Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Immunity ; 56(5): 893-894, 2023 May 09.
Article in English | MEDLINE | ID: mdl-37163986
2.
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
3.
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
4.
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
5.
Eur J Immunol ; 43(11): 2845-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23881845

ABSTRACT

T-cell development depends upon interactions between thymocytes and thymic epithelial cells (TECs). The engagement of delta-like 4 (DL4) on TECs by Notch1 expressed by blood-borne BM-derived precursors is essential for T-cell commitment in the adult thymus. In contrast to the adult, the earliest T-cell progenitors in the embryo originate in the fetal liver and migrate to the nonvascularized fetal thymus via chemokine signals. Within the fetal thymus, some T-cell precursors undergo programmed TCRγ and TCRδ rearrangement and selection, giving rise to unique γδ T cells. Despite these fundamental differences between fetal and adult T-cell lymphopoiesis, we show here that DL4-mediated Notch signaling is essential for the development of both αß and γδ T-cell lineages in the embryo. Deletion of the DL4 gene in fetal TECs results in an early block in αß T-cell development and a dramatic reduction of all γδ T-cell subsets in the fetal thymus. In contrast to the adult, no dramatic deviation of T-cell precursors to alternative fates was observed in the fetal thymus in the absence of Notch signaling. Taken together, our data reveal a common requirement for DL4-mediated Notch signaling in fetal and adult thymopoiesis.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Lymphopoiesis , Membrane Proteins/metabolism , Precursor Cells, T-Lymphoid/immunology , T-Lymphocyte Subsets/immunology , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal/immunology , Calcium-Binding Proteins , Cell Lineage , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Receptor, Notch1/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction/immunology , Thymus Gland/embryology
6.
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
7.
J Immunol ; 186(4): 2529-34, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21257968

ABSTRACT

Although the importance of the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome in health and disease is well appreciated, a precise characterization of NLRP3 expression is yet undetermined. To this purpose, we generated a knock-in mouse in which the Nlrp3 coding sequence was substituted for the GFP (enhanced GFP [egfp]) gene. In this way, the expression of eGFP is driven by the endogenous regulatory elements of the Nlrp3 gene. In this study, we show that eGFP expression indeed mirrors that of NLRP3. Interestingly, splenic neutrophils, macrophages, and, in particular, monocytes and conventional dendritic cells showed robust eGFP fluorescence, whereas lymphoid subsets, eosinophils, and plasmacytoid dendritic cells showed negligible eGFP levels. NLRP3 expression was highly inducible in macrophages, both by MyD88- and Trif-dependent pathways. In vivo, when mice were challenged with diverse inflammatory stimuli, differences in both the number of eGFP-expressing cells and fluorescence intensity were observed in the draining lymph node. Thus, NLRP3 levels at the site of adaptive response initiation are controlled by recruitment of NLRP3-expressing cells and by NLRP3 induction.


Subject(s)
Carrier Proteins/biosynthesis , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carrier Proteins/genetics , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Fluorescent Dyes/metabolism , Gene Knock-In Techniques , Genes, Reporter/immunology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/deficiency , Green Fluorescent Proteins/genetics , Hematopoietic Stem Cells/pathology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Spleen/immunology , Spleen/metabolism , Spleen/pathology
8.
Innate Immun ; 17(3): 338-52, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20501515

ABSTRACT

Dendritic cells (DCs) serve as a link between the innate and adaptive immune systems. The activation state of DCs is crucial in this role. However, when DCs are isolated from lymphoid tissues, purified and placed in culture they undergo 'spontaneous' activation. The basis of this was explored, using up-regulation of DC surface MHC II, CD40, CD80 and CD86 as indicators of DC activation. No evidence was found for DC damage during isolation or for microbial products causing the activation. The culture activation of spleen DCs differed from that of Langerhans cells when released from E-cadherin-mediated adhesions, since E-cadherin was not detected and activation still occurred with ß-catenin null DCs. Much of the activation could be attributed to DC-DC interactions. Although increases in surface MHC II levels occurred under all culture conditions tested, the increase in expression of CD40, CD80 and CD86 was much less under culture conditions where such interactions were minimised. DC-to-DC contact under the artificial conditions of high DC concentration in culture induced the production of soluble factors and these, in turn, induced the up-regulation of co-stimulatory molecules on the DC surface.


Subject(s)
Antigens, CD/metabolism , Cell Communication , Cytokines/metabolism , Dendritic Cells/metabolism , Histocompatibility Antigens Class II/metabolism , Animals , Antigens, CD/genetics , Cadherins/metabolism , Cell Adhesion , Cell Communication/immunology , Cell Differentiation , Cells, Cultured , Chimera , Cytokines/genetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Histocompatibility Antigens Class II/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen , Up-Regulation/immunology , beta Catenin/genetics
9.
J Immunol ; 185(2): 1177-85, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20566828

ABSTRACT

In adaptive immunity, Th17 lymphocytes produce the IL-17 and IL-22 cytokines that stimulate mucosal antimicrobial defenses and tissue repair. In this study, we observed that the TLR5 agonist flagellin induced swift and transient transcription of genes encoding IL-17 and IL-22 in lymphoid, gut, and lung tissues. This innate response also temporarily enhanced the expression of genes associated with the antimicrobial Th17 signature. The source of the Th17-related cytokines was identified as novel populations of CD3(neg)CD127(+) immune cells among which CD4-expressing cells resembling lymphoid tissue inducer cells. We also demonstrated that dendritic cells are essential for expression of Th17-related cytokines and so for stimulation of innate cells. These data define that TLR-induced activation of CD3(neg)CD127(+) cells and production of Th17-related cytokines may be crucial for the early defenses against pathogen invasion of host tissues.


Subject(s)
Interleukin-17/immunology , Interleukins/immunology , Mucous Membrane/immunology , Signal Transduction/immunology , Spleen/immunology , Toll-Like Receptor 5/immunology , Animals , CD3 Complex/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flagellin/pharmacology , Flow Cytometry , Gene Expression/drug effects , Gene Expression/immunology , Ileum/drug effects , Ileum/immunology , Ileum/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Interleukins/genetics , Interleukins/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , Mucous Membrane/cytology , Mucous Membrane/metabolism , Oligonucleotide Array Sequence Analysis , Spleen/cytology , Spleen/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , Interleukin-22
10.
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
11.
PLoS One ; 5(2): e9258, 2010 Feb 18.
Article in English | MEDLINE | ID: mdl-20174635

ABSTRACT

BACKGROUND: The Notch pathway is essential for proper epidermal differentiation during embryonic skin development. Moreover, skin specific loss of Notch signaling in the embryo results in skin barrier defects accompanied by a B-lymphoproliferative disease. However, much less is known about the consequences of loss of Notch signaling after birth. METHODOLOGY AND PRINCIPAL FINDINGS: To study the function of Notch signaling in the skin of adult mice, we made use of a series of conditional gene targeted mice that allow inactivation of several components of the Notch signaling pathway specifically in the skin. We demonstrate that skin-specific inactivation of Notch1 and Notch2 simultaneously, or RBP-J, induces the development of a severe form of atopic dermatitis (AD), characterized by acanthosis, spongiosis and hyperkeratosis, as well as a massive dermal infiltration of eosinophils and mast cells. Likewise, patients suffering from AD, but not psoriasis or lichen planus, have a marked reduction of Notch receptor expression in the skin. Loss of Notch in keratinocytes induces the production of thymic stromal lymphopoietin (TSLP), a cytokine deeply implicated in the pathogenesis of AD. The AD-like associated inflammation is accompanied by a myeloproliferative disorder (MPD) characterized by an increase in immature myeloid populations in the bone marrow and spleen. Transplantation studies revealed that the MPD is cell non-autonomous and caused by dramatic microenvironmental alterations. Genetic studies demontrated that G-CSF mediates the MPD as well as changes in the bone marrow microenvironment leading to osteopenia. SIGNIFICANCE: Our data demonstrate a critical role for Notch in repressing TSLP production in keratinocytes, thereby maintaining integrity of the skin and the hematopoietic system.


Subject(s)
Dermatitis, Atopic/physiopathology , Myeloproliferative Disorders/physiopathology , Receptors, Notch/physiology , Signal Transduction/physiology , Skin/physiopathology , Animals , Cytokines/metabolism , Dermatitis, Atopic/genetics , Dermatitis, Atopic/mortality , Flow Cytometry , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Immunoglobulins , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, Transgenic , Models, Biological , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/mortality , Receptor, Notch1/genetics , Receptor, Notch1/physiology , Receptor, Notch2/genetics , Receptor, Notch2/physiology , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Skin/metabolism , Skin/pathology , Survival Analysis , Survival Rate , Thymic Stromal Lymphopoietin
12.
J Immunol ; 183(11): 7212-22, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19915064

ABSTRACT

It is well established that Notch signaling plays a critical role at multiple stages of T cell development and activation. However, detailed analysis of the cellular and molecular events associated with Notch signaling in T cells is hampered by the lack of reagents that can unambiguously measure cell surface Notch receptor expression. Using novel rat mAbs directed against the extracellular domains of Notch1 and Notch2, we find that Notch1 is already highly expressed on common lymphoid precursors in the bone marrow and remains at high levels during intrathymic maturation of CD4(-)CD8(-) thymocytes. Notch1 is progressively down-regulated at the CD4(+)CD8(+) and mature CD4(+) or CD8(+) thymic stages and is expressed at low levels on peripheral T cells. Immunofluorescence staining of thymus cryosections further revealed a localization of Notch1(+)CD25(-) cells adjacent to the thymus capsule. Notch1 was up-regulated on peripheral T cells following activation in vitro with anti-CD3 mAbs or infection in vivo with lymphocytic chorio-meningitis virus or Leishmania major. In contrast to Notch1, Notch2 was expressed at intermediate levels on common lymphoid precursors and CD117(+) early intrathymic subsets, but disappeared completely at subsequent stages of T cell development. However, transient up-regulation of Notch2 was also observed on peripheral T cells following anti-CD3 stimulation. Collectively our novel mAbs reveal a dynamic regulation of Notch1 and Notch2 surface expression during T cell development and activation. Furthermore they provide an important resource for future analysis of Notch receptors in various tissues including the hematopoietic system.


Subject(s)
Antibodies, Monoclonal/immunology , Lymphocyte Activation/immunology , Receptor, Notch1/biosynthesis , Receptor, Notch2/biosynthesis , Animals , Antibody Specificity , Cell Differentiation , Cell Membrane/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Receptor, Notch1/immunology , Receptor, Notch2/immunology , Signal Transduction/immunology , Stem Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
13.
J Immunol ; 182(8): 4641-8, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342639

ABSTRACT

Valpha14 invariant (Valpha14i) NKT cells are a subset of regulatory T cells that utilize a semi-invariant TCR to recognize glycolipids associated with monomorphic CD1d molecules. During development in the thymus, CD4(+)CD8(+) Valpha14i NKT precursors recognizing endogenous CD1d-associated glycolipids on other CD4(+)CD8(+) thymocytes are selected to undergo a maturation program involving sequential expression of CD44 and NK-related markers such as NK1.1. The molecular requirements for Valpha14i NKT cell maturation, particularly at early developmental stages, remain poorly understood. In this study, we show that CD4-Cre-mediated T cell-specific inactivation of c-Myc, a broadly expressed transcription factor with a wide range of biological activities, selectively impairs Valpha14i NKT cell development without perturbing the development of conventional T cells. In the absence of c-Myc, Valpha14i NKT cell precursors are blocked at an immature CD44(low)NK1.1(-) stage in a cell autonomous fashion. Residual c-Myc-deficient immature Valpha14i NKT cells appear to proliferate normally, cannot be rescued by transgenic expression of BCL-2, and exhibit characteristic features of immature Valpha14i NKT cells such as high levels of preformed IL-4 mRNA and the transcription factor promyelocytic leukemia zinc finger. Collectively our data identify c-Myc as a critical transcription factor that selectively acts early in Valpha14i NKT cell development to promote progression beyond the CD44(low)NK1.1(-) precursor stage.


Subject(s)
Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation/immunology , Haplotypes/immunology , Immunologic Memory/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Transgenic , Phenotype , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , Time Factors
14.
J Immunol ; 181(12): 8199-203, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050235

ABSTRACT

Interactions between Notch1 receptors on lymphoid progenitors and Delta-like 4 (DL4) ligands on cortical thymic epithelial cells (cTEC) are essential for T cell lineage commitment, expansion, and maturation in the thymus. Using a novel mAb against DL4, we show that DL4 levels on cTEC are very high in the fetal and neonatal thymus when thymocyte expansion is maximal but decrease dramatically in the adult when steady-state homeostasis is attained. Analysis of mutant mouse strains where thymocyte development is blocked at different stages indicates that lymphostromal interactions ("thymus crosstalk") are required for DL4 down-regulation on cTEC. Reconstitution of thymocyte development in these mutant mice further suggests that maturation of thymocytes to the CD4(+)CD8(+) stage and concomitant expansion are needed to promote DL4 down-regulation on cTEC. Collectively, our data support a model where thymic crosstalk quantitatively regulates the rate of Notch1-dependent thymopoiesis by controlling DL4 expression levels on cTEC.


Subject(s)
Cell Communication/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Membrane Proteins/biosynthesis , Thymus Gland/cytology , Thymus Gland/immunology , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Calcium-Binding Proteins , Cell Communication/genetics , Down-Regulation/immunology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Rats , Receptor, Notch1/physiology , Stromal Cells/cytology , Stromal Cells/immunology , Stromal Cells/metabolism , Thymus Gland/embryology , Thymus Gland/metabolism
15.
Cell Stem Cell ; 3(6): 611-24, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-19041778

ABSTRACT

Myc activity is emerging as a key element in acquisition and maintenance of stem cell properties. We have previously shown that c-Myc deficiency results in accumulation of defective hematopoietic stem cells (HSCs) due to niche-dependent differentiation defects. Here we report that immature HSCs coexpress c-myc and N-myc mRNA at similar levels. Although conditional deletion of N-myc in the bone marrow does not affect hematopoiesis, combined deficiency of c-Myc and N-Myc (dKO) results in pancytopenia and rapid lethality. Interestingly, proliferation of HSCs depends on both myc genes during homeostasis, but is c-Myc/N-Myc independent during bone marrow repair after injury. Strikingly, while most dKO hematopoietic cells undergo apoptosis, only self-renewing HSCs accumulate the cytotoxic molecule Granzyme B, normally employed by the innate immune system, thereby revealing an unexpected mechanism of stem cell apoptosis. Collectively, Myc activity (c-Myc and N-Myc) controls crucial aspects of HSC function including proliferation, differentiation, and survival.


Subject(s)
Cell Differentiation/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , Animals , Cell Lineage/genetics , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Graft Survival/genetics , Granzymes/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Pancytopenia/genetics , Pancytopenia/physiopathology , Signal Transduction/genetics , Stress, Physiological/genetics
16.
Eur J Immunol ; 37(11): 3220-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17918204

ABSTRACT

Although it is well established that early expression of TCRbeta transgenes in the thymus leads to efficient inhibition of both endogenous TCRbeta and TCRgamma rearrangement (also known as allelic and "isotypic" exclusion, respectively) the role of pTalpha in these processes remains controversial. Here, we have systematically re-evaluated this issue using three independent strains of TCRbeta-transgenic mice that differ widely in transgene expression levels, and a sensitive intracellular staining assay that detects endogenous TCRVbeta expression in individual immature thymocytes. In the absence of pTalpha, both allelic and isotypic exclusion were reversed in all three TCRbeta-transgenic strains, clearly demonstrating a general requirement for pre-TCR signaling in the inhibition of endogenous TCRbeta and TCRgamma rearrangement. Both allelic and isotypic exclusion were pTalpha dose dependent when transgenic TCRbeta levels were subphysiological. Moreover, pTalpha-dependent allelic and isotypic exclusion occurred in both alphabeta and gammadelta T cell lineages, indicating that pre-TCR signaling can potentially be functional in gammadelta precursors. Finally, levels of endogenous RAG1 and RAG2 were not down-regulated in TCRbeta-transgenic immature thymocytes undergoing allelic or isotypic exclusion. Collectively, our data reveal a critical but lineage-nonspecific role for pTalpha in mediating both allelic and isotypic exclusion in TCRbeta-transgenic mice.


Subject(s)
Cell Lineage/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Genes, T-Cell Receptor beta , Membrane Glycoproteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Alleles , Animals , Blotting, Western , Flow Cytometry , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
J Immunol ; 177(9): 6038-43, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056529

ABSTRACT

During thymus development, immature T cells become committed to two distinct lineages based upon expression of alphabeta or gammadelta TCR. In the alphabeta lineage, developing thymocytes progressively extinguish transcription of the TCRgamma genes by a poorly understood process known as gamma silencing. We show that alphabeta lineage thymocytes in mice lacking a functional pre-TCR undergo limited proliferation and fail to silence TCRgamma genes during development. Stimulation of pre-TCR-deficient immature thymocytes with anti-CD3 Abs does not directly down-regulate TCRgamma transcription but restores TCRgamma silencing following proliferation. Collectively our data reveal an important role for pre-TCR induced proliferation in activating the TCRgamma silencer in alphabeta lineage thymocytes, a process that may reinforce alphabeta or gammadelta lineage commitment.


Subject(s)
Gene Silencing , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocyte Subsets/immunology , Thymus Gland/growth & development , Animals , Antibodies/pharmacology , CD3 Complex/immunology , Cell Lineage/genetics , Cell Proliferation , Mice , Receptors, Antigen, T-Cell, alpha-beta/agonists , Receptors, Antigen, T-Cell, gamma-delta/agonists , Silencer Elements, Transcriptional/genetics , T-Lymphocyte Subsets/drug effects , Thymus Gland/cytology , Thymus Gland/immunology
18.
J Immunol ; 172(11): 6922-30, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15153511

ABSTRACT

Activation of dendritic cells (DC) by microbial products via Toll-like receptors (TLR) is instrumental in the induction of immunity. In particular, TLR signaling plays a major role in the instruction of Th1 responses. The development of Th2 responses has been proposed to be independent of the adapter molecule myeloid differentiation factor 88 (MyD88) involved in signal transduction by TLRs. In this study we show that flagellin, the bacterial stimulus for TLR5, drives MyD88-dependent Th2-type immunity in mice. Flagellin promotes the secretion of IL-4 and IL-13 by Ag-specific CD4(+) T cells as well as IgG1 responses. The Th2-biased responses are associated with the maturation of DCs, which are shown to express TLR5. Flagellin-mediated DC activation requires MyD88 and induces NF-kappaB-dependent transcription and the production of low levels of proinflammatory cytokines. In addition, the flagellin-specific response is characterized by the lack of secretion of the Th1-promoting cytokine IL-12 p70. In conclusion, this study suggests that flagellin and, more generally, TLR ligands can control Th2 responses in a MyD88-dependent manner.


Subject(s)
Antigens, Differentiation/physiology , Flagellin/pharmacology , Receptors, Immunologic/physiology , Th2 Cells/immunology , Adaptor Proteins, Signal Transducing , Animals , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/physiology , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Membrane Glycoproteins/physiology , Mice , Mice, Inbred Strains , Myeloid Differentiation Factor 88 , Ovalbumin/immunology , Protein Subunits/biosynthesis , Receptors, Cell Surface/physiology , Toll-Like Receptor 5 , Toll-Like Receptors
19.
J Immunol ; 170(10): 4926-32, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12734335

ABSTRACT

There has been controversy over the possible lymphoid origin of certain dendritic cell (DC) subtypes. To resolve this issue, DC and plasmacytoid pre-DC isolated from normal mouse tissues were analyzed for transient (mRNA) and permanent (DNA rearrangement) markers of early stages of lymphoid development. About 27% of the DNA of CD8(+) DC from thymus, and 22-35% of the DNA of plasmacytoid pre-DC from spleen and thymus, was found to contain IgH gene D-J rearrangements, compared with 40% for T cells. However, the DC DNA did not contain IgH gene V-D-J rearrangements nor T cell Ag receptor beta gene D-J rearrangements. The same DC lineage populations containing IgH D-J rearrangements expressed mRNA for CD3 chains, and for pre-T alpha. In contrast, little of the DNA of the conventional DC derived from spleen, lymph nodes, or skin, whether CD8(+) or CD8(-), contained IgH D-J rearrangements and splenic conventional DC expressed very little CD3 epsilon or pre-T alpha mRNA. Therefore, many plasmacytoid pre-DC and thymic CD8(+) DC have shared early steps of development with the lymphoid lineages, and differ in origin from conventional peripheral DC.


Subject(s)
CD3 Complex , Dendritic Cells/cytology , Lymphoid Tissue/cytology , Plasma Cells/cytology , Thymus Gland/cytology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Complementarity Determining Regions/biosynthesis , Complementarity Determining Regions/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, T-Cell Receptor alpha , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis/genetics , Phagocytosis/immunology , Plasma Cells/immunology , Plasma Cells/metabolism , RNA, Messenger/biosynthesis , Radiation Chimera/immunology , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism
20.
Blood ; 100(8): 2852-7, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12351395

ABSTRACT

The developmental origin of dendritic cells (DCs) is controversial. In the mouse CD8alpha(+) and CD8alpha(-) DC subsets are often considered to be of lymphoid and myeloid origin respectively, although evidence on this point is conflicting. Very recently a novel CD11c(+) B220(+) DC subset has been identified that appears to be the murine counterpart to interferon alpha (IFNalpha)-producing human plasmacytoid DCs (PDCs). We show here that CD11c(+) B220(+) mouse PDCs, like human PDCs, are present in the thymus and express T lineage markers such as CD8alpha and CD4. However, the intrathymic development of PDCs can be completely dissociated from immature T lineage cells in mixed chimeras established with bone marrow cells from mice deficient for either Notch-1 or T-cell factor 1, two independent mutations that severely block early T-cell development. Our data indicate that thymic PDCs do not arise from a bipotential T/DC precursor.


Subject(s)
CD11c Antigen/blood , Dendritic Cells/immunology , Leukocyte Common Antigens/blood , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Chimera/immunology , Humans , Mice , Mice, Inbred C57BL , Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...