Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 106(2): 464-473, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32467144

RESUMEN

Haploinsufficiency for transcription factor KLF1 causes a variety of human erythroid phenotypes, such as the In(Lu) blood type, increased HbA2 levels, and hereditary persistence of fetal hemoglobin. Severe dominant congenital dyserythropoietic anemia IV (OMIM 613673) is associated with the KLF1 p.E325K variant. CDA-IV patients display ineffective erythropoiesis and hemolysis resulting in anemia, accompanied by persistent high levels of embryonic and fetal hemoglobin. The mouse Nan strain carries a variant in the orthologous residue, KLF1 p.E339D. Klf1Nan causes dominant hemolytic anemia with many similarities to CDA-IV. Here we investigated the impact of Klf1Nan on the developmental expression patterns of the endogenous beta-like and alpha-like globins, and the human beta-like globins carried on a HBB locus transgene. We observe that the switch from primitive, yolk sac-derived, erythropoiesis to definitive, fetal liver-derived, erythropoiesis is delayed in Klf1wt/Nan embryos. This is reflected in globin expression patterns measured between E12.5 and E14.5. Cultured Klf1wt/Nan E12.5 fetal liver cells display growth- and differentiation defects. These defects likely contribute to the delayed appearance of definitive erythrocytes in the circulation of Klf1wt/Nan embryos. After E14.5, expression of the embryonic/fetal globin genes is silenced rapidly. In adult Klf1wt/Nan animals, silencing of the embryonic/fetal globin genes is impeded, but only minute amounts are expressed. Thus, in contrast to human KLF1 p.E325K, mouse KLF1 p.E339D does not lead to persistent high levels of embryonic/fetal globins. Our results support the notion that KLF1 affects gene expression in a variant-specific manner, highlighting the necessity to characterize KLF1 variant-specific phenotypes of patients in detail.


Asunto(s)
Anemia Diseritropoyética Congénita , Factores de Transcripción de Tipo Kruppel , Adulto , Animales , Diferenciación Celular , Eritropoyesis/genética , Hemoglobinas , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones
2.
EMBO J ; 32(3): 473-86, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23299939

RESUMEN

The TREX complex couples nuclear pre-mRNA processing with mRNA export and contains multiple protein components, including Uap56, Alyref, Cip29 and the multi-subunit THO complex. Here, we have identified Chtop as a novel TREX component. We show that both Chtop and Alyref activate the ATPase and RNA helicase activities of Uap56 and that Uap56 functions to recruit both Alyref and Chtop onto mRNA. As observed with the THO complex subunit Thoc5, Chtop binds to the NTF2-like domain of Nxf1, and this interaction requires arginine methylation of Chtop. Using RNAi, we show that co-knockdown of Alyref and Chtop results in a potent mRNA export block. Chtop binds to Uap56 in a mutually exclusive manner with Alyref, and Chtop binds to Nxf1 in a mutually exclusive manner with Thoc5. However, Chtop, Thoc5 and Nxf1 exist in a single complex in vivo. Together, our data indicate that TREX and Nxf1 undergo dynamic remodelling, driven by the ATPase cycle of Uap56 and post-translational modifications of Chtop.


Asunto(s)
Exodesoxirribonucleasas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , ARN Helicasas DEAD-box/metabolismo , Metilación de ADN , Cartilla de ADN/genética , Prueba de Complementación Genética , Humanos , Hibridación Fluorescente in Situ , Complejos Multiproteicos/genética , Proteínas Nucleares/genética , Interferencia de ARN , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/genética
3.
Mol Cell Proteomics ; 11(11): 1263-73, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22872859

RESUMEN

Chromatin target of Prmt1 (Chtop) is a vertebrate-specific chromatin-bound protein that plays an important role in transcriptional regulation. As its mechanism of action remains unclear, we identified Chtop-interacting proteins using a biotinylation-proteomics approach. Here we describe the identification and initial characterization of Five Friends of Methylated Chtop (5FMC). 5FMC is a nuclear complex that can only be recruited by Chtop when the latter is arginine-methylated by Prmt1. It consists of the co-activator Pelp1, the Sumo-specific protease Senp3, Wdr18, Tex10, and Las1L. Pelp1 functions as the core of 5FMC, as the other components become unstable in the absence of Pelp1. We show that recruitment of 5FMC to Zbp-89, a zinc-finger transcription factor, affects its sumoylation status and transactivation potential. Collectively, our data provide a mechanistic link between arginine methylation and (de)sumoylation in the control of transcriptional activity.


Asunto(s)
Arginina/metabolismo , Cromatina/metabolismo , Proteínas Co-Represoras/metabolismo , Cisteína Endopeptidasas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Animales , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Humanos , Metilación , Ratones , Modelos Biológicos , Péptido Hidrolasas/metabolismo , Unión Proteica , Estabilidad Proteica
4.
Sci Rep ; 12(1): 336, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013432

RESUMEN

Haploinsufficiency for the erythroid-specific transcription factor KLF1 is associated with hereditary persistence of fetal hemoglobin (HPFH). Increased HbF ameliorates the symptoms of ß-hemoglobinopathies and downregulation of KLF1 activity has been proposed as a potential therapeutic strategy. However, the feasibility of this approach has been challenged by the observation that KLF1 haploinsufficient individuals with the same KLF1 variant, within the same family, display a wide range of HbF levels. This phenotypic variability is not readily explained by co-inheritance of known HbF-modulating variants in the HBB, HBS1L-MYB and/or BCL11A loci. We studied cultured erythroid progenitors obtained from Maltese individuals in which KLF1 p.K288X carriers display HbF levels ranging between 1.3 and 12.3% of total Hb. Using a combination of gene expression analysis, chromatin accessibility assays and promoter activity tests we find that variation in expression of the wildtype KLF1 allele may explain a significant part of the variability in HbF levels observed in KLF1 haploinsufficiency. Our results have general bearing on the variable penetrance of haploinsufficiency phenotypes and on conflicting interpretations of pathogenicity of variants in other transcriptional regulators such as EP300, GATA2 and RUNX1.


Asunto(s)
Epigénesis Genética , Epigenoma , Epigenómica , Eritroblastos/metabolismo , Haploinsuficiencia , Hemoglobinopatías/genética , Factores de Transcripción de Tipo Kruppel/genética , Células Cultivadas , Secuenciación de Inmunoprecipitación de Cromatina , Eritroblastos/patología , Eritropoyesis/genética , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Predisposición Genética a la Enfermedad , Hemoglobinopatías/sangre , Hemoglobinopatías/diagnóstico , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Malta , Penetrancia , Fenotipo , Cultivo Primario de Células , RNA-Seq
5.
Blood Adv ; 5(9): 2339-2349, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33938942

RESUMEN

The BCL11A gene encodes a transcriptional repressor with essential functions in multiple tissues during human development. Haploinsufficiency for BCL11A causes Dias-Logan syndrome (OMIM 617101), an intellectual developmental disorder with hereditary persistence of fetal hemoglobin (HPFH). Due to the severe phenotype, disease-causing variants in BCL11A occur de novo. We describe a patient with a de novo heterozygous variant, c.1453G>T, in the BCL11A gene, resulting in truncation of the BCL11A-XL protein (p.Glu485X). The truncated protein lacks the 3 C-terminal DNA-binding zinc fingers and the nuclear localization signal, rendering it inactive. The patient displayed high fetal hemoglobin (HbF) levels (12.1-18.7% of total hemoglobin), in contrast to the parents who had HbF levels of 0.3%. We used cultures of patient-derived erythroid progenitors to determine changes in gene expression and chromatin accessibility. In addition, we investigated DNA methylation of the promoters of the γ-globin genes HBG1 and HBG2. HUDEP1 and HUDEP2 cells were used as models for fetal and adult human erythropoiesis, respectively. Similar to HUDEP1 cells, the patient's cells displayed Assay for Transposase-Accessible Chromatin (ATAC) peaks at the HBG1/2 promoters and significant expression of HBG1/2 genes. In contrast, HBG1/2 promoter methylation and genome-wide gene expression profiling were consistent with normal adult erythropoiesis. We conclude that HPFH is the major erythroid phenotype of constitutive BCL11A haploinsufficiency. Given the essential functions of BCL11A in other hematopoietic lineages and the neuronal system, erythroid-specific targeting of the BCL11A gene has been proposed for reactivation of γ-globin expression in ß-hemoglobinopathy patients. Our data strongly support this approach.


