Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 16(7): 1718-1734, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34143974

ABSTRACT

Across species, hematopoietic stem and progenitor cells (HSPCs) arise during embryogenesis from a specialized arterial population, termed hemogenic endothelium. Here, we describe a mechanistic role for the epigenetic regulator, Enhancer of zeste homolog-1 (Ezh1), in vertebrate HSPC production via regulation of hemogenic commitment. Loss of ezh1 in zebrafish embryos favored acquisition of hemogenic (gata2b) and HSPC (runx1) fate at the expense of the arterial program (ephrinb2a, dll4). In contrast, ezh1 overexpression blocked hematopoietic progression via maintenance of arterial gene expression. The related Polycomb group subunit, Ezh2, functioned in a non-redundant, sequential manner, whereby inhibition had no impact on arterial identity, but was capable of blocking ezh1-knockdown-associated HSPC expansion. Single-cell RNA sequencing across ezh1 genotypes revealed a dropout of ezh1+/- cells among arterial endothelium associated with positive regulation of gene transcription. Exploitation of Ezh1/2 modulation has potential functional relevance for improving in vitro HSPC differentiation from induced pluripotent stem cell sources.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Hemangioblasts/metabolism , Hematopoietic Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Embryo, Nonmammalian/metabolism , Endothelial Cells/metabolism , Gene Knockdown Techniques , Hematopoiesis , Loss of Function Mutation , Lymphocytes/metabolism , Mice , RNA-Seq , Single-Cell Analysis
2.
Dev Cell ; 56(5): 571-572, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33689767

ABSTRACT

In this issue of Developmental Cell, Weinreb et al. reveal that loss of the DEAD-box helicase Ddx41 unexpectedly triggers an R-loop-mediated sterile inflammatory cascade which drives HSPC production during embryonic development. Human studies suggest mechanistic conservation for inflammation in DDX41-associated hematologic disease, uncovering a potential route for future therapeutic intervention.


Subject(s)
DEAD-box RNA Helicases , R-Loop Structures , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA Helicases , Humans , Nucleotidyltransferases
3.
Dev Cell ; 55(2): 133-149.e6, 2020 10 26.
Article in English | MEDLINE | ID: mdl-32810442

ABSTRACT

Embryonic hematopoietic stem and progenitor cells (HSPCs) robustly proliferate while maintaining multilineage potential in vivo; however, an incomplete understanding of spatiotemporal cues governing their generation has impeded robust production from human induced pluripotent stem cells (iPSCs) in vitro. Using the zebrafish model, we demonstrate that NLRP3 inflammasome-mediated interleukin-1-beta (IL1ß) signaling drives HSPC production in response to metabolic activity. Genetic induction of active IL1ß or pharmacologic inflammasome stimulation increased HSPC number as assessed by in situ hybridization for runx1/cmyb and flow cytometry. Loss of inflammasome components, including il1b, reduced CD41+ HSPCs and prevented their expansion in response to metabolic cues. Cell ablation studies indicated that macrophages were essential for initial inflammasome stimulation of Il1rl1+ HSPCs. Significantly, in human iPSC-derived hemogenic precursors, transient inflammasome stimulation increased multilineage hematopoietic colony-forming units and T cell progenitors. This work establishes the inflammasome as a conserved metabolic sensor that expands HSPC production in vivo and in vitro.


Subject(s)
Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Inflammasomes/metabolism , Animals , Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development/physiology , Hematopoiesis/physiology , Humans , Zebrafish/embryology
4.
Nat Biotechnol ; 37(8): 962, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31312048

ABSTRACT

In the version of this article initially published, the second NIH grant "R24-DK49216" to author George Q. Daley contained an error. The grant number should have read U54DK110805. The error has been corrected in the HTML and PDF versions of the article.

5.
Nat Biotechnol ; 37(7): 810-818, 2019 07.
Article in English | MEDLINE | ID: mdl-31267104

ABSTRACT

A major challenge for stem cell engineering is achieving a holistic understanding of the molecular networks and biological processes governing cell differentiation. To address this challenge, we describe a computational approach that combines gene expression analysis, previous knowledge from proteomic pathway informatics and cell signaling models to delineate key transitional states of differentiating cells at high resolution. Our network models connect sparse gene signatures with corresponding, yet disparate, biological processes to uncover molecular mechanisms governing cell fate transitions. This approach builds on our earlier CellNet and recent trajectory-defining algorithms, as illustrated by our analysis of hematopoietic specification along the erythroid lineage, which reveals a role for the EGF receptor family member, ErbB4, as an important mediator of blood development. We experimentally validate this prediction and perturb the pathway to improve erythroid maturation from human pluripotent stem cells. These results exploit an integrative systems perspective to identify new regulatory processes and nodes useful in cell engineering.


Subject(s)
Cell Engineering , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Systems Biology/methods , Algorithms , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Computational Biology/methods , Erythrocytes , Erythropoiesis , Flow Cytometry , Gene Expression Regulation , Gene Regulatory Networks , Humans , Mice , Receptor, ErbB-4/metabolism , Signal Transduction , Zebrafish
6.
Stem Cells ; 34(2): 431-44, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26418893

ABSTRACT

Adult-repopulating hematopoietic stem cells (HSCs) emerge in low numbers in the midgestation mouse embryo from a subset of arterial endothelium, through an endothelial-to-hematopoietic transition. HSC-producing arterial hemogenic endothelium relies on the establishment of embryonic blood flow and arterial identity, and requires ß-catenin signaling. Specified prior to and during the formation of these initial HSCs are thousands of yolk sac-derived erythro-myeloid progenitors (EMPs). EMPs ensure embryonic survival prior to the establishment of a permanent hematopoietic system, and provide subsets of long-lived tissue macrophages. While an endothelial origin for these HSC-independent definitive progenitors is also accepted, the spatial location and temporal output of yolk sac hemogenic endothelium over developmental time remain undefined. We performed a spatiotemporal analysis of EMP emergence, and document the morphological steps of the endothelial-to-hematopoietic transition. Emergence of rounded EMPs from polygonal clusters of Kit(+) cells initiates prior to the establishment of arborized arterial and venous vasculature in the yolk sac. Interestingly, Kit(+) polygonal clusters are detected in both arterial and venous vessels after remodeling. To determine whether there are similar mechanisms regulating the specification of EMPs with other angiogenic signals regulating adult-repopulating HSCs, we investigated the role of embryonic blood flow and Wnt/ß-catenin signaling during EMP emergence. In embryos lacking a functional circulation, rounded Kit(+) EMPs still fully emerge from unremodeled yolk sac vasculature. In contrast, canonical Wnt signaling appears to be a common mechanism regulating hematopoietic emergence from hemogenic endothelium. These data illustrate the heterogeneity in hematopoietic output and spatiotemporal regulation of primary embryonic hemogenic endothelium.


Subject(s)
Endothelium, Vascular/metabolism , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells/metabolism , Wnt Proteins/metabolism , Yolk Sac/metabolism , Animals , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Mice , Mice, Transgenic , Yolk Sac/blood supply , Yolk Sac/cytology
7.
Cell Rep ; 11(12): 1892-904, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26095363

ABSTRACT

Hematopoietic potential arises in mammalian embryos before adult-repopulating hematopoietic stem cells (HSCs). At embryonic day 9.5 (E9.5), we show the first murine definitive erythro-myeloid progenitors (EMPs) have an immunophenotype distinct from primitive hematopoietic progenitors, maturing megakaryocytes and macrophages, and rare B cell potential. EMPs emerge in the yolk sac with erythroid and broad myeloid, but not lymphoid, potential. EMPs migrate to the fetal liver and rapidly differentiate, including production of circulating neutrophils by E11.5. Although the surface markers, transcription factors, and lineage potential associated with EMPs overlap with those found in adult definitive hematopoiesis, they are present in unique combinations or proportions that result in a specialized definitive embryonic progenitor. Furthermore, we find that embryonic stem cell (ESC)-derived hematopoiesis recapitulates early yolk sac hematopoiesis, including primitive, EMP, and rare B cell potential. EMPs do not have long-term potential when transplanted in immunocompromised adults, but they can provide transient adult-like RBC reconstitution.


Subject(s)
Embryonic Development/genetics , Embryonic Stem Cells , Hematopoiesis , Hematopoietic Stem Cells , Animals , Blood Cells/cytology , Cell Lineage , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , Yolk Sac/cytology , Yolk Sac/growth & development
8.
Semin Immunol ; 27(6): 379-87, 2015 12.
Article in English | MEDLINE | ID: mdl-27021646

ABSTRACT

The paradigm that all blood cells are derived from hematopoietic stem cells (HSCs) has been challenged by two findings. First, there are tissue-resident hematopoietic cells, including subsets of macrophages that are not replenished by adult HSCs, but instead are maintained by self-renewal of fetal-derived cells. Second, during embryogenesis, there is a conserved program of HSC-independent hematopoiesis that precedes HSC function and is required for embryonic survival. The presence of waves of HSC-independent hematopoiesis as well as fetal HSCs raises questions about the origin of fetal-derived adult tissue-resident macrophages. In the murine embryo, historical examination of embryonic macrophage and monocyte populations combined with recent reports utilizing genetic lineage-tracing approaches has led to a model of macrophage ontogeny that can be integrated with existing models of hematopoietic ontogeny. The first wave of hematopoiesis contains primitive erythroid, megakaryocyte and macrophage progenitors that arise in the yolk sac, and these macrophage progenitors are the source of early macrophages throughout the embryo, including the liver. A second wave of multipotential erythro-myeloid progenitors (EMPs) also arises in the yolk sac. EMPs colonize the fetal liver, initiating myelopoiesis and forming macrophages. Lineage tracing indicates that this second wave of macrophages are distributed in most fetal tissues, although not appreciably in the brain. Thus, fetal-derived adult tissue-resident macrophages, other than microglia, appear to predominately derive from EMPs. While HSCs emerge at midgestation and colonize the fetal liver, the relative contribution of fetal HSCs to tissue macrophages at later stages of development is unclear. The inclusion of macrophage potential in multiple waves of hematopoiesis is consistent with reports of their functional roles throughout development in innate immunity, phagocytosis, and tissue morphogenesis and remodeling. Understanding the influences of developmental origin, as well as local tissue-specific signals, will be necessary to fully decode the diverse functions and responses of tissue-resident macrophages.


Subject(s)
Hematopoiesis , Macrophages/cytology , Animals , Cell Lineage , Humans , Models, Immunological
9.
Blood Cells Mol Dis ; 51(4): 220-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24095199

ABSTRACT

Erythro-myeloid progenitors (EMP) serve as a major source of hematopoiesis in the developing conceptus prior to the formation of a permanent blood system. In this review, we summarize the current knowledge regarding the emergence, fate, and potential of this hematopoietic stem cell (HSC)-independent wave of hematopoietic progenitors, focusing on the murine embryo as a model system. A better understanding of the temporal and spatial control of hematopoietic emergence in the embryo will ultimately improve our ability to derive hematopoietic stem and progenitor cells from embryonic stem cells and induced pluripotent stem cells to serve therapeutic purposes.


Subject(s)
Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/physiology , Hematopoiesis/physiology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/physiology , Animals , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans
10.
Blood ; 121(6): e5-e13, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23243273

ABSTRACT

Erythroid ontogeny is characterized by overlapping waves of primitive and definitive erythroid lineages that share many morphologic features during terminal maturation but have marked differences in cell size and globin expression. In the present study, we compared global gene expression in primitive, fetal definitive, and adult definitive erythroid cells at morphologically equivalent stages of maturation purified from embryonic, fetal, and adult mice. Surprisingly, most transcriptional complexity in erythroid precursors is already present by the proerythroblast stage. Transcript levels are markedly modulated during terminal erythroid maturation, but housekeeping genes are not preferentially lost. Although primitive and definitive erythroid lineages share a large set of nonhousekeeping genes, annotation of lineage-restricted genes shows that alternate gene usage occurs within shared functional categories, as exemplified by the selective expression of aquaporins 3 and 8 in primitive erythroblasts and aquaporins 1 and 9 in adult definitive erythroblasts. Consistent with the known functions of Aqp3 and Aqp8 as H2O2 transporters, primitive, but not definitive, erythroblasts preferentially accumulate reactive oxygen species after exogenous H2O2 exposure. We have created a user-friendly Web site (http://www.cbil.upenn.edu/ErythronDB) to make these global expression data readily accessible and amenable to complex search strategies by the scientific community.


Subject(s)
Erythroid Cells/metabolism , Erythropoiesis/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Animals , Aquaporin 1/genetics , Aquaporin 3/genetics , Aquaporins/genetics , Cell Lineage/genetics , Cells, Cultured , Erythroblasts/metabolism , Erythrocytes/metabolism , Female , Hematopoietic System/cytology , Hematopoietic System/embryology , Hematopoietic System/growth & development , Mice , Mice, Inbred ICR , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
Blood ; 117(17): 4600-8, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21378272

ABSTRACT

A transient erythromyeloid wave of definitive hematopoietic progenitors (erythroid/myeloid progenitors [EMPs]) emerges in the yolk sac beginning at embryonic day 8.25 (E8.25) and colonizes the liver by E10.5, before adult-repopulating hematopoietic stem cells. At E11.5, we observe all maturational stages of erythroid precursors in the liver and the first definitive erythrocytes in the circulation. These early fetal liver erythroblasts express predominantly adult ß-globins and the definitive erythroid-specific transcriptional modifiers c-myb, Sox6, and Bcl11A. Surprisingly, they also express low levels of "embryonic" ßH1-, but not εy-, globin transcripts. Consistent with these results, RNA polymerase and highly modified histones are found associated with ßH1- and adult globin, but not εy-globin, genes. E11.5 definitive proerythroblasts from mice transgenic for the human ß-globin locus, like human fetal erythroblasts, express predominately human γ-, low ß-, and no ε-globin transcripts. Significantly, E9.5 yolk sac-derived EMPs cultured in vitro have similar murine and human transgenic globin expression patterns. Later liver proerythroblasts express low levels of γ-globin, while adult marrow proerythroblasts express only ß-globin transcripts. We conclude that yolk sac-derived EMPs, the first of 2 origins of definitive erythropoiesis, express a unique pattern of globin genes as they generate the first definitive erythrocytes in the liver of the mammalian embryo.


Subject(s)
Erythroid Cells/cytology , Erythropoiesis/physiology , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/cytology , Liver , beta-Globins/genetics , Animals , Animals, Outbred Strains , Cell Lineage/physiology , Erythroblasts/cytology , Erythrocytes/cytology , GATA1 Transcription Factor/genetics , Hematopoietic Stem Cells/physiology , Humans , Kruppel-Like Transcription Factors/genetics , Liver/cytology , Liver/embryology , Liver/physiology , Mammals , Mice , Mice, Transgenic , Yolk Sac/physiology
12.
Exp Hematol ; 39(4): 434-45, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21291953

ABSTRACT

OBJECTIVE: Clastogenic injury of the erythroid lineage results in anemia, reticulocytopenia, and transient appearance of micronucleated reticulocytes. However, the micronucleated reticulocyte dose-response in murine models is only linear to 2 Gy total body irradiation and paradoxically decreases at higher exposures, suggesting complex radiation effects on erythroid intermediates. To better understand this phenomenon, we investigated the kinetics and apoptotic response of the erythron to sublethal radiation injury. MATERIALS AND METHODS: We analyzed the response to 1 and 4 Gy total body irradiation of erythroid progenitors and precursors using colony assays and imaging flow cytometry, respectively. We also investigated cell cycling and apoptotic gene expression of the steady-state erythron. RESULTS: After 1 Gy total body irradiation, erythroid progenitors and precursors were partially depleted. In contrast, essentially all bone marrow erythroid progenitors and precursors were lost within 2 days after 4 Gy irradiation. Imaging flow cytometry analysis revealed preferential loss of phenotypic erythroid colony-forming units and proerythroblasts immediately after sublethal irradiation. Furthermore, these populations underwent radiation-induced apoptosis, without changes in steady-state cellular proliferation, at much higher frequencies than later-stage erythroid precursors. Primary erythroid precursor maturation is associated with marked Bcl-xL upregulation and Bax and Bid downregulation. CONCLUSIONS: Micronucleated reticulocyte loss after higher sublethal radiation exposures results from rapid depletion of erythroid progenitors and precursors. This injury reveals that erythroid colony-forming units and proerythroblasts constitute a particularly proapoptotic compartment within the erythron. We conclude that the functional transition of primary proerythroblasts to later-stage erythroid precursors is characterized by a shift from a proapoptotic to an antiapoptotic phenotype.


Subject(s)
Cell Differentiation/radiation effects , Erythroid Cells/radiation effects , Radiation Injuries, Experimental/physiopathology , Animals , Apoptosis/radiation effects , BH3 Interacting Domain Death Agonist Protein/genetics , Cell Cycle/radiation effects , Dose-Response Relationship, Radiation , Erythroblasts/cytology , Erythroblasts/metabolism , Erythroblasts/radiation effects , Erythroid Cells/cytology , Erythroid Cells/metabolism , Flow Cytometry , Gene Expression Regulation/radiation effects , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Whole-Body Irradiation , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
13.
Blood ; 117(9): 2708-17, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21127173

ABSTRACT

In the hematopoietic hierarchy, only stem cells are thought to be capable of long-term self-renewal. Erythroid progenitors derived from fetal or adult mammalian hematopoietic tissues are capable of short-term, or restricted (10(2)- to 10(5)-fold), ex vivo expansion in the presence of erythropoietin, stem cell factor, and dexamethasone. Here, we report that primary erythroid precursors derived from early mouse embryos are capable of extensive (10(6)- to 10(60)-fold) ex vivo proliferation. These cells morphologically, immunophenotypically, and functionally resemble proerythroblasts, maintaining both cytokine dependence and the potential, despite prolonged culture, to generate enucleated erythrocytes after 3-4 maturational cell divisions. This capacity for extensive erythroblast self-renewal is temporally associated with the emergence of definitive erythropoiesis in the yolk sac and its transition to the fetal liver. In contrast, hematopoietic stem cell-derived definitive erythropoiesis in the adult is associated almost exclusively with restricted ex vivo self-renewal. Primary primitive erythroid precursors, which lack significant expression of Kit and glucocorticoid receptors, lack ex vivo self-renewal capacity. Extensively self-renewing erythroblasts, despite their near complete maturity within the hematopoietic hierarchy, may ultimately serve as a renewable source of red cells for transfusion therapy.


Subject(s)
Cell Differentiation , Erythroblasts/cytology , Fetus/cytology , Mammals/embryology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/pharmacology , Dexamethasone/pharmacology , Erythroblasts/drug effects , Erythroblasts/metabolism , Humans , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Yolk Sac/cytology , Yolk Sac/drug effects , Yolk Sac/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...