Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Transplantation ; 100(8): 1667-74, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27222932

ABSTRACT

BACKGROUND: Current transplant immunosuppression regimens have numerous limitations. Recent evidence suggests histone deacetylase inhibitors (HDACis) may represent a class of drug with immunosuppressive properties. This study compares cyclosporin A (CyA) with the pan-HDACi suberoylanilide hydroxamic acid (SAHA) and a novel HDAC6-specific inhibitor (KA1010) in models of alloreactivity. METHODS: Proliferation and mixed lymphocyte reaction (MLR)-based assays were used to determine the immunosuppressive effect of compounds, and a murine model of allogeneic skin transplantation was adopted to assess the in vivo effects of HDAC6 inhibition. RESULTS: KA1010 displayed superior inhibitory effects on the activation of peripheral mononuclear cells using in vitro models of transplantation. In a 1-way MLR, KA1010 (5 µΜ) reduced parent cell proliferation from 92% to 64% (P = 0.001). A 2-way MLR, adopting IFN-γ production as a marker of alloresponse, resulting in up to 91% reduction. Dose-response curves revealed dose-dependent profiles with greater potency of HDACis over CyA (IC50 values of 82.0 nM and 13.4 nM for KA1010 and SAHA).Mice treated with KA1010 displayed no significant features of skin allograft rejection upon histological analysis at 70 days and graft survival of 80% in subjects treated with 160 mg/kg. Immunological assessment, revealed a significant increase in CD4CD25forkhead box P3 regulatory T cells (from 18% to 25%, P = 0.0002) and a corresponding reduction in CD4 T cells (from 58% to 42%, P = 0.0009). CONCLUSIONS: HDAC6 may represent an optimal target for future immunosuppressant therapeutics with a particular role in transplantation. In this article, we have demonstrated a superior immunosuppressive effect of KA1010 over both CyA and SAHA, in the models of allotransplantation adopted.


Subject(s)
Aminopyridines/pharmacology , Graft Rejection/prevention & control , Graft Survival/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Immunosuppressive Agents/pharmacology , Leukocytes, Mononuclear/drug effects , Skin Transplantation/adverse effects , Skin/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyclosporine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Graft Rejection/enzymology , Graft Rejection/immunology , Graft Rejection/pathology , Histone Deacetylase 6 , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Targeted Therapy , Signal Transduction/drug effects , Skin/enzymology , Skin/immunology , Skin/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Time Factors , Transplantation, Homologous , Vorinostat
2.
J Immunol ; 195(6): 2675-82, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26254339

ABSTRACT

In the thymus, medullary thymic epithelial cells (mTEC) regulate T cell tolerance via negative selection and Foxp3(+) regulatory T cell (Treg) development, and alterations in the mTEC compartment can lead to tolerance breakdown and autoimmunity. Both the receptor activator for NF-κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) axis and expression of the transcriptional regulator Aire are involved in the regulation of thymus medullary microenvironments. However, their impact on the mechanisms controlling mTEC homeostasis is poorly understood, as are the processes that enable the thymus medulla to support the balanced production of mTEC-dependent Foxp3(+) Treg. In this study, we have investigated the control of mTEC homeostasis and examined how this process impacts the efficacy of Foxp3(+) Treg development. Using newly generated RANK Venus reporter mice, we identify distinct RANK(+) subsets that reside within both the mTEC(hi) and mTEC(lo) compartments and that represent direct targets of OPG-mediated control. Moreover, by mapping OPG expression to a subset of Aire(+) mTEC, our data show how cis- and trans-acting mechanisms are able to control the thymus medulla by operating on multiple mTEC targets. Finally, we show that whereas the increase in mTEC availability in OPG-deficient (Tnfrsf11b(-/-)) mice impacts the intrathymic Foxp3(+) Treg pool by enhancing peripheral Treg recirculation back to the thymus, it does not alter the number of de novo Rag2pGFP(+)Foxp3(+) Treg that are generated. Collectively, our study defines patterns of RANK expression within the thymus medulla, and it shows that mTEC homeostasis is not a rate-limiting step in intrathymic Foxp3(+) Treg production.


Subject(s)
Lymphopoiesis/immunology , Osteoprotegerin/genetics , RANK Ligand/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/metabolism , Animals , Autoimmunity/immunology , Cells, Cultured , DNA-Binding Proteins/genetics , Epithelial Cells , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Immune Tolerance/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , Organ Culture Techniques , Osteoprotegerin/biosynthesis , Osteoprotegerin/immunology , RANK Ligand/biosynthesis , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Thymus Gland/immunology , Transcription Factors/biosynthesis , AIRE Protein
3.
J Immunol ; 193(3): 1204-12, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24990081

ABSTRACT

αßT cell development depends upon serial migration of thymocyte precursors through cortical and medullary microenvironments, enabling specialized stromal cells to provide important signals at specific stages of their development. Although conventional αßT cells are subject to clonal deletion in the medulla, entry into the thymus medulla also fosters αßT cell differentiation. For example, during postnatal periods, the medulla is involved in the intrathymic generation of multiple αßT cell lineages, notably the induction of Foxp3(+) regulatory T cell development and the completion of invariant NKT cell development. Although migration of conventional αßT cells to the medulla is mediated by the chemokine receptor CCR7, how other T cell subsets gain access to medullary areas during their normal development is not clear. In this study, we show that combining a panel of thymocyte maturation markers with cell surface analysis of CCR7 and CCR4 identifies distinct stages in the development of multiple αßT cell lineages in the thymus. Although Aire regulates expression of the CCR4 ligands CCL17 and CCL22, we show that CCR4 is dispensable for thymocyte migration and development in the adult thymus, demonstrating defective T cell development in Aire(-/-) mice is not because of a loss of CCR4-mediated migration. Moreover, we reveal that CCR7 controls the development of invariant NKT cells by enabling their access to IL-15 trans-presentation in the thymic medulla and influences the balance of early and late intrathymic stages of Foxp3(+) regulatory T cell development. Collectively, our data identify novel roles for CCR7 during intrathymic T cell development, highlighting its importance in enabling multiple αßT cell lineages to access the thymic medulla.


Subject(s)
Cell Differentiation/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, CCR4/physiology , Receptors, CCR7/physiology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Adaptive Immunity , Animals , Biomarkers/analysis , Cell Lineage/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR4/deficiency , Receptors, CCR7/deficiency , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology
4.
J Immunol ; 192(6): 2659-66, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24510964

ABSTRACT

In the thymus, interactions with both cortical and medullary microenvironments regulate the development of self-tolerant conventional CD4(+) and CD8(+) αßT cells expressing a wide range of αßTCR specificities. Additionally, the cortex is also required for the development of invariant NKT (iNKT) cells, a specialized subset of T cells that expresses a restricted αßTCR repertoire and is linked to the regulation of innate and adaptive immune responses. Although the role of the cortex in this process is to enable recognition of CD1d molecules expressed by CD4(+)CD8(+) thymocyte precursors, the requirements for additional thymus microenvironments during iNKT cell development are unknown. In this study, we reveal a role for medullary thymic epithelial cells (mTECs) during iNKT cell development in the mouse thymus. This requirement for mTECs correlates with their expression of genes required for IL-15 trans-presentation, and we show that soluble IL-15/IL-15Rα complexes restore iNKT cell development in the absence of mTECs. Furthermore, mTEC development is abnormal in iNKT cell-deficient mice, and early stages in iNKT cell development trigger receptor activator for NF-κB ligand-mediated mTEC development. Collectively, our findings demonstrate that intrathymic iNKT cell development requires stepwise interactions with both the cortex and the medulla, emphasizing the importance of thymus compartmentalization in the generation of both diverse and invariant αßT cells. Moreover, the identification of a novel requirement for iNKT cells in thymus medulla development further highlights the role of both innate and adaptive immune cells in thymus medulla formation.


Subject(s)
Cell Differentiation/immunology , Cellular Microenvironment/immunology , Epithelial Cells/immunology , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cellular Microenvironment/drug effects , Cellular Microenvironment/genetics , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Flow Cytometry , Interleukin-15/administration & dosage , Interleukin-15/genetics , Interleukin-15/immunology , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , RANK Ligand/immunology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Interleukin-15/administration & dosage , Receptors, Interleukin-15/genetics , Receptors, Interleukin-15/immunology , Reverse Transcriptase Polymerase Chain Reaction , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factor RelB/genetics , Transcription Factor RelB/immunology , Transcription Factor RelB/metabolism
5.
Curr Top Microbiol Immunol ; 373: 19-47, 2014.
Article in English | MEDLINE | ID: mdl-23612988

ABSTRACT

The development of CD4(+) helper and CD8(+) cytotoxic T-cells expressing the αß form of the T-cell receptor (αßTCR) takes place in the thymus, a primary lymphoid organ containing distinct cortical and medullary microenvironments. While the cortex represents a site of early T-cell precursor development, and the positive selection of CD4(+)8(+) thymocytes, the thymic medulla plays a key role in tolerance induction, ensuring that thymic emigrants are purged of autoreactive αßTCR specificities. In recent years, advances have been made in understanding the development and function of thymic medullary epithelial cells, most notably the subset defined by expression of the Autoimmune Regulator (Aire) gene. Here, we summarize current knowledge of the developmental mechanisms regulating thymus medulla development, and examine the role of the thymus medulla in recessive (negative selection) and dominant (T-regulatory cell) tolerance.


Subject(s)
Thymus Gland/physiology , Animals , Cell Differentiation , Cell Lineage , Epithelial Cells/physiology , Hematopoietic Stem Cells/cytology , Humans , Immune Tolerance , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology
6.
PLoS One ; 8(9): e74019, 2013.
Article in English | MEDLINE | ID: mdl-24069263

ABSTRACT

Thymocytes expressing the invariant Vγ5 γδT-cell receptor represent progenitors of dendritic epidermal T-cells (DETC) that play an important immune surveillance role in the skin. In contrast to the bulk of αßT-cell development, Vγ5(+) DETC progenitor development occurs exclusively in fetal thymus. Whilst αßT-cell development is known to require chemokine receptor mediated migration through distinct thymus regions, culminating in medullary entry and thymic egress, the importance and control of intrathymic migration for DETC progenitors is unclear. We recently revealed a link between Vγ5(+) DETC progenitor development and medullary thymic epithelial cells expressing Aire, a known regulator of thymic chemokine expression, demonstrating that normal Vγ5(+) DETC progenitor development requires regulated intramedullary positioning. Here we investigate the role of chemokines and their receptors during intrathymic Vγ5(+) DETC progenitor development and establishment of the DETC pool in the skin. We report that thymic medullary accumulation of Vγ5(+) DETC progenitors is a G-protein coupled receptor dependent process. However, this process occurs independently of Aire's influences on intrathymic chemokines, and in the absence of CCR4 and CCR7 expression by DETC progenitors. In contrast, analysis of epidermal γδT-cells at neonatal and adult stages in CCR4(-/-) mice reveals that reduced numbers of DETC in adult epidermis are not a consequence of diminished intrathymic embryonic development, nor deficiencies in initial epidermal seeding in the neonate. Collectively, our data reveal differences in the chemokine receptor requirements for intrathymic migration of αß and invariant γδT-cells, and highlight a differential role for CCR4 in the maintenance, but not initial seeding, of DETC in the epidermis.


Subject(s)
Epidermal Cells , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR4/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Epidermis/immunology , Epidermis/metabolism , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, CCR4/genetics , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/metabolism , AIRE Protein
7.
J Exp Med ; 210(4): 675-81, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23530124

ABSTRACT

A key role of the thymic medulla is to negatively select autoreactive CD4(+) and CD8(+) thymocytes, a process important for T cell tolerance induction. However, the involvement of the thymic medulla in other aspects of αß T cell development, including the generation of Foxp3(+) natural regulatory T cells (nTreg cells) and the continued maturation of positively selected conventional αß T cells, is unclear. We show that newly generated conventional CD69(+)Qa2(-) CD4 single-positive thymocytes mature to the late CD69(-)Qa2(+) stage in the absence of RelB-dependent medullary thymic epithelial cells (mTECs). Furthermore, an increasing ability to continue maturation extrathymically is observed within the CD69(+)CCR7(-/lo)CCR9(+) subset of conventional SP4 thymocytes, providing evidence for an independence from medullary support by the earliest stages after positive selection. In contrast, Foxp3(+) nTreg cell development is medullary dependent, with mTECs fostering the generation of Foxp3(-)CD25(+) nTreg cell precursors at the CD69(+)CCR7(+)CCR9(-) stage. Our results demonstrate a differential requirement for the thymic medulla in relation to CD4 conventional and Foxp3(+) thymocyte lineages, in which an intact mTEC compartment is a prerequisite for Foxp3(+) nTreg cell development through the generation of Foxp3(-)CD25(+) nTreg cell precursors.


Subject(s)
Cell Differentiation/physiology , Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Forkhead Transcription Factors/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, CCR/genetics , Receptors, CCR/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , T-Lymphocytes, Regulatory/cytology , Thymocytes/cytology , Thymus Gland/cytology , Transcription Factor RelB/genetics , Transcription Factor RelB/immunology
8.
Eur J Immunol ; 43(3): 589-94, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23299414

ABSTRACT

In the adult thymus, the development of self-tolerant thymocytes requires interactions with thymic epithelial cells (TECs). Although both cortical and medullary TECs (cTECs/mTECs) are known to arise from common bipotent TEC progenitors, the phenotype of these progenitors and the timing of the emergence of these distinct lineages remain unclear. Here, we have investigated the phenotype and developmental properties of bipotent TEC progenitors during cTEC/mTEC lineage development. We show that TEC progenitors can undergo a stepwise acquisition of first cTEC and then mTEC hallmarks, resulting in the emergence of a progenitor population simultaneously expressing the cTEC marker CD205 and the mTEC regulator Receptor Activator of NF-κB (RANK). In vivo analysis reveals the capacity of CD205(+) TECs to generate functionally competent cortical and medullary microenvironments containing both cTECs and Aire(+) mTECs. Thus, TEC development involves a stage in which bipotent progenitors can co-express hallmarks of the cTEC and mTEC lineages through sequential acquisition, arguing against a simple binary model in which both lineages diverge simultaneously from bipotent lineage negative TEC progenitors. Rather, our data reveal an unexpected overlap in the phenotypic properties of these bipotent TECs with their lineage-restricted counterparts.


Subject(s)
Antigens, CD/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Lineage/immunology , Immunophenotyping , Mice , Minor Histocompatibility Antigens , Receptor Activator of Nuclear Factor-kappa B/metabolism , AIRE Protein
9.
J Immunol ; 189(12): 5519-26, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23152561

ABSTRACT

T cell tolerance in the thymus is a key step in shaping the developing T cell repertoire. Thymic medullary epithelial cells play multiple roles in this process, including negative selection of autoreactive thymocytes, influencing thymic dendritic cell positioning, and the generation of Foxp3(+) regulatory T cells. Previous studies show that medullary thymic epithelial cell (mTEC) development involves hemopoietic cross-talk, and numerous TNFR superfamily members have been implicated in this process. Whereas CD40 and RANK represent key examples, interplay between these receptors, and the individual cell types providing their ligands at both fetal and adult stages of thymus development, remain unclear. In this study, by analysis of the cellular sources of receptor activator for NF-κB ligand (RANKL) and CD40L during fetal and adult cross-talk in the mouse, we show that the innate immune cell system drives initial fetal mTEC development via expression of RANKL, but not CD40L. In contrast, cross-talk involving the adaptive immune system involves both RANKL and CD40L, with analysis of distinct subsets of intrathymic CD4(+) T cells revealing a differential contribution of CD40L by conventional, but not Foxp3(+) regulatory, T cells. We also provide evidence for a stepwise involvement of TNFRs in mTEC development, with CD40 upregulation induced by initial RANK signaling subsequently controlling proliferation within the mTEC compartment. Collectively, our findings show how multiple hemopoietic cell types regulate mTEC development through differential provision of RANKL/CD40L during ontogeny, revealing molecular differences in fetal and adult hemopoietic cross-talk. They also suggest a stepwise process of mTEC development, in which RANK is a master player in controlling the availability of other TNFR family members.


Subject(s)
CD40 Ligand/metabolism , Cellular Senescence/immunology , Gene Expression Regulation, Developmental/immunology , RANK Ligand/biosynthesis , Receptor Cross-Talk/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/genetics , CD40 Ligand/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Senescence/genetics , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Fetus/immunology , Immunity, Innate/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , RANK Ligand/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Thymus Gland/metabolism
10.
Immunity ; 36(3): 427-37, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22425250

ABSTRACT

The thymic medulla provides a specialized microenvironment for the negative selection of T cells, with the presence of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) during the embryonic-neonatal period being both necessary and sufficient to establish long-lasting tolerance. Here we showed that emergence of the first cohorts of Aire(+) mTECs at this key developmental stage, prior to αß T cell repertoire selection, was jointly directed by Rankl(+) lymphoid tissue inducer cells and invariant Vγ5(+) dendritic epidermal T cell (DETC) progenitors that are the first thymocytes to express the products of gene rearrangement. In turn, generation of Aire(+) mTECs then fostered Skint-1-dependent, but Aire-independent, DETC progenitor maturation and the emergence of an invariant DETC repertoire. Hence, our data attributed a functional importance to the temporal development of Vγ5(+) γδ T cells during thymus medulla formation for αß T cell tolerance induction and demonstrated a Rank-mediated reciprocal link between DETC and Aire(+) mTEC maturation.


Subject(s)
Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Transcription Factors/immunology , Animals , Cell Differentiation/immunology , Cellular Microenvironment , Epithelial Cells/immunology , Female , Fetus/cytology , Fetus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Signal Transduction/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transcription Factors/deficiency , Transcription Factors/genetics , AIRE Protein
11.
Blood ; 119(11): 2545-51, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22271449

ABSTRACT

In vivo imaging has revolutionized understanding of the spatiotemporal complexity that subserves the generation of successful effector and regulatory immune responses. Until now, invasive surgery has been required for microscopic access to lymph nodes (LNs), making repeated imaging of the same animal impractical and potentially affecting lymphocyte behavior. To allow longitudinal in vivo imaging, we conceived the novel approach of transplanting LNs into the mouse ear pinna. Transplanted LNs maintain the structural and cellular organization of conventional secondary lymphoid organs. They participate in lymphocyte recirculation and exhibit the capacity to receive and respond to local antigenic challenge. The same LN could be repeatedly imaged through time without the requirement for surgical exposure, and the dynamic behavior of the cells within the transplanted LN could be characterized. Crucially, the use of blood vessels as fiducial markers also allowed precise re-registration of the same regions for longitudinal imaging. Thus, we provide the first demonstration of a method for repeated, noninvasive, in vivo imaging of lymphocyte behavior.


Subject(s)
Diagnostic Imaging , Immune System/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphatic Diseases/pathology , Animals , Antigen Presentation/immunology , Flow Cytometry , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Longitudinal Studies , Lymphatic Diseases/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Photons , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology
12.
PLoS One ; 7(12): e53416, 2012.
Article in English | MEDLINE | ID: mdl-23300927

ABSTRACT

The thymus imparts a developmental imprint upon T cells, screening beneficial and self-tolerant T cell receptor (TCR) specificities. Cortical thymic epithelial cells (CTEC) present self-peptide self-MHC complexes to thymocytes, positively selecting those with functional TCRs. Importantly, CTEC generate diverse self-peptides through highly specific peptide processing. The array of peptides utilized for positive selection appears to play a key role in shaping TCR repertoire and influencing T cell functionality. Whilst self-peptide diversity influences T cell development, the precise source of proteins generating such self-peptide arrays remains unknown, the abundance of apoptotic thymocytes failing thymic selection may provide such a pool of self-proteins. In relation to this notion, whilst it has been previously demonstrated that CTEC expression of the endocytic receptor CD205 facilitates binding and uptake of apoptotic thymocytes, the possible role of CD205 during intrathymic T cell development has not been studied. Here, we directly address the role of CD205 in normal thymocyte development and selection. Through analysis of both polyclonal and monoclonal transgenic TCR T-cell development in the context of CD205 deficiency, we demonstrate that CD205 does not play an overt role in T cell development or selection.


Subject(s)
Antigens, CD/genetics , Lectins, C-Type/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Cell Surface/genetics , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Antigens, CD/metabolism , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Receptors, Antigen, T-Cell/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism
13.
Mech Ageing Dev ; 132(5): 249-56, 2011 May.
Article in English | MEDLINE | ID: mdl-21549744

ABSTRACT

Members of the Wnt family of secreted glyco-lipo-proteins affect intrathymic T-cell development and are abundantly secreted by thymic epithelial cells (TECs) that create the specific microenvironment for thymocytes to develop into mature T-cells. During ageing, Wnt expression declines allowing adipoid involution of the thymic epithelium leading to reduced naïve T-cell output. The protein kinase C (PKC) family of serine-threonine kinases is involved in numerous intracellular biochemical processes, including Wnt signal transduction. In the present study, PKCδ expression is shown to increase with age and to co-localise with Wnt receptors Frizzled (Fz)-4 and -6. It is also demonstrated that connective tissue growth factor (CTGF) is a Wnt-4 target gene and is potentially involved in a negative feed-back loop of Wnt signal regulation. Down-regulation of Wnt-4 expression and activation of multiple repressor pathways suppressing ß-catenin dependent signalling in TECs contribute to the initiation of thymic senescence.


Subject(s)
Cellular Senescence/physiology , Epithelial Cells/metabolism , Signal Transduction/physiology , Thymus Gland/metabolism , Wnt Proteins/metabolism , Animals , Cell Line , Epithelial Cells/cytology , Frizzled Receptors/metabolism , Gene Expression Regulation, Enzymologic/physiology , Humans , Mice , Mice, Inbred BALB C , Protein Kinase C-delta/biosynthesis , Receptors, G-Protein-Coupled/metabolism , T-Lymphocytes/metabolism , Thymus Gland/cytology , beta Catenin/metabolism
14.
Rejuvenation Res ; 14(3): 241-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21453014

ABSTRACT

Glucocorticoids are widely used immunosuppressive drugs in treatment of autoimmune diseases and hematological malignancies. Glucocorticoids are particularly effective immune suppressants, because they induce rapid peripheral T cell and thymocyte apoptosis resulting in impaired T cell-dependent immune responses. Although glucocorticoids can induce apoptotic cell death directly in developing thymocytes, how exogenous glucocorticoids affect the thymic epithelial network that provides the microenvironment for T cell development is still largely unknown. In the present work, we show that primary thymic epithelial cells (TECs) express glucocorticoid receptors and that high-dosage dexamethasone induces degeneration of the thymic epithelium within 24 h of treatment. Changes in organ morphology are accompanied by a decrease in the TEC transcription factor FoxN1 and its regulator Wnt-4 parallel with upregulation of lamina-associated polypeptide 2α and peroxisome proliferator activator receptor γ, two characteristic molecular markers for adipose thymic involution. Overexpression of Wnt-4, however, can prevent upregulation of adipose differentiation-related aging markers, suggesting an important role of Wnt-4 in thymic senescence.


Subject(s)
Cellular Senescence/drug effects , Cytoprotection/drug effects , Dexamethasone/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Thymus Gland/cytology , Wnt4 Protein/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Animals , Cell Line , Cell Transdifferentiation/drug effects , Dexamethasone/administration & dosage , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Mice , Mice, Inbred BALB C , Receptors, Glucocorticoid/metabolism
15.
J Immunol ; 186(9): 5227-35, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21421850

ABSTRACT

The entry of T cell progenitors to the thymus marks the beginning of a multistage developmental process that culminates in the generation of self-MHC-restricted CD4(+) and CD8(+) T cells. Although multiple factors including the chemokine receptors CCR7 and CCR9 are now defined as important mediators of progenitor recruitment and colonization in both the fetal and adult thymi, the heterogeneity of thymus-colonizing cells that contribute to development of the T cell pool is complex and poorly understood. In this study, in conjunction with lineage potential assays, we perform phenotypic and genetic analyses on thymus-settling progenitors (TSP) isolated from the embryonic mouse thymus anlagen and surrounding perithymic mesenchyme, including simultaneous gene expression analysis of 14 hemopoietic regulators using single-cell multiplex RT-PCR. We show that, despite the known importance of CCL25-CCR9 mediated thymic recruitment of T cell progenitors, embryonic PIR(+)c-Kit(+) TSP can be subdivided into CCR9(+) and CCR9(-) subsets that differ in their requirements for a functional thymic microenvironment for thymus homing. Despite these differences, lineage potential studies of purified CCR9(+) and CCR9(-) TSP reveal a common bias toward T cell-committed progenitors, and clonal gene expression analysis reveals a genetic consensus that is evident between and within single CCR9(+) and CCR9(-) TSP. Collectively, our data suggest that although the earliest T cell progenitors may display heterogeneity with regard to their requirements for thymus colonization, they represent a developmentally homogeneous progenitor pool that ensures the efficient generation of the first cohorts of T cells during thymus development.


Subject(s)
Cell Lineage , Gene Expression Profiling , Lymphoid Progenitor Cells/cytology , Lymphopoiesis , Receptors, CCR/metabolism , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Apoptosis/immunology , Cell Differentiation/immunology , Cell Separation , Clone Cells , Embryo, Mammalian , Flow Cytometry , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microdissection , Receptors, CCR/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/embryology
16.
J Immunol ; 185(8): 4769-76, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20861360

ABSTRACT

The thymic medulla represents a key site for the induction of T cell tolerance. In particular, autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) provide a spectrum of tissue-restricted Ags that, through both direct presentation and cross-presentation by dendritic cells, purge the developing T cell repertoire of autoimmune specificities. Despite this role, the mechanisms of Aire(+) mTEC development remain unclear, particularly those stages that occur post-Aire expression and represent mTEC terminal differentiation. In this study, in mouse thymus, we analyze late-stage mTEC development in relation to the timing and requirements for Aire and involucrin expression, the latter a marker of terminally differentiated epithelium including Hassall's corpuscles. We show that Aire expression and terminal differentiation within the mTEC lineage are temporally separable events that are controlled by distinct mechanisms. We find that whereas mature thymocytes are not essential for Aire(+) mTEC development, use of an inducible ZAP70 transgenic mouse line--in which positive selection can be temporally controlled--demonstrates that the emergence of involucrin(+) mTECs critically depends upon the presence of mature single positive thymocytes. Finally, although initial formation of Aire(+) mTECs depends upon RANK signaling, continued mTEC development to the involucrin(+) stage maps to activation of the LTα-LTßR axis by mature thymocytes. Collectively, our results reveal further complexity in the mechanisms regulating thymus medulla development and highlight the role of distinct TNFRs in initial and terminal differentiation stages in mTECs.


Subject(s)
Cell Differentiation/immunology , Epithelial Cells/cytology , Lymphotoxin-alpha/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Animals , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Protein Precursors/immunology , Protein Precursors/metabolism , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Self Tolerance/immunology , T-Lymphocytes/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , AIRE Protein
17.
PLoS One ; 5(5): e10701, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20502698

ABSTRACT

Age-associated thymic involution has considerable physiological impact by inhibiting de novo T-cell selection. This impaired T-cell production leads to weakened immune responses. Yet the molecular mechanisms of thymic stromal adipose involution are not clear. Age-related alterations also occur in the murine thymus providing an excellent model system. In the present work structural and molecular changes of the murine thymic stroma were investigated during aging. We show that thymic epithelial senescence correlates with significant destruction of epithelial network followed by adipose involution. We also show in purified thymic epithelial cells the age-related down-regulation of Wnt4 (and subsequently FoxN1), and the prominent increase in LAP2alpha expression. These senescence-related changes of gene expression are strikingly similar to those observed during mesenchymal to pre-adipocyte differentiation of fibroblast cells suggesting similar molecular background in epithelial cells. For molecular level proof-of-principle stable LAP2alpha and Wnt4-over-expressing thymic epithelial cell lines were established. LAP2alpha over-expression provoked a surge of PPARgamma expression, a transcription factor expressed in pre-adipocytes. In contrast, additional Wnt4 decreased the mRNA level of ADRP, a target gene of PPARgamma. Murine embryonic thymic lobes have also been transfected with LAP2alpha- or Wnt4-encoding lentiviral vectors. As expected LAP2alpha over-expression increased, while additional Wnt4 secretion suppressed PPARgamma expression. Based on these pioneer experiments we propose that decreased Wnt activity and increased LAP2alpha expression provide the molecular basis during thymic senescence. We suggest that these molecular changes trigger thymic epithelial senescence accompanied by adipose involution. This process may either occur directly where epithelium can trans-differentiate into pre-adipocytes; or indirectly where first epithelial to mesenchymal transition (EMT) occurs followed by subsequent pre-adipocyte differentiation. The latter version fits better with literature data and is supported by the observed histological and molecular level changes.


Subject(s)
Cellular Senescence , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Membrane Proteins/metabolism , Thymus Gland/metabolism , Thymus Gland/pathology , Wnt Proteins/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Cell Line , Embryo, Mammalian/metabolism , Epithelium/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Biological , Organ Culture Techniques , Reproducibility of Results , Thymus Gland/embryology , Transfection , Wnt4 Protein
18.
Eur J Immunol ; 40(2): 359-65, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19950181

ABSTRACT

Lymphoid tissue inducer cells (LTi) play an important role in the development of lymphoid tissue in embryos. Adult CD4(+)CD3(-) LTi-like cells present a similar phenotype and gene expression to their embryonic counterpart and have important roles in CD4(+) T-cell memory and lymphoid tissue recovery following viral infection. However, adult LTi-like cells are heterogeneous populations and the factors that regulate their survival and accumulation within secondary lymphoid organs remain unclear, in particular whether the T-zone stroma is involved. Here we report the identification and characterization of a distinct subset of podoplanin(+) murine splenic stromal cells that support adult LTi-like cell survival. We have identified and isolated CD45(-)podoplanin(+) stromal cell populations which have a similar but distinct phenotype to T-zone reticular cells in LN. CD45(-)podoplanin(+) fibroblast-like cells mediate LTi-like cell survival in vitro; surprisingly this was not dependent upon IL-7 as revealed through blocking Ab experiments and studies using LTi-like cells unable to respond to gamma chain cytokines. Our findings show that adult LTi-like cells require extrinsic signals from podoplanin(+) splenic stromal cells to survive and suggest that IL-7 is not necessary to mediate their survival in the adult spleen.


Subject(s)
Interleukin-7/metabolism , Membrane Glycoproteins/metabolism , Stromal Cells/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Cell Survival , Cells, Cultured , Female , Flow Cytometry , Fluorescent Antibody Technique , Interleukin-7/genetics , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Lymphoid Tissue/cytology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Spleen/cytology , Stromal Cells/cytology , T-Lymphocytes, Helper-Inducer/cytology , Time Factors
19.
Eur J Immunol ; 39(9): 2395-402, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19662637

ABSTRACT

Cortical and medullary thymic epithelial cells provide essential signals for a normal programme of T-cell development. Current models of thymus development suggest that thymocyte-derived signals play an important role in establishing thymic microenvironments, a process termed thymus crosstalk. Studies on CD3epsilontg26 mice lacking intrathymic T-cell progenitors provided evidence that normal development of the thymic cortex depends upon thymocyte-derived signals. Importantly, the reported failure to effectively reconstitute adult CD3epsilontg26 mice raised the possibility that such crosstalk must occur within a developmental window, and that closure of this window during the postnatal period renders thymic epithelium refractory to crosstalk signals and unable to effectively impose T-cell selection. We have re-investigated the timing of provision of crosstalk in relation to development of functional thymic microenvironments. We show that transfer of either fetal precursors or adult T-committed precursors into adult CD3epsilontg26 mice initiates key parameters of successful thymic reconstitution including thymocyte development and emigration, restoration of cortical and medullary epithelial architecture, and establishment of thymic tolerance mechanisms including maturation of Foxp3(+) Treg and autoimmune regulator-expressing medullary epithelium. Collectively, our data argue against a temporal window of thymocyte crosstalk, and instead demonstrates continued receptiveness of thymic epithelium for the formation of functionally competent thymic microenvironments.


Subject(s)
Epithelial Cells/immunology , Fetus/immunology , Lymphoid Progenitor Cells/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , CD3 Complex/genetics , Epithelial Cells/metabolism , Hematopoiesis, Extramedullary , Immune Tolerance , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology
20.
Eur J Immunol ; 39(7): 1694-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19582736

ABSTRACT

Thymic epithelial cells (TEC) are essential components of the thymus that guide and control the development and TCR repertoire selection of T cells. Previously, TEC have been considered as postmitotic cells that, once generated during ontogeny, were maintained in their mature state. Recently, it has become clear that TEC can be generated from common or committed medullary and cortical TEC progenitor cells in ontogeny, that stages of immature and mature TEC are phenotypically separable, and that TEC undergo a rapid turnover in a matter of a few weeks. All of these findings strongly suggest that in the adult thymus mature TEC are constantly regenerated from a pool of stem or progenitor cells, a view that renders the thymus structure potentially much more dynamic than previously thought. However, the identity of "thymus stem cells" is elusive, and developmental stages of TEC development are only beginning to be elucidated.


Subject(s)
Epithelial Cells/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Epithelial Cells/cytology , Humans , Models, Biological , Stem Cells/cytology , Stem Cells/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...