Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Immunol ; 21(12): 1574-1584, 2020 12.
Article in English | MEDLINE | ID: mdl-33077975

ABSTRACT

A classical view of blood cell development is that multipotent hematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. Lin-c-Kit+Sca-1+Flt3+ cells, termed lymphoid-primed multipotent progenitors (LMPPs), have lost megakaryocyte and erythroid potential but are heterogeneous in their fate. Here, through single-cell RNA sequencing, we identify the expression of Dach1 and associated genes in this fraction as being coexpressed with myeloid/stem genes but inversely correlated with lymphoid genes. Through generation of Dach1-GFP reporter mice, we identify a transcriptionally and functionally unique Dach1-GFP- subpopulation within LMPPs with lymphoid potential with low to negligible classic myeloid potential. We term these 'lymphoid-primed progenitors' (LPPs). These findings define an early definitive branch point of lymphoid development in hematopoiesis and a means for prospective isolation of LPPs.


Subject(s)
Biomarkers , Eye Proteins/metabolism , Genomics , Lymphoid Progenitor Cells/metabolism , Single-Cell Analysis , Animals , Cells, Cultured , Computational Biology/methods , Eye Proteins/genetics , Gene Expression Profiling , Genomics/methods , Hematopoiesis/genetics , High-Throughput Nucleotide Sequencing , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Knockout , Mice, Transgenic , Proteomics , Single-Cell Analysis/methods
2.
Nature ; 614(7947): 343-348, 2023 02.
Article in English | MEDLINE | ID: mdl-36697821

ABSTRACT

Transcriptional enhancer elements are responsible for orchestrating the temporal and spatial control over gene expression that is crucial for programming cell identity during development1-3. Here we describe a novel enhancer element that is important for regulating the expression of Prox1 in lymphatic endothelial cells. This evolutionarily conserved enhancer is bound by key lymphatic transcriptional regulators including GATA2, FOXC2, NFATC1 and PROX1. Genome editing of the enhancer to remove five nucleotides encompassing the GATA2-binding site resulted in perinatal death of homozygous mutant mice due to profound lymphatic vascular defects. Lymphatic endothelial cells in enhancer mutant mice exhibited reduced expression of genes characteristic of lymphatic endothelial cell identity and increased expression of genes characteristic of haemogenic endothelium, and acquired the capacity to generate haematopoietic cells. These data not only reveal a transcriptional enhancer element important for regulating Prox1 expression and lymphatic endothelial cell identity but also demonstrate that the lymphatic endothelium has haemogenic capacity, ordinarily repressed by Prox1.


Subject(s)
Endothelial Cells , Enhancer Elements, Genetic , Hematopoiesis , Lymphatic Vessels , Animals , Mice , Endothelial Cells/metabolism , Enhancer Elements, Genetic/genetics , Hematopoiesis/genetics , Homeodomain Proteins/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Transcription Factors/metabolism
3.
Blood ; 143(4): 342-356, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-37922495

ABSTRACT

ABSTRACT: Glycoprotein Ibα (GPIbα) is expressed on the surface of platelets and megakaryocytes (MKs) and anchored to the membrane skeleton by filamin A (flnA). Although GPIb and flnA have fundamental roles in platelet biogenesis, the nature of this interaction in megakaryocyte biology remains ill-defined. We generated a mouse model expressing either human wild-type (WT) GPIbα (hGPIbαWT) or a flnA-binding mutant (hGPIbαFW) and lacking endogenous mouse GPIbα. Mice expressing the mutant GPIbα transgene exhibited macrothrombocytopenia with preserved GPIb surface expression. Platelet clearance was normal and differentiation of MKs to proplatelets was unimpaired in hGPIbαFW mice. The most striking abnormalities in hGPIbαFW MKs were the defective formation of the demarcation membrane system (DMS) and the redistribution of flnA from the cytoplasm to the peripheral margin of MKs. These abnormalities led to disorganized internal MK membranes and the generation of enlarged megakaryocyte membrane buds. The defective flnA-GPIbα interaction also resulted in misdirected release of buds away from the vasculature into bone marrow interstitium. Restoring the linkage between flnA and GPIbα corrected the flnA redistribution within MKs and DMS ultrastructural defects as well as restored normal bud size and release into sinusoids. These studies define a new mechanism of macrothrombocytopenia resulting from dysregulated MK budding. The link between flnA and GPIbα is not essential for the MK budding process, however, it plays a major role in regulating the structure of the DMS, bud morphogenesis, and the localized release of buds into the circulation.


Subject(s)
Megakaryocytes , Platelet Glycoprotein GPIb-IX Complex , Thrombocytopenia , Animals , Humans , Mice , Blood Platelets/metabolism , Cytoplasm/metabolism , Filamins/genetics , Filamins/metabolism , Megakaryocytes/metabolism , Morphogenesis , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombocytopenia/genetics , Thrombocytopenia/metabolism
4.
Genomics ; 116(2): 110793, 2024 03.
Article in English | MEDLINE | ID: mdl-38220132

ABSTRACT

Single-cell RNA sequencing (scRNA-Seq) has emerged as a powerful tool for understanding cellular heterogeneity and function. However the choice of sample multiplexing reagents can impact data quality and experimental outcomes. In this study, we compared various multiplexing reagents, including MULTI-Seq, Hashtag antibody, and CellPlex, across diverse sample types such as human peripheral blood mononuclear cells (PBMCs), mouse embryonic brain and patient-derived xenografts (PDXs). We found that all multiplexing reagents worked well in cell types robust to ex vivo manipulation but suffered from signal-to-noise issues in more delicate sample types. We compared multiple demultiplexing algorithms which differed in performance depending on data quality. We find that minor improvements to laboratory workflows such as titration and rapid processing are critical to optimal performance. We also compared the performance of fixed scRNA-Seq kits and highlight the advantages of the Parse Biosciences kit for fragile samples. Highly multiplexed scRNA-Seq experiments require more sequencing resources, therefore we evaluated CRISPR-based destruction of non-informative genes to enhance sequencing value. Our comprehensive analysis provides insights into the selection of appropriate sample multiplexing reagents and protocols for scRNA-Seq experiments, facilitating more accurate and cost-effective studies.


Subject(s)
Leukocytes, Mononuclear , Single-Cell Analysis , Humans , Animals , Mice , RNA-Seq , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Algorithms , Gene Expression Profiling/methods
5.
Blood ; 139(15): 2355-2360, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35148538

ABSTRACT

Whether increasing platelet counts in fetal and neonatal alloimmune thrombocytopenia (FNAIT) is effective at preventing intracerebral hemorrhage (ICH) has been a subject of debate. The crux of the matter has been whether thrombocytopenia is the major driver of ICH in diseases such as FNAIT. We recently demonstrated in mice that severe thrombocytopenia was sufficient to drive ICH in utero and in early neonatal life. It remains unclear what degree of thrombocytopenia is required to drive ICH and for how long after birth thrombocytopenia can cause ICH. By inducing a thrombocytopenic range, we demonstrate that there is a large buffer zone of mild thrombocytopenia that does not result in ICH, that ICH becomes probabilistic at 40% of the normal platelet number, and that ICH becomes fully penetrant below 10% of the normal platelet number. We also demonstrate that although the neonatal mouse is susceptible to thrombocytopenia-induced ICH, this sensitivity is rapidly lost between postnatal days 7 and 14. These findings provide important insights into the risk of in utero ICH with varying degrees of thrombocytopenia and into defining the developmental high-risk period for thrombocytopenia-driven ICH in a mouse model of FNAIT.


Subject(s)
Antigens, Human Platelet , Thrombocytopenia, Neonatal Alloimmune , Animals , Cerebral Hemorrhage , Female , Fetus , Humans , Mice , Pregnancy , Prenatal Care
6.
Blood ; 138(10): 885-897, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34189583

ABSTRACT

Intracerebral hemorrhage (ICH) has a devastating impact on the neonatal population. Whether thrombocytopenia is sufficient to cause ICH in neonates is still being debated. In this study, we comprehensively investigated the consequences of severe thrombocytopenia on the integrity of the cerebral vasculature by using 2 orthogonal approaches: by studying embryogenesis in the Nfe2-/- mouse line and by using biologics (anti-GP1Bα antibodies) to induce severe thrombocytopenia at defined times during development. By using a mouse model, we acquired data demonstrating that platelets are required throughout fetal development and into neonatal life for maintaining the integrity of the cerebral vasculature to prevent hemorrhage and that the location of cerebral hemorrhage is dependent on when thrombocytopenia occurs during development. Importantly, this study demonstrates that fetal and neonatal thrombocytopenia-associated ICH occurs within regions of the brain which, in humans, could lead to neurologic damage.


Subject(s)
Cerebral Hemorrhage/metabolism , Fetus/metabolism , Thrombocytopenia/metabolism , Animals , Animals, Newborn , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/pathology , Fetus/pathology , Mice , Mice, Knockout , Patient Acuity , Thrombocytopenia/genetics , Thrombocytopenia/pathology
7.
Immunol Cell Biol ; 99(10): 1006-1010, 2021 11.
Article in English | MEDLINE | ID: mdl-34664303

ABSTRACT

We hypothesize that thrombosis with thrombocytopenia syndrome recently described after administration of adenovirus-vectored vaccines for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) occurs as a result of the unique properties of the adenovirus vectors, which can have widespread biodistribution throughout the body. The antigen is delivered to megakaryocyte cells, which act as part of the primary immune system and distribute the antigen within progeny platelets, also a key component of the immune system. The interaction of the antigen induces preformed antiplatelet factor 4 (PF4) antibodies to bind to PF4-heparan sulfate complexes in the absence of exogenous heparin, at sites where the heparan sulfate concentration in the vascular glycocalyx is optimal for complex formation, causing thrombosis and thrombocytopenia as observed clinically. This hypothesis is testable in cell culture and animal models, and potentially in vivo, and if proven correct has significant implications for vaccine development and our understanding of the links between the coagulation and immune systems.


Subject(s)
COVID-19 , Thrombocytopenia , Thrombosis , Vaccines , Adenoviridae , Animals , Humans , SARS-CoV-2 , Tissue Distribution , Vaccination
8.
Genes Dev ; 25(3): 251-62, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21245161

ABSTRACT

Although many genes are known to be critical for early hematopoiesis in the embryo, it remains unclear whether distinct regulatory pathways exist to control hematopoietic specification versus hematopoietic stem cell (HSC) emergence and function. Due to their interaction with key regulators of hematopoietic commitment, particular interest has focused on the role of the ETS family of transcription factors; of these, ERG is predicted to play an important role in the initiation of hematopoiesis, yet we do not know if or when ERG is required. Using in vitro and in vivo models of hematopoiesis and HSC development, we provide strong evidence that ERG is at the center of a distinct regulatory program that is not required for hematopoietic specification or differentiation but is critical for HSC maintenance during embryonic development. We show that, from the fetal period, ERG acts as a direct upstream regulator of Gata2 and Runx1 gene activity. Without ERG, physiological HSC maintenance fails, leading to the rapid exhaustion of definitive hematopoiesis.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Oncogene Proteins/metabolism , Animals , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/metabolism , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Oncogene Proteins/genetics , Transcription Factors , Transcriptional Regulator ERG
9.
Blood ; 126(6): 807-16, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-25995324

ABSTRACT

The thrombopoietic environment of the neonate is established during prenatal life; therefore, a comprehensive understanding of platelet-forming cell development during embryogenesis is critical to understanding the etiology of early-onset thrombocytopenia. The recent discovery that the first platelet-forming cells of the conceptus are not megakaryocytes (MKs) but diploid platelet-forming cells (DPFCs) revealed a previously unappreciated complexity in thrombopoiesis. This raises important questions, including the following. When do conventional MKs appear? Do pathogenic genetic lesions of adult MKs affect DPFCs? What role does myeloproliferative leukemia virus (MPL), a key regulator of adult megakaryopoiesis, play in prenatal platelet-forming lineages? We performed a comprehensive study to determine the spatial and temporal appearance of prenatal platelet-forming lineages. We demonstrate that DPFCs originate in the yolk sac and then rapidly migrate to other extra- and intraembryonic tissues. Using gene disruption models of Gata1 and Nfe2, we demonstrate that perturbing essential adult MK genes causes an analogous phenotype in the early embryo before the onset of hematopoietic stem/progenitor cell-driven (definitive) hematopoiesis. Finally, we present the surprising finding that DPFC and MK commitment from their respective precursors is MPL independent in vivo but that completion of MK differentiation and establishment of the prenatal platelet mass is dependent on MPL expression.


Subject(s)
Blood Platelets/metabolism , Gene Expression Regulation, Developmental , Megakaryocytes/metabolism , Receptors, Thrombopoietin/genetics , Thrombopoiesis/genetics , Yolk Sac/metabolism , Animals , Blood Platelets/cytology , Cell Differentiation , Cell Lineage/genetics , Embryonic Development/genetics , GATA1 Transcription Factor/deficiency , GATA1 Transcription Factor/genetics , Gene Deletion , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Megakaryocytes/cytology , Mice , Mice, Knockout , NF-E2 Transcription Factor, p45 Subunit/deficiency , NF-E2 Transcription Factor, p45 Subunit/genetics , Receptors, Thrombopoietin/metabolism , Transcription, Genetic , Yolk Sac/cytology , Yolk Sac/growth & development
10.
Blood ; 124(17): 2725-9, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25079356

ABSTRACT

In this study, we test the assumption that the hematopoietic progenitor/colony-forming cells of the embryonic yolk sac (YS), which are endowed with megakaryocytic potential, differentiate into the first platelet-forming cells in vivo. We demonstrate that from embryonic day (E) 8.5 all megakaryocyte (MK) colony-forming cells belong to the conventional hematopoietic progenitor cell (HPC) compartment. Although these cells are indeed capable of generating polyploid MKs, they are not the source of the first platelet-forming cells. We show that proplatelet formation first occurs in a unique and previously unrecognized lineage of diploid platelet-forming cells, which develop within the YS in parallel to HPCs but can be specified in the E8.5 Runx1-null embryo despite the absence of the progenitor cell lineage.


Subject(s)
Cell Lineage/genetics , Diploidy , Embryo, Mammalian/metabolism , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/metabolism , Polyploidy , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Megakaryocyte Progenitor Cells/cytology , Megakaryocytes/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Time Factors , Transcriptome , Yolk Sac/cytology , Yolk Sac/embryology , Yolk Sac/metabolism
11.
Blood ; 131(2): 158-159, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29326349
12.
Development ; 138(6): 1017-31, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21343360

ABSTRACT

Definitive hematopoietic stem cells (HSCs) lie at the foundation of the adult hematopoietic system and provide an organism throughout its life with all blood cell types. Several tissues demonstrate hematopoietic activity at early stages of embryonic development, but which tissue is the primary source of these important cells and what are the early embryonic ancestors of definitive HSCs? Here, we review recent advances in the field of HSC research that have shed light on such questions, while setting them into a historical context, and discuss key issues currently circulating in this field.


Subject(s)
Embryonic Development/physiology , Hematopoietic Stem Cells/physiology , Hematopoietic System/embryology , Adult , Animals , Embryo Research , Embryo, Mammalian , Hematopoiesis/physiology , Humans , Models, Biological
13.
Stem Cell Reports ; 19(8): 1189-1204, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39094562

ABSTRACT

It has been proposed that adult hematopoiesis is sustained by multipotent progenitors (MPPs) specified during embryogenesis. Adult-like hematopoietic stem cell (HSC) and MPP immunophenotypes are present in the fetus, but knowledge of their functional capacity is incomplete. We found that fetal MPP populations were functionally similar to adult cells, albeit with some differences in lymphoid output. Clonal assessment revealed that lineage biases arose from differences in patterns of single-/bi-lineage differentiation. Long-term (LT)- and short-term (ST)-HSC populations were distinguished from MPPs according to capacity for clonal multilineage differentiation. We discovered that a large cohort of long-term repopulating units (LT-RUs) resides within the ST-HSC population; a significant portion of these were labeled using Flt3-cre. This finding has two implications: (1) use of the CD150+ LT-HSC immunophenotype alone will significantly underestimate the size and diversity of the LT-RU pool and (2) LT-RUs in the ST-HSC population have the attributes required to persist into adulthood.


Subject(s)
Cell Lineage , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Animals , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Cell Differentiation , Fetus/cytology , Immunophenotyping , Hematopoiesis , Clone Cells/cytology
14.
Blood ; 127(19): 2267-8, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27206663
15.
Nat Cell Biol ; 24(8): 1211-1225, 2022 08.
Article in English | MEDLINE | ID: mdl-35902769

ABSTRACT

Mouse haematopoietic stem cells (HSCs) first emerge at embryonic day 10.5 (E10.5), on the ventral surface of the dorsal aorta, by endothelial-to-haematopoietic transition. We investigated whether mesenchymal stem cells, which provide an essential niche for long-term HSCs (LT-HSCs) in the bone marrow, reside in the aorta-gonad-mesonephros and contribute to the development of the dorsal aorta and endothelial-to-haematopoietic transition. Here we show that mesoderm-derived PDGFRA+ stromal cells (Mesp1der PSCs) contribute to the haemogenic endothelium of the dorsal aorta and populate the E10.5-E11.5 aorta-gonad-mesonephros but by E13.5 were replaced by neural-crest-derived PSCs (Wnt1der PSCs). Co-aggregating non-haemogenic endothelial cells with Mesp1der PSCs but not Wnt1der PSCs resulted in activation of a haematopoietic transcriptional programme in endothelial cells and generation of LT-HSCs. Dose-dependent inhibition of PDGFRA or BMP, WNT and NOTCH signalling interrupted this reprogramming event. Together, aorta-gonad-mesonephros Mesp1der PSCs could potentially be harnessed to manufacture LT-HSCs from endothelium.


Subject(s)
Hemangioblasts , Mesonephros , Animals , Aorta , Hematopoiesis/genetics , Hematopoietic Stem Cells , Mesoderm , Mice
16.
Nat Cell Biol ; 23(3): 219-231, 2021 03.
Article in English | MEDLINE | ID: mdl-33649477

ABSTRACT

Regulation of haematopoietic stem and progenitor cell (HSPC) fate is crucial during homeostasis and under stress conditions. Here we examine the aetiology of the Flt3 ligand (Flt3L)-mediated increase of type 1 conventional dendritic cells (cDC1s). Using cellular barcoding we demonstrate this occurs through selective clonal expansion of HSPCs that are primed to produce cDC1s and not through activation of cDC1 fate by other HSPCs. In particular, multi/oligo-potent clones selectively amplify their cDC1 output, without compromising the production of other lineages, via a process we term tuning. We then develop Divi-Seq to simultaneously profile the division history, surface phenotype and transcriptome of individual HSPCs. We discover that Flt3L-responsive HSPCs maintain a proliferative 'early progenitor'-like state, leading to the selective expansion of multiple transitional cDC1-primed progenitor stages that are marked by Irf8 expression. These findings define the mechanistic action of Flt3L through clonal tuning, which has important implications for other models of 'emergency' haematopoiesis.


Subject(s)
Cell Proliferation/drug effects , Dendritic Cells/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Membrane Proteins/pharmacology , RNA-Seq , Single-Cell Analysis , Transcriptome/drug effects , Animals , Cell Lineage , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
17.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32706855

ABSTRACT

How platelets are produced by megakaryocytes in vivo remains controversial despite more than a century of investigation. Megakaryocytes readily produce proplatelet structures in vitro; however, visualization of platelet release from proplatelets in vivo has remained elusive. We show that within the native prenatal and adult environments, the frequency and rate of proplatelet formation is incompatible with the physiological demands of platelet replacement. We resolve this inconsistency by performing in-depth analysis of plasma membrane budding, a cellular process that has previously been dismissed as a source of platelet production. Our studies demonstrate that membrane budding results in the sustained release of platelets directly into the peripheral circulation during both fetal and adult life without induction of cell death or proplatelet formation. In support of this model, we demonstrate that in mice deficient for NF-E2 (the thrombopoietic master regulator), the absence of membrane budding correlates with failure of in vivo platelet production. Accordingly, we propose that membrane budding, rather than proplatelet formation, supplies the majority of the platelet biomass.


Subject(s)
Blood Platelets/cytology , Cell Membrane/metabolism , Animals , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Bone Marrow Cells/cytology , Cell Lineage , Cell Membrane/ultrastructure , Databases as Topic , Embryo, Mammalian/cytology , Fetus/cytology , Gene Expression Regulation , Imaging, Three-Dimensional , Integrases/metabolism , Liver/embryology , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice, Inbred C57BL , Ploidies , Reproducibility of Results , Skull/cytology
18.
Genesis ; 47(5): 346-51, 2009 May.
Article in English | MEDLINE | ID: mdl-19370754

ABSTRACT

Cellular reaggregation methods have been used to generate in vitro organotypic cultures as a means to elucidate the cellular and molecular requirements of organogenesis. However, reproducibility from experiment to experiment has remained problematic and furthermore, current protocols do not support reaggregation of many important tissues. Here, using the thymus as a model organ, we present a novel reaggregation method termed "compaction reaggregation" that offers improved kinetics of reaggregation and greatly improved efficiency. Using compaction reaggregation we have been able to reaggregate the aorta-gonad- mesonephros region, a tissue that previously proved refractory to commonly used reaggregation methods, enabling the study of hematopoietic stem cell emergence and expansion. Additionally, compaction reaggregation permits the juxtaposition of different cell layers within the aggregated structure thus providing the means to study inductive interactions between different cell populations in vitro.


Subject(s)
Cell Aggregation/physiology , Cell Lineage , Thymus Gland/cytology , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Keratins/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Organ Culture Techniques/methods , Pregnancy , Thymus Gland/embryology , Thymus Gland/metabolism
19.
Stem Cell Reports ; 12(5): 1056-1068, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30956115

ABSTRACT

Definitive hematopoietic stem cells (HSCs) first emerge in the aorta-gonad-mesonephros (AGM) region in both mice and humans. An ex vivo culture approach has enabled recapitulation and analysis of murine HSC development. Knowledge of early human HSC development is hampered by scarcity of tissue: analysis of both CFU-C and HSC development in the human embryo is limited. Here, we characterized the spatial distribution and temporal kinetics of CFU-C development within early human embryonic tissues. We then sought to adapt the murine ex vivo culture system to recapitulate human HSC development. We show robust expansion of CFU-Cs and maintenance, but no significant expansion, of human HSCs in culture. Furthermore, we demonstrate that HSCs emerge predominantly in the middle section of the dorsal aorta in our culture system. We conclude that there are important differences between early mouse and human hematopoiesis, which currently hinder the quest to recapitulate human HSC development ex vivo.


Subject(s)
Aorta/cytology , Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Spatio-Temporal Analysis , Stem Cells/cytology , Animals , Cell Proliferation/physiology , Cells, Cultured , Colony-Forming Units Assay , Embryo, Mammalian/embryology , Gonads/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , Heterografts , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
20.
Nat Commun ; 7: 10784, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26952187

ABSTRACT

During embryonic development, adult haematopoietic stem cells (HSCs) emerge preferentially in the ventral domain of the aorta in the aorta-gonad-mesonephros (AGM) region. Several signalling pathways such as Notch, Wnt, Shh and RA are implicated in this process, yet how these interact to regulate the emergence of HSCs has not previously been described in mammals. Using a combination of ex vivo and in vivo approaches, we report here that stage-specific reciprocal dorso-ventral inductive interactions and lateral input from the urogenital ridges are required to drive HSC development in the aorta. Our study strongly suggests that these inductive interactions in the AGM region are mediated by the interplay between spatially polarized signalling pathways. Specifically, Shh produced in the dorsal region of the AGM, stem cell factor in the ventral and lateral regions, and BMP inhibitory signals in the ventral tissue are integral parts of the regulatory system involved in the development of HSCs.


Subject(s)
Aorta/metabolism , Gonads/metabolism , Hematopoietic Stem Cells/metabolism , Mesonephros/metabolism , Signal Transduction , Animals , Aorta/embryology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Female , Gonads/embryology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Male , Mesonephros/embryology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL