Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Immunol ; 202(12): 3434-3446, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31068388

RESUMEN

Development of lymphoid progenitors requires a coordinated regulation of gene expression, DNA replication, and gene rearrangement. Chromatin-remodeling activities directed by SWI/SNF2 superfamily complexes play important roles in these processes. In this study, we used a conditional knockout mouse model to investigate the role of Smarca5, a member of the ISWI subfamily of such complexes, in early lymphocyte development. Smarca5 deficiency results in a developmental block at the DN3 stage of αß thymocytes and pro-B stage of early B cells at which the rearrangement of Ag receptor loci occurs. It also disturbs the development of committed (CD73+) γδ thymocytes. The αß thymocyte block is accompanied by massive apoptotic depletion of ß-selected double-negative DN3 cells and premitotic arrest of CD4/CD8 double-positive cells. Although Smarca5-deficient αß T cell precursors that survived apoptosis were able to undergo a successful TCRß rearrangement, they exhibited a highly abnormal mRNA profile, including the persistent expression of CD44 and CD25 markers characteristic of immature cells. We also observed that the p53 pathway became activated in these cells and that a deficiency of p53 partially rescued the defect in thymus cellularity (in contrast to early B cells) of Smarca5-deficient mice. However, the activation of p53 was not primarily responsible for the thymocyte developmental defects observed in the Smarca5 mutants. Our results indicate that Smarca5 plays a key role in the development of thymocytes undergoing ß-selection, γδ thymocytes, and also B cell progenitors by regulating the transcription of early differentiation programs.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Linfocitos B/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Células Progenitoras Linfoides/fisiología , Linfocitos T/fisiología , Timocitos/fisiología , Adenosina Trifosfatasas/genética , Animales , Diferenciación Celular , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Selección Clonal Mediada por Antígenos , Reordenamiento Génico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
2.
Int J Mol Sci ; 21(6)2020 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-32197313

RESUMEN

ISWI chromatin remodeling ATPase SMARCA5 (SNF2H) is a well-known factor for its role in regulation of DNA access via nucleosome sliding and assembly. SMARCA5 transcriptionally inhibits the myeloid master regulator PU.1. Upregulation of SMARCA5 was previously observed in CD34+ hematopoietic progenitors of acute myeloid leukemia (AML) patients. Since high levels of SMARCA5 are necessary for intensive cell proliferation and cell cycle progression of developing hematopoietic stem and progenitor cells in mice, we reasoned that removal of SMARCA5 enzymatic activity could affect the cycling or undifferentiated state of leukemic progenitor-like clones. Indeed, we observed that CRISPR/cas9-mediated SMARCA5 knockout in AML cell lines (S5KO) inhibited the cell cycle progression. We also observed that the SMARCA5 deletion induced karyorrhexis and nuclear budding as well as increased the ploidy, indicating its role in mitotic division of AML cells. The cytogenetic analysis of S5KO cells revealed the premature chromatid separation. We conclude that deleting SMARCA5 in AML blocks leukemic proliferation and chromatid cohesion.


Asunto(s)
Adenosina Trifosfatasas/deficiencia , Proliferación Celular , Cromátides , Proteínas Cromosómicas no Histona/deficiencia , Técnicas de Inactivación de Genes , Leucemia Mieloide Aguda , Proteínas de Neoplasias , Adenosina Trifosfatasas/metabolismo , Línea Celular Tumoral , Cromátides/genética , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Femenino , Humanos , Células K562 , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/metabolismo
3.
Development ; 143(11): 1937-47, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27246713

RESUMEN

Ocular lens morphogenesis is a model for investigating mechanisms of cellular differentiation, spatial and temporal gene expression control, and chromatin regulation. Brg1 (Smarca4) and Snf2h (Smarca5) are catalytic subunits of distinct ATP-dependent chromatin remodeling complexes implicated in transcriptional regulation. Previous studies have shown that Brg1 regulates both lens fiber cell differentiation and organized degradation of their nuclei (denucleation). Here, we employed a conditional Snf2h(flox) mouse model to probe the cellular and molecular mechanisms of lens formation. Depletion of Snf2h induces premature and expanded differentiation of lens precursor cells forming the lens vesicle, implicating Snf2h as a key regulator of lens vesicle polarity through spatial control of Prox1, Jag1, p27(Kip1) (Cdkn1b) and p57(Kip2) (Cdkn1c) gene expression. The abnormal Snf2h(-/-) fiber cells also retain their nuclei. RNA profiling of Snf2h(-/) (-) and Brg1(-/-) eyes revealed differences in multiple transcripts, including prominent downregulation of those encoding Hsf4 and DNase IIß, which are implicated in the denucleation process. In summary, our data suggest that Snf2h is essential for the establishment of lens vesicle polarity, partitioning of prospective lens epithelial and fiber cell compartments, lens fiber cell differentiation, and lens fiber cell nuclear degradation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Diferenciación Celular , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Embrión de Mamíferos/metabolismo , Cristalino/citología , Cristalino/embriología , Animales , Autofagia , Compartimento Celular , Ciclo Celular , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción del Choque Térmico , Ratones Noqueados , Mitofagia , Modelos Biológicos , Mutación/genética , Proteínas Nucleares/metabolismo , Factor de Transcripción PAX6/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma/genética
4.
Stem Cells ; 35(6): 1614-1623, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28276606

