Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
J Immunol ; 202(4): 1137-1144, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30651344

ABSTRACT

Thymus autonomy is the capacity of the thymus to maintain T lymphocyte development and export independently of bone marrow contribution. Prolonging thymus autonomy was shown to be permissive to the development of T cell acute lymphoblastic leukemia (T-ALL), similar to the human disease. In this study, performing thymus transplantation experiments in mice, we report that thymus autonomy can occur in several experimental conditions, and all are permissive to T-ALL. We show that wild type thymi maintain their function of T lymphocyte production upon transplantation into recipients with several genotypes (and corresponding phenotypic differences), i.e., Rag2 - / - γc - / -, γc - / -, Rag2 - / - IL-7rα - / -, and IL-7rα - / - We found that the cellularity of the thymus grafts is influenced exclusively by the genotype of the host, i.e., IL-7rα-/- versus γc -/- Nonetheless, the difference in cellularity detected in thymus autonomy bore no impact on onset, incidence, immunophenotype, or pathologic condition of T-ALL. In all tested conditions, T-ALL reached an incidence of 80%, demonstrating that thymus autonomy bears a high risk of leukemia. We also analyzed the microbiota composition of the recipients and their genetic background, but none of the differences found influenced the development of T-ALL. Taken together, our data support that IL-7 drives cellular turnover non-cell autonomously, which is required for prevention of T-ALL. We found no influence for T-ALL in the specific combination of the genotypic mutations tested (including the developmental block caused by Rag deficiency), in microbiota composition, or minor differences in the genetic background of the strains.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Interleukin-7/immunology , Thymus Gland/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics
2.
J Clin Immunol ; 38(6): 727-732, 2018 08.
Article in English | MEDLINE | ID: mdl-30105620

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is curative for severe combined immunodeficiency (SCID), but data on long-term impact of pre-HSCT chemotherapy, immune reconstitution and quality of life (QoL) of specific SCID genotypes are limited. We evaluated the long-term immune-reconstitution, health outcome and QoL in IL7Rα SCID, Artemis and RAG1 and 2 SCID survivors > 2 years post-HSCT in our center. Clinical data and immune reconstitution parameters were collated, and patients/families answered PedsQL generic core scale v4.0 questionnaires. Thirty-nine patients with a diagnosis of IL7Rα SCID (17 patients), Artemis SCID (8 patients) and RAG1/2 SCID (13 patients) had undergone HSCT with median age at last follow up for IL7Rα SCID, 14 years (range 4-27) and Artemis and RAG1/2 SCID, 10 years (range 2-18). Many patients have ongoing medical issues at latest follow-up [IL7Rα (73%), Artemis (85%), RAG1/2 (55%)]. Artemis SCID patients experienced more sequela than RAG1/2 SCID. Conditioned recipients with Artemis and RAG SCID had more CD4+ naïve lymphocytes compared to unconditioned recipients. All patients except those of IL7Rα SCID reported lower QoL; further subset group analysis showed parents and Artemis and RAG1/2 survivors without ongoing medical issues reported normal QoL. Conditioned recipients have superior long-term thymopoiesis, chimerism and immunoglobulin-independence. QoL was normal in those who did not have medical issues at long-term follow-up.


Subject(s)
DNA-Binding Proteins/deficiency , Endonucleases/deficiency , Homeodomain Proteins/genetics , Nuclear Proteins/deficiency , Quality of Life , Receptors, Interleukin-7/deficiency , Severe Combined Immunodeficiency/epidemiology , Severe Combined Immunodeficiency/etiology , Adolescent , Adult , Child , Child, Preschool , Disease Susceptibility , Female , Hematopoietic Stem Cell Transplantation , Humans , Longitudinal Studies , Male , Outcome Assessment, Health Care , Retrospective Studies , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/therapy , Time Factors , Young Adult
3.
Cell Death Dis ; 9(3): 273, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449560

ABSTRACT

Interleukin 7 receptor (il7r), a transmembrane receptor, belongs to the type I cytokine receptor family. Il7r is involved in the pathogenesis of neurodegenerative disorders, such as multiple sclerosis. Targeted knockdown of il7r leads to delayed myelination, highlighting the potential role of il7r in the development of the nervous system. Zebrafish is an ideal model for the study of neurogenesis; moreover, the il7r gene is highly conserved between zebrafish and human. The aim of the present study was to investigate the novel function of il7r in neurogenesis. First, an il7r -/- homozygous mutant line was generated by clustered regularly interspaced short palindromic repeats (CRISPR)-associated 9 (CRISPR/Cas9) technology. Second, the gross development of il7r-/- mutants revealed remarkably smaller eyes and delayed retinal neurodifferentiation. Third, microarray analysis revealed that genes associated with the phototransduction signalling pathway were strongly down-regulated in il7r -/- mutants. Finally, the results from behavioural tests indicated that visual function was impaired in il7r -/- mutant larvae. Overall, our data demonstrate that a lack of il7r retards the development of the retina. Thus, il7r is an essential molecule for maintaining normal retinal development in zebrafish.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knockdown Techniques , Neurogenesis , Receptors, Interleukin-7/deficiency , Retinal Neurons/metabolism , Zebrafish Proteins/deficiency , Zebrafish/metabolism , Animals , Animals, Genetically Modified , CRISPR-Associated Protein 9/metabolism , Gene Expression Regulation, Developmental , Mutation , Photic Stimulation , Receptors, Interleukin-7/genetics , Signal Transduction , Vision, Ocular , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
Biol Blood Marrow Transplant ; 21(10): 1721-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26151303

ABSTRACT

Graft-versus-host disease (GVHD) impairs immune reconstitution after allogeneic stem cell transplantation (allo-SCT) and effective therapies aimed at restoring T cell counts in GVHD patients have yet to be developed. During GVHD, CD4(+) T cell reconstitution is particularly affected and current models hold that GVHD insult to the peripheral lymphoid niche is responsible for this effect. Here, we show that naïve CD4(+) T cell homeostatic proliferation (HP) is lost during GVHD because of low systemic IL-7 and impaired dendritic cell (DC) regeneration. We assessed factors involved in DC differentiation and found that although fms-like tyrosine kinase 3 ligand (Flt3-L) levels were normal, stromal-derived factor-1α (SDF-1α) was diminished in the blood of GVHD mice. Unlike Flt3-L treatment, the administration of SDF-1α specifically increased CD8α(+) DC numbers and did not worsen GVHD. Importantly, CD4(+) T cell HP was enhanced only when IL-7 and SDF-1α or Flt3L were coadministered, confirming the crucial role of DCs and IL-7 in restoring CD4(+) T cell regeneration during GVHD. Altogether, our results indicate that CD8α(+) DCs are part of the peripheral niche that controls CD4(+) T cell HP and that their depletion, combined with low systemic IL-7, explains how GVHD constrains naïve CD4(+) T cell reconstitution after allo-SCT.


Subject(s)
Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , Chemokine CXCL12/therapeutic use , Graft vs Host Disease/drug therapy , Interleukin-7/therapeutic use , Membrane Proteins/therapeutic use , Adoptive Transfer , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/transplantation , Chemokine CXCL12/blood , Chemokine CXCL12/deficiency , Dendritic Cells/immunology , Drug Synergism , Drug Therapy, Combination , Female , Graft vs Host Disease/blood , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Interleukin-7/deficiency , Interleukin-7/physiology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/deficiency , Recombinant Proteins/therapeutic use , Stromal Cells/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Homologous
6.
Pediatr Infect Dis J ; 34(3): 301-3, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25191848

ABSTRACT

The management of patients with IL-7Rα deficient, T-B+NK+ severe combined immunodeficiency (SCID) is a challenge in absence of adequate diagnostic and treatment modalities. An infant diagnosed as SCID at 6 months of age, who received only supportive treatment is described. We present a different perspective of SCID that is managed in a low-middle income country with lack of resources for definitive therapy.


Subject(s)
Receptors, Interleukin-7/deficiency , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Humans , Infant , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Male , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/diagnosis
7.
PLoS One ; 9(2): e88771, 2014.
Article in English | MEDLINE | ID: mdl-24551160

ABSTRACT

IL-7 is a critical cytokine for lymphocyte development. Recent work has highlighted critical roles for IL-7 signaling in mature T cell homeostasis and function, but its role in B cells is less well characterized. Using a knock-in mouse possessing a Tyr to Phe mutation at position 449 (IL-7Rα(449F/449F) mice) within the cytoplasmic SH2-binding motif of IL-7Rα, we evaluated the role of IL-7Rα Y449 motif in spleen B cells. IL-7Rα(449F/449F) mice had reduced numbers and increased death of follicular B cells compared to WT, but had significantly more follicular cells than IL-7Rα(-/-). The death of IL-7Rα(449F/449F) follicular cells was not due to a failure to respond to BAFF or lower levels of BAFF, a critical B cell survival factor. Marginal zone B cells were unaffected by the IL-7Rα(449F/449F) mutation. Any role for TSLP was ruled out, as TSLPR(-/-) mice had an identical B cell phenotype to wild-type mice. Bone marrow chimeras and the absence of IL-7Rα on B cells suggested that IL-7 did not directly regulate mature B cells, but that an IL-7-responsive cell was influencing B cells. IL-7 was also critical at the checkpoint between the T1 and T2 stages in the spleen. IL-7Rα(-/-) mice fail to develop T2 cells, but IL-7Rα(449F/449F) show a reduction compared to WT but not complete absence of T2 cells. We also tested the functional responses of IL-7Rα(449F/449F) to antigens and infection and found no difference in antibody responses to T-dependent or T-independent antigens, or to Influenza/A. IL-7 was important for generation of antibody responses to the intestinal worm H. polygyrus and for naive levels of IgA. Taken together, this suggests that IL-7 regulates follicular B cell numbers and survival in a cell-extrinsic manner, via a bone-marrow derived cell, but is not critical for antibody production outside the gut.


Subject(s)
B-Lymphocytes/immunology , Interleukin-7/immunology , Receptors, Interleukin-7/immunology , Signal Transduction/immunology , Amino Acid Substitution , Animals , Antibodies, Viral/blood , Antigens, Viral/blood , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , B-Lymphocytes/cytology , Cell Survival , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Gene Knock-In Techniques , Immunoglobulin G/blood , Influenza A virus/immunology , Interleukin-7/genetics , Mice , Mice, Transgenic , Protein Interaction Domains and Motifs , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , Spleen/cytology , Spleen/immunology , Thymic Stromal Lymphopoietin
8.
Immunobiology ; 218(7): 960-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23312956

ABSTRACT

Recent evidence indicates that monocytes and macrophages express T cell receptor (TCR)αß-like combinatorial immune receptors. Here, we demonstrate the presence of a second recombinatorial immunoreceptor, which is structurally based on the TCR γ- and δ-chains, in human and murine monocytes and differentially activated macrophages (referred to here as TCRL(m)γδ). In vitro, infection of macrophages with mycobacteria and gram positive or gram negative bacteria induced expression of donor-specific and differential TCRL(m)Vδ repertoires indicating that the novel immunoreceptor represents a dynamic flexible host defense system that responds to bacterial challenge. In vivo, we find that TCRL(m)γδ bearing macrophages, which express highly restricted repertoires of the antigen-binding Vδ chain, accumulate in the cerebrospinal fluid in acute bacterial meningitis and in advanced lesions of atherosclerosis. These results identify an as yet unrecognized monocyte/macrophage subpopulation that bears combinatorial TCRL(m)γδ immune receptors, and is associated with both acute and chronic inflammatory diseases. Moreover, they indicate that the monocytic lineage uses the same bipartite system of TCRαß/TCRγδ-based combinatorial immune receptors that is present in T cells. Our findings suggest specific roles of monocytes/macrophages in various inflammatory conditions and lend further evidence that flexible immune recognition in higher vertebrates operates on a broader cellular basis than previously thought.


Subject(s)
Adaptive Immunity , Bacterial Infections/immunology , Gene Expression/immunology , Macrophages/immunology , Meningitis, Bacterial/immunology , Monocytes/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Acute Disease , Aged , Animals , Antigens, Bacterial/immunology , Atherosclerosis/immunology , Atherosclerosis/pathology , Bacterial Infections/microbiology , Bacterial Infections/pathology , Escherichia coli/immunology , Humans , Immunophenotyping , Macrophages/cytology , Macrophages/microbiology , Male , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/pathology , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/microbiology , Mycobacterium bovis/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
9.
Blood ; 121(1): 64-71, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23152541

ABSTRACT

The mouse thymus supports T-cell development, but also contains non-T-cell lineages such as dendritic cells, macrophages, and granulocytes that are necessary for T-cell repertoire selection and apoptotic thymocyte clearance. Early thymic progenitors (ETPs) are not committed to the T-cell lineage, as demonstrated by both in vitro and in vivo assays. Whether ETPs realize non-T-cell lineage potentials in vivo is not well understood and indeed is controversial. In the present study, we investigated whether ETPs are the major precursors of any non-T-lineage cells in the thymus. We analyzed the development of these populations under experimental circumstances in which ETPs are nearly absent due to either abrogated thymic settling or inhibition of early thymic development by genetic ablation of IL-7 receptorα or Hes1. Results obtained using multiple in vivo approaches indicate that the majority of thymic granulocytes derive from ETPs. These data indicate that myelolymphoid progenitors settle the thymus and thus clarify the pathways by which stem cells give rise to downstream blood cell lineages.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/physiology , Lymphopoiesis/physiology , Receptors, Interleukin-7/physiology , T-Lymphocyte Subsets/cytology , Thymus Gland/cytology , Animals , Animals, Congenic , Basic Helix-Loop-Helix Transcription Factors/deficiency , Bone Marrow Transplantation , Cell Lineage , Cell Movement , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Homeodomain Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Radiation Chimera , Receptors, Interleukin-7/deficiency , Thymus Gland/growth & development , Transcription Factor HES-1
10.
J Exp Med ; 209(8): 1401-8, 2012 Jul 30.
Article in English | MEDLINE | ID: mdl-22778388

ABSTRACT

Thymus transplants can correct deficiencies of the thymus epithelium caused by the complete DiGeorge syndrome or FOXN1 mutations. However, thymus transplants were never used to correct T cell-intrinsic deficiencies because it is generally believed that thymocytes have short intrinsic lifespans. This notion is based on thymus transplantation experiments where it was shown that thymus-resident cells were rapidly replaced by progenitors originating in the bone marrow. In contrast, here we show that neonatal thymi transplanted into interleukin 7 receptor-deficient hosts harbor populations with extensive capacity to self-renew, and maintain continuous thymocyte generation and export. These thymus transplants reconstitute the full diversity of peripheral T cell repertoires one month after surgery, which is the earliest time point studied. Moreover, transplantation experiments performed across major histocompatibility barriers show that allogeneic transplanted thymi are not rejected, and allogeneic cells do not induce graft-versus-host disease; transplants induced partial or total protection to infection. These results challenge the current dogma that thymocytes cannot self-renew, and indicate a potential use of neonatal thymus transplants to correct T cell-intrinsic deficiencies. Finally, as found with mature T cells, they show that thymocyte survival is determined by the competition between incoming progenitors and resident cells.


Subject(s)
Bone Marrow Cells/cytology , Thymocytes/cytology , Thymus Gland/transplantation , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Graft vs Host Disease/prevention & control , Mice , Mice, Inbred BALB C , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Transplantation, Homologous
11.
J Autoimmun ; 37(3): 217-27, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21745722

ABSTRACT

Type 1 diabetes (T1D) develops as a consequence of a progressive autoimmune response that destroys insulin-producing ß-cells in pancreatic islets. Because of their role(s) in controlling immune responses, considerable effort has been directed toward resolving whether regulatory T cells (Tregs) offer a clinical treatment to restore tolerance in T1D. We previously reported that in vitro-induced adaptive Treg cells (aTregs) can reverse T1D and persist as protective memory cells in the NOD mouse model. In the current study, we investigated mechanisms that regulate aTregs. We found that these FoxP3(+) aTregs expressed high levels of the IL-7 receptor, IL-7Rα, without the high affinity receptor for IL-2, CD25, which is found on natural Treg cells (nTregs). IL-7Rα expression was mirrored by the dependency of aTregs on IL-7 for persistence. IL-10 and TGF-ß, effector cytokines of aTregs, were not essential for their maintenance at the level of systemic antibody blocking. Nevertheless, IL-10 modulated cytokine production by aTregs and TGF-ß was critical for protection. aTregs were found to infiltrate islets and the expression of integrin-ß7 was required for their localization in the pancreas. Furthermore, blocking aTreg entry into the pancreas prevented their control of diabetogenic effector T cells, implying the need for local control of the autoimmune response. The distinct homeostatic regulation of aTregs independently of a response to IL-2, which is defective in T1D patients, suggests that these cells represent a translatable candidate to control the autoimmune response.


Subject(s)
Adaptive Immunity , Diabetes Mellitus, Type 1/immunology , Integrin beta Chains/immunology , Interleukin-7/immunology , Pancreas/immunology , Receptors, Interleukin-7/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory , Adoptive Transfer , Animals , Autoimmunity , Cell Differentiation/immunology , Cell Movement/immunology , Diabetes Mellitus, Type 1/metabolism , Female , Humans , Integrin beta Chains/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7/deficiency , Interleukin-7/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Pancreas/metabolism , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
12.
J Immunol ; 186(4): 2623-32, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21217010

ABSTRACT

We have previously demonstrated that IL-7 is essential for the persistence of colitis as a survival factor of colitogenic IL-7Rα-expressing memory CD4(+) T cells. Because IL-7Rα is broadly expressed on various immune cells, it is possible that the persistence of colitogenic CD4(+) T cells is affected by other IL-7Rα-expressing non-T cells. To test this hypothesis, we conducted two adoptive transfer colitis experiments using IL-7Rα(-/-) CD4(+)CD25(-) donor cells and IL-7Rα(-/-) × RAG-2(-/-) recipient mice, respectively. First, IL-7Rα expression on colitic lamina propria (LP) CD4(+) T cells was significantly higher than on normal LP CD4(+) T cells, whereas expression on other colitic LP immune cells, (e.g., NK cells, macrophages, myeloid dendritic cells) was conversely lower than that of paired LP cells in normal mice, resulting in predominantly higher expression of IL-7Rα on colitogenic LP CD4(+) cells, which allows them to exclusively use IL-7. Furthermore, RAG-2(-/-) mice transferred with IL-7Rα(-/-) CD4(+)CD25(-) T cells did not develop colitis, although LP CD4(+) T cells from mice transferred with IL-7Rα(-/-) CD4(+)CD25(-) T cells were differentiated to CD4(+)CD44(high)CD62L(-) effector-memory T cells. Finally, IL-7Rα(-/-) × RAG-2(-/-) mice transferred with CD4(+)CD25(-) T cells developed colitis similar to RAG-2(-/-) mice transferred with CD4(+)CD25(-) T cells. These results suggest that IL-7Rα expression on colitogenic CD4(+) T cells, but not on other cells, is essential for the development of chronic colitis. Therefore, therapeutic approaches targeting the IL-7/IL-7R signaling pathway in colitogenic CD4(+) T cells may be feasible for the treatment of inflammatory bowel diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Immunologic Memory , Receptors, Interleukin-7/biosynthesis , Up-Regulation/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cells, Cultured , Chronic Disease , Colitis/genetics , Colitis/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Genetic Predisposition to Disease , Immunologic Memory/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , Up-Regulation/genetics
13.
J Immunol ; 186(4): 1981-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21239710

ABSTRACT

Lymphopenia-induced proliferation (LIP) is a proliferative program initiated in response to T cell insufficiency caused by acute or chronic immunodepletion. Studies of lymphopenic mice have demonstrated that the cytokine IL-7 and TCR signaling are critical for LIP. We examined how these two factors impact T cell proliferation following transfer into moderately lymphopenic mice. In this study, we show that moderate lymphopenia (∼25% of wild-type lymphocytes) of IL-7Rα knock-in mutant (IL-7Rα(449F)) mice supports T cell proliferation, although with decreased frequency and kinetics compared with cells transferred to severely lymphopenic (5% of wild-type lymphocytes) IL-7Rα(-/-) hosts. Although previous studies have demonstrated that elevated IL-7 levels play an important role in LIP, IL-7 availability was not elevated in IL-7Rα(449F) mice. However, moderate lymphopenia increased access of transferred T cells to self-peptide presented on APCs that can trigger TCR signaling and proliferation. Importantly, we did not detect significant changes in TCR Vß usage of proliferated T cells recovered from either moderately or severely lymphopenic hosts. Our work demonstrates that polyclonal T cells retain a diverse TCR repertoire following proliferation mediated by either self-peptide-MHC interaction alone or in combination with IL-7, and that T cell reconstitution is most efficient in the presence of increased IL-7 availability.


Subject(s)
Cell Proliferation , Interleukin-7/biosynthesis , Lymphopenia/immunology , Lymphopenia/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Up-Regulation/immunology , Animals , Biological Availability , Chronic Disease , Cytokines/metabolism , Gene Knock-In Techniques , Humans , Interleukin-7/metabolism , Interleukin-7/physiology , Lymphocyte Depletion , Lymphopenia/genetics , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , Receptors, Interleukin-7/biosynthesis , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , Spleen/immunology , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/metabolism , Up-Regulation/genetics
14.
J Immunol ; 186(3): 1343-7, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187442

ABSTRACT

The transcription factor Krüppel-like factor 2 (KLF2) controls the emigration of conventional T cells from the thymus through its regulation of the cell surface receptor S1P1. Prior to KLF2 expression, developing T cells require a positive selection signal through the TCR. However, following positive selection there are time, spatial, and maturational events that occur before KLF2 is finally upregulated and emigration occurs. We are interested in determining the signals that upregulate KLF2 and allow thymocytes to emigrate into circulation and whether they are linked to functional maturation. In endothelial cells KLF2 expression has been shown to be dependent on the mitogen-activated protein kinase ERK5. Furthermore, it has been reported that IL-7 signaling leads to the phosphorylation of ERK5. Thus, we hypothesized that IL-7R signaling through ERK5 could drive the expression of KLF2. In this study, we provide evidence that this hypothesis is incorrect. We also found that CD8 lineage specification occurred normally in the absence of IL-7R signaling, in contrast to a recently proposed model. We showed that both CD4 and CD8 T cells complete maturation and express KLF2 independently of ERK5 and IL-7.


Subject(s)
Cell Differentiation/immunology , Cell Movement/immunology , Interleukin-7/physiology , Mitogen-Activated Protein Kinase 7/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 7/deficiency , Mitogen-Activated Protein Kinase 7/genetics , Receptors, Interleukin-7/antagonists & inhibitors , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism
15.
J Exp Med ; 206(5): 1009-17, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19414553

ABSTRACT

Two subsets of natural and adaptive regulatory T (T reg) cells have been described, but the identity of adaptive type 1 regulatory (Tr1)-like cells in humans is unclear. We analyzed a subset of human blood CD4(+) T cells--CD45RA(-)CD25(-)interleukin (IL)-7 receptor (R)(-) cells--that rapidly secreted high levels of IL-10 together with interferon gamma, but produced little IL-2. These IL-7R(-) T cells were rare, anergic, and largely Foxp3(-). They expressed low levels of Bcl-2 but high levels of Ki-67 and ICOS, suggesting that they have been recently activated in vivo. Consistently, they responded selectively to persistent foreign and self-antigens under steady-state conditions. Unlike natural CD25(+) T reg cells, IL-7R(-) cells suppressed naive and memory T cell proliferation in an IL-10-dependent fashion, and they required strong T cell receptor stimulation for suppression. To our knowledge, this is the first report that identifies Tr1-like cells in human blood. These IL-10-secreting cells have characteristics of chronically activated Th1 effector cells and are distinct from CD25(+) T reg cells.


Subject(s)
Interferon-gamma/immunology , Interleukin-10/immunology , Receptors, Interleukin-7/deficiency , T-Lymphocytes, Regulatory/immunology , Antigens, CD/immunology , Autoantigens/immunology , Genes, T-Cell Receptor , Humans , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation , Receptors, CCR7/deficiency , Receptors, CCR7/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
16.
Blood ; 113(23): 5793-800, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19357399

ABSTRACT

Interleukin-7 (IL-7) plays a central role in the homeostasis of the T-cell compartment by regulating T-cell survival and proliferation. Whether IL-7 can influence T-cell receptor (TCR) signaling in T cells remains controversial. Here, using IL-7-deficient hosts and TCR-transgenic T cells that conditionally express IL-7R, we examined antigen-specific T-cell responses in vitro and in vivo to viral infection and lymphopenia to determine whether IL-7 signaling influences TCR-triggered cell division events. In vitro, we could find no evidence that IL-7 signaling could costimulate T-cell activation over a broad range of conditions, suggesting that IL-7 does not directly tune TCR signaling. In vivo, however, we found an acute requirement for IL-7 signaling for efficiently triggering T-cell responses to influenza A virus challenge. Furthermore, we found that IL-7 was required for the enhanced homeostatic TCR signaling that drives lymphopenia-induced proliferation by a mechanism involving efficient contacts of T cells with dendritic cells. Consistent with this, saturating antigen-presenting capacity in vivo overcame the triggering defect in response to cognate peptide. Thus, we demonstrate a novel role for IL-7 in regulating T cell-dendritic cell interactions that is essential for both T-cell homeostasis and activation in vivo.


Subject(s)
Dendritic Cells/immunology , Homeostasis/immunology , Interleukin-7/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigens/immunology , Dendritic Cells/metabolism , Interleukin-7/deficiency , Interleukin-7/genetics , Interleukin-7/metabolism , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , Signal Transduction/immunology , Substrate Specificity/immunology , T-Lymphocytes/metabolism
17.
J Exp Med ; 206(4): 761-78, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19289576

ABSTRACT

Thymic T cell progenitor (TCP) importation is a periodic, gated event that is dependent on the expression of functional P-selectin ligands on TCPs. Occupancy of intrathymic TCP niches is believed to negatively regulate TCP importation, but the nature of this feedback mechanism is not yet resolved. We show that P-selectin and CCL25 are periodically expressed in the thymus and are essential parts of the thymic gate-keeping mechanism. Periodicity of thymic TCP receptivity and the size of the earliest intrathymic TCP pool were dependent on the presence of functional P-selectin ligand on TCPs. Furthermore, we show that the numbers of peripheral blood lymphocytes directly affected thymic P-selectin expression and TCP receptivity. We identified sphingosine-1-phosphate (S1P) as one feedback signal that could mediate influence of the peripheral lymphocyte pool on thymic TCP receptivity. Our findings suggest a model whereby thymic TCP importation is controlled by both early thymic niche occupancy and the peripheral lymphocyte pool via S1P.


Subject(s)
P-Selectin/genetics , Serine Endopeptidases/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Bone Marrow Cells/immunology , Homeostasis , Lymphocytes/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Proprotein Convertases , RNA/genetics , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , Serine Endopeptidases/blood , Transcription, Genetic
18.
J Exp Med ; 206(3): 607-22, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19273629

ABSTRACT

Many dendritic cells (DCs) in the normal mouse thymus are generated intrathymically from common T cell/DC progenitors. However, our previous work suggested that at least 50% of thymic DCs originate independently of these progenitors. We now formally demonstrate by parabiotic, adoptive transfer, and developmental studies that two of the three major subsets of thymic DCs originate extrathymically and continually migrate to the thymus, where they occupy a finite number of microenvironmental niches. The thymus-homing DCs consisted of immature plasmacytoid DCs (pDCs) and the signal regulatory protein alpha-positive (Sirpalpha(+)) CD11b(+) CD8alpha(-) subset of conventional DCs (cDCs), both of which could take up and transport circulating antigen to the thymus. The cDCs of intrathymic origin were mostly Sirpalpha(-) CD11b(-) CD8alpha(hi) cells. Upon arrival in the thymus, the migrant pDCs enlarged and up-regulated CD11c, major histocompatibility complex II (MHC II), and CD8alpha, but maintained their plasmacytoid morphology. In contrast, the migrant cDCs proliferated extensively, up-regulated CD11c, MHC II, and CD86, and expressed dendritic processes. The possible functional implications of these findings are discussed.


Subject(s)
Cell Movement , Dendritic Cells/cytology , Dendritic Cells/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Adoptive Transfer , Aging/immunology , Animals , Antigens/immunology , Blood Cells/cytology , Blood Cells/immunology , Cell Differentiation , Cell Proliferation , Chimerism , Homeodomain Proteins/metabolism , Inflammation/immunology , Mice , Microspheres , Models, Immunological , Monocytes/cytology , Monocytes/immunology , Ovalbumin/immunology , Parabiosis , Phenotype , Protein Transport , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/metabolism
19.
Blood ; 113(17): 3961-8, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19246338

ABSTRACT

Interleukin-7 (IL-7) is crucial for the development of T and B lymphocytes from common lymphoid progenitors (CLPs) and for the maintenance of mature T lymphocytes. Its in vivo role for dendritic cells (DCs) has been poorly defined. Here, we investigated whether IL-7 is important for the development or maintenance of different DC types. Bone marrow-derived DCs expressed the IL-7 receptor (IL-7R) and survived significantly longer in the presence of IL-7. Migratory DCs (migDCs) isolated from lymph nodes also expressed IL-7R. Surprisingly, IL-7R was not required for their maintenance but indirectly for their development. Conventional DCs (cDCs) and plasmacytoid DCs (pDCs) resident in lymph nodes and spleen were IL-7R(-). Using mixed bone marrow chimeras, we observed an intrinsic requirement for IL-7R signals in their development. As the number of CLPs but not myeloid progenitors was reduced in the absence of IL-7 signals, we propose that a large fraction of cDCs and pDCs derives from CLPs and shares not only the lymphoid origin but also the IL-7 requirement with lymphocyte precursors.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Interleukin-7/immunology , Interleukin-7/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Gene Expression Regulation/immunology , Interleukin-7/deficiency , Interleukin-7/genetics , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/deficiency , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , Signal Transduction/immunology
20.
Proc Natl Acad Sci U S A ; 106(2): 552-7, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19122139

ABSTRACT

Gene knockout experiments in mice have suggested a hierarchical model of early B cell commitment wherein E2A proteins (E47 and E12) activate early B cell factor (Ebf1), which in turn activates expression of the B cell commitment factor, Pax5. In IL-7 receptor alpha (IL-7Ralpha) knockout mice, B cell development is blocked before B-lineage commitment at the prepro-B cell stage in adult animals. In IL-7Ralpha(-/-) prepro-B cells, E47 is expressed and yet is insufficient to transcriptionally activate the putative downstream target gene, Ebf1. In this study, we show that further increases of E47 expression in IL-7Ralpha(-/-) prepro-B cells fails to activate Ebf1, but rather leads to a dramatic induction of the E2A inhibitory factors, Id2 and Id3. In contrast, enforced expression of Ebf1 in IL-7Ralpha(-/-) bone marrow potently down-regulates Id2 and Id3 mRNA expression and restores B cell differentiation in vivo. Down-regulation of both Id2 and Id3 during B cell specification is essential in that overexpression of either Id2 or Id3 in wild-type bone marrow blocks B cell specification at the prepro-B cell stage. Collectively, these studies suggest a model where Ebf1 induction specifies the B cell fate by dramatically increasing activity of E47 at the posttranslational level.


Subject(s)
B-Lymphocytes/cytology , Down-Regulation , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Proteins/genetics , Receptors, Interleukin-7/deficiency , TCF Transcription Factors/physiology , Trans-Activators/physiology , Animals , Bone Marrow , Cell Differentiation , Cell Lineage , Gene Expression Regulation , Inhibitor of Differentiation Protein 2/physiology , Inhibitor of Differentiation Proteins/physiology , Interleukin-7/deficiency , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid , RNA, Messenger/analysis , TCF Transcription Factors/genetics , Transcription Factor 7-Like 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...