Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 336, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013432

RESUMO

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


Assuntos
Epigênese Genética , Epigenoma , Epigenômica , Eritroblastos/metabolismo , Haploinsuficiência , Hemoglobinopatias/genética , Fatores de Transcrição Kruppel-Like/genética , Células Cultivadas , Sequenciamento de Cromatina por Imunoprecipitação , Eritroblastos/patologia , Eritropoese/genética , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Predisposição Genética para Doença , Hemoglobinopatias/sangue , Hemoglobinopatias/diagnóstico , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Malta , Penetrância , Fenótipo , Cultura Primária de Células , RNA-Seq
2.
Blood Adv ; 5(9): 2339-2349, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33938942

RESUMO

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


Assuntos
Haploinsuficiência , Proteínas Nucleares , Adulto , Proteínas de Transporte/genética , Humanos , Proteínas Nucleares/genética , Fenótipo , Proteínas Repressoras/genética
3.
Haematologica ; 106(2): 464-473, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32467144

RESUMO

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


Assuntos
Anemia Diseritropoética Congênita , Fatores de Transcrição Kruppel-Like , Adulto , Animais , Diferenciação Celular , Eritropoese/genética , Hemoglobinas , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos
4.
J Leukoc Biol ; 2020 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-33289106

RESUMO

Dendritic cells (DCs) are key immune modulators and are able to mount immune responses or tolerance. DC differentiation and activation imply a plethora of molecular and cellular responses, including transcriptional changes. PU.1 is a highly expressed transcription factor in DCs and coordinates relevant aspects of DC biology. Due to their role as immune regulators, DCs pose as a promising immunotherapy tool. However, some of their functional features, such as survival, activation, or migration, are compromised due to the limitations to simulate in vitro the physiologic DC differentiation process. A better knowledge of transcriptional programs would allow the identification of potential targets for manipulation with the aim of obtaining "qualified" DCs for immunotherapy purposes. Most of the current knowledge regarding DC biology derives from studies using mouse models, which not always find a parallel in human. In the present study, we dissect the PU.1 transcriptional regulome and interactome in mouse and human DCs, in the steady state or LPS activated. The PU.1 transcriptional regulome was identified by performing PU.1 chromatin immunoprecipitation followed by high-throughput sequencing and pairing these data with RNAsequencing data. The PU.1 interactome was identified by performing PU.1 immunoprecipitation followed by mass spectrometry analysis. Our results portray PU.1 as a pivotal factor that plays an important role in the regulation of genes required for proper DC activation and function, and assures the repression of nonlineage genes. The interspecies differences between human and mouse DCs are surprisingly substantial, highlighting the need to study the biology of human DCs.

5.
Hum Genomics ; 14(1): 39, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-33066815

RESUMO

The expression of the human ß-like globin genes follows a well-orchestrated developmental pattern, undergoing two essential switches, the first one during the first weeks of gestation (ε to γ), and the second one during the perinatal period (γ to ß). The γ- to ß-globin gene switching mechanism includes suppression of fetal (γ-globin, HbF) and activation of adult (ß-globin, HbA) globin gene transcription. In hereditary persistence of fetal hemoglobin (HPFH), the γ-globin suppression mechanism is impaired leaving these individuals with unusual elevated levels of fetal hemoglobin (HbF) in adulthood. Recently, the transcription factors KLF1 and BCL11A have been established as master regulators of the γ- to ß-globin switch. Previously, a genomic variant in the KLF1 gene, identified by linkage analysis performed on twenty-seven members of a Maltese family, was found to be associated with HPFH. However, variation in the levels of HbF among family members, and those from other reported families carrying genetic variants in KLF1, suggests additional contributors to globin switching. ASF1B was downregulated in the family members with HPFH. Here, we investigate the role of ASF1B in γ- to ß-globin switching and erythropoiesis in vivo. Mouse-human interspecies ASF1B protein identity is 91.6%. By means of knockdown functional assays in human primary erythroid cultures and analysis of the erythroid lineage in Asf1b knockout mice, we provide evidence that ASF1B is a novel contributor to steady-state erythroid differentiation, and while its loss affects the balance of globin expression, it has no major role in hemoglobin switching.


Assuntos
Proteínas de Ciclo Celular/genética , Eritropoese/genética , Chaperonas de Histonas/genética , Globinas beta/genética , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Células HEK293 , Chaperonas de Histonas/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos Knockout , Polimorfismo de Nucleotídeo Único , Interferência de RNA , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , gama-Globinas/genética
6.
Blood ; 136(3): 269-278, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32396940

RESUMO

The oxygen transport function of hemoglobin (HB) is thought to have arisen ∼500 million years ago, roughly coinciding with the divergence between jawless (Agnatha) and jawed (Gnathostomata) vertebrates. Intriguingly, extant HBs of jawless and jawed vertebrates were shown to have evolved twice, and independently, from different ancestral globin proteins. This raises the question of whether erythroid-specific expression of HB also evolved twice independently. In all jawed vertebrates studied to date, one of the HB gene clusters is linked to the widely expressed NPRL3 gene. Here we show that the nprl3-linked hb locus of a jawless vertebrate, the river lamprey (Lampetra fluviatilis), shares a range of structural and functional properties with the equivalent jawed vertebrate HB locus. Functional analysis demonstrates that an erythroid-specific enhancer is located in intron 7 of lamprey nprl3, which corresponds to the NPRL3 intron 7 MCS-R1 enhancer of jawed vertebrates. Collectively, our findings signify the presence of an nprl3-linked multiglobin gene locus, which contains a remote enhancer that drives globin expression in erythroid cells, before the divergence of jawless and jawed vertebrates. Different globin genes from this ancestral cluster evolved in the current NPRL3-linked HB genes in jawless and jawed vertebrates. This provides an explanation of the enigma of how, in different species, globin genes linked to the same adjacent gene could undergo convergent evolution.


Assuntos
Eritrócitos/metabolismo , Evolução Molecular , Proteínas de Peixes , Regulação da Expressão Gênica/fisiologia , Hemoglobinas , Lampreias , Animais , Proteínas de Peixes/biossíntese , Proteínas de Peixes/genética , Hemoglobinas/biossíntese , Hemoglobinas/genética , Lampreias/genética , Lampreias/metabolismo , Família Multigênica
7.
Epigenetics Chromatin ; 12(1): 33, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31164147

RESUMO

BACKGROUND: Both tissue-specific and ubiquitously expressed transcription factors, such as Sp-family members, are required for correct development. However, the molecular details of how ubiquitous factors are involved in programming tissue-specific chromatin and thus participate in developmental processes are still unclear. We previously showed that embryonic stem cells lacking Sp1 DNA-binding activity (Sp1ΔDBD/ΔDBD cells) are able to differentiate into early blood progenitors despite the inability of Sp1 to bind chromatin without its DNA-binding domain. However, gene expression during differentiation becomes progressively deregulated, and terminal differentiation is severely compromised. RESULTS: Here, we studied the cooperation of Sp1 with its closest paralogue Sp3 in hematopoietic development and demonstrate that Sp1 and Sp3 binding sites largely overlap. The complete absence of either Sp1 or Sp3 or the presence of the Sp1 DNA-binding mutant has only a minor effect on the pattern of distal accessible chromatin sites and their transcription factor binding motif content, suggesting that these mutations do not affect tissue-specific chromatin programming. Sp3 cooperates with Sp1ΔDBD/ΔDBD to enable hematopoiesis, but is unable to do so in the complete absence of Sp1. Using single-cell gene expression analysis, we show that the lack of Sp1 DNA binding leads to a distortion of cell fate decision timing, indicating that stable chromatin binding of Sp1 is required to maintain robust differentiation trajectories. CONCLUSIONS: Our findings highlight the essential contribution of ubiquitous factors such as Sp1 to blood cell development. In contrast to tissue-specific transcription factors which are required to direct specific cell fates, loss of Sp1 leads to a widespread deregulation in timing and coordination of differentiation trajectories during hematopoietic specification.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular/genética , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Transcrição Gênica
8.
PLoS One ; 14(3): e0208659, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30921348

RESUMO

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


Assuntos
Anemia Hemolítica/genética , Células Eritroides/citologia , Fatores de Transcrição Kruppel-Like/genética , Proteína ran de Ligação ao GTP/genética , Substituição de Aminoácidos , Animais , Diferenciação Celular , Células Cultivadas , Cromatina/metabolismo , Modelos Animais de Doenças , Embrião não Mamífero/metabolismo , Células Eritroides/metabolismo , Eritropoese , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Análise de Sequência de RNA/métodos , Proteína ran de Ligação ao GTP/metabolismo
10.
Am J Blood Res ; 4(1): 27-32, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25232502

RESUMO

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

11.
Cell Rep ; 4(3): 589-600, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23911284

RESUMO

Here, we show that transcription factors bound to regulatory sequences can be identified by purifying these unique sequences directly from mammalian cells in vivo. Using targeted chromatin purification (TChP), a double-pull-down strategy with a tetracycline-sensitive "hook" bound to a specific promoter, we identify transcription factors bound to the repressed γ-globin gene-associated regulatory regions. After validation of the binding, we show that, in human primary erythroid cells, knockdown of a number of these transcription factors induces γ-globin gene expression. Reactivation of γ-globin gene expression ameliorates the symptoms of ß-thalassemia and sickle cell disease, and these factors provide potential targets for the development of therapeutics for treating these patients.


Assuntos
Cromatina/isolamento & purificação , Técnicas de Silenciamento de Genes/métodos , Animais , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Humanos , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Proteômica/métodos , Transcrição Gênica , Globinas beta/genética , Globinas beta/isolamento & purificação , Globinas beta/metabolismo
12.
Blood ; 121(13): 2553-62, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23361909

RESUMO

B-cell lymphoma 11A (BCL11A) downregulation in human primary adult erythroid progenitors results in elevated expression of fetal γ-globin. Recent reports showed that BCL11A expression is activated by KLF1, leading to γ-globin repression. To study regulation of erythropoiesis and globin expression by KLF1 and BCL11A in an in vivo model, we used mice carrying a human ß-globin locus transgene with combinations of Klf1 knockout, Bcl11a floxed, and EpoR(Cre) knockin alleles. We found a higher percentage of reticulocytes in adult Klf1(wt/ko) mice and a mild compensated anemia in Bcl11a(cko/cko) mice. These phenotypes were more pronounced in compound Klf1(wt/ko)::Bcl11a(cko/cko) mice. Analysis of Klf1(wt/ko), Bcl11a(cko/cko), and Klf1(wt/ko)::Bcl11a(cko/cko) mutant embryos demonstrated increased expression of mouse embryonic globins during fetal development. Expression of human γ-globin remained high in Bcl11a(cko/cko) embryos during fetal development, and this was further augmented in Klf1(wt/ko)::Bcl11a(cko/cko) embryos. After birth, expression of human γ-globin and mouse embryonic globins decreased in Bcl11a(cko/cko) and Klf1(wt/ko)::Bcl11a(cko/cko) mice, but the levels remained much higher than those observed in control animals. Collectively, our data support an important role for the KLF1-BCL11A axis in erythroid maturation and developmental regulation of globin expression.


Assuntos
Proteínas de Transporte/genética , Eritropoese/genética , Genes de Troca/genética , Globinas/genética , Fatores de Transcrição Kruppel-Like/genética , Proteínas Nucleares/genética , Animais , Proteínas de Ligação a DNA , Embrião de Mamíferos , Eritropoese/fisiologia , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Rearranjo Gênico/genética , Rearranjo Gênico/fisiologia , Genes de Troca/fisiologia , Humanos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Repressoras , Reticulocitose/genética , Reticulocitose/fisiologia , Baço/citologia , Baço/embriologia , Baço/metabolismo
13.
Mol Cell Proteomics ; 11(11): 1263-73, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22872859

RESUMO

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


Assuntos
Arginina/metabolismo , Cromatina/metabolismo , Proteínas Correpressoras/metabolismo , Cisteína Endopeptidases/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Sumoilação , Fatores de Transcrição/metabolismo , Animais , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Humanos , Metilação , Camundongos , Modelos Biológicos , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Estabilidade Proteica
14.
PLoS Genet ; 6(11): e1001203, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21085687

RESUMO

SUMO modification of transcription factors is linked to repression of transcription. The physiological significance of SUMO attachment to a particular transcriptional regulator, however, is largely unknown. We have employed the ubiquitously expressed murine transcription factor Sp3 to analyze the role of SUMOylation in vivo. We generated mice and mouse embryonic fibroblasts (MEFs) carrying a subtle point mutation in the SUMO attachment sequence of Sp3 (IKEE(553)D mutation). The E(553)D mutation impedes SUMOylation of Sp3 at K(551)in vivo, without affecting Sp3 protein levels. Expression profiling revealed that spermatocyte-specific genes, such as Dmc1 and Dnahc8, and neuronal genes, including Paqr6, Rims3, and Robo3, are de-repressed in non-testicular and extra-neuronal mouse tissues and in mouse embryonic fibroblasts expressing the SUMOylation-deficient Sp3E(553)D mutant protein. Chromatin immunoprecipitation experiments show that transcriptional de-repression of these genes is accompanied by the loss of repressive heterochromatic marks such as H3K9 and H4K20 tri-methylation and impaired recruitment of repressive chromatin-modifying enzymes. Finally, analysis of the DNA methylation state of the Dmc1, Paqr6, and Rims3 promoters by bisulfite sequencing revealed that these genes are highly methylated in Sp3wt MEFs but are unmethylated in Sp3E(553)D MEFs linking SUMOylation of Sp3 to tissue-specific CpG methylation. Our results establish SUMO conjugation to Sp3 as a molecular beacon for the assembly of repression machineries to maintain tissue-specific transcriptional gene silencing.


Assuntos
Inativação Gênica , Neurônios/metabolismo , Fator de Transcrição Sp3/genética , Espermatócitos/metabolismo , Sumoilação/genética , Animais , Cromatina/metabolismo , Metilação de DNA/genética , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Marcação de Genes , Masculino , Meiose/genética , Camundongos , Camundongos Mutantes , Neurônios/citologia , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fator de Transcrição Sp3/metabolismo
15.
Nat Genet ; 42(9): 801-5, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20676099

RESUMO

Hereditary persistence of fetal hemoglobin (HPFH) is characterized by persistent high levels of fetal hemoglobin (HbF) in adults. Several contributory factors, both genetic and environmental, have been identified but others remain elusive. HPFH was found in 10 of 27 members from a Maltese family. We used a genome-wide SNP scan followed by linkage analysis to identify a candidate region on chromosome 19p13.12-13. Sequencing revealed a nonsense mutation in the KLF1 gene, p.K288X, which ablated the DNA-binding domain of this key erythroid transcriptional regulator. Only family members with HPFH were heterozygous carriers of this mutation. Expression profiling on primary erythroid progenitors showed that KLF1 target genes were downregulated in samples from individuals with HPFH. Functional assays suggested that, in addition to its established role in regulating adult globin expression, KLF1 is a key activator of the BCL11A gene, which encodes a suppressor of HbF expression. These observations provide a rationale for the effects of KLF1 haploinsufficiency on HbF levels.


Assuntos
Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Adulto , Células Cultivadas , Eritropoese/genética , Família , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Humanos , Perda de Heterozigosidade/fisiologia , Masculino , Linhagem , Fatores de Transcrição/genética , Transfecção
16.
Blood ; 116(20): 4349-52, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-20688955

RESUMO

An estimated 6% to 7% of the earth's population carries a mutation affecting red blood cell function. The ß-thalassemias and sickle cell disease are the most common monogenic disorders caused by these mutations. Increased levels of γ-globin ameliorate the severity of these diseases because fetal hemoglobin (HbF; α2γ2) can effectively replace adult hemoglobin (HbA; α2ß2) and counteract polymerization of sickle hemoglobin (HbS; α2ß(S)2). Therefore, understanding the molecular mechanism of globin switching is of biologic and clinical importance. Here, we show that the recently identified chromatin factor Friend of Prmt1 (FOP) is a critical modulator of γ-globin gene expression. Knockdown of FOP in adult erythroid progenitors strongly induces HbF. Importantly, γ-globin expression can be elevated in cells from ß-thalassemic patients by reducing FOP levels. These observations identify FOP as a novel therapeutic target in ß-hemoglobinopathies.


Assuntos
Hemoglobina Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Hemoglobina Fetal/metabolismo , Humanos , Camundongos , Proteínas Nucleares/genética , Fatores de Transcrição/genética
17.
PLoS One ; 5(3): e9587, 2010 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-20221402

RESUMO

BACKGROUND: The zinc finger protein Sp2 (specificity protein 2) is a member of the glutamine-rich Sp family of transcription factors. Despite its close similarity to Sp1, Sp3 and Sp4, Sp2 does not bind to DNA or activate transcription when expressed in mammalian cell lines. The expression pattern and the biological relevance of Sp2 in the mouse are unknown. METHODOLOGY/PRINCIPAL FINDINGS: Whole-mount in situ hybridization of mouse embryos between E7.5 and E9.5 revealed abundant expression in most embryonic and extra-embryonic tissues. In order to unravel the biological relevance of Sp2, we have targeted the Sp2 gene by a tri-loxP strategy. Constitutive Sp2null and conditional Sp2cko knockout alleles were obtained by crossings with appropriate Cre recombinase expressing mice. Constitutive disruption of the mouse Sp2 gene (Sp2null) resulted in severe growth retardation and lethality before E9.5. Mouse embryonic fibroblasts (MEFs) derived from Sp2null embryos at E9.5 failed to grow. Cre-mediated ablation of Sp2 in Sp2cko/cko MEFs obtained from E13.5 strongly impaired cell proliferation. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that Sp2 is essential for early mouse development and autonomous proliferation of MEFs in culture. Comparison of the Sp2 knockout phenotype with the phenotypes of Sp1, Sp3 and Sp4 knockout strains shows that, despite their structural similarity and evolutionary relationship, all four glutamine-rich members of the Sp family of transcription factors have distinct non-redundant functions in vivo.


Assuntos
Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator de Transcrição Sp2/fisiologia , Animais , Proliferação de Células , DNA/metabolismo , Evolução Molecular , Feminino , Hibridização In Situ , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Fatores de Tempo , Transcrição Gênica
18.
Mol Cell Biol ; 30(1): 260-72, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19858291

RESUMO

We describe the isolation and characterization of Friend of Prmt1 (Fop), a novel chromatin target of protein arginine methyltransferases. Human Fop is encoded by C1orf77, a gene of previously unknown function. We show that Fop is tightly associated with chromatin, and that it is modified by both asymmetric and symmetric arginine methylation in vivo. Furthermore, Fop plays an important role in the ligand-dependent activation of estrogen receptor target genes, including TFF1 (pS2). Fop depletion results in an almost complete block of estradiol-induced promoter occupancy by the estrogen receptor. Our data indicate that Fop recruitment to the promoter is an early critical event in the activation of estradiol-dependent transcription.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Arginina/metabolismo , Biotinilação , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Estradiol/farmacologia , Humanos , Metilação , Camundongos , Proteínas Nucleares/genética , Mapeamento de Interação de Proteínas , Proteínas Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/genética , Proteômica , Proteínas Repressoras/metabolismo , Especificidade por Substrato , Fatores de Transcrição , Ativação Transcricional
19.
Blood ; 109(12): 5481-90, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17327407

RESUMO

Three Gata transcription factors (Gata1, -2, and -3) are essential for hematopoiesis. These factors are thought to play distinct roles because they do not functionally replace each other. For instance, Gata2 messenger RNA (mRNA) expression is highly elevated in Gata1-null erythroid cells, yet this does not rescue the defect. Here, we test whether Gata2 and -3 transgenes rescue the erythroid defect of Gata1-null mice, if expressed in the appropriate spatiotemporal pattern. Gata1, -2, and -3 transgenes driven by beta-globin regulatory elements, directing expression to late stages of differentiation, fail to rescue erythropoiesis in Gata1-null mutants. In contrast, when controlled by Gata1 regulatory elements, directing expression to the early stages of differentiation, Gata1, -2, and -3 do rescue the Gata1-null phenotype. The dramatic increase of endogenous Gata2 mRNA in Gata1-null progenitors is not reflected in Gata2 protein levels, invoking translational regulation. Our data show that the dynamic spatiotemporal regulation of Gata factor levels is more important than their identity and provide a paradigm for developmental control mechanisms that are hard-wired in cis-regulatory elements.


Assuntos
Eritropoese , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica/fisiologia , Animais , Fatores de Transcrição GATA/análise , Fatores de Transcrição GATA/deficiência , Fator de Transcrição GATA1/deficiência , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA3/genética , Camundongos , Camundongos Knockout , Biossíntese de Proteínas , RNA Mensageiro/análise , Sequências Reguladoras de Ácido Nucleico/fisiologia , Transgenes
20.
Genes Dev ; 18(20): 2485-90, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15489291

RESUMO

Three-dimensional organization of a gene locus is important for its regulation, as recently demonstrated for the beta-globin locus. When actively expressed, the cis-regulatory elements of the beta-globin locus are in proximity in the nuclear space, forming a compartment termed the Active Chromatin Hub (ACH). However, it is unknown which proteins are involved in ACH formation. Here, we show that EKLF, an erythroid transcription factor required for adult beta-globin gene transcription, is also required for ACH formation. We conclude that transcription factors can play an essential role in the three-dimensional organization of gene loci.


Assuntos
Cromatina/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Globinas/genética , Fatores de Transcrição/genética , Animais , Reagentes de Ligações Cruzadas , DNA/metabolismo , Primers do DNA , DNA Complementar/genética , Proteínas de Ligação a DNA/metabolismo , Formaldeído , Globinas/química , Globinas/metabolismo , Fatores de Transcrição Kruppel-Like , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase/métodos , Mapeamento por Restrição , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...