Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Immunol ; 11(3): 257-64, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20118929

ABSTRACT

Immature CD4(+)CD8(+) (double-positive (DP)) thymocytes are signaled via T cell antigen receptors (TCRs) to undergo positive selection and become responsive to intrathymic cytokines such as interleukin 7 (IL-7). We report here that cytokine signaling is required for positively selected thymocytes to express the transcription factor Runx3, specify CD8 lineage choice and differentiate into cytotoxic-lineage T cells. In DP thymocytes genetically engineered to be cytokine responsive, IL-7 signaling induced TCR-unsignaled DP thymocytes to express Runx3 and to differentiate into mature CD8(+) T cells, completely circumventing positive selection. We conclude that TCR-mediated positive selection converts DP cells into cytokine-responsive thymocytes, but it is subsequent signaling by intrathymic cytokines that specifies CD8 lineage choice and promotes differentiation into cytotoxic-lineage T cells.


Subject(s)
Cytokines/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Count , Cell Differentiation/immunology , Cell Lineage , Core Binding Factor Alpha 3 Subunit/immunology , Flow Cytometry , Interleukin-7/immunology , Mice , Mice, Knockout , Mice, Transgenic , STAT5 Transcription Factor/immunology , Signal Transduction
2.
J Immunol ; 188(12): 5859-66, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22593613

ABSTRACT

All T cells are dependent on IL-7 for their development and for homeostasis. Foxp3(+) regulatory T cells (Tregs) are unique among T cells in that they are dependent on IL-2. Whether such IL-2 dependency is distinct from or in addition to an IL-7 requirement has been a confounding issue, particularly because of the absence of an adequate experimental system to address this question. In this study, we present a novel in vivo mouse model where IL-2 expression is intact but IL-7 expression was geographically limited to the thymus. Consequently, IL-7 is not available in peripheral tissues. Such mice were generated by introducing a thymocyte-specific IL-7 transgene onto an IL-7 null background. In these mice, T cell development in the thymus, including Foxp3(+) Treg numbers, was completely restored, which correlates with the thymus-specific expression of transgenic IL-7. In peripheral cells, however, IL-7 expression was terminated, which resulted in a general paucity of T cells and a dramatic reduction of Foxp3(+) Treg numbers. Loss of Tregs was further accompanied by a significant reduction in Foxp3(+) expression levels. These data suggest that peripheral IL-7 is not only necessary for Treg survival but also for upregulating Foxp3 expression. Collectively, we assessed the effect of a selective peripheral IL-7 deficiency in the presence of a fully functional thymus, and we document a critical requirement for in vivo IL-7 in T cell maintenance and specifically in Foxp3(+) cell homeostasis.


Subject(s)
Forkhead Transcription Factors/immunology , Homeostasis/immunology , Interleukin-7/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Flow Cytometry , Fluorescent Antibody Technique , Forkhead Transcription Factors/metabolism , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
3.
J Immunol ; 188(10): 4866-75, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22504644

ABSTRACT

Differentiation and maintenance of recirculating effector memory CD8 T cells (T(EM)) depends on prolonged cognate Ag stimulation. Whether similar pathways of differentiation exist for recently identified tissue-resident effector memory T cells (T(RM)), which contribute to rapid local protection upon pathogen re-exposure, is unknown. Memory CD8αß(+) T cells within small intestine epithelium are well-characterized examples of T(RM), and they maintain a long-lived effector-like phenotype that is highly suggestive of persistent Ag stimulation. This study sought to define the sources and requirements for prolonged Ag stimulation in programming this differentiation state, including local stimulation via cognate or cross-reactive Ags derived from pathogens, microbial flora, or dietary proteins. Contrary to expectations, we found that prolonged cognate Ag stimulation was dispensable for intestinal T(RM) ontogeny. In fact, chronic antigenic stimulation skewed differentiation away from the canonical intestinal T cell phenotype. Resident memory signatures, CD69 and CD103, were expressed in many nonlymphoid tissues including intestine, stomach, kidney, reproductive tract, pancreas, brain, heart, and salivary gland and could be driven by cytokines. Moreover, TGF-ß-driven CD103 expression was required for T(RM) maintenance within intestinal epithelium in vivo. Thus, induction and maintenance of long-lived effector-like intestinal T(RM) differed from classic models of T(EM) ontogeny and were programmed through a novel location-dependent pathway that was required for the persistence of local immunological memory.


Subject(s)
Cell Differentiation/immunology , Epitopes, T-Lymphocyte/physiology , Immunologic Memory/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Line , Female , Immunophenotyping , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , Time Factors , Tissue Distribution/immunology
4.
Int Immunol ; 24(10): 661-71, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22899673

ABSTRACT

IL-7 signaling is required for thymocyte development and its loss has a severe deleterious effect on thymus function. Thymocyte-stromal cell interactions and other mechanisms tightly regulate IL-7 expression. We show that disruption of that regulation by over-expression of IL-7 inhibits T-cell development and promotes extensive B-cell lymphopoiesis in the thymus. Our data reveal that high levels of IL-7 negate Notch-1 function in thymocytes found in IL-7 transgenic mice and in co-culture with OP9-DL1 cells. While high levels of IL-7R are present on thymocytes, increased suppressor of cytokine signaling-1 expression blunts IL-7 downstream signaling, resulting in hypo-phosphorylation of proteins in the PI3K-Akt pathway. Consequently, GSK3ß remains active and inhibits Notch-1 signaling as observed by decreased Hes-1 and Deltex expression in thymic progenitors. This is the first demonstration that high levels of IL-7 antagonize Notch-1 signaling and suggest that IL-7 may affect T- versus B-lineage choice in the thymus.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Interleukin-7/immunology , Lymphopoiesis , Receptor, Notch1/metabolism , T-Lymphocytes/cytology , Thymocytes/cytology , Animals , Coculture Techniques , Interleukin-7/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Stromal Cells/cytology , Stromal Cells/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology , Thymus Gland/metabolism
5.
J Exp Med ; 201(10): 1647-57, 2005 May 16.
Article in English | MEDLINE | ID: mdl-15897278

ABSTRACT

Destruction of the host intestinal epithelium by donor effector T cell populations is a hallmark of graft-versus-host disease (GVHD), but the underlying mechanisms remain obscure. We demonstrate that CD8(+) T cells expressing CD103, an integrin conferring specificity for the epithelial ligand E-cadherin, play a critical role in this process. A TCR transgenic GVHD model was used to demonstrate that CD103 is selectively expressed by host-specific CD8(+) T cell effector populations (CD8 effectors) that accumulate in the host intestinal epithelium during GVHD. Although host-specific CD8 effectors infiltrated a wide range of host compartments, only those infiltrating the intestinal epithelium expressed CD103. Host-specific CD8 effectors expressing a TGF-beta dominant negative type II receptor were defective in CD103 expression on entry into the intestinal epithelium, which indicates local TGF-beta activity as a critical regulating factor. Host-specific CD8 effectors deficient in CD103 expression successfully migrated into the host intestinal epithelium but were retained at this site much less efficiently than wild-type host-specific CD8 effectors. The relevance of these events to GVHD pathogenesis is supported by the finding that CD103-deficient CD8(+) T cells were strikingly defective in transferring intestinal GVHD pathology and mortality. Collectively, these data document a pivotal role for TGF-beta-dependent CD103 expression in dictating the gut tropism, and hence the destructive potential, of CD8(+) T cells during GVHD pathogenesis.


Subject(s)
Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Integrin alpha Chains/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/immunology , Animals , Cadherins/immunology , Cell Movement/genetics , Cell Movement/immunology , Graft vs Host Disease/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Transforming Growth Factor beta/immunology
6.
Blood ; 112(8): 3255-63, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18694999

ABSTRACT

Although studies have demonstrated that androgen withdrawal increases thymic size, molecular mechanisms underlying this expansion remain largely unknown. We show that decreased androgen signaling leads to enhanced immigration of bone marrow T-cell precursors, as manifested by both an early increase of early thymic progenitors (ETP) and improved uptake of adoptively transferred quantified precursors into congenic castrated hosts. We provide evidence that the ETP niche is enhanced after androgen withdrawal by proliferation of UEA(+) thymic epithelial cells (TEC) and increased TEC production of CCL25, a ligand critical for ETP entry. Moreover, the greatest increase in CCL25 production is by UEA(+) TEC, linking function of this subset with the increase in ETP immigration. Furthermore, blockade of CCL25 abrogated the effects of castration by impairing ETP entry, retarding immature thymocyte development, limiting increase of thymic size, and impairing increase of thymopoiesis. Taken together, these findings describe a cohesive mechanism underlying increased thymic productivity after androgen withdrawal.


Subject(s)
Androgens/metabolism , Chemokines, CC/metabolism , Thymus Gland/metabolism , Animals , Cell Movement , Cell Proliferation , Cell Separation , Cohort Studies , Epithelial Cells/metabolism , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Models, Biological , Protein Binding , Thymus Gland/cytology
7.
Exp Hematol ; 52: 40-49, 2017 08.
Article in English | MEDLINE | ID: mdl-28552733

ABSTRACT

Impaired immune reconstitution after hematopoietic stem cell transplantation (HSCT) is attributed in part to impaired thymopoiesis. Recent data suggest that precursor input may be a point of regulation for the thymus. We hypothesized that administration of FLT3 ligand (FLT3L) would enhance thymopoiesis after adoptive transfer of aged, FLT3L-treated bone marrow (BM) to aged, Lupron-treated hosts by increasing murine HSC (Lin[minus]Sca1+c-Kit+ [LSK] cells) trafficking and survival. In murine models of aged and young hosts, we show that FLT3L enhances thymopoiesis in aged, Lupron-treated hosts through increased survival and export of LSK cells via CXCR4 regulation. In addition, we elucidate an underlying mechanism of FLT3L action on BM LSK cells-FLT3L drives LSK cells into the stromal niche using Hoescht (Ho) dye perimortem. In summary, we show that FLT3L administration leads to: (1) increased LSK cells and early thymocyte progenitor precursors that can enhance thymopoiesis after transplantation and androgen withdrawal, (2) mobilization of LSK cells through downregulation of CXCR4, (3) enhanced BM stem cell survival associated with Bcl-2 upregulation, and (4) BM stem cell enrichment through increased trafficking to the BM niche. Therefore, we show a mechanism by which FLT3L activity on hematopoeitic and thymic progenitor cells may contribute to thymic recovery. These data have potential clinical relevance to enhance thymic reconstitution after cytoreductive therapy.


Subject(s)
Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Receptors, CXCR4/metabolism , Stem Cell Niche , Thymus Gland/metabolism , Animals , Bone Marrow/metabolism , Cell Survival , Female , Flow Cytometry , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Thymus Gland/cytology
8.
Sci Rep ; 7(1): 4043, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28642474

ABSTRACT

Deuterated water (2H2O) is a label commonly used for safe quantitative measurement of deuterium enrichment into DNA of proliferating cells. More recently, it has been used for labeling proteins and other biomolecules. Our in vitro - in vivo research reports important stable isotopic labeling enrichment differences into the DNA nucleosides and their isotopologues (e.g. deoxyadenosine (dA) M + 1, dA M + 2, dA M + 3), as well as tumor cell proliferation effects for various forms of commercially available stable heavy water (2H2O, H218O, and 2H218O). Using an in vitro mouse thymus tumor cell line, we determined that H218O provides superior DNA labeling enrichment quantitation, as measured by GC-positive chemical ionization (PCI)-MS/MS. In addition, at higher but physiologically relevant doses, both 2H218O and 2H2O down modulated mouse thymus tumor cell proliferation, whereas H218O water had no observable effects on cell proliferation. The in vivo labeling studies, where normal mouse bone marrow cells (i.e. high turnover) were evaluated post labeling, demonstrated DNA enrichments concordant with measurements from the in vitro studies. Our research also reports a headspace-GC-NCI-MS method, which rapidly and quantitatively measures stable heavy water levels in total body water.


Subject(s)
DNA Replication/drug effects , Deuterium Oxide/pharmacology , Isotope Labeling , Animals , Cell Line, Tumor , Cell Proliferation , DNA/chemistry , DNA/genetics , Mass Spectrometry , Mice
9.
Cancer Res ; 64(18): 6524-9, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374963

ABSTRACT

Tumorigenesis in rodents, as well as in humans, has been shown to be a multistep process, with each step reflecting an altered gene product or gene regulatory process leading to autonomy of cell growth. Initial genetic mutations are often associated with dysfunctional growth regulation, as is demonstrated in several transgenic mouse models. These changes are often followed by alterations in tumor suppressor gene function, allowing unchecked cell cycle progression and, by genomic instability, additional genetic mutations responsible for tumor metastasis. Here we show that reduced transforming growth factor-beta signaling in T lymphocytes leads to a rapid expansion of a CD8+ memory T-cell population and a subsequent transformation to leukemia/lymphoma as shown by multiple criteria, including peripheral blood cell counts histology, T-cell receptor monoclonality, and host transferability. Furthermore, spectral karyotype analysis of the tumors shows that the tumors have various chromosomal aberrations. These results suggest that reduced transforming growth factor-beta signaling acts as a primary carcinogenic event, allowing uncontrolled proliferation with consequent accumulation of genetic defects and leukemic transformation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Leukemia, T-Cell/immunology , Lymphoproliferative Disorders/immunology , Transforming Growth Factor beta/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Chromosome Aberrations , Immunologic Memory , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Antigen, T-Cell/immunology , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction
10.
Clin Transl Sci ; 2(4): 279-85, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19750208

ABSTRACT

Thymic epithelial cells (TECs) and dendritic cells are essential for the maintenance of thymopoiesis. Because these stromal elements define the progenitor niche, provide critical survival signals and growth factors, and direct positive and negative selection, detailed study of these populations is necessary to understand important elements for thymic renewal after cytotoxic injury. Study of TEC is currently hindered by lengthy enzymatic separation techniques with decreased viability. We present a new rapid separation technique that yields consistent viable TEC numbers in a quarter of the prior preparation time. Using this new procedure, we identify changes in stroma populations following total body irradiation (TBI). By flow cytometry, we show that TBI significantly depletes UEA+ medullary TEC, while sparing Ly51+ CD45- cells. Further characterization of the Ly51+ subset reveals enrichment of fibroblasts (CD45- Ly51+ MHCII-), while cortical TECs (CD45- Ly51+ MHCII+) were markedly reduced. Dendritic cells (CD11lc+ CD45+) were also decreased following TBI. These data suggest that cytotoxic preparative regimens may impair thymic renewal by reducing critical populations of cortical and medullary TEC, and that such thymic damage can be assessed by this new rapid separation technique, thereby providing a means of assessing optimal conditioning pretransplantfor enhancing thymic-dependent immune reconstitution posttranspiant.


Subject(s)
Stromal Cells/cytology , Thymus Gland/pathology , Animals , Cell Separation , Dendritic Cells/cytology , Endothelial Cells/cytology , Epithelial Cells/cytology , Female , Flow Cytometry/methods , Immunohistochemistry/methods , Leukocyte Common Antigens/biosynthesis , Mice , Microscopy, Fluorescence/methods , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/immunology , Whole-Body Irradiation
11.
Nat Immunol ; 8(10): 1049-59, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17873878

ABSTRACT

T cell immunity requires the long-term survival of T cells that are capable of recognizing self antigens but are not overtly autoreactive. How this balance is achieved remains incompletely understood. Here we identify a homeostatic mechanism that transcriptionally tailors CD8 coreceptor expression in individual CD8+ T cells to the self-specificity of their clonotypic T cell receptor (TCR). 'Coreceptor tuning' results from interplay between cytokine and TCR signals, such that signals from interleukin 7 and other common gamma-chain cytokines transcriptionally increase CD8 expression and thereby promote TCR engagement of self ligands, whereas TCR signals impair common gamma-chain cytokine signaling and thereby decrease CD8 expression. This dynamic interplay induces individual CD8+ T cells to express CD8 in quantities appropriate for the self-specificity of their TCR, promoting the engagement of self ligands, yet avoiding autoreactivity.


Subject(s)
CD8 Antigens/genetics , Interleukin-7/pharmacology , Receptors, Antigen, T-Cell/physiology , Signal Transduction/physiology , Transcription, Genetic , Animals , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Cytokines/pharmacology , Enhancer Elements, Genetic , Homeostasis , Humans , Mice , Up-Regulation
12.
J Immunol ; 176(1): 118-27, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365402

ABSTRACT

Ocular pigment epithelia contribute to immune privilege by suppressing T cell activation and converting T cells into regulatory T regulatory cells (Tregs) that inhibit bystander T cell activation. Iris pigment epithelium (IPE) does so through direct cell-cell contact with naive T cells, and this suppressive contact is via interactions between B7 expressed constitutively on IPE cells and CTLA-4 expressed on a subpopulation of CD8+ T cells. We have now examined whether TGFbeta is required in this process. We report that IPE produces both soluble and membrane-bound active TGFbeta, but that only the latter is actually delivered to CD8+ T cells. In turn, these T cells become IPE Tregs by up-regulating their own expression of B7-1/B7-2 and soluble and membrane-bound TGFbeta. IPE Tregs through their expression of B7 are able to engage CTLA-4+ bystander T cells, and thus precisely, target delivery of membrane-bound TGFbeta. We propose that this mechanism of suppression via TGFbeta ensures that soluble active TGFbeta is not released into the ocular microenvironment where it can have unregulated and deleterious effects, including elevation of intraocular pressure and development of glaucoma.


Subject(s)
Antigens, Differentiation/immunology , CD8 Antigens/immunology , Iris/immunology , Lymphocyte Activation/immunology , Pigment Epithelium of Eye/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD , Antigens, Differentiation/genetics , CTLA-4 Antigen , Cell Communication/immunology , Cells, Cultured , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Pigment Epithelium of Eye/immunology , Pigments, Biological/immunology , RNA, Messenger/analysis , Receptors, Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/biosynthesis
13.
Blood ; 108(8): 2789-95, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16788095

ABSTRACT

T-cell subpopulations, defined by their expression of CD4, CD8, naive, and memory cell-surface markers, occupy distinct homeostatic compartments that are regulated primarily by cytokines. CD8+ memory T cells, as defined by CD44(hi) surface expression, are dependent on IL-15 as a positive regulator of their homeostatic maintenance. Manipulation of IL-15 signaling through gene aberration, overexpression, or receptor alterations has been shown to dramatically affect T-cell homeostasis, with overexpression leading to fatal leukemia. Here we show that TGF-beta is the critical negative regulator of murine CD8+ memory T-cell homeostasis with direct opposition to the positive effects of IL-15. This negative regulation is mediated, at least in part, by the ability of TGF-beta to modulate expression of the beta-chain of the IL-15 receptor, thus establishing a central axis between these 2 cytokines for homeostatic control of CD8+ memory T-cell populations. These data establish TGF-beta as a critical and dominant tumor-suppressor pathway opposing IL-15-mediated CD8+ T-cell expansion and potential malignant transformation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-15/metabolism , Transforming Growth Factor beta/metabolism , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Genes, T-Cell Receptor , Homeostasis , Immunologic Memory , Interleukin-15/deficiency , Interleukin-15/genetics , Interleukin-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Interleukin-15 , Receptors, Interleukin-2/chemistry , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics
14.
J Immunol ; 174(1): 60-7, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15611228

ABSTRACT

Epithelial cells in the thymus produce IL-7, an essential cytokine that promotes the survival, differentiation, and proliferation of thymocytes. We identified IL-7-expressing thymic epithelial cells (TECs) throughout ontogeny and in the adult mouse thymus by in situ hybridization analysis. IL-7 expression is initiated in the thymic fated domain of the early primordium by embryonic day 11.5 and is expressed in a Foxn1-independent pathway. Marked changes occur in the localization and regulation of IL-7-expressing TECs during development. IL-7-expressing TECs are present throughout the early thymic rudiment. In contrast, a major population of IL-7-expressing TECs is localized to the medulla in the adult thymus. Using mouse strains in which thymocyte development is arrested at various stages, we show that fetal and postnatal thymi differ in the frequency and localization of IL-7-expressing TECs. Whereas IL-7 expression is initiated independently of hemopoietic-derived signals during thymic organogenesis, thymocyte-derived signals play an essential role in regulating IL-7 expression in the adult TEC compartment. Moreover, different thymocyte subsets regulate the expression of IL-7 and keratin 5 in adult cortical epithelium, suggesting that despite phenotypic similarities, the cortical TEC compartments of wild-type and RAG-1(-/-) mice are developmentally and functionally distinct.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/metabolism , Interleukin-7/biosynthesis , Thymus Gland/embryology , Thymus Gland/growth & development , Animals , Embryo, Mammalian , Epithelial Cells/cytology , Flow Cytometry , Genes, RAG-1/immunology , Immunohistochemistry , In Situ Hybridization , Mice , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/cytology
15.
Blood ; 104(4): 1110-9, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15130942

ABSTRACT

Interleukin 7 (IL-7) is critical in maintaining thymic-dependent and thymic-independent pathways of T-cell homeostasis. T-cell receptor (TCR) rearrangement excision circles (TRECs) have been used as markers for recent thymic emigrants (RTEs) in assessing human thymic function. To study the thymic and peripheral effects of IL-7 on RTEs, we measured TREC content and peripheral naive T-cell subsets and turnover in IL-7-treated mice. Short-term administration of IL-7 into thymus-intact mice resulted in increased total TREC numbers, consistent with RTE accumulation. Decreases in TREC frequency were attributable to dilution secondary to increased cell turnover. Significantly, IL-7 administration into thymectomized mice resulted in patterns of decreased TREC frequency and increased total TREC number similar to those in IL-7-treated thymus-intact mice. Distinct patterns of naive cell and RTE distribution among peripheral immune organs and altered expression of CD11a were observed following IL-7 treatment in thymus-intact and thymectomized mice. These results demonstrate (1) that total TREC number and not TREC frequency accurately reflects quantitative changes in RTEs; (2) that short-term IL-7 administration results in preferential accumulations of RTEs among peripheral immune organs, accounting for the increase in TRECs in the total peripheral lymphoid pool; and (3) no evidence for regulation of thymic function by short-term IL-7 administration.


Subject(s)
Cell Movement/drug effects , Interleukin-7/pharmacology , Lymphoid Tissue/cytology , Thymus Gland/cytology , Animals , CD11a Antigen/analysis , Gene Rearrangement, T-Lymphocyte , Homeostasis/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Spleen/cytology , T-Lymphocyte Subsets , Thymectomy , Thymus Gland/physiology
16.
J Immunol ; 172(1): 214-21, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14688328

ABSTRACT

CD103 is an integrin with specificity for the epithelial cell-specific ligand, E-cadherin. Recent studies indicate that CD103 expression endows peripheral CD8 cells with a unique capacity to access the epithelial compartments of organ allografts. In the present study we used a nonvascularized mouse renal allograft model to 1) define the mechanisms regulating CD103 expression by graft-infiltrating CD8 effector populations, and 2) identify the cellular compartments in which this occurs. We report that CD8 cells responding to donor alloantigens in host lymphoid compartments do not initially express CD103, but dramatically up-regulate CD103 expression to high levels subsequent to migration to the graft site. CD103+CD8+ cells that infiltrated renal allografts exhibited a classic effector phenotype and were selectively localized to the graft site. CD8 cells expressing low levels of CD103 were also present in lymphoid compartments, but three-color analyses revealed that these are almost exclusively of naive phenotype. Adoptive transfer studies using TCR-transgenic CD8 cells demonstrated that donor-specific CD8 cells rapidly and uniformly up-regulate CD103 expression following entry into the graft site. Donor-specific CD8 cells expressing a dominant negative TGF-beta receptor were highly deficient in CD103 expression following migration to the graft, thereby implicating TGF-beta activity as a dominant controlling factor. The relevance of these data to conventional (vascularized) renal transplantation is confirmed. These data support a model in which TGF-beta activity present locally at the graft site plays a critical role in regulating CD103 expression, and hence the epitheliotropism, of CD8 effector populations that infiltrate renal allografts.


Subject(s)
Antigens, CD/biosynthesis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Integrin alpha Chains/biosynthesis , Kidney Transplantation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Movement/immunology , Female , Graft Rejection/immunology , Graft Rejection/pathology , Immunophenotyping , Integrin alpha Chains/metabolism , Kidney Cortex/blood supply , Kidney Cortex/pathology , Kidney Cortex/transplantation , Kidney Transplantation/methods , Kidney Transplantation/pathology , Male , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/transplantation , Transforming Growth Factor beta/physiology , Transplantation, Homologous/immunology , Transplantation, Homologous/methods , Transplantation, Homologous/pathology
17.
Blood ; 104(13): 4165-72, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15328149

ABSTRACT

Interleukin-7 receptor (IL-7R) levels are tightly controlled during ontogeny: high on double-negative (DN) cells, absent on double-positive (DP) cells, and high once again on thymocytes undergoing positive selection. To determine if loss of IL-7-mediated survival signals in DP cells is necessary for normal antigen-specific selection, we created T-lineage-specific IL-7R alpha chain (IL-7Ralpha) transgenic (Tg) mice in which IL-7R is expressed throughout ontogeny. There was no effect of the IL-7Ralpha Tg on negative selection. Surprisingly, however, although the thymi of IL-7Ralpha Tg mice were comparable at birth, there was a decrease in thymocyte number as the mice aged. This was found to be due to competition between DN and IL-7R-expressing DP cells for endogenous IL-7, which resulted in decreased levels of Bcl-2 in DN cells, increased DN apoptosis, and decreased DN cell number. Therefore, the down-regulation of IL-7R on DP cells is an "altruistic" act required for maintaining an adequate supply of local IL-7 for DN cells.


Subject(s)
Lymphopoiesis/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , T-Lymphocytes/immunology , Animals , Apoptosis/immunology , DNA, Complementary/genetics , Gene Expression Regulation/immunology , In Situ Nick-End Labeling , Interleukin-7/immunology , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Selection, Genetic , Thymus Gland/immunology
18.
Blood ; 104(12): 3804-12, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15292057

ABSTRACT

Graft-versus-host disease (GVHD) is a major cause of morbidity and mortality after bone marrow transplantation (BMT). CD4(+)CD25(+) immune regulatory T cells (Tregs), long recognized for their critical role in induction and maintenance of self-tolerance and prevention of autoimmunity, are also important in the regulation of immune responses in allogeneic bone marrow (BM) and solid organ transplantation. Published data indicate that ex vivo activated and expanded donor Tregs result in significant inhibition of lethal GVHD. This study provides a direct comparison of LSel(hi) and LSel(lo) Tregs for GVHD inhibition and for the promotion of allogeneic BM engraftment. Imaging of green fluorescent protein-positive effectors in GVHD control mice and LSel(hi) and LSel(lo) Treg-treated mice vividly illustrate the multisystemic nature of GVHD and the profound inhibition of GVHD by LSel(hi) Tregs. Data indicate that LSel(hi) Tregs interfere with the activation and expansion of GVHD effector T cells in secondary lymphoid organs early after BMT. Either donor- or host-type LSel(hi), but not LSel(lo), Tregs potently increased donor BM engraftment in sublethally irradiated mice, an event occurring independently of transforming growth factor beta signaling of host T cells. These data indicate that Treg cellular therapy warrants clinical consideration for the inhibition of GVHD and the promotion of alloengraftment.


Subject(s)
Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes/transplantation , Graft Rejection/prevention & control , Graft vs Host Disease/prevention & control , L-Selectin , Animals , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/methods , CD4-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Graft Survival , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , L-Selectin/analysis , Lymphocyte Transfusion/methods , Mice , Mice, Inbred Strains , Receptors, Interleukin-2 , Survival Analysis , Transforming Growth Factor beta/physiology
19.
Blood ; 104(5): 1419-27, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15155461

ABSTRACT

To study interleukin-7 (IL-7) in early thymocyte development, we generated mice transgenic (Tg) for the IL-7 gene under control of the lck proximal promoter. Founder line TgA, with the lowest level of IL-7 overexpression, showed enhanced alphabeta T-cell development. In contrast, in the highest overexpressing founder line, TgB, alphabeta T-cell development was disturbed with a block at the earliest intrathymic precursor stage. This was due to decreased progenitor proliferation as assessed by Ki-67 staining and in vivo bromodeoxyuridine (BrdU) incorporation. Bcl-2 was up-regulated in T-cell-committed progenitors in all Tg lines, and accounted for greater numbers of double positive (DP), CD4 single positive (SP), and CD8SP thymocytes in TgA mice where, in contrast to TgB mice, thymocyte progenitor proliferation was normal. Mixed marrow chimeras using TgB(+) and congenic mice as donors, and experiments using anti-IL-7 monoclonal antibody (MAb) in vivo, confirmed the role of IL-7 protein in the observed TgB phenotype. In conclusion, at low Tg overexpression, IL-7 enhanced alphabeta T-cell development by increasing thymocyte progenitor survival, while at high overexpression IL-7 reduces their proliferation, inducing a dramatic block in DP production. These results show for the first time in vivo a dose effect of IL-7 on alphabeta T-cell development and have implications for IL-7 in the clinical setting.


Subject(s)
Gene Expression Regulation, Developmental/immunology , Interleukin-7/genetics , Thymus Gland/embryology , Thymus Gland/physiology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Division/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/physiology , Leukocyte Common Antigens/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Stem Cells/cytology , Stem Cells/physiology , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Thymus Gland/cytology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL