked chromatin by EZH2 inhibition represents a promising approach to improve the efficacy of DNA-damaging cytotoxic agents in patients with AML. This strategy might allow for a lowering of chemotherapy doses, with a consequent reduction of treatment-related side effects in elderly patients with AML or those with significant comorbidities. SIGNIFICANCE: Pharmacological inhibition of EZH2 renders DNA of AML cells more accessible to cytotoxic agents, facilitating leukemia suppression with reduced doses of chemotherapy.See related commentary by Adema and Colla, p. 359.


Asunto(s)
Cromatina/metabolismo , Histonas/metabolismo , Leucemia Mieloide Aguda/genética , Animales , Humanos , Ratones
11.
J Biol Chem ; 285(38): 29434-45, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20622260

RESUMEN

In metastatic cancer cells, the process of invasion is regulated by several transcription factors that induce changes required for migration and resistance to apoptosis. Slug (SNAI2, Snail2) is involved in epithelial mesenchymal transition in physiological and in pathological contexts. We show here that in embryonic kidney, colon carcinoma, chronic myeloid leukemia-blast crisis, and in neuroblastoma cells, expression of Slug is transcriptionally regulated by c-Myb via Myb binding sites in the 5'-flanking region and in the first intron of the slug gene. In embryonic kidney and neuroblastoma cells, c-Myb induced vimentin, fibronectin, and N-cadherin expression and membrane ruffling via actin polymerization consistent with the acquisition of a mesenchymal-like phenotype. Furthermore, down-regulation of endogenous c-Myb levels in colon carcinoma cells led to increased expression of E-cadherin and reduced levels of vimentin. Some of these changes are predominantly Slug-dependent as Slug silencing via RNA interference (RNAi) reverts the cells to a quasi-parental condition. Changes in gene expression and morphology induced by c-Myb-activated Slug correlated with increased ability to migrate (embryonic kidney) and to invade through a Matrigel membrane (embryonic kidney, colon carcinoma, neuroblastoma). c-Myb-dependent Slug expression was also essential for the homing of chronic myeloid leukemia K562 cells to the bone marrow. In summary, we show here that the proto-oncogene c-Myb controls Slug transcription in tumor cells of different origin. Such a regulatory pathway contributes to the acquisition of invasive properties that are important for the metastatic process.


Asunto(s)
Médula Ósea/patología , Proteínas Proto-Oncogénicas c-myb/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Etopósido/farmacología , Citometría de Flujo , Humanos , Intrones/genética , Ratones , Ratones SCID , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/fisiopatología , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myb/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
12.
J Biol Chem ; 285(40): 30837-50, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20659895

RESUMEN

The transcription factor C/EBPα is more potent than C/EBPß in inducing granulocitic differentiation and inhibiting BCR/ABL-expressing cells. We took a "domain swapping" approach to assess biological effects, modulation of gene expression, and binding to C/EBPα-regulated promoters by wild-type and chimeric C/EBPα/C/EBPß proteins. Wild-type and N-C/EBPα+ C/EBPß-DBD induced transcription of the granulocyte-colony stimulating factor receptor (G-CSFR) gene, promoted differentiation, and suppressed proliferation of K562 cells vigorously; instead, wild-type C/EBPß and N-C/EBPß+C/EBPα-DBD had modest effects, although they bound the G-CSFR promoter like wild-type C/EBPα and N-C/EBPα+C/EBPß-DBD. Chimeric proteins consisting of the TAD of VP16 and the DBD of C/EBPα or C/EBPß inhibited proliferation and induced differentiation of K562 cells as effectively as wild-type C/EBPα. Gene expression profiles induced by C/EBPα resembled those modulated by N-C/EBPα+C/EBPß-DBD, whereas C/EBPß induced a pattern similar to that of N-C/EBPß+C/EBPα-DBD. C/EBPα activation induced changes in the expression of more cell cycle- and apoptosis-related genes than the other proteins and enhanced Imatinib-induced apoptosis of K562 cells. Expression of FOXO3a, a novel C/EBPα-regulated gene, was required for apoptosis but not for differentiation induction or proliferation inhibition of K562 cells.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Ciclo Celular , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Transcripción Genética , Apoptosis/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular/genética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/genética , Humanos , Células K562 , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
13.
Genes (Basel) ; 12(9)2021 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-34573335

RESUMEN

Ph+ ALL is a poor-prognosis leukemia subtype driven by the BCR-ABL1 oncogene, either the p190- or the p210-BCR/ABL isoform in a 70:30 ratio. Tyrosine Kinase inhibitors (TKIs) are the drugs of choice in the therapy of Ph+ ALL. In combination with standard chemotherapy, TKIs have markedly improved the outcome of Ph+ ALL, in particular if this treatment is followed by bone marrow transplantation. However, resistance to TKIs develops with high frequency, causing leukemia relapse that results in <5-year overall survival. Thus, new therapies are needed to address relapsed/TKI-resistant Ph+ ALL. We have shown that expression of cell cycle regulatory kinase CDK6, but not of the highly related CDK4 kinase, is required for the proliferation and survival of Ph+ ALL cells. Comparison of leukemia suppression induced by treatment with the clinically-approved dual CDK4/6 inhibitor palbociclib versus CDK6 silencing revealed that the latter treatment was markedly more effective, probably reflecting inhibition of CDK6 kinase-independent effects. Thus, we developed CDK4/6-targeted proteolysis-targeting chimeras (PROTACs) that preferentially degrade CDK6 over CDK4. One compound termed PROTAC YX-2-107, which degrades CDK6 by recruiting the Cereblon ubiquitin ligase, markedly suppressed leukemia burden in mice injected with de novo or TKI-resistant Ph+ ALL. The effect of PROTAC YX-2-107 was comparable or superior to that of palbociclib. The development of CDK6-selective PROTACs represents an effective strategy to exploit the "CDK6 dependence" of Ph+ ALL cells while sparing a high proportion of normal hematopoietic progenitors that depend on both CDK6 and CDK6 for their survival. In combination with other agents, CDK6-selective PROTACs may be valuable components of chemotherapy-free protocols for the therapy of Ph+ ALL and other CDK6-dependent hematological malignancies.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 6 Dependiente de la Ciclina , Terapia Molecular Dirigida/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo
14.
Nat Commun ; 12(1): 6469, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753942

RESUMEN

Subunit switches in the BAF chromatin remodeler are essential during development. ARID1B and its paralog ARID1A encode for mutually exclusive BAF subunits. De novo ARID1B haploinsufficient mutations cause neurodevelopmental disorders, including Coffin-Siris syndrome, which is characterized by neurological and craniofacial features. Here, we leveraged ARID1B+/- Coffin-Siris patient-derived iPSCs and modeled cranial neural crest cell (CNCC) formation. We discovered that ARID1B is active only during the first stage of this process, coinciding with neuroectoderm specification, where it is part of a lineage-specific BAF configuration (ARID1B-BAF). ARID1B-BAF regulates exit from pluripotency and lineage commitment by attenuating thousands of enhancers and genes of the NANOG and SOX2 networks. In iPSCs, these enhancers are maintained active by ARID1A-containing BAF. At the onset of differentiation, cells transition from ARID1A- to ARID1B-BAF, eliciting attenuation of the NANOG/SOX2 networks and triggering pluripotency exit. Coffin-Siris patient cells fail to perform the ARID1A/ARID1B switch, and maintain ARID1A-BAF at the pluripotency enhancers throughout all stages of CNCC formation. This leads to persistent NANOG/SOX2 activity which impairs CNCC formation. Despite showing the typical neural crest signature (TFAP2A/SOX9-positive), ARID1B-haploinsufficient CNCCs are also aberrantly NANOG-positive. These findings suggest a connection between ARID1B mutations, neuroectoderm specification and a pathogenic mechanism for Coffin-Siris syndrome.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína Homeótica Nanog/metabolismo , Cresta Neural/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Proteínas de Unión al ADN/genética , Citometría de Flujo , Células HEK293 , Humanos , Mutación/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética
15.
Blood ; 111(9): 4771-9, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18227349

RESUMEN

The c-Myb gene encodes a transcription factor required for proliferation and survival of normal myeloid progenitors and leukemic blast cells. Targeting of c-Myb by antisense oligodeoxynucleotides has suggested that myeloid leukemia blasts (including chronic myelogenous leukemia [CML]-blast crisis cells) rely on c-Myb expression more than normal progenitors, but a genetic approach to assess the requirement of c-Myb by p210(BCR/ABL)-transformed hematopoietic progenitors has not been taken. We show here that loss of a c-Myb allele had modest effects (20%-28% decrease) on colony formation of nontransduced progenitors, while the effect on p210(BCR/ABL)-expressing Lin(-) Sca-1(+) and Lin(-) Sca-1(+)Kit(+) cells was more pronounced (50%-80% decrease). Using a model of CML-blast crisis, mice (n = 14) injected with p210(BCR/ABL)-transduced p53(-/-)c-Myb(w/w) marrow cells developed leukemia rapidly and had a median survival of 26 days, while only 67% of mice (n = 12) injected with p210(BCR/ABL)-transduced p53(-/-)c-Myb(w/d) marrow cells died of leukemia with a median survival of 96 days. p210(BCR/ABL)-transduced c-Myb(w/w) and c-Myb(w/d) marrow progenitors expressed similar levels of the c-Myb-regulated genes c-Myc and cyclin B1, while those of Bcl-2 were reduced. However, ectopic Bcl-2 expression did not enhance colony formation of p210(BCR/ABL)-transduced c-Myb(w/d) Lin(-)Sca-1(+)Kit(+) cells. Together, these studies support the requirement of c-Myb for p210(BCR/ABL)-dependent leukemogenesis.


Asunto(s)
Transformación Celular Neoplásica , Proteínas de Fusión bcr-abl/fisiología , Células Madre Hematopoyéticas/patología , Leucemia/etiología , Proteínas Proto-Oncogénicas c-myb/fisiología , Animales , Proteínas de Fusión bcr-abl/administración & dosificación , Proteínas de Fusión bcr-abl/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Leucemia/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Transducción Genética
16.
Blood ; 112(5): 1942-50, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18550858

RESUMEN

Ectopic C/EBPalpha expression in p210(BCR/ABL)-expressing hematopoietic cells induces granulocytic differentiation, inhibits proliferation, and suppresses leukemogenesis. To assess the underlying mechanisms, C/EBPalpha targets were identified by microarray analyses. Upon C/EBPalpha activation, expression of c-Myb and GATA-2 was repressed in 32D-BCR/ABL, K562, and chronic myelogenous leukemia (CML) blast crisis (BC) primary cells but only c-Myb levels decreased slightly in CD34(+) normal progenitors. The role of these 2 genes for the effects of C/EBPalpha was assessed by perturbing their expression in K562 cells. Ectopic c-Myb expression blocked the proliferation inhibition- and differentiation-inducing effects of C/EBPalpha, whereas c-Myb siRNA treatment enhanced C/EBPalpha-mediated proliferation inhibition and induced changes in gene expression indicative of monocytic differentiation. Ectopic GATA-2 expression suppressed the proliferation inhibitory effect of C/EBPalpha but blocked in part the effect on differentiation; GATA-2 siRNA treatment had no effects on C/EBPalpha induction of differentiation but inhibited proliferation of K562 cells, alone or upon C/EBPalpha activation. In summary, the effects of C/EBPalpha in p210(BCR/ABL)-expressing cells depend, in part, on transcriptional repression of c-Myb and GATA-2. Since perturbation of c-Myb and GATA-2 expression has nonidentical consequences for proliferation and differentiation of K562 cells, the effects of C/EBPalpha appear to involve dif-ferent transcription-regulated targets.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/farmacología , Proteínas de Fusión bcr-abl/biosíntesis , Factor de Transcripción GATA2/genética , Genes myb/efectos de los fármacos , Secuencia de Bases , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cartilla de ADN/genética , Proteínas de Fusión bcr-abl/genética , Genes abl , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , ARN Interferente Pequeño/genética , Transcripción Genética/efectos de los fármacos , Transfección
17.
Blood Cancer Discov ; 1(1): 48-67, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32974613

RESUMEN

Persistence of drug-resistant quiescent leukemic stem cells (LSC) and impaired natural killer (NK) cell immune response account for relapse of chronic myelogenous leukemia (CML). Inactivation of protein phosphatase 2A (PP2A) is essential for CML-quiescent LSC survival and NK cell antitumor activity. Here we show that MIR300 has antiproliferative and PP2A-activating functions that are dose dependently differentially induced by CCND2/CDK6 and SET inhibition, respectively. MIR300 is upregulated in CML LSCs and NK cells by bone marrow microenvironment (BMM) signals to induce quiescence and impair immune response, respectively. Conversely, BCR-ABL1 downregulates MIR300 in CML progenitors to prevent growth arrest and PP2A-mediated apoptosis. Quiescent LSCs escape apoptosis by upregulating TUG1 long noncoding RNA that uncouples and limits MIR300 function to cytostasis. Genetic and pharmacologic MIR300 modulation and/or PP2A-activating drug treatment restore NK cell activity, inhibit BMM-induced growth arrest, and selectively trigger LSC apoptosis in vitro and in patient-derived xenografts; hence, the importance of MIR300 and PP2A activity for CML development and therapy.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , MicroARNs , Humanos , Células Asesinas Naturales , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , MicroARNs/genética , Células Madre Neoplásicas , Inhibidores de Proteínas Quinasas/metabolismo , Proteína Fosfatasa 2/genética , Microambiente Tumoral/genética
18.
Mol Cancer ; 8: 58, 2009 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-19646290

RESUMEN

BACKGROUND: Akt/PKB is a serine/threonine kinase that has attracted much attention because of its central role in regulating cell proliferation, survival, motility and angiogenesis. Activation of Akt in breast cancer portends aggressive tumour behaviour, resistance to hormone-, chemo-, and radiotherapy-induced apoptosis and it is correlated with decreased overall survival. Recent studies have identified novel tumor-specific substrates of Akt that may provide new diagnostic and prognostic markers and serve as therapeutic targets. This study was undertaken to identify pAkt-interacting proteins and to assess their biological roles in breast cancer cells. RESULTS: We confirmed that one of the pAkt interacting proteins is the Elongation Factor EF1alpha. EF1alpha contains a putative Akt phosphorylation site, but is not phosphorylated by pAkt1 or pAkt2, suggesting that it may function as a modulator of pAkt activity. Indeed, downregulation of EF1alpha expression by siRNAs led to markedly decreased expression of pAkt1 and to less extent of pAkt2 and was associated with reduced proliferation, survival and invasion of HCC1937 cells. Proliferation and survival was further reduced by combining EF1alpha siRNAs with specific pAkt inhibitors whereas EF1alpha downregulation slightly attenuated the decreased invasion induced by Akt inhibitors. CONCLUSION: We show here that EF1alpha is a pAkt-interacting protein which regulates pAkt levels. Since EF1alpha is often overexpressed in breast cancer, the consequences of EF1alpha increased levels for proliferation, survival and invasion will likely depend on the relative concentration of Akt1 and Akt2.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor 1 de Elongación Peptídica/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Supervivencia Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoprecipitación , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , Factor 1 de Elongación Peptídica/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Interferencia de ARN
19.
Int J Cancer ; 125(11): 2547-55, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19623650

RESUMEN

Stage 4 neuroblastoma (NB) is a devastating childhood cancer whose poor outcome has remained essentially unchanged in the last 20 years. Receptor tyrosine kinases have important roles in the control of proliferation, differentiation and apoptosis of NB cells. Thus, we tested the activity of second-generation tyrosine kinase inhibitor Dasatinib in human NB cell lines in vitro and in an orthotopic mouse model. Dasatinib inhibited cell viability with an IC(50) in the submicromolar range in 7 of 10 tested cell lines. In sensitive cells, Dasatinib reduced anchorage-independent growth and, in some instances, induced senescence and apoptosis. In HTLA-230 cells, Dasatinib treatment caused down-regulation of c-Kit and c-Src phosphorylation in conjunction with strong inhibition of Erk1/2 and Akt activity. To test the efficacy of Dasatinib in vivo, HTLA-230 and SY5Y cells were orthotopically injected in the adrenal gland of nude mice and drug treatments carried out until day 40. In mice injected with HTLA-230 cells, tumour growth was significantly inhibited at the dose of 30 mg/(kg day) when treatment was started 7 days after injection. In animals injected with SY5Y cells that were exquisitely sensitive in vitro (IC(50)= 92 nM), the antitumour effect of Dasatinib was observed at the dose of 60 mg/(kg day) but only when treatment was started 1 day after injection. However, the anti-tumour effect of Dasatinib in vivo was partial in both orthotopic models, emphasizing the importance of testing candidate new drugs in animal environments closely mimicking the human tumour.


Asunto(s)
Modelos Animales de Enfermedad , Neuroblastoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Senescencia Celular , Dasatinib , Femenino , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Técnicas In Vitro , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fosforilación , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Clin Cancer Res ; 14(14): 4622-30, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18628477

RESUMEN

PURPOSE: We assessed the relevance of Slug (SNAI2) for apoptosis resistance and invasion potential of neuroblastoma cells in vitro and in vivo. EXPERIMENTAL DESIGN: We evaluated the effect of imatinib mesylate on invasion and analyzed the genes modulated by imatinib mesylate treatment in neuroblastoma cells. Slug expression, inhibited by imatinib mesylate treatment, was knocked down in neuroblastoma cells by RNA interference, and the effects on invasion and apoptosis were evaluated in vitro. A pseudometastatic model of neuroblastoma in severe combined immunodeficient mice was used to assess the effects of Slug silencing alone or in combination with imatinib mesylate treatment on metastasis development. RESULTS: Microarray analysis revealed that several genes, including Slug, were down-regulated by imatinib mesylate. Slug expression was detectable in 8 of 10 human neuroblastoma cell lines. Two Slug-expressing cell lines were infected with a vector encoding a microRNA to Slug mRNA. Infected cells with reduced levels of Slug were tested for the expression of apoptosis-related genes (p53, Bax, and Bcl-2) identified previously as Slug targets. Bcl-2 was down-regulated in Slug-interfered cells. Slug down-regulation increased sensitivity to apoptosis induced by imatinib mesylate, etoposide, or doxorubicin. Invasion of Slug-silenced cells was reduced in vitro. Animals injected with Slug-silenced cells had fewer tumors than controls and the inhibition of tumor growth was even higher in animals treated with imatinib mesylate. CONCLUSIONS: Slug down-regulation facilitates apoptosis induced by proapoptotic drugs in neuroblastoma cells and decreases their invasion capability in vitro and in vivo. Slug inhibition, possibly combined with imatinib mesylate, may represent a novel strategy for treatment of metastatic neuroblastoma.


Asunto(s)
Apoptosis/fisiología , Invasividad Neoplásica , Neuroblastoma/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/farmacología , Benzamidas , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Mesilato de Imatinib , Ratones , Neuroblastoma/tratamiento farmacológico , Análisis de Secuencia por Matrices de Oligonucleótidos , Piperazinas/farmacología , Pirimidinas/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA