Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Exp Ther Med ; 16(5): 4277-4282, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30344701

ABSTRACT

Administration of bone marrow-derived mesenchymal stem cells (MSCs) is a possible treatment for graft-versus-host disease (GVHD) following allogeneic hematopoietic stem cell transplantation and other inflammatory conditions. To address the mechanism of immunosuppression by MSCs, in particular those derived from adipose tissue (AMSCs), AMSCs were isolated from three different mouse strains, and the suppressive capacity of the AMSCs thus obtained to suppress interferon (IFN)-γ generation in mixed lymphocyte reaction cultures serving as an in vitro model of GVHD were assessed. It was revealed that the AMSCs had a potent capacity to suppress IFN-γ production regardless of their strain of origin and that such suppression was not associated with production of interleukin-10. In addition, the results demonstrated that ß2-microglobulin (ß2m)-deficient AMSCs from ß2m-/- mice were also potent suppressor cells, verifying the fact that the mechanism underlying the suppression by AMSCs is independent of major histocompatibility complex (MHC) class I expression or MHC compatibility. As AMSCs appear to have immunosuppressive properties, AMSCs may be a useful source of biological suppressor cells for the control of GVHD in humans.

2.
Genes Dev ; 30(22): 2475-2485, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27913604

ABSTRACT

In general, cell fate is determined primarily by transcription factors, followed by epigenetic mechanisms fixing the status. While the importance of transcription factors controlling cell fate has been well characterized, epigenetic regulation of cell fate maintenance remains to be elucidated. Here we provide an obvious fate conversion case, in which the inactivation of polycomb-medicated epigenetic regulation results in conversion of T-lineage progenitors to the B-cell fate. In T-cell-specific Ring1A/B-deficient mice, T-cell development was severely blocked at an immature stage. We found that these developmentally arrested T-cell precursors gave rise to functional B cells upon transfer to immunodeficient mice. We further demonstrated that the arrest was almost completely canceled by additional deletion of Pax5 These results indicate that the maintenance of T-cell fate critically requires epigenetic suppression of the B-lineage gene program.


Subject(s)
B-Lymphocytes/cytology , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/genetics , Gene Silencing , Polycomb-Group Proteins/metabolism , T-Lymphocytes/cytology , Animals , Cell Lineage , Gene Deletion , Gene Expression Regulation, Developmental , Immunoglobulin Heavy Chains/genetics , Mice, Inbred C57BL , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic/genetics , Ubiquitin-Protein Ligases/genetics
3.
Appl Biochem Biotechnol ; 180(8): 1559-1573, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27406037

ABSTRACT

Transplantation of hematopoietic stem and progenitor cells (HSCs) i.e., self-renewing cells that retain multipotentiality, is now a widely performed therapy for many hematopoietic diseases. However, these cells are present in low number and are subject to replicative senescence after extraction; thus, the acquisition of sufficient numbers of cells for transplantation requires donors able to provide repetitive blood samples and/or methods of expanding cell numbers without disturbing cell multipotentiality. Previous studies have shown that HSCs maintain their multipotentiality and self-renewal activity if TCF3 transcription function is blocked under B cell differentiating conditions. Taking advantage of this finding to devise a new approach to HSC expansion in vitro, we constructed an episomal expression vector that specifically targets and transiently represses the TCF3 gene. This consisted of a vector encoding a transcription activator-like effector (TALE) fused to a Krüppel-associated box (KRAB) repressor. We showed that this TALE-KRAB vector repressed expression of an exogenous reporter gene in HEK293 and COS-7 cell lines and, more importantly, efficiently repressed endogenous TCF3 in a human B lymphoma cell line. These findings suggest that this vector can be used to maintain multipotentiality in HSC being subjected to a long-term expansion regimen prior to transplantation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Targeting , Repressor Proteins/metabolism , Transcription Activator-Like Effectors/metabolism , Animals , COS Cells , Chlorocebus aethiops , Gene Deletion , Genes, Reporter , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Luciferases/metabolism , Luminescent Proteins/metabolism , Plasmids/metabolism , Transfection , Red Fluorescent Protein
4.
PLoS Genet ; 12(1): e1005776, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26789017

ABSTRACT

Thymic medullary regions are formed in neonatal mice as islet-like structures, which increase in size over time and eventually fuse a few weeks after birth into a continuous structure. The development of medullary thymic epithelial cells (TEC) is dependent on NF-κB associated signaling though other signaling pathways may contribute. Here, we demonstrate that Stat3-mediated signals determine medullary TEC cellularity, architectural organization and hence the size of the medulla. Deleting Stat3 expression selectively in thymic epithelia precludes the postnatal enlargement of the medulla retaining a neonatal architecture of small separate medullary islets. In contrast, loss of Stat3 expression in cortical TEC neither affects the cellularity or organization of the epithelia. Activation of Stat3 is mainly positioned downstream of EGF-R as its ablation in TEC phenocopies the loss of Stat3 expression in these cells. These results indicate that Stat3 meditated signal via EGF-R is required for the postnatal development of thymic medullary regions.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells , ErbB Receptors/genetics , STAT3 Transcription Factor/biosynthesis , Animals , Embryonic Development , ErbB Receptors/biosynthesis , Flow Cytometry , Gene Expression Regulation, Developmental , Mice , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes/metabolism , Thymocytes/metabolism , Thymus Gland/growth & development , Thymus Gland/metabolism
5.
Stem Cell Reports ; 5(5): 716-727, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26607950

ABSTRACT

Self-renewal potential and multipotency are hallmarks of a stem cell. It is generally accepted that acquisition of such stemness requires rejuvenation of somatic cells through reprogramming of their genetic and epigenetic status.We show here that a simple block of cell differentiation is sufficient to induce and maintain stem cells. By overexpression of the transcriptional inhibitor ID3 in murine hematopoietic progenitor cells and cultivation under B cell induction conditions, the cells undergo developmental arrest and enter a self-renewal cycle. These cells can be maintained in vitro almost indefinitely, and the long-term cultured cells exhibit robust multi-lineage reconstitution when transferred into irradiated mice. These cells can be cloned and re-expanded with 50% plating efficiency, indicating that virtually all cells are self-renewing. Equivalent progenitors were produced from human cord blood stem cells, and these will ultimately be useful as a source of cells for immune cell therapy.


Subject(s)
Cell Cycle Checkpoints , Hematopoietic Stem Cells/cytology , Leukocytes/cytology , Animals , Cell Lineage , Cells, Cultured , Fetal Blood/cytology , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells/cytology , Mice , Mice, Inbred C57BL
6.
Immunol Rev ; 238(1): 23-36, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20969582

ABSTRACT

Most hematology and immunology textbooks describe that the first branch point from the hematopoietic stem cells (HSCs) produces two progenitors, one for myelo-erythroid cells and the other for lymphoid cells including T and B cells. This model is based on the concept that the blood cell family can be subdivided into two major lineages, a myelo-erythroid lineage and a lymphoid lineage. Several alternative models have been proposed during the last three decades. We proposed the myeloid-based model in 2001, in which myeloid potential is retained in an early stage of branches toward erythroid, T-, and B-cell lineages. In this review, we focus on the point that cell differentiation models have two different facets: as a map of developmental potential and a cell fate map. These two are expressed in other words as a map for lineage restriction and a map for physiological production routes. We argue that a map of potential is first and foremost essential for the study of molecular mechanisms of lineage commitment, which is the least clarified aspect of cell differentiation. The validity of the myeloid-based model of hematopoiesis will be discussed in reference to these two issues, developmental potential and cell fate.


Subject(s)
Cell Lineage/immunology , Hematopoiesis/immunology , Models, Immunological , Myeloid Progenitor Cells/immunology , Thymus Gland/embryology , Animals , Antigens, Differentiation/immunology , Humans , T-Lymphocytes/immunology , Thymus Gland/immunology
7.
Science ; 329(5987): 93-6, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20595615

ABSTRACT

In early T cell development, progenitors retaining the potential to generate myeloid and natural killer lineages are eventually determined to a specific T cell lineage. The molecular mechanisms that drive this determination step remain unclarified. We show that, when murine hematopoietic progenitors were cultured on immobilized Notch ligand DLL4 protein in the presence of a cocktail of cytokines including interleukin-7, progenitors developing toward T cells were arrested and the arrested cells entered a self-renewal cycle, maintaining non-T lineage potentials. Reduced concentrations of interleukin-7 promoted T cell lineage determination. A similar arrest and self-renewal of progenitors were observed in thymocytes of mice deficient in the transcription factor Bcl11b. Our study thus identifies the earliest checkpoint during T cell development and shows that it is Bcl11b-dependent.


Subject(s)
Cell Lineage , Hematopoietic Stem Cells/physiology , Lymphopoiesis , Precursor Cells, T-Lymphoid/physiology , Repressor Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , Coculture Techniques , Gene Expression Regulation, Developmental , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hematopoietic Stem Cells/cytology , Interleukin-7/metabolism , Liver/embryology , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology , Repressor Proteins/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics , Up-Regulation
8.
Int Immunol ; 22(2): 65-70, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20053701

ABSTRACT

Blood cells comprise very diverse cell types with a wide range of crucial functions; however, they share a common progenitor cell type-the hematopoietic stem cell (HSC). Clarifying how HSCs differentiate into these diverse cell types is important for understanding how they attain their various functions and offers the potential for therapeutic manipulation. Various theories exist about how HSCs diversify; in particular, one model (the 'classical' model) proposes that lymphocytes and myelo-erythroid lineages branch separately at an early stage of hematopoiesis, whereas another model (the 'myeloid-based' model) proposes that the myeloid potential is retained for much longer among cells that can become lymphocytes. This article describes and compares these models and outlines recent evidence supporting the myeloid-based model.


Subject(s)
Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/physiology , Models, Biological , Myeloid Cells/physiology , Adult Stem Cells/physiology , Animals , Fetal Stem Cells/physiology , Humans , Lymphocytes/physiology
9.
Blood ; 115(2): 261-4, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19828700

ABSTRACT

Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) is a treatment option for patients with hematopoietic malignancies that is hampered by treatment-related morbidity and mortality, in part the result of opportunistic infections, a direct consequence of delayed T-cell recovery. Thymic output can be improved by facilitation of thymic immigration, known to require precommitment of CD34(+) cells. We demonstrate that Delta-like ligand-mediated predifferentiation of mobilized CD34(+) cells in vitro results in a population of thymocyte-like cells arrested at a T/natural killer (NK)-cell progenitor stage. On intrahepatic transfer to Rag2(-/-)gamma(c)(-/-) mice, these cells selectively home to the thymus and differentiate toward surface T-cell receptor-alphabeta(+) mature T cells considerably faster than animals transplanted with noncultured CD34(+) cells. This finding creates the opportunity to develop an early T-cell reconstitution therapy to combine with HSCT.


Subject(s)
Antigens, CD34 , Killer Cells, Natural/metabolism , Lymphoid Progenitor Cells/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Knockout , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transplantation, Heterologous , Transplantation, Homologous
10.
Trends Immunol ; 30(5): 193-200, 2009 May.
Article in English | MEDLINE | ID: mdl-19356980

ABSTRACT

The concept that blood cells arising from hematopoietic stem cells (HSC) can be subdivided into two major lineages, a myelo-erythroid and a lymphoid lineage, has long persisted. Indeed, it has become almost axiomatic that the first branch point from the HSC produces two progenitors, one for myelo-erythroid cells and the other for lymphoid cells. However, recent studies have provided a battery of findings that cannot be explained by this classical model. We will outline how this classical model arose before describing how we came to propose an alternative 'myeloid-based model', in which myeloid potential is retained in erythroid, T, and B cell branches even after these lineages have segregated from each other.


Subject(s)
Cell Lineage/physiology , Hematopoiesis/physiology , Models, Biological , Animals , Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Humans , Lymphoid Progenitor Cells/cytology , Myeloid Progenitor Cells/cytology , Phylogeny
11.
Mol Immunol ; 46(8-9): 1756-67, 2009 May.
Article in English | MEDLINE | ID: mdl-19250680

ABSTRACT

The development and maintenance of thymic microenvironments depends on sustained crosstalk signals derived from developing thymocytes. However, the molecular basis for the initial phase in the lymphoid dependent development of thymic epithelial cells (TECs) remains unclear. Here we show that similarly to regular thymocytes, developing B cells enforced to express the Notch ligand Delta-like-1 (DLL1) efficiently induce the non-polarized, three-dimensional (3D) meshwork architecture of cortical TECs in fetal thymic organ culture. Moreover, the DLL1-overexpressing B cells induce well-developed distinct medullae. Such medullae also arose in lobes reconstituted with Rag2(-/-) thymocytes overexpressing DLL1. Our present findings thus strongly suggest that Notch signaling from thymocytes to TECs induces TEC development at an early phase of thymic organogenesis. The present approach using non-T lineage cells for the in vitro construction of thymic environments may also provide a novel tool for thymus regeneration and T cell production in immunocompromised individuals.


Subject(s)
Epithelial Cells/metabolism , Receptors, Notch/metabolism , Thymus Gland/embryology , Thymus Gland/metabolism , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Cell Lineage/genetics , Cell Lineage/physiology , Cells, Cultured , Embryo, Mammalian , Female , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Organogenesis/genetics , Organogenesis/physiology , Pregnancy , Receptors, Notch/genetics , Receptors, Notch/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Thymus Gland/physiology , Transfection
12.
Eur J Immunol ; 39(1): 96-112, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19089815

ABSTRACT

Previous studies on the MHC class-specific differentiation of CD4(+)CD8(+) thymocytes into CD4(+) and CD8(+) T cells have focused on the role of coreceptor molecules. However, CD4 and CD8 T cells develop according to their MHC class specificities even in these mice lacking coreceptors. This study investigated the possibility that lineage is determined not only by coreceptors, but is also guided by the way how MHC molecules are presented. MHC class II molecules possess a highly conserved Cys in their transmembrane domain, which is palmitoylated and thereby associates with lipid rafts, whereas neither palmitoylation nor raft association was observed with MHC class I molecules. The generation of CD4 T cells was impaired and that of CD8 T cells was augmented when the rafts on the thymic epithelial cells were disrupted. This was due to the conversion of MHC class II-specific thymocytes from the CD4 lineage to CD8. The ability of I-A(d) molecule to associate with rafts was lost when its transmembrane Cys was replaced. The development of DO11.10 thymocytes recognizing this mutant I-A(dm) was converted from CD4 to CD8. These results suggest that the CD4 lineage commitment is directed by the raft-associated presentation of MHC class II molecules.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Lineage/immunology , Histocompatibility Antigens Class II/immunology , Membrane Microdomains/immunology , Thymus Gland/immunology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cholesterol/immunology , Cholesterol/metabolism , Epithelial Cells/immunology , Histocompatibility Antigens Class I/immunology , Lipids/immunology , Lipoylation/immunology , Mice , Molecular Sequence Data , Organ Culture Techniques , Sequence Alignment , Thymus Gland/cytology
13.
Nature ; 452(7188): 768-72, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-18401412

ABSTRACT

During haematopoiesis, pluripotent haematopoietic stem cells are sequentially restricted to give rise to a variety of lineage-committed progenitors. The classical model of haematopoiesis postulates that, in the first step of differentiation, the stem cell generates common myelo-erythroid progenitors and common lymphoid progenitors (CLPs). However, our previous studies in fetal mice showed that myeloid potential persists even as the lineage branches segregate towards T and B cells. We therefore proposed the 'myeloid-based' model of haematopoiesis, in which the stem cell initially generates common myelo-erythroid progenitors and common myelo-lymphoid progenitors. T-cell and B-cell progenitors subsequently arise from common myelo-lymphoid progenitors through myeloid-T and myeloid-B stages, respectively. However, it has been unclear whether this myeloid-based model is also valid for adult haematopoiesis. Here we provide clonal evidence that the early cell populations in the adult thymus contain progenitors that have lost the potential to generate B cells but retain substantial macrophage potential as well as T-cell, natural killer (NK)-cell and dendritic-cell potential. We also show that such T-cell progenitors can give rise to macrophages in the thymic environment in vivo. Our findings argue against the classical dichotomy model in which T cells are derived from CLPs; instead, they support the validity of the myeloid-based model for both adult and fetal haematopoiesis.


Subject(s)
Aging/physiology , Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Myeloid Cells/cytology , T-Lymphocytes/cytology , Animals , B-Lymphocytes/cytology , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Fetus , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/cytology , Macrophages/cytology , Macrophages/metabolism , Mice , Models, Biological , Myeloid Cells/metabolism , Stromal Cells/cytology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/embryology , Thymus Gland/transplantation
14.
Eur J Haematol ; 80(2): 151-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18005390

ABSTRACT

OBJECTIVE: For transplantation of cord blood (CB) cells, it is important to select a CB sample that can reconstitute not only myelo-erythropoiesis but also lymphopoiesis in recipients. However, until now the reconstitution ability of CB samples has been assessed by colony forming unit-culture (CFU-C) assay or by simply counting CD34+ cells. The present study aims at establishing a method capable of assessing the potential of T lymphopoieses of CB samples. METHODS: CD34+ CD38- cells sorted from CB were cultured on a monolayer of murine stromal cell line TSt-4, transduced with the human Delta-like 1 gene. RESULTS: Immature T cells expressing CD5 and/or CD7 were generated in the culture. As these immature T cells can easily be discriminated from mature T cells that are included in the mononuclear cell population (MNCs), we can use the MNCs as starting material for quantification of progenitors capable of generating T cells (TGP). By applying a limiting dilution analysis, we succeeded in determining the frequency of TGP in MNCs. It was found that the ratios for the number of TGP vs. that of CFU-C differ among CB samples maximally by 3.5 times. CONCLUSION: The present assay system provides a novel tool for the evaluation of CB samples, especially for their T-cell-generating potential.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Fetal Blood/cytology , Stem Cells/cytology , T-Lymphocytes/cytology , ADP-ribosyl Cyclase 1/biosynthesis , Animals , Antigens, CD34/biosynthesis , Antigens, CD7/biosynthesis , CD5 Antigens/biosynthesis , Calcium-Binding Proteins , Flow Cytometry , Gene Rearrangement , Humans , Intercellular Signaling Peptides and Proteins/genetics , Leukocytes, Mononuclear/cytology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Stromal Cells/metabolism
15.
J Immunol ; 179(6): 3699-706, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785806

ABSTRACT

Loss of dendritic cell potential is one of the major events in intrathymic T cell development, during which the progenitors become determined to the T cell lineage. However, it remains unclear whether this event occurs in synchrony with another important event, TCRbeta chain gene rearrangement, which has been considered the definitive sign of irreversible T cell lineage commitment. To address this issue, we used transgenic mice in which GFP expression is controlled by the lck proximal promoter. We found that the double-negative (DN) 2 stage can be subdivided into GFP- and GFP+ populations, representing functionally different developmental stages in that the GFP-DN2, but not GFP+DN2, cells retain dendritic cell potential. The GFP+DN2 cells were found to undergo several rounds of proliferation before the initiation of TCRbeta rearrangement as evidenced by the diversity of D-Jbeta rearrangements seen in T cells derived from a single GFP+DN2 progenitor. These results indicated that the determination step of progenitors to the T cell lineage is a separable event from TCRbeta rearrangement.


Subject(s)
Cell Lineage/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cell Differentiation/immunology , Cell Division/genetics , Cell Division/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Down-Regulation/genetics , Down-Regulation/immunology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Trans-Activators/antagonists & inhibitors , Trans-Activators/biosynthesis
16.
EMBO J ; 24(23): 4052-60, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16292344

ABSTRACT

T cells are produced in the thymus from progenitors of extrathymic origin. As no specific markers are available, the developmental pathway of progenitors preceding thymic colonization remains unclear. Here we show that progenitors in murine fetal liver and blood, which are capable of giving rise to T cells, NK cells and dendritic cells, but not B cells, can be isolated by their surface expression of paired immunoglobulin-like receptors (PIR). PIR expression is maintained until the earliest intrathymic stage, then downregulated before the onset of CD25 expression. Unlike intrathymic progenitors, generation of prethymic PIR(+) progenitors does not require Hes1-mediated Notch signaling. These findings disclose a prethymic stage of T-cell development programmed for immigration of the thymus, which is genetically separable from intrathymic stages.


Subject(s)
Cell Differentiation/immunology , Receptors, Immunologic/biosynthesis , Stem Cells/cytology , Stem Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Lineage/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Homeodomain Proteins/physiology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Liver/cytology , Liver/embryology , Liver/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Receptors, Immunologic/genetics , Receptors, Notch/physiology , Signal Transduction/physiology , Stem Cells/metabolism , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Transcription Factor HES-1
17.
J Immunol ; 175(9): 5848-56, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16237077

ABSTRACT

T cell progenitors in the adult thymus (AT) are not well characterized. In the present study, we show that the earliest progenitors in the murine AT are, like those in fetal thymus (FT), unable to generate B or myeloid cells, but still retain the ability to generate NK cells and dendritic cells. However, AT progenitors are distinct from those in FT or fetal liver, in that they are able to produce approximately 100 times larger numbers of T cells than progenitors in fetuses. Such a capability to generate a large number of T cells was mainly attributed to their potential to extensively proliferate before the TCRbeta chain gene rearrangement. We propose that the AT is colonized by T/NK/dendritic cell tripotential progenitors with much higher potential to form diversity in TCRbeta chains than FT progenitors.


Subject(s)
Cell Lineage , Dendritic Cells/cytology , Fetus/cytology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/cytology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , B-Lymphocytes/cytology , Cell Differentiation , Mice , Mice, Inbred C57BL , Thymus Gland/immunology
18.
J Immunol ; 174(5): 2525-32, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15728458

ABSTRACT

It remains controversial whether the thymus-colonizing progenitors are committed to the T cell lineage. A major problem that has impeded the characterization of thymic immigrants has been that the earliest intrathymic progenitors thus far identified do not necessarily represent the genuine thymic immigrants, because their developmental potential should have been influenced by contact with the thymic microenvironment. In the present study, we examined the developmental potential of the ontogenically earliest thymic progenitors of day 11 murine fetus. These cells reside in the surrounding mesenchymal region and have not encountered thymic epithelial components. Flow cytometric and immunohistochemical analyses demonstrated that these cells are exclusively Lin(-)c-kit(+)IL-7R(+). Limiting dilution analyses disclosed that the progenitors with T cell potential were abundant, while those with B cell potential were virtually absent in the region of day 11 thymic anlage. Clonal analyses reveled that they are restricted to T, NK, and dendritic cell lineages. Each progenitor was capable of forming a large number of precursors that may clonally accommodate highly diverse TCRbeta chains. These results provide direct evidence that the progenitors restricted to the T/NK/dendritic cell lineage selectively immigrate into the thymus.


Subject(s)
Cell Movement/immunology , Dendritic Cells/cytology , Hematopoietic Stem Cells/immunology , Killer Cells, Natural/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , Thymus Gland/embryology , Thymus Gland/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Proliferation , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Receptors, Interleukin-7/biosynthesis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/metabolism
19.
Methods Mol Med ; 105: 345-58, 2005.
Article in English | MEDLINE | ID: mdl-15492406

ABSTRACT

It has long been unclear how the pluripotent hematopoietic stem cell is restricted to the major lineage progenitors including the progenitors for myeloid, T- and B-cells. This is the result of the absence of a methodology capable of determining the developmental potential of individual progenitors to generate these major lineage cells. We have established such an assay system, termed the multilineage progenitor assay, as a modification of the fetal thymic organ culture system. By examining cells from murine fetal tissues with this assay, we have succeeded in elucidating the process of lineage restrictions in early hematopoiesis.


Subject(s)
Cell Lineage/physiology , Colony-Forming Units Assay/methods , Embryo, Mammalian/physiology , Myeloid Progenitor Cells/physiology , Thymus Gland/physiology , Animals , Cell Differentiation/physiology , Embryo, Mammalian/cytology , Lymphopoiesis/physiology , Mice , Myelopoiesis/physiology , Thymus Gland/cytology
20.
Blood ; 103(2): 530-7, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14512296

ABSTRACT

During murine fetal development, hemato-poietic progenitors start to colonize the thymic anlage at day 11 of gestation via blood stream. The present study aims at identifying the earliest prethymic progenitors in circulation. Here, we show that the interleukin-7 receptor-positive (IL-7R+) cells in Lin- c-kit+ population are circulating exclusively between days 11 and 14 of fetal age. Clonal analysis revealed that these IL-7R+ cells mostly contain T-cell lineage-restricted progenitors (p-Ts). The proportion of circulating p-Ts reaches 30% of the total p-Ts during these fetal ages, whereas virtually all B-cell lineage-restricted progenitors stay in the fetal liver, suggesting that the p-Ts are selectively released to the circulation. The circulating p-Ts retain the potential to generate natural killer cells and dendritic cells and exhibit extensive proliferation before the occurrence of T-cell receptor beta (TCRbeta) chain gene rearrangement. We propose that the wave of p-Ts in fetal blood disclosed by this study represents the ontogenically earliest thymic immigrants.


Subject(s)
Fetal Blood/cytology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Base Sequence , DNA Primers , Embryonic and Fetal Development , Female , Fetal Blood/drug effects , Fetal Blood/immunology , Gestational Age , Interleukins/pharmacology , Liver/cytology , Liver/embryology , Liver/immunology , Mice , Mice, Inbred C57BL , Models, Animal , Organ Culture Techniques , Placenta , Pregnancy , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Factor/pharmacology , T-Lymphocytes/cytology , Thymus Gland/embryology , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...