Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nat Immunol ; 24(4): 637-651, 2023 04.
Article in English | MEDLINE | ID: mdl-36959291

ABSTRACT

Thymocytes bearing autoreactive T cell receptors (TCRs) are agonist-signaled by TCR/co-stimulatory molecules to either undergo clonal deletion or to differentiate into specialized regulatory T (Treg) or effector T (Teff) CD4+ cells. How these different fates are achieved during development remains poorly understood. We now document that deletion and differentiation are agonist-signaled at different times during thymic selection and that Treg and Teff cells both arise after clonal deletion as alternative lineage fates of agonist-signaled CD4+CD25+ precursors. Disruption of agonist signaling induces CD4+CD25+ precursors to initiate Foxp3 expression and become Treg cells, whereas persistent agonist signaling induces CD4+CD25+ precursors to become IL-2+ Teff cells. Notably, we discovered that transforming growth factor-ß induces Foxp3 expression and promotes Treg cell development by disrupting weaker agonist signals and that Foxp3 expression is not induced by IL-2 except under non-physiological in vivo conditions. Thus, TCR signaling disruption versus persistence is a general mechanism of lineage fate determination in the thymus that directs development of agonist-signaled autoreactive thymocytes.


Subject(s)
Clonal Deletion , Thymocytes , Thymocytes/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , CD4-Positive T-Lymphocytes/metabolism , Thymus Gland/metabolism , Receptors, Antigen, T-Cell/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism
2.
Nat Immunol ; 18(11): 1218-1227, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945245

ABSTRACT

T cell antigen receptor (TCR) signaling in the thymus initiates positive selection, but the CD8+-lineage fate is thought to be induced by cytokines after TCR signaling has ceased, although this remains controversial and unproven. We have identified four cytokines (IL-6, IFN-γ, TSLP and TGF-ß) that did not signal via the common γ-chain (γc) receptor but that, like IL-7 and IL-15, induced expression of the lineage-specifying transcription factor Runx3d and signaled the generation of CD8+ T cells. Elimination of in vivo signaling by all six of these 'lineage-specifying cytokines' during positive selection eliminated Runx3d expression and completely abolished the generation of CD8+ single-positive thymocytes. Thus, this study proves that signaling during positive selection by lineage-specifying cytokines is responsible for all CD8+-lineage-fate 'decisions' in the thymus.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Lineage/immunology , Cytokines/immunology , Thymus Gland/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , Cytokines/metabolism , Flow Cytometry , Gene Expression/immunology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
3.
Nat Immunol ; 17(12): 1415-1423, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27668801

ABSTRACT

Major histocompatibility complex class I (MHC I) positive selection of CD8+ T cells in the thymus requires that T cell antigen receptor (TCR) signaling end in time for cytokines to induce Runx3d, the CD8-lineage transcription factor. We examined the time required for these events and found that the overall duration of positive selection was similar for all CD8+ thymocytes in mice, despite markedly different TCR signaling times. Notably, prolonged TCR signaling times were counter-balanced by accelerated Runx3d induction by cytokines and accelerated differentiation into CD8+ T cells. Consequently, lineage errors did not occur except when MHC I-TCR signaling was so prolonged that the CD4-lineage-specifying transcription factor ThPOK was expressed, preventing Runx3d induction. Thus, our results identify a compensatory signaling mechanism that prevents lineage-fate errors by dynamically modulating Runx3d induction rates during MHC I positive selection.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Clonal Selection, Antigen-Mediated , Core Binding Factor Alpha 3 Subunit/metabolism , Histocompatibility Antigens Class I/metabolism , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , Cytokines/metabolism , Histocompatibility Antigens Class I/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transcription Factors
4.
Cell ; 154(6): 1326-41, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034254

ABSTRACT

Thymic selection requires signaling by the protein tyrosine kinase Lck to generate T cells expressing αß T cell antigen receptors (TCR). For reasons not understood, the thymus selects only αßTCR that are restricted by major histocompatibility complex (MHC)-encoded determinants. Here, we report that Lck proteins that were coreceptor associated promoted thymic selection of conventionally MHC-restricted TCR, but Lck proteins that were coreceptor free promoted thymic selection of MHC-independent TCR. Transgenic TCR with MHC-independent specificity for CD155 utilized coreceptor-free Lck to signal thymic selection in the absence of MHC, unlike any transgenic TCR previously described. Thus, the thymus can select either MHC-restricted or MHC-independent αßTCR depending on whether Lck is coreceptor associated or coreceptor free. We conclude that the intracellular state of Lck determines the specificity of thymic selection and that Lck association with coreceptor proteins during thymic selection is the mechanism by which MHC restriction is imposed on a randomly generated αßTCR repertoire.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , T-Lymphocytes/cytology , Thymocytes/metabolism , Thymus Gland/metabolism , Animals , Major Histocompatibility Complex , Mice , Receptors, Antigen, T-Cell, alpha-beta , Receptors, Virus , Signal Transduction , T-Lymphocytes/metabolism , Thymus Gland/immunology
5.
Nat Immunol ; 14(2): 143-51, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23242416

ABSTRACT

The maintenance of naive CD8(+) T cells is necessary for lifelong immunocompetence but for unknown reasons requires signaling via both interleukin 7 (IL-7) and the T cell antigen receptor (TCR). We now report that naive CD8(+) T cells required IL-7 signaling to be intermittent, not continuous, because prolonged IL-7 signaling induced naive CD8(+) T cells to proliferate, produce interferon-γ (IFN-γ) and undergo IFN-γ-triggered cell death. Homeostatic engagement of the TCR interrupted IL-7 signaling and thereby supported the survival and quiescence of CD8(+) T cells. However, CD8(+) T cells with insufficient TCR affinity for self ligands received prolonged IL-7 signaling and died during homeostasis. In this study we identified regulation of the duration of IL-7 signaling by homeostatic engagement of the TCR as the basis for in vivo CD8(+) T cell homeostasis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeostasis/immunology , Interleukin-7/genetics , Receptors, Antigen, T-Cell/genetics , Signal Transduction/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Death/immunology , Cell Proliferation , Cell Survival/immunology , Gene Expression Regulation , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-7/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Time Factors
6.
Nat Immunol ; 13(6): 569-78, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22544394

ABSTRACT

Clonal deletion of autoreactive thymocytes is important for self-tolerance, but the intrathymic signals that induce clonal deletion have not been clearly identified. We now report that clonal deletion during negative selection required CD28-mediated costimulation of autoreactive thymocytes at the CD4(+)CD8(lo) intermediate stage of differentiation. Autoreactive thymocytes were prevented from undergoing clonal deletion by either a lack of CD28 costimulation or transgenic overexpression of the antiapoptotic factors Bcl-2 or Mcl-1, with surviving thymocytes differentiating into anergic CD4(-)CD8(-) double-negative thymocytes positive for the T cell antigen receptor αß subtype (TCRαß) that 'preferentially' migrated to the intestine, where they re-expressed CD8α and were sequestered as CD8αα(+) intraepithelial lymphocytes (IELs). Our study identifies costimulation by CD28 as the intrathymic signal required for clonal deletion and identifies CD8αα(+) IELs as the developmental fate of autoreactive thymocytes that survive negative selection.


Subject(s)
Cell Differentiation/immunology , Clonal Deletion/immunology , Receptors, Antigen, T-Cell/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , CD28 Antigens/immunology , CD4 Antigens/immunology , CD8 Antigens/immunology , Flow Cytometry , Immune Tolerance/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , Thymocytes/cytology , Thymus Gland/cytology
7.
Immunity ; 40(6): 910-23, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24909888

ABSTRACT

The common γ-chain (γc) plays a central role in signaling by IL-2 and other γc-dependent cytokines. Here we report that activated T cells produce an alternatively spliced form of γc mRNA that results in protein expression and secretion of the γc extracellular domain. The soluble form of γc (sγc) is present in serum and directly binds to IL-2Rß and IL-7Rα proteins on T cells to inhibit cytokine signaling and promote inflammation. sγc suppressed IL-7 signaling to impair naive T cell survival during homeostasis and exacerbated Th17-cell-mediated inflammation by inhibiting IL-2 signaling upon T cell activation. Reciprocally, the severity of Th17-cell-mediated inflammatory diseases was markedly diminished in mice lacking sγc. Thus, sγc expression is a naturally occurring immunomodulator that regulates γc cytokine signaling and controls T cell activation and differentiation.


Subject(s)
Alternative Splicing/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunoglobulin gamma-Chains/immunology , Inflammation/immunology , Th17 Cells/immunology , Animals , Autoimmunity , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/immunology , Immunoglobulin gamma-Chains/blood , Immunoglobulin gamma-Chains/genetics , Immunomodulation , Interleukin-2 Receptor beta Subunit/immunology , Interleukin-5 Receptor alpha Subunit/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/immunology , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction/immunology
8.
Immunity ; 38(6): 1116-28, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23746651

ABSTRACT

Immune tolerance requires regulatory T (Treg) cells to prevent autoimmune disease, with the transcription factor Foxp3 functioning as the critical regulator of Treg cell development and function. We report here that Foxp3 was lethal to developing Treg cells in the thymus because it induced a unique proapoptotic protein signature (Puma⁺⁺⁺p-Bim⁺⁺p-JNK⁺⁺DUSP6⁻) and repressed expression of prosurvival Bcl-2 molecules. However, Foxp3 lethality was prevented by common gamma chain (γc)-dependent cytokine signals that were present in the thymus in limiting amounts sufficient to support only ∼1 million Treg cells. Consequently, most newly arising Treg cells in the thymus were deprived of this signal and underwent Foxp3-induced death, with Foxp3⁺CD25⁻ Treg precursor cells being the most susceptible. Thus, we identify Foxp3 as a proapoptotic protein that requires developing Treg cells to compete with one another for limiting amounts of γc-dependent survival signals in the thymus.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cytokines/immunology , Forkhead Transcription Factors/metabolism , Interleukin Receptor Common gamma Subunit/immunology , T-Lymphocytes, Regulatory/physiology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Survival , Cells, Cultured , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Lymphopoiesis/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
9.
EMBO J ; 31(2): 366-77, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22036949

ABSTRACT

Immature thymocytes are bipotential cells that are signalled during positive selection to become either helper- or cytotoxic-lineage T cells. By tracking expression of lineage determining transcription factors during positive selection, we now report that the Cd8 coreceptor gene locus co-opts any coreceptor protein encoded within it to induce thymocytes to express the cytotoxic-lineage factor Runx3 and to adopt the cytotoxic-lineage fate, findings we refer to as 'coreceptor gene imprinting'. Specifically, encoding CD4 proteins in the endogenous Cd8 gene locus caused major histocompatibility complex class II-specific thymocytes to express Runx3 during positive selection and to differentiate into CD4(+) cytotoxic-lineage T cells. Our findings further indicate that coreceptor gene imprinting derives from the dynamic regulation of specific cis Cd8 gene enhancer elements by positive selection signals in the thymus. Thus, for coreceptor-dependent thymocytes, lineage fate is determined by Cd4 and Cd8 coreceptor gene loci and not by the specificity of T-cell antigen receptor/coreceptor signalling. This study identifies coreceptor gene imprinting as a critical determinant of lineage fate determination in the thymus.


Subject(s)
CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , Lymphopoiesis/genetics , T-Lymphocyte Subsets/cytology , Thymocytes/cytology , Animals , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , Cell Lineage/genetics , Clonal Selection, Antigen-Mediated , Core Binding Factor Alpha 3 Subunit/biosynthesis , Core Binding Factor Alpha 3 Subunit/genetics , Cytokines/physiology , DNA, Recombinant/genetics , Enhancer Elements, Genetic/genetics , Gene Knock-In Techniques , Genes, Synthetic , Mice , Mice, Inbred C57BL , Mice, Transgenic , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics
10.
Crit Rev Immunol ; 35(1): 59-76, 2015.
Article in English | MEDLINE | ID: mdl-25746048

ABSTRACT

Development and central tolerance of T lymphocytes in the thymus requires both TCR signals and collaboration with signals generated through costimulatory molecule interactions. In this review, we discuss the importance of CD28-CD80/86 and CD40-CD40L costimulatory interactions in promoting normal thymic development. This discussion includes roles in the generation of a normal thymic medulla, in the development of specific T-cells subsets, including iNKT and T regulatory cells, and in the generation of a tolerant mature T-cell repertoire. We discuss recent contributions to the understanding of CD28-CD80/86 and CD40-CD40L costimulatory interactions in thymic development, and we highlight the ways in which the many important roles mediated by these interactions collaborate to promote normal thymic development.


Subject(s)
B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD28 Antigens/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Epithelial Cells/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Animals , Cell Differentiation , Humans , Immune Tolerance , Receptor Cross-Talk , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
11.
J Immunol ; 192(6): 2892-903, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24523508

ABSTRACT

Expression of MHC class I molecules, which provide immune surveillance against intracellular pathogens, is higher on lymphoid cells than on any other cell types. In T cells, this is a result of activation of class I transcription by the T cell enhanceosome consisting of Runx1, CBFß, and LEF1. We now report that MHC class I transcription in T cells also is enhanced by Foxp3, resulting in higher levels of class I in CD4(+)CD25(+) T regulatory cells than in conventional CD4(+)CD25(-) T cells. Interestingly, the effect of Foxp3 regulation of MHC class I transcription is cell type specific: Foxp3 increases MHC class I expression in T cells but represses it in epithelial tumor cells. In both cell types, Foxp3 targets the upstream IFN response element and downstream core promoter of the class I gene. Importantly, expression of MHC class I contributes to the function of CD4(+)CD25(+) T regulatory cells by enhancing immune suppression, both in in vitro and in vivo. These findings identify MHC class I genes as direct targets of Foxp3 whose expression augments regulatory T cell function.


Subject(s)
Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Base Sequence , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HeLa Cells , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Jurkat Cells , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , T-Lymphocytes, Regulatory/metabolism , Thymocytes/immunology , Thymocytes/metabolism , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
12.
Eur J Immunol ; 43(9): 2283-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712827

ABSTRACT

γ-Chain (γc) cytokine receptor signaling is required for the development of all lymphocytes. Why γc signaling plays such an essential role is not fully understood, but induction of the serine/threonine kinase Pim1 is considered a major downstream event of γc as Pim1 prevents apoptosis and increases metabolic activity. Consequently, we asked whether Pim1 overexpression would suffice to restore lymphocyte development in γc-deficient mice. By analyzing Pim1-transgenic γc-deficient mice (Pim1(Tg) γc(KO) ), we show that Pim1 promoted T-cell development and survival in the absence of γc. Interestingly, such effects were largely limited to CD4(+) lineage αß T cells as CD4(+) T-cell numbers improved to near normal levels but CD8(+) T cells remained severely lymphopenic. Notably, Pim1 over-expression failed to promote development and survival of any T-lineage cells other than αß T cells, as we observed complete lack of γδ, NKT, FoxP3(+) T regulatory cells and TCR-ß(+) CD8αα IELs in Pim1(Tg) γc(KO) mice. Collectively, these results uncover distinct requirements for γc signaling between CD4(+) αß T cells and all other T-lineage cells, and they identify Pim1 as a novel effector molecule sufficient to drive CD4(+) αß T-cell development and survival in the absence of γc cytokine receptor signaling.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chemokines, C/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Receptors, Cytokine/metabolism , Animals , CD8 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Cell Survival , Chemokines, C/deficiency , Forkhead Transcription Factors/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells , Proto-Oncogene Proteins c-pim-1/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , T-Lymphocytes, Regulatory
13.
Blood ; 130(23): 2448-2450, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29217698
14.
Blood ; 119(22): 5155-63, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22403258

ABSTRACT

CTLA-4 proteins contribute to the suppressor function of regulatory T cells (Tregs), but the mechanism by which they do so remains incompletely understood. In the present study, we assessed CTLA-4 protein function in both Tregs and conventional (Tconv) CD4(+) T cells. We report that CTLA-4 proteins are responsible for all 3 characteristic Treg functions of suppression, TCR hyposignaling, and anergy. However, Treg suppression and anergy only required the external domain of CTLA-4, whereas TCR hyposignaling required its internal domain. Surprisingly, TCR hyposignaling was neither required for Treg suppression nor anergy because costimulatory blockade by the external domain of CTLA-4 was sufficient for both functions. We also report that CTLA-4 proteins were localized in Tregs in submembrane vesicles that rapidly recycled to/from the cell surface, whereas CTLA-4 proteins in naive Tconv cells were retained in Golgi vesicles away from the cell membrane and had no effect on Tconv cell function. However, TCR signaling of Tconv cells released CTLA-4 proteins from Golgi retention and caused activated Tconv cells to acquire suppressor function. Therefore, the results of this study demonstrate the importance of intracellular localization for CTLA-4 protein function and reveal that CTLA-4 protein externalization imparts suppressor function to both regulatory and conventional CD4(+) T cells.


Subject(s)
CTLA-4 Antigen/immunology , Cell Membrane/immunology , Clonal Anergy/physiology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/physiology , T-Lymphocytes, Regulatory/immunology , Animals , CTLA-4 Antigen/genetics , Cell Membrane/genetics , Golgi Apparatus/genetics , Golgi Apparatus/immunology , Lymphocyte Activation/physiology , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory/cytology
15.
Int Immunol ; 25(12): 671-81, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24048955

ABSTRACT

In addition to TCR signaling, the activation and proliferation of naive T cells require CD28-mediated co-stimulation. Once engaged, CD28 is phosphorylated and can then activate signaling pathways by recruiting molecules to its YMNM motif and two PxxP motifs. In this study, we analyzed the relationship between tyrosine phosphorylation and the co-stimulatory function of CD28 in murine primary CD4(+) T cells. Tyrosine phosphorylation is decreased in CD28 where the N-terminal PxxP motif is mutated (nPA). In cells expressing nPA, activation of Akt and functional co-stimulation were decreased. In contrast, where the C-terminal PxxP motif is mutated, tyrosine phosphorylation and activation of the ERK, Akt and NF-κB were intact, but proliferation and IL-2 production were decreased. Using the Y(189) to F mutant, we also demonstrated that in naive CD4(+) T cells, tyrosine at position 189 in the YMNM motif is critical for both tyrosine phosphorylation and the functional co-stimulatory effects of CD28. This mutation did not affect unfractionated T-cell populations. Overall, our data suggest that CD28 signaling uses tyrosine phosphorylation-dependent and phosphorylation-independent pathways.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , Animals , CD28 Antigens/chemistry , CD28 Antigens/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunophenotyping , Interleukin-2/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , NF-kappa B/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-akt/metabolism , Sequence Alignment , Tyrosine/metabolism
16.
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
17.
Science ; 382(6670): 534-541, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37917689

ABSTRACT

CD8 T cell tolerance is thought to result from clonal deletion of autoreactive thymocytes before they differentiate into mature CD8 T cells in the thymus. However, we report that, in mice, CD8 T cell tolerance instead results from premature thymic eviction of immature autoreactive CD8 thymocytes into the periphery, where they differentiate into self-tolerant mature CD8 T cells. Premature thymic eviction is triggered by T cell receptor (TCR)-driven down-regulation of the transcriptional repressor Gfi1, which induces expression of sphingosine-1-phosphate receptor-1 (S1P1) on negatively selected immature CD8 thymocytes. Thus, premature thymic eviction is the basis for CD8 T cell tolerance and is the mechanism responsible for the appearance in the periphery of mature CD8 T cells bearing autoreactive TCRs that are absent from the thymus.


Subject(s)
CD8-Positive T-Lymphocytes , Clonal Deletion , Peripheral Tolerance , Thymus Gland , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Transcription Factors/metabolism , Male , Female
18.
JCI Insight ; 6(21)2021 11 08.
Article in English | MEDLINE | ID: mdl-34747370

ABSTRACT

Foxp3+ Tregs are potent immunosuppressive CD4+ T cells that are critical to maintain immune quiescence and prevent autoimmunity. Both the generation and maintenance of Foxp3+ Tregs depend on the cytokine IL-2. Hence, the expression of the IL-2 receptor α-chain (CD25) is not only considered a specific marker, but also a nonredundant requirement for Tregs. Here, we report that Foxp3+ Tregs in the small intestine (SI) epithelium, a critical barrier tissue, are exempt from such an IL-2 requirement, since they had dramatically downregulated CD25 expression, showed minimal STAT5 phosphorylation ex vivo, and were unable to respond to IL-2 in vitro. Nonetheless, SI epithelial Tregs survived and were present at the same frequency as in other lymphoid organs, and they retained potent suppressor function that was associated with high levels of CTLA-4 expression and the production of copious amounts of IL-10. Moreover, adoptive transfer experiments of Foxp3+ Tregs revealed that such IL-2-independent survival and effector functions were imposed by the SI epithelial tissue, suggesting that tissue adaptation is a mechanism that tailors the effector function and survival requirements of Foxp3+ Tregs specific to the tissue environment.


Subject(s)
Epithelium/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-2/metabolism , Intestine, Small/physiopathology , T-Lymphocytes, Regulatory/metabolism , Animals , Homeostasis , Humans , Mice
19.
iScience ; 23(8): 101421, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32791329

ABSTRACT

IL-7 receptor signaling is essential for the generation and maintenance of conventional T cells. Immunosuppressive Foxp3+ Treg cells, however, express uniquely low amounts of the IL-7-proprietary IL-7Rα so that they are impaired in IL-7 signaling. Because Treg cells depend on IL-2, the loss of IL-7Rα has been considered irrelevant for Treg cells. In contrast, here, we report that IL-7Rα downregulation is necessary to maximize IL-2R signaling. Although IL-7Rα overexpression promoted IL-7 signaling, unexpectedly, IL-2 signaling was suppressed in the same cells. Mechanistically, we found that γc, which is a receptor subunit shared by IL-7R and IL-2R, directly binds and pre-associates with IL-7Rα, thus limiting its availability for IL-2R binding. Consequently, overexpression of signaling-deficient, tailless IL-7Rα proteins inhibited IL-2R signaling, demonstrating that IL-7Rα sequesters γc and suppresses IL-2R signaling by extracellular interactions. Collectively, these results reveal a previously unappreciated regulatory mechanism of IL-2 receptor signaling that is governed by IL-7Rα abundance.

20.
Front Immunol ; 11: 1216, 2020.
Article in English | MEDLINE | ID: mdl-32612609

ABSTRACT

MHC-independent αßTCRs (TCRs) recognize conformational epitopes on native self-proteins and arise in mice lacking both MHC and CD4/CD8 coreceptor proteins. Although naturally generated in the thymus, these TCRs resemble re-engineered therapeutic chimeric antigen receptor (CAR) T cells in their specificity for MHC-independent ligands. Here we identify naturally arising MHC-independent TCRs reactive to three native self-proteins (CD48, CD102, and CD155) involved in cell adhesion. We report that naturally arising MHC-independent TCRs require high affinity TCR-ligand engagements in the thymus to signal positive selection and that high affinity positive selection generates a peripheral TCR repertoire with limited diversity and increased self-reactivity. We conclude that the affinity of TCR-ligand engagements required to signal positive selection in the thymus inversely determines the diversity and self-tolerance of the mature TCR repertoire that is selected.


Subject(s)
Clonal Selection, Antigen-Mediated , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Self Tolerance/immunology , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/physiology , Animals , Antigens, CD/metabolism , CD8 Antigens/immunology , Cell Adhesion Molecules/metabolism , Ligands , Lymphocyte Function-Associated Antigen-1/metabolism , Major Histocompatibility Complex/genetics , Mice , Mice, Knockout , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL