Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Blood ; 113(22): 5456-65, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19346495

ABSTRACT

The basic helix-loop-helix transcription factor Scl/Tal1 controls the development and subsequent differentiation of hematopoietic stem cells (HSCs). However, because few Scl target genes have been validated to date, the underlying mechanisms have remained largely unknown. In this study, we have used ChIP-Seq technology (coupling chromatin immunoprecipitation with deep sequencing) to generate a genome-wide catalog of Scl-binding events in a stem/progenitor cell line, followed by validation using primary fetal liver cells and comprehensive transgenic mouse assays. Transgenic analysis provided in vivo validation of multiple new direct Scl target genes and allowed us to reconstruct an in vivo validated network consisting of 17 factors and their respective regulatory elements. By coupling ChIP-Seq in model cell lines with in vivo transgenic validation and sophisticated bioinformatic analysis, we have identified a widely applicable strategy for the reconstruction of stem cell regulatory networks in which biologic material is otherwise limiting. Moreover, in addition to revealing multiple previously unrecognized links to known HSC regulators, as well as novel links to genes not previously implicated in HSC function, comprehensive transgenic analysis of regulatory elements provided substantial new insights into the transcriptional control of several important hematopoietic regulators, including Cbfa2t3h/Eto2, Cebpe, Nfe2, Zfpm1/Fog1, Erg, Mafk, Gfi1b, and Myb.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Proto-Oncogene Proteins/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites/genetics , Cells, Cultured , Embryo, Mammalian/blood supply , Gene Expression Profiling , Genome , Humans , Mice , Mice, Transgenic , Models, Biological , Protein Binding , Proto-Oncogene Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription, Genetic
2.
Stem Cells ; 28(10): 1751-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20715180

ABSTRACT

A common feature of early embryo cells from the inner cell mass (ICM) and of ESCs is an absolute dependence on an atypical cell cycle in which the G1 phase is shortened to preserve their self-renewing and pluripotent nature. The transcription factor B-Myb has been attributed a role in proliferation, in particular during the G2/M phases of the cell cycle. Intriguingly, B-Myb levels in ICM/ESCs are greater than 100 times compared with those in normal proliferating cells, suggesting a particularly important function for this transcription factor in pluripotent stem cells. B-Myb is essential for embryo development beyond the preimplantation stage, but its role in ICM/ESCs remains unclear. Using a combination of mouse genetics, single DNA fiber analyses and high-resolution three-dimensional (3D) imaging, we demonstrate that B-Myb has no influence on the expression of pluripotency factors, but instead B-Myb ablation leads to stalling of replication forks and superactivation of replication factories that result in disorganization of the replication program and an increase in double-strand breaks. These effects are partly due to aberrant transcriptional regulation of cell cycle proliferation factors, namely c-Myc and FoxM1, which dictate normal S phase progression. We conclude that B-Myb acts crucially during the S phase in ESCs by facilitating proper progression of replication, thereby protecting the cells from genomic damage. Our findings have particular relevance in the light of the potential therapeutic application of ESCs and the need to maintain their genomic integrity.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , S Phase/genetics , Trans-Activators/metabolism , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation , Chromatin Immunoprecipitation , Flow Cytometry , Fluorescent Antibody Technique , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Mice , Microscopy, Confocal , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Trans-Activators/genetics
3.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30877232

ABSTRACT

Mutations at the N- or C-terminus of C/EBPα are frequent in acute myeloid leukaemia (AML) with normal karyotype. Here, we investigate the role of the transcription factor Myb in AMLs driven by different combinations of CEBPA mutations. Using knockdown of Myb in murine cell lines modelling the spectrum of CEBPA mutations, we show that the effect of reduced Myb depends on the mutational status of the two Cebpa alleles. Importantly, Myb knockdown fails to override the block in myeloid differentiation in cells with biallelic N-terminal C/EBPα mutations, demonstrating for the first time that the dependency on Myb is much lower in AML with this mutational profile. By comparing gene expression following Myb knockdown and chromatin immunoprecipitation sequencing data for the binding of C/EBPα isoforms, we provide evidence for a functional cooperation between C/EBPα and Myb in the maintenance of AML. This co-dependency breaks down when both alleles of CEBPA harbour N-terminal mutations, as a subset of C/EBPα-regulated genes only bind the short p30 C/EBPα isoform and, unlike other C/EBPα-regulated genes, do so without a requirement for Myb.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation/genetics , Proto-Oncogene Proteins c-myb/genetics , Alleles , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Mice , Phenotype , Protein Isoforms/genetics , RNA, Small Interfering/genetics , Transfection
4.
Cancer Res ; 78(20): 5767-5779, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30082276

ABSTRACT

Myelodysplastic syndromes (MDS) are a heterogeneous group of diseases characterized by blood cytopenias that occur as a result of somatic mutations in hematopoietic stem cells (HSC). MDS leads to ineffective hematopoiesis, and as many as 30% of patients progress to acute myeloid leukemia (AML). The mechanisms by which mutations accumulate in HSC during aging remain poorly understood. Here we identify a novel role for MYBL2 in DNA double-strand break (DSB) repair in HSC. In patients with MDS, low MYBL2 levels associated with and preceded transcriptional deregulation of DNA repair genes. Stem/progenitor cells from these patients display dysfunctional DSB repair kinetics after exposure to ionizing radiation (IR). Haploinsufficiency of Mybl2 in mice also led to a defect in the repair of DSBs induced by IR in HSC and was characterized by unsustained phosphorylation of the ATM substrate KAP1 and telomere fragility. Our study identifies MYBL2 as a crucial regulator of DSB repair and identifies MYBL2 expression levels as a potential biomarker to predict cellular response to genotoxic treatments in MDS and to identify patients with defects in DNA repair. Such patients with worse prognosis may require a different therapeutic regimen to prevent progression to AML.Significance: These findings suggest MYBL2 levels may be used as a biological biomarker to determine the DNA repair capacity of hematopoietic stem cells from patients with MDS and as a clinical biomarker to inform decisions regarding patient selection for treatments that target DNA repair.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/20/5767/F1.large.jpg Cancer Res; 78(20); 5767-79. ©2018 AACR.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , Hematopoietic Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Cell Proliferation , Comet Assay , DNA Repair , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genotype , Humans , Kinetics , Mice , Mice, Inbred C57BL , Myelodysplastic Syndromes/metabolism , Phosphorylation , Radiation, Ionizing
5.
Exp Hematol ; 34(5): 654-63, 2006 May.
Article in English | MEDLINE | ID: mdl-16647571

ABSTRACT

OBJECTIVE: The differentiation of megakaryocytes is characterized by polyploidization and cytoplasmic maturation leading to platelet production. Studying these processes is hindered by the paucity of bone marrow megakaryocytes and their precursors. We describe a method for the expansion and purification of committed megakaryocyte progenitors and demonstrate their usefulness by studying changes in the expression of Ets and GATA family transcription factors throughout megakaryocytopoiesis. METHODS: A two-step serum-free method was developed. Cells isolated using this method were analyzed for surface marker expression by flow cytometry, and for their ability to differentiate using single-cell culture. Purified progenitors were induced to differentiate and analyzed with respect to their ploidy by flow cytometry and expression of specific genes by RT-PCR. RESULTS: A population of Lin- c-kit+ CD45+ CD41+ CD31+ CD34low CD9low FcgammaRII/IIIlow Sca-1med/low committed megakaryocyte progenitors was purified. These cells could be differentiated efficiently, achieving ploidy of up to 128N. Analysis of RNA demonstrated the expected increases in expression of key megakaryocyte-associated genes. RT-PCR analysis also revealed that a range of Ets and GATA factors are expressed, their individual levels and patterns of expression varying widely. Surprisingly, we find that GATA-6 is specifically expressed in late differentiated megakaryocytes and has the potential to regulate megakaryocyte-expressed genes in cooperation with Ets factors. CONCLUSION: Purified primary megakaryocytic progenitors are able to differentiate as a cohort into fully mature megakaryocytes. The number of cells obtainable, and the synchrony of the differentiation process, facilitates analysis of the dynamics of molecular processes involved in megakaryocytopoiesis. The expression pattern of Ets and GATA family transcription factors reveals the complexity of the involvement of these key megakaryocytic regulators. The finding of GATA-6 expression and demonstration of its functional activity suggests a novel mechanism for the regulation of certain genes late in megakaryocytopoiesis.


Subject(s)
Cell Differentiation , GATA6 Transcription Factor/physiology , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Proto-Oncogene Proteins c-ets/physiology , Animals , Base Sequence , DNA Primers , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
6.
PLoS One ; 10(9): e0138257, 2015.
Article in English | MEDLINE | ID: mdl-26382271

ABSTRACT

The MYB transcription factor plays critical roles in normal and malignant haematopoiesis. We previously showed that MYB was a direct activator of FLT3 expression within the context of acute myeloid leukaemia. During normal haematopoiesis, increasing levels of FLT3 expression determine a strict hierarchy within the haematopoietic stem and early progenitor compartment, which associates with lymphoid and myeloid commitment potential. We use the conditional deletion of the Myb gene to investigate the influence of MYB in Flt3 transcriptional regulation within the haematopoietic stem cell (HSC) hierarchy. In accordance with previous report, in vivo deletion of Myb resulted in rapid biased differentiation of HSC with concomitant loss of proliferation capacity. We find that loss of MYB activity also coincided with decreased FLT3 expression. At the chromatin level, the Flt3 promoter is primed in immature HSC, but occupancy of further intronic elements determines expression. Binding to these locations, MYB and C/EBPα need functional cooperation to activate transcription of the locus. This cooperation is cell context dependent and indicates that MYB and C/EBPα activities are inter-dependent in controlling Flt3 expression to influence lineage commitment of multipotential progenitors.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Hematopoietic Stem Cells/metabolism , Oncogene Proteins v-myb/physiology , fms-Like Tyrosine Kinase 3/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Gene Expression Regulation , Hematopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins v-myb/genetics , fms-Like Tyrosine Kinase 3/metabolism
7.
PLoS One ; 7(8): e43300, 2012.
Article in English | MEDLINE | ID: mdl-22952660

ABSTRACT

Product of the Itga2b gene, CD41 contributes to hematopoietic stem cell (HSC) and megakaryocyte/platelet functions. CD41 expression marks the onset of definitive hematopoiesis in the embryo where it participates in regulating the numbers of multipotential progenitors. Key to platelet aggregation, CD41 expression also characterises their precursor, the megakaryocyte, and is specifically up regulated during megakaryopoiesis. Though phenotypically unique, megakaryocytes and HSC share numerous features, including key transcription factors, which could indicate common sub-regulatory networks. In these respects, Itga2b can serve as a paradigm to study features of both developmental-stage and HSC- versus megakaryocyte-specific regulations. By comparing different cellular contexts, we highlight a mechanism by which internal promoters participate in Itga2b regulation. A developmental process connects epigenetic regulation and promoter switching leading to CD41 expression in HSC. Interestingly, a similar process can be observed at the Mpl locus, which codes for another receptor that defines both HSC and megakaryocyte identities. Our study shows that Itga2b expression is controlled by lineage-specific networks and associates with H4K8ac in megakaryocyte or H3K27me3 in the multipotential hematopoietic cell line HPC7. Correlating with the decrease in H3K27me3 at the Itga2b Iocus, we find that following commitment to megakaryocyte differentiation, the H3K27 demethylase Jmjd3 up-regulation influences both Itga2b and Mpl expression.


Subject(s)
Hematopoiesis/physiology , Integrin alpha2/metabolism , Receptors, Thrombopoietin/biosynthesis , Cell Differentiation , Cell Line , Cell Lineage , Chromatin Immunoprecipitation , Cluster Analysis , DNA/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Hematopoiesis/genetics , Humans , Megakaryocytes/cytology , Models, Genetic , Oligonucleotide Array Sequence Analysis , Phenotype , Platelet Membrane Glycoprotein IIb/metabolism , Promoter Regions, Genetic , Recombinant Proteins/metabolism
8.
Mol Cell Proteomics ; 6(3): 548-64, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17186946

ABSTRACT

The platelet surface is poorly characterized due to the low abundance of many membrane proteins and the lack of specialist tools for their investigation. In this study we identified novel human platelet and mouse megakaryocyte membrane proteins using specialist proteomics and genomics approaches. Three separate methods were used to enrich platelet surface proteins prior to identification by liquid chromatography and tandem mass spectrometry: lectin affinity chromatography, biotin/NeutrAvidin affinity chromatography, and free flow electrophoresis. Many known, abundant platelet surface transmembrane proteins and several novel proteins were identified using each receptor enrichment strategy. In total, two or more unique peptides were identified for 46, 68, and 22 surface membrane, intracellular membrane, and membrane proteins of unknown subcellular localization, respectively. The majority of these were single transmembrane proteins. To complement the proteomics studies, we analyzed the transcriptome of a highly purified preparation of mature primary mouse megakaryocytes using serial analysis of gene expression in view of the increasing importance of mutant mouse models in establishing protein function in platelets. This approach identified all of the major classes of platelet transmembrane receptors, including multitransmembrane proteins. Strikingly 17 of the 25 most megakaryocyte-specific genes (relative to 30 other serial analysis of gene expression libraries) were transmembrane proteins, illustrating the unique nature of the megakaryocyte/platelet surface. The list of novel plasma membrane proteins identified using proteomics includes the immunoglobulin superfamily member G6b, which undergoes extensive alternate splicing. Specific antibodies were used to demonstrate expression of the G6b-B isoform, which contains an immunoreceptor tyrosine-based inhibition motif. G6b-B undergoes tyrosine phosphorylation and association with the SH2 domain-containing phosphatase, SHP-1, in stimulated platelets suggesting that it may play a novel role in limiting platelet activation.


Subject(s)
Blood Platelets/chemistry , Megakaryocytes/chemistry , Membrane Proteins/analysis , Animals , Cell Membrane/chemistry , Cells, Cultured , Chromatography, Affinity , Gene Expression , Genomics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Platelet Activation , Proteomics , RNA/metabolism , Receptors, Immunologic/analysis , Receptors, Immunologic/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL