Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 24(22): 13472-13480, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33058427

RESUMEN

TG-Interacting Factor 1 (Tgif1) affects proliferation and differentiation of myeloid cells and regulates self-renewal of haematopoietic stem cells (HSCs). To determine its impact on leukaemic haematopoiesis, we induced acute or chronic myeloid leukaemias (AML or CML) in mice by enforced expression of MLL-AF9 or BCR-ABL, respectively, in Tgif1+/+ or Tgif1-/- haematopoietic stem and progenitor cells (HSPCs) and transplanted them into syngeneic recipients. We find that loss of Tgif1 accelerates leukaemic progression and shortens survival in mice with either AML or CML. Leukaemia-initiating cells (LICs) occur with higher frequency in AML among mice transplanted with MLL-AF9-transduced Tgif1-/- HSPCs than with Tgif1+/+ BMCs. Moreover, AML in mice generated with Tgif1-/- HSPCs are chemotherapy resistant and relapse more rapidly than those whose AML arose in Tgif1+/+ HSPCs. Whole transcriptome analysis shows significant alterations in gene expression profiles associated with transforming growth factor-beta (TGF-beta) and retinoic acid (RA) signalling pathways because of Tgif1 loss. These findings indicate that Tgif1 has a protective role in myeloid leukaemia initiation and progression, and its anti-leukaemic contributions are connected to TGF-beta- and RA-driven functions.


Asunto(s)
Biomarcadores de Tumor , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/mortalidad , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Proteínas Represoras/deficiencia , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas de Homeodominio , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/diagnóstico , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Ratones Noqueados , Pronóstico , Recurrencia , Eliminación de Secuencia
2.
J Immunol ; 193(9): 4654-62, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25238756

RESUMEN

Transcriptional mechanisms governing hematopoietic stem cell (HSC) quiescence, self-renewal, and differentiation are not fully understood. Sequence-specific ssDNA-binding protein 2 (SSBP2) is a candidate acute myelogenous leukemia (AML) suppressor gene located at chromosome 5q14. SSBP2 binds the transcriptional adaptor protein Lim domain-binding protein 1 (LDB1) and enhances LDB1 stability to regulate gene expression. Notably, Ldb1 is essential for HSC specification during early development and maintenance in adults. We previously reported shortened lifespan and greater susceptibility to B cell lymphomas and carcinomas in Ssbp2(-/-) mice. However, whether Ssbp2 plays a regulatory role in normal HSC function and leukemogenesis is unknown. In this study, we provide several lines of evidence to demonstrate a requirement for Ssbp2 in the function and transcriptional program of hematopoietic stem and progenitor cells (HSPCs) in vivo. We found that hematopoietic tissues were hypoplastic in Ssbp2(-/-) mice, and the frequency of lymphoid-primed multipotent progenitor cells in bone marrow was reduced. Other significant features of these mice were delayed recovery from 5-fluorouracil treatment and diminished multilineage reconstitution in lethally irradiated bone marrow recipients. Dramatic reduction of Notch1 transcripts and increased expression of transcripts encoding the transcription factor E2a and its downstream target Cdkn1a also distinguished Ssbp2(-/-) HSPCs from wild-type HSPCs. Finally, a tendency toward coordinated expression of SSBP2 and the AML suppressor NOTCH1 in a subset of the Cancer Genome Atlas AML cases suggested a role for SSBP2 in AML pathogenesis. Collectively, our results uncovered a critical regulatory function for SSBP2 in HSPC gene expression and function.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Estrés Fisiológico , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Médula Ósea/metabolismo , Médula Ósea/patología , Trasplante de Médula Ósea , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Expresión Génica , Hematopoyesis/genética , Homeostasis/genética , Inmunofenotipificación , Ratones , Ratones Noqueados , Fenotipo , Receptor Notch1/genética , Receptor Notch1/metabolismo
3.
Biol Blood Marrow Transplant ; 20(7): 1014-20, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24704384

RESUMEN

Multiple myeloma is the most frequent indication for high-dose melphalan (HDM) chemotherapy with autologous stem cell transplantation (ASCT). Gastrointestinal symptoms represent the most significant nonhematological toxicity of HDM. However, specific, especially genetic, predictors of their incidence or clinical severity are lacking. The amino acid transporters LAT1 and LAT2 encoded by the SLC7A5 and SLC7A8 genes, respectively, are the principal mediators of melphalan uptake into cells. To determine whether genetic variability at these loci contributed to interindividual differences in the development of gastrointestinal complications of HDM, we analyzed single nucleotide polymorphisms (SNPs) in these genes in 135 patients with multiple myeloma treated with HDM and ASCT and correlated these with the need for total parenteral nutrition (TPN). Seven SNPs in SLC7A5 and 20 in SLC7A8 were genotyped. Multiple analyses indicated that 1 SNP in the first intron of SLC7A5, rs4240803, was significantly associated with TPN use (odds ratio = .45, 95% confidence interval, .25 to .79; P = .007). Further, every haplotype that correlated with TPN requirement included this SNP. These results suggest that variability in melphalan transport affects mucosal injury after HDM. This finding could help in individualizing the dose of this effective and widely used chemotherapeutic agent for multiple myeloma.


Asunto(s)
Antineoplásicos/efectos adversos , Enfermedades Gastrointestinales/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Transportador de Aminoácidos Neutros Grandes 1/genética , Melfalán/efectos adversos , Mieloma Múltiple/terapia , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Estudios de Casos y Controles , Terapia Combinada , Femenino , Enfermedades Gastrointestinales/genética , Predisposición Genética a la Enfermedad , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Masculino , Melfalán/administración & dosificación , Melfalán/farmacocinética , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Polimorfismo de Nucleótido Simple , Trasplante Autólogo
4.
Neurotherapeutics ; 18(2): 827-844, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33844154

RESUMEN

Schizophrenia is a complex condition associated with perceptual disturbances, decreased motivation and affect, and disrupted cognition. Individuals living with schizophrenia may experience myriad poor outcomes, including impairment in independent living and function as well as decreased life expectancy. Though existing treatments may offer benefit, many individuals still experience treatment resistant and disabling symptoms. In light of the negative outcomes associated with schizophrenia and the limitations in currently available treatments, there is a significant need for novel therapeutic interventions. Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation technique that can modulate the activity of discrete cortical regions, allowing direct manipulation of local brain activation and indirect manipulation of the target's associated neural networks. rTMS has been studied in schizophrenia for the treatment of auditory hallucinations, negative symptoms, and cognitive deficits, with mixed results. The field's inability to arrive at a consensus on the use rTMS in schizophrenia has stemmed from a variety of issues, perhaps most notably the significant heterogeneity amongst existing trials. In addition, it is likely that factors specific to schizophrenia, rather than the rTMS itself, have presented barriers to the interpretation of existing results. However, advances in approaches to rTMS as a biologic probe and therapeutic, many of which include the integration of neuroimaging with rTMS, offer hope that this technology may still play a role in improving the understanding and treatment of schizophrenia.


Asunto(s)
Encéfalo/diagnóstico por imagen , Neuroimagen/tendencias , Esquizofrenia/diagnóstico por imagen , Esquizofrenia/terapia , Estimulación Magnética Transcraneal/tendencias , Encéfalo/fisiopatología , Predicción , Humanos , Neuroimagen/métodos , Neuronavegación/métodos , Neuronavegación/tendencias , Esquizofrenia/fisiopatología , Estimulación Magnética Transcraneal/métodos , Resultado del Tratamiento
5.
Biochim Biophys Acta ; 1779(5): 347-55, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18455519

RESUMEN

TG-interacting factor (TGIF) is a homeobox transcriptional repressor that has been implicated in holoprosencephaly and various types of cancer, including leukemias. In this study, we provide the first detailed description of the TGIF locus characterizing 12 TGIF splice isoforms. These isoforms have similar open reading frames but different 5' untranslated regions. TGIF expression data are presented from multiple tissues, cell lines and primary leukemia cells. Isoform-specific real-time PCR analysis showed that even though these isoforms were broadly expressed all except isoform 4, had very low level of expression. In fact, isoform 4 was the predominant TGIF isoform expressed in all tissues analyzed. Since TGIF, levels have recently implicated to play a role in acute myelogenous leukemia we proceeded to characterize the minimal promoter region of isoform 4 as a first step in understanding mechanisms of TGIF expression. As expected for homeobox genes, the minimal promoter region for isoform 4 has multiple Sp1 binding sites and a CpG island raising the possibility that the low TGIF expression seen in some AML patients and leukemia cell lines may be secondary to methylation. Further characterization of expression from this promoter using 5-Aza-2'-deoxycytidine treatment and transient expression assays showed that decreased TGIF expression is likely secondary to active repression and not because of promoter methylation. A detailed characterization of this complex locus is important as it may help to clarify the functions of this gene in brain development and leukemia biology.


Asunto(s)
Empalme Alternativo , Proteínas de Homeodominio/genética , Leucemia Mieloide Aguda/genética , Proteínas Represoras/genética , Región de Flanqueo 5' , Línea Celular , Línea Celular Tumoral , Clonación Molecular , Genómica , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Represoras/metabolismo
6.
Biochem Biophys Res Commun ; 390(2): 295-301, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19799863

RESUMEN

The TAL1 (or SCL) gene, originally discovered through its involvement by a chromosomal translocation in T-cell acute lymphoblastic leukemia, encodes a basic helix-loop-helix (bHLH) transcription factor essential for hematopoietic and vascular development. To identify its interaction partners, we expressed a tandem epitope-tagged protein in murine erythroleukemia (MEL) cells and characterized affinity-purified Tal1-containing complexes by liquid chromatography-tandem mass spectrometry analysis. In addition to known interacting proteins, two proteins related to the Eight-Twenty-One (ETO) corepressor, Eto2/Mtg16 and Mtgr1, were identified from the peptide fragments analyzed. Tal1 interaction with Eto2 and Mtgr1 was verified by coimmunoprecipitation analysis in Tal1, Eto2-, and Mtgr1-transfected COS-7 cells, MEL cells expressing V5 epitope-tagged Tal1 protein, and non-transfected MEL cells. Mapping analysis with Gal4 fusion proteins demonstrated a requirement for the bHLH domain of Tal1 and TAF110 domain of Eto2 for their interaction, and transient transfection and glutathione S-transferase pull-down analysis showed that Mtgr1 and Eto2 enhanced the other's association with Tal1. Enforced expression of Eto2 in differentiating MEL cells inhibited the promoter of the Protein 4.2 (P4.2) gene, a direct target of TAL1 in erythroid progenitors, and transduction of Eto2 and Mtgr1 augmented Tal1-mediated gene repression. Finally, chromatin immunoprecipitation analysis revealed that Eto2 occupancy of the P4.2 promoter in MEL cells decreased with differentiation, in parallel with a decline in Eto2 protein abundance. These results identify Eto2 and Mtgr1 as authentic interaction partners of Tal1 and suggest they act as heteromeric corepressors of this bHLH transcription factor during erythroid differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Precursoras Eritroides/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Ratones , Proteína 1 de la Leucemia Linfocítica T Aguda
7.
Mol Cell Biol ; 26(7): 2615-25, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16537906

RESUMEN

Blood and vascular cells are generated during early embryogenesis from a common precursor, the hemangioblast. The stem cell leukemia gene (SCL/tal 1) encodes a basic helix-loop-helix transcription factor that is essential for the normal development of blood progenitors and blood vessels. We have previously characterized a panel of SCL enhancers including the +19 element, which directs expression to hematopoietic stem cells and endothelium. Here we demonstrate that SCL is expressed in bone primordia during embryonic development and in adult osteoblasts. Despite consistent expression in cells of the osteogenic lineage, SCL protein is not required for bone specification of embryonic stem cells. In transgenic mice, the SCL +19 core enhancer directed reporter gene expression to vascular smooth muscle and bone in addition to blood and endothelium. A 644-bp fragment containing the SCL +19 core enhancer was active in both blood and bone cell lines and was bound in vivo by a common array of Ets and GATA transcription factors. Taken together with the recent observation that a common progenitor can give rise to blood and bone cells, our results suggest that the SCL +19 enhancer targets a mesodermal progenitor capable of generating hematopoietic, vascular, and osteoblastic progeny.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sangre/metabolismo , Huesos/citología , Huesos/metabolismo , Elementos de Facilitación Genéticos/genética , Proteínas Proto-Oncogénicas/genética , Transcripción Genética/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Factor de Transcripción GATA2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Ratones , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Proteínas Nucleares , Osteogénesis , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Esqueleto , Células Madre/citología , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/metabolismo
8.
Clin Cancer Res ; 14(21): 7095-101, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18981008

RESUMEN

PURPOSE: Recruitment of histone deacetylases (HDAC) is a mechanism of transcriptional repression implicated in the differentiation block in acute myeloid leukemia (AML). We hypothesized that the HDAC inhibitor romidepsin could cause transcriptional derepression, up-regulation of specific target genes in AML, and differentiation of the leukemic clone. The primary objectives of the study were to evaluate the safety and efficacy of romidepsin in advanced AML. EXPERIMENTAL DESIGN: Twenty patients were stratified into cohort A or B based on the absence or presence of chromosomal abnormalities known to recruit HDACs, including those involving core binding factor (CBF). Romidepsin was administered i.v. at 13 mg/m(2)/d on days 1, 8, and 15 of a 28-day cycle. Pharmacodynamic endpoints were evaluated at serial time points. RESULTS: Common adverse effects noted were grade 1 to 2 nausea, anorexia, and fatigue. No objective evidence of antileukemic activity was seen in cohort A. In cohort B, although there were no clinical responses by standard criteria, antileukemic activity was observed in 5 of 7 patients. Two patients had clearance of bone marrow blasts and 3 patients had a >50% decrease in bone marrow blasts. Furthermore, in cohort B, at 24 h, there was a significant increase in MDR1 (P=0.005), p15 (P=0.01), and p14 (P<0.0001) expression. In cohort A, although there was a trend toward up-regulation of MDR1, p15, and p14 expression, these changes were not statistically significant. CONCLUSION: Romidepsin has differential antileukemic and molecular activity in CBF AML. Development of this agent in CBF AML should focus on combinations that target related mechanisms of gene silencing such as DNA methylation.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Factores de Unión al Sitio Principal/metabolismo , Depsipéptidos/uso terapéutico , Inhibidores de Histona Desacetilasas , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Aberraciones Cromosómicas , Estudios de Cohortes , Depsipéptidos/efectos adversos , Evaluación de Medicamentos , Inhibidores Enzimáticos/uso terapéutico , Femenino , Histona Desacetilasas/efectos adversos , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Persona de Mediana Edad
9.
Biochem Biophys Res Commun ; 373(2): 303-8, 2008 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-18565323

RESUMEN

A family of single-stranded DNA-binding proteins (or SSBPs) has been shown to augment the function of LIM-homeodomain (LIM-HD) transcription factors in embryogenesis by interaction with LIM domain-binding protein-1 (LDB1). No DNA-binding complex has been described, however, containing a LIM-HD protein, LDB1, and SSBP, and the mechanism by which SSBPs affect LIM-HD function had not been elucidated. Through use of electrophoretic mobility shift, antibody supershift, and ChIP analyses, we show that an Lhx2-Ldb1-Ssbp3 complex binds a specific element in the Lhx2 target gene Cga (encoding the alpha subunit of glycoprotein hormones) in the alphaT3-1 pituitary cell line. Using overexpression and knockdown approaches, we demonstrate that SSBP3 inhibits Lhx2 and Ldb1 turnover, stimulates assembly of this DNA-binding complex, promotes its recruitment to the Cga promoter, and enhances Cga transcription. These studies provide novel insights into the regulation of pituitary gene expression and LIM-HD function more generally.


Asunto(s)
ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Hipófisis/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica , Hormonas Glicoproteicas de Subunidad alfa/genética , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas con Dominio LIM , Proteínas con Homeodominio LIM , Ratones , Hipófisis/citología , Regiones Promotoras Genéticas , Factores de Transcripción/antagonistas & inhibidores
10.
FEBS J ; 274(19): 5137-46, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17803681

RESUMEN

A major contributing factor to the development of breast cancer is decreased functional expression of breast cancer susceptibility gene 1, BRCA1. Another key contributor to tumorigenesis is hypoxia. Here we show that hypoxia increased the nuclear localization of BRCA1 in MCF-7 and MDA-MB-468 human breast cancer cell lines without changing its steady-state expression level. Nuclear accumulation of BRCA1 was not evident in MCF-12A or HMEC (human mammary epithelial cell) nonmalignant mammary epithelial cells under the same conditions. Hypoxia also increased the cell surface expression of TRAIL on MDA-MB-468 cells. Neutralization of TRAIL precluded the hypoxia-induced accumulation of BRCA1 in the nucleus, whereas exogenously administered TRAIL mimicked the effect. Treatment of MDA-MB-468 cells with TRAIL resulted in a dose- and time-dependent increase in apoptosis. Furthermore, TRAIL-induced apoptosis in HCC1937 cells, which harbor a BRCA1 mutation, increased synergistically when wild-type BRCA1 was reconstituted in the cells, and downregulation of BRCA1 expression in MDA-MB-468 cells reduced the apoptotic response to TRAIL. These data provide a novel link between hypoxia, TRAIL and BRCA1, and suggest that this relationship may be especially relevant to the potential use of TRAIL as a chemotherapeutic agent.


Asunto(s)
Apoptosis/fisiología , Proteína BRCA1/metabolismo , Neoplasias de la Mama/patología , Hipoxia de la Célula , Núcleo Celular/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Proteína BRCA1/genética , Línea Celular Tumoral , Humanos , Mutagénesis
11.
Leuk Lymphoma ; 48(9): 1728-35, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17786708

RESUMEN

Between January 1990 and April 2001, 115 patients received high-dose chemotherapy (HDT) followed by autologous stem cell transplantation (ASCT) for relapsed or refractory Hodgkin lymphoma (HL). With a median follow-up of 58 months (range, 1 - 175 months), 5-year progression-free survival (PFS) and overall survival (OS) were 46% and 58%, respectively. Twelve patients with primary refractory disease had a 5-year PFS of 41% and OS of 58%, not significantly different from those of the remaining cohort. Early and overall regimen related mortality were 7% and 16%, respectively. Male gender (P = 0.04) and a time to relapse (TTR) < 12 months (P = 0.03) were associated with decreased OS by univariate analysis. In multivariate analysis, TTR < 12 months remained statistically significant (P = 0.04). We have confirmed that HDT and ASCT result in long-term survival for a proportion of patients with relapsed or refractory HL. All patients, including those with primary refractory disease, benefited from HDT and ASCT.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Enfermedad de Hodgkin/terapia , Adolescente , Adulto , Terapia Combinada , Femenino , Enfermedad de Hodgkin/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Primarias Secundarias/etiología , Pronóstico , Recurrencia , Trasplante Autólogo , Trasplante Homólogo , Resultado del Tratamiento
12.
Mol Cell Biol ; 23(21): 7585-99, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14560005

RESUMEN

The TAL1 (or SCL) gene, originally identified from its involvement by a recurrent chromosomal translocation, encodes a basic helix-loop-helix transcription factor essential for erythropoiesis. Although presumed to regulate transcription, its target genes are largely unknown. We show here that a nuclear complex containing TAL1, its DNA-binding partner E47, zinc finger transcription factor GATA-1, LIM domain protein LMO2, and LIM domain-binding protein Ldb1 transactivates the protein 4.2 (P4.2) gene through two E box GATA elements in its proximal promoter. Binding of this complex to DNA was dependent on the integrity of both E box and GATA sites and was demonstrated to occur on the P4.2 promoter in cells. Maximal transcription in transiently transfected cells required both E box GATA elements and expression of all five components of the complex. This complex was shown, in addition, to be capable of linking in solution double-stranded oligonucleotides corresponding to the two P4.2 E box GATA elements. This DNA-linking activity required Ldb1 and increased with dimethyl sulfoxide-induced differentiation of murine erythroleukemia (MEL) cells. In contrast, enforced expression in MEL cells of dimerization-defective mutant Ldb1, as well as wild-type Ldb1, significantly decreased E box GATA DNA-binding activities, P4.2 promoter activity, and accumulation of P4.2 and beta-globin mRNAs. These studies define a physiologic target for a TAL1- and GATA-1-containing ternary complex and reveal a positive role for Ldb1 in erythroid gene expression and differentiation.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Eritrocitos/metabolismo , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Genes Reguladores , Secuencias Hélice-Asa-Hélice , Proteínas con Dominio LIM , Leucemia Eritroblástica Aguda , Proteínas de la Membrana , Ratones , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/genética , Células Tumorales Cultivadas
13.
Biochem J ; 399(2): 297-304, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16800816

RESUMEN

SWI/SNF complexes are involved in both activation and repression of transcription. While one of two homologous ATPases, Brg1 [Brm (Brahma)-related gene 1] or Brm, is required for their chromatin remodelling function, less is known about how these complexes are recruited to DNA. We recently established that a DNA-binding complex containing TAL1/SCL, E47, GATA-1, LMO2 and Ldb1 stimulates P4.2 (protein 4.2) transcription in erythroid progenitors via two E box-GATA elements in the gene's proximal promoter. We show here that the SWI/SNF protein Brg1 is also associated with this complex and that both the E box and GATA DNA-binding sites in these elements are required for Brg1 recruitment. Further, Brg1 occupancy of the P4.2 promoter decreased with terminal erythroid differentiation in association with increased P4.2 transcription, while enforced expression of Brg1 in murine erythroleukaemia cells reduced P4.2 gene expression. Overexpression of Brg1 was associated with increased occupancy of the P4.2 promoter by the nuclear co-repressor mSin3A and HDAC2 (histone deacetylase 2) and with reduced histone H3 and H4 acetylation. Finally, a specific HDAC inhibitor attenuated Brg1-directed repression of P4.2 promoter activity in transfected cells. These results provide insight into the mechanism by which SWI/SNF proteins are recruited to promoters and suggest that transcription of P4.2, and most likely other genes, is actively repressed until the terminal differentiation of erythroid progenitors.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Células Precursoras Eritroides/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Acetilación , Animales , Diferenciación Celular , ADN/metabolismo , ADN Helicasas , Células Precursoras Eritroides/citología , Histona Desacetilasa 2 , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica , Complejo Correpresor Histona Desacetilasa y Sin3 , Células Tumorales Cultivadas
14.
Exp Hematol ; 51: 47-62, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28410882

RESUMEN

The hormone erythropoietin (Epo) is required for erythropoiesis, yet its molecular mechanism of action remains poorly understood, particularly with respect to chromatin dynamics. To investigate how Epo modulates the erythroid epigenome, we performed epigenetic profiling using an ex vivo murine cell system that undergoes synchronous erythroid maturation in response to Epo stimulation. Our findings define the repertoire of Epo-modulated enhancers, illuminating a new facet of Epo signaling. First, a large number of enhancers rapidly responded to Epo stimulation, revealing a cis-regulatory network of Epo-responsive enhancers. In contrast, most of the other identified enhancers remained in an active acetylated state during Epo signaling, suggesting that most erythroid enhancers are established at an earlier precursor stage. Second, we identified several hundred super-enhancers that were linked to key erythroid genes, such as Tal1, Bcl11a, and Mir144/451. Third, experimental and computational validation revealed that many predicted enhancer regions were occupied by TAL1 and enriched with DNA-binding motifs for GATA1, KLF1, TAL1/E-box, and STAT5. Additionally, many of these cis-regulatory regions were conserved evolutionarily and displayed correlated enhancer:promoter acetylation. Together, these findings define a cis-regulatory enhancer network for Epo signaling during erythropoiesis, and provide the framework for future studies involving the interplay of epigenetics and Epo signaling.


Asunto(s)
Reprogramación Celular/fisiología , Epigénesis Genética/fisiología , Células Eritroides/metabolismo , Eritropoyesis/fisiología , Eritropoyetina/metabolismo , Transducción de Señal/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas de Unión al ADN , Epigenómica , Células Eritroides/citología , Eritropoyetina/genética , Femenino , Factor de Transcripción GATA1/biosíntesis , Factor de Transcripción GATA1/genética , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Endogámicos BALB C , MicroARNs/biosíntesis , MicroARNs/genética , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras , Factor de Transcripción STAT5/biosíntesis , Factor de Transcripción STAT5/genética , Proteína 1 de la Leucemia Linfocítica T Aguda
15.
J Biochem ; 140(1): 105-19, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16815859

RESUMEN

LIM-domain-binding proteins (CLIM/NLI/Ldb) are nuclear cofactors for LIM homeodomain transcription factors (LIM-HDs) and LIM-only proteins (LMOs). The LIM-interaction domain (LID) of Ldb is located in the carboxy-terminal region and encoded by the last exon (exon 10) of Ldb genes. It is known that the mammalian CLIM1/Ldb2 gene has a splice isoform, named CLIM1b, lacking the LID. However, little is known about the nature of CLIM1b or the evolutionary conservation of this type of alternative splicing in amphibians and teleost fish. Here, we demonstrate that splice isoforms lacking the LID are also present in the Ldb1 genes of mammals, chick, and Xenopus, as well as in fish paralog Ldb4. All these splicing variations occur in intron 9 and exon 10. We observed that Ldb4b (splice isoform lacking LID) is localized in the nucleus when expressed in mammalian culture cells, and binds to Ldb4a (splice isoform containing LID) but not directly to LIM proteins. However, Ldb4b binds to LMO4 via Ldb4a when coexpressed in culture cells. We also found that mouse Ldb1b lacks the ability to activate protein 4.2 promoter, which is stimulated by LMO2 and Ldb1. These findings suggest that splice isoforms of Ldb lacking LID are potential regulators of Ldb function.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Isoformas de Proteínas/genética , Proteínas de Xenopus/genética , Proteínas de Pez Cebra/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Regulación de la Expresión Génica , Proteínas con Dominio LIM , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia , Transcripción Genética/efectos de los fármacos
16.
PLoS One ; 11(10): e0164804, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27780223

RESUMEN

Squamous cell carcinoma of the head and neck (HNSCC) accounts for more than 300,000 deaths worldwide per year as a consequence of tumor cell invasion of adjacent structures or metastasis. LIM-only protein 4 (LMO4) and LIM-domain binding protein 1 (LDB1), two directly interacting transcriptional adaptors that have important roles in normal epithelial cell differentiation, have been associated with increased metastasis, decreased differentiation, and shortened survival in carcinoma of the breast. Here, we implicate two LDB1-binding proteins, single-stranded binding protein 2 (SSBP2) and 3 (SSBP3), in controlling LMO4 and LDB1 protein abundance in HNSCC and in regulating specific tumor cell functions in this disease. First, we found that the relative abundance of LMO4, LDB1, and the two SSBPs correlated very significantly in a panel of human HNSCC cell lines. Second, expression of these proteins in tumor primaries and lymph nodes involved by metastasis were concordant in 3 of 3 sets of tissue. Third, using a Matrigel invasion and organotypic reconstruct assay, CRISPR/Cas9-mediated deletion of LDB1 in the VU-SCC-1729 cell line, which is highly invasive of basement membrane and cellular monolayers, reduced tumor cell invasiveness and migration, as well as proliferation on tissue culture plastic. Finally, inactivation of the LDB1 gene in these cells decreased growth and vascularization of xenografted human tumor cells in vivo. These data show that LMO4, LDB1, and SSBP2 and/or SSBP3 regulate metastasis, proliferation, and angiogenesis in HNSCC and provide the first evidence that SSBPs control LMO4 and LDB1 protein abundance in a cancer context.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Células Escamosas/patología , Proteínas de Unión al ADN/metabolismo , Proteínas con Dominio LIM/metabolismo , Neoplasias de la Boca/patología , Factores de Transcripción/metabolismo , Animales , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias de la Boca/metabolismo , Metástasis de la Neoplasia , Trasplante de Neoplasias , Técnicas de Cultivo de Tejidos
17.
Blood ; 112(13): 4786-7, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19064734
18.
Hum Pathol ; 36(5): 494-504, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15948116

RESUMEN

Anaplastic large cell lymphoma (ALCL) is an aggressive large T- or null-cell lymphoma. Most ALCLs arising in children and young adults express a constitutively active receptor tyrosine kinase, anaplastic lymphoma kinase (ALK). Anaplastic large cell lymphomas lacking ALK are clinically heterogeneous and their pathogenesis is unknown. This study is the first complementary DNA (cDNA) microarray analysis using RNA extracted from tumor tissue (7 ALK+ ALCLs and 7 ALK- ALCLs) to identify genes differentially expressed or shared between the ALK+ and ALK- tumors. Unsupervised hierarchical clustering using the top 11 most statistically significant discriminator cDNAs correctly grouped all ALK+ and ALK- tumors. Hierarchical clustering analysis using the 44 cDNAs with the greatest differential expression between ALK+ and ALK- RNAs grouped 6 of 7 ALK+ ALCLs together and 1 ALK+ ALCL with the ALK- group. In general, ALK+ tumors overexpress genes encoding signal transduction molecules (SYK , LYN , CDC37) and underexpress transcription factor genes (including HOXC6 and HOX A3 ) compared with the ALK- group. Cyclin D3 was overexpressed in the ALK+ group and the cell cycle inhibitor p19INK4D was decreased in the ALK- group, suggesting different mechanisms of promoting G 1 /S transition. Both groups had similar proliferation rates. Genes highly expressed in both ALK- and ALK+ ALCLs included kinases (LCK, protein kinase C, vav2, and NKIAMRE) and antiapoptotic molecules, suggesting possible common pathogenetic mechanisms as well.


Asunto(s)
Expresión Génica , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/genética , Proteínas Tirosina Quinasas/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Quinasa de Linfoma Anaplásico , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Niño , Ciclina D3 , Inhibidor p19 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Ciclinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Tirosina Quinasas Receptoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Exp Hematol ; 40(6): 431-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22269114

RESUMEN

Twenty-six patients with recurrent CD20(+) B-cell lymphoid malignancies received fludarabine, cyclophosphamide, and rituximab-based nonablative conditioning followed by either matched related (n = 18) or unrelated (n = 8) donor allogeneic stem cell transplantation (allo-SCT) between March 2008 and May 2011. Median age of patients at transplantation was 59 years (range, 41-64 years). At diagnosis, 20 (77%) had stage IV disease; 23 (88%) received ≥3 regimens, 14 (54%) received ≥4 regimens, and 4 (15%) had earlier autologous-SCT. All patients had either chemosensitive or stable disease and nine (35%) were in complete remission before transplantation. At the time of analysis, 17 patients were alive with an estimated 2-year overall survival and progression-free survival rate of 63% and nonrelapse mortality of 25%. Grade II to IV acute graft-vs-host-disease occurred in 8 (31%) and chronic graft-vs-host-disease in 6 (23%) patients (extensive, n = 3). Causes of death include progressive disease in four, acute graft-vs-host-disease in two (both after receiving donor lymphocyte infusion for mixed chimerism with residual disease), infection in one, and other (e.g., substance abuse, leukoencephalopathy) in two. Six patients required rehospitalization within 100 days of SCT (mean = 10 days; range, 3-18 days). Our data support fludarabine, cyclophosphamide, and rituximab-based nonablative conditioning allo-SCT in CD20(+) B-cell lymphoid malignancies and it is time to compare this regimen with an alternative reduced-intensity conditioning regimen in B-cell malignancies.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfoma de Células B/tratamiento farmacológico , Acondicionamiento Pretrasplante , Adulto , Antígenos CD20/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Femenino , Herpesvirus Humano 4/fisiología , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/virología , Masculino , Persona de Mediana Edad , Readmisión del Paciente , Análisis de Supervivencia , Resultado del Tratamiento , Activación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA