Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Ther Nucleic Acids ; 35(3): 102229, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38952440

ABSTRACT

p47 phox -deficient chronic granulomatous disease (p47-CGD) is a primary immunodeficiency caused by mutations in the neutrophil cytosolic factor 1 (NCF1) gene, resulting in defective NADPH oxidase function in phagocytes. Due to its complex genomic context, the NCF1 locus is not suited for safe gene editing with current genome editing technologies. Therefore, we developed a targeted NCF1 coding sequence knock-in by CRISPR-Cas9 ribonucleoprotein and viral vector template delivery, to restore p47 phox expression under the control of the endogenous NCF2 locus. NCF2 encodes for p67 phox , an NADPH oxidase subunit that closely interacts with p47 phox and is predominantly expressed in myeloid cells. This approach restored p47 phox expression and NADPH oxidase function in p47-CGD patient hematopoietic stem and progenitor cells (HSPCs) and in p47 phox -deficient mouse HSPCs, with the transgene expression following a myeloid differentiation pattern. Adeno-associated viral vectors performed favorably over integration-deficient lentiviral vectors for template delivery, with fewer off-target integrations and higher correction efficacy in HSPCs. Such myeloid-directed gene editing is promising for clinical CGD gene therapy, as it leads to the co-expression of p47 phox and p67 phox , ensuring spatiotemporal and near-physiological transgene expression in myeloid cells.

2.
Nat Immunol ; 23(4): 505-517, 2022 04.
Article in English | MEDLINE | ID: mdl-35354960

ABSTRACT

Intrinsic and extrinsic cues determine developmental trajectories of hematopoietic stem cells (HSCs) towards erythroid, myeloid and lymphoid lineages. Using two newly generated transgenic mice that report and trace the expression of terminal deoxynucleotidyl transferase (TdT), transient induction of TdT was detected on a newly identified multipotent progenitor (MPP) subset that lacked self-renewal capacity but maintained multilineage differentiation potential. TdT induction on MPPs reflected a transcriptionally dynamic but uncommitted stage, characterized by low expression of lineage-associated genes. Single-cell CITE-seq indicated that multipotency in the TdT+ MPPs is associated with expression of the endothelial cell adhesion molecule ESAM. Stable and progressive upregulation of TdT defined the lymphoid developmental trajectory. Collectively, we here identify a new multipotent progenitor within the MPP4 compartment. Specification and commitment are defined by downregulation of ESAM which marks the progressive loss of alternative fates along all lineages.


Subject(s)
DNA Nucleotidylexotransferase , Hematopoietic Stem Cells , Multipotent Stem Cells , Animals , Cell Differentiation , Cell Lineage/genetics , DNA Nucleotidylexotransferase/genetics , DNA Nucleotidylexotransferase/metabolism , Hematopoietic Stem Cells/physiology , Mice , Mice, Transgenic
3.
Nat Commun ; 10(1): 5734, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31844046

ABSTRACT

System-wide quantification of the cell surface proteotype and identification of extracellular glycosylation sites is challenging when samples are limited. Here, we miniaturize and automate the previously described Cell Surface Capture (CSC) technology, increasing sensitivity, reproducibility and throughput. We use this technology, which we call autoCSC, to create population-specific surfaceome maps of developing mouse B cells and use targeted flow cytometry to uncover developmental cell subpopulations.


Subject(s)
B-Lymphocyte Subsets/classification , Cell Differentiation , High-Throughput Screening Assays/methods , Membrane Proteins/analysis , Proteomics/methods , Animals , B-Lymphocyte Subsets/metabolism , Cell Membrane/metabolism , Flow Cytometry/methods , HT29 Cells , Humans , Membrane Proteins/metabolism , Mice , Primary Cell Culture , Reproducibility of Results
4.
J Exp Med ; 216(3): 638-655, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30765463

ABSTRACT

T cell development is critically dependent on successful rearrangement of antigen-receptor chains. At the ß-selection checkpoint, only cells with a functional rearrangement continue in development. However, how nonselected T cells proceed in their dead-end fate is not clear. We identified low CD27 expression to mark pre-T cells that have failed to rearrange their ß-chain. Expression profiling and single-cell transcriptome clustering identified a developmental trajectory through ß-selection and revealed specific expression of the transcription factor Duxbl at a stage of high recombination activity before ß-selection. Conditional transgenic expression of Duxbl resulted in a developmental block at the DN3-to-DN4 transition due to reduced proliferation and enhanced apoptosis, whereas RNA silencing of Duxbl led to a decrease in apoptosis. Transcriptome analysis linked Duxbl to elevated expression of the apoptosis-inducing Oas/RNaseL pathway. RNaseL deficiency or sustained Bcl2 expression led to a partial rescue of cells in Duxbl transgenic mice. These findings identify Duxbl as a regulator of ß-selection by inducing apoptosis in cells with a nonfunctional rearrangement.


Subject(s)
Homeodomain Proteins/metabolism , T-Lymphocytes/physiology , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Female , Gene Expression Regulation , Homeodomain Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/cytology , Thymus Gland/cytology , Transcription Factors/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
5.
Front Immunol ; 9: 2258, 2018.
Article in English | MEDLINE | ID: mdl-30364182

ABSTRACT

Interleukin-7 (IL-7) and Flt3-ligand (FL) are two cytokines important for the generation of B cells, as manifested by the impaired B cell development in mice deficient for either cytokine or their respective receptors and by the complete block in B cell differentiation in the absence of both cytokines. IL-7 is an important survival and proliferation factor for B cell progenitors, whereas FL acts on several early developmental stages, prior to B cell commitment. We have generated mice constitutively over-expressing both IL-7 and FL. These double transgenic mice develop splenomegaly and lymphadenopathy characterized by tremendously enlarged lymph nodes even in young animals. Lymphoid, myeloid and dendritic cell numbers are increased compared to mice over-expressing either of the two cytokines alone and the effect on their expansion is synergistic, rather than additive. B cell progenitors, early progenitors with myeloid and lymphoid potential (EPLM), common lymphoid progenitors (CLP) and lineage-, Sca1+, kit+ (LSK) cells are all increased not only in the bone marrow but also in peripheral blood, spleen and even lymph nodes. When transplanted into irradiated wild-type mice, lymph node cells show long-term multilineage reconstitution, further confirming the presence of functional hematopoietic progenitors therein. Our double transgenic mouse model shows that sustained and combined over-expression of IL-7 and FL leads to a massive expansion of most bone marrow hematopoietic progenitors and to their associated presence in peripheral lymphoid organs where they reside and potentially differentiate further, thus leading to the synergistic increase in mature lymphoid and myeloid cell numbers. The present study provides further in vivo evidence for the concerted action of IL-7 and FL on lymphopoiesis and suggests that extramedullary niches, including those in lymph nodes, can support the survival and maintenance of hematopoietic progenitors that under physiological conditions develop exclusively in the bone marrow.


Subject(s)
Hematopoietic Stem Cells/immunology , Interleukin-7/immunology , Lymphoid Progenitor Cells/immunology , Membrane Proteins/immunology , Multipotent Stem Cells/immunology , Animals , Cell Proliferation/genetics , Cell Survival/genetics , Cell Survival/immunology , Gene Expression/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukin-7/genetics , Interleukin-7/metabolism , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism
6.
Int J Mol Sci ; 19(7)2018 Jul 21.
Article in English | MEDLINE | ID: mdl-30037064

ABSTRACT

Evidence from studies of the behaviour of stem and progenitor cells and of the influence of cytokines on their fate determination, has recently led to a revised view of the process by which hematopoietic stem cells and their progeny give rise to the many different types of blood and immune cells. The new scenario abandons the classical view of a rigidly demarcated lineage tree and replaces it with a much more continuum-like view of the spectrum of fate options open to hematopoietic stem cells and their progeny. This is in contrast to previous lineage diagrams, which envisaged stem cells progressing stepwise through a series of fairly-precisely described intermediate progenitors in order to close down alternative developmental options. Instead, stem and progenitor cells retain some capacity to step sideways and adopt alternative, closely related, fates, even after they have "made a lineage choice." The stem and progenitor cells are more inherently versatile than previously thought and perhaps sensitive to lineage guidance by environmental cues. Here we examine the evidence that supports these views and reconsider the meaning of cell lineages in the context of a continuum model of stem cell fate determination and environmental modulation.


Subject(s)
Hematopoiesis/physiology , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Cytokines/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans
7.
Immunol Cell Biol ; 96(9): 898-911, 2018 10.
Article in English | MEDLINE | ID: mdl-29637611

ABSTRACT

For more than 30 years, the scheme whereby bone marrow hematopoietic stem cells give rise to the many different types of blood and immune cells has been represented as a lineage tree diagram. In this model, hematopoietic stem cells follow a preferred route to each of the end-cell types and gradually restrict their other lineage options via a series of intermediate oligo-potent progenitors. Recent findings of lineage biases or affiliations within hematopoietic stem and progenitor cells that are either pluripotent or uni-potent show that a continuum of fate options is open to hematopoietic stem cells. These results support the view that in order to close down developmental options, hematopoietic stem cells can make an immediate lineage choice rather than become gradually committed as they progress step-wise through a series of intermediate progenitors. In this scenario, there is inherent versatility in that developing cells are still able to move sideways to adopt an alternative lineage fate. Here, we examine the information that is leading toward this very different viewpoint of blood cell development.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Cytokines/metabolism , Hematopoietic Stem Cells/metabolism , Humans
8.
EMBO J ; 36(24): 3619-3633, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29030486

ABSTRACT

Single-cell RNA sequencing is a powerful technology for assessing heterogeneity within defined cell populations. Here, we describe the heterogeneity of a B220+CD117intCD19-NK1.1- uncommitted hematopoietic progenitor having combined lymphoid and myeloid potential. Phenotypic and functional assays revealed four subpopulations within the progenitor with distinct lineage developmental potentials. Among them, the Ly6D+SiglecH-CD11c- fraction was lymphoid-restricted exhibiting strong B-cell potential, whereas the Ly6D-SiglecH-CD11c- fraction showed mixed lympho-myeloid potential. Single-cell RNA sequencing of these subsets revealed that the latter population comprised a mixture of cells with distinct lymphoid and myeloid transcriptional signatures and identified a subgroup as the potential precursor of Ly6D+SiglecH-CD11c- Subsequent functional assays confirmed that B220+CD117intCD19-NK1.1- single cells are, with rare exceptions, not bipotent for lymphoid and myeloid lineages. A B-cell priming gradient was observed within the Ly6D+SiglecH-CD11c- subset and we propose a herein newly identified subgroup as the direct precursor of the first B-cell committed stage. Therefore, the apparent multipotency of B220+CD117intCD19-NK1.1- progenitors results from underlying heterogeneity at the single-cell level and highlights the validity of single-cell transcriptomics for resolving cellular heterogeneity and developmental relationships among hematopoietic progenitors.


Subject(s)
Hematopoietic Stem Cells/physiology , Sequence Analysis, RNA/methods , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation , Cell Lineage , Female , Gene Expression Profiling , Genetic Heterogeneity , Hematopoietic Stem Cells/cytology , High-Throughput Nucleotide Sequencing , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/physiology , Male , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/physiology , Single-Cell Analysis
9.
Int J Mol Sci ; 18(5)2017 May 12.
Article in English | MEDLINE | ID: mdl-28498310

ABSTRACT

The fms-like tyrosine kinase 3 (Flt3) is a cell surface receptor that is expressed by various hematopoietic progenitor cells (HPC) and Flt3-activating mutations are commonly present in acute myeloid and lymphoid leukemias. These findings underscore the importance of Flt3 to steady-state and malignant hematopoiesis. In this study, the expression of Flt3 protein and Flt3 mRNA by single cells within the hematopoietic stem cell (HSC) and HPC bone marrow compartments of C57/BL6 mice was investigated using flow cytometry and the quantitative reverse transcription polymerase chain reaction. Flt3 was heterogeneously expressed by almost all of the populations studied, including long-term reconstituting HSC and short-term reconstituting HSC. The erythropoietin receptor (EpoR) and macrophage colony-stimulating factor receptor (M-CSFR) were also found to be heterogeneously expressed within the multipotent cell compartments. Co-expression of the mRNAs encoding Flt3 and EpoR rarely occurred within these compartments. Expression of both Flt3 and M-CSFR protein at the surface of single cells was more commonly observed. These results emphasize the heterogeneous nature of HSC and HPC and the new sub-populations identified are important to understanding the origin and heterogeneity of the acute myeloid leukemias.


Subject(s)
Hematopoietic Stem Cells/metabolism , fms-Like Tyrosine Kinase 3/genetics , Animals , Hematopoiesis , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , fms-Like Tyrosine Kinase 3/metabolism
10.
Int J Mol Sci ; 18(6)2017 May 24.
Article in English | MEDLINE | ID: mdl-28538663

ABSTRACT

The cytokine Fms-like tyrosine kinase 3 ligand (FL) is an important regulator of hematopoiesis. Its receptor, Flt3, is expressed on myeloid, lymphoid and dendritic cell progenitors and is considered an important growth and differentiation factor for several hematopoietic lineages. Activating mutations of Flt3 are frequently found in acute myeloid leukemia (AML) patients and associated with a poor clinical prognosis. In the present review we provide an overview of our current knowledge on the role of FL in the generation of blood cell lineages. We examine recent studies on Flt3 expression by hematopoietic stem cells and its potential instructive action at early stages of hematopoiesis. In addition, we review current findings on the role of mutated FLT3 in leukemia and the development of FLT3 inhibitors for therapeutic use to treat AML. The importance of mouse models in elucidating the role of Flt3-ligand in normal and malignant hematopoiesis is discussed.


Subject(s)
Hematopoiesis , Leukemia, Myeloid/metabolism , Membrane Proteins/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Drug Discovery , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Membrane Proteins/genetics , Mutation , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics
11.
Eur J Immunol ; 47(2): 394-405, 2017 02.
Article in English | MEDLINE | ID: mdl-27925658

ABSTRACT

Up to now long-term in vitro growth of pro-B cells was thought to require stromal cells. However, here we show that fetal liver (FL) and bone marrow (BM) derived pro-B cells can be propagated long-term in stromal cell-free cultures supplemented with IL-7, stem cell factor and FLT3 ligand. Within a week, most cells expressed surface CD19, CD79A, λ5, and VpreB antigens and had rearranged immunoglobulin D-J heavy chain genes. Both FL and BM pro-B cells reconstituted the B-cell compartments of immuno-incompetent Rag2-deficient mice, with FL pro-B cells generating follicular, marginal zone (MZB) and B1a B cells, and BM pro-B cells giving rise mainly to MZB cells. Reconstituted Rag2-deficient mice generated significant levels of IgM and IgG antibodies to a type II T-independent antigen; mice reconstituted with FL pro-B cells generated surprisingly high IgG1 titers. Finally, we show for the first time that mice reconstituted with mixtures of pro-B and pro-T cells propagated in stromal cell-free in vitro cultures mounted a T-cell-dependent antibody response. This novel stromal cell-free culture system facilitates our understanding of B-cell development and might be applied clinically.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Precursor Cells, B-Lymphoid/immunology , Animals , Antibody Formation , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , Interleukin-7/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, T-Lymphoid/immunology , Stem Cell Factor/metabolism , Stromal Cells/immunology , Transplantation Chimera
12.
Proc Natl Acad Sci U S A ; 113(50): E8122-E8130, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911806

ABSTRACT

Hematopoietic cells are continuously generated throughout life from hematopoietic stem cells, thus making hematopoiesis a favorable system to study developmental cell lineage commitment. The main factors incorporating environmental signals to developing hematopoietic cells are cytokines, which regulate commitment of hematopoietic progenitors to the different blood lineages by acting either in an instructive or a permissive manner. Fms-like tyrosine kinase-3 (Flt3) ligand (FL) and Interleukin-7 (IL-7) are cytokines pivotal for B-cell development, as manifested by the severely compromised B-cell development in their absence. However, their precise role in regulating B-cell commitment has been the subject of debate. In the present study we assessed the rescue of B-cell commitment in mice lacking IL-7 but simultaneously overexpressing FL. Results obtained demonstrate that FL overexpression in IL-7-deficient mice rescues B-cell commitment, resulting in significant Ebf1 and Pax5 expression in Ly6D+CD135+CD127+CD19- precursors and subsequent generation of normal numbers of CD19+ B-cell progenitors, therefore indicating that IL-7 can be dispensable for commitment to the B-cell lineage. Further analysis of Ly6D+CD135+CD127+CD19- progenitors in IL-7- or FL-deficient mice overexpressing Bcl2, as well as in IL-7 transgenic mice suggests that both FL and IL-7 regulate B-cell commitment in a permissive manner: FL by inducing proliferation of Ly6D+CD135+CD127+CD19- progenitors and IL-7 by providing survival signals to these progenitors.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Lineage/immunology , Interleukin-7/metabolism , Membrane Proteins/metabolism , Animals , Antigens, CD19/metabolism , Antigens, Ly/metabolism , B-Lymphocytes/metabolism , Cell Proliferation , Cell Survival , Female , GPI-Linked Proteins/metabolism , Hematopoiesis/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Interleukin-7/deficiency , Interleukin-7/genetics , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
13.
Acta Cardiol ; 70(2): 233-43, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26148385

ABSTRACT

OBJECTIVE: Spinal cord stimulation (SCS) is an invasive method used for treating patients with refractory angina. Although SCS is recommended by the European Society of Cardiology and American Heart Association (class lib, level of evidence B and C, respectively), the method is used very rarely and data on its effectiveness is limited. The aim of this work was to perform a systematic review of all randomized controlled trials (RCTs) that investigated the effectiveness of SCS as a treatment for refractory angina. METHODS: Medline and Cochrane library databases were searched from their inception to April 2014 for RCTs that investigated the efficacy and safety of SCS in patients with refractory angina. RESULTS: Nine RCTs were included in the systematic review. The included RCTs were categorized into two groups: RCTs comparing SCS either with optimal medical treatment or inactive mode or low stimulation SCS; and those comparing SCS with alternative therapeutic interventions. Follow-up was short-term (1-6 months) in most studies, showing no major complications. Two studies reported a neutral effect regarding mortality. Regarding efficacy, most RCTs were in favour of SCS mainly in the short term. The most recent, multi-centre RCT reported no significant difference compared to the control group. CONCLUSIONS: RCTs investigating the efficacy of SCS were small and they demonstrated a small effect in angina improvement. Due to great differences in their design the interpretation of the results is complex. Before this method is recommended as a routine therapy for refractory angina, a larger, well-designed, multicentre RCT is needed.


Subject(s)
Angina Pectoris/therapy , Randomized Controlled Trials as Topic , Spinal Cord Stimulation/methods , Humans
14.
Haematologica ; 99(4): 638-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24463214

ABSTRACT

Cytokines are essential regulators of hematopoiesis, acting in an instructive or permissive way. Fms-like tyrosine kinase 3 ligand (FLT3L) is an important cytokine for the development of several hematopoietic populations. Its receptor (FLT3) is expressed on both myeloid and lymphoid progenitors and deletion of either the receptor or its ligand leads to defective developmental potential of hematopoietic progenitors. In vivo administration of FLT3L promotes expansion of progenitors with combined myeloid and lymphoid potential. To investigate further the role of this cytokine in hematopoietic development, we generated transgenic mice expressing high levels of human FLT3L. These transgenic mice displayed a dramatic expansion of dendritic and myeloid cells, leading to splenomegaly and blood leukocytosis. Bone marrow myeloid and lymphoid progenitors were significantly increased in numbers but retained their developmental potential. Furthermore, the transgenic mice developed anemia together with a reduction in platelet numbers. FLT3L was shown to rapidly reduce the earliest erythroid progenitors when injected into wild-type mice, indicating a direct negative role of the cytokine on erythropoiesis. We conclude that FLT3L acts on multipotent progenitors in an instructive way, inducing their development into myeloid/lymphoid lineages while suppressing their megakaryocyte/erythrocyte potential.


Subject(s)
Hematopoiesis/physiology , Membrane Proteins/genetics , Anemia/genetics , Anemia/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukocytosis/genetics , Leukocytosis/metabolism , Lymphatic Diseases/genetics , Lymphatic Diseases/pathology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Splenomegaly/genetics , Splenomegaly/pathology
15.
Proc Natl Acad Sci U S A ; 109(39): 15871-6, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-23019372

ABSTRACT

To better understand the process of B-lymphocyte lineage restriction, we have investigated molecular and functional properties in early B-lineage cells from Pax-5-deficient animals crossed to a B-lineage-restricted reporter mouse, allowing us to identify B-lineage-specified progenitors independently of conventional surface markers. Pax-5 deficiency resulted in a dramatic increase in the frequency of specified progenitor B-cells marked by expression of a λ5 (Igll1) promoter-controlled reporter gene. Gene expression analysis of ex vivo isolated progenitor cells revealed that Pax-5 deficiency has a minor impact on B-cell specification. However, single-cell in vitro differentiation analysis of ex vivo isolated cells revealed that specified B-lineage progenitors still displayed a high degree of plasticity for development into NK or T lineage cells. In contrast, we were unable to detect any major changes in myeloid lineage potential in specified Pax-5-deficient cells. By comparison of gene expression patterns in ex vivo isolated Pax-5- and Ebf-1-deficient progenitors, it was possible to identify a set of B-cell-restricted genes dependent on Ebf-1 but not Pax-5, supporting the idea that B-cell specification and commitment is controlled by distinct regulatory networks.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/physiology , Gene Expression Regulation/immunology , Animals , B-Lymphocytes/cytology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mice , Mice, Transgenic , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Trans-Activators/genetics , Trans-Activators/immunology
16.
Blood ; 118(5): 1283-90, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21652681

ABSTRACT

Deficiencies in the IL-7 signaling pathway result in severe disruptions of lymphoid development in adult mice. To understand more about how IL-7 deficiency impacts early lymphoid development, we have investigated lineage restriction events within the common lymphoid progenitor (CLP) compartment in IL-7 knockout mice. This revealed that although IL-7 deficiency had a minor impact on the development of LY6D(-) multipotent CLPs, the formation of the lineage restricted LY6D(+) CLP population was dramatically reduced. This was reflected in a low-level transcription of B-lineage genes as well as in a loss of functional B-cell commitment. The few Ly6D(+) CLPs developed in the absence of IL-7 displayed increased lineage plasticity and low expression of Ebf-1. Absence of Ebf-1 could be linked to increased plasticity because even though Ly6D(+) cells develop in Ebf-1-deficient mice, these cells retain both natural killer and dendritic cell potential. This reveals that IL-7 is essential for normal development of Ly6D(+) CLPs and that Ebf-1 is crucial for lineage restriction in early lymphoid progenitors.


Subject(s)
Cell Lineage/genetics , Interleukin-7/physiology , Lymphoid Progenitor Cells/physiology , Trans-Activators/physiology , Animals , Antigens, Ly/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Cell Differentiation/genetics , Cells, Cultured , GPI-Linked Proteins/metabolism , Gene Expression Profiling , Interleukin-7/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Trans-Activators/genetics , Trans-Activators/metabolism
17.
J Biol Chem ; 285(47): 36275-84, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20829349

ABSTRACT

The development of lymphoid cells from bone marrow progenitors is dictated by interplay between internal cues such as transcription factors and external signals like the cytokines Flt-3 ligand and Il-7. These proteins are both of large importance for normal lymphoid development; however, it is unclear if they act in direct synergy to expand a transient Il-7R(+)Flt-3(+) population or if the collaboration is created through sequential activities. We report here that Flt-3L and Il-7 synergistically stimulated the expansion of primary Il-7R(+)Flt-3(+) progenitor cells and a hematopoietic progenitor cell line ectopically expressing the receptors. The stimulation resulted in a reduced expression of pro-apoptotic genes and also mediated survival of primary progenitor cells in vitro. However, functional analysis of single cells suggested that the anti-apoptotic effect was additive indicating that the synergy observed mainly depends on stimulation of proliferation. Analysis of downstream signaling events suggested that although Il-7 induced Stat-5 phosphorylation, Flt-3L caused activation of the ERK and AKT signaling pathways. Flt-3L could also drive proliferation in synergy with ectopically expressed constitutively active Stat-5. This synergy could be inhibited with either receptor tyrosine kinase or MAPK inhibitors suggesting that Flt-3L and Il-7 act in synergy by activation of independent signaling pathways to expand early hematopoietic progenitors.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukin-7/pharmacology , STAT5 Transcription Factor/metabolism , Signal Transduction , fms-Like Tyrosine Kinase 3/metabolism , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Drug Synergism , Flow Cytometry , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , fms-Like Tyrosine Kinase 3/genetics
18.
Proc Natl Acad Sci U S A ; 107(37): 16280-5, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805474

ABSTRACT

Chronic myeloid leukemia (CML) is genetically characterized by the Philadelphia (Ph) chromosome, formed through a reciprocal translocation between chromosomes 9 and 22 and giving rise to the constitutively active tyrosine kinase P210 BCR/ABL1. Therapeutic strategies aiming for a cure of CML will require full eradication of Ph chromosome-positive (Ph(+)) CML stem cells. Here we used gene-expression profiling to identify IL-1 receptor accessory protein (IL1RAP) as up-regulated in CML CD34(+) cells and also in cord blood CD34(+) cells as a consequence of retroviral BCR/ABL1 expression. To test whether IL1RAP expression distinguishes normal (Ph(-)) and leukemic (Ph(+)) cells within the CML CD34(+)CD38(-) cell compartment, we established a unique protocol for conducting FISH on small numbers of sorted cells. By using this method, we sorted cells directly into drops on slides to investigate their Ph-chromosome status. Interestingly, we found that the CML CD34(+)CD38(-)IL1RAP(+) cells were Ph(+), whereas CML CD34(+)CD38(-)IL1RAP(-) cells were almost exclusively Ph(-). By performing long-term culture-initiating cell assays on the two cell populations, we found that Ph(+) and Ph(-) candidate CML stem cells could be prospectively separated. In addition, by generating an anti-IL1RAP antibody, we provide proof of concept that IL1RAP can be used as a target on CML CD34(+)CD38(-) cells to induce antibody-dependent cell-mediated cytotoxicity. This study thus identifies IL1RAP as a unique cell surface biomarker distinguishing Ph(+) from Ph(-) candidate CML stem cells and opens up a previously unexplored avenue for therapy of CML.


Subject(s)
Antibodies/immunology , Apoptosis , Interleukin-1 Receptor Accessory Protein/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , ADP-ribosyl Cyclase 1/immunology , Antigens, CD34/immunology , Cell Separation , Fusion Proteins, bcr-abl/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interleukin-1 Receptor Accessory Protein/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Glycoproteins/immunology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/immunology
19.
Blood ; 115(13): 2601-9, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19996414

ABSTRACT

To investigate molecular events involved in the regulation of lymphoid lineage commitment, we crossed lambda5 reporter transgenic mice to Rag1-GFP knockin mice. This allowed us to subfractionate common lymphoid progenitors and pre-pro-B (fraction A) cells into lambda5(-)Rag1(low), lambda5(-)Rag1(high), and lambda5(+)Rag1(high) cells. Clonal in vitro differentiation analysis demonstrated that Rag1(low) cells gave rise to B/T and NK cells. Rag1(high) cells displayed reduced NK-cell potential with preserved capacity to generate B- and T-lineage cells, whereas the lambda5(+) cells were B-lineage restricted. Ebf1 and Pax5 expression was largely confined to the Rag1(high) populations. These cells also expressed a higher level of the surface protein LY6D, providing an additional tool for the analysis of early lymphoid development. These data suggest that the classic common lymphoid progenitor compartment composes a mixture of cells with relatively restricted lineage potentials, thus opening new possibilities to investigate early hematopoiesis.


Subject(s)
Lymphocytes/cytology , Animals , Antigens, Ly/biosynthesis , Antigens, Ly/genetics , Biomarkers , Cell Lineage , Cells, Cultured/cytology , Cells, Cultured/metabolism , Coculture Techniques , Flow Cytometry , GPI-Linked Proteins , Gene Expression Profiling , Gene Knock-In Techniques , Genes, Reporter , Homeodomain Proteins/genetics , Killer Cells, Natural/cytology , Lymphocyte Subsets/cytology , Lymphocytes/metabolism , Lymphopoiesis , Mice , Mice, Transgenic , PAX5 Transcription Factor/biosynthesis , PAX5 Transcription Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Trans-Activators/biosynthesis , Trans-Activators/genetics
20.
J Immunol ; 181(5): 3364-72, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18714008

ABSTRACT

Development of B-lymphoid cells in the bone marrow is a process under strict control of a hierarchy of transcription factors. To understand the development of a B-lymphoid-restricted functional network of transcription factors, we have investigated the cell autonomous role of the transcription factor EBF1 in early B cell development. This revealed that even though transplanted EBF1-deficient fetal liver cells were able to generate common lymphoid progenitors (CLPs) as well as B220(+)CD43(+)AA4.1(+) candidate precursor B cells, none of these populations showed signs of B lineage priming. The isolated CLPs were able to generate T lymphocytes in vitro supporting the idea that the phenotype of EBF1-deficient mice is restricted to the development of the B lineage. Furthermore, EBF deficient CLPs displayed a reduction in Ig H chain recombination as compared with their wild-type counterpart and essentially lacked transcription of B-lineage-associated genes. Among the genes displaying reduced expression in the EBF1 deficient CLPs were the transcription factors Pax5, Pou2af1 (OcaB), and FoxO1 that all appear to be direct genetic targets for EBF1 because their promoters contained functional binding sites for this factor. This leads us to suggest that EBF1 regulates a transcription factor network crucial for B lineage commitment.


Subject(s)
B-Lymphocytes/cytology , Gene Regulatory Networks , Lymphoid Progenitor Cells/cytology , Trans-Activators/physiology , Animals , Cell Lineage , Down-Regulation , Mice , Precursor Cells, B-Lymphoid/cytology , T-Lymphocytes/cytology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...