Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Haematologica ; 108(4): 1053-1067, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35861015

RESUMEN

Although human cell cultures stimulated with dexamethasone suggest that the glucocorticoid receptor (GR) activates stress erythropoiesis, the effects of GR activation on erythropoiesis in vivo remain poorly understood. We characterized the phenotype of a large cohort of patients with Cushing disease, a rare condition associated with elevated cortisol levels. Results from hypercortisolemic patients with active Cushing disease were compared with those obtained from eucortisolemic patients after remission and from volunteers without the disease. Patients with active Cushing disease exhibited erythrocytosis associated with normal hemoglobin F levels. In addition, their blood contained elevated numbers of GR-induced CD163+ monocytes and a unique class of CD34+ cells expressing CD110, CD36, CD133 and the GR-target gene CXCR4. When cultured, these CD34+ cells generated similarly large numbers of immature erythroid cells in the presence and absence of dexamethasone, with raised expression of the GR-target gene GILZ. Of interest, blood from patients with Cushing disease in remission maintained high numbers of CD163+ monocytes and, although their CD34+ cells had a normal phenotype, these cells were unresponsive to added dexamethasone. Collectively, these results indicate that chronic exposure to excess glucocorticoids in vivo leads to erythrocytosis by generating erythroid progenitor cells with a constitutively active GR. Although remission rescues the erythrocytosis and the phenotype of the circulating CD34+ cells, a memory of other prior changes is maintained in remission.


Asunto(s)
Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT) , Policitemia , Humanos , Policitemia/etiología , Células Madre Hematopoyéticas/metabolismo , Glucocorticoides/farmacología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Dexametasona/farmacología , Células Cultivadas
2.
Elife ; 102021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33570494

RESUMEN

Erythroblastic islands are a specialized niche that contain a central macrophage surrounded by erythroid cells at various stages of maturation. However, identifying the precise genetic and transcriptional control mechanisms in the island macrophage remains difficult due to macrophage heterogeneity. Using unbiased global sequencing and directed genetic approaches focused on early mammalian development, we find that fetal liver macrophages exhibit a unique expression signature that differentiates them from erythroid and adult macrophage cells. The importance of erythroid Krüppel-like factor (EKLF)/KLF1 in this identity is shown by expression analyses in EKLF-/- and in EKLF-marked macrophage cells. Single-cell sequence analysis simplifies heterogeneity and identifies clusters of genes important for EKLF-dependent macrophage function and novel cell surface biomarkers. Remarkably, this singular set of macrophage island cells appears transiently during embryogenesis. Together, these studies provide a detailed perspective on the importance of EKLF in the establishment of the dynamic gene expression network within erythroblastic islands in the developing embryo and provide the means for their efficient isolation.


Asunto(s)
Eritropoyesis/genética , Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Macrófagos/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo
3.
Bio Protoc ; 11(23): e4243, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-35005088

RESUMEN

In vivo erythropoiesis occurs in the erythroblast island niche (EBI), comprising of a central macrophage that attaches to and aids the maturation of erythroid progenitors into mature reticulocytes. Macrophages in hematopoietic tissue such as embryonic fetal liver are heterogeneous and express the cell surface protein F4/80. Earlier methods of isolating F4/80+ macrophages from hematopoietic tissue relied on FACS sorting, but the relatively low numbers of F4/80+ cells obtained after FACS sometimes led to poor RNA quality. Additionally, since EBI macrophages are attached to erythroblasts, care must be taken to avoid contamination with bound erythroblasts. We have developed a novel method for isolating F4/80+ cells from E13.5 mouse fetal liver using magnetic nanoparticles, which can be performed on the lab bench. During cell suspension and homogenization, we also add a peptide that disrupts erythroid macrophage interactions and generates F4/80+ single cells free of erythroid contamination. Thus, our protocol generates a population enriched in F4/80+ cells that are healthy and ready for sensitive techniques such as single cell sequencing.

4.
Blood Cells Mol Dis ; 83: 102434, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32311573

RESUMEN

KLF1 (EKLF) is a master regulator of erythropoiesis and controls expression of a wide array of target genes. We interrogated human tissue microarray samples via immunohistological analysis to address whether levels of KLF1 protein are associated with leukemia. We have made the unexpected findings that higher KLF1 levels are correlated with cells containing abnormal chromosomes, and that high KLF1 expression is not limited to acute myeloid leukemia (AML) associated with erythroid/megakaryoblastic differentiation. Expression of KLF1 is associated with poor survival. Further analyses reveal that KLF1 directly regulates a number of genes that play a role in chromosomal integrity. Together these results suggest that monitoring KLF1 levels may provide a new marker for risk stratification and prognosis in patients with AML.


Asunto(s)
Aberraciones Cromosómicas , Regulación Leucémica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Leucemia Mieloide Aguda/genética , Adulto , Animales , Células COS , Chlorocebus aethiops , Estudios de Cohortes , Femenino , Humanos , Factores de Transcripción de Tipo Kruppel/análisis , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Adulto Joven
5.
Cancer Discov ; 9(6): 778-795, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30944118

RESUMEN

Even though the Ten-eleven translocation (TET) enzymes catalyze the generation of 5-hydroxymethylcytosines required for lineage commitment and subsequent differentiation of stem cells into erythroid cells, the mechanisms that link extracellular signals to TET activation and DNA hydroxymethylation are unknown. We demonstrate that hematopoietic cytokines phosphorylate TET2, leading to its activation in erythroid progenitors. Specifically, cytokine receptor-associated JAK2 phosphorylates TET2 at tyrosines 1939 and 1964. Phosphorylated TET2 interacts with the erythroid transcription factor KLF1, and this interaction with TET2 is increased upon exposure to erythropoietin. The activating JAK2V617F mutation seen in myeloproliferative disease patient samples and in mouse models is associated with increased TET activity and cytosine hydroxymethylation as well as genome-wide loss of cytosine methylation. These epigenetic and functional changes are also associated with increased expression of several oncogenic transcripts. Thus, we demonstrate that JAK2-mediated TET2 phosphorylation provides a mechanistic link between extracellular signals and epigenetic changes during hematopoiesis. SIGNIFICANCE: Identification of TET2 phosphorylation and activation by cytokine-stimulated JAK2 links extracellular signals to chromatin remodeling during hematopoietic differentiation. This provides potential avenues to regulate TET2 function in the context of myeloproliferative disorders and myelodysplastic syndromes associated with the JAK2V617F-activating mutation.This article is highlighted in the In This Issue feature, p. 681.


Asunto(s)
Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Hematopoyesis/genética , Janus Quinasa 2/metabolismo , Proteínas Proto-Oncogénicas/genética , Activación Transcripcional , Biomarcadores , Dioxigenasas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Fosforilación
6.
Sci Rep ; 8(1): 6587, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29700354

RESUMEN

Erythroid Krüppel-like Factor (EKLF/KLF1) is an erythroid-enriched transcription factor that plays a global role in all aspects of erythropoiesis, including cell cycle control and differentiation. We queried whether its mutation might play a role in red cell malignancies by genomic sequencing of the KLF1 transcription unit in cell lines, erythroid neoplasms, dysplastic disorders, and leukemia. In addition, we queried published databases from a number of varied sources. In all cases we only found changes in commonly notated SNPs. Our results suggest that if there are mutations in KLF1 associated with erythroid malignancies, they are exceedingly rare.


Asunto(s)
Genética de Población , Factores de Transcripción de Tipo Kruppel/metabolismo , Mutación , Transcripción Genética , Alelos , Diferenciación Celular/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Mapeo Cromosómico , Células Eritroides/citología , Células Eritroides/metabolismo , Eritropoyesis/genética , Variación Genética , Genómica/métodos , Humanos , Factores de Transcripción de Tipo Kruppel/clasificación , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
7.
Eur J Pharmacol ; 776: 9-18, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26879870

RESUMEN

Gamma globin induction remains a promising pharmacological therapeutic treatment mode for sickle cell anemia and beta thalassemia, however Hydroxyurea remains the only FDA approved drug which works via this mechanism. In this regard, we assayed the γ-globin inducing capacity of Cis-vaccenic acid (CVA). CVA induced differentiation of K562, JK1 and transgenic mice primary bone marrow hematopoietic progenitor stem cells. CVA also significantly up-regulated γ-globin gene expression in JK-1 and transgenic mice bone marrow erythroid progenitor stem cells (TMbmEPSCs) but not K562 cells without altering cell viability. Increased γ-globin expression was accompanied by KLF1 suppression in CVA induced JK-1 cells. Erythropoietin induced differentiation of JK-1 cells 24h before CVA induction did not significantly alter CVA induced differentiation and γ-globin expression in JK-1 cells. Inhibition of JK-1 and Transgenic mice bone marrow erythroid progenitor stem cells Fatty acid elongase 5 (Elovl5) and Δ(9) desaturase suppressed the γ-globin inductive effects of CVA. CVA treatment failed to rescue γ-globin expression in Elovl5 and Δ(9)-desaturase inhibited cells 48 h post inhibition in JK-1 cells. The data suggests that CVA directly modulates differentiation of JK-1 and TMbmEPSCs, and indirectly modulates γ-globin gene expression in these cells. Our findings provide important clues for further evaluations of CVA as a potential fetal hemoglobin therapeutic inducer.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Ácidos Oléicos/farmacología , Regulación hacia Arriba/efectos de los fármacos , gamma-Globinas/biosíntesis , Acetiltransferasas/antagonistas & inhibidores , Animales , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyetina/farmacología , Elongasas de Ácidos Grasos , Feto/metabolismo , Hemoglobinas/metabolismo , Humanos , Células K562 , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Transgénicos , Feniltiourea/análogos & derivados , Feniltiourea/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Tiocarbamatos/farmacología , gamma-Globinas/genética , gamma-Globinas/metabolismo
8.
PLoS Genet ; 11(10): e1005526, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26452208

RESUMEN

Circulating red blood cells (RBCs) are essential for tissue oxygenation and homeostasis. Defective terminal erythropoiesis contributes to decreased generation of RBCs in many disorders. Specifically, ineffective nuclear expulsion (enucleation) during terminal maturation is an obstacle to therapeutic RBC production in vitro. To obtain mechanistic insights into terminal erythropoiesis we focused on FOXO3, a transcription factor implicated in erythroid disorders. Using an integrated computational and experimental systems biology approach, we show that FOXO3 is essential for the correct temporal gene expression during terminal erythropoiesis. We demonstrate that the FOXO3-dependent genetic network has critical physiological functions at key steps of terminal erythropoiesis including enucleation and mitochondrial clearance processes. FOXO3 loss deregulated transcription of genes implicated in cell polarity, nucleosome assembly and DNA packaging-related processes and compromised erythroid enucleation. Using high-resolution confocal microscopy and imaging flow cytometry we show that cell polarization is impaired leading to multilobulated Foxo3-/- erythroblasts defective in nuclear expulsion. Ectopic FOXO3 expression rescued Foxo3-/- erythroblast enucleation-related gene transcription, enucleation defects and terminal maturation. Remarkably, FOXO3 ectopic expression increased wild type erythroblast maturation and enucleation suggesting that enhancing FOXO3 activity may improve RBCs production. Altogether these studies uncover FOXO3 as a novel regulator of erythroblast enucleation and terminal maturation suggesting FOXO3 modulation might be therapeutic in disorders with defective erythroid maturation.


Asunto(s)
Eritrocitos/metabolismo , Eritropoyesis/genética , Factores de Transcripción Forkhead/genética , Biología de Sistemas , Animales , Autofagia/genética , Células de la Médula Ósea/metabolismo , Polaridad Celular/genética , Eritroblastos/metabolismo , Eritrocitos/citología , Citometría de Flujo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Homeostasis , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo
9.
Mol Cell Biol ; 35(21): 3726-38, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26303528

RESUMEN

Understanding how transcriptional regulators are themselves controlled is important in attaining a complete picture of the intracellular effects that follow signaling cascades during early development and cell-restricted differentiation. We have addressed this issue by focusing on the regulation of EKLF/KLF1, a zinc finger transcription factor that plays a necessary role in the global regulation of erythroid gene expression. Using biochemical affinity purification, we have identified the DEK oncoprotein as a critical factor that interacts with an essential upstream enhancer element of the EKLF promoter and exerts a positive effect on EKLF levels. This element also binds a core set of erythroid transcription factors, suggesting that DEK is part of a tissue-restricted enhanceosome that contains BMP4-dependent and -independent components. Together with local enrichment of properly coded histones and an open chromatin domain, optimal transcriptional activation of the EKLF locus can be established.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Células Eritroides/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Oncogénicas/metabolismo , Regiones Promotoras Genéticas , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 4/metabolismo , Línea Celular , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/análisis , Regulación de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/análisis , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Oncogénicas/análisis , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Recombinantes/análisis , Proteínas Recombinantes/metabolismo , Transducción de Señal
10.
J Biol Chem ; 290(15): 9929-40, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713074

RESUMEN

Erythroid Kruppel-like factor (EKLF or KLF1) is a transcription factor crucial for red cell development that is directly involved in regulation of a large number of erythroid genes. EKLF serves mostly as an activator of expression of these genes; however, it can act also as a repressor. Here, we present evidence that EKLF interacts with proteins from the PIAS (protein inhibitor of activated STAT) family that convey repressive activity to EKLF in the absence of sumoylation. Our studies identify PIAS3 as a transcriptional corepressor of EKLF for at least a subset of its target genes during erythropoiesis (e.g. ß-globin, α-hemoglobin stabilizing protein). We demonstrate an interaction between EKLF and PIAS proteins confirmed by in vivo coimmunoprecipitation assays with both exogenous and endogenous proteins. We identified an LXXLL signature motif located near the N terminus of PIAS proteins that, although not involved in the EKLF-PIAS3 interaction, is required for the transrepression activity. Knockdown of endogenous PIAS3 accelerates differentiation of both murine erythroleukemia cells, as well as fetal liver cells, whereas an increase in PIAS3 levels inhibits this increase. Using chromatin immunoprecipitation assays, we show that PIAS3 preferentially occupies the ß-globin promoter in undifferentiated murine erythroleukemia cells. Together these results demonstrate that an interaction between EKLF and PIAS3 provides a novel mode of regulation of EKLF activity in the absence of sumolylation and furthermore shows an important involvement of PIAS proteins in erythropoiesis.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , Mutación , Proteínas Inhibidoras de STAT Activados/genética , Activación Transcripcional , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Western Blotting , Células COS , Diferenciación Celular/genética , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , Células K562 , Factores de Transcripción de Tipo Kruppel/metabolismo , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Ratones , Microscopía Confocal , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Inhibidoras de STAT Activados/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sumoilación , Globinas beta/genética , Globinas beta/metabolismo
11.
Exp Hematol ; 43(1): 65-70, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25283745

RESUMEN

Alternative splicing has emerged as a vital way to expand the functional repertoire of a set number of mammalian genes. For example, such changes can dramatically alter the function and cellular localization of transcription factors. With this in mind, we addressed whether EKLF/KLF1 mRNA, coding for a transcription factor that plays a critical role in erythropoietic gene regulation, is alternatively spliced. We find that EKLF mRNA undergoes exon skipping only in primary tissues and that this splice variant (SV) remains at a very low level in both embryonic and adult erythroid cells, as well as during terminal differentiation. The resultant protein is truncated and partially encodes a non-erythroid Krüppel-like factor amino acid sequence. Its overexpression can alter full-length erythroid Krüppel-like factor function at selected promoters. We discuss these results in the context of stress and with respect to recent global studies on the role of alternative splicing during terminal erythroid differentiation.


Asunto(s)
Empalme Alternativo , Células Eritroides/metabolismo , Eritropoyesis/genética , Factores de Transcripción de Tipo Kruppel/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular Tumoral , Linaje de la Célula , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Células K562 , Factores de Transcripción de Tipo Kruppel/fisiología , Leucemia Eritroblástica Aguda/patología , Ratones , Datos de Secuencia Molecular , Flebotomía , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Bazo/metabolismo , Transcripción Genética , Activación Transcripcional
12.
Development ; 141(11): 2245-54, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24866116

RESUMEN

The erythroblastic island provides an important nutritional and survival support niche for efficient erythropoietic differentiation. Island integrity is reliant on adhesive interactions between erythroid and macrophage cells. We show that erythroblastic islands can be formed from single progenitor cells present in differentiating embryoid bodies, and that these correspond to erythro-myeloid progenitors (EMPs) that first appear in the yolk sac of the early developing embryo. Erythroid Krüppel-like factor (EKLF; KLF1), a crucial zinc finger transcription factor, is expressed in the EMPs, and plays an extrinsic role in erythroid maturation by being expressed in the supportive macrophage of the erythroblastic island and regulating relevant genes important for island integrity within these cells. Together with its well-established intrinsic contributions to erythropoiesis, EKLF thus plays a coordinating role between two different cell types whose interaction provides the optimal environment to generate a mature red blood cell.


Asunto(s)
Eritrocitos/citología , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Nicho de Células Madre , Animales , Adhesión Celular , Diferenciación Celular , Membrana Celular/metabolismo , Células Madre Embrionarias/citología , Eritroblastos/citología , Células Precursoras Eritroides/citología , Eritropoyesis/fisiología , Macrófagos/citología , Ratones , Regiones Promotoras Genéticas , Reticulocitos/citología , Células Madre/citología , Saco Vitelino/fisiología , Dedos de Zinc
13.
Int J Biochem Cell Biol ; 50: 112-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24594363

RESUMEN

To clarify the role of HDACs in erythropoiesis, expression, activity and function of class I (HDAC1, HDAC2, HDAC3) and class IIa (HDAC4, HDAC5) HDACs during in vitro maturation of human erythroblasts were compared. During erythroid maturation, expression of HDAC1, HDAC2 and HDAC3 remained constant and activity and GATA1 association (its partner of the NuRD complex), of HDAC1 increased. By contrast, HDAC4 content drastically decreased and HDAC5 remained constant in content but decreased in activity. In erythroid cells, pull down experiments identified the presence of a novel complex formed by HDAC5, GATA1, EKLF and pERK which was instead undetectable in cells of the megakaryocytic lineage. With erythroid maturation, association among HDAC5, GATA1 and EKLF persisted but levels of pERK sharply decreased. Treatment of erythroleukemic cells with inhibitors of ERK phosphorylation reduced by >90% the total and nuclear content of HDAC5, GATA1 and EKLF, suggesting that ERK phosphorylation is required for the formation of this complex. Based on the function of class IIa HDACs as chaperones of other proteins to the nucleus and the erythroid-specificity of HDAC5 localization, this novel HDAC complex was named nuclear remodeling shuttle erythroid (NuRSERY). Exposure of erythroid cells to the class II-selective HDAC inhibitor (HDACi) APHA9 increased γ/(γ+ß) globin expression ratios (Mai et al., 2007), suggesting that NuRSERY may regulate globin gene expression. In agreement with this hypothesis, exposure of erythroid cells to APHA9 greatly reduced the association among HDAC5, GATA1 and EKLF. Since exposure to APHA9 did not affect survival rates or p21 activation, NuRSERY may represent a novel, possibly less toxic, target for epigenetic therapies of hemoglobinopaties and other disorders.


Asunto(s)
Células Eritroides/metabolismo , Factor de Transcripción GATA1/metabolismo , Histona Desacetilasas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , eIF-2 Quinasa/metabolismo , Diferenciación Celular/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Eritroblastos/citología , Eritroblastos/enzimología , Eritroblastos/patología , Células Eritroides/citología , Células Eritroides/enzimología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células K562 , Megacariocitos/citología , Megacariocitos/enzimología , Megacariocitos/metabolismo , Fosforilación
14.
Blood Cells Mol Dis ; 51(2): 71-5, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23522491

RESUMEN

KLF1 is an erythroid specific transcription factor that is involved in erythroid lineage commitment, globin switching and terminal red blood cell maturation. Various mutations of KLF1 have been identified in humans, which have led to both benign and pathological phenotypes. The E325K mutation, within the second zinc finger of the KLF1 gene, has been shown to cause a new form of congenital dyserythropoietic anemia (CDA) now labeled as CDA type IV. We report the fourth documented case of this mutation, and propose a clinical diagnostic model to better identify this disease in other patients. Our patient is a Taiwanese child who presented to us at 8years of age with severe hemolytic anemia, splenomegaly, elevated fetal hemoglobin (HbF), iron overload, and dyserythropoiesis in the bone marrow. KLF1 sequence analysis revealed a G-to-A transition in one allele of exon 3, which resulted in the substitution of a glutamate 325 by a lysine. Flow cytometry analysis revealed decreased protein expression of CD44 on the red blood cells, and decreased red blood cell deformability as measured using an ektacytometer. Blood typing revealed his red blood cells to be Co(a-b-), In(b-), LW(ab-) and Lu(b+), even though DNA testing predicted that he would be Co(a+b-) and LW(a+b-). This newly discovered CDA combines features of a hemoglobinopathy, RBC membrane defect and hereditary persistence of HbF (HPFH) which are not seen in the previous types of CDA. Increased awareness of this phenotype may improve the more prompt and accurate diagnosis of these patients.


Asunto(s)
Anemia Diseritropoyética Congénita/diagnóstico , Anemia Diseritropoyética Congénita/genética , Pueblo Asiatico/genética , Factores de Transcripción de Tipo Kruppel/genética , Mutación , Anemia Diseritropoyética Congénita/terapia , Médula Ósea/patología , Niño , Análisis Mutacional de ADN , Membrana Eritrocítica/metabolismo , Humanos , Masculino , Fragilidad Osmótica/genética , Taiwán
15.
Mol Cell Biol ; 33(1): 4-13, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23090966

RESUMEN

Erythroid Krüppel-like factor (EKLF or KLF1) is a transcriptional regulator that plays a critical role in lineage-restricted control of gene expression. KLF1 expression and activity are tightly controlled in a temporal and differentiation stage-specific manner. The mechanisms by which KLF1 is regulated encompass a range of biological processes, including control of KLF1 RNA transcription, protein stability, localization, and posttranslational modifications. Intact KLF1 regulation is essential to correctly regulate erythroid function by gene transcription and to maintain hematopoietic lineage homeostasis by ensuring a proper balance of erythroid/megakaryocytic differentiation. In turn, KLF1 regulates erythroid biology by a wide variety of mechanisms, including gene activation and repression by regulation of chromatin configuration, transcriptional initiation and elongation, and localization of gene loci to transcription factories in the nucleus. An extensive series of biochemical, molecular, and genetic analyses has uncovered some of the secrets of its success, and recent studies are highlighted here. These reveal a multilayered set of control mechanisms that enable efficient and specific integration of transcriptional and epigenetic controls and that pave the way for proper lineage commitment and differentiation.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Dedos de Zinc/inmunología , Dedos de Zinc/fisiología
16.
J Biol Chem ; 287(44): 36720-31, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22982397

RESUMEN

During erythropoiesis, erythropoietin stimulates induction of erythroid transcription factors that activate expression of erythroid genes including the erythropoietin receptor (EPO-R) that results in increased sensitivity to erythropoietin. DNA binding of the basic helix-loop-helix transcription factor, TAL1/SCL, is required for normal erythropoiesis. A link between elevated TAL1 and excessive erythrocytosis is suggested by erythroid progenitor cells from a patient that exhibits unusually high sensitivity to erythropoietin with concomitantly elevated TAL1 and EPO-R expression. We found that TAL1 regulates EPO-R expression mediated via three conserved E-box binding motifs (CAGCTG) in the EPO-R 5' untranslated transcribed region. TAL1 increases association of the GATA-1·TAL1·LMO2·LDB1 transcription activation complex to the region that includes the transcription start site and the 5' GATA and 3' E-box motifs flanking the EPO-R transcription start site suggesting that TAL1 promotes accessibility of this region. Nucleosome shifting has been demonstrated to facilitate TAL1 but not GATA-1 binding to regulate target gene expression. Accordingly, we observed that with induced expression of EPO-R in hemotopoietic progenitor cells, nucleosome phasing shifts to increase the linker region containing the EPO-R transcription start site and TAL1 binds to the flanking 5' GATA and 3' E-box regions of the promoter. These data suggest that TAL1 binds to the EPO-R promoter to activate EPO-R expression and provides a potential link to elevated EPO-R expression leading to hypersensitivity to erythropoietin and the resultant excessive erythrocytosis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica , Policitemia/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Eritropoyetina/metabolismo , Adulto , Antígenos CD/metabolismo , Sitios de Unión , Estudios de Casos y Controles , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Análisis Mutacional de ADN , Células Eritroides/metabolismo , Eritropoyetina/fisiología , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Expresión Génica , Genes Reporteros , Células Madre Hematopoyéticas/metabolismo , Humanos , Janus Quinasa 2/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Masculino , Mutación Missense , Policitemia/genética , Policitemia/patología , Regiones Promotoras Genéticas , Unión Proteica , Receptores de Eritropoyetina/genética , Proteína 1 de la Leucemia Linfocítica T Aguda , Globinas beta/genética , Globinas beta/metabolismo
17.
J Biol Chem ; 287(19): 15193-204, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22393050

RESUMEN

Erythroid Krüppel-like factor (EKLF; KLF1) is an erythroid-specific transcription factor required for the transcription of genes that regulate erythropoiesis. In this paper, we describe the identification of a novel EKLF interactor, Ppm1b, a serine-threonine protein phosphatase that has been implicated in the attenuation of NFκB signaling and the regulation of Cdk9 phosphorylation status. We show that Ppm1b interacts with EKLF via its PEST1 sequence. However, its genetic regulatory role is complex. Using a promoter-reporter assay in an erythroid cell line, we show that Ppm1b superactivates EKLF at the ß-globin and BKLF promoters, dependent on intact Ppm1b phosphatase activity. Conversely, depletion of Ppm1b in CD34(+) cells leads to a higher level of endogenous ß-globin gene activation after differentiation. We also observe that Ppm1b likely has an indirect role in regulating EKLF turnover via its zinc finger domain. Together, these studies show that Ppm1b plays a multilayered role in regulating the availability and optimal activity of the EKLF protein in erythroid cells.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Células Madre Hematopoyéticas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Antígenos CD4/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Humanos , Inmunoprecipitación , Células K562 , Factores de Transcripción de Tipo Kruppel/genética , Datos de Secuencia Molecular , Fosfoproteínas Fosfatasas/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteína Fosfatasa 2C , Interferencia de ARN , Homología de Secuencia de Aminoácido , Transcripción Genética , Globinas beta/genética
18.
Proc Natl Acad Sci U S A ; 107(39): 16846-51, 2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20837550

RESUMEN

Pharmacological treatments designed to reactivate fetal γ-globin can lead to an effective and successful clinical outcome in patients with hemoglobinopathies. However, new approaches remain highly desired because such treatments are not equally effective for all patients, and toxicity issues remain. We have taken a systematic approach to develop an embedded chimeric peptide nucleic acid (PNA) that effectively enters the cell and the nucleus, binds to its target site at the human fetal γ-globin promoter, and reactivates this transcript in adult transgenic mouse bone marrow and human primary peripheral blood cells. In vitro and in vivo DNA-binding assays in conjunction with live-cell imaging have been used to establish and optimize chimeric PNA design parameters that lead to successful gene activation. Our final molecule contains a specific γ-promoter-binding PNA sequence embedded within two amino acid motifs: one leads to efficient cell/nuclear entry, and the other generates transcriptional reactivation of the target. These embedded PNAs overcome previous limitations and are generally applicable to the design of in vivo transcriptional activation reagents that can be directed to any promoter region of interest and are of direct relevance to clinical applications that would benefit from such a need.


Asunto(s)
Diseño de Fármacos , Expresión Génica/efectos de los fármacos , Hemoglobinopatías/tratamiento farmacológico , Ácidos Nucleicos de Péptidos/farmacología , gamma-Globinas/genética , Animales , Células de la Médula Ósea/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Hemoglobinopatías/genética , Humanos , Ratones , Ácidos Nucleicos de Péptidos/química , Ácidos Nucleicos de Péptidos/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos
19.
Proc Natl Acad Sci U S A ; 106(11): 4213-8, 2009 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-19251649

RESUMEN

The architectural layout of a eukaryotic RNA polymerase II core promoter plays a role in general transcriptional activation. However, its role in tissue-specific expression is not known. For example, differing modes of its recognition by general transcription machinery can provide an additional layer of control within which a single tissue-restricted transcription factor may operate. Erythroid Kruppel-like factor (EKLF) is a hematopoietic-specific transcription factor that is critical for the activation of subset of erythroid genes. We find that EKLF interacts with TATA binding protein-associated factor 9 (TAF9), which leads to important consequences for expression of adult beta-globin. First, TAF9 functionally supports EKLF activity by enhancing its ability to activate the beta-globin gene. Second, TAF9 interacts with a conserved beta-globin downstream promoter element, and ablation of this interaction by beta-thalassemia-causing mutations decreases its promoter activity and disables superactivation. Third, depletion of EKLF prevents recruitment of TAF9 to the beta-globin promoter, whereas depletion of TAF9 drastically impairs beta-promoter activity. However, a TAF9-independent mode of EKLF transcriptional activation is exhibited by the alpha-hemoglobin-stabilizing protein (AHSP) gene, which does not contain a discernable downstream promoter element. In this case, TAF9 does not enhance EKLF activity and depletion of TAF9 has no effect on AHSP promoter activation. These studies demonstrate that EKLF directs different modes of tissue-specific transcriptional activation depending on the architecture of its target core promoter.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/fisiología , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/metabolismo , Activación Transcripcional , Proteínas Sanguíneas , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Chaperonas Moleculares , Mutación , Regiones Promotoras Genéticas , Distribución Tisular , Factores de Transcripción , Globinas beta/biosíntesis , Globinas beta/genética , Talasemia beta/genética
20.
Eur J Haematol ; 82(6): 466-76, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19220418

RESUMEN

OBJECTIVES: The erythroid Kruppel-like factor (EKLF) is an essential transcription factor for beta-type globin gene switching, and specifically activates transcription of the adult beta-globin gene promoter. We sought to determine if EKLF is also required for activation of the gamma-globin gene by short-chain fatty acid (SCFA) derivatives, which are now entering clinical trials. METHODS: The functional and physical interaction of EKLF and co-regulatory molecules with the endogenous human globin gene promoters was studied in primary human erythroid progenitors and cell lines, using chromatin immunoprecipitation (ChIP) assays and genetic manipulation of the levels of EKLF and co-regulators. RESULTS AND CONCLUSIONS: Knockdown of EKLF prevents SCFA-induced expression of the gamma-globin promoter in a stably expressed microLCRbeta(pr)R(luc) (A)gamma(pr)F(luc) cassette, and prevents induction of the endogenous gamma-globin gene in primary human erythroid progenitors. EKLF is actively recruited to endogenous gamma-globin gene promoters after exposure of primary human erythroid progenitors, and murine hematopoietic cell lines, to SCFA derivatives. The core ATPase BRG1 subunit of the human SWI/WNF complex, a ubiquitous multimeric complex that regulates gene expression by remodeling nucleosomal structure, is also required for gamma-globin gene induction by SCFA derivatives. BRG1 is actively recruited to the endogenous gamma-globin promoter of primary human erythroid progenitors by exposure to SCFA derivatives, and this recruitment is dependent upon the presence of EKLF. These findings demonstrate that EKLF, and the co-activator BRG1, previously demonstrated to be required for definitive or adult erythropoietic patterns of globin gene expression, are co-opted by SCFA derivatives to activate the fetal globin genes.


Asunto(s)
Ácidos Grasos Volátiles/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , gamma-Globinas/genética , Animales , Células Cultivadas , ADN Helicasas/fisiología , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal , Humanos , Factores de Transcripción de Tipo Kruppel/fisiología , Ratones , Proteínas Nucleares/fisiología , Transporte de Proteínas , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA