Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
1.
Mucosal Immunol ; 8(2): 340-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25138665

ABSTRACT

Innate lymphoid cells (ILC) are RAG-independent lymphocytes with important roles in innate immunity, and include group-1 (natural killer (NK) cell, ILC1), group-2 (ILC2), and group-3 (lymphoid tissue inducer (LTi), NCR(+) ILC3) subsets. Group-3 ILC express Rorγt, produce interleukin (IL)-22, and are critically important in the normal function of mucosal tissues. Here, we describe a novel model cell line for the study of ILC function and differentiation. The parental MNK cell line, derived from NKR-P1B(+) fetal thymocytes, shows a capacity to differentiate in γc cytokines. One IL-7-responsive subline, designated MNK-3, expresses Rorγt and produces high levels of IL-22 in response to IL-23 and IL-1ß stimulation. MNK-3 cells display surface markers and transcript expression characteristic of group-3 ILC, including IL-7Rα (CD127), c-kit (CD117), CCR6, Thy1 (CD90), RANK, RANKL, and lymphotoxin (LTα1ß2). Using an in vitro assay of LTi cell activity, MNK-3 cells induce ICAM-1 and VCAM-1 expression on stromal cells in a manner dependent upon LTα1ß2 expression. A second IL-2-responsive subline, MNK-1, expresses several NK cell receptors, perforin and granzymes, and shows some cytotoxic activity. Thus, MNK-1 cells serve as a model of ILC1/NK development and differentiation, whereas MNK-3 cells provide an attractive in vitro system to study the function of ILC3/LTi cells.


Subject(s)
Cell Differentiation/immunology , Immunity, Innate , Lymphocytes/cytology , Lymphocytes/immunology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cell Lineage , Cluster Analysis , Cytokines/metabolism , Gene Expression Profiling , Gene Expression Regulation , Immunophenotyping , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism
2.
Blood ; 98(13): 3635-42, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739167

ABSTRACT

During ontogeny, the hematopoietic system is established from mesoderm-derived precursors; however, molecular events regulating the onset of hematopoiesis are not well characterized. Several members of the transforming growth factor beta (TGF-beta) superfamily have been implicated as playing a role during mesoderm specification and hematopoiesis. CD105 (endoglin) is an accessory receptor for members of the TGF-beta superfamily. Here it is reported that during the differentiation of murine embryonic stem (ES) cells in vitro, hematopoietic commitment within Flk1(+) mesodermal precursor populations is characterized by CD105 expression. In particular, CD105 is expressed during the progression from the Flk1(+)CD45(-) to Flk1(-)CD45(+) stage. The developmentally regulated expression of CD105 suggests that it may play a role during early hematopoiesis from Flk1(+) precursors. To determine whether CD105 plays a functional role during early hematopoietic development, the potential of CD105-deficient ES cells to differentiate into various hematopoietic lineages in vitro was assessed. In the absence of CD105, myelopoiesis and definitive erythropoiesis were severely impaired. In contrast, lymphopoiesis appeared to be only mildly affected. Thus, these findings suggest that the regulated expression of CD105 functions to support lineage-specific hematopoietic development from Flk1(+) precursors.


Subject(s)
Embryo, Mammalian/cytology , Gene Expression , Hematopoiesis , Hematopoietic Stem Cells/cytology , Mesoderm/cytology , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Growth Factor/analysis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/physiology , Animals , Antigens, CD , Cell Differentiation , Endoglin , Erythropoiesis , Gene Deletion , Gene Expression Regulation, Developmental , Granulocytes , Hematopoietic Stem Cells/chemistry , Leukocyte Common Antigens/analysis , Lymphocytes , Mice , Receptors, Cell Surface , Receptors, Vascular Endothelial Growth Factor
3.
J Immunol ; 167(9): 4966-73, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11673503

ABSTRACT

During thymocyte development, high-affinity/avidity TCR engagement leads to the induction of negative selection and apoptosis, while lower TCR affinity-avidity interactions lead to positive selection and survival. To elucidate how these extracellular interactions are translated into intracellular signals that distinguish between positive and negative selection, we developed a culture system in which naive double-positive thymocytes were either induced to differentiate along the CD8(+) lineage pathway or were triggered for clonal deletion. Using this system, we show that sustained low level activation of extracellular signal-regulated kinases (ERKs) promotes positive selection, whereas strong but transient ERK activation is coupled with negatively selecting stimuli. Importantly, similar ERK activation profiles were demonstrated during positive selection for strong agonist ligands presented at low concentrations or weak agonist ligands. This is consistent with the affinity/avidity model and a role for strong or weak agonists during positive selection. Surprisingly, the addition of a pharmacological inhibitor which blocks ERK activation prevented the induction of negative selection. These data suggest that the duration and strength of the TCR signal is involved in discriminating between positive and negative selection.


Subject(s)
MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases/physiology , T-Lymphocytes/physiology , Animals , Calcium/metabolism , Clonal Deletion , DNA-Binding Proteins/physiology , Enzyme Activation , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/physiology , Receptors, Antigen, T-Cell/physiology , Time Factors
4.
J Exp Med ; 194(6): 757-68, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11560992

ABSTRACT

The dual specificity kinases mitogen-activated protein kinase (MAPK) kinase (MKK)7 and MKK4 are the only molecules known to directly activate the stress kinases stress-activated protein kinases (SAPKs)/c-Jun N-terminal kinases (JNKs) in response to environmental or mitogenic stimuli. To examine the physiological role of MKK7 in hematopoietic cells, we used a gene targeting strategy to mutate MKK7 in murine T and B cells and non-lymphoid mast cells. Loss of MKK7 in thymocytes and mature B cells results in hyperproliferation in response to growth factor and antigen receptor stimulation and increased thymic cellularity. Mutation of mkk7 in mast cells resulted in hyperproliferation in response to the cytokines interleukin (IL)-3 and stem cell factor (SCF). SAPK/JNK activation was completely abolished in the absence of MKK7, even though expression of MKK4 was strongly upregulated in mkk7(-/-) mast cell lines, and phosphorylation of MKK4 occurred normally in response to multiple stress stimuli. Loss of MKK7 did not affect activation of extracellular signal-regulated kinase (ERK)1/2 or p38 MAPK. mkk7(-/-) mast cells display reduced expression of JunB and the cell cycle inhibitor p16INK4a and upregulation of cyclinD1. Reexpression of p16INK4a in mkk7(-/-) mast cells abrogates the hyperproliferative response. Apoptotic responses to a variety of stimuli were not affected. Thus, MKK7 is an essential and specific regulator of stress-induced SAPK/JNK activation in mast cells and MKK7 negatively regulates growth factor and antigen receptor-driven proliferation in hematopoietic cells. These results indicate that the MKK7-regulated stress signaling pathway can function as negative regulator of cell growth in multiple hematopoietic lineages.


Subject(s)
B-Lymphocytes/cytology , MAP Kinase Kinase 4 , Mast Cells/cytology , Mitogen-Activated Protein Kinase Kinases/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Growth Factor/metabolism , T-Lymphocytes/cytology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Division , Enzyme Activation , Gene Targeting , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Interleukin-3/metabolism , Interleukin-3/pharmacology , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 7 , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Thymus Gland/cytology
5.
Blood ; 97(8): 2269-77, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290587

ABSTRACT

Cytokines play an essential role during early T-cell development. However, the mechanisms controlling cytokine signaling in developing thymocytes have not been elucidated. Cytokine receptor signaling can be modulated by suppressor of cytokine signaling-1 (SOCS-1), which acts as a negative regulator of Janus kinases. SOCS-1 is normally expressed throughout thymocyte development; however, retroviral-mediated overexpression of SOCS-1 in fetal liver-derived hematopoietic progenitors prevented their progression beyond the earliest stage of T-cell development. Further analysis revealed that SOCS-1 expression is transiently suppressed following pre-T-cell receptor (TCR) signaling. Moreover, constitutive expression of SOCS-1 abrogated pre-TCR- mediated expansion of immature thymocytes but did not interfere with differentiation. These findings reveal that SOCS-1 serves to regulate cytokine signaling at critical checkpoints during early T-cell development.


Subject(s)
Carrier Proteins/physiology , Repressor Proteins , Signal Transduction , T-Lymphocyte Subsets/metabolism , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Differentiation , Cell Division , Cell Lineage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukin Receptor Common gamma Subunit , Interleukin-7/physiology , Liver/embryology , Lymphocyte Activation , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , Organ Culture Techniques , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta , Receptors, Interleukin-7/physiology , Recombinant Fusion Proteins/physiology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , T-Lymphocyte Subsets/cytology , Thymus Gland/cytology , Thymus Gland/embryology
6.
Nature ; 410(6828): 549-54, 2001 Mar 29.
Article in English | MEDLINE | ID: mdl-11279485

ABSTRACT

Programmed cell death is a fundamental requirement for embryogenesis, organ metamorphosis and tissue homeostasis. In mammals, release of mitochondrial cytochrome c leads to the cytosolic assembly of the apoptosome-a caspase activation complex involving Apaf1 and caspase-9 that induces hallmarks of apoptosis. There are, however, mitochondrially regulated cell death pathways that are independent of Apaf1/caspase-9. We have previously cloned a molecule associated with programmed cell death called apoptosis-inducing factor (AIF). Like cytochrome c, AIF is localized to mitochondria and released in response to death stimuli. Here we show that genetic inactivation of AIF renders embryonic stem cells resistant to cell death after serum deprivation. Moreover, AIF is essential for programmed cell death during cavitation of embryoid bodies-the very first wave of cell death indispensable for mouse morphogenesis. AIF-dependent cell death displays structural features of apoptosis, and can be genetically uncoupled from Apaf1 and caspase-9 expression. Our data provide genetic evidence for a caspase-independent pathway of programmed cell death that controls early morphogenesis.


Subject(s)
Apoptosis/physiology , Flavoproteins/physiology , Membrane Proteins/physiology , Mitochondria/physiology , Animals , Apoptosis Inducing Factor , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspases/metabolism , Cell Differentiation , Chimera , Embryo, Mammalian/cytology , Embryonic and Fetal Development/physiology , Female , Flavoproteins/genetics , Gene Targeting , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Morphogenesis , Proteins/physiology , Recombination, Genetic , Stem Cells
7.
Proc Natl Acad Sci U S A ; 98(2): 609-14, 2001 Jan 16.
Article in English | MEDLINE | ID: mdl-11149941

ABSTRACT

Pre-T cell receptor (preTCR)-derived signals mediate the transition of thymocytes from the CD4(-) CD8(-) double-negative (DN) to CD4(+) CD8(+) double-positive stage of T lymphocyte development. This progression, termed beta-selection, is limited to thymocytes that have generated a functional TCR-beta chain able to associate with pTalpha to form the preTCR complex. Formation of the preTCR complex not only induces differentiation, survival, and proliferation of DN thymocytes; it also inhibits further TCR-beta gene rearrangement through an ill-defined process known as allelic exclusion. The signaling pathways controlling this critical developmental checkpoint have not been characterized. Here we demonstrate that formation of the preTCR complex leads to the activation of protein kinase C (PKC), and that activation of PKC is necessary for the differentiation and expansion of DN thymocytes. Importantly, we also show that allelic exclusion at the TCR-beta gene loci is enforced by PKC-mediated signals. These results define PKC as a central mediator of both differentiation and allelic exclusion during thymocyte development.


Subject(s)
Alleles , Clonal Deletion/physiology , Hematopoiesis/physiology , Isoenzymes/physiology , Protein Kinase C/physiology , T-Lymphocyte Subsets/cytology , Thymus Gland/cytology , Adaptor Proteins, Signal Transducing , Animals , Biolistics , Cell Differentiation , DNA-Binding Proteins , Enzyme Activation , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Genes, Dominant , Genes, Reporter , Isoenzymes/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Mice , Mice, Knockout , Organ Culture Techniques , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoproteins/physiology , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-raf/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection , Type C Phospholipases/physiology , ZAP-70 Protein-Tyrosine Kinase
9.
J Immunol ; 165(10): 5566-72, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11067911

ABSTRACT

During alphabeta T cell development, CD4(-)CD8(-) thymocytes first express pre-TCR (pTalpha/TCR-beta) before their differentiation to the CD4(+)CD8(+) stage. Positive selection of self-tolerant T cells is then determined by the alphabeta TCR expressed on CD4(+)CD8(+) thymocytes. Conceivably, an overlap in surface expression of these two receptors would interfere with the delicate balance of thymic selection. Therefore, a mechanism ensuring the sequential expression of pre-TCR and TCR must function during thymocyte development. In support of this notion, we demonstrate that expression of TCR-alpha by immature thymocytes terminates the surface expression of pre-TCR. Our results reveal that expression of TCR-alpha precludes the formation of pTalpha/TCR-beta dimers within the endoplasmic reticulum, leading to the displacement of pre-TCR from the cell surface. These findings illustrate a novel posttranslational mechanism for the regulation of pre-TCR expression, which may ensure that alphabeta TCR expression on thymocytes undergoing selection is not compromised by the expression of pre-TCR.


Subject(s)
Membrane Glycoproteins/metabolism , Protein Precursors/antagonists & inhibitors , Protein Precursors/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/antagonists & inhibitors , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , T-Lymphocyte Subsets/metabolism , Animals , Binding, Competitive/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Dimerization , Disulfides/metabolism , Gene Expression Regulation/immunology , Genes, T-Cell Receptor alpha/physiology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , Protein Precursors/metabolism , Protein Processing, Post-Translational/immunology , RNA, Messenger/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocyte Subsets/cytology , Tumor Cells, Cultured
11.
J Immunol ; 164(4): 1730-3, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657617

ABSTRACT

We recently described a population of fetal thymocytes with a CD117+NK1.1+CD90lowCD25- phenotype, which were shown to contain committed T cell and NK cell progenitors. However, the characterization of a single cell with a restricted T and NK cell precursor potential was lacking. Here, using an in vitro model for T and NK cell differentiation, we provide conclusive evidence demonstrating the existence of a clonal lineage-restricted T and NK cell progenitor. These results establish that fetal thymocytes with a CD117+NK1.1+CD90lowCD25- phenotype represent bipotent T and NK cell progenitors.


Subject(s)
Killer Cells, Natural/immunology , Proteins , Stem Cells/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/embryology , Thymus Gland/immunology , Animals , Antigens/biosynthesis , Antigens, Ly , Antigens, Surface , Cell Lineage/immunology , Clone Cells , Colony-Forming Units Assay , Immunophenotyping , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lectins, C-Type , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B , Organ Culture Techniques , Protein Biosynthesis , Proto-Oncogene Proteins c-kit/biosynthesis , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology
12.
Dev Immunol ; 8(1): 31-45, 2000.
Article in English | MEDLINE | ID: mdl-11293810

ABSTRACT

Information regarding the intracellular signaling processes that occur during the development of T cells has largely been obtained with the use of transgenic mouse models, which although providing invaluable information are time consuming and costly. To this end, we have developed a novel system that facilitates the in vivo analysis of signal transduction pathways during T-lymphocyte development. This approach uses reporter-plasmids for the detection of intracellular signals mediated by the mitogen-activated protein kinase or cyclic AMP-dependent protein kinase. Reporter-plasmids are transfected into thymocytes in fetal thymic organ culture by accelerated DNA/particle bombardment (gene gun), and the activation of a signaling pathway is determined in the form of a standard luciferase assay. Importantly, this powerful technique preserves the structural integrity of the thymus, and will provide an invaluable tool to study how thymocytes respond to normal environmental stimuli encountered during differentiation within the thymic milieu. Thus, this method allows for the monitoring of signals that occur in a biological time frame, such as during differentiation, and within the natural environment of differentiating cells.


Subject(s)
T-Lymphocytes/metabolism , Thymus Gland/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins , Genes, Reporter , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/metabolism , Organ Culture Techniques , Signal Transduction , T-Lymphocytes/cytology , Thymus Gland/cytology , Transformation, Genetic
13.
J Exp Med ; 190(11): 1647-56, 1999 Dec 06.
Article in English | MEDLINE | ID: mdl-10587355

ABSTRACT

The first checkpoint in T cell development occurs between the CD4(-)CD8(-) and CD4(+)CD8(+) stages and is associated with formation of the pre-T cell receptor (TCR). The signaling mechanisms that drive this progression remain largely unknown. Here, we show that extracellular signal-regulated kinases (ERKs)-1/2 are activated upon engagement of the pre-TCR. Using a novel experimental system, we demonstrate that expression of the pre-TCR by developing thymocytes induces ERK-1/2 activation within the thymus. In addition, the activation of this pre-TCR signaling cascade is mediated through Lck. These findings directly link pre-TCR complex formation with specific downstream signaling components in vivo.


Subject(s)
DNA-Binding Proteins/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Enzyme Activation , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Mice , Mice, Knockout , Mice, SCID , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Receptor-CD3 Complex, Antigen, T-Cell/analysis , Signal Transduction , T-Lymphocyte Subsets/immunology , Thymoma/genetics , Thymoma/immunology , Thymus Gland/immunology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology , Transfection , Tumor Cells, Cultured
14.
Proc Natl Acad Sci U S A ; 96(17): 9797-802, 1999 Aug 17.
Article in English | MEDLINE | ID: mdl-10449774

ABSTRACT

To study molecular events involved in B lymphocyte development and V(D)J rearrangement, we have established an efficient system for the differentiation of embryonic stem (ES) cells into mature Ig-secreting B lymphocytes. Here, we show that B lineage cells generated in vitro from ES cells are functionally analogous to normal fetal liver-derived or bone marrow-derived B lineage cells at three important developmental stages: first, they respond to Flt-3 ligand during an early lymphopoietic progenitor stage; second, they become targets for Abelson murine leukemia virus (A-MuLV) infection at a pre-B cell stage; third, they secrete Ig upon stimulation with lipopolysaccharide at a mature mitogen-responsive stage. Moreover, the ES cell-derived A-MuLV-transformed pre-B (EAB) cells are phenotypically and functionally indistinguishable from standard A-MuLV-transformed pre-B cells derived from infection of mouse fetal liver or bone marrow. Notably, EAB cells possess functional V(D)J recombinase activity. In particular, the generation of A-MuLV transformants from ES cells will provide an advantageous system to investigate genetic modifications that will help to elucidate molecular mechanisms in V(D)J recombination and in A-MuLV-mediated transformation.


Subject(s)
B-Lymphocytes/physiology , Gene Rearrangement, B-Lymphocyte , Hematopoietic Stem Cells/physiology , Abelson murine leukemia virus , Animals , Cell Differentiation/drug effects , Coculture Techniques , Hematopoiesis , Hematopoietic Stem Cells/drug effects , Ligands , Membrane Proteins/pharmacology , Mice , Polymerase Chain Reaction
15.
J Immunol ; 162(10): 5917-23, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10229828

ABSTRACT

The mouse NK1.1 Ag originally defined as NK cell receptor (NKR)-P1C (CD161) mediates NK cell activation. Here, we show that another member of the mouse CD161 family, NKR-P1B, represents a novel NK1.1 Ag. In contrast to NKR-P1C, which functions as an activating receptor, NKR-P1B inhibits NK cell activation. Association of NKR-P1B with Src homology 2-containing protein tyrosine phosphatase-1 provides a molecular mechanism for this inhibition. The existence of these two NK1.1 Ags with opposite functions suggests a potential role for NKR-P1 molecules, such as those of the Ly-49 gene family, in regulating NK cell function.


Subject(s)
Antigens, Surface/metabolism , Antigens/metabolism , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Lectins, C-Type , Proteins/metabolism , Receptors, Immunologic/metabolism , Amino Acid Sequence , Animals , Antigens/genetics , Antigens, Ly , Antigens, Surface/genetics , Blood Cells/immunology , Fetal Blood/immunology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Models, Immunological , Molecular Sequence Data , Multigene Family , NK Cell Lectin-Like Receptor Subfamily B , Phosphorylation , Protein Binding , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Proteins/genetics , Receptors, Immunologic/genetics , SH2 Domain-Containing Protein Tyrosine Phosphatases , Sequence Homology, Amino Acid , Species Specificity , src Homology Domains
16.
Immunol Rev ; 165: 63-74, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9850852

ABSTRACT

T cells and natural killer (NK) cells are presumed to share a common intrathymic precursor. The development of conventional alpha beta T lymphocytes begins within the early fetal thymus, after the colonization of multipotent CD117+ precursors. Irrevocable commitment to the T lineage is marked by thymus-induced expression of CD25. However, the contribution of the fetal thymus to NK lineage commitment and differentiation remains largely unappreciated. Recently, we demonstrated that the development of functional mouse NK cells occurs first in the fetal thymus. Moreover, the appearance of mature fetal thymic NK cells (NK1.1+/CD117-) is preceded by a thymus-induced developmental stage (NK1.1+/CD117+) that marks lineage commitment of multipotent hematopoietic precursors to the T and NK-cell fates. Commitment to the T/NK bipotent stage is induced by fetal thymic stroma, but is not thymus dependent. Recent data indicate that CD90+/CD117lo fetal blood prothymocytes exhibit NK lineage potential and are phenotypically and functionally identical to fetal thymic NK1.1+/CD117+ progenitors. This finding also indicates that full commitment of circulating precursors to the T-cell lineage occurs after thymus colonization. In this review, we discuss recent insights into the cellular and molecular events involved in fetal mouse T and NK lineage commitment and differentiation to unipotent progenitors.


Subject(s)
Killer Cells, Natural/cytology , T-Lymphocytes/cytology , Animals , Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells , Humans , Lymphocytes , Mice , Thymus Gland/cytology , Thymus Gland/embryology
17.
J Immunol ; 161(12): 6544-51, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9862680

ABSTRACT

We recently identified a stage in fetal ontogeny (NK1.1+/CD117+) that defines committed progenitors for T and NK lymphocytes. These cells are found in the fetal thymus as early as day 13 of gestation, but are absent in the fetal liver. Nonetheless, multipotent precursors derived from both the fetal thymus and fetal liver are capable of rapidly differentiating to the NK1.1+ stage upon transfer into fetal thymic organ culture (FTOC). This suggests that expression of NK1.1 marks a thymus-induced lineage commitment event. We now report that a subset of the most immature fetal thymocytes (NK1.1-/CD117+) is capable of up-regulating NK1.1 expression spontaneously upon short-term in vitro culture. Interestingly, fetal liver-derived CD117+ precursors remain NK1.1- upon similar culture. Spontaneous up-regulation of NK1.1 surface expression is minimally affected by transcriptional blockade, mitogen-induced activation, or exposure of these cells to exogenous cytokines or stromal cells. These data suggest that induction of NK1.1 expression on cultured thymocytes may be predetermined by exposure to the thymic microenvironment in vivo. Importantly, multipotent CD117+ thymocytes subdivided on the basis of NK1.1 expression after short-term in vitro culture show distinct precursor potential in lymphocyte lineage reconstitution assays. This demonstrates that even the earliest precursor thymocyte population, although phenotypically homogeneous, contains a functionally heterogeneous subset of lineage-committed progenitors. These findings characterize a thymus-induced pathway in the control of lymphocyte lineage commitment to the T and NK cell fates.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens/biosynthesis , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/metabolism , Protein Biosynthesis , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Animals , Antigens/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Ly , Antigens, Surface , Cell Differentiation , Cell Lineage , Cells, Cultured , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/cytology , Lectins, C-Type , Liver/cytology , Liver/embryology , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B , Organ Culture Techniques , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , T-Lymphocyte Subsets/cytology , Thymus Gland/embryology , Transcription, Genetic
18.
Immunity ; 9(2): 187-97, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9729039

ABSTRACT

We recently identified a fetal thymic developmental stage (NK1.1+/CD117(lo)) that characterizes committed T/NK progenitors. We now report the existence of phenotypically and functionally identical T/NK progenitors in mouse fetal blood and spleen but not in fetal liver. These precursors are indistinguishable from previously characterized fetal blood "prothymocytes" (CD90+/CD117(lo)), with the exception that they express NK1.1, lack markers associated with T lineage commitment, maintain a germline TCRbeta locus, and can give rise to both T and NK cells. Moreover, NK1.1+/CD90+/CD117(lo) fetal blood precursors are present in athymic nude mice. These results suggest that the T/NK lineage commitment pathway is thymus-independent. In contrast, full commitment to the alphabeta T lineage does not precede thymus colonization.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/physiology , Animals , B-Lymphocytes/cytology , Bone Marrow Cells/cytology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Coculture Techniques , Female , Fetal Blood/cytology , Gene Expression , Killer Cells, Natural/cytology , Killer Cells, Natural/physiology , Mice , Mice, Nude , Organ Culture Techniques , Polymerase Chain Reaction , Receptors, Antigen, T-Cell, alpha-beta/genetics , Spleen/cytology , Spleen/physiology , Stromal Cells/cytology , Thymus Gland/cytology
19.
J Immunol ; 160(2): 744-53, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9580246

ABSTRACT

Natural killer (NK) cells mediate MHC-unrestricted cytolysis of virus-infected cells and tumor cells. In the adult mouse, NK cells are bone marrow-derived lymphocytes that mature predominantly in extrathymic locations but have also been suggested to share a common intrathymic progenitor with T lymphocytes. However, mature NK cells are thought to be absent in mouse fetal ontogeny. We report the existence of thymocytes with a mature NK cell phenotype (NK1.1+/CD117-) as early as day 13 of gestation, approximately 3 days before the appearance of CD4+/CD8+ cells in T lymphocyte development. These mature fetal thymic NK cells express genes associated with NK cell effector function and, when freshly isolated, display MHC-unrestricted cytolytic activity in vitro. Moreover, the capacity of fetal thymic NK cells for sustained growth both in vitro and in vivo, in addition to their close phenotypic resemblance to early precursor thymocytes, confounds previous assessments of NK lineage precursor function. Thus, mature NK cells may have been inadvertently included in previous attempts to identify multipotent and bipotent precursor thymocytes. These results provide the first evidence of functional NK lymphocytes in mouse fetal ontogeny and demonstrate that NK cell maturation precedes alpha beta T cell development in the fetal thymus.


Subject(s)
Embryonic and Fetal Development/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Division/immunology , Cell Separation , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Gene Expression Regulation, Developmental/immunology , Immunophenotyping , Killer Cells, Natural/metabolism , Major Histocompatibility Complex/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/immunology , T-Lymphocyte Subsets/immunology
20.
J Immunol ; 160(4): 1735-41, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9469431

ABSTRACT

C4Dlow cells are a population of lymphoid lineage-restricted progenitor cells representing the earliest precursors present in the adult thymus. Paradoxically, thymic progenitors with a similar phenotype in fetal mice and adult RAG-2-deficient (RAG-2-/-) mice lack this characteristic low-level expression of CD4. We now show that radiation-induced differentiation of CD4+ CD8+ double positive thymocytes in RAG-2-/- mice results in the appearance of low levels of CD4 on thymocytes that are phenotypically identical to C4Dlow progenitor cells present in the normal adult thymus. This suggests that CD4 surface expression can be passively transferred from double positive cells to early progenitor thymocytes. Analysis of mixed bone marrow chimeras, reconstituted with hematopoietic stem cells from both CD4-/- (CD45.2) and CD4wt (CD45.1) congenic mice, revealed a CD4low phenotype on cells derived from CD4-/- bone marrow cells. Furthermore, these CD4-/- -derived "C4Dlow" progenitors were capable of reconstituting lymphocyte-depleted fetal thymi, with all thymocytes displaying a CD4-/- phenotype. This directly demonstrates that genetically CD4-deficient thymic progenitor cells can passively acquire a C4Dlow phenotype. Moreover, CD4 expression on C4Dlow progenitor thymocytes is sensitive to mild acid treatment, indicating that CD4 may not exist as an integral cell surface molecule on this thymocyte population. Our findings demonstrate that low-level CD4 surface expression can be passively acquired by intrathymic progenitor cells from the surrounding thymic microenvironment, suggesting that other cell surface molecules expressed at low levels may also result from an acquired phenotype.


Subject(s)
CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/classification , Stem Cells/classification , Stem Cells/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Bone Marrow/immunology , CD4 Antigens/biosynthesis , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera/immunology , Stem Cells/metabolism , Thymus Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL