Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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
2.
Haematologica ; 104(6): 1189-1201, 2019 06.
Article in English | MEDLINE | ID: mdl-30679325

ABSTRACT

The t(4;11)(q21;q23) translocation is associated with high-risk infant pro-B-cell acute lymphoblastic leukemia and arises prenatally during embryonic/fetal hematopoiesis. The developmental/pathogenic contribution of the t(4;11)-resulting MLL-AF4 (MA4) and AF4-MLL (A4M) fusions remains unclear; MA4 is always expressed in patients with t(4;11)+ B-cell acute lymphoblastic leukemia, but the reciprocal fusion A4M is expressed in only half of the patients. Because prenatal leukemogenesis manifests as impaired early hematopoietic differentiation, we took advantage of well-established human embryonic stem cell-based hematopoietic differentiation models to study whether the A4M fusion cooperates with MA4 during early human hematopoietic development. Co-expression of A4M and MA4 strongly promoted the emergence of hemato-endothelial precursors, both endothelial- and hemogenic-primed. Double fusion-expressing hemato-endothelial precursors specified into significantly higher numbers of both hematopoietic and endothelial-committed cells, irrespective of the differentiation protocol used and without hijacking survival/proliferation. Functional analysis of differentially expressed genes and differentially enriched H3K79me3 genomic regions by RNA-sequencing and H3K79me3 chromatin immunoprecipitation-sequencing, respectively, confirmed a hematopoietic/endothelial cell differentiation signature in double fusion-expressing hemato-endothelial precursors. Importantly, chromatin immunoprecipitation-sequencing analysis revealed a significant enrichment of H3K79 methylated regions specifically associated with HOX-A cluster genes in double fusion-expressing differentiating hematopoietic cells. Overall, these results establish a functional and molecular cooperation between MA4 and A4M fusions during human hematopoietic development.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hematopoiesis/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Animals , Apoptosis/genetics , Cell Cycle/genetics , Coculture Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Methylation , Mice , Mice, Knockout
3.
Stem Cells ; 35(2): 507-521, 2017 02.
Article in English | MEDLINE | ID: mdl-27615355

ABSTRACT

Stable reconstitution of vascular endothelial beds upon transplantation of progenitor cells represents an important challenge due to the paucity and generally limited integration/expansion potential of most identified vascular related cell subsets. We previously showed that mouse fetal liver (FL) hemato/vascular cells from day 12 of gestation (E12), expressing the Stem Cell Leukaemia (SCL) gene enhancer transgene (SCL-PLAP+ cells), had robust endothelial engraftment potential when transferred to the blood stream of newborns or adult conditioned recipients, compared to the scarce vascular contribution of adult bone marrow cells. However, the specific SCL-PLAP+ hematopoietic or endothelial cell subset responsible for the long-term reconstituting endothelial cell (LTR-EC) activity and its confinement to FL developmental stages remained unknown. Using a busulfan-treated newborn transplantation model, we show that LTR-EC activity is restricted to the SCL-PLAP+ VE-cadherin+ CD45- cell population, devoid of hematopoietic reconstitution activity and largely composed by Lyve1+ endothelial-committed cells. SCL-PLAP+ Ve-cadherin+ CD45- cells contributed to the liver sinusoidal endothelium and also to the heart, kidney and lung microvasculature. LTR-EC activity was detected at different stages of FL development, yet marginal activity was identified in the adult liver, revealing unknown functional differences between fetal and adult liver endothelial/endothelial progenitors. Importantly, the observations that expanding donor-derived vascular grafts colocalize with proliferating hepatocyte-like cells and participate in the systemic circulation, support their functional integration into young livers. These findings offer new insights into the engraftment, phonotypical, and developmental characterization of a novel endothelial/endothelial progenitor cell subtype with multiorgan LTR-EC activity, potentially instrumental for the treatment/genetic correction of vascular diseases. Stem Cells 2017;35:507-521.


Subject(s)
Endothelial Cells/cytology , Fetus/cytology , Fetus/embryology , Liver/embryology , Animals , Antigens, CD/metabolism , Blood Vessels/transplantation , Cadherins/metabolism , Cell Aggregation , Cell Line , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Hematopoiesis , Leukocyte Common Antigens/metabolism , Mice , Organ Specificity , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism
4.
Blood ; 128(15): 1928-1939, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27554085

ABSTRACT

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


Subject(s)
Embryo, Mammalian/metabolism , Endoglin/metabolism , Enhancer Elements, Genetic/physiology , Hematopoiesis/physiology , Molecular Probes/metabolism , Animals , Cell Line , Embryo, Mammalian/cytology , Endoglin/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Molecular Probes/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism
6.
Cancer Cell ; 23(2): 200-14, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23410974

ABSTRACT

Most patients with acute lymphoblastic leukemia (ALL) fail current treatments highlighting the need for better therapies. Because oncogenic signaling activates a p53-dependent DNA damage response and apoptosis, leukemic cells must devise appropriate countermeasures. We show here that growth factor independence 1 (Gfi1) can serve such a function because Gfi1 ablation exacerbates p53 responses and lowers the threshold for p53-induced cell death. Specifically, Gfi1 restricts p53 activity and expression of proapoptotic p53 targets such as Bax, Noxa (Pmaip1), and Puma (Bbc3). Subsequently, Gfi1 ablation cures mice from leukemia and limits the expansion of primary human T-ALL xenografts in mice. This suggests that targeting Gfi1 could improve the prognosis of patients with T-ALL or other lymphoid leukemias.


Subject(s)
Apoptosis , DNA Damage/genetics , DNA-Binding Proteins/physiology , Lymphoma, B-Cell/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , Animals , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, Notch1/genetics , Xenograft Model Antitumor Assays
7.
Blood ; 120(19): 4006-17, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22932805

ABSTRACT

The coding single nucleotide polymorphism GFI136N in the human gene growth factor independence 1 (GFI1) is present in 3%-7% of whites and increases the risk for acute myeloid leukemia (AML) by 60%. We show here that GFI136N, in contrast to GFI136S, lacks the ability to bind to the Gfi1 target gene that encodes the leukemia-associated transcription factor Hoxa9 and fails to initiate histone modifications that regulate HoxA9 expression. Consistent with this, AML patients heterozygous for the GFI136N variant show increased HOXA9 expression compared with normal controls. Using ChipSeq, we demonstrate that GFI136N specific epigenetic changes are also present in other genes involved in the development of AML. Moreover, granulomonocytic progenitors, a bone marrow subset from which AML can arise in humans and mice, show a proliferative expansion in the presence of the GFI136N variant. In addition, granulomonocytic progenitors carrying the GFI136N variant allele have altered gene expression patterns and differ in their ability to grow after transplantation. Finally, GFI136N can accelerate a K-RAS driven fatal myeloproliferative disease in mice. Our data suggest that the presence of a GFI136N variant allele induces a preleukemic state in myeloid precursors by deregulating the expression of Hoxa9 and other AML-related genes.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Homeodomain Proteins/genetics , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics , Animals , Cluster Analysis , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Hematopoiesis/genetics , Histones/metabolism , Humans , Mice , Mice, Transgenic , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/mortality , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/metabolism
8.
EMBO J ; 30(19): 4059-70, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21873977

ABSTRACT

The transcription factor RUNX1 is essential to establish the haematopoietic gene expression programme; however, the mechanism of how it activates transcription of haematopoietic stem cell (HSC) genes is still elusive. Here, we obtained novel insights into RUNX1 function by studying regulation of the human CD34 gene, which is expressed in HSCs. Using transgenic mice carrying human CD34 PAC constructs, we identified a novel downstream regulatory element (DRE), which is bound by RUNX1 and is necessary for human CD34 expression in long-term (LT)-HSCs. Conditional deletion of Runx1 in mice harbouring human CD34 promoter-DRE constructs abrogates human CD34 expression. We demonstrate by chromosome conformation capture assays in LT-HSCs that the DRE physically interacts with the human CD34 promoter. Targeted mutagenesis of RUNX binding sites leads to perturbation of this interaction and decreased human CD34 expression in LT-HSCs. Overall, our in vivo data provide novel evidence about the role of RUNX1 in mediating interactions between distal and proximal elements of the HSC gene CD34.


Subject(s)
Antigens, CD34/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Animals , Bone Marrow Transplantation , Chromatin/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Fetal Blood/cytology , Genotype , HL-60 Cells , Humans , Mice , Mice, Transgenic , Models, Biological , Regulatory Sequences, Nucleic Acid/genetics
9.
Cancer Cell ; 20(1): 39-52, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21741595

ABSTRACT

Pathways defining susceptibility of normal cells to oncogenic transformation may be valuable therapeutic targets. We characterized the cell of origin and its critical pathways in MN1-induced leukemias. Common myeloid (CMP) but not granulocyte-macrophage progenitors (GMP) could be transformed by MN1. Complementation studies of CMP-signature genes in GMPs demonstrated that MN1-leukemogenicity required the MEIS1/AbdB-like HOX-protein complex. ChIP-sequencing identified common target genes of MN1 and MEIS1 and demonstrated identical binding sites for a large proportion of their chromatin targets. Transcriptional repression of MEIS1 targets in established MN1 leukemias demonstrated antileukemic activity. As MN1 relies on but cannot activate expression of MEIS1/AbdB-like HOX proteins, transcriptional activity of these genes determines cellular susceptibility to MN1-induced transformation and may represent a promising therapeutic target.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genes, Dominant/genetics , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred C57BL , Models, Biological , Myeloid Ecotropic Viral Integration Site 1 Protein , Promoter Regions, Genetic/genetics , Protein Binding
10.
Science ; 329(5987): 85-9, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20538915

ABSTRACT

T cells develop in the thymus and are critical for adaptive immunity. Natural killer (NK) lymphocytes constitute an essential component of the innate immune system in tumor surveillance, reproduction, and defense against microbes and viruses. Here, we show that the transcription factor Bcl11b was expressed in all T cell compartments and was indispensable for T lineage development. When Bcl11b was deleted, T cells from all developmental stages acquired NK cell properties and concomitantly lost or decreased T cell-associated gene expression. These induced T-to-natural killer (ITNK) cells, which were morphologically and genetically similar to conventional NK cells, killed tumor cells in vitro, and effectively prevented tumor metastasis in vivo. Therefore, ITNKs may represent a new cell source for cell-based therapies.


Subject(s)
Cell Lineage , Killer Cells, Natural/physiology , Lymphopoiesis , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Genes, T-Cell Receptor beta , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphopoiesis/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...