Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
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
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.
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
4.
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
5.
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
6.
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
7.
Nucleic Acids Res ; 45(3): 1130-1143, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28180284

RESUMO

The rules of engagement between zinc finger transcription factors and DNA have been partly defined by in vitro DNA-binding and structural studies, but less is known about how these rules apply in vivo. Here, we demonstrate how a missense mutation in the second zinc finger of Krüppel-like factor-1 (KLF1) leads to degenerate DNA-binding specificity in vivo, resulting in ectopic transcription and anemia in the Nan mouse model. We employed ChIP-seq and 4sU-RNA-seq to identify aberrant DNA-binding events genome wide and ectopic transcriptional consequences of this binding. We confirmed novel sequence specificity of the mutant recombinant zinc finger domain by performing biophysical measurements of in vitro DNA-binding affinity. Together, these results shed new light on the mechanisms by which missense mutations in DNA-binding domains of transcription factors can lead to autosomal dominant diseases.


Assuntos
DNA/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Transcriptoma/genética , Dedos de Zinco/genética , Animais , Linhagem Celular , Sobrevivência Celular/genética , Células Eritroides/metabolismo , Eritropoese/genética , Humanos , Fatores de Transcrição Kruppel-Like/química , Camundongos , Modelos Genéticos , Modelos Moleculares , Proteínas Mutantes/química , Mutação de Sentido Incorreto , Ligação Proteica
8.
Blood ; 128(12): 1631-41, 2016 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-27480112

RESUMO

The mechanisms regulating the sequential steps of terminal erythroid differentiation remain largely undefined, yet are relevant to human anemias that are characterized by ineffective red cell production. Erythroid Krüppel-like Factor (EKLF/KLF1) is a master transcriptional regulator of erythropoiesis that is mutated in a subset of these anemias. Although EKLF's function during early erythropoiesis is well studied, its role during terminal differentiation has been difficult to functionally investigate due to the impaired expression of relevant cell surface markers in Eklf(-/-) erythroid cells. We have circumvented this problem by an innovative use of imaging flow cytometry to investigate the role of EKLF in vivo and have performed functional studies using an ex vivo culture system that enriches for terminally differentiating cells. We precisely define a previously undescribed block during late terminal differentiation at the orthochromatic erythroblast stage for Eklf(-/-) cells that proceed beyond the initial stall at the progenitor stage. These cells efficiently decrease cell size, condense their nucleus, and undergo nuclear polarization; however, they display a near absence of enucleation. These late-stage Eklf(-/-) cells continue to cycle due to low-level expression of p18 and p27, a new direct target of EKLF. Surprisingly, both cell cycle and enucleation deficits are rescued by epistatic reintroduction of either of these 2 EKLF target cell cycle inhibitors. We conclude that the cell cycle as regulated by EKLF during late stages of differentiation is inherently critical for enucleation of erythroid precursors, thereby demonstrating a direct functional relationship between cell cycle exit and nuclear expulsion.


Assuntos
Núcleo Celular/metabolismo , Embrião de Mamíferos/metabolismo , Eritroblastos/metabolismo , Fatores de Transcrição Kruppel-Like/fisiologia , Animais , Sítios de Ligação , Pontos de Checagem do Ciclo Celular , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Eritroblastos/citologia , Eritropoese/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Blood ; 127(15): 1856-62, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-26903544

RESUMO

Until recently our approach to analyzing human genetic diseases has been to accurately phenotype patients and sequence the genes known to be associated with those phenotypes; for example, in thalassemia, the globin loci are analyzed. Sequencing has become increasingly accessible, and thus a larger panel of genes can be analyzed and whole exome and/or whole genome sequencing can be used when no variants are found in the candidate genes. By using such approaches in patients with unexplained anemias, we have discovered that a broad range of hitherto unrelated human red cell disorders are caused by variants in KLF1, a master regulator of erythropoiesis, which were previously considered to be extremely rare causes of human genetic disease.


Assuntos
Eritrócitos/citologia , Eritropoese/genética , Fatores de Transcrição Kruppel-Like/genética , Anemia Hemolítica/genética , Animais , Antígenos de Grupos Sanguíneos , Exoma , Deleção de Genes , Regulação da Expressão Gênica , Variação Genética , Heme/química , Hemoglobinopatias/genética , Humanos , Hidropisia Fetal/genética , Ferro/química , Camundongos , Fenótipo , Estrutura Terciária de Proteína , Piruvato Quinase/deficiência , Análise de Sequência de DNA , Globinas beta/genética
10.
PLoS Genet ; 11(10): e1005526, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26452208

RESUMO

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.


Assuntos
Eritrócitos/metabolismo , Eritropoese/genética , Fatores de Transcrição Forkhead/genética , Biologia de Sistemas , Animais , Autofagia/genética , Células da Medula Óssea/metabolismo , Polaridade Celular/genética , Eritroblastos/metabolismo , Eritrócitos/citologia , Citometria de Fluxo , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Homeostase , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo
11.
Curr Opin Hematol ; 24(3): 183-190, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28157724

RESUMO

PURPOSE OF REVIEW: Transcriptional regulators provide the molecular and biochemical basis for the cell specific properties and characteristics that follow from their central role in establishing tissue-restricted expression. Precise and sequential control of terminal cell divisions, nuclear condensation, and enucleation are defining characteristics within erythropoietic differentiation. This review is focused on KLF1, a central global regulator of this process. RECENT FINDINGS: Studies in the past year have brought a number of proteins that are targets of KLF1 regulation into focus with respect to their roles in terminal erythroid differentiation. Many of these are involved in fine control of the cell cycle at both early (E2F2, Cyclin A2) and later (p18, p27, p19) stages of differentiation, or are directly involved in enucleation (p18, p27). Dramatic biophysical changes controlled at the nuclear lamin by caspase 3 enable histone release and nuclear condensation, whereas dematin association with structural proteins alters the timing of enucleation. Conditional ablation of mDia2 has established its role in late stage cell cycle and enucleation. SUMMARY: Transcription factors such as KLF1, along with epigenetic modifiers, play crucial roles in establishing the proper onset and progression of terminal differentiation events. Studies from the past year show a remarkable multifaceted convergence on cell cycle control, and establish that the orthochromatic erythroblast stage is a critical nodal point for many of the effects on enucleation. These studies are relevant to understanding the underlying causes of anemia and hematologic disease where defective enucleation predicts a poor clinical outcome.


Assuntos
Eritropoese , Fatores de Transcrição Kruppel-Like/metabolismo , Animais , Ciclo Celular/genética , Diferenciação Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica
12.
Development ; 141(11): 2245-54, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24866116

RESUMO

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.


Assuntos
Eritrócitos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/metabolismo , Nicho de Células-Tronco , Animais , Adesão Celular , Diferenciação Celular , Membrana Celular/metabolismo , Células-Tronco Embrionárias/citologia , Eritroblastos/citologia , Células Precursoras Eritroides/citologia , Eritropoese/fisiologia , Macrófagos/citologia , Camundongos , Regiões Promotoras Genéticas , Reticulócitos/citologia , Células-Tronco/citologia , Saco Vitelino/fisiologia , Dedos de Zinco
13.
14.
Proc Natl Acad Sci U S A ; 111(37): 13337-42, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25197097

RESUMO

The binding of chromatin-associated proteins and incorporation of histone variants correlates with alterations in gene expression. These changes have been particularly well analyzed at the mammalian ß-globin locus, where transcription factors such as erythroid Krüppel-like factor (EKLF), which is also known as Krüppel-like factor 1 (KLF1), play a coordinating role in establishing the proper chromatin structure and inducing high-level expression of adult ß-globin. We had previously shown that EKLF preferentially interacts with histone H3 and that the H3.3 variant is differentially recruited to the ß-globin promoter. We now find that a novel interaction between EKLF and the histone cell cycle regulation defective homolog A (HIRA) histone chaperone accounts for these effects. HIRA is not only critical for ß-globin expression but is also required for activation of the erythropoietic regulators EKLF and GATA binding protein 1 (GATA1). Our results provide a mechanism by which transcription factor-directed recruitment of a generally expressed histone chaperone can lead to tissue-restricted changes in chromatin components, structure, and transcription at specific genomic sites during differentiation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Regulação da Expressão Gênica , Chaperonas de Histonas/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição/metabolismo , Globinas beta/genética , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/deficiência , Diferenciação Celular/genética , Células-Tronco Embrionárias/metabolismo , Células Eritroides/metabolismo , Eritropoese , Chaperonas de Histonas/química , Chaperonas de Histonas/deficiência , Fatores de Transcrição Kruppel-Like/química , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Deleção de Sequência , Fatores de Transcrição/química , Fatores de Transcrição/deficiência , Ativação Transcricional/genética , Dedos de Zinco , Globinas beta/metabolismo
15.
J Biol Chem ; 290(15): 9929-40, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25713074

RESUMO

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.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , Mutação , Proteínas Inibidoras de STAT Ativados/genética , Ativação Transcricional , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Western Blotting , Células COS , Diferenciação Celular/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , Células K562 , Fatores de Transcrição Kruppel-Like/metabolismo , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Camundongos , Microscopia Confocal , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sumoilação , Globinas beta/genética , Globinas beta/metabolismo
16.
Blood ; 124(5): 672-3, 2014 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-25082863

RESUMO

In this issue of Blood, Liu et al gain an understanding of phenotypic variability in hemoglobinopathies. They find that mutations in Krüppel-like factor-1 (KLF1) are significantly more prevalent in patients with ß-thalassemia than previously recognized and correlate with a milder phenotype. This supports the emerging concept that monoallelic KLF1 mutations can play a modulatory role in hemoglobinopathies.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , Mutação , Talassemia beta/epidemiologia , Talassemia beta/genética , Feminino , Humanos , Masculino
17.
Proc Natl Acad Sci U S A ; 108(26): 10484-9, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21670263

RESUMO

Erythroid Krüppel-like factor (EKLF) plays an important role in erythroid development by stimulating ß-globin gene expression. We have examined the details by which the minimal transactivation domain (TAD) of EKLF (EKLFTAD) interacts with several transcriptional regulatory factors. We report that EKLFTAD displays homology to the p53TAD and, like the p53TAD, can be divided into two functional subdomains (EKLFTAD1 and EKLFTAD2). Based on sequence analysis, we found that EKLFTAD2 is conserved in KLF2, KLF4, KLF5, and KLF15. In addition, we demonstrate that EKLFTAD2 binds the amino-terminal PH domain of the Tfb1/p62 subunit of TFIIH (Tfb1PH/p62PH) and four domains of CREB-binding protein/p300. The solution structure of the EKLFTAD2/Tfb1PH complex indicates that EKLFTAD2 binds Tfb1PH in an extended conformation, which is in contrast to the α-helical conformation seen for p53TAD2 in complex with Tfb1PH. These studies provide detailed mechanistic information into EKLFTAD functions as well as insights into potential interactions of the TADs of other KLF proteins. In addition, they suggest that not only have acidic TADs evolved so that they bind using different conformations on a common target, but that transitioning from a disordered to a more ordered state is not a requirement for their ability to bind multiple partners.


Assuntos
Fatores de Transcrição Kruppel-Like/química , Sequência de Aminoácidos , Sítios de Ligação , Calorimetria , Clonagem Molecular , Humanos , Células K562 , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Conformação Proteica , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/metabolismo
18.
J Biol Chem ; 287(19): 15193-204, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22393050

RESUMO

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.


Assuntos
Células Precursoras Eritroides/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Antígenos CD4/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Imunoprecipitação , Células K562 , Fatores de Transcrição Kruppel-Like/genética , Dados de Sequência Molecular , Fosfoproteínas Fosfatases/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteína Fosfatase 2C , Interferência de RNA , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Globinas beta/genética
19.
J Biol Chem ; 287(44): 36720-31, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22982397

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica , Policitemia/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores da Eritropoetina/metabolismo , Adulto , Antígenos CD/metabolismo , Sítios de Ligação , Estudos de Casos e Controles , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Análise Mutacional de DNA , Células Eritroides/metabolismo , Eritropoetina/fisiologia , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Expressão Gênica , Genes Reporter , Células-Tronco Hematopoéticas/metabolismo , Humanos , Janus Quinase 2/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Masculino , Mutação de Sentido Incorreto , Policitemia/genética , Policitemia/patologia , Regiões Promotoras Genéticas , Ligação Proteica , Receptores da Eritropoetina/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Globinas beta/genética , Globinas beta/metabolismo
20.
Blood Cells Mol Dis ; 51(2): 71-5, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23522491

RESUMO

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.


Assuntos
Anemia Diseritropoética Congênita/diagnóstico , Anemia Diseritropoética Congênita/genética , Povo Asiático/genética , Fatores de Transcrição Kruppel-Like/genética , Mutação , Anemia Diseritropoética Congênita/terapia , Medula Óssea/patologia , Criança , Análise Mutacional de DNA , Membrana Eritrocítica/metabolismo , Humanos , Masculino , Fragilidade Osmótica/genética , Taiwan
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa