Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Adv Exp Med Biol ; 1459: 217-242, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39017846

RESUMO

Erythroid Krüppel-like factor (KLF1), first discovered in 1992, is an erythroid-restricted transcription factor (TF) that is essential for terminal differentiation of erythroid progenitors. At face value, KLF1 is a rather inconspicuous member of the 26-strong SP/KLF TF family. However, 30 years of research have revealed that KLF1 is a jack of all trades in the molecular control of erythropoiesis. Initially described as a one-trick pony required for high-level transcription of the adult HBB gene, we now know that it orchestrates the entire erythroid differentiation program. It does so not only as an activator but also as a repressor. In addition, KLF1 was the first TF shown to be directly involved in enhancer/promoter loop formation. KLF1 variants underlie a wide range of erythroid phenotypes in the human population, varying from very mild conditions such as hereditary persistence of fetal hemoglobin and the In(Lu) blood type in the case of haploinsufficiency, to much more serious non-spherocytic hemolytic anemias in the case of compound heterozygosity, to dominant congenital dyserythropoietic anemia type IV invariably caused by a de novo variant in a highly conserved amino acid in the KLF1 DNA-binding domain. In this chapter, we present an overview of the past and present of KLF1 research and discuss the significance of human KLF1 variants.


Assuntos
Eritropoese , Fatores de Transcrição Kruppel-Like , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Humanos , Eritropoese/genética , Animais , Diferenciação Celular/genética , Células Eritroides/metabolismo , Células Eritroides/citologia , Mutação
2.
J Biol Chem ; 299(12): 105489, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38000658

RESUMO

EKLF/KLF1 is an essential transcription factor that plays a global role in erythroid transcriptional activation. Regulation of KLF1 is of interest, as it displays a highly restricted expression pattern, limited to erythroid cells and its progenitors. Here we use biochemical affinity purification to identify the DDX5/p68 protein as an activator of KLF1 by virtue of its interaction with the erythroid-specific DNAse hypersensitive site upstream enhancer element (EHS1). We further show that this protein associates with DEK and CTCF. We postulate that the range of interactions of DDX5/p68 with these and other proteins known to interact with this element render it part of the enhanseosome complex critical for optimal expression of KLF1 and enables the formation of a proper chromatin configuration at the Klf1 locus. These individual interactions provide quantitative contributions that, in sum, establish the high-level activity of the Klf1 promoter and suggest they can be selectively manipulated for clinical benefit.


Assuntos
RNA Helicases DEAD-box , Elementos Facilitadores Genéticos , Fatores de Transcrição Kruppel-Like , Eritropoese , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo
3.
Front Cell Dev Biol ; 11: 1148013, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37113767

RESUMO

Introduction: Congenital dyserythropoietic anaemia (CDA) type IV has been associated with an amino acid substitution, Glu325Lys (E325K), in the transcription factor KLF1. These patients present with a range of symptoms, including the persistence of nucleated red blood cells (RBCs) in the peripheral blood which reflects the known role for KLF1 within the erythroid cell lineage. The final stages of RBCs maturation and enucleation take place within the erythroblastic island (EBI) niche in close association with EBI macrophages. It is not known whether the detrimental effects of the E325K mutation in KLF1 are restricted to the erythroid lineage or whether deficiencies in macrophages associated with their niche also contribute to the disease pathology. Methods: To address this question, we generated an in vitro model of the human EBI niche using induced pluripotent stem cells (iPSCs) derived from one CDA type IV patient as well as two iPSC lines genetically modified to express an KLF1-E325K-ERT2 protein that could be activated with 4OH-tamoxifen. The one patient iPSC line was compared to control lines from two healthy donors and the KLF1-E325K-ERT2 iPSC line to one inducible KLF1-ERT2 line generated from the same parental iPSCS. Results: The CDA patient-derived iPSCs and iPSCs expressing the activated KLF1-E325K-ERT2 protein showed significant deficiencies in the production of erythroid cells with associated disruption of some known KLF1 target genes. Macrophages could be generated from all iPSC lines but when the E325K-ERT2 fusion protein was activated, we noted the generation of a slightly less mature macrophage population marked by CD93. A subtle trend in their reduced ability to support RBC enucleation was also associated with macrophages carrying the E325K-ERT2 transgene. Discussion: Taken together these data support the notion that the clinically significant effects of the KLF1-E325K mutation are primarily associated with deficiencies in the erythroid lineage but it is possible that deficiencies in the niche might have the potential to exacerbate the condition. The strategy we describe provides a powerful approach to assess the effects of other mutations in KLF1 as well as other factors associated with the EBI niche.

4.
Blood ; 141(25): 3039-3054, 2023 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-37084386

RESUMO

Red blood cell disorders can result in severe anemia. One such disease congenital dyserythropoietic anemia IV (CDA IV) is caused by the heterozygous mutation E325K in the transcription factor KLF1. However, studying the molecular basis of CDA IV is severely impeded by the paucity of suitable and adequate quantities of material from patients with anemia and the rarity of the disease. We, therefore, took a novel approach, creating a human cellular disease model system for CDA IV that accurately recapitulates the disease phenotype. Next, using comparative proteomics, we reveal extensive distortion of the proteome and a wide range of disordered biological processes in CDA IV erythroid cells. These include downregulated pathways the governing cell cycle, chromatin separation, DNA repair, cytokinesis, membrane trafficking, and global transcription, and upregulated networks governing mitochondrial biogenesis. The diversity of such pathways elucidates the spectrum of phenotypic abnormalities that occur with CDA IV and impairment to erythroid cell development and survival, collectively explaining the CDA IV disease phenotype. The data also reveal far more extensive involvement of KLF1 in previously assigned biological processes, along with novel roles in the regulation of intracellular processes not previously attributed to this transcription factor. Overall, the data demonstrate the power of such a model cellular system to unravel the molecular basis of disease and how studying the effects of a rare mutation can reveal fundamental biology.


Assuntos
Anemia Diseritropoética Congênita , Humanos , Anemia Diseritropoética Congênita/genética , Mutação , Regulação da Expressão Gênica , Fenótipo , Fatores de Transcrição/genética
5.
Haematologica ; 108(4): 1053-1067, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35861015

RESUMO

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.


Assuntos
Hipersecreção Hipofisária de ACTH , Policitemia , Humanos , Policitemia/etiologia , Células-Tronco Hematopoéticas/metabolismo , Glucocorticoides/farmacologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Dexametasona/farmacologia , Células Cultivadas
6.
Cell Rep ; 41(12): 111830, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36543143

RESUMO

EKLF/Klf1 is a zinc-finger transcription activator essential for erythroid lineage commitment and terminal differentiation. Using ChIP-seq, we investigate EKLF DNA binding and transcription activation mechanisms during mouse embryonic erythropoiesis. We utilize the Nan/+ mouse that expresses the EKLF-E339D (Nan) variant mutated in its conserved zinc-finger region and address the mechanism of hypomorphic and neomorphic changes in downstream gene expression. First, we show that Nan-EKLF limits normal EKLF binding to a subset of its sites. Second, we find that ectopic binding of Nan-EKLF occurs largely at enhancers and activates transcription through pioneering activity. Third, we find that for a subset of ectopic targets, gene activation is achieved in Nan/+ only by Nan-EKLF binding to distal enhancers, leading to RNA polymerase II pause-release. These results have general applicability to understanding how a DNA binding variant factor confers dominant disruptive effects on downstream gene expression even in the presence of its normal counterpart.


Assuntos
Fatores de Transcrição Kruppel-Like , RNA Polimerase II , Camundongos , Animais , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Eritropoese/genética , DNA/metabolismo , Zinco
7.
Bio Protoc ; 12(3): e4329, 2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-35284601

RESUMO

[This corrects the article DOI: 10.21769/BioProtoc.4243.].

8.
Front Genet ; 12: 756028, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34880902

RESUMO

During definitive erythropoiesis, maturation of erythroid progenitors into enucleated reticulocytes requires the erythroblastic island (EBI) niche comprising a central macrophage attached to differentiating erythroid progenitors. Normally, the macrophage provides a nurturing environment for maturation of erythroid cells. Its critical physiologic importance entails aiding in recovery from anemic insults, such as systemic stress or acquired disease. Considerable interest in characterizing the central macrophage of the island niche led to the identification of putative cell surface markers enriched in island macrophages, enabling isolation and characterization. Recent studies focus on bulk and single cell transcriptomics of the island macrophage during adult steady-state erythropoiesis and embryonic erythropoiesis. They reveal that the island macrophage is a distinct cell type but with widespread cellular heterogeneity, likely suggesting distinct developmental origins and biological function. These studies have also uncovered transcriptional programs that drive gene expression in the island macrophage. Strikingly, the master erythroid regulator EKLF/Klf1 seems to also play a major role in specifying gene expression in island macrophages, including a putative EKLF/Klf1-dependent transcription circuit. Our present review and analysis of mouse single cell genetic patterns suggest novel expression characteristics that will enable a clear enrichment of EBI subtypes and resolution of island macrophage heterogeneity. Specifically, the discovery of markers such as Epor, and specific features for EKLF/Klf1-expressing island macrophages such as Sptb and Add2, or for SpiC-expressing island macrophage such as Timd4, or for Maf/Nr1h3-expressing island macrophage such as Vcam1, opens exciting possibilities for further characterization of these unique macrophage cell types in the context of their critical developmental function.

9.
Elife ; 102021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33570494

RESUMO

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.


Assuntos
Eritropoese/genética , Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Macrófagos/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo
10.
Bio Protoc ; 11(23): e4243, 2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-35005088

RESUMO

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.

11.
Mutat Res Rev Mutat Res ; 786: 108336, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33339573

RESUMO

The etiology and severity of anemia, a common blood disorder, are diverse. Dominant mutations in Krüppel-like factor 1 (KLF1/EKLF) underlie the molecular basis for some of them. KLF1 is a zinc finger transcription factor that plays an essential role in red blood cell proliferation and differentiation. Mutations have been identified in the KLF1 gene that cause hematologic diseases. Two of these alter one allele but generate an extreme phenotype: the mouse Nan mutation (E339D) leads to hemolytic neonatal anemia with hereditary spherocytosis, and the human CDA mutation (E325K) causes congenital dyserythropoietic anemia (CDA) type IV. These modify functionally important amino acids in the zinc finger DNA-binding domain at positions involved in direct interactions with regulatory elements of KLF1's target genes. Although the two dominant mutations alter the same evolutionarily conserved glutamic acid residue, the substitutions are not equivalent and lead to divergent consequences for the molecular mechanisms underlying activity of these mutants, particularly in recognition and interaction with their unique binding sites. Consequently, the properties of the protein are transformed such that it acquires novel dominant characteristics whose effects may not be limited to the erythroid compartment. KLF1 mutants cause loss-of-function/haploinsufficiency effects on some KLF1 wild-type target genes, while at the same time gain-of-function effects activate ectopic sites and neomorphic gene expression. Such anomalies not only lead to intrinsic red cell problems, but also to expression of non-erythroid genes that systemically disturb organ development. This review highlights recent molecular, biochemical, and genetic studies of KLF1 mutants, particularly the dramatic consequences that come from just a single amino acid change. The study of these variants provides an important contribution to the overall understanding of the DNA-protein interface of the zinc finger subtype of transcription factors, and the potential clinical consequences of what might appear to be a minor change in sequence.


Assuntos
Anemia/genética , Fatores de Transcrição Kruppel-Like/genética , Sítios de Ligação , Diferenciação Celular , Humanos , Mutação de Sentido Incorreto , Fenótipo
12.
13.
Stem Cell Res ; 45: 101837, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32413789

RESUMO

Hemoglobin production during mammalian development is characterized by temporal switches of the genes coding for the α- and ß-globin chains. Defects in this controlled process can lead to hemoglobinapathies such as sickle cell disease and ß-thalassemia. The ability of human embryonic stem cells (hESC) to proceed through hematopoiesis could provide a clinically useful source of red blood cells. However, hESC-derived red cells exhibit an embryonic/fetal, but not adult, mode of hemoglobin expression. The resource described here is a hESC line engineered to express a reporter from its adult globin promoter, providing a screening platform for small molecules that lead to efficient induction of adult globin.


Assuntos
Edição de Genes , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Adulto , Animais , Linhagem Celular , Células-Tronco Embrionárias , Hematopoese , Humanos , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética
14.
Blood Cells Mol Dis ; 83: 102434, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32311573

RESUMO

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.


Assuntos
Aberrações Cromossômicas , Regulação Leucêmica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Leucemia Mieloide Aguda/genética , Adulto , Animais , Células COS , Chlorocebus aethiops , Estudos de Coortes , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/análise , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Adulto Jovem
15.
Mol Cell Biol ; 40(5)2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31818881

RESUMO

Krüppel-like factor 1 (KLF1/EKLF) is a transcription factor that globally activates genes involved in erythroid cell development. Various mutations are identified in the human KLF1 gene. The E325K mutation causes congenital dyserythropoietic anemia (CDA) type IV, characterized by severe anemia and non-erythroid-cell-related symptoms. The CDA mutation is in the second zinc finger of KLF1 at a position functionally involved in its interactions with DNA. The molecular parameters of how CDA-KLF1 exerts its biological effects have not been addressed. Here, using an in vitro selection strategy, we determined the preferred DNA-binding site for CDA-KLF1. Binding to the deduced consensus sequence is supported by in vitro gel shifts and by in vivo functional reporter gene studies. Two significant changes compared to wild-type (WT) binding are observed: G is selected as the middle nucleotide, and the 3' portion of the consensus sequence is more degenerate. As a consequence, CDA-KLF1 did not bind the WT consensus sequence. However, activation of ectopic sites is promoted. Continuous activation of WT target genes occurs if they fortuitously contain the novel CDA site nearby. Our findings provide a molecular understanding of how a single mutation in the KLF1 zinc finger exerts effects on erythroid physiology in CDA type IV.


Assuntos
Anemia Diseritropoética Congênita/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Mutação , Animais , Sítios de Ligação , Células COS , Chlorocebus aethiops , DNA/metabolismo , Genes Reporter , Humanos , Células K562 , Luciferases/genética , Oligonucleotídeos/metabolismo , Reprodutibilidade dos Testes , Dedos de Zinco
16.
Cancer Discov ; 9(6): 778-795, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30944118

RESUMO

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.


Assuntos
Citocinas/metabolismo , Proteínas de Ligação a DNA/genética , Hematopoese/genética , Janus Quinase 2/metabolismo , Proteínas Proto-Oncogênicas/genética , Ativação Transcricional , Biomarcadores , Dioxigenases , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fosforilação
17.
Haematologica ; 104(12): 2372-2380, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30872368

RESUMO

Congenital dyserythropoietic anemia type IV is caused by a heterozygous mutation, Glu325Lys (E325K), in the KLF1 transcription factor. Molecular characteristics of this disease have not been clarified, partly due to its rarity. We expanded erythroid cells from a patient's peripheral blood and analyzed its global expression pattern. We find that a large number of erythroid pathways are disrupted, particularly those related to membrane transport, globin regulation, and iron utilization. The altered genetics lead to significant deficits in differentiation. Glu325 is within the KLF1 zinc finger domain at an amino acid critical for site specific DNA binding. The change to Lys is predicted to significantly alter the target site recognition sequence, both by subverting normal recognition and by enabling interaction with novel sites. Consistent with this, we find high level ectopic expression of genes not normally present in the red cell. These altered properties explain patients' clinical and phenotypic features, and elucidate the dominant character of the mutation.


Assuntos
Anemia Diseritropoética Congênita/genética , Anemia Diseritropoética Congênita/patologia , Células Eritroides/patologia , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Fatores de Transcrição Kruppel-Like/genética , Mutação , Diferenciação Celular , Células Eritroides/metabolismo , Humanos
18.
Sci Rep ; 8(1): 12793, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143664

RESUMO

Anemic Nan mice carry a mutation (E339D) in the second zinc finger of erythroid transcription factor KLF1. Nan-KLF1 fails to bind a subset of normal KLF1 targets and ectopically binds a large set of genes not normally engaged by KLF1, resulting in a corrupted fetal liver transcriptome. Here, we performed RNAseq using flow cytometric-sorted spleen erythroid precursors from adult Nan and WT littermates rendered anemic by phlebotomy to identify global transcriptome changes specific to the Nan Klf1 mutation as opposed to anemia generally. Mutant Nan-KLF1 leads to extensive and progressive transcriptome corruption in adult spleen erythroid precursors such that stress erythropoiesis is severely compromised. Terminal erythroid differentiation is defective in the bone marrow as well. Principle component analysis reveals two major patterns of differential gene expression predicting that defects in basic cellular processes including translation, cell cycle, and DNA repair could contribute to disordered erythropoiesis and anemia in Nan. Significant erythroid precursor stage specific changes were identified in some of these processes in Nan. Remarkably, however, despite expression changes in large numbers of associated genes, most basic cellular processes were intact in Nan indicating that developing red cells display significant physiological resiliency and establish new homeostatic set points in vivo.


Assuntos
Envelhecimento/patologia , Anemia/genética , Anemia/patologia , Diferenciação Celular/genética , Eritropoese/genética , Fatores de Transcrição Kruppel-Like/genética , Mutação/genética , Transcriptoma/genética , Animais , Sequência de Bases , Ciclo Celular/genética , Dano ao DNA , Células Eritroides/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Ontologia Genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/embriologia , Fígado/metabolismo , Camundongos , Camundongos Mutantes , Mitofagia/genética , Anotação de Sequência Molecular , Análise de Componente Principal , Espécies Reativas de Oxigênio/metabolismo , Baço/embriologia , Baço/metabolismo
19.
Sci Rep ; 8(1): 6587, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29700354

RESUMO

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.


Assuntos
Genética Populacional , Fatores de Transcrição Kruppel-Like/metabolismo , Mutação , Transcrição Gênica , Alelos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Mapeamento Cromossômico , Células Eritroides/citologia , Células Eritroides/metabolismo , Eritropoese/genética , Variação Genética , Genômica/métodos , Humanos , Fatores de Transcrição Kruppel-Like/classificação , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas
20.
Development ; 144(3): 430-440, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28143845

RESUMO

Transcription factor control of cell-specific downstream targets can be significantly altered when the controlling factor is mutated. We show that the semi-dominant neonatal anemia (Nan) mutation in the EKLF/KLF1 transcription factor leads to ectopic expression of proteins that are not normally expressed in the red blood cell, leading to systemic effects that exacerbate the intrinsic anemia in the adult and alter correct development in the early embryo. Even when expressed as a heterozygote, the Nan-EKLF protein accomplishes this by direct binding and aberrant activation of genes encoding secreted factors that exert a negative effect on erythropoiesis and iron use. Our data form the basis for a novel mechanism of physiological deficiency that is relevant to human dyserythropoietic anemia and likely other disease states.


Assuntos
Anemia Neonatal/genética , Fatores de Transcrição Kruppel-Like/genética , Mutação , Substituição de Aminoácidos , Anemia Neonatal/sangue , Anemia Neonatal/embriologia , Animais , Animais Recém-Nascidos , Citocinas/sangue , DNA/genética , DNA/metabolismo , Modelos Animais de Doenças , Eritrócitos/metabolismo , Eritropoese/genética , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Humanos , Fatores de Transcrição Kruppel-Like/sangue , Fatores de Transcrição Kruppel-Like/deficiência , Camundongos , Camundongos Knockout , Camundongos Mutantes , Modelos Biológicos , Proteínas Musculares/sangue , Proteínas Mutantes/sangue , Proteínas Mutantes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA