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










Publication year range
1.
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
2.
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
3.
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
4.
Immunol Lett ; 198: 60-65, 2018 06.
Article in English | MEDLINE | ID: mdl-29709545

ABSTRACT

Dipeptidyl peptidase 9 (DPP9) is a ubiquitously expressed intracellular prolyl peptidase implicated in immunoregulation. However, its physiological relevance in the immune system remains largely unknown. We investigated the role of DPP9 enzyme in immune system by characterizing DPP9 knock-in mice expressing a catalytically inactive S729A mutant of DPP9 enzyme (DPP9ki/ki mice). DPP9ki/ki mice show reduced number of lymphoid and myeloid cells in fetal liver and postnatal blood but their hematopoietic cells are fully functional and able to reconstitute lymphoid and myeloid lineages even in competitive mixed chimeras. These studies demonstrate that inactivation of DPP9 enzymatic activity does not lead to any perturbations in mouse hematopoiesis.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Hematopoiesis/immunology , Hematopoietic Stem Cells/physiology , Cell Count , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Gene Knock-In Techniques , Hematopoietic Stem Cells/cytology , Lymphocytes/cytology , Myeloid Cells/cytology
5.
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
6.
Front Immunol ; 8: 418, 2017.
Article in English | MEDLINE | ID: mdl-28450862

ABSTRACT

Thymic epithelial cells (TECs) are the main components of the thymic stroma that support and control T-cell development. Preparative regimens using DNA-damaging agents, such as total body irradiation and/or chemotherapeutic drugs, that are necessary prior to bone marrow transplantation (BMT) have profound deleterious effects on the hematopoietic system, including the thymic stroma, which may be one of the main causes for the prolonged periods of T-cell deficiency and the inefficient T cell reconstitution that are common following BMT. The DNA damage response (DDR) is a complex signaling network that allows cells to respond to all sorts of genotoxic insults. Hypoxia is known to modulate the DDR and play a role affecting the survival capacity of different cell types. In this study, we have characterized in detail the DDR of cortical and medullary TEC lines and their response to ionizing radiation, as well as the effects of hypoxia on their DDR. Although both mTECs and cTECs display relatively high radio-resistance, mTEC cells have an increased survival capacity to ionizing radiation (IR)-induced DNA damage, and hypoxia specifically decreases the radio-resistance of mTECs by upregulating the expression of the pro-apoptotic factor Bim. Analysis of the expression of TEC functional factors by primary mouse TECs showed a marked decrease of highly important genes for TEC function and confirmed cTECs as the most affected cell type by IR. These findings have important implications for improving the outcomes of BMT and promoting successful T cell reconstitution.

7.
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
8.
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
9.
Eur J Immunol ; 45(3): 932-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25408420

ABSTRACT

T-cell lymphopenia following BM transplantation or diseases such as AIDS result in immunodeficiency. Novel approaches to ameliorate this situation are urgently required. Herein, we describe a novel stromal cell free culture system in which Lineage(-) Sca1(+)c-kit(+) BM hematopoietic progenitors very efficiently differentiate into pro-T cells. This culture system consists of plate-bound Delta-like 4 Notch ligand and the cytokines SCF and IL-7. The pro-T cells developing in these cultures express CD25, CD117, and partially CD44; express cytoplasmic CD3ε; and have their TCRß locus partially D-J rearranged. They could be expanded for over 3 months and used to reconstitute the T-cell compartments of sublethally irradiated T-cell-deficient CD3ε(-/-) mice or lethally irradiated WT mice. Pro-T cells generated in this system could partially correct the T-cell lymphopenia of pre-Tα(-/-) mice. However, reconstituted CD3ε(-/-) mice suffered from a wasting disease that was prevented by co-injection of purified CD4(+) CD25(high) WT Treg cells. In a T-cell-sufficient or T-lymphopenic setting, the development of disease was not observed. Thus, this in vitro culture system represents a powerful tool to generate large numbers of pro-T cells for transplantation and possibly with clinical applications.


Subject(s)
Cell Culture Techniques/methods , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Precursor Cells, T-Lymphoid/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/immunology , Adaptor Proteins, Signal Transducing , Animals , Antigens, CD/genetics , Antigens, CD/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Calcium-Binding Proteins , Cells, Cultured , Female , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Precursor Cells, T-Lymphoid/cytology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Stromal Cells , T-Lymphocytes, Regulatory/cytology
10.
Immunol Lett ; 160(2): 113-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24472602

ABSTRACT

CD19 plays a crucial role in mature B cell development as best exemplified by the finding that CD19 deficient mice have severely reduced mature B cell compartments (Engel et al., 1995; Rickert et al., 1995). In the present study we show that the transition into the mature B cell compartments is heavily dependent on the correct amount of CD19 expression. Thus, Nup-98-HoxB4 immortalized hematopoietic stem cells (HSCs) over-expressing CD19 show upon transplantation an impaired pro/pre B to immature B cell transition in the bone marrow, whereas Nup-98-HoxB4 HSCs expressing a shRNA that down-modulates CD19 expression show upon transplantation a strongly reduced mature B cell compartment. Overall our findings indicate that too high CD19 expression might result into too strong BCR signaling in the bone marrow and therefore causing negative selection. Too low CD19 expression might result into too little BCR signaling and thereby preventing the B cells to enter the mature pool (absence of positive selection).


Subject(s)
Antigens, CD19/genetics , Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/metabolism , Pre-B Cell Receptors/genetics , Precursor Cells, B-Lymphoid/metabolism , Animals , Antigens, CD19/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation , Female , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Immunophenotyping , Male , Mice , Mice, Knockout , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/immunology , Pre-B Cell Receptors/immunology , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...