Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Curr Opin Cell Biol ; 85: 102255, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37806296

ABSTRACT

The hematopoietic system is one of the earliest tissues to develop. De novo generation of hematopoietic progenitor and stem cells occurs through a transdifferentiation of (hemogenic) endothelial cells to hematopoietic identity, resulting in the formation of intra-aortic hematopoietic cluster (IAHC) cells. Heterogeneity of IAHC cell phenotypes and functions has stymied the field in its search for the transcriptional program of emerging hematopoietic stem cells (HSCs), given that an individual IAHC cannot be simultaneously examined for function and transcriptome. Several models could account for this heterogeneity, including a novel model suggesting that the transcriptomes of individual emerging IAHC cells are in an unstable/metastable state, with pivotal hematopoietic transcription factors expressed dynamically due to transcriptional pulsing and combinatorial activities. The question remains - how is functional hematopoietic cell fate established - is the process stochastic? This article touches upon these important issues, which may be relevant to the field's inability to make HSCs ex vivo.


Subject(s)
Endothelial Cells , Hematopoietic Stem Cells , Cell Differentiation , Cell Transdifferentiation/physiology , Stochastic Processes
2.
Exp Hematol ; 118: 1-11, 2023 02.
Article in English | MEDLINE | ID: mdl-36529317

ABSTRACT

The adult-definitive hematopoietic hierarchy and hematopoietic stem cells (HSCs) residing in the bone marrow are established during embryonic development. In mouse, human, and many other mammals, it is the sudden formation of so-called intra-aortic/arterial hematopoietic clusters (IAHCs) that best signifies and visualizes this de novo generation of HSCs and hematopoietic progenitor cells (HPCs). Cluster cells arise through an endothelial-to-hematopoietic transition and, for some time, express markers/genes of both tissue types, whilst acquiring more hematopoietic features and losing endothelial ones. Among several hundreds of IAHC cells, the midgestation mouse embryo contains only very few bona fide adult-repopulating HSCs, suggestive of a challenging cell fate to achieve. Most others are HPCs of various types, some of which have the potential to mature into HSCs in vitro. Based on the number of cells that reveal hematopoietic function, a fraction of IAHC cells is uncharacterized. This review aims to explore the current state of knowledge on IAHC cells. We will describe markers useful for isolation and characterization of these fleetingly produced, yet vitally important, cells and for the refined enrichment of the HSCs they contain, and speculate on the role of some IAHC cells that are as-yet functionally uncharacterized.


Subject(s)
Embryo, Mammalian , Hematopoietic Stem Cells , Pregnancy , Female , Mice , Animals , Humans , Cell Differentiation/physiology , Hematopoietic Stem Cells/metabolism , Embryo, Mammalian/metabolism , Mammals
3.
Hemasphere ; 6(6): e737, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35647488

ABSTRACT

The hierarchical framework of the adult blood system as we know it from current medical and hematology textbooks, displays a linear branching network of dividing and differentiated cells essential for the growth and maintenance of the healthy organism. This view of the hierarchy has evolved over the last 75 years. An amazing increase in cellular complexity has been realized; however, innovative single-cell technologies continue to uncover essential cell types and functions in animal models and the human blood system. The most potent cell of the hematopoietic hierarchy is the hematopoietic stem cell. Stem cells for adult tissues are the long-lived self-renewing cellular component, which ensure that differentiated tissue-specific cells are maintained and replaced through the entire adult lifespan. Although much blood research is focused on hematopoietic tissue homeostasis, replacement and regeneration during adult life, embryological studies have widened and enriched our understanding of additional developmental hierarchies and interacting cells of this life-sustaining tissue. Here, we review the current state of knowledge of the hierarchical organization and the vast heterogeneity of the hematopoietic system from embryonic to adult stages.

4.
Blood Adv ; 5(3): 829-842, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33560396

ABSTRACT

Integrated molecular signals regulate cell fate decisions in the embryonic aortic endothelium to drive hematopoietic stem cell (HSC) generation during development. The G-protein-coupled receptor 56 (Gpr56, also called Adgrg1) is the most highly upregulated receptor gene in cells that take on hematopoietic fate and is expressed by adult bone marrow HSCs. Despite the requirement for Gpr56 in hematopoietic stem/progenitor cell (HS/PC) generation in zebrafish embryos and the highly upregulated expression of GPR56 in treatment-resistant leukemic patients, its function in normal mammalian hematopoiesis remains unclear. Here, we examine the role of Gpr56 in HS/PC development in Gpr56 conditional knockout (cKO) mouse embryos and Gpr knockout (KO) embryonic stem cell (ESC) hematopoietic differentiation cultures. Our results show a bias toward myeloid differentiation of Gpr56 cKO fetal liver HSCs and an increased definitive myeloid progenitor cell frequency in Gpr56KO ESC differentiation cultures. Surprisingly, we find that mouse Gpr97 can rescue Gpr56 morphant zebrafish hematopoietic generation, and that Gpr97 expression is upregulated in mouse Gpr56 deletion models. When both Gpr56 and Gpr97 are deleted in ESCs, no or few hematopoietic PCs (HPCs) are generated upon ESC differentiation. Together, our results reveal novel and redundant functions for these 2 G-protein coupled receptors in normal mammalian hematopoietic cell development and differentiation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Zebrafish , Animals , Cell Differentiation , Hematopoiesis/genetics , Hematopoietic Stem Cells , Humans , Mice , Receptors, G-Protein-Coupled/genetics , Zebrafish/genetics
5.
Development ; 147(23)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323375

ABSTRACT

The central nervous system hosts parenchymal macrophages, known as microglia, and non-parenchymal macrophages, collectively termed border-associated macrophages (BAMs). Microglia, but not BAMs, were reported to be absent in mice lacking a conserved Csf1r enhancer: the fms-intronic regulatory element (FIRE). However, it is unknown whether FIRE deficiency also impacts BAM arrival and/or maintenance. Here, we show that macrophages in the ventricular system of the brain, including Kolmer's epiplexus macrophages, are absent in Csf1rΔFIRE/ΔFIRE mice. Stromal choroid plexus BAMs are also considerably reduced. During normal development, we demonstrate that intracerebroventricular macrophages arrive from embryonic day 10.5, and can traverse ventricular walls in embryonic slice cultures. In Csf1rΔFIRE/ΔFIRE embryos, the arrival of both primitive microglia and intracerebroventricular macrophages was eliminated, whereas the arrival of cephalic mesenchyme and stromal choroid plexus BAMs was only partially restricted. Our results provide new insights into the development and regulation of different CNS macrophage populations.


Subject(s)
Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Macrophages/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Brain/growth & development , Brain/metabolism , Central Nervous System/growth & development , Embryo, Mammalian , Introns/genetics , Mice , Microglia/metabolism , Parenchymal Tissue/growth & development , Parenchymal Tissue/metabolism , Regulatory Sequences, Nucleic Acid
6.
EMBO J ; 39(8): e104270, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32149421

ABSTRACT

Hematopoietic stem cells (HSCs) develop from the hemogenic endothelium in cluster structures that protrude into the embryonic aortic lumen. Although much is known about the molecular characteristics of the developing hematopoietic cells, we lack a complete understanding of their origin and the three-dimensional organization of the niche. Here, we use advanced live imaging techniques of organotypic slice cultures, clonal analysis, and mathematical modeling to show the two-step process of intra-aortic hematopoietic cluster (IACH) formation. First, a hemogenic progenitor buds up from the endothelium and undergoes division forming the monoclonal core of the IAHC. Next, surrounding hemogenic cells are recruited into the IAHC, increasing their size and heterogeneity. We identified the Notch ligand Dll4 as a negative regulator of the recruitment phase of IAHC. Blocking of Dll4 promotes the entrance of new hemogenic Gfi1+ cells into the IAHC and increases the number of cells that acquire HSC activity. Mathematical modeling based on our data provides estimation of the cluster lifetime and the average recruitment time of hemogenic cells to the cluster under physiologic and Dll4-inhibited conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Aorta/embryology , Calcium-Binding Proteins/genetics , Cell Division , Endothelial Progenitor Cells/physiology , Female , Hemangioblasts/physiology , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Models, Theoretical
7.
Blood ; 134(22): 1929-1940, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31697805

ABSTRACT

Along with the aorta-gonad-mesonephros region, the head is a site of hematopoietic stem and progenitor cell (HS/PC) development in the mouse embryo. Macrophages are present in both these embryonic hemogenic sites, and recent studies indicate a functional interaction of macrophages with hematopoietic cells as they are generated in the aorta. Whereas brain macrophages or "microglia" are known to affect neuronal patterning and vascular circuitry in the embryonic brain, it is unknown whether macrophages play a role in head hematopoiesis. Here, we characterize head macrophages and examine whether they affect the HS/PC output of the hindbrain-branchial arch (HBA) region of the mouse embryo. We show that HBA macrophages are CD45+F4/80+CD11b+Gr1- and express the macrophage-specific Csf1r-GFP reporter. In the HBA of chemokine receptor-deficient (Cx3cr1-/-) embryos, a reduction in erythropoiesis is concomitant with a decrease in HBA macrophage percentages. In cocultures, we show that head macrophages boost hematopoietic progenitor cell numbers from HBA endothelial cells > twofold, and that the proinflammatory factor tumor necrosis factor-α is produced by head macrophages and influences HBA hematopoiesis in vitro. Taken together, head macrophages play a positive role in HBA erythropoiesis and HS/PC expansion and/or maturation, acting as microenvironmental cellular regulators in hematopoietic development.


Subject(s)
Embryo, Mammalian/embryology , Erythropoiesis/physiology , Head/embryology , Hematopoietic Stem Cells/metabolism , Macrophages/metabolism , Animals , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Male , Mice , Mice, Knockout
8.
Elife ; 82019 02 13.
Article in English | MEDLINE | ID: mdl-30758286

ABSTRACT

The origins and functions of kidney macrophages in the adult have been explored, but their roles during development remain largely unknown. Here we characterise macrophage arrival, localisation, heterogeneity, and functions during kidney organogenesis. Using genetic approaches to ablate macrophages, we identify a role for macrophages in nephron progenitor cell clearance as mouse kidney development begins. Throughout renal organogenesis, most kidney macrophages are perivascular and express F4/80 and CD206. These macrophages are enriched for mRNAs linked to developmental processes, such as blood vessel morphogenesis. Using antibody-mediated macrophage-depletion, we show macrophages support vascular anastomoses in cultured kidney explants. We also characterise a subpopulation of galectin-3+ (Gal3+) myeloid cells within the developing kidney. Our findings may stimulate research into macrophage-based therapies for renal developmental abnormalities and have implications for the generation of bioengineered kidney tissues.


Subject(s)
Galectin 3/genetics , Kidney/growth & development , Nephrons/growth & development , Organogenesis/genetics , Animals , Calcium-Binding Proteins/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Kidney/metabolism , Lectins, C-Type/genetics , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice , Nephrons/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/metabolism
9.
J Exp Med ; 215(1): 233-248, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29217535

ABSTRACT

Cell fate is established through coordinated gene expression programs in individual cells. Regulatory networks that include the Gata2 transcription factor play central roles in hematopoietic fate establishment. Although Gata2 is essential to the embryonic development and function of hematopoietic stem cells that form the adult hierarchy, little is known about the in vivo expression dynamics of Gata2 in single cells. Here, we examine Gata2 expression in single aortic cells as they establish hematopoietic fate in Gata2Venus mouse embryos. Time-lapse imaging reveals rapid pulsatile level changes in Gata2 reporter expression in cells undergoing endothelial-to-hematopoietic transition. Moreover, Gata2 reporter pulsatile expression is dramatically altered in Gata2+/- aortic cells, which undergo fewer transitions and are reduced in hematopoietic potential. Our novel finding of dynamic pulsatile expression of Gata2 suggests a highly unstable genetic state in single cells concomitant with their transition to hematopoietic fate. This reinforces the notion that threshold levels of Gata2 influence fate establishment and has implications for transcription factor-related hematologic dysfunctions.


Subject(s)
Cell Differentiation , GATA2 Transcription Factor/genetics , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Single-Cell Analysis , Animals , Female , Fluorescent Antibody Technique , GATA2 Transcription Factor/metabolism , Gene Expression , Genes, Reporter , Male , Mice , Mice, Transgenic , Phenotype , Single-Cell Analysis/methods
11.
FEBS Lett ; 590(22): 3975-3986, 2016 11.
Article in English | MEDLINE | ID: mdl-27543859

ABSTRACT

The development of the hematopoietic system during early embryonic stages occurs in spatially and temporally distinct waves. Hematopoietic stem cells (HSC), the most potent and self-renewing cells of this system, are produced in the final 'definitive' wave of hematopoietic cell generation. In contrast to HSCs in the adult, which differentiate via intermediate progenitor populations to produce functional blood cells, the generation of hematopoietic cells in the embryo prior to HSC generation occurs in the early waves by producing blood cells without intermediate progenitors (such as the 'primitive' hematopoietic cells). The lineage relationship between the early hematopoietic cells and the cells giving rise to HSCs, the genetic networks controlling their emergence, and the precise temporal determination of HSC fate remain topics of intense research and debate. This Review article discusses the current knowledge on the step-wise embryonic establishment of the adult hematopoietic system, examines the roles of pivotal intrinsic regulators in this process, and raises questions concerning the temporal onset of HSC fate determination.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells , Adult , Animals , Cell Lineage/genetics , Humans , Mice
12.
Blood ; 128(15): 1928-1939, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27554085

ABSTRACT

Enhancers are the primary determinants of cell identity, and specific promoter/enhancer combinations of Endoglin (ENG) have been shown to target blood and endothelium in the embryo. Here, we generated a series of embryonic stem cell lines, each targeted with reporter constructs driven by specific promoter/enhancer combinations of ENG, to evaluate their discriminative potential and value as molecular probes of the corresponding transcriptome. The Eng promoter (P) in combination with the -8/+7/+9-kb enhancers, targeted cells in FLK1 mesoderm that were enriched for blast colony forming potential, whereas the P/-8-kb enhancer targeted TIE2+/c-KIT+/CD41- endothelial cells that were enriched for hematopoietic potential. These fractions were isolated using reporter expression and their transcriptomes profiled by RNA-seq. There was high concordance between our signatures and those from embryos with defects at corresponding stages of hematopoiesis. Of the 6 genes that were upregulated in both hemogenic mesoderm and hemogenic endothelial fractions targeted by the reporters, LRP2, a multiligand receptor, was the only gene that had not previously been associated with hematopoiesis. We show that LRP2 is indeed involved in definitive hematopoiesis and by doing so validate the use of reporter gene-coupled enhancers as probes to gain insights into transcriptional changes that facilitate cell fate transitions.


Subject(s)
Embryo, Mammalian/metabolism , Endoglin/metabolism , Enhancer Elements, Genetic/physiology , Hematopoiesis/physiology , Molecular Probes/metabolism , Animals , Cell Line , Embryo, Mammalian/cytology , Endoglin/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Molecular Probes/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism
13.
Dev Biol ; 416(1): 34-41, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27235813

ABSTRACT

Hematopoietic cell generation in the midgestation mouse embryo occurs through the natural transdifferentiation of temporally and spatially restricted set of hemogenic endothelial cells. These cells take on hematopoietic fate in the aorta, vitelline and umbilical arteries and appear as hematopoietic cell clusters that emerge from the vascular wall. Genetic and live imaging data have supported this. Recently, the embryonic head has been shown to contain fully functional hematopoietic stem cells (HSC). By lineage tracing, cerebrovascular specific endothelial cells were shown to contribute to the postnatal mouse hematopoietic system. Since Ly6aGFP is a marker of all HSCs, some hematopoietic cluster cells and hemogenic endothelial cells in the midgestation mouse aorta, we examine here whether embryonic head HSCs and vascular endothelial cells are positive for this marker. Whereas some head vasculature, single hematopoietic cells and all HSCs are Ly6aGFP expressing, we do not find clusters of hematopoietic cells emerging from the cerebrovasculature that are characteristic of endothelial-to-hematopoietic transition.


Subject(s)
Antigens, Ly/analysis , Head/embryology , Membrane Proteins/analysis , Animals , Antigens, Differentiation/analysis , Female , Green Fluorescent Proteins , Hematopoietic Stem Cells , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
Stem Cell Reports ; 6(3): 383-95, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26923823

ABSTRACT

Hematopoietic stem cells (HSC), the self-renewing cells of the adult blood differentiation hierarchy, are generated during embryonic stages. The first HSCs are produced in the aorta-gonad-mesonephros (AGM) region of the embryo through endothelial to a hematopoietic transition. BMP4 and Hedgehog affect their production and expansion, but it is unknown whether they act to affect the same HSCs. In this study using the BRE GFP reporter mouse strain that identifies BMP/Smad-activated cells, we find that the AGM harbors two types of adult-repopulating HSCs upon explant culture: One type is BMP-activated and the other is a non-BMP-activated HSC type that is indirectly controlled by Hedgehog signaling through the VEGF pathway. Transcriptomic analyses demonstrate that the two HSC types express distinct but overlapping genetic programs. These results revealing the bifurcation in HSC types at early embryonic stages in the AGM explant model suggest that their development is dependent upon the signaling molecules in the microenvironment.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Hedgehog Proteins/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Smad Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism
15.
Blood ; 127(11): 1426-37, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26834239

ABSTRACT

The Gata2 transcription factor is a pivotal regulator of hematopoietic cell development and maintenance, highlighted by the fact that Gata2 haploinsufficiency has been identified as the cause of some familial cases of acute myelogenous leukemia/myelodysplastic syndrome and in MonoMac syndrome. Genetic deletion in mice has shown that Gata2 is pivotal to the embryonic generation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). It functions in the embryo during endothelial cell to hematopoietic cell transition to affect hematopoietic cluster, HPC, and HSC formation. Gata2 conditional deletion and overexpression studies show the importance of Gata2 levels in hematopoiesis, during all developmental stages. Although previous studies of cell populations phenotypically enriched in HPCs and HSCs show expression of Gata2, there has been no direct study of Gata2 expressing cells during normal hematopoiesis. In this study, we generate a Gata2Venus reporter mouse model with unperturbed Gata2 expression to examine the hematopoietic function and transcriptome of Gata2 expressing and nonexpressing cells. We show that all the HSCs are Gata2 expressing. However, not all HPCs in the aorta, vitelline and umbilical arteries, and fetal liver require or express Gata2. These Gata2-independent HPCs exhibit a different functional output and genetic program, including Ras and cyclic AMP response element-binding protein pathways and other Gata factors, compared with Gata2-dependent HPCs. Our results, indicating that Gata2 is of major importance in programming toward HSC fate but not in all cells with HPC fate, have implications for current reprogramming strategies.


Subject(s)
Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Aorta/cytology , Aorta/embryology , Bacterial Proteins/analysis , Bacterial Proteins/genetics , Cell Lineage , Cells, Cultured , Cellular Reprogramming Techniques , GATA2 Transcription Factor/deficiency , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/physiology , Genes, Reporter , Genetic Vectors/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/classification , Hematopoietic Stem Cells/physiology , Liver/cytology , Liver/embryology , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transcriptome , Transgenes , Umbilical Arteries/cytology , Umbilical Arteries/embryology
17.
Nat Commun ; 6: 8040, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26282601

ABSTRACT

Adult haematopoiesis is the outcome of distinct haematopoietic stem cell (HSC) subtypes with self-renewable repopulating ability, but with different haematopoietic cell lineage outputs. The molecular basis for this heterogeneity is largely unknown. BMP signalling regulates HSCs as they are first generated in the aorta-gonad-mesonephros region, but at later developmental stages, its role in HSCs is controversial. Here we show that HSCs in murine fetal liver and the bone marrow are of two types that can be prospectively isolated--BMP activated and non-BMP activated. Clonal transplantation demonstrates that they have distinct haematopoietic lineage outputs. Moreover, the two HSC types differ in intrinsic genetic programs, thus supporting a role for the BMP signalling axis in the regulation of HSC heterogeneity and lineage output. Our findings provide insight into the molecular control mechanisms that define HSC types and have important implications for reprogramming cells to HSC fate and treatments targeting distinct HSC types.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/physiology , Signal Transduction/physiology , Animals , Benzofurans , Bone Morphogenetic Proteins/genetics , Embryo, Mammalian , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Quinolines
18.
J Exp Med ; 212(1): 93-106, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25547674

ABSTRACT

Hematopoietic stem cells (HSCs) are generated via a natural transdifferentiation process known as endothelial to hematopoietic cell transition (EHT). Because of small numbers of embryonal arterial cells undergoing EHT and the paucity of markers to enrich for hemogenic endothelial cells (ECs [HECs]), the genetic program driving HSC emergence is largely unknown. Here, we use a highly sensitive RNAseq method to examine the whole transcriptome of small numbers of enriched aortic HSCs, HECs, and ECs. Gpr56, a G-coupled protein receptor, is one of the most highly up-regulated of the 530 differentially expressed genes. Also, highly up-regulated are hematopoietic transcription factors, including the "heptad" complex of factors. We show that Gpr56 (mouse and human) is a target of the heptad complex and is required for hematopoietic cluster formation during EHT. Our results identify the processes and regulators involved in EHT and reveal the surprising requirement for Gpr56 in generating the first HSCs.


Subject(s)
Cell Transdifferentiation/genetics , Endothelial Cells/metabolism , Gene Expression Profiling/methods , Hematopoietic Stem Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Animals , CHO Cells , COS Cells , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cricetulus , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endothelial Cells/cytology , Female , Gene Ontology , Hematopoietic Stem Cells/cytology , Humans , In Situ Hybridization , Mice, Inbred C57BL , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA/methods , Up-Regulation
19.
J Exp Med ; 210(13): 2843-50, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24297996

ABSTRACT

Knowledge of the key transcription factors that drive hematopoietic stem cell (HSC) generation is of particular importance for current hematopoietic regenerative approaches and reprogramming strategies. Whereas GATA2 has long been implicated as a hematopoietic transcription factor and its dysregulated expression is associated with human immunodeficiency syndromes and vascular integrity, it is as yet unknown how GATA2 functions in the generation of HSCs. HSCs are generated from endothelial cells of the major embryonic vasculature (aorta, vitelline, and umbilical arteries) and are found in intra-aortic hematopoietic clusters. In this study, we find that GATA2 function is essential for the generation of HSCs during the stage of endothelial-to-hematopoietic cell transition. Specific deletion of Gata2 in Vec (Vascular Endothelial Cadherin)-expressing endothelial cells results in a deficiency of long-term repopulating HSCs and intra-aortic cluster cells. By specific deletion of Gata2 in Vav-expressing hematopoietic cells (after HSC generation), we further show that GATA2 is essential for HSC survival. This is in contrast to the known activity of the RUNX1 transcription factor, which functions only in the generation of HSCs, and highlights the unique requirement for GATA2 function in HSCs throughout all developmental stages.


Subject(s)
GATA2 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Alleles , Animals , Apoptosis , Cell Separation , Cell Survival , Flow Cytometry , Gene Deletion , Mice , Mice, Knockout , Mice, Transgenic , Stem Cells
20.
Cell Stem Cell ; 9(6): 541-52, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22136929

ABSTRACT

Hematopoietic stem cells (HSCs) and an earlier wave of definitive erythroid/myeloid progenitors (EMPs) differentiate from hemogenic endothelial cells in the conceptus. EMPs can be generated in vitro from embryonic or induced pluripotent stem cells, but efforts to produce HSCs have largely failed. The formation of both EMPs and HSCs requires the transcription factor Runx1 and its non-DNA binding partner core binding factor ß (CBFß). Here we show that the requirements for CBFß in EMP and HSC formation in the conceptus are temporally and spatially distinct. Panendothelial expression of CBFß in Tek-expressing cells was sufficient for EMP formation, but was not adequate for HSC formation. Expression of CBFß in Ly6a-expressing cells, on the other hand, was sufficient for HSC, but not EMP, formation. The data indicate that EMPs and HSCs differentiate from distinct populations of hemogenic endothelial cells, with Ly6a expression specifically marking the HSC-generating hemogenic endothelium.


Subject(s)
Core Binding Factor beta Subunit/metabolism , Endothelial Cells/physiology , Erythroid Cells/metabolism , Hematopoietic Stem Cells/physiology , Myeloid Cells/physiology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Cell Differentiation/physiology , Cell Lineage , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Endothelial Cells/cytology , Erythroid Cells/cytology , Hematopoietic Stem Cells/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Myeloid Cells/cytology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...