Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Immunol ; 212(2): 284-294, 2024 01 15.
Article in English | MEDLINE | ID: mdl-37991420

ABSTRACT

There is considerable interest in therapeutically engaging human γδ T cells. However, due to the unique TCRs of human γδ T cells, studies from animal models have provided limited directly applicable insights, and human γδ T cells from key immunological tissues remain poorly characterized. In this study, we investigated γδ T cells from human spleen tissue. Compared to blood, where Vδ2+Vγ9+ T cells are the dominant subset, splenic γδ T cells included a variety of TCR types, with Vδ1+ T cells typically being the most frequent. Intracellular cytokine staining revealed that IFN-γ was produced by a substantial fraction of splenic γδ T cells, IL-17A by a small fraction, and IL-4 was minimal. Primary splenic γδ T cells frequently expressed NKG2D (NK group 2 member D) and CD16, whereas expression of DNAM-1 (DNAX accessory molecule 1), CD28, PD-1, TIGIT, and CD94 varied according to subset, and there was generally little expression of natural cytotoxicity receptors, TIM-3, LAG-3, or killer Ig-like receptors. In vitro expansion was associated with marked changes in expression of these activating and inhibitory receptors. Analysis of functional responses of spleen-derived Vδ2+Vγ9+, Vδ1+Vγ9+, and Vδ1+Vγ9- T cell lines to recombinant butyrophilin BTN2A1 and BTN3A1 demonstrated that both Vδ2+Vγ9+ and Vδ1+Vγ9+ T cells were capable of responding to the extracellular domain of BTN2A1, whereas the addition of BTN3A1 only markedly enhanced the responses of Vδ2+Vγ9+ T cells. Conversely, Vδ1+Vγ9+ T cells appeared more responsive than Vδ2+Vγ9+ T cells to TCR-independent NKG2D stimulation. Thus, despite shared recognition of BTN2A1, differential effects of BTN3A1 and coreceptors may segregate target cell responses of Vδ2+Vγ9+ and Vδ1+Vγ9+ T cells.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , Spleen , Animals , Humans , Spleen/metabolism , Butyrophilins , NK Cell Lectin-Like Receptor Subfamily K , T-Lymphocytes , Antigens, CD
2.
J Immunol ; 209(8): 1475-1480, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36096643

ABSTRACT

Vγ9Vδ2+ T cell-targeted immunotherapy is of interest to harness its MHC-independent cytotoxic potential against a variety of cancers. Recent studies have identified heterodimeric butyrophilin (BTN) 2A1 and BTN3A1 as the molecular entity providing "signal 1" to the Vγ9Vδ2 TCR, but "signal 2" costimulatory requirements remain unclear. Using a tumor cell-free assay, we demonstrated that a BTN2A1/3A1 heterodimeric fusion protein activated human Vγ9Vδ2+ T cells, but only in the presence of costimulatory signal via CD28 or NK group 2 member D. Nonetheless, addition of a bispecific γδ T cell engager BTN2A1/3A1-Fc-CD19scFv alone enhanced granzyme B-mediated killing of human CD19+ lymphoma cells when cocultured with Vγ9Vδ2+ T cells, suggesting expression of costimulatory ligand(s) on tumor cells is sufficient to satisfy the "signal 2" requirement. These results highlight the parallels of signal 1 and signal 2 requirements in αß and γδ T cell activation and demonstrate the utility of heterodimeric BTNs to promote targeted activation of γδ T cells.


Subject(s)
CD28 Antigens , Receptors, Antigen, T-Cell, gamma-delta , Antigens, CD/metabolism , Butyrophilins/metabolism , Granzymes , Humans , Ligands , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Antigen, T-Cell, gamma-delta/metabolism
3.
Immunity ; 38(6): 1105-15, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23791645

ABSTRACT

How hematopoietic stem cells (HSCs) produce particular lineages is insufficiently understood. We searched for key factors that direct HSC to lymphopoiesis. Comparing gene expression profiles for HSCs and early lymphoid progenitors revealed that Satb1, a global chromatin regulator, was markedly induced with lymphoid lineage specification. HSCs from Satb1-deficient mice were defective in lymphopoietic activity in culture and failed to reconstitute T lymphopoiesis in wild-type recipients. Furthermore, Satb1 transduction of HSCs and embryonic stem cells robustly promoted their differentiation toward lymphocytes. Whereas genes that encode Ikaros, E2A, and Notch1 were unaffected, many genes involved in lineage decisions were regulated by Satb1. Satb1 expression was reduced in aged HSCs with compromised lymphopoietic potential, but forced Satb1 expression partly restored that potential. Thus, Satb1 governs the initiating process central to the replenishing of lymphoid lineages. Such activity in lymphoid cell generation may be of clinical importance and useful to overcome immunosenescence.


Subject(s)
Hematopoietic Stem Cells/physiology , Lymphopoiesis , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Survival/genetics , Cells, Cultured , Cellular Senescence/genetics , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation , Humans , Lymphopoiesis/genetics , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Transgenes/genetics
4.
Genome Res ; 23(12): 2030-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24013550

ABSTRACT

Memory is a hallmark of adaptive immunity, wherein lymphocytes mount a superior response to a previously encountered antigen. It has been speculated that epigenetic alterations in memory lymphocytes contribute to their functional distinction from their naive counterparts. However, the nature and extent of epigenetic alterations in memory compartments remain poorly characterized. Here we profile the DNA methylome and the transcriptome of B-lymphocyte subsets representing stages of the humoral immune response before and after antigen exposure in vivo from multiple humans. A significant percentage of activation-induced losses of DNA methylation mapped to transcription factor binding sites. An additional class of demethylated loci mapped to Alu elements across the genome and accompanied repression of DNA methyltransferase 3A. The activation-dependent DNA methylation changes were largely retained in the progeny of activated B cells, generating a similar epigenetic signature in downstream memory B cells and plasma cells with distinct transcriptional programs. These findings provide insights into the methylation dynamics of the genome during cellular differentiation in an immune response.


Subject(s)
Alu Elements , B-Lymphocytes/immunology , DNA Methylation , Lymphocyte Activation/genetics , Regulatory Elements, Transcriptional/genetics , Adaptive Immunity/genetics , Adaptive Immunity/immunology , B-Lymphocytes/metabolism , Binding Sites/genetics , Cell Differentiation/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Genome, Human , Humans , Immunologic Memory/genetics , Plasma Cells/immunology , Plasma Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Biochem J ; 459(3): 505-12, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24576085

ABSTRACT

The tandem PHD (plant homeodomain) fingers of the CHD4 (chromodomain helicase DNA-binding protein 4) ATPase are epigenetic readers that bind either unmodified histone H3 tails or H3K9me3 (histone H3 trimethylated at Lys9). This dual function is necessary for the transcriptional and chromatin remodelling activities of the NuRD (nucleosome remodelling and deacetylase) complex. In the present paper, we show that calixarene-based supramolecular hosts disrupt binding of the CHD4 PHD2 finger to H3K9me3, but do not affect the interaction of this protein with the H3K9me0 (unmodified histone H3) tail. A similar inhibitory effect, observed for the association of chromodomain of HP1γ (heterochromatin protein 1γ) with H3K9me3, points to a general mechanism of methyl-lysine caging by calixarenes and suggests a high potential for these compounds in biochemical applications. Immunofluorescence analysis reveals that the supramolecular agents induce changes in chromatin organization that are consistent with their binding to and disruption of H3K9me3 sites in living cells. The results of the present study suggest that the aromatic macrocyclic hosts can be used as a powerful new tool for characterizing methylation-driven epigenetic mechanisms.


Subject(s)
Calixarenes/pharmacology , Chromatin Assembly and Disassembly/drug effects , Drug Design , Histones/antagonists & inhibitors , Indicators and Reagents/pharmacology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/antagonists & inhibitors , Models, Molecular , Autoantigens/chemistry , Autoantigens/genetics , Autoantigens/metabolism , Calixarenes/chemical synthesis , Calixarenes/chemistry , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic/drug effects , HEK293 Cells , Histones/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/chemistry , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Indicators and Reagents/chemical synthesis , Indicators and Reagents/chemistry , Lysine/analogs & derivatives , Lysine/metabolism , Methylation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/chemistry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs/drug effects , Protein Processing, Post-Translational , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
6.
J Exp Med ; 203(8): 1867-73, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16880261

ABSTRACT

The mechanism of lineage commitment from hematopoietic stem cells (HSCs) is not well understood. Although commitment to either the lymphoid or the myeloid lineage is popularly viewed as the first step of lineage restriction from HSCs, this model of hematopoietic differentiation has recently been challenged. The previous identification of multipotent progenitors (MPPs) that can produce lymphocytes and granulocyte/macrophages (GMs) but lacks erythroid differentiation ability suggests the existence of an alternative HSC differentiation program. Contribution to different hematopoietic lineages by these MPPs under physiological conditions, however, has not been carefully examined. In this study, we performed a refined characterization of MPPs by subfractionating three distinct subsets based on Flt3 and vascular cell adhesion molecule 1 expression. These MPP subsets differ in their ability to give rise to erythroid and GM lineage cells but are equally potent in lymphoid lineage differentiation in vivo. The developmental hierarchy of these MPP subsets demonstrates the sequential loss of erythroid and then GM differentiation potential during early hematopoiesis. Our results suggest that the first step of lineage commitment from HSCs is not simply a selection between the lymphoid and the myeloid lineage.


Subject(s)
Cell Lineage , Lymphocytes/cytology , Macrophages/cytology , Multipotent Stem Cells/cytology , Myeloid Progenitor Cells/cytology , Animals , Cell Differentiation , Erythroid Cells/cytology , Mice , Mice, Inbred C57BL , Vascular Cell Adhesion Molecule-1/metabolism , fms-Like Tyrosine Kinase 3/immunology
7.
J Exp Med ; 201(8): 1197-203, 2005 Apr 18.
Article in English | MEDLINE | ID: mdl-15837809

ABSTRACT

Cytokine receptor signals have been suggested to stimulate cell differentiation during hemato/lymphopoiesis. Such action, however, has not been clearly demonstrated. Here, we show that adult B cell development in IL-7(-/-) and IL-7R alpha(2/-) mice is arrested at the pre-pro-B cell stage due to insufficient expression of the B cell-specific transcription factor EBF and its target genes, which form a transcription factor network in determining B lineage specification. EBF expression is restored in IL-7(-/-) pre-pro-B cells upon IL-7 stimulation or in IL-7R alpha(-/-) pre-pro-B cells by activation of STAT5, a major signaling molecule downstream of the IL-7R signaling pathway. Furthermore, enforced EBF expression partially rescues B cell development in IL-7R alpha(-/-) mice. Thus, IL-7 receptor signaling is a participant in the formation of the transcription factor network during B lymphopoiesis by up-regulating EBF, allowing stage transition from the pre-pro-B to further maturational stages.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/biosynthesis , Lymphopoiesis , Receptors, Interleukin-7/metabolism , Trans-Activators/biosynthesis , Animals , B-Lymphocytes/drug effects , DNA-Binding Proteins/metabolism , Interleukin-7/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Milk Proteins/metabolism , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , STAT5 Transcription Factor , Signal Transduction , Trans-Activators/metabolism , Up-Regulation
8.
Blood ; 113(23): 5757-64, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19363218

ABSTRACT

Lymphoid and myeloid lineage segregation is a major developmental step during early hematopoiesis from hematopoietic stem cells. It is not clear, however, whether multipotent progenitors (MPPs) adopt a lymphoid or myeloid fate through stochastic mechanisms, or whether this process can be regulated by extracellular stimuli. In this study, we show that lymphoid lineage specification occurs in MPPs before lymphoid lineage priming, during which MPPs migrate from the proximal to the distal region relative to the endosteum of the bone marrow. Lymphoid-specified MPPs have low myeloid differentiation potential in vivo, but potently differentiate into myeloid cells in vitro. When treated with pertussis toxin, an inhibitor of G protein-coupled receptor signaling, lymphoid-specified MPPs regain in vivo myeloid potential, and their localization is dispersed in the bone marrow. These results clearly demonstrate that specific microenvironments that favorably support lymphoid or myeloid lineage development exist at structurally distinct regions in the bone marrow.


Subject(s)
Cell Lineage/drug effects , Hematopoietic Stem Cells/cytology , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Lymphoid Tissue/cytology , Lymphoid Tissue/drug effects , Multipotent Stem Cells/cytology , Pertussis Toxin/pharmacology , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cells, Cultured , Granulocytes/cytology , Granulocytes/drug effects , Granulocytes/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Lymphoid Tissue/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
9.
J Immunother Cancer ; 8(2)2020 10.
Article in English | MEDLINE | ID: mdl-33004541

ABSTRACT

BACKGROUND: Combination treatment with chemotherapy and immune checkpoint inhibitors (ICIs) has demonstrated meaningful clinical benefit to patients. However, chemotherapy-induced damage to the immune system can potentially diminish the efficacy of chemotherapy/ICI combinations. Trilaciclib, a highly potent, selective and reversible cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor in development to preserve hematopoietic stem and progenitor cells and immune system function during chemotherapy, has demonstrated proof of concept in recent clinical trials. Furthermore, CDK4/6 inhibition has been shown to augment T-cell activation and antitumor immunity in preclinical settings. Therefore, addition of trilaciclib has the potential to further enhance the efficacy of chemotherapy and ICI combinations. METHODS: In murine syngeneic tumor models, a schedule of 3 weekly doses of trilaciclib was combined with chemotherapy/ICI regimens to assess the effect of transient CDK4/6 inhibition on antitumor response and intratumor T-cell proliferation and function. Peripheral T-cell status was also analyzed in patients with small cell lung cancer (SCLC) treated with chemotherapy with or without trilaciclib to gain insights into the effect of transient exposure of trilaciclib on T-cell activation. RESULTS: Preclinically, the addition of trilaciclib to chemotherapy/ICI regimens enhanced antitumor response and overall survival compared with chemotherapy and ICI combinations alone. This effect is associated with the modulation of the proliferation and composition of T-cell subsets in the tumor microenvironment and increased effector function. Transient exposure of trilaciclib in patients with SCLC during chemotherapy treatment both preserved and increased peripheral lymphocyte counts and enhanced T-cell activation, suggesting that trilaciclib not only preserved but also enhanced immune system function. CONCLUSIONS: Transient CDK4/6 inhibition by trilaciclib was sufficient to enhance and prolong the duration of the antitumor response by chemotherapy/ICI combinations, suggesting a role for the transient cell cycle arrest of tumor immune infiltrates in remodeling the tumor microenvironment. These results provide a rationale for combining trilaciclib with chemotherapy/ICI regimens to improve antitumor efficacy in patients with cancer.


Subject(s)
Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lymphocyte Activation/drug effects , Small Cell Lung Carcinoma/drug therapy , Animals , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/pathology , Mice , Small Cell Lung Carcinoma/pathology
10.
Environ Health Perspect ; 122(3): 262-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24316720

ABSTRACT

BACKGROUND: Diethylstilbestrol (DES) is a synthetic estrogen associated with adverse effects on reproductive organs. DES-induced toxicity of the mouse seminal vesicle (SV) is mediated by estrogen receptor α (ERα), which alters expression of seminal vesicle secretory protein IV (Svs4) and lactoferrin (Ltf) genes. OBJECTIVES: We examined a role for nuclear receptor activity in association with DNA methylation and altered gene expression. METHODS: We used the neonatal DES exposure mouse model to examine DNA methylation patterns via bisulfite conversion sequencing in SVs of wild-type (WT) and ERα-knockout (αERKO) mice. RESULTS: The DNA methylation status at four specific CpGs (-160, -237, -306, and -367) in the Svs4 gene promoter changed during mouse development from methylated to unmethylated, and DES prevented this change at 10 weeks of age in WT SV. At two specific CpGs (-449 and -459) of the Ltf gene promoter, DES altered the methylation status from methylated to unmethylated. Alterations in DNA methylation of Svs4 and Ltf were not observed in αERKO SVs, suggesting that changes of methylation status at these CpGs are ERα dependent. The methylation status was associated with the level of gene expression. In addition, gene expression of three epigenetic modifiers-DNMT3A, MBD2, and HDAC2-increased in the SV of DES-exposed WT mice. CONCLUSION: DES-induced hormonal toxicity resulted from altered gene expression of Svs4 and Ltf associated with changes in DNA methylation that were mediated by ERα. Alterations in gene expression of DNMT3A, MBD2, and HDAC2 in DES-exposed male mice may be involved in mediating the changes in methylation status in the SV. CITATION: Li Y, Hamilton KJ, Lai AY, Burns KA, Li L, Wade PA, Korach KS. 2014. Diethylstilbestrol (DES)-stimulated hormonal toxicity is mediated by ERα alteration of target gene methylation patterns and epigenetic modifiers (DNMT3A, MBD2, and HDAC2) in the mouse seminal vesicle. Environ Health Perspect 122:262-268; http://dx.doi.org/10.1289/ehp.1307351.


Subject(s)
DNA Methylation , Diethylstilbestrol/toxicity , Environmental Exposure , Epigenesis, Genetic , Estrogen Receptor alpha/genetics , Gene Expression , Seminal Vesicles/drug effects , Animals , DNA Methylation/drug effects , Estrogen Receptor alpha/metabolism , Lactoferrin/genetics , Lactoferrin/metabolism , Male , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Seminal Vesicle Secretory Proteins/genetics , Seminal Vesicle Secretory Proteins/metabolism , Seminal Vesicles/metabolism
11.
Nat Rev Cancer ; 11(8): 588-96, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21734722

ABSTRACT

The nucleosome remodelling and histone deacetylase (NuRD; also known as Mi-2) complex regulates gene expression at the level of chromatin. The NuRD complex has been identified - using both genetic and molecular analyses - as a key determinant of differentiation in mouse embryonic stem cells and during development in various model systems. Similar to other chromatin remodellers, such as SWI/SNF and Polycomb complexes, NuRD has also been implicated in the regulation of transcriptional events that are integral to oncogenesis and cancer progression. Emerging molecular details regarding the recruitment of NuRD to specific loci during development, and the modulation of these events in cancer, are used to illustrate how the inappropriate localization of the complex could contribute to tumour biology.


Subject(s)
Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Neoplasms/genetics , Neoplasms/physiopathology , Animals , Chromatin , Chromatin Assembly and Disassembly , DNA Methylation , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Silencing , Genomic Instability , Humans , Neoplasms/drug therapy , Promoter Regions, Genetic
12.
J Exp Med ; 207(9): 1939-50, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20733034

ABSTRACT

Aberrant DNA methylation commonly occurs in cancer cells where it has been implicated in the epigenetic silencing of tumor suppressor genes. Additional roles for DNA methylation, such as transcriptional activation, have been predicted but have yet to be clearly demonstrated. The BCL6 oncogene is implicated in the pathogenesis of germinal center-derived B cell lymphomas. We demonstrate that the intragenic CpG islands within the first intron of the human BCL6 locus were hypermethylated in lymphoma cells that expressed high amounts of BCL6 messenger RNA (mRNA). Inhibition of DNA methyltransferases decreased BCL6 mRNA abundance, suggesting a role for these methylated CpGs in positively regulating BCL6 transcription. The enhancer-blocking transcription factor CTCF bound to this intronic region in a methylation-sensitive manner. Depletion of CTCF by short hairpin RNA in neoplastic plasma cells that do not express BCL6 resulted in up-regulation of BCL6 transcription. These data indicate that BCL6 expression is maintained during lymphomagenesis in part through DNA methylation that prevents CTCF-mediated silencing.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Lymphoma, B-Cell/metabolism , Repressor Proteins/metabolism , CCCTC-Binding Factor , Cell Line, Tumor , CpG Islands , DNA-Binding Proteins/genetics , Humans , Introns , Lymphoma, B-Cell/genetics , Oncogenes , Protein Binding , Proto-Oncogene Proteins c-bcl-6 , Repressor Proteins/genetics , Transcription, Genetic
13.
Semin Immunol ; 20(4): 207-12, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18583148

ABSTRACT

Until the past few years, it has been thought that lymphoid and myeloid lineage segregation represents the first step of lineage restriction during hematopoiesis from hematopoietic stem cell. Recent investigation of the cell populations within multipotent progenitors in the bone marrow has led to new understanding of how hematopoietic stem cells diversify into different hematopoietic cell types. This review focuses on the recent advances in understanding the developmental events that occur during hematopoietic stem cell specification into the T and B lymphocyte lineages in adult mice.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Hematopoietic Stem Cells/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/cytology , Hematopoietic Stem Cells/cytology , Humans , Mice , T-Lymphocytes/cytology
14.
J Immunol ; 181(1): 383-92, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18566404

ABSTRACT

IL-7 plays a critical role in B cell fate decision by regulating early B cell factor (EBF) expression. However, it was not clear when IL-7 stimulation is necessary in hemato-/lymphopoiesis in adult mice. Here we show that pre-proB cells derived from IL-7-/- mice have lost B cell potential, despite up-regulation of EBF expression following IL-7 stimulation. Pre-proB cells from wild-type mice can give rise to proB cells in the absence of IL-7. In this case, EBF up-regulation during the transition from the pre-proB to proB stages occurs normally. In contrast, EBF expression by IL-7-/- pre-proB cells after IL-7 stimulation is approximately 20 times lower than wild-type pre-proB cells. In addition, only multipotent progenitors with higher levels of ectopic EBF can give rise to proB cells in the absence of IL-7. Therefore, the primary function of IL-7 before the pre-proB stage in B cell development is to maintain the EBF expression level above a certain threshold, which is necessary for pre-proB cells to further transit to the proB stage.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Interleukin-7/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Lymphopoiesis/immunology , Trans-Activators/metabolism , Animals , B-Lymphocytes/metabolism , Interleukin-7/deficiency , Interleukin-7/genetics , Interleukin-7/metabolism , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/metabolism , Signal Transduction , Time Factors , Trans-Activators/genetics , Trans-Activators/immunology
15.
Proc Natl Acad Sci U S A ; 104(15): 6311-6, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17404232

ABSTRACT

The thymus requires continuous replenishment of progenitors from the bone marrow (BM) to sustain T cell development. However, it remains unclear which hematopoietic progenitors downstream from hematopoietic stem cells in the BM home to the thymus in adult mice. In this work, we demonstrate that although multiple BM populations have intrinsic T lineage differentiation potential, a small subset of multipotent progenitors (MPPs) expressing CCR9 preferentially homes to the thymus. These CCR9(+) MPPs are phenotypically similar to the most immature early T lineage progenitors (ETPs) in the thymus and are present in the peripheral blood. Similar to ETPs, CCR9(+) MPPs undergo Notch signaling, as indicated by higher expression of Notch1 and downstream target Hes1 genes compared with other MPP subsets. Furthermore, CCR9(+) MPPs possess differentiation potential similar to that of ETPs, with very limited granulocyte/macrophage differentiation potential, but they can differentiate into T, B, and dendritic cells. These characteristics implicate CCR9(+) MPPs as the BM precursors of the earliest thymic progenitors. In addition, our data suggest that before transition from BM to thymus, MPPs are lymphoid-specified and primed for T lineage differentiation.


Subject(s)
Cell Differentiation/immunology , Hematopoietic Stem Cells/cytology , Multipotent Stem Cells/cytology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Flow Cytometry , Homeodomain Proteins/metabolism , Immunophenotyping , Mice , Receptor, Notch1/metabolism , Receptors, CCR , Receptors, Chemokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Thymus Gland/immunology , Transcription Factor HES-1
16.
J Immunol ; 178(9): 5717-26, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17442955

ABSTRACT

Thymocytes undergoing TCRbeta gene rearrangements are maintained in a low or nonproliferating state during early T cell development. This block in cell cycle progression is not released until the expression of a functional pre-TCR, which is composed of a successfully rearranged TCRbeta-chain and the Pre-Talpha-chain. The regulatory molecules responsible for the coordination of these differentiation and proliferation events are currently unknown. E2A and HEB are structurally and functionally related basic helix-loop-helix transcription factors involved in T cell development. To reveal the function of E2A and HEB through the stage of pre-TCR expression and alleviate functional compensation between E2A and HEB, we use a double-conditional knockout model. The simultaneous deletion of E2A and HEB in developing thymocytes leads to a severe developmental block before pre-TCR expression and a dramatic reduction of Pre-Talpha expression. These developmentally arrested thymocytes exhibit increased proliferation in vivo and dramatic expansion ex vivo in response to IL-7 signaling. These results suggest that E2A and HEB are not only critical for T cell differentiation but also necessary to retain developing thymocytes in cell cycle arrest before pre-TCR expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Lymphocyte Activation/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology , TCF Transcription Factors/physiology , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle/genetics , Cell Proliferation , Gene Expression Regulation , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics , Genes, T-Cell Receptor beta/genetics , Interleukin-7/physiology , Lymphocyte Count , Mice , Mice, Knockout , Sequence Deletion , TCF Transcription Factors/genetics , Transcription Factor 7-Like 1 Protein
17.
Proc Natl Acad Sci U S A ; 103(3): 672-7, 2006 Jan 17.
Article in English | MEDLINE | ID: mdl-16407117

ABSTRACT

Lymphoid lineage-committed progenitors, such as common lymphoid progenitors (CLPs), maintain a latent myeloid differentiation potential, which can be initiated by stimulation through exogenously expressed cytokine receptors, including IL-2 receptors. Here we show that the transcription factor CCAAT enhancer-binding protein-alpha (C/EBPalpha) is promptly up-regulated in CLPs upon ectopic IL-2 stimulation. Enforced C/EBPalpha expression is sufficient to initiate myeloid differentiation from CLPs, as well as from proT and proB cells, even though proB cells do not give rise to myeloid cells after ectopic IL-2 stimulation. Expression of Pax5, a B lymphoid-affiliated transcription factor, is completely suppressed by enforced C/EBPalpha but not by ectopic IL-2 stimulation in proB cells. Introduction of Pax5 blocks ectopic IL-2 receptor-mediated myeloid lineage conversion in CLPs. These data suggest that C/EBPalpha is a proximal target of cytokine-induced lineage conversion in lymphoid progenitors. Furthermore, complete loss of Pax5 expression triggered by up-regulation of C/EBPalpha is a critical event for lineage conversion from lymphoid to myeloid lineage in CLPs and proB cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/physiology , Lymphocytes/physiology , Myeloid Cells/physiology , PAX5 Transcription Factor/physiology , Stem Cells/physiology , Animals , CCAAT-Enhancer-Binding Protein-alpha/biosynthesis , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Cell Lineage/physiology , Cells, Cultured , Down-Regulation/genetics , Down-Regulation/physiology , Humans , Interleukin-2 Receptor beta Subunit , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , PAX5 Transcription Factor/antagonists & inhibitors , PAX5 Transcription Factor/biosynthesis , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Stem Cells/cytology , Up-Regulation/genetics , Up-Regulation/physiology
18.
J Immunol ; 175(8): 5016-23, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16210604

ABSTRACT

Mechanisms of lymphoid and myeloid lineage choice by hemopoietic stem cells remain unclear. In this study we show that the multipotent progenitor (MPP) population, which is immediately downstream of hemopoietic stem cells, is heterogeneous and can be subdivided in terms of VCAM-1 expression. VCAM-1(+) MPPs were fully capable of differentiating into both lymphoid and myeloid lineages. In contrast, VCAM-1(-) MPPs gave rise to lymphocytes predominately in vivo. T and B cell development from VCAM-1(-) MPPs was 1 wk faster than that from VCAM-1(+) MPPs. Furthermore, VCAM-1(+) MPPs gave rise to common myeloid progenitors and VCAM-1(-) MPPs in vivo, indicating that VCAM-1(-) MPPs are progenies of VCAM-1(+) MPPs. VCAM-1(-) MPPs, in turn, developed into lymphoid lineage-restricted common lymphoid progenitors. These results establish a hierarchy of developmental relationship between MPP subsets and lymphoid and myeloid progenitors. In addition, VCAM-1(+) MPPs may represent the branching point between the lymphoid and myeloid lineages.


Subject(s)
Bone Marrow Cells/enzymology , Multipotent Stem Cells/enzymology , fms-Like Tyrosine Kinase 3/biosynthesis , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Lymphoid Tissue/cytology , Lymphoid Tissue/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/classification , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Myeloid Cells/cytology , Myeloid Cells/enzymology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , fms-Like Tyrosine Kinase 3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL