Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Genome Biol ; 24(1): 152, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37370129

ABSTRACT

BACKGROUND: Platelets and erythrocytes constitute over 95% of all hematopoietic stem cell output. However, the clonal dynamics of HSC contribution to these lineages remains largely unexplored. RESULTS: We use lentiviral genetic labeling of mouse hematopoietic stem cells to quantify output from all lineages, nucleate, and anucleate, simultaneously linking these with stem and progenitor cell transcriptomic phenotypes using single-cell RNA-sequencing. We observe dynamic shifts of clonal behaviors through time in same-animal peripheral blood and demonstrate that acute platelet depletion shifts the output of multipotent hematopoietic stem cells to the exclusive production of platelets. Additionally, we observe the emergence of new myeloid-biased clones, which support short- and long-term production of blood cells. CONCLUSIONS: Our approach enables kinetic studies of multi-lineage output in the peripheral blood and transcriptional heterogeneity of individual hematopoietic stem cells. Our results give a unique insight into hematopoietic stem cell reactivation upon platelet depletion and of clonal dynamics in both steady state and under stress.


Subject(s)
Blood Platelets , Hematopoiesis , Mice , Animals , Cell Lineage , Kinetics , Hematopoietic Stem Cells , Clone Cells , Cell Differentiation
2.
Development ; 148(3)2021 02 03.
Article in English | MEDLINE | ID: mdl-33462113

ABSTRACT

Macrophages are components of the innate immune system with key roles in tissue inflammation and repair. It is now evident that macrophages also support organogenesis, but few studies have characterized their identity, ontogeny and function during heart development. Here, we show that the distribution and prevalence of resident macrophages in the subepicardial compartment of the developing heart coincides with the emergence of new lymphatics, and that macrophages interact closely with the nascent lymphatic capillaries. Consequently, global macrophage deficiency led to extensive vessel disruption, with mutant hearts exhibiting shortened and mis-patterned lymphatics. The origin of cardiac macrophages was linked to the yolk sac and foetal liver. Moreover, the Cx3cr1+ myeloid lineage was found to play essential functions in the remodelling of the lymphatic endothelium. Mechanistically, macrophage hyaluronan was required for lymphatic sprouting by mediating direct macrophage-lymphatic endothelial cell interactions. Together, these findings reveal insight into the role of macrophages as indispensable mediators of lymphatic growth during the development of the mammalian cardiac vasculature.


Subject(s)
Heart/growth & development , Lymphatic Vessels , Macrophages/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , Cell Adhesion , Cell Line , Endothelial Cells , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Humans , Inflammation , Lymphangiogenesis , Macrophages/immunology , Mice , Mice, Inbred C57BL , Organogenesis/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Yolk Sac
3.
Nature ; 554(7690): 106-111, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29298288

ABSTRACT

Rare multipotent haematopoietic stem cells (HSCs) in adult bone marrow with extensive self-renewal potential can efficiently replenish all myeloid and lymphoid blood cells, securing long-term multilineage reconstitution after physiological and clinical challenges such as chemotherapy and haematopoietic transplantations. HSC transplantation remains the only curative treatment for many haematological malignancies, but inefficient blood-lineage replenishment remains a major cause of morbidity and mortality. Single-cell transplantation has uncovered considerable heterogeneity among reconstituting HSCs, a finding that is supported by studies of unperturbed haematopoiesis and may reflect different propensities for lineage-fate decisions by distinct myeloid-, lymphoid- and platelet-biased HSCs. Other studies suggested that such lineage bias might reflect generation of unipotent or oligopotent self-renewing progenitors within the phenotypic HSC compartment, and implicated uncoupling of the defining HSC properties of self-renewal and multipotency. Here we use highly sensitive tracking of progenitors and mature cells of the megakaryocyte/platelet, erythroid, myeloid and B and T cell lineages, produced from singly transplanted HSCs, to reveal a highly organized, predictable and stable framework for lineage-restricted fates of long-term self-renewing HSCs. Most notably, a distinct class of HSCs adopts a fate towards effective and stable replenishment of a megakaryocyte/platelet-lineage tree but not of other blood cell lineages, despite sustained multipotency. No HSCs contribute exclusively to any other single blood-cell lineage. Single multipotent HSCs can also fully restrict towards simultaneous replenishment of megakaryocyte, erythroid and myeloid lineages without executing their sustained lymphoid lineage potential. Genetic lineage-tracing analysis also provides evidence for an important role of platelet-biased HSCs in unperturbed adult haematopoiesis. These findings uncover a limited repertoire of distinct HSC subsets, defined by a predictable and hierarchical propensity to adopt a fate towards replenishment of a restricted set of blood lineages, before loss of self-renewal and multipotency.


Subject(s)
Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Multipotent Stem Cells/cytology , Animals , Antigens, CD34 , B-Lymphocytes/cytology , Blood Platelets/cytology , CD48 Antigen/deficiency , Cell Self Renewal , Erythroid Cells/cytology , Female , Hematopoietic Stem Cells/metabolism , Male , Megakaryocytes/cytology , Mice , Multipotent Stem Cells/metabolism , Myeloid Cells/cytology , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , T-Lymphocytes/cytology
4.
Cancer Cell ; 30(3): 404-417, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27622333

ABSTRACT

More than 80% of patients with the refractory anemia with ring sideroblasts subtype of myelodysplastic syndrome (MDS) have mutations in Splicing Factor 3B, Subunit 1 (SF3B1). We generated a conditional knockin mouse model of the most common SF3B1 mutation, Sf3b1(K700E). Sf3b1(K700E) mice develop macrocytic anemia due to a terminal erythroid maturation defect, erythroid dysplasia, and long-term hematopoietic stem cell (LT-HSC) expansion. Sf3b1(K700E) myeloid progenitors and SF3B1-mutant MDS patient samples demonstrate aberrant 3' splice-site selection associated with increased nonsense-mediated decay. Tet2 loss cooperates with Sf3b1(K700E) to cause a more severe erythroid and LT-HSC phenotype. Furthermore, the spliceosome modulator, E7017, selectively kills SF3B1(K700E)-expressing cells. Thus, SF3B1(K700E) expression reflects the phenotype of the mutation in MDS and may be a therapeutic target in MDS.


Subject(s)
Erythropoiesis/physiology , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Spliceosomes/physiology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Erythropoiesis/genetics , Hematopoietic Stem Cells/physiology , Humans , Mice , Mice, Transgenic , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Phosphoproteins/deficiency , Phosphoproteins/metabolism , Point Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Splicing , RNA Splicing Factors/deficiency , RNA Splicing Factors/metabolism
5.
Cancer Cell ; 26(4): 509-20, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25242043

ABSTRACT

The casein kinase 1A1 gene (CSNK1A1) is a putative tumor suppressor gene located in the common deleted region for del(5q) myelodysplastic syndrome (MDS). We generated a murine model with conditional inactivation of Csnk1a1 and found that Csnk1a1 haploinsufficiency induces hematopoietic stem cell expansion and a competitive repopulation advantage, whereas homozygous deletion induces hematopoietic stem cell failure. Based on this finding, we found that heterozygous inactivation of Csnk1a1 sensitizes cells to a CSNK1 inhibitor relative to cells with two intact alleles. In addition, we identified recurrent somatic mutations in CSNK1A1 on the nondeleted allele of patients with del(5q) MDS. These studies demonstrate that CSNK1A1 plays a central role in the biology of del(5q) MDS and is a promising therapeutic target.


Subject(s)
Casein Kinase I/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 5 , Myelodysplastic Syndromes/genetics , Aged , Animals , Base Sequence , Casein Kinase I/genetics , DNA Primers , Female , Flow Cytometry , Haploinsufficiency , Humans , Male , Mice , Mutation , Polymerase Chain Reaction , Young Adult
6.
Proc Natl Acad Sci U S A ; 107(40): 17315-20, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20855591

ABSTRACT

Acetaminophen (APAP) toxicity is the most common drug-induced cause of acute liver failure in the United States. The only available treatment, N-acetylcysteine (NAC), has a limited time window of efficacy, indicating a need for additional therapeutic options. Zebrafish have emerged as a powerful tool for drug discovery. Here, we developed a clinically relevant zebrafish model of APAP toxicity. APAP depleted glutathione stores, elevated aminotransferase levels, increased apoptosis, and caused dose-dependent hepatocyte necrosis. These outcomes were limited by NAC and conserved in zebrafish embryos. In a targeted embryonic chemical screen, prostaglandin E2 (PGE2) was identified as a potential therapeutic agent; in the adult, PGE2 similarly decreased APAP-associated toxicity. Significantly, when combined with NAC, PGE2 extended the time window for a successful intervention, synergistically reducing apoptosis, improving liver enzymes, and preventing death. Use of a wnt reporter zebrafish line and chemical genetic epistasis showed that the effects of PGE2 are mediated through the wnt signaling pathway. Zebrafish can be used as a clinically relevant toxicological model amenable to the identification of additional therapeutics and biomarkers of APAP injury; our data suggest combinatorial PGE2 and NAC treatment would be beneficial for patients with APAP-induced liver damage.


Subject(s)
Acetaminophen/toxicity , Acetylcysteine , Chemical and Drug Induced Liver Injury , Dinoprostone/metabolism , Liver Failure, Acute , Signal Transduction/physiology , Zebrafish , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Analgesics, Non-Narcotic/toxicity , Animals , Animals, Genetically Modified , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Genes, Reporter , Glutathione/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Failure, Acute/drug therapy , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Proteome/analysis , Zebrafish/anatomy & histology , Zebrafish/physiology
7.
Cancer Cell ; 17(6): 584-96, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20541703

ABSTRACT

We report a Jak2V617F knockin mouse myeloproliferative neoplasm (MPN) model resembling human polycythemia vera (PV). The MPN is serially transplantable and we demonstrate that the hematopoietic stem cell (HSC) compartment has the unique capacity for disease initiation but does not have a significant selective competitive advantage over wild-type HSCs. In contrast, myeloid progenitor populations are expanded and skewed toward the erythroid lineage, but cannot transplant the disease. Treatment with a JAK2 kinase inhibitor ameliorated the MPN phenotype, but did not eliminate the disease-initiating population. These findings provide insights into the consequences of JAK2 activation on HSC differentiation and function and have the potential to inform therapeutic approaches to JAK2V617F-positive MPN.


Subject(s)
Amino Acid Substitution , Hematopoietic Stem Cells/pathology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Animals , Antigens, CD/metabolism , Bone Marrow/pathology , Bone Marrow Cells/drug effects , Bone Marrow Transplantation , Cell Count , Cell Differentiation/genetics , Disease Models, Animal , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Erythropoietin/pharmacology , Gene Expression/genetics , Gene Expression Profiling , Hematocrit , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Heterozygote , Humans , Janus Kinase 2/antagonists & inhibitors , Megakaryocyte Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/pathology , Megakaryocyte-Erythroid Progenitor Cells/drug effects , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Megakaryocyte-Erythroid Progenitor Cells/pathology , Megakaryocyte-Erythroid Progenitor Cells/transplantation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myeloproliferative Disorders/drug therapy , Polycythemia Vera/genetics , Polycythemia Vera/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Spleen/drug effects , Spleen/pathology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Survival Analysis
8.
Cell ; 137(4): 736-48, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450519

ABSTRACT

During vertebrate embryogenesis, hematopoietic stem cells (HSCs) arise in the aorta-gonads-mesonephros (AGM) region. We report here that blood flow is a conserved regulator of HSC formation. In zebrafish, chemical blood flow modulators regulated HSC development, and silent heart (sih) embryos, lacking a heartbeat and blood circulation, exhibited severely reduced HSCs. Flow-modifying compounds primarily affected HSC induction after the onset of heartbeat; however, nitric oxide (NO) donors regulated HSC number even when treatment occurred before the initiation of circulation, and rescued HSCs in sih mutants. Morpholino knockdown of nos1 (nnos/enos) blocked HSC development, and its requirement was shown to be cell autonomous. In the mouse, Nos3 (eNos) was expressed in HSCs in the AGM. Intrauterine Nos inhibition or embryonic Nos3 deficiency resulted in a reduction of hematopoietic clusters and transplantable murine HSCs. This work links blood flow to AGM hematopoiesis and identifies NO as a conserved downstream regulator of HSC development.


Subject(s)
Blood Physiological Phenomena , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Zebrafish
9.
Cell ; 136(6): 1136-47, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19303855

ABSTRACT

Interactions between developmental signaling pathways govern the formation and function of stem cells. Prostaglandin (PG) E2 regulates vertebrate hematopoietic stem cells (HSC). Similarly, the Wnt signaling pathway controls HSC self-renewal and bone marrow repopulation. Here, we show that wnt reporter activity in zebrafish HSCs is responsive to PGE2 modulation, demonstrating a direct interaction in vivo. Inhibition of PGE2 synthesis blocked wnt-induced alterations in HSC formation. PGE2 modified the wnt signaling cascade at the level of beta-catenin degradation through cAMP/PKA-mediated stabilizing phosphorylation events. The PGE2/Wnt interaction regulated murine stem and progenitor populations in vitro in hematopoietic ES cell assays and in vivo following transplantation. The relationship between PGE2 and Wnt was also conserved during regeneration of other organ systems. Our work provides in vivo evidence that Wnt activation in stem cells requires PGE2, and suggests the PGE2/Wnt interaction is a master regulator of vertebrate regeneration and recovery.


Subject(s)
Dinoprostone/metabolism , Embryonic Development , Hematopoietic Stem Cells/metabolism , Wnt Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Proliferation , Cell Survival , Embryonic Stem Cells/metabolism , Liver/physiology , Mice , Regeneration , Signal Transduction , Zebrafish/embryology , beta Catenin/metabolism
10.
Dev Biol ; 320(1): 161-74, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18585699

ABSTRACT

Developmental signaling pathways hold the keys to unlocking the promise of adult tissue regeneration, and to inhibiting carcinogenesis. Patients with mutations in the Adenomatous Polyposis Coli (APC) gene are at increased risk of developing hepatoblastoma, an embryonal form of liver cancer, suggesting that Wnt affects hepatic progenitor cells. To elucidate the role of APC loss and enhanced Wnt activity in liver development, we examined APC mutant and wnt inducible transgenic zebrafish. APC(+/-) embryos developed enlarged livers through biased induction of hepatic gene programs and increased proliferation. Conversely, APC(-/-) embryos formed no livers. Blastula transplantations determined that the effects of APC loss were cell autonomous. Induction of wnt modulators confirmed biphasic consequences of wnt activation: endodermal pattern formation and gene expression required suppression of wnt signaling in early somitogenesis; later, increased wnt activity altered endodermal fate by enhancing liver growth at the expense of pancreas formation; these effects persisted into the larval stage. In adult APC(+/-) zebrafish, increased wnt activity significantly accelerated liver regeneration after partial hepatectomy. Similarly, liver regeneration was significantly enhanced in APC(Min/+) mice, indicating the conserved effect of Wnt pathway activation in liver regeneration across vertebrate species. These studies reveal an important and time-dependent role for wnt signaling during liver development and regeneration.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Liver/embryology , Mutation/genetics , Signal Transduction , Wnt Proteins/metabolism , Zebrafish/embryology , Animals , Apoptosis , Body Patterning , Cell Lineage , Cell Proliferation , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endoderm/cytology , Endoderm/embryology , Hepatectomy , Hepatocytes/cytology , Liver/cytology , Liver Regeneration , Phenotype , Stem Cells/cytology , Time Factors , beta Catenin/metabolism
11.
Cell Cycle ; 6(24): 3054-7, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18075310

ABSTRACT

We have recently demonstrated through a chemical screen in the zebrafish embryo that prostaglandin E2 (PGE2) is an evolutionarily conserved regulator of hematopoietic stem cell (HSC) number. These results have further been confirmed by in vitro and in vivo studies in the murine model. Bioactive PGE2 derivatives have potential clinical application to accelerate recovery of the hematopoietic system following chemotherapy or irradiation. Ex vivo expansion of HSCs prior to stem cell transplantation may improve reconstitution of hematopoiesis and immune function. This article aims to summarize current knowledge of PGE2-mediated regulation of blood cell homeostasis as well as to discuss the proposed use of PGE2 to expand hematopoietic stem cells for transplantation in the clinical setting.


Subject(s)
Dinoprostone/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , Bone Marrow , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Signal Transduction , Zebrafish
12.
Nature ; 447(7147): 1007-11, 2007 Jun 21.
Article in English | MEDLINE | ID: mdl-17581586

ABSTRACT

Haematopoietic stem cell (HSC) homeostasis is tightly controlled by growth factors, signalling molecules and transcription factors. Definitive HSCs derived during embryogenesis in the aorta-gonad-mesonephros region subsequently colonize fetal and adult haematopoietic organs. To identify new modulators of HSC formation and homeostasis, a panel of biologically active compounds was screened for effects on stem cell induction in the zebrafish aorta-gonad-mesonephros region. Here, we show that chemicals that enhance prostaglandin (PG) E2 synthesis increased HSC numbers, and those that block prostaglandin synthesis decreased stem cell numbers. The cyclooxygenases responsible for PGE2 synthesis were required for HSC formation. A stable derivative of PGE2 improved kidney marrow recovery following irradiation injury in the adult zebrafish. In murine embryonic stem cell differentiation assays, PGE2 caused amplification of multipotent progenitors. Furthermore, ex vivo exposure to stabilized PGE2 enhanced spleen colony forming units at day 12 post transplant and increased the frequency of long-term repopulating HSCs present in murine bone marrow after limiting dilution competitive transplantation. The conserved role for PGE2 in the regulation of vertebrate HSC homeostasis indicates that modulation of the prostaglandin pathway may facilitate expansion of HSC number for therapeutic purposes.


Subject(s)
Dinoprostone/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Homeostasis/drug effects , Vertebrates , Animals , Cell Differentiation/drug effects , Core Binding Factor Alpha 2 Subunit/genetics , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Gene Expression Regulation/drug effects , Mice , Proto-Oncogene Proteins c-myb/genetics , Vertebrates/embryology , Zebrafish/embryology , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...