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










Publication year range
1.
Cell Rep ; 31(7): 107655, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433964

ABSTRACT

Transcription factors (TFs) play a pivotal role in determining cell states, yet our understanding of the causative relationship between TFs and cell states is limited. Here, we systematically examine the state changes of human pluripotent embryonic stem cells (hESCs) by the large-scale manipulation of single TFs. We establish 2,135 hESC lines, representing three clones each of 714 doxycycline (Dox)-inducible genes including 481 TFs, and obtain 26,998 microscopic cell images and 2,174 transcriptome datasets-RNA sequencing (RNA-seq) or microarrays-48 h after the presence or absence of Dox. Interestingly, the expression of essentially all the genes, including genes located in heterochromatin regions, are perturbed by these TFs. TFs are also characterized by their ability to induce differentiation of hESCs into specific cell lineages. These analyses help to provide a way of classifying TFs and identifying specific sets of TFs for directing hESC differentiation into desired cell types.


Subject(s)
Human Embryonic Stem Cells/metabolism , Transcription Factors/metabolism , Cell Differentiation/physiology , Cell Line , Human Embryonic Stem Cells/cytology , Humans , Single-Cell Analysis/methods
2.
Stem Cell Reports ; 4(3): 445-58, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25733018

ABSTRACT

Mechanisms underlying age-related defects within lymphoid-lineages remain poorly understood. We previously reported that sex steroid ablation (SSA) induced lymphoid rejuvenation and enhanced recovery from hematopoietic stem cell (HSC) transplantation (HSCT). We herein show that, mechanistically, SSA induces hematopoietic and lymphoid recovery by functionally enhancing both HSC self-renewal and propensity for lymphoid differentiation through intrinsic molecular changes. Our transcriptome analysis revealed further hematopoietic support through rejuvenation of the bone marrow (BM) microenvironment, with upregulation of key hematopoietic factors and master regulatory factors associated with aging such as Foxo1. These studies provide important cellular and molecular insights into understanding how SSA-induced regeneration of the hematopoietic compartment can underpin recovery of the immune system following damaging cytoablative treatments. These findings support a short-term strategy for clinical use of SSA to enhance the production of lymphoid cells and HSC engraftment, leading to improved outcomes in adult patients undergoing HSCT and immune depletion in general.


Subject(s)
Cell Differentiation , Gonadal Steroid Hormones/antagonists & inhibitors , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lymphopoiesis/physiology , Regeneration , Animals , Cell Count , Cell Differentiation/genetics , Cell Movement , Cell Self Renewal , Gene Expression Profiling , Gene Expression Regulation, Developmental , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Regeneration/genetics , Stem Cell Niche
3.
J Immunol ; 184(11): 6014-24, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20483779

ABSTRACT

Cytotoxic antineoplastic therapy is used to treat malignant disease but results in long-term immunosuppression in postpubertal and adult individuals, leading to increased incidence and severity of opportunistic infections. We have previously shown that sex steroid ablation (SSA) reverses immunodeficiencies associated with age and hematopoietic stem cell transplantation in both autologous and allogeneic settings. In this study, we have assessed the effects of SSA by surgical castration on T cell recovery of young male mice following cyclophosphamide treatment as a model for the impact of chemotherapy. SSA increased thymic cellularity, involving all of the thymocyte subsets and early T lineage progenitors. It also induced early repair of damage to the thymic stromal microenvironment, which is crucial to the recovery of a fully functional T cell-based immune system. These functional changes in thymic stromal subsets included enhanced production of growth factors and chemokines important for thymopoiesis, which preceded increases in both thymocyte and stromal cellularity. These effects collectively translated to an increase in peripheral and splenic naive T cells. In conclusion, SSA enhances T cell recovery following cyclophosphamide treatment of mice, at the level of the thymocytes and their stromal niches. This provides a new approach to immune reconstitution following antineoplastic therapy.


Subject(s)
Antineoplastic Agents/toxicity , Cyclophosphamide/toxicity , Gonadal Steroid Hormones/immunology , Orchiectomy , T-Lymphocytes/immunology , Animals , Cell Separation , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Steroids , T-Lymphocytes/drug effects , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/immunology
4.
J Immunol ; 180(8): 5384-92, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18390720

ABSTRACT

Medullary thymic epithelial cells (mTEC) play an important and unique role in central tolerance, expressing tissue-restricted Ags (TRA) which delete thymocytes autoreactive to peripheral organs. Since deficiencies in this cell type or activity can lead to devastating autoimmune diseases, it is important to understand the factors which regulate mTEC differentiation and function. Lymphotoxin (LT) ligands and the LTbetaR have been recently shown to be important regulators of mTEC biology; however, the precise role of this pathway in the thymus is not clear. In this study, we have investigated the impact of this signaling pathway in greater detail, focusing not only on mTEC but also on other thymic stromal cell subsets. LTbetaR expression was found in all TEC subsets, but the highest levels were detected in MTS-15(+) thymic fibroblasts. Rather than directing the expression of the autoimmune regulator Aire in mTEC, we found LTbetaR signals were important for TRA expression in a distinct population of mTEC characterized by low levels of MHC class II (mTEC(low)), as well as maintenance of MTS-15(+) fibroblasts. In addition, thymic stromal cell subsets from LT-deficient mice exhibit defects in chemokine production similar to that found in peripheral lymphoid organs of Lta(-/-) and Ltbr(-/-) mice. Thus, we propose a broader role for LTalpha1beta2-LTbetaR signaling in the maintenance of the thymic microenvironments, specifically by regulating TRA and chemokine expression in mTEC(low) for efficient induction of central tolerance.


Subject(s)
Cytokines/metabolism , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/metabolism , Stromal Cells/metabolism , Thymus Gland/metabolism , Transcription Factors/metabolism , Animals , Clonal Deletion/immunology , Cytokines/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression , Immune Tolerance , Lymphotoxin beta Receptor/immunology , Lymphotoxin-alpha/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Stromal Cells/cytology , Stromal Cells/immunology , Thymus Gland/immunology , Transcription Factors/immunology , AIRE Protein
5.
Adv Immunol ; 99: 59-94, 2008.
Article in English | MEDLINE | ID: mdl-19117532

ABSTRACT

Functionally competent immune system includes a functionally competent T-cell repertoire that is reactive to foreign antigens but is tolerant to self-antigens. The repertoire of T cells is primarily formed in the thymus through positive and negative selection of developing thymocytes. Immature thymocytes that undergo V(D)J recombination of T-cell antigen receptor (TCR) genes and that express the virgin repertoire of TCRs are generated in thymic cortex. The recent discovery of thymoproteasomes, a molecular complex specifically expressed in cortical thymic epithelial cells (cTEC), has revealed a unique role of cTEC in cuing the further development of immature thymocytes in thymic cortex, possibly by displaying unique self-peptides that induce positive selection. Cortical thymocytes that receive TCR-mediated positive selection signals are destined to survive for further differentiation and are induced to express CCR7, a chemokine receptor. Being attracted to CCR7 ligands expressed by medullary thymic epithelial cells (mTEC), CCR7-expressing positively selected thymocytes relocate to thymic medulla. The medullary microenvironment displays another set of unique self-peptides for trimming positively selected T-cell repertoire to establish self-tolerance, via promiscuous expression of tissue-specific antigens by mTEC and efficient antigen presentation by dendritic cells. Recent results demonstrate that tumor necrosis factor (TNF) superfamily ligands, including receptor activating NF-kappaB ligand (RANKL), CD40L, and lymphotoxin, are produced by positively selected thymocytes and pivotally regulate mTEC development and thymic medulla formation.


Subject(s)
Dendritic Cells/immunology , Epithelial Cells/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, CCR7/immunology , Self Tolerance/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Clonal Deletion/immunology , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Humans , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR7/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism
6.
J Immunol Methods ; 329(1-2): 56-66, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17988680

ABSTRACT

The microenvironment of the thymus consists of functionally discrete niches composed of distinct stromal cell subsets. Clinically relevant changes affecting T-cell differentiation occur within these niches with age and injury caused by irradiation and chemotherapy treatments. The study of thymic stromal cells has been hampered by the technical difficulty in isolating significant numbers of this important population. Here we present an improved protocol for enzymatic isolation of stromal cells that enables comparative flow cytometric analyses and their purification for downstream cellular or molecular analysis. Fractions analyzed throughout enzymatic digestion of the thymus revealed that various stromal subsets are isolated at characteristic intervals. This highlights the importance of pooling all cells isolated from the thymus for numerical and phenotypic analysis to avoid biased representation of subpopulations. We also describe refined magnetic bead separation techniques that yield almost pure preparations of CD45(-) stroma. Sorting of these suspensions using defined markers enabled purification of the major epithelial subsets, confirmed by keratin staining and PCR analysis. This three-step procedure represents a rapid, reproducible method for the unbiased purification of the stromal cells that direct thymic T-cell differentiation.


Subject(s)
Cell Separation , Epithelial Cells , Flow Cytometry , Stromal Cells , Thymus Gland/cytology , Animals , Epithelial Cells/chemistry , Epithelial Cells/immunology , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Immunomagnetic Separation , Keratins/analysis , Leukocyte Common Antigens/analysis , Mice , Mice, Inbred C57BL , Phenotype , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/chemistry , Stromal Cells/immunology , Temperature , Thymus Gland/chemistry , Thymus Gland/immunology
7.
J Immunol ; 178(8): 4956-65, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404277

ABSTRACT

T cell differentiation in the thymus is dependent upon signals from thymic stromal cells. Most studies into the nature of these signals have focused only on the support provided by the thymic epithelium, but there is an emerging view that other stromal cells such as mesenchymal fibroblasts may also be involved. Study of the latter has been hindered by a lack of appropriate markers, particularly those allowing their isolation. In this study, we describe a new surface marker of thymic stroma, MTS-15, and demonstrate its specificity for fibroblasts and a subset of endothelial cells. Coculture experiments showed that the determinant could be transferred between cells. Extensive biochemical analysis demonstrated that the Ag bound by MTS-15 was the glycosphingolipid Forssman determinant, consistent with the distribution observed. Transcriptional analysis of purified MTS-15(+) thymic fibroblasts revealed a unique expression profile for a number of chemokines and growth factors important to thymocyte and epithelial cell development. In a model of cyclophosphamide-induced thymic involution and regeneration, fibroblasts were found to expand extensively and express growth factors important to epithelial proliferation and increased T cell production just before thymic regeneration. Overall, this study identifies a useful marker of thymic fibroblasts and highlights this subpopulation as a key player in thymic function by virtue of their support of both thymocytes and epithelial cells.


Subject(s)
Antibodies, Monoclonal/immunology , Fibroblasts/chemistry , Stromal Cells/chemistry , Thymus Gland/cytology , Animals , Biomarkers , Cells, Cultured , Gas Chromatography-Mass Spectrometry , Gene Expression Profiling , Immunohistochemistry , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Regeneration , Thymus Gland/physiology , Tissue Distribution
8.
Blood ; 109(9): 3803-11, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17213286

ABSTRACT

The systemic administration of keratinocyte growth factor (KGF) enhances T-cell lymphopoiesis in normal mice and mice that received a bone marrow transplant. KGF exerts protection to thymic stromal cells from cytoablative conditioning and graft-versus-host disease-induced injury. However, little is known regarding KGF's molecular and cellular mechanisms of action on thymic stromal cells. Here, we report that KGF induces in vivo a transient expansion of both mature and immature thymic epithelial cells (TECs) and promotes the differentiation of the latter type of cells. The increased TEC numbers return within 2 weeks to normal values and the microenvironment displays a normal architectural organization. Stromal changes initiate an expansion of immature thymocytes and permit regular T-cell development at an increased rate and for an extended period of time. KGF signaling in TECs activates both the p53 and NF-kappaB pathways and results in the transcription of several target genes necessary for TEC function and T-cell development, including bone morphogenetic protein 2 (BMP2), BMP4, Wnt5b, and Wnt10b. Signaling via the canonical BMP pathway is critical for the KGF effects. Taken together, these data provide new insights into the mechanism(s) of action of exogenous KGF on TEC function and thymopoiesis.


Subject(s)
Cell Proliferation , Epithelial Cells/immunology , Fibroblast Growth Factor 7/immunology , Lymphopoiesis/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Bone Morphogenetic Proteins/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Epithelial Cells/cytology , Female , Fibroblast Growth Factor 7/pharmacology , Lymphopoiesis/drug effects , Mice , NF-kappa B/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology , Tumor Suppressor Protein p53/immunology
9.
Blood ; 108(12): 3777-85, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16896157

ABSTRACT

Despite the importance of thymic stromal cells to T-cell development, relatively little is known about their biology. Here, we use single-cell analysis of stromal cells to analyze extensive changes in the number and composition of thymic stroma throughout life, revealing a surprisingly dynamic population. Phenotypic progression of thymic epithelial subsets was assessed at high resolution in young mice to provide a developmental framework. The cellular and molecular requirements of adult epithelium were studied, using various mutant mice to demonstrate new cross talk checkpoints dependent on RelB in the cortex and CD40 in the medulla. With the use of Ki67 and BrdU labeling, the turnover of thymic epithelium was found to be rapid, but then diminished on thymic involution. The various defects in stromal turnover and composition that accompanied involution were rapidly reversed following sex steroid ablation. Unexpectedly, mature cortical and medullary epithelium showed a potent capacity to stimulate naive T cells, comparable to that of thymic dendritic cells. Overall, these studies show that the thymic stroma is a surprisingly dynamic population and may have a more direct role in negative selection than previously thought.


Subject(s)
Epithelial Cells/physiology , Thymus Gland/physiology , Aging/physiology , Animals , CD40 Antigens/metabolism , Dendritic Cells/cytology , Dendritic Cells/physiology , Epithelial Cells/cytology , Ki-67 Antigen/metabolism , Mice , Mice, Mutant Strains , Stromal Cells/cytology , Stromal Cells/physiology , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Thymus Gland/cytology , Transcription Factor RelB/metabolism
10.
Immunity ; 24(2): 165-77, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16473829

ABSTRACT

Immature CD4+CD8+ thymocytes, which are generated in the thymic cortex, are induced upon positive selection to differentiate into mature T lymphocytes and relocate to the thymic medulla. It was recently shown that a chemokine signal via CCR7 is essential for the cortex-to-medulla migration of positively selected thymocytes in the thymus. However, the role of the cortex-to-medulla migration in T cell development and selection has remained unclear. The present study shows that the developmental kinetics and the thymic export of mature thymocytes were undisturbed in adult mice lacking CCR7 or its ligands (CCR7L). The inhibition of sphingosine-1-phosphate-mediated lymphocyte egress from the thymus led to the accumulation of mature thymocytes in the cortex of CCR7- or CCR7L-deficient mice, unlike the accumulation in the medulla of normal mice, thereby suggesting that mature thymocytes may be exported directly from the cortex in the absence of CCR7 signals. However, the thymocytes that were generated in the absence of CCR7 or CCR7L were potent in causing autoimmune dacryoadenitis and sialadenitis in mice and were thus incapable of establishing central tolerance to organ-specific antigens. These results indicate that CCR7-mediated cortex-to-medulla migration of thymocytes is essential for establishing central tolerance rather than for supporting the maturation or export of thymocytes.


Subject(s)
Autoimmunity , Cell Movement , Chemotaxis, Leukocyte , Immune Tolerance , Receptors, Chemokine/physiology , Thymus Gland/cytology , Animals , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR7 , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Thymus Gland/immunology , Thymus Gland/metabolism
11.
Curr Opin Immunol ; 17(2): 137-43, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15766672

ABSTRACT

T-cell development in the thymus is a stepwise process, mediated by a variety of stromal cells in different regions of the organ. Although the cellular composition of the thymic microenvironment has been known for over a decade, the molecular cues that govern its formation are only beginning to be understood. Stromal-derived chemokines attract T-cell precursors to the thymus and direct maturing thymocytes to appropriate niches for their further development. Reciprocal signals from developing T cells provide crosstalk that is essential for establishment and maintenance of the thymic microenvironment. Elucidation of the molecular players involved and their context within the organ is the challenge for the field today. This knowledge could then be translated to clinical restoration of thymic function and T-cell reconstitution.


Subject(s)
Homeostasis/physiology , Thymus Gland/physiology , Animals , Chemokines/physiology , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/physiology , Gonadal Steroid Hormones/physiology , Growth Hormone/physiology , Humans , Interleukins/physiology , Somatomedins/physiology , Thymus Gland/embryology , Thymus Gland/immunology
12.
J Immunol ; 174(4): 1862-70, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699112

ABSTRACT

Autoimmune regulator (AIRE) gene mutation is responsible for the development of organ-specific autoimmune disease with monogenic autosomal recessive inheritance. Although Aire has been considered to regulate the elimination of autoreactive T cells through transcriptional control of tissue-specific Ags in thymic epithelial cells, other mechanisms of AIRE-dependent tolerance remain to be investigated. We have established Aire-deficient mice and examined the mechanisms underlying the breakdown of self-tolerance. The production and/or function of immunoregulatory T cells were retained in the Aire-deficient mice. The mice developed Sjogren's syndrome-like pathologic changes in the exocrine organs, and this was associated with autoimmunity against a ubiquitous protein, alpha-fodrin. Remarkably, transcriptional expression of alpha-fodrin was retained in the Aire-deficient thymus. These results suggest that Aire regulates the survival of autoreactive T cells beyond transcriptional control of self-protein expression in the thymus, at least against this ubiquitous protein. Rather, Aire may regulate the processing and/or presentation of self-proteins so that the maturing T cells can recognize the self-Ags in a form capable of efficiently triggering autoreactive T cells. With the use of inbred Aire-deficient mouse strains, we also demonstrate the presence of some additional factor(s) that determine the target-organ specificity of the autoimmune disease caused by Aire deficiency.


Subject(s)
Autoantigens/immunology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Animals , Autoantibodies/biosynthesis , Autoantigens/biosynthesis , Carrier Proteins/immunology , Exocrine Glands/immunology , Exocrine Glands/metabolism , Exocrine Glands/pathology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mice, Nude , Microfilament Proteins/immunology , Organ Specificity/genetics , Organ Specificity/immunology , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/pathology , Self Tolerance/genetics , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology , Sjogren's Syndrome/pathology , Species Specificity , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Thymus Gland/pathology , Transcription Factors/physiology , AIRE Protein
13.
Blood ; 105(1): 31-9, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15358618

ABSTRACT

During embryonic development, T-lymphoid precursor cells colonize the thymus. Chemoattraction by the fetal thymus is thought to mediate T-precursor cell colonization. However, the molecules that attract T-precursor cells to the thymus remain unclear. By devising time-lapse visualization in culture, the present results show that alymphoid fetal thymus lobes attract T-precursor cells from fetal liver or fetal blood. CD4(-)CD8(-)CD25(-)CD44+ fetal thymocytes retained the activity to specifically re-enter the thymus. The attraction was predominantly due to I-A-expressing thymic epithelial cells and was mediated by pertussis toxin-sensitive G-protein signals. Among the chemokines produced by the fetal thymus, CCL21, CCL25, and CXCL12 could attract CD4(-)CD8(-)CD25(-)CD44+ fetal thymocytes. However, fetal thymus colonization was markedly diminished by neutralizing antibodies specific for CCL21 and CCL25, but not affected by anti-CXCL12 antibody. Fetal thymus colonization was partially defective in CCL21-deficient plt/plt mice and was further diminished by anti-CCL25 antibody. These results indicate that CCL21 is involved in the recruitment of T-cell precursors to the fetal thymus and suggest that the combination of CCL21 and CCL25 plays a major role in fetal thymus colonization.


Subject(s)
Chemokines, CC/metabolism , Stem Cells/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/embryology , Thymus Gland/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Cells, Cultured , Chemokine CCL21 , Chemokine CXCL12 , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/immunology , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Chemotaxis, Leukocyte , Fetal Blood/cytology , Fetus/embryology , Fetus/immunology , Fetus/metabolism , Histocompatibility Antigens Class II/immunology , Liver/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/immunology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Time Factors
14.
Methods Mol Biol ; 290: 117-33, 2005.
Article in English | MEDLINE | ID: mdl-15361659

ABSTRACT

Fetal thymus organ culture (FTOC) is a unique and powerful culture system that allows intrathymic T-lymphocyte development in vitro. T-cell development in FTOC well represents fetal thymocyte development in vivo. Here, we describe the basic method for FTOC as well as several related techniques, including the reconstitution of thymus lobes with T-lymphoid progenitor cells, high-oxygen submersion culture, time-lapse visualization of thymic emigration, reaggregation culture, and retrovirus-mediated gene transfer to developing thymocytes in FTOC.


Subject(s)
T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Female , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Pregnancy
15.
Dev Dyn ; 231(2): 425-31, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15366020

ABSTRACT

Here, we investigated the expression of the claudin family of tight junction transmembrane proteins in the developing mouse submandibular gland. Data obtained by reverse transcriptase-polymerase chain reaction, Western blot, and immunofluorescence microscopy showed the expression and localization of claudin-3 to -8, -10, and -11 at epithelial tight junctions. Examination of the glands taken from embryonic day (E) 14, E16, and newborn mice revealed differential expression patterns of these claudins in the developing epithelium. Claudin-3, -5, and -7 were expressed in all of the luminal epithelial cells of the ducts at all of the developmental stages examined and in those of terminal tubules at E16 and later. Claudin-4 was expressed mainly in the ducts at all the developmental stages. The expression of claudin-6 and -8 was also restricted to the ducts at E14 and E16; but after birth, the former was undetectable, whereas the latter was expressed in both the ducts and terminal tubules. Claudin-10 and -11 were detectable mainly in the terminal tubules at E16 and later. In addition to being found in the epithelium, claudin-5 was also expressed in certain mesenchymal cells, probably endothelial cells. These results will provide a valuable resource for further investigation of tubulogenesis and physiological regulation of claudin-based tight junctions.


Subject(s)
Membrane Proteins/metabolism , Submandibular Gland/embryology , Submandibular Gland/metabolism , Tight Junctions/metabolism , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Membrane Proteins/genetics , Mice , Submandibular Gland/cytology
16.
J Exp Med ; 200(4): 493-505, 2004 Aug 16.
Article in English | MEDLINE | ID: mdl-15302902

ABSTRACT

Upon TCR-mediated positive selection, developing thymocytes relocate within the thymus from the cortex to the medulla for further differentiation and selection. However, it is unknown how this cortex-medulla migration of thymocytes is controlled and how it controls T cell development. Here we show that in mice deficient for CCR7 or its ligands mature single-positive thymocytes are arrested in the cortex and do not accumulate in the medulla. These mutant mice are defective in forming the medullary region of the thymus. Thymic export of T cells in these mice is compromised during the neonatal period but not in adulthood. Thymocytes in these mice show no defects in maturation, survival, and negative selection to ubiquitous antigens. TCR engagement of immature cortical thymocytes elevates the cell surface expression of CCR7. These results indicate that CCR7 signals are essential for the migration of positively selected thymocytes from the cortex to the medulla. CCR7-dependent cortex-medulla migration of thymocytes plays a crucial role in medulla formation and neonatal T cell export but is not essential for maturation, survival, negative selection, and adult export of thymocytes.


Subject(s)
Cell Differentiation/physiology , Cell Movement/physiology , Receptors, Chemokine/metabolism , Signal Transduction/physiology , Thymus Gland/physiology , Animals , DNA Primers , Eosine Yellowish-(YS) , Flow Cytometry , Fluorescence , Hematoxylin , Mice , Mice, Inbred C57BL , Receptors, CCR7 , Receptors, Chemokine/physiology , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/growth & development
17.
J Immunol ; 172(4): 2067-75, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14764671

ABSTRACT

Physical contact between thymocytes and the thymic stroma is essential for T cell maturation and shapes the T cell repertoire in the periphery. Stromal elements that control these processes still remain elusive. We used a mouse strain with mutant NF-kappaB-inducing kinase (NIK) to examine the mechanisms underlying the breakdown of self-tolerance. This NIK-mutant strain manifests autoimmunity and disorganized thymic structure with abnormal expression of Rel proteins in the stroma. Production of immunoregulatory T cells that control autoreactive T cells was impaired in NIK-mutant mice. The autoimmune disease seen in NIK-mutant mice was reproduced in athymic nude mice by grafting embryonic thymus from NIK-mutant mice, and this was rescued by supply of exogenous immunoregulatory T cells. Impaired production of immunoregulatory T cells by thymic stroma without normal NIK was associated with altered expression of peripheral tissue-restricted Ags, suggesting an essential role of NIK in the thymic microenvironment in the establishment of central tolerance.


Subject(s)
NF-kappa B/biosynthesis , Protein Serine-Threonine Kinases/physiology , Self Tolerance , Thymus Gland/enzymology , Thymus Gland/immunology , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Fetal Tissue Transplantation/immunology , Fetal Tissue Transplantation/pathology , Gene Expression Regulation, Developmental/immunology , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Mutant Strains , Mice, Nude , Mutation , NF-kappa B/physiology , Organ Culture Techniques , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, Interleukin-2/biosynthesis , Self Tolerance/genetics , Stromal Cells/enzymology , Stromal Cells/immunology , Stromal Cells/transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/transplantation , Thymus Gland/embryology , Thymus Gland/transplantation , NF-kappaB-Inducing Kinase
18.
J Immunol ; 171(8): 4113-20, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14530333

ABSTRACT

The arylhydrocarbon receptor nuclear translocator (ARNT) is a member of the basic helix-loop-helix, PER-ARNT-SIM family of heterodimeric transcription factors, and serves as a dimerization partner for arylhydrocarbon receptor (AHR) and hypoxia-inducible factor-1alpha. To assess the function of ARNT in T cells, we disrupted the Arnt gene specifically in T cells of mice by conditional gene targeting using T cell-specific p56(lck)-Cre (Lck-Cre) transgenic Arnt-floxed mice. Thus generated, T cell-specific Arnt-disrupted mice (Lck-Cre;Arnt(flox/Delta) transgenic mice) exhibited complete loss of the expression of ARNT protein only in T cells, and were viable and appeared normal. The Arnt-disrupted T cells in the thymus were phenotypically and histologically normal. The Arnt-deficient T cells in the spleen were capable of responding to TCR stimulation in vitro. However, unlike normal mice in which exposure to the environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an AHR ligand, resulted in thymic involution, the thymus of Lck-Cre;Arnt(flox/Delta) mice were resistant to TCDD treatment in vivo. In contrast, benzo(a)pyrene, another AHR ligand, still caused thymic involution in Lck-Cre;Arnt(flox/Delta) mice. Finally, fetal thymus organ culture using Lck-Cre;Arnt(flox/Delta) and K5-Cre;Arnt(flox/Delta) (epithelial cell-specific Arnt-disrupted mice) showed that thymocytes rather than thymic epithelial cells are predominantly responsible for TCDD-induced thymic atrophy. Our results indicate that ARNT in T lineage cells is essential for TCDD-mediated thymic involution.


Subject(s)
DNA-Binding Proteins , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/drug effects , Thymus Gland/pathology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Atrophy/chemically induced , Atrophy/genetics , Atrophy/immunology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Crosses, Genetic , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Gene Targeting , Immunity, Innate/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Polychlorinated Dibenzodioxins/metabolism , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism
19.
J Immunol ; 171(7): 3394-400, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14500633

ABSTRACT

TCR ligation by the self-peptide-associated MHC molecules is essential for T cell development in the thymus, so that class II MHC-deficient mice do not generate CD4(+)CD8(-) T cells. The present results show that the administration of anti-TCR mAb into class II MHC-deficient mice restores the generation of CD4(+)CD8(-) T cells in vivo. The CD4 T cells were recovered in the thymus, peripheral blood, and the spleen, indicating that the anti-TCR treatment is sufficient for peripheral supply of newly generated CD4 T cells. Unlike peripheral CD4 T cells that disappeared within 5 wk after the treatment, CD4(+)CD8(-) thymocytes remained undiminished even after 5 wk, suggesting that CD4 T cells in the thymus are maintained separately from circulating CD4 T cells and even without class II MHC molecules. It was also found that the mass of medullary region in the thymus, which was reduced in class II MHC-deficient mice, was restored by the anti-TCR administration, suggesting that the medulla for CD4(+)CD8(-) thymocytes is formed independently of the medulla for CD4(-)CD8(+) thymocytes. These results indicate that in vivo anti-TCR treatment in class II MHC-deficient mice restores the generation of circulating CD4 T cells and optimal formation of the medulla in the thymus, suggesting that anti-TCR Ab may be useful for clinical treatment of class II MHC deficiencies.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Genes, T-Cell Receptor beta/immunology , Histocompatibility Antigens Class II/genetics , Lymphocyte Activation/genetics , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Animals, Newborn , CD4 Antigens/biosynthesis , CD4 Antigens/blood , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/blood , CD8 Antigens/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Division/genetics , Cell Division/immunology , Fetus , Histocompatibility Antigens Class II/metabolism , Hybridomas , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/embryology , Thymus Gland/pathology
20.
Immunity ; 16(2): 205-18, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11869682

ABSTRACT

Most T lymphocytes are generated within the thymus. It is unclear, however, how newly generated T cells relocate out of the thymus to the circulation. The present study shows that a CC chemokine CCL19 attracts mature T cells out of the fetal thymus organ culture. Another CC chemokine CCL21, which shares CCR7 with CCL19 but has a unique C-terminal extension containing positively charged amino acids, failed to show involvement in thymic emigration. Neonatal appearance of circulating T cells was defective in CCL19-neutralized mice as well as in CCR7-deficient mice but not in CCL21-neutralized mice. In the thymus, CCL19 is predominantly localized in the medulla including endothelial venules. These results indicate a CCL19- and CCR7-dependent pathway of thymic emigration, which represents a major pathway of neonatal T cell export.


Subject(s)
Chemokines, CC/physiology , Chemotaxis/physiology , Receptors, Chemokine/physiology , T-Lymphocytes/physiology , Thymus Gland/cytology , Animals , Animals, Newborn , Chemokine CCL19 , Chemokine CCL21 , Chemokines, CC/genetics , Gene Expression , Humans , Ligands , Mice , Mice, Knockout , Neutralization Tests , Organ Culture Techniques , Receptors, CCR7 , Receptors, Chemokine/genetics , T-Lymphocytes/cytology , Thymus Gland/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...