Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Elife ; 122024 May 29.
Article in English | MEDLINE | ID: mdl-38809590

ABSTRACT

Hematopoietic stem cells emerge in the embryo from an aortic-derived tissue called the hemogenic endothelium (HE). The HE appears to give birth to cells of different nature and fate but the molecular principles underlying this complexity are largely unknown. Here we show, in the zebrafish embryo, that two cell types emerge from the aortic floor with radically different morphodynamics. With the support of live imaging, we bring evidence suggesting that the mechanics underlying the two emergence types rely, or not, on apicobasal polarity establishment. While the first type is characterized by reinforcement of apicobasal polarity and maintenance of the apical/luminal membrane until release, the second type emerges via a dynamic process reminiscent of trans-endothelial migration. Interfering with Runx1 function suggests that the balance between the two emergence types depends on tuning apicobasal polarity at the level of the HE. In support of this and unexpectedly, we show that Pard3ba - one of the four Pard3 proteins expressed in the zebrafish - is sensitive to interference with Runx1 activity, in aortic endothelial cells. This supports the idea of a signaling cross talk controlling cell polarity and its associated features, between aortic and hemogenic cells. In addition, using new transgenic fish lines that express Junctional Adhesion Molecules and functional interference, we bring evidence for the essential role of ArhGEF11/PDZ-RhoGEF in controlling the HE-endothelial cell dynamic interface, including cell-cell intercalation, which is ultimately required for emergence completion. Overall, we highlight critical cellular and dynamic events of the endothelial-to-hematopoietic transition that support emergence complexity, with a potential impact on cell fate.


In mammals and other animals with backbones, the cells that will make up blood and immune cells are generated during a very narrow timeframe in embryonic development. These cells, called hematopoietic stem cells and progenitors (or HSPCs for short), emerge from tissue known as hemogenic endothelium that makes up the floor of early blood vessels. For HPSCs to eventually specialise into different types of blood and immune cells, they require diverse migratory and homing properties that, ultimately, will determine the specific type of functions they exert. An important question for scientists studying the development of different blood and immune cell types is when this commitment to functional diversity is established. It could, for example, arise due to cells in the hemogenic endothelium having different origins. Alternatively, the signals that generate hemogenic endothelium cells could be responsible. It is also possible that both explanations are true, and that having different mechanisms involved ensures diversity in populations of HSPCs. To investigate differences between the HSPCs emerging from the hemogenic endothelium, Torcq et al. studied zebrafish embryos that had been modified so that one of the proteins involved in sensing cell polarity ­ where the top and bottom of the cell are located ­ was fluorescent. Live imaging of the embryos showed that two types of cells, with striking differences in morphology, emerge from the hemogenic tissue. In addition, one cell type displays the same polarity as the other vessel cells, whereas the other does not. Torcq et al. also present evidence suggesting that the signals responsible for controlling this cell polarity are provided by surrounding blood vessel cells, supporting the idea of an interplay between the different cell types. The finding that two different cell types emerge from the hemogenic endothelium, reveals a potential new source of diversity in HSPCs. Ultimately, this is expected to contribute to their functional complexity, resulting in both long-term stem cells that retain their full regenerative potential into adulthood and more specialized blood and immune cells.


Subject(s)
Cell Polarity , Core Binding Factor Alpha 2 Subunit , Hematopoietic Stem Cells , Zebrafish Proteins , Zebrafish , Zebrafish/embryology , Animals , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoietic Stem Cells/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hemangioblasts/metabolism , Hemangioblasts/cytology , Hemangioblasts/physiology , Embryo, Nonmammalian/metabolism , Animals, Genetically Modified
2.
Sci China Life Sci ; 64(12): 2073-2087, 2021 12.
Article in English | MEDLINE | ID: mdl-34181164

ABSTRACT

During embryogenesis, hematopoietic stem progenitor cells (HSPCs) are believed to be derived from hemogenic endothelial cells (HECs). Moreover, arterial feature is proposed to be a prerequisite for HECs to generate HSPCs with lymphoid potential. Although the molecular basis of hematopoietic stem cell-competent HECs has been delicately elucidated within the embryo proper, the functional and molecular characteristics of HECs in the extraembryonic yolk sac (YS) remain largely unresolved. In this study, we initially identified six molecularly different endothelial populations in the midgestational YS through integrated analysis of several single-cell RNA sequencing (scRNA-seq) datasets and validated the arterial vasculature distribution of Gja5+ ECs using a Gja5-EGFP reporter mouse model. Further, we explored the hemogenic potential of different EC populations based on their Gja5-EGFP and CD44 expression levels. The hemogenic potential was ubiquitously detected in spatiotemporally different vascular beds on embryonic days (E)8.5-E9.5 and gradually concentrated in CD44-positive ECs from E10.0. Unexpectedly, B-lymphoid potential was detected in the YS ECs as early as E8.5 regardless of their arterial features. Furthermore, the capacity for generating hematopoietic progenitors with in vivo lymphoid potential was found in nonarterial as well as arterial YS ECs on E10.0-E10.5. Importantly, the distinct identities of E10.0-E10.5 HECs between YS and intraembryonic caudal region were revealed by further scRNA-seq analysis. Cumulatively, these findings extend our knowledge regarding the hemogenic potential of ECs from anatomically and molecularly different vascular beds, providing a theoretical basis for better understanding the sources of HSPCs during mammalian development.


Subject(s)
Hemangioblasts/physiology , Hematopoietic Stem Cells/physiology , Yolk Sac/blood supply , Animals , Gene Expression Profiling , Mice , Mice, Inbred Strains , Sequence Analysis, RNA
3.
Mech Ageing Dev ; 195: 111461, 2021 04.
Article in English | MEDLINE | ID: mdl-33600833

ABSTRACT

The research on neurodegenerative diseases (NeuroDegD) has been traditionally focused on later life stages. There is now an increasing evidence, that they may be programmed during early development. Here, we propose that NeuroDegD are the result of the complex process of imprinting on fetal hemogenic endothelium, from which the microglial cells make to origin. The central role of placenta and epigenetic mechanisms (methylation of DNA, histone modifications and regulation by non-coding RNAs) in mediating the short and long-term effects has been also described. Precisely, it reports their role in impacting plasticity and memory of microglial cells. In addition, we also underline the necessity of further studies for clearing all mechanisms involved and developing epigenetic methods for identifying potential targets as biomarkers, and for developing preventive measures. Such biomarkers might be used to identify individuals at risk to NeuroDegD. Finally, the sex dependence of fetal programming process has been discussed. It might justify the sex differences in the epidemiologic, imaging, biomarkers, and pathology studies of these pathologies. The discovery of related mechanisms might have important clinical implications in both the etiology of disorders and the management of pregnant women for encouraging healthy long-term outcomes for their children, and future generations. Impending research on the mechanisms related to transgenerational transmission of prenatal stress might consent the development and application of therapies and/or intervention strategies for these disorders in humans.


Subject(s)
Epigenesis, Genetic , Fetal Development/physiology , Hemangioblasts/physiology , Microglia/physiology , Neurodegenerative Diseases , Biological Variation, Individual , Biomarkers/analysis , Cell Plasticity , Gene Expression Regulation, Developmental , Humans , Molecular Imprinting , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/prevention & control , Risk Assessment , Sex Characteristics
4.
Annu Rev Physiol ; 83: 17-37, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33035429

ABSTRACT

Embryonic definitive hematopoiesis generates hematopoietic stem and progenitor cells (HSPCs) essential for establishment and maintenance of the adult blood system. This process requires the specification of a subset of vascular endothelial cells to become blood-forming, or hemogenic, and the subsequent endothelial-to-hematopoietic transition to generate HSPCs therefrom. The mechanisms that regulate these processes are under intensive investigation, as their recapitulation in vitro from human pluripotent stem cells has the potential to generate autologous HSPCs for clinical applications. In this review, we provide an overview of hemogenic endothelial cell development and highlight the molecular events that govern hemogenic specification of vascular endothelial cells and the generation of multilineage HSPCs from hemogenic endothelium. We also discuss the impact of hemogenic endothelial cell development on adult hematopoiesis.


Subject(s)
Endothelial Cells/physiology , Endothelium, Vascular/physiology , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/physiology , Hemangioblasts/physiology , Hematopoiesis/physiology , Humans
5.
Blood ; 136(7): 845-856, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32392346

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) in the bone marrow are derived from a small population of hemogenic endothelial (HE) cells located in the major arteries of the mammalian embryo. HE cells undergo an endothelial to hematopoietic cell transition, giving rise to HSPCs that accumulate in intra-arterial clusters (IAC) before colonizing the fetal liver. To examine the cell and molecular transitions between endothelial (E), HE, and IAC cells, and the heterogeneity of HSPCs within IACs, we profiled ∼40 000 cells from the caudal arteries (dorsal aorta, umbilical, vitelline) of 9.5 days post coitus (dpc) to 11.5 dpc mouse embryos by single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin sequencing. We identified a continuous developmental trajectory from E to HE to IAC cells, with identifiable intermediate stages. The intermediate stage most proximal to HE, which we term pre-HE, is characterized by increased accessibility of chromatin enriched for SOX, FOX, GATA, and SMAD motifs. A developmental bottleneck separates pre-HE from HE, with RUNX1 dosage regulating the efficiency of the pre-HE to HE transition. A distal candidate Runx1 enhancer exhibits high chromatin accessibility specifically in pre-HE cells at the bottleneck, but loses accessibility thereafter. Distinct developmental trajectories within IAC cells result in 2 populations of CD45+ HSPCs; an initial wave of lymphomyeloid-biased progenitors, followed by precursors of hematopoietic stem cells (pre-HSCs). This multiomics single-cell atlas significantly expands our understanding of pre-HSC ontogeny.


Subject(s)
Cell Differentiation , Endothelium/embryology , Hemangioblasts/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Embryo, Mammalian , Endothelium/cytology , Endothelium/metabolism , Female , Gene Dosage/physiology , Gene Expression Regulation, Developmental , Hemangioblasts/cytology , Hematopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , RNA-Seq/methods
6.
Crit Care ; 24(1): 71, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32122366

ABSTRACT

BACKGROUND: A dysbalanced coagulation system is part of the pathological host response to infection in sepsis. Activation of pro-coagulant pathways and attenuation of anti-coagulant activity ultimately lead to microvascular stasis and consequent organ failure. No treatment approaches specifically targeting this axis are available. We explored the effects of therapeutic plasma exchange (TPE) on microvascular coagulation dysbalance in septic shock. METHODS: We conducted a prospective single-center study enrolling 31 patients with early septic shock (onset < 12 h) requiring high doses of norepinephrine (NE > 0.4 µg/kg/min). Clinical and biochemical data, including measurement of protein C; a disintegrin and metalloprotease with a thrombospondin type 1 motif, member 13 (ADAMTS13); and von Willebrand factor antigen (vWF:Ag), were obtained before and after TPE against fresh frozen plasma. RESULTS: Antithrombotic acting proteins such as antithrombin-III (ATIII) and protein C were markedly reduced in septic patients, but their activity increased after TPE (ATIII, 51% (41-61) vs. 63% (48-70), p = 0.029; protein C, 47% (38-60) vs. 62% (54-69), p = 0.029). Median ADAMTS13 activity was increased by TPE from 27 (21-42) % before to 47 (38-62) % after TPE (p < 0.001). In contrast, vWF:Ag was elevated and could be reduced by TPE (353 (206-492) IU/dL vs. 170 (117-232) IU/dL, p < 0.001). Regression analysis yielded a correlation between ADAMTS13 activity and platelet count (p = 0.001, R2 = 0.316). CONCLUSIONS: Septic shock was associated with activation of pro-coagulant pathways and simultaneous depletion of anti-coagulant factors. TPE partially attenuated this dysbalance by removing pro- and by replacing anti-coagulant factors. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03065751. Retrospectively registered on 28 February 2017.


Subject(s)
Blood Coagulation/physiology , Hemangioblasts/physiology , Plasma Exchange/methods , Shock, Septic/blood , ADAMTS13 Protein/analysis , ADAMTS13 Protein/blood , Adult , Antithrombin III/analysis , Female , Hemangioblasts/enzymology , Humans , Male , Middle Aged , Prospective Studies , Shock, Septic/physiopathology , von Willebrand Factor/analysis
7.
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
8.
Nat Commun ; 10(1): 3577, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31395869

ABSTRACT

Haematopoietic stem cells are generated from the haemogenic endothelium (HE) located in the floor of the dorsal aorta (DA). Despite being integral to arteries, it is controversial whether HE and arterial endothelium share a common lineage. Here, we present a transgenic zebrafish runx1 reporter line to isolate HE and aortic roof endothelium (ARE)s, excluding non-aortic endothelium. Transcriptomic analysis of these populations identifies Runx1-regulated genes and shows that HE initially expresses arterial markers at similar levels to ARE. Furthermore, runx1 expression depends on prior arterial programming by the Notch ligand dll4. Runx1-/- mutants fail to downregulate arterial genes in the HE, which remains integrated within the DA, suggesting that Runx1 represses the pre-existing arterial programme in HE to allow progression towards the haematopoietic fate. These findings strongly suggest that, in zebrafish, aortic endothelium is a precursor to HE, with potential implications for pluripotent stem cell differentiation protocols for the generation of transplantable HSCs.


Subject(s)
Arteries/embryology , Endothelium, Vascular/embryology , Hemangioblasts/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Arteries/cytology , Arteries/metabolism , Cell Lineage , Core Binding Factor Alpha 2 Subunit/genetics , Embryo, Nonmammalian , Embryonic Development , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Knockout Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Exp Hematol ; 68: 2-9, 2018 12.
Article in English | MEDLINE | ID: mdl-30391350

ABSTRACT

The transcription factor RUNX1 is required in the embryo for formation of the adult hematopoietic system. Here, we describe the seminal findings that led to the discovery of RUNX1 and of its critical role in blood cell formation in the embryo from hemogenic endothelium (HE). We also present RNA-sequencing data demonstrating that HE cells in different anatomic sites, which produce hematopoietic progenitors with dissimilar differentiation potentials, are molecularly distinct. Hemogenic and non-HE cells in the yolk sac are more closely related to each other than either is to hemogenic or non-HE cells in the major arteries. Therefore, a major driver of the different lineage potentials of the committed erythro-myeloid progenitors that emerge in the yolk sac versus hematopoietic stem cells that originate in the major arteries is likely to be the distinct molecular properties of the HE cells from which they are derived. We used bioinformatics analyses to predict signaling pathways active in arterial HE, which include the functionally validated pathways Notch, Wnt, and Hedgehog. We also used a novel bioinformatics approach to assemble transcriptional regulatory networks and predict transcription factors that may be specifically involved in hematopoietic cell formation from arterial HE, which is the origin of the adult hematopoietic system.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Hemangioblasts/physiology , Hematopoiesis/physiology , Animals , Arteries/cytology , Arteries/embryology , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/deficiency , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/physiology , Drosophila Proteins/genetics , Fetal Blood/physiology , Gene Expression Regulation, Developmental , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/virology , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Knockout , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Transcription, Genetic , Yolk Sac/cytology
10.
Development ; 145(5)2018 03 12.
Article in English | MEDLINE | ID: mdl-29530939

ABSTRACT

During ontogeny, hematopoietic stem and progenitor cells arise from hemogenic endothelium through an endothelial-to-hematopoietic transition that is strictly dependent on the transcription factor RUNX1. Although it is well established that RUNX1 is essential for the onset of hematopoiesis, little is known about the role of RUNX1 dosage specifically in hemogenic endothelium and during the endothelial-to-hematopoietic transition. Here, we used the mouse embryonic stem cell differentiation system to determine if and how RUNX1 dosage affects hemogenic endothelium differentiation. The use of inducible Runx1 expression combined with alterations in the expression of the RUNX1 co-factor CBFß allowed us to evaluate a wide range of RUNX1 levels. We demonstrate that low RUNX1 levels are sufficient and necessary to initiate an effective endothelial-to-hematopoietic transition. Subsequently, RUNX1 is also required to complete the endothelial-to-hematopoietic transition and to generate functional hematopoietic precursors. In contrast, elevated levels of RUNX1 are able to drive an accelerated endothelial-to-hematopoietic transition, but the resulting cells are unable to generate mature hematopoietic cells. Together, our results suggest that RUNX1 dosage plays a pivotal role in hemogenic endothelium maturation and the establishment of the hematopoietic system.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Endothelium, Vascular/physiology , Gene Dosage/physiology , Hemangioblasts/physiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/physiology , Gene Expression Regulation , Mice , Mice, Knockout
11.
Stem Cells Dev ; 27(8): 524-533, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29583085

ABSTRACT

Specification of endothelial cells (ECs) into arterial, venous, and lymphatic cells is a crucial process of vascular development, and expanding our knowledge about EC specification from human pluripotent stem cells (hPSCs) will aid the design of optimal strategies for producing desired types of ECs for therapies. In our prior studies, we revealed that hPSC-derived VE-cadherin(V)+CD31+CD34+ ECs are heterogeneous and include at least three major subsets with distinct hemogenic properties: V+CD43/235a-CD73- hemogenic endothelial progenitors (HEPs), V+CD43loCD235a+73- angiogenic hematopoietic progenitors (AHPs), and V+CD43/235a-73+ non-HEPs. In this study, using angiogenesis assays, we demonstrated that ECs within these subsets have distinct endothelial colony- and tube-forming properties, proliferative and migratory properties, and endothelial nitric oxide synthase and inflammatory cytokine production potentials. Culture of isolated subsets in arterial, venous, and lymphatic conditions revealed that AHPs are skewed toward lymphatic, HEPs toward arterial, and non-HEPs toward venous differentiation in vitro. These findings suggest that selection and enhancement of production of a particular EC subset may aid in generating desirable EC populations with arterial, venous, or lymphatic properties from hPSCs.


Subject(s)
Cell Lineage/physiology , Hemangioblasts/cytology , Neovascularization, Physiologic , Pluripotent Stem Cells/cytology , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation , Cell Line , Cell Movement , Cell Proliferation , Colony-Forming Units Assay , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Hemangioblasts/physiology , Humans , Leukosialin/genetics , Leukosialin/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pluripotent Stem Cells/physiology
12.
Microbiol Mol Biol Rev ; 82(1)2018 06.
Article in English | MEDLINE | ID: mdl-29436479

ABSTRACT

Comprising the majority of leukocytes in humans, neutrophils are the first immune cells to respond to inflammatory or infectious etiologies and are crucial participants in the proper functioning of both innate and adaptive immune responses. From their initial appearance in the liver, thymus, and spleen at around the eighth week of human gestation to their generation in large numbers in the bone marrow at the end of term gestation, the differentiation of the pluripotent hematopoietic stem cell into a mature, segmented neutrophil is a highly controlled process where the transcriptional regulators C/EBP-α and C/EBP-ε play a vital role. Recent advances in neutrophil biology have clarified the life cycle of these cells and revealed striking differences between neonatal and adult neutrophils based on fetal maturation and environmental factors. Here we detail neutrophil ontogeny, granulopoiesis, and neutrophil homeostasis and highlight important differences between neonatal and adult neutrophil populations.


Subject(s)
Gene Expression Regulation/immunology , Hematopoiesis/immunology , Homeostasis/immunology , Neutrophils/physiology , Adult , Age Factors , Animals , Apoptosis/immunology , Cell Death/immunology , Cytoplasmic Granules/physiology , Extracellular Traps/immunology , Hemangioblasts/physiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Homeostasis/genetics , Humans , Immunity, Innate , Infant, Newborn , Neutrophils/immunology , Phagocytosis , Regulatory Elements, Transcriptional/immunology
13.
Reprod Toxicol ; 73: 96-104, 2017 10.
Article in English | MEDLINE | ID: mdl-28789864

ABSTRACT

Ethanol's effect on embryonic vasculogenesis and its underlying mechanism is obscure. Using VE-cadherin in situ hybridization, we found blood islands formation was inhibited in area opaca, but abnormal VE-cadherin+ cells were seen in area pellucida. We hypothesise ethanol may affect blood island progenitor cell migration and differentiation. DiI and in vitro experiments revealed ethanol inhibited cell migration, Quantitative PCR analysis revealed that ethanol exposure enhanced cell differentiation in area pellucida of HH5 chick embryos and repressed cell differentiation in area pellucida of HH8 chick embryos. By exposing to 2,2'-azobis-amidinopropane dihydrochloride, a ROS inducer, which gave a similar anti-vasculogenesis effect as ethanol and this anti-vasculogenesis effect could be reversed by vitamin C. Overall, exposing early chick embryos to ethanol represses blood island progenitor cell migration but disturbed differentiation at a different stage, so that the disorder of blood island formation occurs through excess ROS production and altered vascular-associated gene expression.


Subject(s)
Chick Embryo/drug effects , Ethanol/toxicity , Hemangioblasts/drug effects , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Chick Embryo/embryology , Embryonic Development/drug effects , Hemangioblasts/physiology , Reactive Oxygen Species/metabolism
14.
Int J Dev Biol ; 61(3-4-5): 329-335, 2017.
Article in English | MEDLINE | ID: mdl-28621430

ABSTRACT

In vertebrates, definitive hematopoietic stem cells (HSCs) first emerge in the ventral wall of the aorta in the Aorta-Gonad-Mesonephros (AGM) region of the embryo, where they differentiate from a specialized type of endothelium termed Hemogenic Endothelium (HE). While the transition from HE to hematopoietic tissue has received much experimental attention, much less is known regarding generation of HE itself. The current study investigates the emergence of the HE in the chick embryo aorta. Using the HE marker Runx1 as well as a new chicken-reactive antibody to the endothelial marker VE-Cadherin, we document the relationship between the emerging HE and surrounding tissues, particularly the coelomic epithelium (CE) and CE-derived sub-aortic mesenchyme. In addition, the fate of the CE cells was traced by electroporation of a GFP-expressing plasmid into the CE, followed by analysis using immunofluorescence and in situ hybridization. We make the novel observation that CE-derived mesenchyme transiently invades through the ventral wall of the aorta during the period of establishment of HE and just prior to the emergence of hematopoietic cell clusters in the ventral aortic wall. These observations emphasize a hitherto unappreciated dynamism in the aortic wall during the period of HE generation, and open the door to future studies regarding the role of invasive CE-derived cells during aortic hematopoiesis.


Subject(s)
Aorta/embryology , Aorta/physiology , Hemangioblasts/physiology , Hematopoiesis , Mesenchymal Stem Cells/cytology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Lineage , Chick Embryo , Core Binding Factor Alpha 2 Subunit/metabolism , Endothelium, Vascular/metabolism , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Mesoderm/metabolism , Mesonephros , Microscopy, Fluorescence
15.
Elife ; 62017 03 08.
Article in English | MEDLINE | ID: mdl-28271994

ABSTRACT

Novel regenerative therapies may stem from deeper understanding of the mechanisms governing cardiovascular lineage diversification. Using enhancer mapping and live imaging in avian embryos, and genetic lineage tracing in mice, we investigated the spatio-temporal dynamics of cardiovascular progenitor populations. We show that expression of the cardiac transcription factor Nkx2.5 marks a mesodermal population outside of the cardiac crescent in the extraembryonic and lateral plate mesoderm, with characteristics of hemogenic angioblasts. Extra-cardiac Nkx2.5 lineage progenitors migrate into the embryo and contribute to clusters of CD41+/CD45+ and RUNX1+ cells in the endocardium, the aorta-gonad-mesonephros region of the dorsal aorta and liver. We also demonstrated that ectopic expression of Nkx2.5 in chick embryos activates the hemoangiogenic gene expression program. Taken together, we identified a hemogenic angioblast cell lineage characterized by transient Nkx2.5 expression that contributes to hemogenic endothelium and endocardium, suggesting a novel role for Nkx2.5 in hemoangiogenic lineage specification and diversification.


Subject(s)
Aorta/embryology , Endocardium/embryology , Hemangioblasts/physiology , Homeobox Protein Nkx-2.5/metabolism , Animals , Chick Embryo , Mice , Spatio-Temporal Analysis
16.
Methods ; 101: 65-72, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26439174

ABSTRACT

The generation of hematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) remains a major goal for regenerative medicine and disease modeling. However, hPSC differentiation cultures produce mostly hematopoietic progenitors belonging to the embryonic HSC-independent hematopoietic program, which may not be relevant or accurate for modeling normal and disease-state adult hematopoietic processes. Through a stage-specific directed differentiation approach, it is now possible to generate exclusively definitive hematopoietic progenitors from hPSCs showing characteristics of the more developmentally advanced fetal hematopoiesis. Here, we summarize recent efforts at generating hPSC-derived definitive hematopoiesis through embryoid body differentiation under defined conditions. Embryoid bodies are generated through enzymatic dissociation of hPSCs from matrigel-coated plasticware, followed by recombinant BMP4, driving mesoderm specification. Definitive hematopoiesis is specified by a GSK3ß-inhibitor, followed by recombinant VEGF and supportive hematopoietic cytokines. The CD34+ cells obtained using this method are then suitable for hematopoietic assays for definitive hematopoietic potential.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Antigens, CD34/metabolism , Bone Morphogenetic Protein 4/physiology , Cell Culture Techniques , Cell Line , Cytokines/physiology , Embryoid Bodies/cytology , Embryoid Bodies/physiology , Hemangioblasts/physiology , Hematopoiesis , Humans , Vascular Endothelial Growth Factor A/physiology
17.
Sci China Life Sci ; 58(12): 1256-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26612042

ABSTRACT

Hematopoietic stem cells (HSCs) are specified and generated during the embryonic development and have remarkable potential to replenish the full set of blood cell lineages. Researchers have long been interested in clarifying the molecular events involved in HSC specification. Many studies have reported the development of methods for generating functional hematopoietic cells from pluripotent stem cells (PSCs-embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs)) for decades. However, the generation of HSCs with robust long-term repopulation potential remains a swingeing challenge, of which a major factor contributing to this failure is the difficulty to define the intraembryonic signals related to the specification of HSCs. Since HSCs directly derive from hemogenic endothelium, in this review, we summarize both in vivo and in vitro studies on conserved signaling pathways that control the specification of HSCs from hemogenic endothelial cells.


Subject(s)
Cell Differentiation/physiology , Hemangioblasts/physiology , Hematopoietic Stem Cells/physiology , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein 4/physiology , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Humans , Models, Biological , Receptors, Notch/physiology , Wnt Signaling Pathway/physiology
18.
Nat Commun ; 6: 7739, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26204127

ABSTRACT

Changes in cell fate and identity are essential for endothelial-to-haematopoietic transition (EHT), an embryonic process that generates the first adult populations of haematopoietic stem cells (HSCs) from hemogenic endothelial cells. Dissecting EHT regulation is a critical step towards the production of in vitro derived HSCs. Yet, we do not know how distinct endothelial and haematopoietic fates are parsed during the transition. Here we show that genes required for arterial identity function later to repress haematopoietic fate. Tissue-specific, temporally controlled, genetic loss of arterial genes (Sox17 and Notch1) during EHT results in increased production of haematopoietic cells due to loss of Sox17-mediated repression of haematopoietic transcription factors (Runx1 and Gata2). However, the increase in EHT can be abrogated by increased Notch signalling. These findings demonstrate that the endothelial haematopoietic fate switch is actively repressed in a population of endothelial cells, and that derepression of these programs augments haematopoietic output.


Subject(s)
Blood Vessels/embryology , Core Binding Factor Alpha 2 Subunit/metabolism , GATA2 Transcription Factor/metabolism , HMGB Proteins/physiology , Hemangioblasts/physiology , SOXF Transcription Factors/physiology , Animals , Female , Genes, Reporter , Hematopoiesis , Mice , Pregnancy , Receptor, Notch1/metabolism
19.
Development ; 142(6): 1050-61, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25758220

ABSTRACT

The adult blood system is established by hematopoietic stem cells (HSCs), which arise during development from an endothelial-to-hematopoietic transition of cells comprising the floor of the dorsal aorta. Expression of aortic runx1 has served as an early marker of HSC commitment in the zebrafish embryo, but recent studies have suggested that HSC specification begins during the convergence of posterior lateral plate mesoderm (PLM), well before aorta formation and runx1 transcription. Further understanding of the earliest stages of HSC specification necessitates an earlier marker of hemogenic endothelium. Studies in mice have suggested that GATA2 might function at early stages within hemogenic endothelium. Two orthologs of Gata2 exist in zebrafish: gata2a and gata2b. Here, we report that gata2b expression initiates during the convergence of PLM, becoming restricted to emerging HSCs. We observe Notch-dependent gata2b expression within the hemogenic subcompartment of the dorsal aorta that is in turn required to initiate runx1 expression. Our results indicate that Gata2b functions within hemogenic endothelium from an early stage, whereas Gata2a functions more broadly throughout the vascular system.


Subject(s)
Body Patterning/physiology , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/physiology , Hemangioblasts/physiology , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Aorta/cytology , Aorta/embryology , Bacterial Proteins , Core Binding Factor Alpha 2 Subunit/metabolism , DNA Primers/genetics , Flow Cytometry , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Luminescent Proteins , Mesoderm/embryology , Oligonucleotides, Antisense/genetics , Real-Time Polymerase Chain Reaction , Time-Lapse Imaging , Zebrafish Proteins/metabolism , Red Fluorescent Protein
20.
Biotechnol Lett ; 37(6): 1315-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25700820

ABSTRACT

Human hemangioblasts exist only during the early embryonic developmental stage thereby limiting the adult cellular source from which to obtain such cells for study. To overcome this, hemangioblast studies have focused on utilizing human embryonic stem cell (hESC) derivatives but current methods are cell-line dependent. Single cell dissociation of a hESC colony quickly led to cell death in most hESC lines due to enzyme treatment which, in turn, reduced induction potential and hemangioblast differentiation efficiency. Therefore, we sought to effectively improve the process of cell dissociation that is adaptable to various hESC lines and increase the initial induction potential of embryoid body (hEB). As a result, we determined an effective cell dissociation method through a comparison study involving various reagents which demonstrated successful dissociation regardless of cell line and enhanced hemangioblast differentiation efficiency.


Subject(s)
Cell Differentiation , Cytological Techniques/methods , Embryoid Bodies , Hemangioblasts/physiology , Human Embryonic Stem Cells/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...