RESUMEN

The imitation switch nuclear ATPase Smarca5 (Snf2h) is one of the most conserved chromatin remodeling factors. It exists in a variety of oligosubunit complexes that move DNA with respect to the histone octamer to generate regularly spaced nucleosomal arrays. Smarca5 interacts with different accessory proteins and represents a molecular motor for DNA replication, repair, and transcription. We deleted Smarca5 at the onset of definitive hematopoiesis (Vav1-iCre) and observed that animals die during late fetal development due to anemia. Hematopoietic stem and progenitor cells accumulated but their maturation toward erythroid and myeloid lineages was inhibited. Proerythroblasts were dysplastic while basophilic erythroblasts were blocked in G2/M and depleted. Smarca5 deficiency led to increased p53 levels, its activation at two residues, one associated with DNA damage (S15Ph °s ) second with CBP/p300 (K376Ac ), and finally activation of the p53 targets. We also deleted Smarca5 in committed erythroid cells (Epor-iCre) and observed that animals were anemic postnatally. Furthermore, 4-hydroxytamoxifen-mediated deletion of Smarca5 in the ex vivo cultures confirmed its requirement for erythroid cell proliferation. Thus, Smarca5 plays indispensable roles during early hematopoiesis and erythropoiesis. Stem Cells 2017;35:1614-1623.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Diferenciación Celular , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Adenosina Trifosfatasas/deficiencia , Anemia/patología , Animales , Ciclo Celular , Proliferación Celular , Proteínas Cromosómicas no Histona/deficiencia , Daño del ADN/genética , Células Eritroides/citología , Eritropoyesis , Eliminación de Gen , Genotipo , Hematopoyesis , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
5.
EMBO J ; 30(21): 4450-64, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21897363

RESUMEN

The oncogenic cluster miR-17-92 encodes seven related microRNAs that regulate cell proliferation, apoptosis and development. Expression of miR-17-92 cluster is decreased upon cell differentiation. Here, we report a novel mechanism of the regulation of miR-17-92 cluster. Using transgenic PU.1(-/-) myeloid progenitors we show that upon macrophage differentiation, the transcription factor PU.1 induces the secondary determinant Egr2 which, in turn, directly represses miR-17-92 expression by recruiting histone demethylase Jarid1b leading to histone H3 lysine K4 demethylation within the CpG island at the miR-17-92 promoter. Conversely, Egr2 itself is targeted by miR-17-92, indicating existence of mutual regulatory relationship between miR-17-92 and Egr2. Furthermore, restoring EGR2 levels in primary acute myeloid leukaemia blasts expressing elevated levels of miR-17-92 and low levels of PU.1 and EGR2 leads to downregulation of miR-17-92 and restored expression of its targets p21CIP1 and BIM. We propose that upon macrophage differentiation PU.1 represses the miR-17-92 cluster promoter by an Egr-2/Jarid1b-mediated H3K4 demethylation mechanism whose deregulation may contribute to leukaemic states.


Asunto(s)
Diferenciación Celular/genética , Epigénesis Genética/fisiología , Macrófagos/fisiología , MicroARNs/genética , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Animales , Secuencia de Bases , Células Cultivadas , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/fisiología , Silenciador del Gen/fisiología , Técnicas de Transferencia de Gen , Células HL-60 , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Macrófagos/metabolismo , Ratones , MicroARNs/metabolismo , Modelos Biológicos , Familia de Multigenes/genética , Células 3T3 NIH , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Largo no Codificante , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Homología de Secuencia de Ácido Nucleico , Transactivadores/genética , Transactivadores/metabolismo , Transfección
6.
Commun Biol ; 7(1): 244, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38424235

RESUMEN

The formation of hematopoietic cells relies on the chromatin remodeling activities of ISWI ATPase SMARCA5 (SNF2H) and its complexes. The Smarca5 null and conditional alleles have been used to study its functions in embryonic and organ development in mice. These mouse model phenotypes vary from embryonic lethality of constitutive knockout to less severe phenotypes observed in tissue-specific Smarca5 deletions, e.g., in the hematopoietic system. Here we show that, in a gene dosage-dependent manner, the hypomorphic allele of SMARCA5 (S5tg) can rescue not only the developmental arrest in hematopoiesis in the hCD2iCre model but also the lethal phenotypes associated with constitutive Smarca5 deletion or Vav1iCre-driven conditional knockout in hematopoietic progenitor cells. Interestingly, the latter model also provided evidence for the role of SMARCA5 expression level in hematopoietic stem cells, as the Vav1iCre S5tg animals accumulate stem and progenitor cells. Furthermore, their hematopoietic stem cells exhibited impaired lymphoid lineage entry and differentiation. This observation contrasts with the myeloid lineage which is developing without significant disturbances. Our findings indicate that animals with low expression of SMARCA5 exhibit normal embryonic development with altered lymphoid entry within the hematopoietic stem cell compartment.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Ratones , Animales , Células Madre Hematopoyéticas/metabolismo , Hematopoyesis/genética , Diferenciación Celular/genética , Adenosina Trifosfatasas/metabolismo
7.
Blood ; 117(14): 3816-25, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21296997

RESUMEN

Elevated levels of microRNA miR-155 represent a candidate pathogenic factor in chronic B-lymphocytic leukemia (B-CLL). In this study, we present evidence that MYB (v-myb myeloblastosis viral oncogene homolog) is overexpressed in a subset of B-CLL patients. MYB physically associates with the promoter of miR-155 host gene (MIR155HG, also known as BIC, B-cell integration cluster) and stimulates its transcription. This coincides with the hypermethylated histone H3K4 residue and spread hyperacetylation of H3K9 at MIR155HG promoter. Our data provide evidence of oncogenic activities of MYB in B-CLL that include its stimulatory role in MIR155HG transcription.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/genética , MicroARNs/genética , Proteínas Oncogénicas v-myb/fisiología , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Células HeLa , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Análisis por Micromatrices , Proteínas Oncogénicas v-myb/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Transcripción Genética/fisiología , Transfección , Células Tumorales Cultivadas
8.
Cells ; 11(2)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35053339

RESUMEN

The mechanisms by which myelodysplastic syndrome (MDS) cells resist the effects of hypomethylating agents (HMA) are currently the subject of intensive research. A better understanding of mechanisms by which the MDS cell becomes to tolerate HMA and progresses to acute myeloid leukemia (AML) requires the development of new cellular models. From MDS/AML cell lines we developed a model of 5-azacytidine (AZA) resistance whose stability was validated by a transplantation approach into immunocompromised mice. When investigating mRNA expression and DNA variants of the AZA resistant phenotype we observed deregulation of several cancer-related pathways including the phosphatidylinosito-3 kinase signaling. We have further shown that these pathways can be modulated by specific inhibitors that, while blocking the proliferation of AZA resistant cells, are unable to increase their sensitivity to AZA. Our data reveal a set of molecular mechanisms that can be targeted to expand therapeutic options during progression on AZA therapy.


Asunto(s)
Azacitidina/farmacología , Resistencia a Antineoplásicos , Modelos Biológicos , Animales , ADN de Neoplasias/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Ratones , Ratones SCID , Anotación de Secuencia Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Transcriptoma/genética
9.
Front Biosci ; 13: 6126-34, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508648

RESUMEN

Chromatin structure and its changes or maintenance throughout developmental checkpoints play indispensable role in organismal homeostasis. Chromatin remodeling factors of the SWI/SNF2 superfamily use ATP hydrolysis to change DNA-protein contacts, and their loss-of-function or inappropriate increase leads to distinct human pathologic states. In this review, we focus on the translational view of human pathologic physiology involving SWI/SNF2 superfamily, combining latest finding from basic and clinical research. We discuss in mechanistic terms the consequences resulting from dose alteration of the SWI/SNF2 superfamily ATPases and emphasize the necessity of future human subject-based studies.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Enfermedades Genéticas Congénitas/metabolismo , Leucemia/metabolismo , Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , ADN/metabolismo , ADN Helicasas/genética , ADN de Neoplasias/metabolismo , Enfermedades Genéticas Congénitas/genética , Histonas/metabolismo , Humanos , Leucemia/genética , Neoplasias/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética
10.
Nat Commun ; 5: 4181, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24946904

RESUMEN

Chromatin compaction mediates progenitor to post-mitotic cell transitions and modulates gene expression programs, yet the mechanisms are poorly defined. Snf2h and Snf2l are ATP-dependent chromatin remodelling proteins that assemble, reposition and space nucleosomes, and are robustly expressed in the brain. Here we show that mice conditionally inactivated for Snf2h in neural progenitors have reduced levels of histone H1 and H2A variants that compromise chromatin fluidity and transcriptional programs within the developing cerebellum. Disorganized chromatin limits Purkinje and granule neuron progenitor expansion, resulting in abnormal post-natal foliation, while deregulated transcriptional programs contribute to altered neural maturation, motor dysfunction and death. However, mice survive to young adulthood, in part from Snf2l compensation that restores Engrailed-1 expression. Similarly, Purkinje-specific Snf2h ablation affects chromatin ultrastructure and dendritic arborization, but alters cognitive skills rather than motor control. Our studies reveal that Snf2h controls chromatin organization and histone H1 dynamics for the establishment of gene expression programs underlying cerebellar morphogenesis and neural maturation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cerebelo/embriología , Ensamble y Desensamble de Cromatina/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Histonas/metabolismo , Morfogénesis/fisiología , Células-Madre Neurales/fisiología , Análisis de Varianza , Animales , Western Blotting , Bromodesoxiuridina , Inmunoprecipitación de Cromatina , Femenino , Fluorescencia , Galactósidos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Indoles , Masculino , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Microscopía Electrónica de Transmisión , Morfogénesis/genética , Células-Madre Neurales/metabolismo , Células de Purkinje/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Prueba de Desempeño de Rotación con Aceleración Constante , Cloruro de Tolonio
11.
Mol Cancer Res ; 7(10): 1693-703, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19825991

RESUMEN

Hematopoietic transcription factors GATA-1 and PU.1 bind each other on DNA to block transcriptional programs of undesired lineage during hematopoietic commitment. Murine erythroleukemia (MEL) cells that coexpress GATA-1 and PU.1 are blocked at the blast stage but respond to molecular removal (downregulation) of PU.1 or addition (upregulation) of GATA-1 by inducing terminal erythroid differentiation. To test whether GATA-1 blocks PU.1 in MEL cells, we have conditionally activated a transgenic PU.1 protein fused with the estrogen receptor ligand-binding domain (PUER), resulting in activation of a myeloid transcriptional program. Gene expression arrays identified components of the PU.1-dependent transcriptome negatively regulated by GATA-1 in MEL cells, including CCAAT/enhancer binding protein alpha (Cebpa) and core-binding factor, beta subunit (Cbfb), which encode two key hematopoietic transcription factors. Inhibition of GATA-1 by small interfering RNA resulted in derepression of PU.1 target genes. Chromatin immunoprecipitation and reporter assays identified PU.1 motif sequences near Cebpa and Cbfb that are co-occupied by PU.1 and GATA-1 in the leukemic blasts. Significant derepression of Cebpa and Cbfb is achieved in MEL cells by either activation of PU.1 or knockdown of GATA-1. Furthermore, transcriptional regulation of these loci by manipulating the levels of PU.1 and GATA-1 involves quantitative increases in a transcriptionally active chromatin mark: acetylation of histone H3K9. Collectively, we show that either activation of PU.1 or inhibition of GATA-1 efficiently reverses the transcriptional block imposed by GATA-1 and leads to the activation of a myeloid transcriptional program directed by PU.1.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Transformación Celular Neoplásica/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Factor de Transcripción GATA1/genética , Leucemia/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Transformación Celular Neoplásica/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Factor de Transcripción GATA1/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/fisiopatología , Células Mieloides/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Elementos Reguladores de la Transcripción/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Activación Transcripcional/genética
12.
Biometals ; 19(5): 453-60, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16937251

RESUMEN

We have found and sequenced a significant part of the previously described tellurite resistance determinant on mini-Mu derivative pPR46, named pNT3B, originally cloned from a large conjugative plasmid pTE53, found in Escherichia coli. This plasmid contains genes essential for tellurite resistance, together with the protective region bearing genes terX, Y, W, and the conserved spacing region bearing several ORFs of unknown function. Computer analysis of obtained sequence revealed a close similarity to the formerly described ter operons found on the Serratia marcescens plasmid R478 and the chromosome of Escherichia coli O157:H7. This finding confirms the presence of a whole region on the large conjugative plasmid that pTE53 originated from a uropathogenic E. coli strain, and suggests its possible role in horizontal gene transfer, resulting in the development of new pathogenic E. coli strains.


Asunto(s)
Farmacorresistencia Bacteriana/fisiología , Escherichia coli O157 , Plásmidos/genética , Telurio/metabolismo , Biología Computacional , Medios de Cultivo/química , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Transferencia de Gen Horizontal , Humanos , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Operón
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA