Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Immunol ; 209(1): 77-92, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35705252

ABSTRACT

The zinc-finger transcription factor GATA-3 plays a crucial role during early T cell development and also dictates later T cell differentiation outcomes. However, its role and collaboration with the Notch signaling pathway in the induction of T lineage specification and commitment have not been fully elucidated. We show that GATA-3 deficiency in mouse hematopoietic progenitors results in an early block in T cell development despite the presence of Notch signals, with a failure to upregulate Bcl11b expression, leading to a diversion along a myeloid, but not a B cell, lineage fate. GATA-3 deficiency in the presence of Notch signaling results in the apoptosis of early T lineage cells, as seen with inhibition of CDK4/6 (cyclin-dependent kinases 4 and 6) function, and dysregulated cyclin-dependent kinase inhibitor 2b (Cdkn2b) expression. We also show that GATA-3 induces Bcl11b, and together with Bcl11b represses Cdkn2b expression; however, loss of Cdkn2b failed to rescue the developmental block of GATA-3-deficient T cell progenitor. Our findings provide a signaling and transcriptional network by which the T lineage program in response to Notch signals is realized.


Subject(s)
GATA3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes , Animals , Cell Differentiation , Cell Lineage , Cyclin-Dependent Kinase Inhibitor Proteins , Gene Regulatory Networks , Mice , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/metabolism
2.
J Immunol ; 202(10): 2837-2842, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30962294

ABSTRACT

Lymphoid specification is the process by which hematopoietic stem cells (HSCs) and their progeny become restricted to differentiation through the lymphoid lineages. The basic helix-loop-helix transcription factors E2A and Lyl1 form a complex that promotes lymphoid specification. In this study, we demonstrate that Tal1, a Lyl1-related basic helix-loop-helix transcription factor that promotes T acute lymphoblastic leukemia and is required for HSC specification, erythropoiesis, and megakaryopoiesis, is a negative regulator of murine lymphoid specification. We demonstrate that Tal1 limits the expression of multiple E2A target genes in HSCs and controls the balance of myeloid versus T lymphocyte differentiation potential in lymphomyeloid-primed progenitors. Our data provide insight into the mechanisms controlling lymphocyte specification and may reveal a basis for the unique functions of Tal1 and Lyl1 in T acute lymphoblastic leukemia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Differentiation/immunology , Lymphoid Progenitor Cells/immunology , Myeloid Progenitor Cells/immunology , Neoplasm Proteins/immunology , T-Cell Acute Lymphocytic Leukemia Protein 1/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics
3.
Sci Immunol ; 3(22)2018 04 27.
Article in English | MEDLINE | ID: mdl-29703840

ABSTRACT

All innate lymphoid cells (ILCs) require the small helix-loop-helix transcription factor ID2, but the functions of ID2 are not well understood in these cells. We show that mature natural killer (NK) cells, the prototypic ILCs, developed in mice lacking ID2 but remained as precursor CD27+CD11b- cells that failed to differentiate into CD27-CD11b+ cytotoxic effectors. We show that ID2 limited chromatin accessibility at E protein binding sites near naïve T lymphocyte-associated genes including multiple chemokine receptors, cytokine receptors, and signaling molecules and altered the NK cell response to inflammatory cytokines. In the absence of ID2, CD27+CD11b- NK cells expressed ID3, a helix-loop-helix protein associated with naïve T cells, and they transitioned from a CD8 memory precursor-like to a naïve-like chromatin accessibility state. We demonstrate that ID3 was required for the development of ID2-deficient NK cells, indicating that completely unfettered E protein function is incompatible with NK cell development. These data solidify the roles of ID2 and ID3 as mediators of effector and naïve gene programs, respectively, and revealed a critical role for ID2 in promoting a chromatin state and transcriptional program in CD27+CD11b- NK cells that supports cytotoxic effector differentiation and cytokine responses.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Differentiation/immunology , Inhibitor of Differentiation Protein 2/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/immunology , Chromatin/metabolism , Gene Expression Regulation/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/immunology , Inhibitor of Differentiation Proteins/metabolism , Killer Cells, Natural/metabolism , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/metabolism
4.
Eur J Immunol ; 47(5): 800-805, 2017 05.
Article in English | MEDLINE | ID: mdl-28276053

ABSTRACT

Group 1 innate lymphoid cells include natural killer (NK) cells and ILC1s, which mediate the response to intracellular pathogens. Thymic NK (tNK) cells were described with hybrid features of immature NK cells and ILC1 but whether these cells are related to NK cells or ILC1 has not been fully investigated. We report that murine tNK cells expressed the NK-cell associated transcription factor EOMES and developed independent of the essential ILC1 factor TBET, confirming their placement within the NK lineage. Moreover, tNK cells resemble NK cells rather than ILC1 in their requirements for the E protein transcription factor inhibitor ID2. We provide further insight into the mechanisms governing tNK-cell development by showing that the transcription factor ETS1 prevented tNK cell acquisition of the conventional NK-cell maturation markers CD11b and KLRG1. Our data reveal few ILC1 in the thymus and clarify the identity and developmental requirements of tNK cells.


Subject(s)
Killer Cells, Natural/physiology , Lymphocytes/physiology , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , CD11b Antigen/genetics , CD11b Antigen/immunology , CD11b Antigen/metabolism , Cell Differentiation , Cell Lineage , Immunity, Innate , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Killer Cells, Natural/immunology , Lectins, C-Type , Lymphocytes/immunology , Mice , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , T-Box Domain Proteins/genetics , Thymocytes/cytology , Thymocytes/physiology , Thymus Gland/cytology , Transcription Factors/genetics
6.
J Immunol ; 194(7): 3191-200, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710912

ABSTRACT

The chemokine receptor CCR9 controls the immigration of multipotent hematopoietic progenitor cells into the thymus to sustain T cell development. Postimmigration, thymocytes downregulate CCR9 and migrate toward the subcapsular zone where they recombine their TCR ß-chain and γ-chain gene loci. CCR9 is subsequently upregulated and participates in the localization of thymocytes during their selection for self-tolerant receptor specificities. Although the dynamic regulation of CCR9 is essential for early T cell development, the mechanisms controlling CCR9 expression have not been determined. In this article, we show that key regulators of T cell development, Notch1 and the E protein transcription factors E2A and HEB, coordinately control the expression of Ccr9. E2A and HEB bind at two putative enhancers upstream of Ccr9 and positively regulate CCR9 expression at multiple stages of T cell development. In contrast, the canonical Notch signaling pathway prevents the recruitment of p300 to the putative Ccr9 enhancers, resulting in decreased acetylation of histone H3 and a failure to recruit RNA polymerase II to the Ccr9 promoter. Although Notch signaling modestly modulates the binding of E proteins to one of the two Ccr9 enhancers, we found that Notch signaling represses Ccr9 in T cell lymphoma lines in which Ccr9 transcription is independent of E protein function. Our data support the hypothesis that activation of Notch1 has a dominant-negative effect on Ccr9 transcription and that Notch1 and E proteins control the dynamic expression of Ccr9 during T cell development.


Subject(s)
Gene Expression Regulation , Lymphoid Progenitor Cells/metabolism , Receptors, CCR/genetics , Receptors, Notch , Signal Transduction , T-Lymphocyte Subsets/metabolism , Transcription, Genetic , Animals , Antigens, Surface/metabolism , Binding Sites , Cell Line , Cell Movement/genetics , Cell Movement/immunology , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Transgenic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocyte Subsets/immunology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
7.
Immunol Rev ; 238(1): 93-109, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20969587

ABSTRACT

Lymphopoiesis generates mature B, T, and NK lymphocytes from hematopoietic stem cells via a series of increasingly restricted developmental intermediates. The transcriptional networks that regulate these fate choices are composed of both common and lineage-specific components, which combine to create a cellular context that informs the developmental response to external signals. E proteins are an important factor during lymphopoiesis, and E2A in particular is required for normal T- and B-cell development. Although the other E proteins, HEB and E2-2, are expressed during lymphopoiesis and can compensate for some of E2A's activity, E2A proteins have non-redundant functions during early T-cell development and at multiple checkpoints throughout B lymphopoiesis. More recently, a role for E2A has been demonstrated in the generation of lymphoid-primed multipotent progenitors and shown to favor their specification toward lymphoid over myeloid lineages. This review summarizes both our current understanding of the wide-ranging functions of E proteins during the development of adaptive lymphocytes and the novel functions of E2A in orchestrating a lymphoid-biased cellular context in early multipotent progenitors.


Subject(s)
B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Gene Expression Regulation, Developmental , Multipotent Stem Cells/immunology , T-Lymphocytes/immunology , Animals , Cell Lineage , GATA3 Transcription Factor/immunology , Gene Expression Regulation, Developmental/immunology , Humans , Lymphoid Progenitor Cells/immunology , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Protein Binding
8.
Proc Natl Acad Sci U S A ; 105(34): 12429-34, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18719121

ABSTRACT

Tumor necrosis factor receptor 1-associated death domain protein (TRADD) is the core adaptor recruited to TNF receptor 1 (TNFR1) upon TNFalpha stimulation. In cells from TRADD-deficient mice, TNFalpha-mediated apoptosis and TNFalpha-stimulated NF-kappaB, JNK, and ERK activation are defective. TRADD is also important for germinal center formation, DR3-mediated costimulation of T cells, and TNFalpha-mediated inflammatory responses in vivo. TRADD deficiency does not enhance IFNgamma-induced signaling. Importantly, TRADD has a novel role in TLR3 and TLR4 signaling. TRADD participates in the TLR4 complex formed upon LPS stimulation, and TRADD-deficient macrophages show impaired cytokine production in response to TLR ligands in vitro. Thus, TRADD is a multifunctional protein crucial both for TNFR1 signaling and other signaling pathways relevant to immune responses.


Subject(s)
Signal Transduction , TNF Receptor-Associated Death Domain Protein/metabolism , Toll-Like Receptor 4/metabolism , Animals , Apoptosis , Germinal Center , Inflammation , Mice , Mice, Knockout , NF-kappa B/metabolism , T-Lymphocytes , TNF Receptor-Associated Death Domain Protein/physiology , Toll-Like Receptor 3/metabolism , Tumor Necrosis Factor-alpha/pharmacology
9.
Methods Mol Biol ; 380: 73-81, 2007.
Article in English | MEDLINE | ID: mdl-17876088

ABSTRACT

Mature hematopoietic cells, like all other terminally differentiated lineages, arise during ontogeny via a series of increasingly restricted intermediates. Hematopoietic progenitors derive from the mesoderm, which gives rise to hemangioblasts that can differentiate into endothelial or endocardial precursors, or hematopoietic stem cells (HSCs). These HSCs, in turn, may either self-renew or differentiate into lineage-restricted progenitors, and ultimately mature effector cells. The ability to generate most hematopoietic lineages in a two-dimensional in vitro environment has facilitated our study of this complex process. Until recently, T lymphocytes were the exception, and appeared to require the specific three-dimensional microenvironment of the thymus to develop. However, here we describe a protocol for the generation of immunocompetent T lymphocytes from embryonic stem cells (ESCs) in vitro, within the two-dimensional microenvironment provided by OP9 bone marrow stromal cells that have been transduced to express the Notch ligand Delta-like-1. This procedure will facilitate further study of T lymphocytes by providing a model system in which the effects of genetic and environmental manipulations of ESC-derived progenitors can be examined, and the mechanisms of tolerance potentially dissected, in vitro.


Subject(s)
Cell Differentiation/immunology , Embryonic Stem Cells/cytology , T-Lymphocytes/cytology , Animals , Cell Culture Techniques/methods , Coculture Techniques/methods , Embryonic Stem Cells/immunology , Flow Cytometry/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , T-Lymphocytes/immunology
10.
Methods Mol Biol ; 330: 113-21, 2006.
Article in English | MEDLINE | ID: mdl-16846020

ABSTRACT

Mature hematopoietic cells, like all other terminally differentiated lineages, arise during ontogeny via a series of increasingly restricted intermediates. Hematopoietic progenitors have their origin in the mesoderm, which gives rise to hemangioblasts that can differentiate into endothelial or endocardial precursors or hematopoietic stem cells. These hematopoietic stem cells in turn may either self-renew or differentiate into lineage-restricted progenitors and ultimately mature effector cells. The ability to generate most hematopoietic lineages in a two-dimensional environment in vitro has facilitated our study of this complex process. Until recently, the T-lymphocyte lineage was the exception and appeared to require the specialized three-dimensional microenvironment of the thymus to develop. However, here we describe a protocol for the generation of T lymphocytes from embryonic stem cells in vitro, within a two-dimensional microenvironment, provided by OP9 bone marrow stromal cells. This procedure will facilitate further study of early T lymphopoiesis by providing a simple model system in which the effects of genetic and environmental manipulations of embryonic stem cell-derived progenitors can be examined without requiring other more complex in vivo or in vitro experimental approaches.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/physiology , T-Lymphocytes/physiology , Animals , Cell Lineage , Coculture Techniques/methods , Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred Strains , T-Lymphocytes/cytology
11.
J Immunol ; 176(9): 5267-75, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16621992

ABSTRACT

The bone marrow and thymus, although both hemopoietic environments, induce very distinct differentiation outcomes. The former supports hemopoietic stem cell self-renewal and multiple hemopoietic lineages, while the latter supports T lymphopoiesis almost exclusively. This distinction suggests that the thymic environment acts to restrict the hemopoietic fates available to thymic immigrants. In this study, we demonstrate that the addition of the Notch ligand Delta-like-1 (Dll-1) to an in vitro system that otherwise supports myelopoiesis, greatly reduces the myelopoietic potential of stem cells or uncommitted progenitors. In contrast, committed myeloid progenitors mature regardless of the presence of Dll-1. The block in myelopoiesis is the direct result of Notch signaling within the hemopoietic progenitor, and Dll-1-induced signals cause a rapid increase in the expression of the zinc finger transcription factor GATA-2. Importantly, in the absence of GATA-2, Dll-1-induced signals fail to inhibit commitment to the myeloid fate. Taken together, our results support a role for GATA-2 in allowing Dll-1 to restrict non-T cell lineage differentiation outcomes.


Subject(s)
GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Myelopoiesis , Receptors, Notch/metabolism , Cells, Cultured , Gene Expression Regulation , Signal Transduction , Transcription Factors/metabolism
12.
Methods Mol Biol ; 290: 135-47, 2005.
Article in English | MEDLINE | ID: mdl-15361660

ABSTRACT

Lymphocytes arise during ontogeny via a series of increasingly restricted intermediates. Initially, the mesoderm gives rise to hemangioblasts, which can differentiate into endothelial precursors, or hematopoietic stem cells (HSCs). HSCs can either self-renew or differentiate into lineage-restricted progenitors and, ultimately, to mature effector cells. This complex process is only beginning to be understood, and the ability to generate lymphocytes from embryonic stem (ES) cells in vitro will facilitate further study by providing a model system in which the effects of genetic and environmental manipulations of ES-cell-derived progenitors can be examined. In this protocol, we describe procedures for generating either B- and NK- or T-lymphocytes from mouse ES cells in vitro.


Subject(s)
Embryo, Mammalian/cytology , Lymphocytes/cytology , Stem Cells/cytology , Animals , Coculture Techniques , Lymphocytes/classification , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
13.
Nat Immunol ; 5(4): 410-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15034575

ABSTRACT

Embryonic stem cells (ESCs) have the potential to serve as a renewable source of transplantable tissue-specific stem cells. However, the molecular cues necessary to direct the differentiation of ESCs toward specific cell lineages remain obscure. Here we report the successful induction of ESC differentiation into mature functional T lymphocytes with a simple in vitro coculture system. The directed differentiation of ESCs into T cells required the engagement of Notch receptors by Delta-like 1 ligand (DL1) expressed on the OP9-DL1 stromal cell line. We found a normal program of T cell differentiation in ESC-OP9-DL1 cell cocultures. ESC-derived T cell progenitors effectively reconstituted the T cell compartment of immunodeficient mice, enabling an effective response to a viral infection. These findings provide a powerful tool for the molecular analysis of T cell development and open new avenues for the development of immunotherapeutic approaches using defined sources of stem cells.


Subject(s)
Cell Differentiation/physiology , Stem Cells/physiology , T-Lymphocytes/physiology , Animals , Cell Differentiation/immunology , Coculture Techniques , Gene Expression Profiling , Mice , Stem Cells/immunology , T-Lymphocytes/immunology
14.
Blood ; 102(5): 1649-53, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12738664

ABSTRACT

Embryonic stem (ES) cells can differentiate into most blood cells in vitro, providing a powerful model system to study hematopoiesis. However, ES cell-derived T lymphocytes have not been generated in vitro, and it was unresolved whether such potential is absent or merely difficult to isolate. Because the latter case might result from rapid commitment to non-T-cell fates, we isolated ES cell-derived prehematopoietic precursors for reconstitution of fetal thymic organ cultures. We found a transient Flk1+CD45- subset of these precursors generated T lymphocytes in vitro, and the use of reaggregate thymic organ cultures greatly enhanced reconstitution frequency. These findings reveal that ES cells can exhibit in vitro T-cell potential, but this is restricted to early stages of ES cell differentiation. Moreover, the results support the notion that the thymic microenvironment can induce T-cell differentiation from a subset of prehematopoietic progenitors and suggest deficient migration into intact thymi hindered previous attempts to generate T cells in vitro from ES cell-derived progenitors. These findings demonstrate that a defined subset of ES cells has the potential to generate T cells in vitro and could contribute to greater understanding of the molecular events of hematopoietic induction and T-cell lineage commitment.


Subject(s)
Hematopoietic Stem Cells/cytology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Cell Aggregation/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Coculture Techniques , Female , Fetus , Hematopoietic Stem Cells/chemistry , Leukocyte Common Antigens/analysis , Mice , Mice, Congenic , Mice, Inbred C57BL , Organ Culture Techniques , Pregnancy , T-Lymphocytes/chemistry , Thymus Gland/embryology , Vascular Endothelial Growth Factor Receptor-2/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...