Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 25(6): 1007-1019, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816617

ABSTRACT

Rare multipotent stem cells replenish millions of blood cells per second through a time-consuming process, passing through multiple stages of increasingly lineage-restricted progenitors. Although insults to the blood-forming system highlight the need for more rapid blood replenishment from stem cells, established models of hematopoiesis implicate only one mandatory differentiation pathway for each blood cell lineage. Here, we establish a nonhierarchical relationship between distinct stem cells that replenish all blood cell lineages and stem cells that replenish almost exclusively platelets, a lineage essential for hemostasis and with important roles in both the innate and adaptive immune systems. These distinct stem cells use cellularly, molecularly and functionally separate pathways for the replenishment of molecularly distinct megakaryocyte-restricted progenitors: a slower steady-state multipotent pathway and a fast-track emergency-activated platelet-restricted pathway. These findings provide a framework for enhancing platelet replenishment in settings in which slow recovery of platelets remains a major clinical challenge.


Subject(s)
Blood Platelets , Cell Differentiation , Hematopoietic Stem Cells , Megakaryocytes , Blood Platelets/immunology , Blood Platelets/metabolism , Animals , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Cell Differentiation/immunology , Megakaryocytes/cytology , Cell Lineage , Mice, Inbred C57BL , Hematopoiesis , Thrombopoiesis , Mice, Knockout , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/immunology
2.
Nat Immunol ; 24(6): 915-924, 2023 06.
Article in English | MEDLINE | ID: mdl-37081147

ABSTRACT

Immune cell locomotion is associated with amoeboid migration, a flexible mode of movement, which depends on rapid cycles of actin polymerization and actomyosin contraction1. Many immune cells do not necessarily require integrins, the major family of adhesion receptors in mammals, to move productively through three-dimensional tissue spaces2,3. Instead, they can use alternative strategies to transmit their actin-driven forces to the substrate, explaining their migratory adaptation to changing external environments4-6. However, whether these generalized concepts apply to all immune cells is unclear. Here, we show that the movement of mast cells (immune cells with important roles during allergy and anaphylaxis) differs fundamentally from the widely applied paradigm of interstitial immune cell migration. We identify a crucial role for integrin-dependent adhesion in controlling mast cell movement and localization to anatomical niches rich in KIT ligand, the major mast cell growth and survival factor. Our findings show that substrate-dependent haptokinesis is an important mechanism for the tissue organization of resident immune cells.


Subject(s)
Actins , Integrins , Animals , Integrins/metabolism , Actins/metabolism , Mast Cells/metabolism , Cell Movement , Leukocytes/metabolism , Cell Adhesion , Mammals/metabolism
3.
Nat Immunol ; 17(6): 666-676, 2016 06.
Article in English | MEDLINE | ID: mdl-27043410

ABSTRACT

According to current models of hematopoiesis, lymphoid-primed multi-potent progenitors (LMPPs) (Lin(-)Sca-1(+)c-Kit(+)CD34(+)Flt3(hi)) and common myeloid progenitors (CMPs) (Lin(-)Sca-1(+)c-Kit(+)CD34(+)CD41(hi)) establish an early branch point for separate lineage-commitment pathways from hematopoietic stem cells, with the notable exception that both pathways are proposed to generate all myeloid innate immune cell types through the same myeloid-restricted pre-granulocyte-macrophage progenitor (pre-GM) (Lin(-)Sca-1(-)c-Kit(+)CD41(-)FcγRII/III(-)CD150(-)CD105(-)). By single-cell transcriptome profiling of pre-GMs, we identified distinct myeloid differentiation pathways: a pathway expressing the gene encoding the transcription factor GATA-1 generated mast cells, eosinophils, megakaryocytes and erythroid cells, and a pathway lacking expression of that gene generated monocytes, neutrophils and lymphocytes. These results identify an early hematopoietic-lineage bifurcation that separates the myeloid lineages before their segregation from other hematopoietic-lineage potential.


Subject(s)
Cell Differentiation , Cell Lineage , Lymphocytes/physiology , Myeloid Cells/physiology , Myeloid Progenitor Cells/physiology , Animals , Antigens, CD/metabolism , Cells, Cultured , Computational Biology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoiesis , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Sequence Analysis, RNA , Single-Cell Analysis , Tissue Array Analysis , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
4.
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
5.
Blood ; 142(19): 1622-1632, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37562000

ABSTRACT

A critical regulatory role of hematopoietic stem cell (HSC) vascular niches in the bone marrow has been implicated to occur through endothelial niche cell expression of KIT ligand. However, endothelial-derived KIT ligand is expressed in both a soluble and membrane-bound form and not unique to bone marrow niches, and it is also systemically distributed through the circulatory system. Here, we confirm that upon deletion of both the soluble and membrane-bound forms of endothelial-derived KIT ligand, HSCs are reduced in mouse bone marrow. However, the deletion of endothelial-derived KIT ligand was also accompanied by reduced soluble KIT ligand levels in the blood, precluding any conclusion as to whether the reduction in HSC numbers reflects reduced endothelial expression of KIT ligand within HSC niches, elsewhere in the bone marrow, and/or systemic soluble KIT ligand produced by endothelial cells outside of the bone marrow. Notably, endothelial deletion, specifically of the membrane-bound form of KIT ligand, also reduced systemic levels of soluble KIT ligand, although with no effect on stem cell numbers, implicating an HSC regulatory role primarily of soluble rather than membrane KIT ligand expression in endothelial cells. In support of a role of systemic rather than local niche expression of soluble KIT ligand, HSCs were unaffected in KIT ligand deleted bones implanted into mice with normal systemic levels of soluble KIT ligand. Our findings highlight the need for more specific tools to unravel niche-specific roles of regulatory cues expressed in hematopoietic niche cells in the bone marrow.


Subject(s)
Endothelial Cells , Stem Cell Factor , Mice , Animals , Stem Cell Factor/metabolism , Hematopoietic Stem Cells/metabolism , Bone Marrow/metabolism , Bone and Bones , Stem Cell Niche , Bone Marrow Cells/metabolism
6.
Nat Immunol ; 13(4): 412-9, 2012 Feb 19.
Article in English | MEDLINE | ID: mdl-22344248

ABSTRACT

The stepwise commitment from hematopoietic stem cells in the bone marrow to T lymphocyte-restricted progenitors in the thymus represents a paradigm for understanding the requirement for distinct extrinsic cues during different stages of lineage restriction from multipotent to lineage-restricted progenitors. However, the commitment stage at which progenitors migrate from the bone marrow to the thymus remains unclear. Here we provide functional and molecular evidence at the single-cell level that the earliest progenitors in the neonatal thymus had combined granulocyte-monocyte, T lymphocyte and B lymphocyte lineage potential but not megakaryocyte-erythroid lineage potential. These potentials were identical to those of candidate thymus-seeding progenitors in the bone marrow, which were closely related at the molecular level. Our findings establish the distinct lineage-restriction stage at which the T cell lineage-commitment process transits from the bone marrow to the remote thymus.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage/immunology , Lymphoid Progenitor Cells/cytology , Myeloid Cells/cytology , Precursor Cells, B-Lymphoid/cytology , T-Lymphocytes/cytology , Animals , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Thymus Gland/cytology
7.
EMBO Rep ; 23(9): e53221, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35848459

ABSTRACT

The effect of radiation therapy on tumor vasculature has long been a subject of debate. Increased oxygenation and perfusion have been documented during radiation therapy. Conversely, apoptosis of endothelial cells in irradiated tumors has been proposed as a major contributor to tumor control. To examine these contradictions, we use multiphoton microscopy in two murine tumor models: MC38, a highly vascularized, and B16F10, a moderately vascularized model, grown in transgenic mice with tdTomato-labeled endothelium before and after a single (15 Gy) or fractionated (5 × 3 Gy) dose of radiation. Unexpectedly, even these high doses lead to little structural change of the perfused vasculature. Conversely, non-perfused vessels and blind ends are substantially impaired after radiation accompanied by apoptosis and reduced proliferation of their endothelium. RNAseq analysis of tumor endothelial cells confirms the modification of gene expression in apoptotic and cell cycle regulation pathways after irradiation. Therefore, we conclude that apoptosis of tumor endothelial cells after radiation does not impair vascular structure.


Subject(s)
Endothelial Cells , Neoplasms , Animals , Apoptosis , Endothelial Cells/metabolism , Endothelium/metabolism , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/radiotherapy , Radiation, Ionizing
8.
J Immunol ; 208(2): 358-370, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34903641

ABSTRACT

Dendritic cells (DCs) are heterogeneous immune regulators involved in autoimmune diseases. Epigenomic mechanisms orchestrating DC development and DC subset diversification remain insufficiently understood but could be important to modulate DC fate for clinical purposes. By combining whole-genome methylation assessment with the analysis of mice expressing reduced DNA methyltransferase 1 levels, we show that distinct DNA methylation levels and patterns are required for the development of plasmacytoid DC and conventional DC subsets. We provide clonal in vivo evidence for DC lineage establishment at the stem cell level, and we show that a high DNA methylation threshold level is essential for Flt3-dependent survival of DC precursors. Importantly, reducing methylation predominantly depletes plasmacytoid DC and alleviates systemic lupus erythematosus in an autoimmunity mouse model. This study shows how DNA methylation regulates the production of DC subsets and provides a potential rationale for targeting autoimmune disease using hypomethylating agents.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/genetics , Dendritic Cells/immunology , Homeostasis/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Autoimmunity/genetics , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout
9.
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
10.
Nature ; 535(7611): 299-302, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27411635

ABSTRACT

The mechanisms underlying haematopoietic lineage decisions remain disputed. Lineage-affiliated transcription factors with the capacity for lineage reprogramming, positive auto-regulation and mutual inhibition have been described as being expressed in uncommitted cell populations. This led to the assumption that lineage choice is cell-intrinsically initiated and determined by stochastic switches of randomly fluctuating cross-antagonistic transcription factors. However, this hypothesis was developed on the basis of RNA expression data from snapshot and/or population-averaged analyses. Alternative models of lineage choice therefore cannot be excluded. Here we use novel reporter mouse lines and live imaging for continuous single-cell long-term quantification of the transcription factors GATA1 and PU.1 (also known as SPI1). We analyse individual haematopoietic stem cells throughout differentiation into megakaryocytic-erythroid and granulocytic-monocytic lineages. The observed expression dynamics are incompatible with the assumption that stochastic switching between PU.1 and GATA1 precedes and initiates megakaryocytic-erythroid versus granulocytic-monocytic lineage decision-making. Rather, our findings suggest that these transcription factors are only executing and reinforcing lineage choice once made. These results challenge the current prevailing model of early myeloid lineage choice.


Subject(s)
Cell Differentiation , Cell Lineage , GATA1 Transcription Factor/metabolism , Myeloid Cells/cytology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Erythrocytes/cytology , Feedback, Physiological , Female , Genes, Reporter , Granulocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Male , Megakaryocytes/cytology , Mice , Models, Biological , Monocytes/cytology , Reproducibility of Results , Single-Cell Analysis , Stochastic Processes
11.
Blood ; 133(8): 816-819, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30301719

ABSTRACT

The molecular mechanisms governing the transition from hematopoietic stem cells (HSCs) to lineage-committed progenitors remain poorly understood. Transcription factors (TFs) are powerful cell intrinsic regulators of differentiation and lineage commitment, while cytokine signaling has been shown to instruct the fate of progenitor cells. However, the direct regulation of differentiation-inducing hematopoietic TFs by cell extrinsic signals remains surprisingly difficult to establish. PU.1 is a master regulator of hematopoiesis and promotes myeloid differentiation. Here we report that tumor necrosis factor (TNF) can directly and rapidly upregulate PU.1 protein in HSCs in vitro and in vivo. We demonstrate that in vivo, niche-derived TNF is the principal PU.1 inducing signal in HSCs and is both sufficient and required to relay signals from inflammatory challenges to HSCs.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/metabolism , Myelopoiesis , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Hematopoietic Stem Cells/pathology , Inflammation/metabolism , Inflammation/pathology , Mice , Stem Cell Niche
12.
Int J Cancer ; 146(12): 3410-3422, 2020 06 15.
Article in English | MEDLINE | ID: mdl-31721195

ABSTRACT

RuvBL1 is an AAA+ ATPase whose expression in hepatocellular carcinoma (HCC) correlates with a poor prognosis. In vitro models suggest that targeting RuvBL1 could be an effective strategy against HCC. However, the role of RuvBL1 in the onset and progression of HCC remains unknown. To address this question, we developed a RuvBL1hep+/- mouse model and evaluated the outcome of DEN-induced liver carcinogenesis up to 12 months of progression. We found that RuvBL1 haploinsufficiency initially delayed the onset of liver cancer, due to a reduced hepatocyte turnover in RuvBL1hep+/- mice. However, RuvBL1hep+/- mice eventually developed HCC nodules that, with aging, grew larger than in the control mice. Moreover, RuvBL1hep+/- mice developed hepatic insulin resistance and impaired glucose homeostasis. We could determine that RuvBL1 regulates insulin signaling through the Akt/mTOR pathway in liver physiology in vivo as well as in normal hepatocytic and HCC cells in vitro. Whole transcriptome analysis of mice livers confirmed the major role of RuvBL1 in the regulation of hepatic glucose metabolism. Finally, RuvBL1 expression was found significantly correlated to glucose metabolism and mTOR signaling by bioinformatic analysis of human HCC sample from the publicly available TGCA database. These data uncover a role of RuvBL1 at the intersection of liver metabolism, hepatocyte proliferation and HCC development, providing a molecular rationale for its overexpression in liver cancer.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , DNA Helicases/genetics , Insulin Resistance/genetics , Liver Neoplasms/genetics , Liver/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cohort Studies , DNA Helicases/metabolism , Datasets as Topic , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Disease Models, Animal , Disease Progression , Disease-Free Survival , Glucose/metabolism , Haploinsufficiency , Hepatocytes/metabolism , Humans , Insulin/metabolism , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
13.
Blood ; 131(15): 1712-1719, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29339402

ABSTRACT

Although an essential role for canonical Notch signaling in generation of hematopoietic stem cells in the embryo and in thymic T-cell development is well established, its role in adult bone marrow (BM) myelopoiesis remains unclear. Some studies, analyzing myeloid progenitors in adult mice with inhibited Notch signaling, implicated distinct roles of canonical Notch signaling in regulation of progenitors for the megakaryocyte, erythroid, and granulocyte-macrophage cell lineages. However, these studies might also have targeted other pathways. Therefore, we specifically deleted, in adult BM, the transcription factor recombination signal-binding protein J κ (Rbpj), through which canonical signaling from all Notch receptors converges. Notably, detailed progenitor staging established that canonical Notch signaling is fully dispensable for all investigated stages of megakaryocyte, erythroid, and myeloid progenitors in steady state unperturbed hematopoiesis, after competitive BM transplantation, and in stress-induced erythropoiesis. Moreover, expression of key regulators of these hematopoietic lineages and Notch target genes were unaffected by Rbpj deficiency in BM progenitor cells.


Subject(s)
Bone Marrow/metabolism , Erythropoiesis , Myelopoiesis , Receptors, Notch/metabolism , Signal Transduction , Stress, Physiological , Animals , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mice, Transgenic , Receptors, Notch/genetics
14.
EMBO Rep ; 19(10)2018 10.
Article in English | MEDLINE | ID: mdl-30166337

ABSTRACT

Few studies report on the in vivo requirement for hematopoietic niche factors in the mammalian embryo. Here, we comprehensively analyze the requirement for Kit ligand (Kitl) in the yolk sac and aorta-gonad-mesonephros (AGM) niche. In-depth analysis of loss-of-function and transgenic reporter mouse models show that Kitl-deficient embryos harbor decreased numbers of yolk sac erythro-myeloid progenitor (EMP) cells, resulting from a proliferation defect following their initial emergence. This EMP defect causes a dramatic decrease in fetal liver erythroid cells prior to the onset of hematopoietic stem cell (HSC)-derived erythropoiesis, and a reduction in tissue-resident macrophages. Pre-HSCs in the AGM require Kitl for survival and maturation, but not proliferation. Although Kitl is expressed widely in all embryonic hematopoietic niches, conditional deletion in endothelial cells recapitulates germline loss-of-function phenotypes in AGM and yolk sac, with phenotypic HSCs but not EMPs remaining dependent on endothelial Kitl upon migration to the fetal liver. In conclusion, our data establish Kitl as a critical regulator in the in vivoAGM and yolk sac endothelial niche.


Subject(s)
Embryonic Development/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Stem Cell Factor/genetics , Animals , Aorta/growth & development , Cell Lineage/genetics , Cell Proliferation/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Erythropoiesis/genetics , Gene Expression Regulation, Developmental/genetics , Gonads/growth & development , Mesonephros/growth & development , Mice , Mice, Transgenic , Stem Cell Niche/genetics , Yolk Sac/growth & development
15.
Haematologica ; 104(11): 2215-2224, 2019 11.
Article in English | MEDLINE | ID: mdl-30975913

ABSTRACT

Somatic mutations in acute myeloid leukemia are acquired sequentially and hierarchically. First, pre-leukemic mutations, such as t(8;21) that encodes AML1-ETO, are acquired within the hematopoietic stem cell (HSC) compartment, while signaling pathway mutations, including KRAS activating mutations, are late events acquired during transformation of leukemic progenitor cells and are rarely detectable in HSC. This raises the possibility that signaling pathway mutations are detrimental to clonal expansion of pre-leukemic HSC. To address this hypothesis, we used conditional genetics to introduce Aml1-ETO and K-RasG12D into murine HSC, either individually or in combination. In the absence of activated Ras, Aml1-ETO-expressing HSC conferred a competitive advantage. However, activated K-Ras had a marked detrimental effect on Aml1-ETO-expressing HSC, leading to loss of both phenotypic and functional HSC. Cell cycle analysis revealed a loss of quiescence in HSC co-expressing Aml1-ETO and K-RasG12D, accompanied by an enrichment in E2F and Myc target gene expression and depletion of HSC self-renewal-associated gene expression. These findings provide a mechanistic basis for the observed absence of KRAS signaling mutations in the pre-malignant HSC compartment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoietic Stem Cells/metabolism , Mutation , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , Animals , Cell Proliferation/genetics , Gene Expression , Gene Expression Profiling , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Transgenic , Models, Animal , Models, Biological , Precancerous Conditions/genetics , Precancerous Conditions/metabolism
16.
J Immunol ; 199(9): 3031-3041, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28939758

ABSTRACT

The mechanisms leading to autoimmune and inflammatory diseases in the CNS have not been elucidated. The environmental triggers of the aberrant presence of CD4+ T cells in the CNS are not known. In this article, we report that abnormal ß-catenin expression in T cells drives a fatal neuroinflammatory disease in mice that is characterized by CNS infiltration of T cells, glial activation, and progressive loss of motor function. We show that enhanced ß-catenin expression in T cells leads to aberrant and Th1-biased T cell activation, enhanced expression of integrin α4ß1, and infiltration of activated T cells into the spinal cord, without affecting regulatory T cell function. Importantly, expression of ß-catenin in mature naive T cells was sufficient to drive integrin α4ß1 expression and CNS migration, whereas pharmacologic inhibition of integrin α4ß1 reduced the abnormal T cell presence in the CNS of ß-catenin-expressing mice. Together, these results implicate deregulation of the Wnt/ß-catenin pathway in CNS inflammation and suggest novel therapeutic strategies for neuroinflammatory disorders.


Subject(s)
Integrin alpha4beta1/immunology , Spinal Cord Diseases/immunology , Spinal Cord/immunology , Th1 Cells/immunology , Wnt Signaling Pathway/immunology , beta Catenin/immunology , Animals , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Integrin alpha4beta1/genetics , Mice , Mice, Knockout , Spinal Cord/pathology , Spinal Cord Diseases/genetics , Spinal Cord Diseases/pathology , Th1 Cells/pathology , Wnt Signaling Pathway/genetics , beta Catenin/genetics
17.
Nature ; 502(7470): 232-6, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23934107

ABSTRACT

The blood system is maintained by a small pool of haematopoietic stem cells (HSCs), which are required and sufficient for replenishing all human blood cell lineages at millions of cells per second throughout life. Megakaryocytes in the bone marrow are responsible for the continuous production of platelets in the blood, crucial for preventing bleeding--a common and life-threatening side effect of many cancer therapies--and major efforts are focused at identifying the most suitable cellular and molecular targets to enhance platelet production after bone marrow transplantation or chemotherapy. Although it has become clear that distinct HSC subsets exist that are stably biased towards the generation of lymphoid or myeloid blood cells, we are yet to learn whether other types of lineage-biased HSC exist or understand their inter-relationships and how differently lineage-biased HSCs are generated and maintained. The functional relevance of notable phenotypic and molecular similarities between megakaryocytes and bone marrow cells with an HSC cell-surface phenotype remains unclear. Here we identify and prospectively isolate a molecularly and functionally distinct mouse HSC subset primed for platelet-specific gene expression, with enhanced propensity for short- and long-term reconstitution of platelets. Maintenance of platelet-biased HSCs crucially depends on thrombopoietin, the primary extrinsic regulator of platelet development. Platelet-primed HSCs also frequently have a long-term myeloid lineage bias, can self-renew and give rise to lymphoid-biased HSCs. These findings show that HSC subtypes can be organized into a cellular hierarchy, with platelet-primed HSCs at the apex. They also demonstrate that molecular and functional priming for platelet development initiates already in a distinct HSC population. The identification of a platelet-primed HSC population should enable the rational design of therapies enhancing platelet output.


Subject(s)
Blood Platelets/cytology , Cell Differentiation , Hematopoietic Stem Cells/cytology , Animals , Cell Lineage/genetics , Female , Gene Expression Regulation, Developmental , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL
18.
Br J Haematol ; 183(4): 588-600, 2018 11.
Article in English | MEDLINE | ID: mdl-30596405

ABSTRACT

Given that FLT3 expression is highly restricted on lymphoid progenitors, it is possible that the established role of FLT3 in the regulation of B and T lymphopoiesis reflects its high expression and role in regulation of lymphoid-primed multipotent progenitors (LMPPs) or common lymphoid progenitors (CLPs). We generated a Flt3 conditional knock-out (Flt3fl/fl) mouse model to address the direct role of FLT3 in regulation of lymphoid-restricted progenitors, subsequent to turning on Rag1 expression, as well as potentially ontogeny-specific roles in B and T lymphopoiesis. Our studies establish a prominent and direct role of FLT3, independently of the established role of FLT3 in regulation of LMPPs and CLPs, in regulation of fetal as well as adult early B cell progenitors, and the early thymic progenitors (ETPs) in adult mice but not in the fetus. Our findings highlight the potential benefit of targeting poor prognosis acute B-cell progenitor leukaemia and ETP leukaemia with recurrent FLT3 mutations using clinical FLT3 inhibitors.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Bone Marrow Cells/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lymphoid Progenitor Cells/pathology , Mice , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thymus Gland/metabolism , Thymus Gland/pathology , fms-Like Tyrosine Kinase 3/genetics
19.
Blood ; 126(10): 1214-23, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26162409

ABSTRACT

In-depth molecular investigation of familial leukemia has been limited by the rarity of recognized cases. This study examines the genetic events initiating leukemia and details the clinical progression of disease across multiple families harboring germ-line CEBPA mutations. Clinical data were collected from 10 CEBPA-mutated families, representing 24 members with acute myeloid leukemia (AML). Whole-exome (WES) and deep sequencing were performed to genetically profile tumors and define patterns of clonal evolution. Germline CEBPA mutations clustered within the N-terminal and were highly penetrant, with AML presenting at a median age of 24.5 years (range, 1.75-46 years). In all diagnostic tumors tested (n = 18), double CEBPA mutations (CEBPAdm) were detected, with acquired (somatic) mutations preferentially targeting the C-terminal. Somatic CEBPA mutations were unstable throughout the disease course, with different mutations identified at recurrence. Deep sequencing of diagnostic and relapse paired samples confirmed that relapse-associated CEBPA mutations were absent at diagnosis, suggesting recurrence was triggered by novel, independent clones. Integrated WES and deep sequencing subsequently revealed an entirely new complement of mutations at relapse, verifying the presentation of a de novo leukemic episode. The cumulative incidence of relapse in familial AML was 56% at 10 years (n = 11), and 3 patients experienced ≥3 disease episodes over a period of 17 to 20 years. Durable responses to secondary therapies were observed, with prolonged median survival after relapse (8 years) and long-term overall survival (10-year overall survival, 67%). Our data reveal that familial CEBPA-mutated AML exhibits a unique model of disease progression, associated with favorable long-term outcomes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Germ-Line Mutation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Adolescent , Adult , Child , Child, Preschool , Disease Progression , Female , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Infant , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Pedigree , Young Adult
20.
Nat Chem Biol ; 11(8): 571-578, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26167872

ABSTRACT

The CEBPA gene is mutated in 9% of patients with acute myeloid leukemia (AML). Selective expression of a short (30-kDa) CCAAT-enhancer binding protein-α (C/EBPα) translational isoform, termed p30, represents the most common type of CEBPA mutation in AML. The molecular mechanisms underlying p30-mediated transformation remain incompletely understood. We show that C/EBPα p30, but not the normal p42 isoform, preferentially interacts with Wdr5, a key component of SET/MLL (SET-domain/mixed-lineage leukemia) histone-methyltransferase complexes. Accordingly, p30-bound genomic regions were enriched for MLL-dependent H3K4me3 marks. The p30-dependent increase in self-renewal and inhibition of myeloid differentiation required Wdr5, as downregulation of the latter inhibited proliferation and restored differentiation in p30-dependent AML models. OICR-9429 is a new small-molecule antagonist of the Wdr5-MLL interaction. This compound selectively inhibited proliferation and induced differentiation in p30-expressing human AML cells. Our data reveal the mechanism of p30-dependent transformation and establish the essential p30 cofactor Wdr5 as a therapeutic target in CEBPA-mutant AML.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Dihydropyridines/pharmacology , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Docking Simulation , Molecular Sequence Data , Molecular Targeted Therapy , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Signal Transduction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL