Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Development ; 151(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37982461

ABSTRACT

Early organogenesis represents a key step in animal development, during which pluripotent cells diversify to initiate organ formation. Here, we sampled 300,000 single-cell transcriptomes from mouse embryos between E8.5 and E9.5 in 6-h intervals and combined this new dataset with our previous atlas (E6.5-E8.5) to produce a densely sampled timecourse of >400,000 cells from early gastrulation to organogenesis. Computational lineage reconstruction identified complex waves of blood and endothelial development, including a new programme for somite-derived endothelium. We also dissected the E7.5 primitive streak into four adjacent regions, performed scRNA-seq and predicted cell fates computationally. Finally, we defined developmental state/fate relationships by combining orthotopic grafting, microscopic analysis and scRNA-seq to transcriptionally determine cell fates of grafted primitive streak regions after 24 h of in vitro embryo culture. Experimentally determined fate outcomes were in good agreement with computationally predicted fates, demonstrating how classical grafting experiments can be revisited to establish high-resolution cell state/fate relationships. Such interdisciplinary approaches will benefit future studies in developmental biology and guide the in vitro production of cells for organ regeneration and repair.


Subject(s)
Gastrulation , Organogenesis , Mice , Animals , Cell Differentiation , Organogenesis/genetics , Primitive Streak , Endothelium , Embryo, Mammalian , Mammals
3.
Nat Commun ; 14(1): 4645, 2023 08 14.
Article in English | MEDLINE | ID: mdl-37580379

ABSTRACT

In mitosis, most transcription factors detach from chromatin, but some are retained and bookmark genomic sites. Mitotic bookmarking has been implicated in lineage inheritance, pluripotency and reprogramming. However, the biological significance of this mechanism in vivo remains unclear. Here, we address mitotic retention of the hemogenic factors GATA2, GFI1B and FOS during haematopoietic specification. We show that GATA2 remains bound to chromatin throughout mitosis, in contrast to GFI1B and FOS, via C-terminal zinc finger-mediated DNA binding. GATA2 bookmarks a subset of its interphase targets that are co-enriched for RUNX1 and other regulators of definitive haematopoiesis. Remarkably, homozygous mice harbouring the cyclin B1 mitosis degradation domain upstream Gata2 partially phenocopy knockout mice. Degradation of GATA2 at mitotic exit abolishes definitive haematopoiesis at aorta-gonad-mesonephros, placenta and foetal liver, but does not impair yolk sac haematopoiesis. Our findings implicate GATA2-mediated mitotic bookmarking as critical for definitive haematopoiesis and highlight a dependency on bookmarkers for lineage commitment.


Subject(s)
Chromatin , GATA2 Transcription Factor , Mitosis , Animals , Mice , Chromosomes/metabolism , DNA , Hematopoiesis/genetics , GATA2 Transcription Factor/genetics
4.
Nat Commun ; 13(1): 773, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140205

ABSTRACT

The transcription factor RUNX1 is a critical regulator of developmental hematopoiesis and is frequently disrupted in leukemia. Runx1 is a large, complex gene that is expressed from two alternative promoters under the spatiotemporal control of multiple hematopoietic enhancers. To dissect the dynamic regulation of Runx1 in hematopoietic development, we analyzed its three-dimensional chromatin conformation in mouse embryonic stem cell (ESC) differentiation cultures. Runx1 resides in a 1.1 Mb topologically associating domain (TAD) demarcated by convergent CTCF motifs. As ESCs differentiate to mesoderm, chromatin accessibility, Runx1 enhancer-promoter (E-P) interactions, and CTCF-CTCF interactions increase in the TAD, along with initiation of Runx1 expression from the P2 promoter. Differentiation to hematopoietic progenitor cells is associated with the formation of tissue-specific sub-TADs over Runx1, a shift in E-P interactions, P1 promoter demethylation, and robust expression from both Runx1 promoters. Deletion of promoter-proximal CTCF sites at the sub-TAD boundaries has no obvious effects on E-P interactions but leads to partial loss of domain structure, mildly affects gene expression, and delays hematopoietic development. Together, our analysis of gene regulation at a large multi-promoter developmental gene reveals that dynamic sub-TAD chromatin boundaries play a role in establishing TAD structure and coordinated gene expression.


Subject(s)
Chromatin/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation , DNA/chemistry , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Mesoderm/metabolism , Mice , Nucleic Acid Conformation , Promoter Regions, Genetic
5.
Cell Rep ; 37(11): 110103, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34910918

ABSTRACT

Hematopoietic stem cells (HSCs) emerge during development from the vascular wall of the main embryonic arteries. The onset of circulation triggers several processes that provide critical external factors for HSC generation. Nevertheless, it is not fully understood how and when the onset of circulation affects HSC emergence. Here we show that in Ncx1-/- mouse embryos devoid of circulation the HSC lineage develops until the phenotypic pro-HSC stage. However, these cells reside in an abnormal microenvironment, fail to activate the hematopoietic program downstream of Runx1, and are functionally impaired. Single-cell transcriptomics shows that during the endothelial-to-hematopoietic transition, Ncx1-/- cells fail to undergo a glycolysis to oxidative phosphorylation metabolic switch present in wild-type cells. Interestingly, experimental activation of glycolysis results in decreased intraembryonic hematopoiesis. Our results suggest that the onset of circulation triggers metabolic changes that allow HSC generation to proceed.


Subject(s)
Cell Differentiation , Cell Lineage , Endothelium, Vascular/pathology , Glycolysis , Hematopoiesis , Hematopoietic Stem Cells/pathology , Sodium-Calcium Exchanger/physiology , Animals , Endothelium, Vascular/metabolism , Female , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Phosphorylation , Single-Cell Analysis , Transcriptome
6.
Nat Commun ; 12(1): 7019, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857757

ABSTRACT

Yolk sac (YS) hematopoiesis is critical for the survival of the embryo and a major source of tissue-resident macrophages that persist into adulthood. Yet, the transcriptional and epigenetic regulation of YS hematopoiesis remains poorly characterized. Here we report that the epigenetic regulator Ezh2 is essential for YS hematopoiesis but dispensable for subsequent aorta-gonad-mesonephros (AGM) blood development. Loss of EZH2 activity in hemogenic endothelium (HE) leads to the generation of phenotypically intact but functionally deficient erythro-myeloid progenitors (EMPs), while the generation of primitive erythroid cells is not affected. EZH2 activity is critical for the generation of functional EMPs at the onset of the endothelial-to-hematopoietic transition but subsequently dispensable. We identify a lack of Wnt signaling downregulation as the primary reason for the production of non-functional EMPs. Together, our findings demonstrate a critical and stage-specific role of Ezh2 in modulating Wnt signaling during the generation of EMPs from YS HE.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Myeloid Progenitor Cells/metabolism , Vesicular Transport Proteins/genetics , Yolk Sac/metabolism , Animals , Cell Differentiation , Embryo, Mammalian , Enhancer of Zeste Homolog 2 Protein/deficiency , Epigenesis, Genetic , Erythroid Cells/cytology , Female , Fetus , Genes, Reporter , Hematopoiesis/genetics , Liver/cytology , Liver/growth & development , Liver/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Myeloid Progenitor Cells/pathology , Primary Cell Culture , Vesicular Transport Proteins/metabolism , Wnt Signaling Pathway , Yolk Sac/cytology , Yolk Sac/growth & development , Red Fluorescent Protein
7.
Genome Res ; 31(7): 1159-1173, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34088716

ABSTRACT

Regulatory interactions mediated by transcription factors (TFs) make up complex networks that control cellular behavior. Fully understanding these gene regulatory networks (GRNs) offers greater insight into the consequences of disease-causing perturbations than can be achieved by studying single TF binding events in isolation. Chromosomal translocations of the lysine methyltransferase 2A (KMT2A) gene produce KMT2A fusion proteins such as KMT2A-AFF1 (previously MLL-AF4), causing poor prognosis acute lymphoblastic leukemias (ALLs) that sometimes relapse as acute myeloid leukemias (AMLs). KMT2A-AFF1 drives leukemogenesis through direct binding and inducing the aberrant overexpression of key genes, such as the anti-apoptotic factor BCL2 and the proto-oncogene MYC However, studying direct binding alone does not incorporate possible network-generated regulatory outputs, including the indirect induction of gene repression. To better understand the KMT2A-AFF1-driven regulatory landscape, we integrated ChIP-seq, patient RNA-seq, and CRISPR essentiality screens to generate a model GRN. This GRN identified several key transcription factors such as RUNX1 that regulate target genes downstream of KMT2A-AFF1 using feed-forward loop (FFL) and cascade motifs. A core set of nodes are present in both ALL and AML, and CRISPR screening revealed several factors that help mediate response to the drug venetoclax. Using our GRN, we then identified a KMT2A-AFF1:RUNX1 cascade that represses CASP9, as well as KMT2A-AFF1-driven FFLs that regulate BCL2 and MYC through combinatorial TF activity. This illustrates how our GRN can be used to better connect KMT2A-AFF1 behavior to downstream pathways that contribute to leukemogenesis, and potentially predict shifts in gene expression that mediate drug response.

8.
Nat Commun ; 12(1): 821, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547282

ABSTRACT

Down syndrome is associated with genome-wide perturbation of gene expression, which may be mediated by epigenetic changes. We perform an epigenome-wide association study on neonatal bloodspots comparing 196 newborns with Down syndrome and 439 newborns without Down syndrome, adjusting for cell-type heterogeneity, which identifies 652 epigenome-wide significant CpGs (P < 7.67 × 10-8) and 1,052 differentially methylated regions. Differential methylation at promoter/enhancer regions correlates with gene expression changes in Down syndrome versus non-Down syndrome fetal liver hematopoietic stem/progenitor cells (P < 0.0001). The top two differentially methylated regions overlap RUNX1 and FLI1, both important regulators of megakaryopoiesis and hematopoietic development, with significant hypermethylation at promoter regions of these two genes. Excluding Down syndrome newborns harboring preleukemic GATA1 mutations (N = 30), identified by targeted sequencing, has minimal impact on the epigenome-wide association study results. Down syndrome has profound, genome-wide effects on DNA methylation in hematopoietic cells in early life, which may contribute to the high frequency of hematological problems, including leukemia, in children with Down syndrome.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Down Syndrome/genetics , Epigenesis, Genetic , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Case-Control Studies , Core Binding Factor Alpha 2 Subunit/metabolism , CpG Islands , DNA Methylation , Down Syndrome/metabolism , Down Syndrome/pathology , Female , Fetus , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Genome, Human , Genome-Wide Association Study , Hematopoietic Stem Cells/pathology , Humans , Infant, Newborn , Liver/metabolism , Liver/pathology , Male , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/metabolism
10.
Nat Cell Biol ; 23(1): 61-74, 2021 01.
Article in English | MEDLINE | ID: mdl-33420489

ABSTRACT

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


Subject(s)
Embryonic Stem Cells/cytology , Hemangioblasts/cytology , Mesoderm/cytology , T-Box Domain Proteins/physiology , Yolk Sac/cytology , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Stem Cells/metabolism , Female , Hemangioblasts/metabolism , Male , Mesoderm/metabolism , Mice, Knockout , Pregnancy , RNA-Seq , Single-Cell Analysis , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Yolk Sac/metabolism
11.
Nucleic Acids Res ; 47(14): 7402-7417, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31127293

ABSTRACT

The CRISPR system is widely used in genome editing for biomedical research. Here, using either dual paired Cas9D10A nickases or paired Cas9 nuclease we characterize unintended larger deletions at on-target sites that frequently evade common genotyping practices. We found that unintended larger deletions are prevalent at multiple distinct loci on different chromosomes, in cultured cells and mouse embryos alike. We observed a high frequency of microhomologies at larger deletion breakpoint junctions, suggesting the involvement of microhomology-mediated end joining in their generation. In populations of edited cells, the distribution of larger deletion sizes is dependent on proximity to sgRNAs and cannot be predicted by microhomology sequences alone.


Subject(s)
CRISPR-Cas Systems , Chromosome Deletion , Chromosomes, Mammalian/genetics , Gene Editing/methods , Sequence Deletion , Animals , Cell Line , Chromosome Breakpoints , Chromosomes, Mammalian/metabolism , DNA End-Joining Repair , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Mice , Models, Genetic , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
12.
Blood ; 131(20): 2223-2234, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29555646

ABSTRACT

Despite the well-established cell-intrinsic role of epigenetic factors in normal and malignant hematopoiesis, their cell-extrinsic role remains largely unexplored. Herein we investigated the hematopoietic impact of inactivating Ezh2, a key component of polycomb repressive complex 2 (PRC2), in the fetal liver (FL) vascular niche. Hematopoietic specific (Vav-iCre) Ezh2 inactivation enhanced FL hematopoietic stem cell (HSC) expansion with normal FL erythropoiesis. In contrast, endothelium (Tie2-Cre) targeted Ezh2 inactivation resulted in embryonic lethality with severe anemia at embryonic day 13.5 despite normal emergence of functional HSCs. Ezh2-deficient FL endothelium overexpressed Mmp9, which cell-extrinsically depleted the membrane-bound form of Kit ligand (mKitL), an essential hematopoietic cytokine, in FL. Furthermore, Mmp9 inhibition in vitro restored mKitL expression along with the erythropoiesis supporting capacity of FL endothelial cells. These data establish that Ezh2 is intrinsically dispensable for FL HSCs and provides proof of principle that modulation of epigenetic regulators in niche components can exert a marked cell-extrinsic impact.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Fetus , Hematopoiesis, Extramedullary , Liver/physiology , Anemia/genetics , Anemia/metabolism , Animals , Biomarkers , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/metabolism , Fluorescent Antibody Technique , Gene Expression , Gene Silencing , Hematopoiesis, Extramedullary/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Phenotype , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Stem Cell Factor/metabolism
13.
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
14.
Adv Exp Med Biol ; 962: 47-64, 2017.
Article in English | MEDLINE | ID: mdl-28299650

ABSTRACT

The de novo generation of hematopoietic stem and progenitor cells (HSPC) occurs solely during embryogenesis from a population of epithelial cells called hemogenic endothelium (HE). During midgestation HE cells in multiple intra- and extraembryonic vascular beds leave the vessel wall as they transition into HSPCs in a process termed the endothelial to hematopoietic transition (EHT). Runx1 expression in HE cells orchestrates the transcriptional switch necessary for the transdifferentiation of endothelial cells into functional HSPCs. Runx1 is widely considered the master regulator of developmental hematopoiesis because it plays an essential function during specification of the hematopoietic lineage during embryogenesis. Here we review the role of Runx1 in embryonic HSPC formation, with a particular focus on its role in hemogenic endothelium.


Subject(s)
Blood Cells/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Development/physiology , Hemangioblasts/metabolism , Animals , Cell Transdifferentiation/physiology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Endothelium, Vascular/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Stem Cells/metabolism
15.
Dev Biol ; 424(2): 236-245, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28189604

ABSTRACT

Hematopoietic stem cells (HSCs) emerge during development via an endothelial-to-hematopoietic transition from hemogenic endothelium of the dorsal aorta (DA). Using in situ hybridization and analysis of a knock-in RedStar reporter, we show that the transcriptional regulator Hhex is expressed in endothelium of the dorsal aorta (DA) and in clusters of putative HSCs as they are specified during murine development. We exploited this observation, using the Hhex locus to define cis regulatory elements, enhancers and interacting transcription factors that are both necessary and sufficient to support gene expression in the emerging HSC. We identify an evolutionarily conserved non-coding region (ECR) in the Hhex locus with the capacity to bind the hematopoietic-affiliated transcriptional regulators Gata2, SCL, Fli1, Pu.1 and Ets1/2. This region is sufficient to drive the expression of a transgenic GFP reporter in the DA endothelium and intra-aortic hematopoietic clusters. GFP-positive AGM cells co-expressed HSC-associated markers c-Kit, CD34, VE-Cadherin, and CD45, and were capable of multipotential differentiation and long term engraftment when transplanted into myelo-ablated recipients. The Hhex ECR was also sufficient to drive expression at additional blood sites including the yolk sac blood islands, fetal liver, vitelline and umbilical arteries and the adult bone marrow, suggesting a common mechanism for Hhex regulation throughout ontogenesis of the blood system. To explore the physiological requirement for the Hhex ECR region during hematoendothelial development, we deleted the ECR element from the endogenous locus in the context of a targeted Hhex-RedStar reporter allele. Results indicate a specific requirement for the ECR in blood-associated Hhex expression during development and further demonstrate a requirement for this region in the adult HSC compartment. Taken together, our results identified the ECR region as an enhancer both necessary and sufficient for gene expression in HSC development and homeostasis. The Hhex ECR thus appears to be a core node for the convergence of the transcription factor network that governs the emergence of HSCs.


Subject(s)
Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Compartmentation , Cell Lineage/genetics , Colony-Forming Units Assay , Conserved Sequence/genetics , Embryo, Mammalian/metabolism , Genetic Loci , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/genetics
16.
Nat Immunol ; 17(12): 1424-1435, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27695000

ABSTRACT

The final stages of restriction to the T cell lineage occur in the thymus after the entry of thymus-seeding progenitors (TSPs). The identity and lineage potential of TSPs remains unclear. Because the first embryonic TSPs enter a non-vascularized thymic rudiment, we were able to directly image and establish the functional and molecular properties of embryonic thymopoiesis-initiating progenitors (T-IPs) before their entry into the thymus and activation of Notch signaling. T-IPs did not include multipotent stem cells or molecular evidence of T cell-restricted progenitors. Instead, single-cell molecular and functional analysis demonstrated that most fetal T-IPs expressed genes of and had the potential to develop into lymphoid as well as myeloid components of the immune system. Moreover, studies of embryos deficient in the transcriptional regulator RBPJ demonstrated that canonical Notch signaling was not involved in pre-thymic restriction to the T cell lineage or the migration of T-IPs.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Lymphoid Progenitor Cells/physiology , Myeloid Progenitor Cells/physiology , Receptors, Notch/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Fetus , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
17.
Dev Cell ; 36(5): 525-39, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26954547

ABSTRACT

Definitive hematopoiesis emerges via an endothelial-to-hematopoietic transition in the embryo and placenta; however, the precursor cells to hemogenic endothelium are not defined phenotypically. We previously demonstrated that the induction of hematopoietic progenitors from fibroblasts progresses through hemogenic precursors that are Prom1(+)Sca1(+)CD34(+)CD45(-) (PS34CD45(-)). Guided by these studies, we analyzed mouse placentas and identified a population with this phenotype. These cells express endothelial markers, are heterogeneous for early hematopoietic markers, and localize to the vascular labyrinth. Remarkably, global gene expression profiles of PS34CD45(-) cells correlate with reprogrammed precursors and establish a hemogenic precursor cell molecular signature. PS34CD45(-) cells are also present in intra-embryonic hemogenic sites. After stromal co-culture, PS34CD45(-) cells give rise to all blood lineages and engraft primary and secondary immunodeficient mice. In summary, we show that reprogramming reveals a phenotype for in vivo precursors to hemogenic endothelium, establishing that direct in vitro conversion informs developmental processes in vivo.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Cellular Reprogramming , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Cells, Cultured , Endothelium/metabolism , Female , Fibroblasts/cytology , Mice , Mice, Inbred C57BL , Pregnancy
18.
Exp Hematol ; 43(10): 821-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26143582

ABSTRACT

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


Subject(s)
Hemoglobinopathies/genetics , Hemoglobinopathies/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Congresses as Topic , Hemoglobinopathies/pathology , Hemoglobinopathies/therapy , Humans
19.
Mol Cell Biol ; 35(12): 2165-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25870111

ABSTRACT

The bone morphogenetic protein (BMP)/SMAD signaling pathway is a critical regulator of angiogenic sprouting and is involved in vascular development in the embryo. SMAD1 and SMAD5, the core mediators of BMP signaling, are vital for this activity, yet little is known about their transcriptional regulation in endothelial cells. Here, we have integrated multispecies sequence conservation, tissue-specific chromatin, in vitro reporter assay, and in vivo transgenic data to identify and validate Smad1+63 and the Smad5 promoter as tissue-specific cis-regulatory elements that are active in the developing endothelium. The activity of these elements in the endothelium was dependent on highly conserved ETS, GATA, and E-box motifs, and chromatin immunoprecipitation showed high levels of enrichment of FLI1, GATA2, and SCL at these sites in endothelial cell lines and E11 dorsal aortas in vivo. Knockdown of FLI1 and GATA2 but not SCL reduced the expression of SMAD1 and SMAD5 in endothelial cells in vitro. In contrast, CD31(+) cKit(-) endothelial cells harvested from embryonic day 9 (E9) aorta-gonad-mesonephros (AGM) regions of GATA2 null embryos showed reduced Smad1 but not Smad5 transcript levels. This is suggestive of a degree of in vivo selection where, in the case of reduced SMAD1 levels, endothelial cells with more robust SMAD5 expression have a selective advantage.


Subject(s)
Endothelium/embryology , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Proto-Oncogene Protein c-fli-1/metabolism , Smad1 Protein/genetics , Smad5 Protein/genetics , Animals , Base Sequence , Cell Line , Endothelium/metabolism , GATA2 Transcription Factor/genetics , Gene Knockdown Techniques , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/genetics
20.
Development ; 141(20): 4018-30, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25252941

ABSTRACT

Transcription factors (TFs) act within wider regulatory networks to control cell identity and fate. Numerous TFs, including Scl (Tal1) and PU.1 (Spi1), are known regulators of developmental and adult haematopoiesis, but how they act within wider TF networks is still poorly understood. Transcription activator-like effectors (TALEs) are a novel class of genetic tool based on the modular DNA-binding domains of Xanthomonas TAL proteins, which enable DNA sequence-specific targeting and the manipulation of endogenous gene expression. Here, we report TALEs engineered to target the PU.1-14kb and Scl+40kb transcriptional enhancers as efficient new tools to perturb the expression of these key haematopoietic TFs. We confirmed the efficiency of these TALEs at the single-cell level using high-throughput RT-qPCR, which also allowed us to assess the consequences of both PU.1 activation and repression on wider TF networks during developmental haematopoiesis. Combined with comprehensive cellular assays, these experiments uncovered novel roles for PU.1 during early haematopoietic specification. Finally, transgenic mouse studies confirmed that the PU.1-14kb element is active at sites of definitive haematopoiesis in vivo and PU.1 is detectable in haemogenic endothelium and early committing blood cells. We therefore establish TALEs as powerful new tools to study the functionality of transcriptional networks that control developmental processes such as early haematopoiesis.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , Cell Differentiation , Coculture Techniques , Endothelial Cells/cytology , Hematopoietic Stem Cells , Humans , K562 Cells , Mice , Mice, Transgenic , Phenotype , Single-Cell Analysis , Transcription Factors/metabolism , Transgenes , Xanthomonas/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL