Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Cell ; 135(6): 1118-29, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-19062086

ABSTRACT

Bone marrow hematopoietic stem cells (HSCs) are crucial to maintain lifelong production of all blood cells. Although HSCs divide infrequently, it is thought that the entire HSC pool turns over every few weeks, suggesting that HSCs regularly enter and exit cell cycle. Here, we combine flow cytometry with label-retaining assays (BrdU and histone H2B-GFP) to identify a population of dormant mouse HSCs (d-HSCs) within the lin(-)Sca1+cKit+CD150+CD48(-)CD34(-) population. Computational modeling suggests that d-HSCs divide about every 145 days, or five times per lifetime. d-HSCs harbor the vast majority of multilineage long-term self-renewal activity. While they form a silent reservoir of the most potent HSCs during homeostasis, they are efficiently activated to self-renew in response to bone marrow injury or G-CSF stimulation. After re-establishment of homeostasis, activated HSCs return to dormancy, suggesting that HSCs are not stochastically entering the cell cycle but reversibly switch from dormancy to self-renewal under conditions of hematopoietic stress.


Subject(s)
Adult Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Antigens, Differentiation/metabolism , Bone Marrow/physiology , Bromouracil/analogs & derivatives , Fluorouracil/metabolism , Green Fluorescent Proteins , Hematopoietic Stem Cells/physiology , Homeostasis , Mice , Mice, Transgenic , Uridine/analogs & derivatives , Uridine/metabolism
2.
Nat Immunol ; 11(8): 717-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20622884

ABSTRACT

ZFP36L1 and ZFP36L2 are RNA-binding proteins (RBPs) that interact with AU-rich elements in the 3' untranslated region of mRNA, which leads to mRNA degradation and translational repression. Here we show that mice that lacked ZFP36L1 and ZFP36L2 during thymopoiesis developed a T cell acute lymphoblastic leukemia (T-ALL) dependent on the oncogenic transcription factor Notch1. Before the onset of T-ALL, thymic development was perturbed, with accumulation of cells that had passed through the beta-selection checkpoint without first expressing the T cell antigen receptor beta-chain (TCRbeta). Notch1 expression was higher in untransformed thymocytes in the absence of ZFP36L1 and ZFP36L2. Both RBPs interacted with evolutionarily conserved AU-rich elements in the 3' untranslated region of Notch1 and suppressed its expression. Our data establish a role for ZFP36L1 and ZFP36L2 during thymocyte development and in the prevention of malignant transformation.


Subject(s)
Nuclear Proteins/deficiency , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Tristetraprolin/deficiency , Amino Acid Sequence , Animals , Butyrate Response Factor 1 , Conserved Sequence , Humans , Immunophenotyping , Kaplan-Meier Estimate , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sequence Alignment , Thymus Gland/growth & development , Transcription, Genetic , Tristetraprolin/genetics , Tristetraprolin/immunology
3.
EMBO J ; 34(15): 2008-24, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26136212

ABSTRACT

Myc controls the metabolic reprogramming that supports effector T cell differentiation. The expression of Myc is regulated by the T cell antigen receptor (TCR) and pro-inflammatory cytokines such as interleukin-2 (IL-2). We now show that the TCR is a digital switch for Myc mRNA and protein expression that allows the strength of the antigen stimulus to determine the frequency of T cells that express Myc. IL-2 signalling strength also directs Myc expression but in an analogue process that fine-tunes Myc quantity in individual cells via post-transcriptional control of Myc protein. Fine-tuning Myc matters and is possible as Myc protein has a very short half-life in T cells due to its constant phosphorylation by glycogen synthase kinase 3 (GSK3) and subsequent proteasomal degradation. We show that Myc only accumulates in T cells exhibiting high levels of amino acid uptake allowing T cells to match Myc expression to biosynthetic demands. The combination of digital and analogue processes allows tight control of Myc expression at the population and single cell level during immune responses.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , Interleukin-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Analysis of Variance , Animals , Blotting, Western , Cloning, Molecular , Flow Cytometry , Leupeptins , Mice , Mice, Transgenic , Mutagenesis , Proto-Oncogene Proteins c-myc/immunology , Pyridines , Pyrimidines , Real-Time Polymerase Chain Reaction
4.
Immunity ; 32(1): 14-27, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20152168

ABSTRACT

The Notch signaling pathway regulates many aspects of embryonic development, as well as differentiation processes and tissue homeostasis in multiple adult organ systems. Disregulation of Notch signaling is associated with several human disorders, including cancer. In the last decade, it became evident that Notch signaling plays important roles within the hematopoietic and immune systems. Notch plays an essential role in the development of embryonic hematopoietic stem cells and influences multiple lineage decisions of developing lymphoid and myeloid cells. Moreover, recent evidence suggests that Notch is an important modulator of T cell-mediated immune responses. In this review, we discuss Notch signaling in hematopoiesis, lymphocyte development, and function as well as in T cell acute lymphoblastic leukemia.


Subject(s)
Receptors, Notch/immunology , Signal Transduction/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Humans , Leukemia-Lymphoma, Adult T-Cell/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
5.
Immunity ; 33(5): 671-84, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21093323

ABSTRACT

Although canonical Notch signaling regulates multiple hematopoietic lineage decisions including T cell and marginal zone B cell fate specification, the downstream molecular mediators of Notch function are largely unknown. We showed here that conditional inactivation of Hes1, a well-characterized Notch target gene, in adult murine bone marrow (BM) cells severely impaired T cell development without affecting other Notch-dependent hematopoietic lineages such as marginal zone B cells. Competitive mixed BM chimeras, intrathymic transfer experiments, and in vitro culture of BM progenitors on Delta-like-expressing stromal cells further demonstrated that Hes1 is required for T cell lineage commitment, but dispensable for Notch-dependent thymocyte maturation through and beyond the beta selection checkpoint. Furthermore, our data strongly suggest that Hes1 is essential for the development and maintenance of Notch-induced T cell acute lymphoblastic leukemia. Collectively, our studies identify Hes1 as a critical but context-dependent mediator of canonical Notch signaling in the hematopoietic system.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Homeodomain Proteins/genetics , Lymphocyte Activation/genetics , Receptors, Notch/genetics , Animals , B-Lymphocytes/immunology , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Transcription Factor HES-1
6.
Blood ; 128(12): 1567-77, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27421959

ABSTRACT

The first definitive hematopoietic stem cells (dHSCs) in the mouse emerge in the dorsal aorta of the embryonic day (E) 10.5 to 11 aorta-gonad-mesonephros (AGM) region. Notch signaling is essential for early HSC development but is dispensable for the maintenance of adult bone marrow HSCs. How Notch signaling regulates HSC formation in the embryo is poorly understood. We demonstrate here that Notch signaling is active in E10.5 HSC precursors and involves both Notch1 and Notch2 receptors, but is gradually downregulated while they progress toward dHSCs at E11.5. This downregulation is accompanied by gradual functional loss of Notch dependency. Thus, as early as at final steps in the AGM region, HSCs begin acquiring the Notch independency characteristic of adult bone marrow HSCs as part of the maturation program. Our data indicate that fine stage-dependent tuning of Notch signaling may be required for the generation of definitive HSCs from pluripotent cells.


Subject(s)
Aorta/embryology , Embryo, Mammalian/cytology , Gonads/embryology , Hematopoietic Stem Cells/cytology , Mesonephros/embryology , Receptor, Notch2/metabolism , Stromal Cells/cytology , Animals , Aorta/metabolism , Cells, Cultured , Embryo, Mammalian/metabolism , Gonads/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Mesonephros/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Stromal Cells/metabolism
7.
J Immunol ; 197(3): 771-82, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27324132

ABSTRACT

Although Notch signaling plays important roles in lineage commitment and differentiation of multiple cell types including conventional T cells, nothing is currently known concerning Notch function in innate-like T cells. We have found that the homeostasis of several well-characterized populations of innate-like T cells including invariant NKT cells (iNKT), CD8ααTCRαß small intestinal intraepithelial lymphocytes, and innate memory phenotype CD8 T cells is controlled by Notch. Notch selectively regulates hepatic iNKT cell survival via tissue-restricted control of B cell lymphoma 2 and IL-7Rα expression. More generally, Notch regulation of innate-like T cell homeostasis involves both cell-intrinsic and -extrinsic mechanisms and relies upon context-dependent interactions with Notch ligand-expressing fibroblastic stromal cells. Collectively, using conditional ablation of Notch receptors on peripheral T cells or Notch ligands on putative fibroblastic stromal cells, we show that Notch signaling is indispensable for the homeostasis of three tissue-restricted populations of innate-like T cells: hepatic iNKT, CD8ααTCRαß small intestinal intraepithelial lymphocytes, and innate memory phenotype CD8 T cells, thus supporting a generalized role for Notch in innate T cell homeostasis.


Subject(s)
Cell Differentiation/immunology , Homeostasis/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Flow Cytometry , Immunohistochemistry , Mice , Mice, Transgenic , Receptors, Notch/metabolism
8.
PLoS Genet ; 11(3): e1005088, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25811463

ABSTRACT

MHC class II (MHCII) genes are transactivated by the NOD-like receptor (NLR) family member CIITA, which is recruited to SXY enhancers of MHCII promoters via a DNA-binding "enhanceosome" complex. NLRC5, another NLR protein, was recently found to control transcription of MHC class I (MHCI) genes. However, detailed understanding of NLRC5's target gene specificity and mechanism of action remained lacking. We performed ChIP-sequencing experiments to gain comprehensive information on NLRC5-regulated genes. In addition to classical MHCI genes, we exclusively identified novel targets encoding non-classical MHCI molecules having important functions in immunity and tolerance. ChIP-sequencing performed with Rfx5(-/-) cells, which lack the pivotal enhanceosome factor RFX5, demonstrated its strict requirement for NLRC5 recruitment. Accordingly, Rfx5-knockout mice phenocopy Nlrc5 deficiency with respect to defective MHCI expression. Analysis of B cell lines lacking RFX5, RFXAP, or RFXANK further corroborated the importance of the enhanceosome for MHCI expression. Although recruited by common DNA-binding factors, CIITA and NLRC5 exhibit non-redundant functions, shown here using double-deficient Nlrc5(-/-)CIIta(-/-) mice. These paradoxical findings were resolved by using a "de novo" motif-discovery approach showing that the SXY consensus sequence occupied by NLRC5 in vivo diverges significantly from that occupied by CIITA. These sequence differences were sufficient to determine preferential occupation and transactivation by NLRC5 or CIITA, respectively, and the S box was found to be the essential feature conferring NLRC5 specificity. These results broaden our knowledge on the transcriptional activities of NLRC5 and CIITA, revealing their dependence on shared enhanceosome factors but their recruitment to distinct enhancer motifs in vivo. Furthermore, we demonstrated selectivity of NLRC5 for genes encoding MHCI or related proteins, rendering it an attractive target for therapeutic intervention. NLRC5 and CIITA thus emerge as paradigms for a novel class of transcriptional regulators dedicated for transactivating extremely few, phylogenetically related genes.


Subject(s)
Genes, MHC Class II , Genes, MHC Class I , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Transcriptional Activation/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Enhancer Elements, Genetic , Gene Expression Regulation , Genome , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/immunology , Promoter Regions, Genetic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trans-Activators/biosynthesis , Trans-Activators/immunology , Transcriptional Activation/immunology
9.
Blood ; 121(6): 918-29, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23134786

ABSTRACT

Hematopoietic stem cells (HSCs) are the most primitive cells in the hematopoietic system and are under tight regulation for self-renewal and differentiation. Notch signals are essential for the emergence of definitive hematopoiesis in mouse embryos and are critical regulators of lymphoid lineage fate determination. However, it remains unclear how Notch regulates the balance between HSC self-renewal and differentiation in the adult bone marrow (BM). Here we report a novel mechanism that prevents HSCs from undergoing premature lymphoid differentiation in BM. Using a series of in vivo mouse models and functional HSC assays, we show that leukemia/lymphoma related factor (LRF) is necessary for HSC maintenance by functioning as an erythroid-specific repressor of Delta-like 4 (Dll4) expression. Lrf deletion in erythroblasts promoted up-regulation of Dll4 in erythroblasts, sensitizing HSCs to T-cell instructive signals in the BM. Our study reveals novel cross-talk between HSCs and erythroblasts, and sheds a new light on the regulatory mechanisms regulating the balance between HSC self-renewal and differentiation.


Subject(s)
DNA-Binding Proteins/genetics , Erythroblasts/metabolism , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Calcium-Binding Proteins , Cell Differentiation/genetics , Cell Proliferation , Cellular Microenvironment/genetics , DNA-Binding Proteins/metabolism , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , T-Lymphocytes/metabolism , Time Factors , Transcription Factors/metabolism , Transcriptome/genetics
10.
J Immunol ; 190(7): 3299-308, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23427253

ABSTRACT

Invariant NKT (iNKT) cells play key roles in host defense by recognizing lipid Ags presented by CD1d. iNKT cells are activated by bacterial-derived lipids and are also strongly autoreactive toward self-lipids. iNKT cell responsiveness must be regulated to maintain effective host defense while preventing uncontrolled stimulation and potential autoimmunity. CD1d-expressing thymocytes support iNKT cell development, but thymocyte-restricted expression of CD1d gives rise to Ag hyperresponsive iNKT cells. We hypothesized that iNKT cells require functional education by CD1d(+) cells other than thymocytes to set their correct responsiveness. In mice that expressed CD1d only on thymocytes, hyperresponsive iNKT cells in the periphery expressed significantly reduced levels of tyrosine phosphatase SHP-1, a negative regulator of TCR signaling. Accordingly, heterozygous SHP-1 mutant mice displaying reduced SHP-1 expression developed a comparable population of Ag hyperresponsive iNKT cells. Restoring nonthymocyte CD1d expression in transgenic mice normalized SHP-1 expression and iNKT cell reactivity. Radiation chimeras revealed that CD1d(+) dendritic cells supported iNKT cell upregulation of SHP-1 and decreased responsiveness after thymic emigration. Hence, dendritic cells functionally educate iNKT cells by tuning SHP-1 expression to limit reactivity.


Subject(s)
Dendritic Cells/metabolism , Natural Killer T-Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Animals , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Cluster Analysis , Dendritic Cells/immunology , Gene Expression Profiling , Gene Expression Regulation , Genomic Imprinting , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Thymus Gland/immunology , Thymus Gland/metabolism
11.
J Immunol ; 191(5): 2344-50, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918982

ABSTRACT

Follicular helper T (TFH) cells are specialized in providing help for B cell differentiation and Ab secretion. Several positive and negative regulators of TFH cell differentiation have been described but their control is not fully understood. In this study, we show that Notch signaling in T cells is a major player in the development and function of TFH cells. T cell-specific gene ablation of Notch1 and Notch2 impaired differentiation of TFH cells in draining lymph nodes of mice immunized with T-dependent Ags or infected with parasites. Impaired TFH cell differentiation correlated with deficient germinal center development and the absence of high-affinity Abs. The impact of loss of Notch on TFH cell differentiation was largely independent of its effect on IL-4. These results show a previously unknown role for Notch in the regulation of TFH cell differentiation and function with implications for the control of this T cell population.


Subject(s)
Cell Differentiation/immunology , Lymphocyte Activation/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Flow Cytometry , Gene Knockdown Techniques , Germinal Center/cytology , Germinal Center/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Helper-Inducer/cytology
12.
PLoS Pathog ; 8(3): e1002560, 2012.
Article in English | MEDLINE | ID: mdl-22396647

ABSTRACT

The protective immune response to intracellular parasites involves in most cases the differentiation of IFNγ-secreting CD4(+) T helper (Th) 1 cells. Notch receptors regulate cell differentiation during development but their implication in the polarization of peripheral CD4(+) T helper 1 cells is not well understood. Of the four Notch receptors, only Notch1 (N1) and Notch2 (N2) are expressed on activated CD4(+) T cells. To investigate the role of Notch in Th1 cell differentiation following parasite infection, mice with T cell-specific gene ablation of N1, N2 or both (N1N2(ΔCD4Cre)) were infected with the protozoan parasite Leishmania major. N1N2(ΔCD4Cre) mice, on the C57BL/6 L. major-resistant genetic background, developed unhealing lesions and uncontrolled parasitemia. Susceptibility correlated with impaired secretion of IFNγ by draining lymph node CD4(+) T cells and increased secretion of the IL-5 and IL-13 Th2 cytokines. Mice with single inactivation of N1 or N2 in their T cells were resistant to infection and developed a protective Th1 immune response, showing that CD4(+) T cell expression of N1 or N2 is redundant in driving Th1 differentiation. Furthermore, we show that Notch signaling is required for the secretion of IFNγ by Th1 cells. This effect is independent of CSL/RBP-Jκ, the major effector of Notch receptors, since L. major-infected mice with a RBP-Jκ deletion in their T cells were able to develop IFNγ-secreting Th1 cells, kill parasites and heal their lesions. Collectively, we demonstrate here a crucial role for RBP-Jκ-independent Notch signaling in the differentiation of a functional Th1 immune response following L. major infection.


Subject(s)
Interferon-gamma/metabolism , Leishmania major/physiology , Leishmaniasis, Cutaneous/immunology , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Th1 Cells/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Host-Parasite Interactions , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Signal Transduction
13.
J Immunol ; 188(8): 3820-8, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22412192

ABSTRACT

Nucleotide-binding oligomerization domain-like receptors (NLRs) are intracellular proteins involved in innate-driven inflammatory responses. The function of the family member NLR caspase recruitment domain containing protein 5 (NLRC5) remains a matter of debate, particularly with respect to NF-κB activation, type I IFN, and MHC I expression. To address the role of NLRC5, we generated Nlrc5-deficient mice (Nlrc5(Δ/Δ)). In this article we show that these animals exhibit slightly decreased CD8(+) T cell percentages, a phenotype compatible with deregulated MHC I expression. Of interest, NLRC5 ablation only mildly affected MHC I expression on APCs and, accordingly, Nlrc5(Δ/Δ) macrophages efficiently primed CD8(+) T cells. In contrast, NLRC5 deficiency dramatically impaired basal expression of MHC I in T, NKT, and NK lymphocytes. NLRC5 was sufficient to induce MHC I expression in a human lymphoid cell line, requiring both caspase recruitment and LRR domains. Moreover, endogenous NLRC5 localized to the nucleus and occupied the proximal promoter region of H-2 genes. Consistent with downregulated MHC I expression, the elimination of Nlrc5(Δ/Δ) lymphocytes by cytotoxic T cells was markedly reduced and, in addition, we observed low NLRC5 expression in several murine and human lymphoid-derived tumor cell lines. Hence, loss of NLRC5 expression represents an advantage for evading CD8(+) T cell-mediated elimination by downmodulation of MHC I levels-a mechanism that may be exploited by transformed cells. Our data show that NLRC5 acts as a key transcriptional regulator of MHC I in lymphocytes and support an essential role for NLRs in directing not only innate but also adaptive immune responses.


Subject(s)
Genes, MHC Class I , Intracellular Signaling Peptides and Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Bone Marrow/immunology , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/immunology , Cell Proliferation , Gene Expression Regulation , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , T-Lymphocytes, Cytotoxic/cytology
14.
Genesis ; 51(10): 717-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23832856

ABSTRACT

The thymus is the site of T cell development. Several stromal and hematopoietic cell types are necessary for the proper function of thymic selection and eventually peripheral immunity. Thymic epithelial cells (TECs) are essential for T cell lineage commitment, expansion, and maturation in the thymus. We were interested in developing an in vivo model in which exogenous gene expression could be transiently induced in embryonic TEC (Tet-On system). To this end, we have generated a bacterial artificial chromosome (BAC) transgenic mouse line in which the reverse tetracycline-dependent transactivator (rtTA) is expressed under the control of the Foxn1 promoter, a transcriptional factor indispensable for TEC development. To analyze the expression pattern and efficiency of this novel mouse model, we crossed the Foxn1-rtTA founder with a Tet-Responsive Element (TRE)-LacZ GFP mouse reporter to obtain a double transgenic mouse. In the presence of doxycycline, rtTA can interact with TRE and induce the expression of GFP and LacZ. In this double transgenic mouse, we observed that GFP expression was high, inducible and limited to TEC in fetal thymus. In contrast, in adult thymus, when TEC development and maturation is completed, GFP was barely detectable. Therefore, Foxn1-rtTA represents a new and efficient transgenic mouse model to induce genes of interest specifically in fetal thymic epithelium.


Subject(s)
Epithelial Cells/metabolism , Forkhead Transcription Factors/genetics , Gene Expression , Mice, Transgenic , Models, Animal , Thymus Gland/embryology , Animals , Cells, Cultured , Chromosomes, Artificial, Bacterial , Epithelium/embryology , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic , Thymus Gland/metabolism
15.
J Neurosci ; 32(16): 5654-66, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22514327

ABSTRACT

The adult mammalian forebrain contains neural stem/progenitor cells (NSCs) that generate neurons throughout life. As in other somatic stem cell systems, NSCs are proposed to be predominantly quiescent and proliferate only sporadically to produce more committed progeny. However, quiescence has recently been shown not to be an essential criterion for stem cells. It is not known whether NSCs show differences in molecular dependence based on their proliferation state. The subventricular zone (SVZ) of the adult mouse brain has a remarkable capacity for repair by activation of NSCs. The molecular interplay controlling adult NSCs during neurogenesis or regeneration is not clear but resolving these interactions is critical in order to understand brain homeostasis and repair. Using conditional genetics and fate mapping, we show that Notch signaling is essential for neurogenesis in the SVZ. By mosaic analysis, we uncovered a surprising difference in Notch dependence between active neurogenic and regenerative NSCs. While both active and regenerative NSCs depend upon canonical Notch signaling, Notch1-deletion results in a selective loss of active NSCs (aNSCs). In sharp contrast, quiescent NSCs (qNSCs) remain after Notch1 ablation until induced during regeneration or aging, whereupon they become Notch1-dependent and fail to fully reinstate neurogenesis. Our results suggest that Notch1 is a key component of the adult SVZ niche, promoting maintenance of aNSCs, and that this function is compensated in qNSCs. Therefore, we confirm the importance of Notch signaling for maintaining NSCs and neurogenesis in the adult SVZ and reveal that NSCs display a selective reliance on Notch1 that may be dictated by mitotic state.


Subject(s)
Adult Stem Cells/physiology , Lateral Ventricles/cytology , Neurogenesis/physiology , Receptor, Notch1/metabolism , Stem Cell Niche/physiology , Adult Stem Cells/drug effects , Animals , Carrier Proteins/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Differentiation/genetics , Cytarabine/pharmacology , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunosuppressive Agents/pharmacology , Intermediate Filament Proteins/genetics , Lateral Ventricles/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurogenesis/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Proteins/genetics , Proteins/metabolism , RNA, Untranslated , Receptor, Notch1/deficiency , Receptors, Estrogen/genetics , Stem Cell Niche/genetics , Tamoxifen/pharmacology , Time Factors
16.
J Exp Med ; 204(2): 331-43, 2007 Feb 19.
Article in English | MEDLINE | ID: mdl-17261636

ABSTRACT

Notch1 (N1) receptor signaling is essential and sufficient for T cell development, and recently developed in vitro culture systems point to members of the Delta family as being the physiological N1 ligands. We explored the ability of Delta1 (DL1) and DL4 to induce T cell lineage commitment and/or maturation in vitro and in vivo from bone marrow (BM) precursors conditionally gene targeted for N1 and/or N2. In vitro DL1 can trigger T cell lineage commitment via either N1 or N2. N1- or N2-mediated T cell lineage commitment can also occur in the spleen after short-term BM transplantation. However, N2-DL1-mediated signaling does not allow further T cell maturation beyond the CD25(+) stage due to a lack of T cell receptor beta expression. In contrast to DL1, DL4 induces and supports T cell commitment and maturation in vitro and in vivo exclusively via specific interaction with N1. Moreover, comparative binding studies show preferential interaction of DL4 with N1, whereas binding of DL1 to N1 is weak. Interestingly, preferential N1-DL4 binding reflects reduced dependence of this interaction on Lunatic fringe, a glycosyl transferase that generally enhances the avidity of Notch receptors for Delta ligands. Collectively, our results establish a hierarchy of Notch-Delta interactions in which N1-DL4 exhibits the greatest capacity to induce and support T cell development.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Hematopoietic Stem Cells/cytology , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction/immunology , T-Lymphocytes/cytology , Animals , DNA Primers , Flow Cytometry , Glycosyltransferases/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Protein Binding , Receptor, Notch2/metabolism , Retroviridae , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells , Transfection
17.
J Immunol ; 187(1): 110-7, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21632721

ABSTRACT

Activating and inhibitory NK receptors regulate the development and effector functions of NK cells via their ITAM and ITIM motifs, which recruit protein tyrosine kinases and phosphatases, respectively. In the T cell lineage, inhibitory Ly49 receptors are expressed by a subset of activated T cells and by CD1d-restricted NKT cells, but virtually no expression of activating Ly49 receptors is observed. Using mice transgenic for the activating receptor Ly49D and its associated ITAM signaling DAP12 chain, we show in this article that Ly49D-mediated ITAM signaling in immature thymocytes impairs development due to a block in maturation from the double negative (DN) to double positive (DP) stages. A large proportion of Ly49D/DAP12 transgenic thymocytes were able to bypass the pre-TCR checkpoint at the DN3 stage, leading to the appearance of unusual populations of DN4 and DP cells that lacked expression of intracellular (ic) TCRß-chain. High levels of CD5 were expressed on ic TCRß(-) DN and DP thymocytes from Ly49D/DAP12 transgenic mice, further suggesting that Ly49D-mediated ITAM signaling mimics physiological ITAM signaling via the pre-TCR. We also observed unusual ic TCRß(-) single positive thymocytes with an immature CD24(high) phenotype that were not found in the periphery. Importantly, thymocyte development was completely rescued by expression of an Ly49A transgene in Ly49D/DAP12 transgenic mice, indicating that Ly49A-mediated ITIM signaling can fully counteract ITAM signaling via Ly49D/DAP12. Collectively, our data indicate that inappropriate ITAM signaling by activating NK receptors on immature thymocytes can subvert T cell development by bypassing the pre-TCR checkpoint.


Subject(s)
Cell Differentiation/immunology , Lymphocyte Activation/immunology , NK Cell Lectin-Like Receptor Subfamily A/physiology , Receptors, Antigen, T-Cell/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Tyrosine/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle/genetics , Cell Cycle/immunology , Cell Differentiation/genetics , H-2 Antigens/genetics , Histocompatibility Antigen H-2D , Humans , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily A/genetics , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/biosynthesis , Signal Transduction/genetics , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
18.
Blood ; 115(22): 4431-8, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20308599

ABSTRACT

The murine gut epithelium contains a large population of thymus-derived intraepithelial lymphocytes (IELs), including both conventional CD4(+) and CD8alphabeta(+) T cells (expressing T-cell receptor alphabeta [TCRalphabeta]) and unconventional CD8alphaalpha(+) T cells (expressing either TCRalphabeta or TCRgammadelta). Whereas conventional IELs are widely accepted to arise from recirculation of activated CD4(+) and CD8alphabeta(+) T cells from the secondary lymphoid organs to the gut, the origin and developmental pathway of unconventional CD8alphaalpha IELs remain controversial. We show here that CD4-Cre-mediated inactivation of c-Myc, a broadly expressed transcription factor with a wide range of biologic activities, selectively impairs the development of CD8alphaalpha TCRalphabeta IELs. In the absence of c-Myc, CD4(-) CD8(-) TCRalphabeta(+) thymic precursors of CD8alphaalpha TCRalphabeta IELs are present but fail to develop on adoptive transfer in immunoincompetent hosts. Residual c-Myc-deficient CD8alphaalpha TCRalphabeta IEL display reduced proliferation and increased apoptosis, which correlate with significantly decreased expression of interleukin-15 receptor subunits and lower levels of the antiapoptotic protein Bcl-2. Transgenic overexpression of human BCL-2 resulted in a pronounced rescue of CD8alphaalpha TCRalphabeta IEL in c-Myc-deficient mice. Taken together, our data support a model in which c-Myc controls the development of CD8alphaalpha TCRalphabeta IELs from thymic precursors by regulating interleukin-15 receptor expression and consequently Bcl-2-dependent survival.


Subject(s)
CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Interleukin-15/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Adoptive Transfer , Animals , Apoptosis , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptors, Interleukin-15/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
19.
Int Immunol ; 23(2): 75-88, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21266499

ABSTRACT

Fas-deficient mice (Fas(lpr/lpr)) and humans have profoundly dysregulated T lymphocyte homeostasis, which manifests as an accumulation of CD4(+) and CD8(+) T cells as well as an unusual population of CD4(-)CD8(-)TCRαß(+) T cells. To date, no unifying model has explained both the increased T-cell numbers and the origin of the CD4(-)CD8(-)TCRαß(+) T cells. As Fas(lpr/lpr) mice raised in a germ-free environment still manifest lymphadenopathy, we considered that this process is primarily driven by recurrent low-avidity TCR signaling in response to self-peptide/MHC as occurs during homeostatic proliferation. In these studies, we developed two independent systems to decrease the number of self-peptide/MHC contacts. First, expression of MHC class I was reduced in OT-I TCR transgenic mice. Although OT-I Fas(lpr/lpr) mice did not develop lymphadenopathy characteristic of Fas(lpr/lpr) mice, in the absence of MHC class I, OT-I Fas(lpr/lpr) T cells accumulated as both CD8(+) and CD4(-)CD8(-) T cells. In the second system, re-expression of ß(2)m limited to thymic cortical epithelial cells of Fas(lpr/lpr) ß(2)m-deficient mice yielded a model in which polyclonal CD8(+) thymocytes entered a peripheral environment devoid of MHC class I. These mice accumulated significantly greater numbers of CD4(-)CD8(-)TCRαß(+) T cells than conventional Fas(lpr/lpr) mice. Thus, Fas shapes the peripheral T-cell repertoire by regulating the survival of a subset of T cells proliferating in response to limited self-peptide/MHC contacts.


Subject(s)
Histocompatibility Antigens Class I/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , fas Receptor/immunology , Animals , CD5 Antigens/immunology , Cell Proliferation , Female , Lymphocyte Activation , Lymphopenia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/immunology
20.
Arterioscler Thromb Vasc Biol ; 31(6): 1377-86, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441138

ABSTRACT

OBJECTIVE: Inflammation and proteolysis crucially contribute to myocardial ischemia and reperfusion injury. The extracellular matrix metalloproteinase inducer EMMPRIN (CD147) and its ligand cyclophilin A (CyPA) may be involved in both processes. The aim of the study was to characterize the role of the CD147 and CyPA interplay in myocardial ischemia/reperfusion (I/R) injury. METHODS AND RESULTS: Immunohistochemistry showed enhanced expression of CD147 and CyPA in myocardial sections from human autopsies of patients who had died from acute myocardial infarction and from mice at 24 hours after I/R. At 24 hours and 7 days after I/R, the infarct size was reduced in CD147(+/-) mice vs CD147(+/+) mice (C57Bl/6), in mice (C57Bl/6) treated with monoclonal antibody anti-CD147 vs control monoclonal antibody, and in CyPA(-/-) mice vs CyPA(+/+) mice (129S6/SvEv), all of which are associated with reduced monocyte and neutrophil recruitment at 24 hours and with a preserved systolic function at 7 days. The combination of CyPA(-/-) mice with anti-CD147 treatment did not yield further protection compared with either inhibition strategy alone. In vitro, treatment with CyPA induced monocyte chemotaxis in a CD147- and phosphatidylinositol 3-kinase-dependent manner and induced monocyte rolling and adhesion to endothelium (human umbilical vein endothelial cells) under flow in a CD147-dependent manner. CONCLUSION: CD147 and its ligand CyPA are inflammatory mediators after myocardial ischemia and reperfusion and represent potential targets to prevent myocardial I/R injury.


Subject(s)
Basigin/physiology , Cyclophilin A/physiology , Myocardial Infarction/metabolism , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/etiology , Systole , Animals , Basigin/analysis , Cell Adhesion , Cell Movement , Cyclophilin A/analysis , Humans , Macrophages/physiology , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/prevention & control , Neutrophils/physiology
SELECTION OF CITATIONS
SEARCH DETAIL