Asunto(s)
Haploinsuficiencia , Proteínas Nucleares , Adulto , Proteínas Portadoras/genética , Humanos , Proteínas Nucleares/genética , Fenotipo , Proteínas Represoras/genética
6.
Mol Cell Biol ; 27(10): 3839-3854, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17353275

RESUMEN

The cooperation of stem cell factor (SCF) and erythropoietin (Epo) is required to induce renewal divisions in erythroid progenitors, whereas differentiation to mature erythrocytes requires the presence of Epo only. Epo and SCF activate common signaling pathways such as the activation of protein kinase B (PKB) and the subsequent phosphorylation and inactivation of Foxo3a. In contrast, only Epo activates Stat5. Both Foxo3a and Stat5 promote erythroid differentiation. To understand the interplay of SCF and Epo in maintaining the balance between renewal and differentiation during erythroid development, we investigated differential Foxo3a target regulation by Epo and SCF. Expression profiling revealed that a subset of Foxo3a targets was not inhibited but was activated by Epo. One of these genes was Cited2. Transcriptional control of Epo/Foxo3a-induced Cited2 was studied and compared with that of the Epo-repressed Foxo3a target Btg1. We show that in response to Epo, the allegedly growth-inhibitory factor Foxo3a associates with the allegedly growth-stimulatory factor Stat5 in the nucleus, which is required for Epo-induced Cited2 expression. In contrast, Btg1 expression is controlled by the cooperation of Foxo3a with cyclic AMP- and Jun kinase-dependent Creb family members. Thus, Foxo3a not only is an effector of PKB but also integrates distinct signals to regulate gene expression in erythropoiesis.


Asunto(s)
Eritropoyesis/fisiología , Eritropoyetina/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Factor de Células Madre/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Células Cultivadas , Análisis por Conglomerados , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Alineación de Secuencia , Transducción de Señal/fisiología , Transactivadores/genética , Transactivadores/metabolismo
7.
J Cell Biol ; 164(2): 175-84, 2004 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-14734530

RESUMEN

Erythropoiesis requires tight control of expansion, maturation, and survival of erythroid progenitors. Because activation of phosphatidylinositol-3-kinase (PI3K) is required for erythropoietin/stem cell factor-induced expansion of erythroid progenitors, we examined the role of the PI3K-controlled Forkhead box, class O (FoxO) subfamily of Forkhead transcription factors. FoxO3a expression and nuclear accumulation increased during erythroid differentiation, whereas untimely induction of FoxO3a activity accelerated differentiation of erythroid progenitors to erythrocytes. We identified B cell translocation gene 1 (BTG1)/antiproliferative protein 2 as a FoxO3a target gene in erythroid progenitors. Promoter studies indicated BTG1 as a direct target of FoxO3a. Expression of BTG1 in primary mouse bone marrow cells blocked the outgrowth of erythroid colonies, which required a domain of BTG1 that binds protein arginine methyl transferase 1. During erythroid differentiation, increased arginine methylation coincided with BTG1 expression. Concordantly, inhibition of methyl transferase activity blocked erythroid maturation without affecting expansion of progenitor cells. We propose FoxO3a-controlled expression of BTG1 and subsequent regulation of protein arginine methyl transferase activity as a novel mechanism controlling erythroid expansion and differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Proteínas de Neoplasias/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Factores de Transcripción/genética , Células 3T3 , Animales , Secuencia de Bases , Células COS , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Clonación Molecular , Cartilla de ADN , Activación Enzimática , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead , Genes Reporteros , Células Madre Hematopoyéticas/citología , Humanos , Luciferasas/genética , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Factores de Transcripción/fisiología
8.
PLoS One ; 14(3): e0208659, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30921348

RESUMEN

Krüppel-like factor 1 (KLF1) is an essential transcription factor for erythroid development, as demonstrated by Klf1 knockout mice which die around E14 due to severe anemia. In humans, >140 KLF1 variants, causing different erythroid phenotypes, have been described. The KLF1 Nan variant, a single amino acid substitution (p.E339D) in the DNA binding domain, causes hemolytic anemia and is dominant over wildtype KLF1. Here we describe the effects of the KLF1 Nan variant during fetal development. We show that Nan embryos have defects in erythroid maturation. RNA-sequencing of the KLF1 Nan fetal liver cells revealed that Exportin 7 (Xpo7) was among the 782 deregulated genes. This nuclear exportin is implicated in terminal erythroid differentiation; in particular it is involved in nuclear condensation. Indeed, KLF1 Nan fetal liver cells had larger nuclei and reduced chromatin condensation. Knockdown of XPO7 in wildtype erythroid cells caused a similar phenotype. We propose that reduced expression of XPO7 is partially responsible for the erythroid defects observed in KLF1 Nan erythroid cells.


Asunto(s)
Anemia Hemolítica/genética , Células Eritroides/citología , Factores de Transcripción de Tipo Kruppel/genética , Proteína de Unión al GTP ran/genética , Sustitución de Aminoácidos , Animales , Diferenciación Celular , Células Cultivadas , Cromatina/metabolismo , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Células Eritroides/metabolismo , Eritropoyesis , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Análisis de Secuencia de ARN/métodos , Proteína de Unión al GTP ran/metabolismo
9.
Hum Gene Ther Methods ; 29(1): 60-74, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29325430

RESUMEN

The ß-hemoglobinopathies sickle cell anemia and ß-thalassemia are the focus of many gene-therapy studies. A key disease parameter is the abundance of globin chains because it indicates the level of anemia, likely toxicity of excess or aberrant globins, and therapeutic potential of induced or exogenous ß-like globins. Reversed-phase high-performance liquid chromatography (HPLC) allows versatile and inexpensive globin quantification, but commonly applied protocols suffer from long run times, high sample requirements, or inability to separate murine from human ß-globin chains. The latter point is problematic for in vivo studies with gene-addition vectors in murine disease models and mouse/human chimeras. This study demonstrates HPLC-based measurements of globin expression (1) after differentiation of the commonly applied human umbilical cord blood-derived erythroid progenitor-2 cell line, (2) in erythroid progeny of CD34+ cells for the analysis of clustered regularly interspaced short palindromic repeats/Cas9-mediated disruption of the globin regulator BCL11A, and (3) of transgenic mice holding the human ß-globin locus. At run times of 8 min for separation of murine and human ß-globin chains as well as of human γ-globin chains, and with routine measurement of globin-chain ratios for 12 nL of blood (tested for down to 0.75 nL) or of 300,000 in vitro differentiated cells, the methods presented here and any variant-specific adaptations thereof will greatly facilitate evaluation of novel therapy applications for ß-hemoglobinopathies.


Asunto(s)
Anemia de Células Falciformes , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Terapia Genética/métodos , Vectores Genéticos , Globinas beta , gamma-Globinas , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Anemia de Células Falciformes/patología , Anemia de Células Falciformes/terapia , Animales , Línea Celular , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Ratones , Ratones Transgénicos , Globinas beta/biosíntesis , Globinas beta/genética , gamma-Globinas/genética
10.
J Clin Endocrinol Metab ; 102(9): 3517-3525, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911146

RESUMEN

Context: Patients with resistance to thyroid hormone (TH) α (RTHα) are characterized by growth retardation, macrocephaly, constipation, and abnormal thyroid function tests. In addition, almost all RTHα patients have mild anemia, the pathogenesis of which is unknown. Animal studies suggest an important role for TH and TH receptor (TR)α in erythropoiesis. Objective: To investigate whether a defect in TRα affects the maturation of red blood cells in RTHα patients. Design, Setting, and Patients: Cultures of primary human erythroid progenitor cells (HEPs), from peripheral blood of RTHα patients (n = 11) harboring different inactivating mutations in TRα (P398R, F397fs406X, C392X, R384H, A382fs388X, A263V, A263S), were compared with healthy controls (n = 11). During differentiation, erythroid cells become smaller, accumulate hemoglobin, and express different cell surface markers. We assessed cell number and cell size, and used cell staining and fluorescence-activated cell sorter analysis to monitor maturation at different time points. Results: After ∼14 days of ex vivo expansion, both control and patient-derived progenitors differentiated spontaneously. However, RTHα-derived cells differentiated more slowly. During spontaneous differentiation, RTHα-derived HEPs were larger, more positive for c-Kit (a proliferation marker), and less positive for glycophorin A (a differentiation marker). The degree of abnormal spontaneous maturation of RTHα-derived progenitors did not correlate with severity of underlying TRα defect. Both control and RTHα-derived progenitors responded similarly when differentiation was induced. T3 exposure accelerated differentiation of both control- and RTHα patient-derived HEPs. Conclusions: Inactivating mutations in human TRα affect the balance between proliferation and differentiation of progenitor cells during erythropoiesis, which may contribute to the mild anemia seen in most RTHα patients.


Asunto(s)
Anemia/genética , Eritropoyesis/genética , Regulación de la Expresión Génica , Receptores alfa de Hormona Tiroidea/genética , Síndrome de Resistencia a Hormonas Tiroideas/genética , Adolescente , Adulto , Anemia/epidemiología , Anemia/fisiopatología , Estudios de Casos y Controles , Células Cultivadas , Niño , Preescolar , Eritrocitos/metabolismo , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , Valores de Referencia , Rol , Células Madre/citología , Células Madre/fisiología , Síndrome de Resistencia a Hormonas Tiroideas/epidemiología , Síndrome de Resistencia a Hormonas Tiroideas/fisiopatología , Adulto Joven
11.
Am J Blood Res ; 4(1): 27-32, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25232502

RESUMEN

Currently, bone marrow transplantation is the only curative treatment for ß-thalassemia and sickle cell disease. In rare cases, sustained and full fetal hemoglobin production was observed in patients after failure of bone marrow transplantation. This rendered the patients transfusion-free, despite genetic disease and transplant rejection. The mechanisms underlying this phenomenon remain unexplored. We have studied a trio (father-mother-child) in which the affected child became transfusion-independent after rejection of an allogeneic bone marrow graft. Remarkably, we found that his non-thalassemic mother also expressed unusually high levels of γ-globin. High HbF in one of the parents may therefore be of prognostic value in these rare cases. Genotyping of the HBB locus and the HbF quantitative trait loci HBS1L-MYB, KLF1 and BCL11A, and protein expression analysis of KLF1 and BCL11A, failed to explain the increased HbF levels, indicating that an as yet unidentified HbF modifier locus may be involved. We hypothesize that epigenetic events brought about by the transplantation procedure allow therapeutic levels of HbF expression in the child. Potential implications of our observations for reactivation of γ-globin expression and interpretation of the French globin gene therapy case are discussed.

12.
Endocrinology ; 150(11): 5163-70, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19797118

RESUMEN

The human monocarboxylate transporter 8 (hMCT8) protein mediates transport of thyroid hormone across the plasma membrane. Association of hMCT8 mutations with severe psychomotor retardation and disturbed thyroid hormone levels has established its physiological relevance, but little is still known about the basic properties of hMCT8. In this study we present evidence that hMCT8 does not form heterodimers with the ancillary proteins basigin, embigin, or neuroplastin, unlike other MCTs. In contrast, it is suggested that MCT8 exists as monomer and homodimer in transiently and stably transfected cells. Apparently hMCT8 forms stable dimers because the complex is resistant to denaturing conditions and dithiothreitol. Cotransfection of wild-type hMCT8 with a mutant lacking amino acids 267-360 resulted in formation of homo-and heterodimers of the variants, indicating that transmembrane domains 4-6 are not involved in the dimerization process. Furthermore, we explored the structural and functional role of the 10 Cys residues in hMCT8. All possible Cys>Ala mutants did not behave differently from wild-type hMCT8 in protein expression, cross-linking experiments with HgCl(2) and transport function. Our findings indicate that individual Cys residues are not important for the function of hMCT8 or suggest that hMCT8 has other yet-undiscovered functions in which cysteines play an essential role.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos/química , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Humanos , Ratones , Datos de Secuencia Molecular , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Mutación , Multimerización de Proteína , Estructura Terciaria de Proteína , Simportadores , Hormonas Tiroideas/metabolismo
13.
Blood ; 110(6): 1933-41, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17505015

RESUMEN

Dendritic cells are key initiators and regulators of the immune response. Dendritic cell commitment and function require orchestrated regulation of transcription. Gata1 is a transcription factor expressed in several hematopoietic lineages. However, Gata1 function has not been explored in the monocytic or dendritic cell compartment. Here, we show that Gata1 is expressed in myeloid and plasmacytoid dendritic cells and that Gata1 ablation affects the survival of dendritic cells. Furthermore, lipopolysaccharide (LPS) stimulation of dendritic cells prompts Gata1 up-regulation, which is accompanied by increased levels of BclX and Ifng. Our findings show that Gata1 is a transcriptional regulator of dendritic cell differentiation and suggest that Gata1 is involved in the dendritic cell and macrophage lineage separation.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Supervivencia Celular , Células Dendríticas/citología , Factor de Transcripción GATA1/fisiología , Animales , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factor de Transcripción GATA1/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/metabolismo , Interferón gamma/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Células Mieloides/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteína bcl-X/metabolismo
14.
Biol Chem ; 385(5): 409-13, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15196000

RESUMEN

LFM-A13, or alpha-cyano-beta-hydroxy-beta-methyl-N-(2,5-dibromophenyl)propenamide, was shown to inhibit Bruton's tyrosine kinase (Btk). Here we show that LFM-A13 efficiently inhibits erythropoietin (Epo)-induced phosphorylation of the erythropoietin receptor, Janus kinase 2 (Jak2) and downstream signalling molecules. However, the tyrosine kinase activity of immunoprecipitated or in vitro translated Btk and Jak2 was equally inhibited by LFM-A13 in in vitro kinase assays. Finally, Epo-induced signal transduction was also inhibited in cells lacking Btk. Taken together, we conclude that LFM-A13 is a potent inhibitor of Jak2 and cannot be used as a specific tyrosine kinase inhibitor to study the role of Btk in Jak2-dependent cytokine signalling.


Asunto(s)
Amidas/farmacología , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Células COS , Janus Quinasa 2 , Fosforilación , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA