Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters










Publication year range
1.
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
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.
Front Immunol ; 9: 1312, 2018.
Article in English | MEDLINE | ID: mdl-29942310

ABSTRACT

For successful bone marrow transplantation (BMT), a preconditioning regime involving chemo and radiotherapy is used that results in DNA damage to both hematopoietic and stromal elements. Following radiation exposure, it is well recognized that a single wave of host-derived thymocytes reconstitutes the irradiated thymus, with donor-derived thymocytes appearing about 7 days post BMT. Our previous studies have demonstrated that, in the presence of donor hematopoietic cells lacking T lineage potential, these host-derived thymocytes are able to generate a polyclonal cohort of functionally mature peripheral T cells numerically comprising ~25% of the peripheral T cell pool of euthymic mice. Importantly, we demonstrated that radioresistant CD44+ CD25+ CD117+ DN2 progenitors were responsible for this thymic auto-reconstitution. Until recently, the mechanisms underlying the radioresistance of DN2 progenitors were unknown. Herein, we have used the in vitro "Plastic Thymus" culture system to perform a detailed investigation of the mechanisms responsible for the high radioresistance of DN2 cells compared with radiosensitive hematopoietic stem cells. Our results indicate that several aspects of DN2 biology, such as (i) rapid DNA damage response (DDR) activation in response to ionizing radiation-induced DNA damage, (ii) efficient repair of DNA double-strand breaks, and (iii) induction of a protective G1/S checkpoint contribute to promoting DN2 cell survival post-irradiation. We have previously shown that hypoxia increases the radioresistance of bone marrow stromal cells in vitro, at least in part by enhancing their DNA double-strand break (DNA DSB) repair capacity. Since the thymus is also a hypoxic environment, we investigated the potential effects of hypoxia on the DDR of DN2 thymocytes. Finally, we demonstrate for the first time that de novo DN2 thymocytes are able to rapidly repair DNA DSBs following thymic irradiation in vivo.

5.
Nat Commun ; 9(1): 2067, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802242

ABSTRACT

Testosterone deficiency in men is associated with increased risk for autoimmunity and increased B cell numbers through unknown mechanisms. Here we show that testosterone regulates the cytokine BAFF, an essential survival factor for B cells. Male mice lacking the androgen receptor have increased splenic B cell numbers, serum BAFF levels and splenic Baff mRNA. Testosterone deficiency by castration causes expansion of BAFF-producing fibroblastic reticular cells (FRCs) in spleen, which may be coupled to lower splenic noradrenaline levels in castrated males, as an α-adrenergic agonist decreases splenic FRC number in vitro. Antibody-mediated blockade of the BAFF receptor or treatment with the neurotoxin 6-hydroxydopamine revert the increased splenic B cell numbers induced by castration. Among healthy men, serum BAFF levels are higher in men with low testosterone. Our study uncovers a previously unrecognized regulation of BAFF by testosterone and raises important questions about BAFF in testosterone-mediated protection against autoimmunity.


Subject(s)
Autoimmune Diseases/metabolism , B-Cell Activating Factor/immunology , B-Cell Activating Factor/metabolism , B-Lymphocytes/immunology , Testosterone/metabolism , Adrenergic alpha-Agonists/pharmacology , Animals , Autoimmune Diseases/immunology , B-Cell Activating Factor/blood , B-Cell Activation Factor Receptor/antagonists & inhibitors , B-Cell Activation Factor Receptor/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Castration , Humans , Male , Mice , Mice, Knockout , Models, Animal , Norepinephrine/metabolism , Oxidopamine/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Testosterone/blood , Testosterone/deficiency , Testosterone/immunology
6.
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
7.
Front Immunol ; 9: 656, 2018.
Article in English | MEDLINE | ID: mdl-29670626

ABSTRACT

Murine chronic graft-versus-host-disease (cGvHD) induced by injection of parental lymphocytes into F1 hybrids results in a disease similar to systemic lupus erythematosus. Here, we have used DBA/2 T cell injection into (C57BL/6 × DBA/2)F1 (BDF1) mice as a model system to test the prophylactic and therapeutic effects of interleukin-2 (IL-2)/anti-IL-2 immune complexes on the course of cGvHD. Our findings demonstrate that pretreatment with Treg inducing JES6/IL-2 complexes render BDF1 mice largely resistant to induction of cGvHD, whereas pretreatment with CD8+ T cell/NK cell inducing S4B6/IL-2 complexes results in a more severe cGvHD. In contrast, treatment with JES6/IL-2 complexes 4 weeks after induction had no beneficial effect on disease symptoms. However, similar treatment with S4B6/IL-2 complexes led to a significant amelioration of the disease. This therapeutic effect seems to be mediated by donor CD8+ T cells. The fact that a much stronger cGvHD is induced in BDF1 mice depleted of donor CD8+ T cells strongly supports this conclusion. The contrasting effects of the two different IL-2 complexes are likely due to different mechanisms.


Subject(s)
Antigen-Antibody Complex/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/therapy , Immunotherapy/methods , Interleukin-2/metabolism , Lupus Erythematosus, Systemic/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/metabolism , Cells, Cultured , Disease Models, Animal , Graft vs Host Disease/immunology , Humans , Lupus Erythematosus, Systemic/immunology , Lymphocyte Transfusion , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Transplantation, Homologous
8.
Nat Commun ; 9(1): 1199, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572442

ABSTRACT

The B cell survival factor (TNFSF13B/BAFF) is often elevated in autoimmune diseases and is targeted in the clinic for the treatment of systemic lupus erythematosus. BAFF contains a loop region designated the flap, which is dispensable for receptor binding. Here we show that the flap of BAFF has two functions. In addition to facilitating the formation of a highly active BAFF 60-mer as shown previously, it also converts binding of BAFF to TNFRSF13C (BAFFR) into a signaling event via oligomerization of individual BAFF-BAFFR complexes. Binding and activation of BAFFR can therefore be targeted independently to inhibit or activate the function of BAFF. Moreover, structural analyses suggest that the flap of BAFF 60-mer temporarily prevents binding of an anti-BAFF antibody (belimumab) but not of a decoy receptor (atacicept). The observed differences in profiles of BAFF inhibition may confer distinct biological and clinical efficacies to these therapeutically relevant inhibitors.


Subject(s)
B-Cell Activating Factor/chemistry , B-Cell Activating Factor/physiology , B-Cell Activation Factor Receptor/chemistry , B-Lymphocytes/cytology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , B-Cell Activating Factor/genetics , Cell Differentiation , Cell Survival , Cross-Linking Reagents/chemistry , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Immunoglobulin Fragments/chemistry , Lymphopenia/metabolism , Male , Mice , Mice, Transgenic , Mutation , Protein Binding , Protein Domains , Recombinant Fusion Proteins/pharmacology
9.
Front Immunol ; 9: 16, 2018.
Article in English | MEDLINE | ID: mdl-29403498

ABSTRACT

The escape of anti-self B cells from tolerance mechanisms like clonal deletion, receptor editing, and anergy results in the production of autoantibodies, which is a hallmark of many autoimmune disorders. In this study, we demonstrate that both germline sequences and somatic mutations contribute to autospecificity of B cell clones. For this issue, we investigated the development of antinuclear autoantibodies (ANAs) and their repertoire in two different mouse models. First, in aging mice that were shown to gain several autoimmune features over time including ANAs. Second, in mice undergoing a chronic graft-versus-host disease (GVHD), thereby developing systemic lupus erythematosus-like symptoms. Detailed repertoire analysis revealed that somatic hypermutations (SHM) were present in all Vh and practically all Vl regions of ANAs generated in these two models. The ANA B cell repertoire in aging mice was restricted, dominated by clonally related Vh1-26/Vk4-74 antibodies. In the collection of GVHD-derived ANAs, the repertoire was less restricted, but the usage of the Vh1-26/Vk4-74 combination was still apparent. Germline conversion showed that the SHM in the 4-74 light chain are deterministic for autoreactivity. Detailed analysis revealed that antinuclear reactivity of these antibodies could be induced by a single amino acid substitution in the CDR1 of the Vk4-74. In both aging B6 and young GVHD mice, conversion of the somatic mutations in the Vh and Vl regions of non Vh1-26/Vk4-74 using antibodies showed that B cells with a germline-encoded V gene could also contribute to the ANA-reactive B cell repertoire. These findings indicate that two distinct pathways generate ANA-producing B cells in both model systems. In one pathway, they are generated by Vh1-26/Vk4-74 expressing B cells in the course of immune responses to an antigen that is neither a nuclear antigen nor any other self-antigen. In the other pathway, ANA-producing B cells are derived from progenitors in the bone marrow that express B cell receptors (BCRs), which bind to nuclear antigens and that escape tolerance induction, possibly as a result of crosslinking of their BCRs by multivalent determinants of nuclear antigens.


Subject(s)
Aging/immunology , Antibodies, Antinuclear/immunology , Antigens, Nuclear/immunology , Autoimmunity/genetics , B-Lymphocytes/immunology , Graft vs Host Disease/immunology , Amino Acid Substitution , Animals , Antibodies, Monoclonal/immunology , Autoimmunity/immunology , Histones/immunology , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Receptors, Antigen, B-Cell/immunology
12.
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
13.
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
14.
Cell Rep ; 18(9): 2189-2202, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28249164

ABSTRACT

B cell activating factor (BAFF) provides B cells with essential survival signals. It binds to three receptors: BAFFR, TACI, and BCMA that are differentially expressed by B cell subsets. BAFFR is early expressed in circulating B cells and provides key signals for further maturation. Here, we report that highly regulated BAFFR processing events modulate BAFF responses. BAFFR processing is triggered by BAFF binding in B cells co-expressing TACI and it is executed by the metalloproteases ADAM10 and ADAM17. The degree of BAFF oligomerization, the expression of ADAM proteins in different B cell subsets, and the activation status of the cell determine the proteases involved in BAFFR processing. Inhibition of ADAM10 augments BAFF-dependent survival of primary human B cells, whereas inhibition of ADAM17 increases BAFFR expression levels on germinal center B cells. Therefore, BAFF-induced processing of BAFFR regulates BAFF-mediated B cell responses in a TACI-dependent manner.


Subject(s)
ADAM Proteins/metabolism , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/metabolism , B-Lymphocyte Subsets/metabolism , Cell Survival/physiology , Peptide Hydrolases/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , Cell Line , Humans , Protein Binding/physiology
15.
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
16.
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
17.
J Immunol ; 196(6): 2561-71, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26851220

ABSTRACT

Flt3 ligand (Flt3L) promotes survival of lymphoid progenitors in the bone marrow and differentiation of dendritic cells (DCs), but its role in regulating innate lymphoid cells (ILCs) during fetal and adult life is not understood. By using Flt3L knockout and transgenic mice, we demonstrate that Flt3L controls ILC numbers by regulating the pool of α4ß7(-) and α4ß7(+) lymphoid tissue inducer cell progenitors in the fetal liver and common lymphoid progenitors in the bone marrow. Deletion of flt3l severely reduced the number of fetal liver progenitors and lymphoid tissue inducer cells in the neonatal intestine, resulting in impaired development of Peyer's patches. In the adult intestine, NK cells and group 2 and 3 ILCs were severely reduced. This effect occurred independently of DCs as ILC numbers were normal in mice in which DCs were constitutively deleted. Finally, we could show that administration of Flt3L increased the number of NKp46(-) group 3 ILCs in wild-type and even in Il7(-/-) mice, which generally have reduced numbers of ILCs. Taken together, Flt3L significantly contributes to ILC and Peyer's patches development by targeting lymphoid progenitor cells during fetal and adult life.


Subject(s)
Immunity, Innate/immunology , Lymphoid Progenitor Cells/immunology , Lymphopoiesis/immunology , Membrane Proteins/immunology , Peyer's Patches/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Separation , Fetus , Flow Cytometry , Immunohistochemistry , Lymphoid Progenitor Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peyer's Patches/cytology
18.
Nat Commun ; 6: 8324, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26477271

ABSTRACT

A widely accepted model posits that activation of enhancers during differentiation goes through a priming step prior to lineage commitment. To investigate the chronology of enhancer repertoire establishment during hematopoiesis, we monitored epigenome dynamics during three developmental stages representing hematopoietic stem cells, B-cell progenitors and mature B-cells. We find that only a minority of enhancers primed in stem cells or progenitors become active at later stages. Furthermore, most enhancers active in differentiated cells were not primed in earlier stages. Thus, the enhancer repertoire is reshaped dynamically during B-cell differentiation and enhancer priming in early stages does not appear to be an obligate step for enhancer activation. Furthermore, our data reveal that heterochromatin and Polycomb-mediated silencing have only a minor contribution in shaping enhancer repertoires during cell differentiation. Together, our data revisit the prevalent model about epigenetic reprogramming during hematopoiesis and give insights into the formation of gene regulatory networks.


Subject(s)
B-Lymphocytes/metabolism , Enhancer Elements, Genetic , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Animals , Chromatin Immunoprecipitation , Epigenomics , Gene Silencing , Heterochromatin/metabolism , Mice, Inbred C57BL , Polycomb-Group Proteins/metabolism
19.
Immunity ; 43(4): 674-89, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26453378

ABSTRACT

Development of dendritic cells (DCs) commences in the bone marrow, from where pre-DCs migrate to peripheral organs to differentiate into mature DCs in situ. However, the factors that regulate organ-specific differentiation to give rise to tissue-specific DC subsets remain unclear. Here we show that the Ras-PI3Kγ-Akt-mTOR signaling axis acted downstream of FLT3L signaling and was required for development of lung CD103(+) DCs and, to a smaller extent, for lung CD11b(+) DCs, but not related DC populations in other non-lymphoid organs. Furthermore, we show that in lymphoid organs such as the spleen, DCs depended on a similar signaling network to respond to FLT3 ligand with overlapping and partially redundant roles for kinases PI3Kγ and PI3Kδ. Thus we identified PI3Kγ as an essential organ-specific regulator of lung DC development and discovered a signaling network regulating tissue-specific DC development mediated by FLT3.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/physiology , Dendritic Cells/cytology , Lung/immunology , Signal Transduction/physiology , fms-Like Tyrosine Kinase 3/physiology , Animals , Apoptosis , Cell Differentiation/physiology , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Dendritic Cells/classification , Heterocyclic Compounds, 3-Ring/pharmacology , Homeostasis/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lung/cytology , Lung/enzymology , Lymphoid Tissue/cytology , Lymphoid Tissue/enzymology , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/physiology , Organ Specificity , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Radiation Chimera , Recombinant Proteins/pharmacology , TOR Serine-Threonine Kinases/physiology
20.
Eur Neuropsychopharmacol ; 25(11): 2049-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26372541

ABSTRACT

Trace amine-associated receptor 1 (TAAR1) activation by selective endogenous agonists modulates dopaminergic neurotransmission. This results in antipsychotic-like behavior in vivo which might be initiated by an interaction of TAAR1 and dopamine D2L receptor (D2R). Here we analyzed the functional link between TAAR1 and D2R using highly potent and selective TAAR1 agonists, and newly generated tools such as TAAR1 knock-out and TAAR1 overexpressing rats as well as specific anti-rat TAAR1 antibodies. We provide data from co-immunoprecipitation experiments supporting a functional interaction of the two receptors in heterologous cells and in brain tissue. Interaction of TAAR1 with D2R altered the subcellular localization of TAAR1 and increased D2R agonist binding affinity. Using specific ß-arrestin 2 (ßArr2) complementation assays we show that the interaction of TAAR1 with D2R reduced ßArr2 recruitment to D2R. In addition, we report that besides Gαs-protein signaling TAAR1 also signals via ßArr2. In the presence of D2R, cAMP signaling of TAAR1 was reduced while its ßArr2 signaling was enhanced, resulting in reduced GSK3ß activation. These results demonstrate that ßArr2 signaling may be an important pathway for TAAR1 function and that the activation of the TAAR1-D2R complex negatively modulates GSK3ß signaling. Given that patients with schizophrenia or bipolar disorder show increased GSK3ß signaling, such a reduction of GSK3ß signaling triggered by the interaction of D2R with activated TAAR1 further supports TAAR1 as a target for the treatment of psychiatric disorders.


Subject(s)
Arrestins/metabolism , Brain/metabolism , Glycogen Synthase Kinase 3/metabolism , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetulus , Cyclic AMP/metabolism , Gene Knockout Techniques , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Rats, Transgenic , Rats, Wistar , Receptors, Dopamine D2/agonists , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , beta-Arrestin 2 , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...