Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
Semin Cell Dev Biol ; 127: 59-67, 2022 07.
Article in English | MEDLINE | ID: mdl-35125239

ABSTRACT

Haematopoietic stem and progenitor cells emerge from specialized haemogenic endothelial cells in select vascular beds during embryonic development. Specification and commitment to the blood lineage, however, occur before endothelial cells are endowed with haemogenic competence, at the time of mesoderm patterning and production of endothelial cell progenitors (angioblasts). Whilst early blood cell fate specification has long been recognized, very little is known about the mechanisms that induce endothelial cell diversification and progressive acquisition of a blood identity by a subset of these cells. Here, we review the endothelial origin of the haematopoietic system and the complex developmental journey of blood-fated angioblasts. We discuss how recent technological advances will be instrumental to examine the diversity of the embryonic anatomical niches, signaling pathways and downstream epigenetic and transcriptional processes controlling endothelial cell heterogeneity and blood cell fate specification. Ultimately, this will give essential insights into the ontogeny of the cells giving rise to haematopoietic stem cells, that may aid in the development of novel strategies for their in vitro production for clinical purposes.


Subject(s)
Hemangioblasts , Cell Differentiation , Cell Lineage , Endothelium , Female , Hemangioblasts/metabolism , Hematopoietic Stem Cells , Humans , Mesoderm/metabolism , Pregnancy
3.
Nat Cell Biol ; 23(1): 61-74, 2021 01.
Article in English | MEDLINE | ID: mdl-33420489

ABSTRACT

Extra-embryonic mesoderm (ExM)-composed of the earliest cells that traverse the primitive streak-gives rise to the endothelium as well as haematopoietic progenitors in the developing yolk sac. How a specific subset of ExM becomes committed to a haematopoietic fate remains unclear. Here we demonstrate using an embryonic stem cell model that transient expression of the T-box transcription factor Eomesodermin (Eomes) governs haemogenic competency of ExM. Eomes regulates the accessibility of enhancers that the transcription factor stem cell leukaemia (SCL) normally utilizes to specify primitive erythrocytes and is essential for the normal development of Runx1+ haemogenic endothelium. Single-cell RNA sequencing suggests that Eomes loss of function profoundly blocks the formation of blood progenitors but not specification of Flk-1+ haematoendothelial progenitors. Our findings place Eomes at the top of the transcriptional hierarchy regulating early blood formation and suggest that haemogenic competence is endowed earlier during embryonic development than was previously appreciated.


Subject(s)
Embryonic Stem Cells/cytology , Hemangioblasts/cytology , Mesoderm/cytology , T-Box Domain Proteins/physiology , Yolk Sac/cytology , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Stem Cells/metabolism , Female , Hemangioblasts/metabolism , Male , Mesoderm/metabolism , Mice, Knockout , Pregnancy , RNA-Seq , Single-Cell Analysis , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Yolk Sac/metabolism
4.
Haematologica ; 106(4): 1106-1119, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32527952

ABSTRACT

The megakaryocyte/erythroid Transient Myeloproliferative Disorder (TMD) in newborns with Down Syndrome (DS) occurs when N-terminal truncating mutations of the hemopoietic transcription factor GATA1, that produce GATA1short protein (GATA1s), are acquired early in development. Prior work has shown that murine GATA1s, by itself, causes a transient yolk sac myeloproliferative disorder. However, it is unclear where in the hemopoietic cellular hierarchy GATA1s exerts its effects to produce this myeloproliferative state. Here, through a detailed examination of hemopoiesis from murine GATA1s ES cells and GATA1s embryos we define defects in erythroid and megakaryocytic differentiation that occur relatively late in hemopoiesis. GATA1s causes an arrest late in erythroid differentiation in vivo, and even more profoundly in ES-cell derived cultures, with a marked reduction of Ter-119 cells and reduced erythroid gene expression. In megakaryopoiesis, GATA1s causes a differentiation delay at a specific stage, with accumulation of immature, kit-expressing CD41hi megakaryocytic cells. In this specific megakaryocytic compartment, there are increased numbers of GATA1s cells in S-phase of cell cycle and reduced number of apoptotic cells compared to GATA1 cells in the same cell compartment. There is also a delay in maturation of these immature GATA1s megakaryocytic lineage cells compared to GATA1 cells at the same stage of differentiation. Finally, even when GATA1s megakaryocytic cells mature, they mature aberrantly with altered megakaryocyte-specific gene expression and activity of the mature megakaryocyte enzyme, acetylcholinesterase. These studies pinpoint the hemopoietic compartment where GATA1s megakaryocyte myeloproliferation occurs, defining where molecular studies should now be focussed to understand the oncogenic action of GATA1s.


Subject(s)
Down Syndrome , Leukemoid Reaction , Animals , Cell Differentiation , GATA1 Transcription Factor/genetics , Humans , Infant, Newborn , Megakaryocytes , Mice
5.
R Soc Open Sci ; 7(1): 191048, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32218938

ABSTRACT

Chromatin remodelling and transcription factors play important roles in lineage commitment and development through control of gene expression. Activation of selected lineage-specific genes and repression of alternative lineage-affiliated genes result in tightly regulated cell differentiation transcriptional programmes. However, the complex functional and physical interplay between transcription factors and chromatin-modifying enzymes remains elusive. Recent evidence has implicated histone demethylases in normal haematopoietic differentiation as well as in malignant haematopoiesis. Here, we report an interaction between H3K4 demethylase JARID1A and the haematopoietic-specific master transcription proteins SCL and GATA1 in red blood cells. Specifically, we observe a direct physical contact between GATA1 and the second PHD domain of JARID1A. This interaction has potential implications for normal and malignant haematopoiesis.

6.
Nat Commun ; 10(1): 1083, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842454

ABSTRACT

VEGFA signaling controls physiological and pathological angiogenesis and hematopoiesis. Although many context-dependent signaling pathways downstream of VEGFA have been uncovered, vegfa transcriptional regulation in vivo remains unclear. Here, we show that the ETS transcription factor, Etv6, positively regulates vegfa expression during Xenopus blood stem cell development through multiple transcriptional inputs. In agreement with its established repressive functions, Etv6 directly inhibits expression of the repressor foxo3, to prevent Foxo3 from binding to and repressing the vegfa promoter. Etv6 also directly activates expression of the activator klf4; reflecting a genome-wide paucity in ETS-binding motifs in Etv6 genomic targets, Klf4 then recruits Etv6 to the vegfa promoter to activate its expression. These two mechanisms (double negative gate and feed-forward loop) are classic features of gene regulatory networks specifying cell fates. Thus, Etv6's dual function, as a transcriptional repressor and activator, controls a major signaling pathway involved in endothelial and blood development in vivo.


Subject(s)
Forkhead Box Protein O3/metabolism , Kruppel-Like Transcription Factors/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics , Xenopus Proteins/metabolism , Xenopus laevis/physiology , Animals , Embryo, Nonmammalian , Endothelium/embryology , Endothelium/metabolism , Forkhead Box Protein O3/antagonists & inhibitors , Forkhead Box Protein O3/genetics , Gene Expression Regulation, Developmental/physiology , Gene Regulatory Networks/physiology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Morpholinos/genetics , Oligonucleotides, Antisense/genetics , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction/physiology , Somites/embryology , Somites/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , ETS Translocation Variant 6 Protein
7.
Nat Commun ; 9(1): 5375, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30560907

ABSTRACT

During development, it is unclear if lineage-fated cells derive from multilineage-primed progenitors and whether active mechanisms operate to restrict cell fate. Here we investigate how mesoderm specifies into blood-fated cells. We document temporally restricted co-expression of blood (Scl/Tal1), cardiac (Mesp1) and paraxial (Tbx6) lineage-affiliated transcription factors in single cells, at the onset of blood specification, supporting the existence of common progenitors. At the same time-restricted stage, absence of SCL results in expansion of cardiac/paraxial cell populations and increased cardiac/paraxial gene expression, suggesting active suppression of alternative fates. Indeed, SCL normally activates expression of co-repressor ETO2 and Polycomb-PRC1 subunits (RYBP, PCGF5) and maintains levels of Polycomb-associated histone marks (H2AK119ub/H3K27me3). Genome-wide analyses reveal ETO2 and RYBP co-occupy most SCL target genes, including cardiac/paraxial loci. Reduction of Eto2 or Rybp expression mimics Scl-null cardiac phenotype. Therefore, SCL-mediated transcriptional repression prevents mis-specification of blood-fated cells, establishing active repression as central to fate determination processes.


Subject(s)
Cell Lineage/physiology , Gene Expression Regulation, Developmental/physiology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Cell Separation/methods , Embryo, Mammalian , Flow Cytometry/methods , Histone Code/physiology , Mesoderm/cytology , Mesoderm/physiology , Mice , Mouse Embryonic Stem Cells , Nuclear Proteins/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/metabolism , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Transcription Factors/genetics
8.
Nat Immunol ; 19(1): 85-97, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29167569

ABSTRACT

The hierarchy of human hemopoietic progenitor cells that produce lymphoid and granulocytic-monocytic (myeloid) lineages is unclear. Multiple progenitor populations produce lymphoid and myeloid cells, but they remain incompletely characterized. Here we demonstrated that lympho-myeloid progenitor populations in cord blood - lymphoid-primed multi-potential progenitors (LMPPs), granulocyte-macrophage progenitors (GMPs) and multi-lymphoid progenitors (MLPs) - were functionally and transcriptionally distinct and heterogeneous at the clonal level, with progenitors of many different functional potentials present. Although most progenitors had the potential to develop into only one mature cell type ('uni-lineage potential'), bi- and rarer multi-lineage progenitors were present among LMPPs, GMPs and MLPs. Those findings, coupled with single-cell expression analyses, suggest that a continuum of progenitors execute lymphoid and myeloid differentiation, rather than only uni-lineage progenitors' being present downstream of stem cells.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling/methods , Lymphoid Progenitor Cells/metabolism , Myeloid Progenitor Cells/metabolism , Single-Cell Analysis/methods , Animals , Cell Lineage/genetics , Cell Separation/methods , Cells, Cultured , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation/methods , Humans , Mice , Transplantation, Heterologous
9.
Blood ; 129(15): 2051-2060, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28179281

ABSTRACT

SCL/TAL1 (stem cell leukemia/T-cell acute lymphoblastic leukemia [T-ALL] 1) is an essential transcription factor in normal and malignant hematopoiesis. It is required for specification of the blood program during development, adult hematopoietic stem cell survival and quiescence, and terminal maturation of select blood lineages. Following ectopic expression, SCL contributes to oncogenesis in T-ALL. Remarkably, SCL's activities are all mediated through nucleation of a core quaternary protein complex (SCL:E-protein:LMO1/2 [LIM domain only 1 or 2]:LDB1 [LIM domain-binding protein 1]) and dynamic recruitment of conserved combinatorial associations of additional regulators in a lineage- and stage-specific context. The finely tuned control of SCL's regulatory functions (lineage priming, activation, and repression of gene expression programs) provides insight into fundamental developmental and transcriptional mechanisms, and highlights mechanistic parallels between normal and oncogenic processes. Importantly, recent discoveries are paving the way to the development of innovative therapeutic opportunities in SCL+ T-ALL.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/pathology , Humans , LIM Domain Proteins/biosynthesis , LIM Domain Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/biosynthesis , Transcription Factors/genetics
10.
J Exp Med ; 213(8): 1513-35, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27377587

ABSTRACT

Our understanding of the perturbation of normal cellular differentiation hierarchies to create tumor-propagating stem cell populations is incomplete. In human acute myeloid leukemia (AML), current models suggest transformation creates leukemic stem cell (LSC) populations arrested at a progenitor-like stage expressing cell surface CD34. We show that in ∼25% of AML, with a distinct genetic mutation pattern where >98% of cells are CD34(-), there are multiple, nonhierarchically arranged CD34(+) and CD34(-) LSC populations. Within CD34(-) and CD34(+) LSC-containing populations, LSC frequencies are similar; there are shared clonal structures and near-identical transcriptional signatures. CD34(-) LSCs have disordered global transcription profiles, but these profiles are enriched for transcriptional signatures of normal CD34(-) mature granulocyte-macrophage precursors, downstream of progenitors. But unlike mature precursors, LSCs express multiple normal stem cell transcriptional regulators previously implicated in LSC function. This suggests a new refined model of the relationship between LSCs and normal hemopoiesis in which the nature of genetic/epigenetic changes determines the disordered transcriptional program, resulting in LSC differentiation arrest at stages that are most like either progenitor or precursor stages of hemopoiesis.


Subject(s)
Antigens, CD34/genetics , Granulocyte-Macrophage Progenitor Cells/metabolism , Leukemia, Myeloid, Acute , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Animals , Antigens, CD34/metabolism , Granulocyte-Macrophage Progenitor Cells/pathology , Heterografts , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/pathology
11.
Sci Rep ; 6: 27195, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27250641

ABSTRACT

Adult-type intraembryonic hematopoiesis arises from specialized endothelial cells of the dorsal aorta (DA). Despite the critical importance of this specialized endothelium for establishment of hematopoietic stem cells and adult hematopoietic lineages, the mechanisms regulating its emergence are incompletely understood. We show that EphrinB2, a principal regulator of endothelial cell function, controls the development of endothelium producing adult-type hematopoiesis. The absence of EphrinB2 impairs DA-derived hematopoiesis. Transmembrane EphrinB2 and its EphB4 receptor interact in the emerging DA, which transiently harbors EphrinB2(+) and EphB4(+) endothelial cells, thereby providing an opportunity for bi-directional cell-to-cell signaling to control the emergence of the hemogenic endothelium. Embryonic Stem (ES) cell-derived EphrinB2(+) cells are enriched with hemogenic endothelial precursors. EphrinB2 silencing impairs ES generation of hematopoietic cells but not generation of endothelial cells. The identification of EphrinB2 as an essential regulator of adult hematopoiesis provides important insight in the regulation of early hematopoietic commitment.


Subject(s)
Aorta/cytology , Ephrin-B2/metabolism , Hemangioblasts/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Aorta/metabolism , Cell Differentiation , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Hemangioblasts/metabolism , Hematopoiesis , Mice , Mouse Embryonic Stem Cells/metabolism , Tissue Culture Techniques
12.
Exp Hematol ; 43(10): 821-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26143582

ABSTRACT

Every 2 years since 1978, an international group of scientists, physicians, and other researchers meet to discuss the latest developments in the underlying etiology, mechanisms of action, and developmental acquisition of cellular and systemic defects exhibited and elicited by the most common inherited human disorders, the hemoglobinopathies. The 19th Hemoglobin Switching Conference, held in September 2014 at St. John's College in Oxford, once again exceeded all expectations by describing cutting edge research in cellular, molecular, developmental, and genomic advances focused on these diseases. The conference comprised about 60 short talks over 3 days by leading investigators in the field. This meeting report describes the highlights of the conference.


Subject(s)
Hemoglobinopathies/genetics , Hemoglobinopathies/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Congresses as Topic , Hemoglobinopathies/pathology , Hemoglobinopathies/therapy , Humans
14.
Cell Rep ; 4(1): 135-47, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23831025

ABSTRACT

Cell fate is governed by combinatorial actions of transcriptional regulators assembling into multiprotein complexes. However, the molecular details of how these complexes form are poorly understood. One such complex, which contains the basic-helix-loop-helix heterodimer SCL:E47 and bridging proteins LMO2:LDB1, critically regulates hematopoiesis and induces T cell leukemia. Here, we report the crystal structure of (SCL:E47)bHLH:LMO2:LDB1LID bound to DNA, providing a molecular account of the network of interactions assembling this complex. This reveals an unexpected role for LMO2. Upon binding to SCL, LMO2 induces new hydrogen bonds in SCL:E47, thereby strengthening heterodimer formation. This imposes a rotation movement onto E47 that weakens the heterodimer:DNA interaction, shifting the main DNA-binding activity onto additional protein partners. Along with biochemical analyses, this illustrates, at an atomic level, how hematopoietic-specific SCL sequesters ubiquitous E47 and associated cofactors and supports SCL's reported DNA-binding-independent functions. Importantly, this work will drive the design of small molecules inhibiting leukemogenic processes.


Subject(s)
DNA/chemistry , Hematopoiesis/genetics , LIM Domain Proteins/chemistry , Molecular Docking Simulation , Transcription, Genetic , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Tumor , DNA/metabolism , HEK293 Cells , Humans , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mice , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Protein Multimerization , Zebrafish
15.
Nucleic Acids Res ; 41(9): 4938-48, 2013 May.
Article in English | MEDLINE | ID: mdl-23519611

ABSTRACT

We report the genomic occupancy profiles of the key hematopoietic transcription factor GATA-1 in pro-erythroblasts and mature erythroid cells fractionated from day E12.5 mouse fetal liver cells. Integration of GATA-1 occupancy profiles with available genome-wide transcription factor and epigenetic profiles assayed in fetal liver cells enabled as to evaluate GATA-1 involvement in modulating local chromatin structure of target genes during erythroid differentiation. Our results suggest that GATA-1 associates preferentially with changes of specific epigenetic modifications, such as H4K16, H3K27 acetylation and H3K4 di-methylation. Furthermore, we used random forest (RF) non-linear regression to predict changes in the expression levels of GATA-1 target genes based on the genomic features available for pro-erythroblasts and mature fetal liver-derived erythroid cells. Remarkably, our prediction model explained a high proportion of 62% of variation in gene expression. Hierarchical clustering of the proximity values calculated by the RF model produced a clear separation of upregulated versus downregulated genes and a further separation of downregulated genes in two distinct groups. Thus, our study of GATA-1 genome-wide occupancy profiles in mouse primary erythroid cells and their integration with global epigenetic marks reveals three clusters of GATA-1 gene targets that are associated with specific epigenetic signatures and functional characteristics.


Subject(s)
Epigenesis, Genetic , Erythropoiesis/genetics , GATA1 Transcription Factor/metabolism , Liver/metabolism , Animals , Cells, Cultured , Erythroid Cells/metabolism , Fetus , Genome , Histones/metabolism , Liver/cytology , Liver/embryology , Mice
16.
Dev Cell ; 24(2): 144-58, 2013 Jan 28.
Article in English | MEDLINE | ID: mdl-23318133

ABSTRACT

VEGFA signaling is critical for endothelial and hematopoietic stem cell (HSC) specification. However, blood defects resulting from perturbation of the VEGFA pathway are always accompanied by impaired vascular/arterial development. Because HSCs derive from arterial cells, it is unclear whether VEGFA directly contributes to HSC specification. This is an important question for our understanding of how HSCs are formed and for developing their production in vitro. Through knockdown of the regulator ETO2 in embryogenesis, we report a specific decrease in expression of medium/long Vegfa isoforms in somites. This leads to absence of Notch1 expression and failure of HSC specification in the dorsal aorta (DA), independently of vessel formation and arterial specification. Vegfa hypomorphs and isoform-specific (medium/long) morphants not only recapitulate this phenotype but also demonstrate that VEGFA short isoform is sufficient for DA development. Therefore, sequential, isoform-specific VEGFA signaling successively induces the endothelial, arterial, and HSC programs in the DA.


Subject(s)
Aorta/embryology , Arteries/embryology , Co-Repressor Proteins/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , Vascular Endothelial Growth Factor A/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Aorta/metabolism , Arteries/metabolism , Co-Repressor Proteins/genetics , Embryonic Development , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Morpholinos , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction , Transcription Factors , Transcription, Genetic , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/growth & development , Xenopus laevis/metabolism
17.
PLoS One ; 7(3): e32981, 2012.
Article in English | MEDLINE | ID: mdl-22396803

ABSTRACT

Because of the scarcity of megakaryocytes in hematopoietic tissues, studying megakaryopoiesis heavily relies on the availability of appropriate cellular models. Here, we report the establishment of a new mouse embryonic stem (ES) cell-derived megakaryocytic cell line, MKD1. The cells are factor-dependent, their cell surface immunophenotype and gene expression profile closely resemble that of primary megakaryocyte progenitors (MkPs) and they further differentiate along the megakaryocyte lineage upon valproic acid treatment. At a functional level, we show that ablation of SCL expression, a transcription factor critical for MkP maturation, leads to gene expression alterations similar to that observed in primary, Scl-excised MkPs. Moreover, the cell line is amenable to biochemical and transcriptional analyses, as we report for GpVI, a direct target of SCL. Thus, the MKD1 cell line offers a pertinent experimental model to study the cellular and molecular mechanisms underlying MkP biology and more broadly megakaryopoiesis.


Subject(s)
Cell Culture Techniques , Embryonic Stem Cells/cytology , Megakaryocyte Progenitor Cells/cytology , Megakaryocytes/cytology , Animals , Cell Differentiation , Cell Line , Cell Lineage , Cells, Cultured , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Immunophenotyping , Mice , Mice, Transgenic , Promoter Regions, Genetic , Stem Cells/cytology , Transcription Factors/metabolism , Transcription, Genetic
18.
Blood ; 118(3): 723-35, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21596846

ABSTRACT

Megakaryopoiesis is a complex process that involves major cellular and nuclear changes and relies on controlled coordination of cellular proliferation and differentiation. These mechanisms are orchestrated in part by transcriptional regulators. The key hematopoietic transcription factor stem cell leukemia (SCL)/TAL1 is required in early hematopoietic progenitors for specification of the megakaryocytic lineage. These early functions have, so far, prevented full investigation of its role in megakaryocyte development in loss-of-function studies. Here, we report that SCL critically controls terminal megakaryocyte maturation. In vivo deletion of Scl specifically in the megakaryocytic lineage affects all key attributes of megakaryocyte progenitors (MkPs), namely, proliferation, ploidization, cytoplasmic maturation, and platelet release. Genome-wide expression analysis reveals increased expression of the cell-cycle regulator p21 in Scl-deleted MkPs. Importantly, p21 knockdown-mediated rescue of Scl-mutant MkPs shows full restoration of cell-cycle progression and partial rescue of the nuclear and cytoplasmic maturation defects. Therefore, SCL-mediated transcriptional control of p21 is essential for terminal maturation of MkPs. Our study provides a mechanistic link between a major hematopoietic transcriptional regulator, cell-cycle progression, and megakaryocytic differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hematopoietic Stem Cells/physiology , Megakaryocytes/physiology , Proto-Oncogene Proteins/metabolism , Thrombocytopenia/physiopathology , Thrombopoiesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Marrow Cells/physiology , Bone Marrow Cells/ultrastructure , Cell Division/physiology , Cell Lineage/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cytoplasm/physiology , Gene Knockdown Techniques , Hematopoietic Stem Cells/ultrastructure , Megakaryocytes/ultrastructure , Mice , Microscopy, Electron , Polyploidy , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , Thrombocytopenia/pathology
19.
Cancer Cell ; 19(1): 138-52, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21251617

ABSTRACT

The relationships between normal and leukemic stem/progenitor cells are unclear. We show that in ∼80% of primary human CD34+ acute myeloid leukemia (AML), two expanded populations with hemopoietic progenitor immunophenotype coexist in most patients. Both populations have leukemic stem cell (LSC) activity and are hierarchically ordered; one LSC population gives rise to the other. Global gene expression profiling shows the LSC populations are molecularly distinct and resemble normal progenitors but not stem cells. The more mature LSC population most closely mirrors normal granulocyte-macrophage progenitors (GMP) and the immature LSC population a previously uncharacterized progenitor functionally similar to lymphoid-primed multipotential progenitors (LMPPs). This suggests that in most cases primary CD34+ AML is a progenitor disease where LSCs acquire abnormal self-renewal potential.


Subject(s)
Granulocyte-Macrophage Progenitor Cells/cytology , Leukemia, Myeloid, Acute/pathology , Lymphoid Progenitor Cells/cytology , Neoplastic Stem Cells/pathology , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Gene Expression Profiling , Graft Survival , Granulocyte-Macrophage Progenitor Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/metabolism , Leukocyte Common Antigens/metabolism , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/transplantation , Transplantation, Heterologous/pathology , Young Adult
20.
Blood ; 117(7): 2146-56, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21076045

ABSTRACT

The LIM only protein 2 (LMO2) is a key regulator of hematopoietic stem cell development whose ectopic expression in T cells leads to the onset of acute lymphoblastic leukemia. Through its LIM domains, LMO2 is thought to function as the scaffold for a DNA-binding transcription regulator complex, including the basic helix-loop-helix proteins SCL/TAL1 and E47, the zinc finger protein GATA-1, and LIM-domain interacting protein LDB1. To understand the role of LMO2 in the formation of this complex and ultimately to dissect its function in normal and aberrant hematopoiesis, we solved the crystal structure of LMO2 in complex with the LID domain of LDB1 at 2.4 Å resolution. We observe a largely unstructured LMO2 kept in register by the LID binding both LIM domains. Comparison of independently determined crystal structures of LMO2 reveals large movements around a conserved hinge between the LIM domains. We demonstrate that such conformational flexibility is necessary for binding of LMO2 to its partner protein SCL/TAL1 in vitro and for the function of this complex in vivo. These results, together with molecular docking and analysis of evolutionarily conserved residues, yield the first structural model of the DNA-binding complex containing LMO2, LDB1, SCL/TAL1, and GATA-1.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Metalloproteins/chemistry , Metalloproteins/genetics , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Oncogenes , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Amino Acid Substitution , Animals , Animals, Genetically Modified , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Crystallography, X-Ray , DNA Primers/genetics , DNA-Binding Proteins/metabolism , Evolution, Molecular , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Hematopoiesis/genetics , Hematopoiesis/physiology , Humans , In Vitro Techniques , LIM Domain Proteins , Metalloproteins/metabolism , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Mutagenesis, Site-Directed , Oncogene Proteins/metabolism , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Static Electricity , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Two-Hybrid System Techniques , Zebrafish/embryology , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL