Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 11(6): 1462-1478, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30503262

RESUMO

The production of blood cells and their precursors from human pluripotent stem cells (hPSCs) in vitro has the potential to make a significant impact upon healthcare provision. We demonstrate that the forward programming of hPSCs through overexpression of GATA1, FLI1, and TAL1 leads to the production of a population of progenitors that can differentiate into megakaryocyte or erythroblasts. Using "rainbow" lentiviral vectors to quantify individual transgene expression in single cells, we demonstrate that the cell fate decision toward an erythroblast or megakaryocyte is dictated by the level of FLI1 expression and is independent of culture conditions. Early FLI1 expression is critical to confer proliferative potential to programmed cells while its subsequent silencing or maintenance dictates an erythroid or megakaryocytic fate, respectively. These committed progenitors subsequently expand and mature into megakaryocytes or erythroblasts in response to thrombopoietin or erythropoietin. Our results reveal molecular mechanisms underlying hPSC forward programming and novel opportunities for application to transfusion medicine.


Assuntos
Linhagem da Célula , Células Eritroides/citologia , Fator de Transcrição GATA1/metabolismo , Megacariócitos/citologia , Células-Tronco Pluripotentes/citologia , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Citocinas/farmacologia , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Eritropoetina/farmacologia , Inativação Gênica , Humanos , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Trombopoetina/farmacologia , Transgenes
3.
J Clin Invest ; 127(3): 814-829, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28134622

RESUMO

Platelets are anuclear cells that are essential for blood clotting. They are produced by large polyploid precursor cells called megakaryocytes. Previous genome-wide association studies in nearly 70,000 individuals indicated that single nucleotide variants (SNVs) in the gene encoding the actin cytoskeletal regulator tropomyosin 4 (TPM4) exert an effect on the count and volume of platelets. Platelet number and volume are independent risk factors for heart attack and stroke. Here, we have identified 2 unrelated families in the BRIDGE Bleeding and Platelet Disorders (BPD) collection who carry a TPM4 variant that causes truncation of the TPM4 protein and segregates with macrothrombocytopenia, a disorder characterized by low platelet count. N-Ethyl-N-nitrosourea-induced (ENU-induced) missense mutations in Tpm4 or targeted inactivation of the Tpm4 locus led to gene dosage-dependent macrothrombocytopenia in mice. All other blood cell counts in Tpm4-deficient mice were normal. Insufficient TPM4 expression in human and mouse megakaryocytes resulted in a defect in the terminal stages of platelet production and had a mild effect on platelet function. Together, our findings demonstrate a nonredundant role for TPM4 in platelet biogenesis in humans and mice and reveal that truncating variants in TPM4 cause a previously undescribed dominant Mendelian platelet disorder.


Assuntos
Plaquetas/metabolismo , Genes Dominantes , Doenças Genéticas Inatas , Mutação de Sentido Incorreto , Trombocitopenia , Tropomiosina , Animais , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Trombocitopenia/genética , Trombocitopenia/metabolismo , Tropomiosina/genética , Tropomiosina/metabolismo
4.
Haematologica ; 102(4): 695-706, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28082341

RESUMO

Gray platelet syndrome is named after the gray appearance of platelets due to the absence of α-granules. It is caused by recessive mutations in NBEAL2, resulting in macrothrombocytopenia and myelofibrosis. Though using the term gray platelets for GATA1 deficiency has been debated, a reduced number of α-granules has been described for macrothrombocytopenia due to GATA1 mutations. We compared platelet size and number of α-granules for two NBEAL2 and two GATA1-deficient patients and found reduced numbers of α-granules for all, with the defect being more pronounced for NBEAL2 deficiency. We further hypothesized that the granule defect for GATA1 is due to a defective control of NBEAL2 expression. Remarkably, platelets from two patients, and Gata1-deficient mice, expressed almost no NBEAL2. The differentiation of GATA1 patient-derived CD34+ stem cells to megakaryocytes showed defective proplatelet and α-granule formation with strongly reduced NBEAL2 protein and ribonucleic acid expression. Chromatin immunoprecipitation sequencing revealed 5 GATA binding sites in a regulatory region 31 kb upstream of NBEAL2 covered by a H3K4Me1 mark indicative of an enhancer locus. Luciferase reporter constructs containing this region confirmed its enhancer activity in K562 cells, and mutagenesis of the GATA1 binding sites resulted in significantly reduced enhancer activity. Moreover, DNA binding studies showed that GATA1 and GATA2 physically interact with this enhancer region. GATA1 depletion using small interfering ribonucleic acid in K562 cells also resulted in reduced NBEAL2 expression. In conclusion, we herein show a long-distance regulatory region with GATA1 binding sites as being a strong enhancer for NBEAL2 expression.


Assuntos
Proteínas Sanguíneas/genética , Elementos Facilitadores Genéticos , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Alelos , Plaquetas/metabolismo , Diferenciação Celular/genética , Fator de Transcrição GATA1/deficiência , Fator de Transcrição GATA1/genética , Expressão Gênica , Genes Recessivos , Genes Reporter , Genes Ligados ao Cromossomo X , Estudos de Associação Genética , Predisposição Genética para Doença , Homozigoto , Humanos , Megacariócitos/citologia , Megacariócitos/metabolismo , Megacariócitos/ultraestrutura , Mutação , Fenótipo , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética , Trombocitopenia/sangue , Trombocitopenia/genética , Trombocitopenia/patologia
5.
Nat Commun ; 7: 11208, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27052461

RESUMO

The production of megakaryocytes (MKs)--the precursors of blood platelets--from human pluripotent stem cells (hPSCs) offers exciting clinical opportunities for transfusion medicine. Here we describe an original approach for the large-scale generation of MKs in chemically defined conditions using a forward programming strategy relying on the concurrent exogenous expression of three transcription factors: GATA1, FLI1 and TAL1. The forward programmed MKs proliferate and differentiate in culture for several months with MK purity over 90% reaching up to 2 × 10(5) mature MKs per input hPSC. Functional platelets are generated throughout the culture allowing the prospective collection of several transfusion units from as few as 1 million starting hPSCs. The high cell purity and yield achieved by MK forward programming, combined with efficient cryopreservation and good manufacturing practice (GMP)-compatible culture, make this approach eminently suitable to both in vitro production of platelets for transfusion and basic research in MK and platelet biology.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Reprogramação Celular , Fator de Transcrição GATA1/genética , Megacariócitos/citologia , Células-Tronco Pluripotentes/citologia , Proteína Proto-Oncogênica c-fli-1/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Plaquetas/citologia , Plaquetas/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Proliferação de Células , Criopreservação/métodos , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lentivirus/genética , Megacariócitos/metabolismo , Análise em Microsséries , Células-Tronco Pluripotentes/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Transdução Genética , Transgenes
6.
Blood ; 125(8): 1292-301, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25533034

RESUMO

Children with Down syndrome (DS) are at increased risk for acute myeloid leukemias (ML-DS) characterized by mixed megakaryocytic and erythroid phenotype and by acquired mutations in the GATA1 gene resulting in a short GATA1s isoform. The chromosome 21 microRNA (miR)-125b cluster has been previously shown to cooperate with GATA1s in transformation of fetal hematopoietic progenitors. In this study, we report that the expression of miR-486-5p is increased in ML-DS compared with non-DS acute megakaryocytic leukemias (AMKLs). miR-486-5p is regulated by GATA1 and GATA1s that bind to the promoter of its host gene ANK1. miR-486-5p is highly expressed in mouse erythroid precursors and knockdown (KD) in ML-DS cells reduced their erythroid phenotype. Ectopic expression and KD of miR-486-5p in primary fetal liver hematopoietic progenitors demonstrated that miR-486-5p cooperates with Gata1s to enhance their self renewal. Consistent with its activation of AKT, overexpression and KD experiments showed its importance for growth and survival of human leukemic cells. Thus, miR-486-5p cooperates with GATA1s in supporting the growth and survival, and the aberrant erythroid phenotype of the megakaryocytic leukemias of DS.


Assuntos
Síndrome de Down/genética , Eritropoese/genética , Leucemia Mieloide Aguda/genética , MicroRNAs/fisiologia , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Pré-Escolar , Síndrome de Down/complicações , Síndrome de Down/fisiopatologia , Células Eritroides/metabolismo , Células HEK293 , Humanos , Células K562 , Leucemia Mieloide Aguda/patologia , Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/genética , Células Tumorais Cultivadas
7.
Development ; 141(20): 4018-30, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25252941

RESUMO

Transcription factors (TFs) act within wider regulatory networks to control cell identity and fate. Numerous TFs, including Scl (Tal1) and PU.1 (Spi1), are known regulators of developmental and adult haematopoiesis, but how they act within wider TF networks is still poorly understood. Transcription activator-like effectors (TALEs) are a novel class of genetic tool based on the modular DNA-binding domains of Xanthomonas TAL proteins, which enable DNA sequence-specific targeting and the manipulation of endogenous gene expression. Here, we report TALEs engineered to target the PU.1-14kb and Scl+40kb transcriptional enhancers as efficient new tools to perturb the expression of these key haematopoietic TFs. We confirmed the efficiency of these TALEs at the single-cell level using high-throughput RT-qPCR, which also allowed us to assess the consequences of both PU.1 activation and repression on wider TF networks during developmental haematopoiesis. Combined with comprehensive cellular assays, these experiments uncovered novel roles for PU.1 during early haematopoietic specification. Finally, transgenic mouse studies confirmed that the PU.1-14kb element is active at sites of definitive haematopoiesis in vivo and PU.1 is detectable in haemogenic endothelium and early committing blood cells. We therefore establish TALEs as powerful new tools to study the functionality of transcriptional networks that control developmental processes such as early haematopoiesis.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Animais , Diferenciação Celular , Técnicas de Cocultura , Células Endoteliais/citologia , Células-Tronco Hematopoéticas , Humanos , Células K562 , Camundongos , Camundongos Transgênicos , Fenótipo , Análise de Célula Única , Fatores de Transcrição/metabolismo , Transgenes , Xanthomonas/metabolismo
8.
Biol Open ; 2(11): 1229-38, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24244860

RESUMO

Comprehensive analysis of cis-regulatory elements is key to understanding the dynamic gene regulatory networks that control embryonic development. While transgenic animals represent the gold standard assay, their generation is costly, entails significant animal usage, and in utero development complicates time-course studies. As an alternative, embryonic stem (ES) cells can readily be differentiated in a process that correlates well with developing embryos. Here, we describe a highly effective platform for enhancer assays using an Hsp68/Venus reporter cassette that targets to the Hprt locus in mouse ES cells. This platform combines the flexibility of Gateway® cloning, live cell trackability of a fluorescent reporter, low background and the advantages of single copy insertion into a defined genomic locus. We demonstrate the successful recapitulation of tissue-specific enhancer activity for two cardiac and two haematopoietic enhancers. In addition, we used this assay to dissect the functionality of the highly conserved Ets/Ets/Gata motif in the Scl+19 enhancer, which revealed that the Gata motif is not required for initiation of enhancer activity. We further confirmed that Gata2 is not required for endothelial activity of the Scl+19 enhancer using Gata2(-/-) Scl+19 transgenic embryos. We have therefore established a valuable toolbox to study gene regulatory networks with broad applicability.

9.
Blood ; 122(15): 2694-703, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23974202

RESUMO

The ETS transcription factor ERG plays a central role in definitive hematopoiesis, and its overexpression in acute myeloid leukemia (AML) is associated with a stem cell signature and poor prognosis. Yet how ERG causes leukemia is unclear. Here we show that pan-hematopoietic ERG expression induces an early progenitor myeloid leukemia in transgenic mice. Integrated genome-scale analysis of gene expression and ERG binding profiles revealed that ERG activates a transcriptional program similar to human AML stem/progenitor cells and to human AML with high ERG expression. This transcriptional program was associated with activation of RAS that was required for leukemia cells growth in vitro and in vivo. We further show that ERG induces expression of the Pim1 kinase oncogene through a novel hematopoietic enhancer validated in transgenic mice and human CD34(+) normal and leukemic cells. Pim1 inhibition disrupts growth and induces apoptosis of ERG-expressing leukemic cells. The importance of the ERG/PIM1 axis is further underscored by the poorer prognosis of AML highly expressing ERG and PIM1. Thus, integrative genomic analysis demonstrates that ERG causes myeloid progenitor leukemia characterized by an induction of leukemia stem cell transcriptional programs. Pim1 and the RAS pathway are potential therapeutic targets of these high-risk leukemias.


Assuntos
Regulação Leucêmica da Expressão Gênica/fisiologia , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Transativadores/genética , Fatores de Transcrição/metabolismo , Animais , Antineoplásicos , Elementos Facilitadores Genéticos/genética , Genômica , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Células Progenitoras Mieloides/fisiologia , Transplante de Neoplasias , Transcrição Gênica/fisiologia , Regulador Transcricional ERG
10.
Blood ; 121(1): 188-96, 2013 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-23160460

RESUMO

Wnt signaling is involved in numerous aspects of vertebrate development and homeostasis, including the formation and function of blood cells. Here, we show that canonical and noncanonical Wnt signaling pathways are present and functional in megakaryocytes (MKs), with several Wnt effectors displaying MK-restricted expression. Using the CHRF288-11 cell line as a model for human MKs, the canonical Wnt3a signal was found to induce a time and dose-dependent increase in ß-catenin expression. ß-catenin accumulation was inhibited by the canonical antagonist dickkopf-1 (DKK1) and by the noncanonical agonist Wnt5a. Whole genome expression analysis demonstrated that Wnt3a and Wnt5a regulated distinct patterns of gene expression in MKs, and revealed a further interplay between canonical and noncanonical Wnt pathways. Fetal liver cells derived from low-density-lipoprotein receptor-related protein 6-deficient mice (LRP6(-/-)), generated dramatically reduced numbers of MKs in culture of lower ploidy (2N and 4N) than wild-type controls, implicating LRP6-dependent Wnt signaling in MK proliferation and maturation. Finally, in wild-type mature murine fetal liver-derived MKs, Wnt3a potently induced proplatelet formation, an effect that could be completely abrogated by DKK1. These data identify novel extrinsic regulators of proplatelet formation, and reveal a profound role for Wnt signaling in platelet production.


Assuntos
Megacariócitos/citologia , Trombopoese/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Plaquetas/citologia , Linhagem Celular , Células Cultivadas/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Fígado/embriologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Proteínas Recombinantes/farmacologia , Trombopoese/genética , Proteínas Wnt/farmacologia , Proteína Wnt3A/farmacologia , beta Catenina/biossíntese , beta Catenina/genética
11.
Blood ; 120(24): 4859-68, 2012 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-22972982

RESUMO

We recently identified 68 genomic loci where common sequence variants are associated with platelet count and volume. Platelets are formed in the bone marrow by megakaryocytes, which are derived from hematopoietic stem cells by a process mainly controlled by transcription factors. The homeobox transcription factor MEIS1 is uniquely transcribed in megakaryocytes and not in the other lineage-committed blood cells. By ChIP-seq, we show that 5 of the 68 loci pinpoint a MEIS1 binding event within a group of 252 MK-overexpressed genes. In one such locus in DNM3, regulating platelet volume, the MEIS1 binding site falls within a region acting as an alternative promoter that is solely used in megakaryocytes, where allelic variation dictates different levels of a shorter transcript. The importance of dynamin activity to the latter stages of thrombopoiesis was confirmed by the observation that the inhibitor Dynasore reduced murine proplatelet for-mation in vitro.


Assuntos
Plaquetas/metabolismo , Dinamina III/genética , Genoma Humano/genética , Proteínas de Homeodomínio/genética , Megacariócitos/metabolismo , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/genética , Animais , Sítios de Ligação/genética , Plaquetas/efeitos dos fármacos , Linhagem Celular Tumoral , Linhagem da Célula/genética , Células Cultivadas , Imunoprecipitação da Cromatina , Expressão Gênica , Variação Genética , Proteínas de Homeodomínio/metabolismo , Humanos , Hidrazonas/farmacologia , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/metabolismo , Contagem de Plaquetas , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Sítio de Iniciação de Transcrição , Transcrição Gênica
12.
PLoS Genet ; 7(6): e1002139, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21738486

RESUMO

Turning genetic discoveries identified in genome-wide association (GWA) studies into biological mechanisms is an important challenge in human genetics. Many GWA signals map outside exons, suggesting that the associated variants may lie within regulatory regions. We applied the formaldehyde-assisted isolation of regulatory elements (FAIRE) method in a megakaryocytic and an erythroblastoid cell line to map active regulatory elements at known loci associated with hematological quantitative traits, coronary artery disease, and myocardial infarction. We showed that the two cell types exhibit distinct patterns of open chromatin and that cell-specific open chromatin can guide the finding of functional variants. We identified an open chromatin region at chromosome 7q22.3 in megakaryocytes but not erythroblasts, which harbors the common non-coding sequence variant rs342293 known to be associated with platelet volume and function. Resequencing of this open chromatin region in 643 individuals provided strong evidence that rs342293 is the only putative causative variant in this region. We demonstrated that the C- and G-alleles differentially bind the transcription factor EVI1 affecting PIK3CG gene expression in platelets and macrophages. A protein-protein interaction network including up- and down-regulated genes in Pik3cg knockout mice indicated that PIK3CG is associated with gene pathways with an established role in platelet membrane biogenesis and thrombus formation. Thus, rs342293 is the functional common variant at this locus; to the best of our knowledge this is the first such variant to be elucidated among the known platelet quantitative trait loci (QTLs). Our data suggested a molecular mechanism by which a non-coding GWA index SNP modulates platelet phenotype.


Assuntos
Cromatina/genética , Estudo de Associação Genômica Ampla , Animais , Plaquetas/metabolismo , Cromossomos Humanos Par 7/genética , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Proteínas de Ligação a DNA/metabolismo , Eritroblastos/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Proteína do Locus do Complexo MDS1 e EVI1 , Macrófagos/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Genéticos , Fenótipo , Proto-Oncogenes , Locos de Características Quantitativas , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo
13.
Exp Hematol ; 39(10): 961-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21781948

RESUMO

One of the key challenges facing biomedical research is to extract biologically meaningful information from the ever-increasing scale and complexity of datasets generated through high-throughput approaches. Hematopoiesis represents one of the most experimentally tractable mammalian organ systems and, therefore, has historically tended to be at the forefront of applying new technologies within biomedical research. The combination of massive parallel sequencing technologies with chromatin-immunoprecipitation (ChIP-Seq) permits genome-scale characterization of histone modification status and identification of the complete set of binding sites for transcription factors. Because transcription factors have long been recognized as essential regulators of cell fate choice in hematopoiesis, ChIP-Seq technology has rapidly entered the arena of modern experimental hematology. Here we review the biological insights gained from ChIP-Seq studies performed in the hematopoietic system since the earliest studies just 4 years ago. A surprisingly large number of different approaches have already been implemented to extract new biological knowledge from ChIP-Seq datasets. By focusing on successful insights from multiple different approaches, we hope to provide stimulating reading for anyone wanting to utilize ChIP-Seq technology within their particular research field.


Assuntos
Imunoprecipitação da Cromatina/métodos , Regulação da Expressão Gênica , Hematopoese/genética , Sequenciamento de Nucleotídeos em Larga Escala , Transcrição Gênica/genética , Animais , Transformação Celular Neoplásica/genética , DNA/metabolismo , Epigênese Genética , Previsões , Células-Tronco Hematopoéticas/metabolismo , Histonas/metabolismo , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Proteínas de Fusão Oncogênica/metabolismo , Ligação Proteica , Fatores de Transcrição/metabolismo
14.
Dev Cell ; 20(5): 597-609, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21571218

RESUMO

Hematopoietic differentiation critically depends on combinations of transcriptional regulators controlling the development of individual lineages. Here, we report the genome-wide binding sites for the five key hematopoietic transcription factors--GATA1, GATA2, RUNX1, FLI1, and TAL1/SCL--in primary human megakaryocytes. Statistical analysis of the 17,263 regions bound by at least one factor demonstrated that simultaneous binding by all five factors was the most enriched pattern and often occurred near known hematopoietic regulators. Eight genes not previously appreciated to function in hematopoiesis that were bound by all five factors were shown to be essential for thrombocyte and/or erythroid development in zebrafish. Moreover, one of these genes encoding the PDZK1IP1 protein shared transcriptional enhancer elements with the blood stem cell regulator TAL1/SCL. Multifactor ChIP-Seq analysis in primary human cells coupled with a high-throughput in vivo perturbation screen therefore offers a powerful strategy to identify essential regulators of complex mammalian differentiation processes.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA2/metabolismo , Genoma Humano/genética , Megacariócitos/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Sítios de Ligação , Diferenciação Celular , Células Cultivadas , Humanos , Megacariócitos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteína 1 de Leucemia Linfocítica Aguda de Células T
15.
Haematologica ; 95(12): 2005-12, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20713462

RESUMO

BACKGROUND: The collagen receptor glycoprotein VI generates activating signals through an immunoreceptor tyrosine-based activating motif on the co-associated Fc receptor gamma chain. Leukocyte-associated immunoglobulin-like receptor-1 also ligates collagen but generates inhibitory signals through immunoreceptor tyrosine-based inhibitory motifs. Thus far, the cellular expression of glycoprotein VI and leukocyte-associated immunoglobulin-like receptor-1 appears mutually exclusive. DESIGN AND METHODS: Using flow cytometry, we studied expression of collagen receptors on differentiating human megakaryocytes. CD34(+) cells were isolated from umbilical cord blood and matured to megakaryocytes in vitro. Freshly isolated bone marrow cells were used to study primary megakaryocytes. Upon cell sorting, cytospins were made to examine cytological characteristics of differentiation. RESULTS: Megakaryocyte maturation is accompanied by up-regulation of glycoprotein VI and down-regulation of leukocyte-associated immunoglobulin-like receptor-1. Interestingly, both in cultures from hematopoietic stem cells and primary cells obtained directly from bone marrow, we identified a subset of morphologically distinct megakaryocytes which co-express glycoprotein VI and leukocyte-associated immunoglobulin-like receptor-1. CONCLUSIONS: This is the first report of a primary cell that co-expresses these collagen receptors with opposite signaling properties. Since megakaryocytes mature in the collagen-rich environment of the bone marrow, these findings may point to a role for leukocyte-associated immunoglobulin-like receptor-1 in the control of megakaryocyte maturation/migration.


Assuntos
Plaquetas/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Imunológicos/metabolismo , Antígenos CD34/metabolismo , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Sangue Fetal/citologia , Citometria de Fluxo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Integrina alfa2beta1/metabolismo , Megacariócitos/metabolismo , Células-Tronco Multipotentes/metabolismo , Receptores de Colágeno/metabolismo , Trombopoese
16.
Blood ; 113(19): e1-9, 2009 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-19228925

RESUMO

Hematopoiesis is a carefully controlled process that is regulated by complex networks of transcription factors that are, in part, controlled by signals resulting from ligand binding to cell-surface receptors. To further understand hematopoiesis, we have compared gene expression profiles of human erythroblasts, megakaryocytes, B cells, cytotoxic and helper T cells, natural killer cells, granulocytes, and monocytes using whole genome microarrays. A bioinformatics analysis of these data was performed focusing on transcription factors, immunoglobulin superfamily members, and lineage-specific transcripts. We observed that the numbers of lineage-specific genes varies by 2 orders of magnitude, ranging from 5 for cytotoxic T cells to 878 for granulocytes. In addition, we have identified novel coexpression patterns for key transcription factors involved in hematopoiesis (eg, GATA3-GFI1 and GATA2-KLF1). This study represents the most comprehensive analysis of gene expression in hematopoietic cells to date and has identified genes that play key roles in lineage commitment and cell function. The data, which are freely accessible, will be invaluable for future studies on hematopoiesis and the role of specific genes and will also aid the understanding of the recent genome-wide association studies.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular/genética , Expressão Gênica , Atlas como Assunto , Linhagem da Célula , Células Cultivadas , Citometria de Fluxo , Perfilação da Expressão Gênica , Hematopoese , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Transcrição/metabolismo
17.
Exp Hematol ; 36(12): 1714-27, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19007685
18.
Br J Haematol ; 141(6): 808-13, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18422784

RESUMO

Congenital amegakaryocytic thrombocytopenia (CAMT) is a rare disorder that presents with severe thrombocytopenia and absence of megakaryocytes in the bone marrow. The disease may develop into bone marrow aplasia. Genetic defects in the gene encoding the thrombopoietin (Tpo) receptor, MPL, are the cause of this disease. In a previous study, we identified four missense mutations in CAMT patients, predicting Arg102Pro, Pro136His, Arg257Cys and Pro635Leu. To investigate whether these mutations result in defective Tpo-binding and/or signalling, full-length wildtype and mutant MPL were transduced into K562 cells. Expression levels and the ability to activate the mitogen-activated protein kinase, Janus kinase-signal transducer and activator of transcription and phosphoinositide-3 kinase pathways upon Tpo-binding were studied. The results predicted that MPL carrying the P136H or P635L mutation was not properly expressed, whereas the R102P and R257C mutations resulted in impaired signal transduction. Our results indicate that a severe clinical course may be expected when these mutations lead to absent Mpl expression or signalling in CAMT patients with missense mutations.


Assuntos
Megacariócitos/patologia , Mutação de Sentido Incorreto , Receptores de Trombopoetina/genética , Trombocitopenia/genética , Substituição de Aminoácidos , Pré-Escolar , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Células K562 , Fosforilação/efeitos dos fármacos , Receptores de Trombopoetina/agonistas , Receptores de Trombopoetina/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transdução de Sinais/efeitos dos fármacos , Trombocitopenia/congênito , Trombocitopenia/patologia , Trombopoetina/farmacologia
19.
Transfusion ; 48(5): 893-901, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18298597

RESUMO

BACKGROUND: The freezing curve currently used for the cryopreservation of peripheral blood stem cell transplants (PBSCTs) has been determined empirically. Although the use of cryopreserved PBSCTs is successful and usually leads to rapid hematopoietic recovery, the freeze-thawing process is known to induce a significant degree of cell death. Furthermore, the infusion of dimethyl sulfoxide (DMSO), used to protect the cells against damage induced by freezing, can cause morbidity. Therefore, optimizing the current cryopreservation protocol (with 10% DMSO and a slow linear cooling curve) with theoretically optimized freezing curves and a lower DMSO concentration might improve the recovery after transplantation. STUDY DESIGN AND METHODS: A theoretical model was used to predict optimal freezing curves for 5 and 10 percent DMSO. CD34+-selected and -unselected PBSCs were cryopreserved with the current or the new freezing curves. Postthaw quality was evaluated by cell viability, colony formation, and megakaryocyte outgrowth. RESULTS: With 10 percent DMSO, the use of the predicted optimal freezing curve resulted in increased postthaw viability of CD34+ cells, colony formation, and megakaryocyte outgrowth. Lowering the DMSO concentration to 5 percent resulted in improved postthaw viability and functionality, which was not further improved by use of the theoretically optimized freezing curve. CONCLUSIONS: Our results indicate that the current cryopreservation method for PBSCTs can be improved by either lowering the DMSO concentration to 5 percent or by using the theoretically optimized freezing curve. Infusion of less DMSO and more viable cells might improve the outcome of PBSCT.


Assuntos
Armazenamento de Sangue/métodos , Preservação de Sangue/métodos , Criopreservação/métodos , Células-Tronco Hematopoéticas/citologia , Antígenos CD34/metabolismo , Sobrevivência Celular , Granulócitos/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Megacariócitos/citologia , Modelos Biológicos , Monócitos/citologia , Dinâmica não Linear
20.
Comput Biol Chem ; 31(3): 178-85, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17499550

RESUMO

Our ability to detect differentially expressed genes in a microarray experiment can be hampered when the number of biological samples of interest is limited. In this situation, we propose the use of information from self-self hybridizations to acuminate our inference of differential expression. A unified modelling strategy is developed to allow better estimation of the error variance. This principle is similar to the use of a pooled variance estimate in the two-sample t-test. The results from real dataset examples suggest that we can detect more genes that are differentially expressed in the combined models. Our simulation study provides evidence that this method increases sensitivity compared to using the information from comparative hybridizations alone, given the same control for false discovery rate. The largest increase in sensitivity occurs when the amount of information in the comparative hybridization is limited.


Assuntos
Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Algoritmos , Plaquetas/metabolismo , Simulação por Computador , DNA Complementar/genética , Eritroblastos/metabolismo , Genótipo , Humanos , Modelos Lineares , Megacariócitos/metabolismo , Hibridização de Ácido Nucleico/métodos , Glicoproteínas da Membrana de Plaquetas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...