Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
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 ; 23(5): 731-742, 2022 05.
Article in English | MEDLINE | ID: mdl-35523960

ABSTRACT

T cell specificity and function are linked during development, as MHC-II-specific TCR signals generate CD4 helper T cells and MHC-I-specific TCR signals generate CD8 cytotoxic T cells, but the basis remains uncertain. We now report that switching coreceptor proteins encoded by Cd4 and Cd8 gene loci functionally reverses the T cell immune system, generating CD4 cytotoxic and CD8 helper T cells. Such functional reversal reveals that coreceptor proteins promote the helper-lineage fate when encoded by Cd4, but promote the cytotoxic-lineage fate when encoded in Cd8-regardless of the coreceptor proteins each locus encodes. Thus, T cell lineage fate is determined by cis-regulatory elements in coreceptor gene loci and is not determined by the coreceptor proteins they encode, invalidating coreceptor signal strength as the basis of lineage fate determination. Moreover, we consider that evolution selected the particular coreceptor proteins that Cd4 and Cd8 gene loci encode to avoid generating functionally reversed T cells because they fail to promote protective immunity against environmental pathogens.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Cell Lineage/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/metabolism
3.
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
4.
J Exp Med ; 216(8): 1749-1761, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31201207

ABSTRACT

Preselection thymocytes are normally retained in the thymic cortex, but the mechanisms responsible remain incompletely understood. We now report that deletion of genes encoding the E-protein transcription factors E2A and HEB disorders chemokine receptor expression on developing thymocytes to allow escape of preselection TCR-CD8+ thymocytes into the periphery. We document that CXCR4 expression normally anchors preselection thymocytes to the thymic cortex via interaction with its ligand CXCL12 on cortical thymic epithelial cells, and that disruption of CXCR4-CXCL12 engagements release preselection thymocytes from the thymic cortex. We further document that CXCR4 expression must be extinguished by TCR-mediated positive selection signals to allow migration of TCR-signaled thymocytes out of the thymic cortex into the medulla. Thus, E-protein transcription factors regulate the ordered expression pattern of chemokine receptors on developing thymocytes, and the interaction of the chemokine receptor CXCR4 with its ligand adheres TCR-unsignaled preselection thymocytes to the thymic cortex.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Receptors, CXCR4/metabolism , Thymocytes/metabolism , Thymus Gland/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CD8 Antigens/metabolism , Cell Differentiation/genetics , Chemokine CXCL12/metabolism , Epithelial Cells/metabolism , Humans , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Receptors, CXCR4/genetics , Signal Transduction/genetics
5.
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
6.
Nat Immunol ; 16(5): 517-24, 2015 May.
Article in English | MEDLINE | ID: mdl-25848867

ABSTRACT

Lethal-7 (let-7) microRNAs (miRNAs) are the most abundant miRNAs in the genome, but their role in developing thymocytes is unclear. We found that let-7 miRNAs targeted Zbtb16 mRNA, which encodes the lineage-specific transcription factor PLZF, to post-transcriptionally regulate PLZF expression and thereby the effector functions of natural killer T cells (NKT cells). Dynamic upregulation of let-7 miRNAs during the development of NKT thymocytes downregulated PLZF expression and directed their terminal differentiation into interferon-γ (IFN-γ)-producing NKT1 cells. Without upregulation of let-7 miRNAs, NKT thymocytes maintained high PLZF expression and terminally differentiated into interleukin 4 (IL-4)-producing NKT2 cells or IL-17-producing NKT17 cells. Upregulation of let-7 miRNAs in developing NKT thymocytes was signaled by IL-15, vitamin D and retinoic acid. Such targeting of a lineage-specific transcription factor by miRNA represents a previously unknown level of developmental regulation in the thymus.


Subject(s)
Cytokines/metabolism , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Natural Killer T-Cells/physiology , Thymocytes/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cytotoxicity, Immunologic/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Promyelocytic Leukemia Zinc Finger Protein , Protein Binding , RNA Processing, Post-Transcriptional , Tretinoin/metabolism , Up-Regulation , Vitamin D/metabolism
7.
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
8.
J Exp Med ; 209(12): 2263-76, 2012 Nov 19.
Article in English | MEDLINE | ID: mdl-23109710

ABSTRACT

The thymus generates T cells with diverse specificities and functions. To assess the contribution of cytokine receptors to the differentiation of T cell subsets in the thymus, we constructed conditional knockout mice in which IL-7Rα or common cytokine receptor γ chain (γ(c)) genes were deleted in thymocytes just before positive selection. We found that γ(c) expression was required to signal the differentiation of MHC class I (MHC-I)-specific thymocytes into CD8(+) cytotoxic lineage T cells and into invariant natural killer T cells but did not signal the differentiation of MHC class II (MHC-II)-specific thymocytes into CD4(+) T cells, even into regulatory Foxp3(+)CD4(+) T cells which require γ(c) signals for survival. Importantly, IL-7 and IL-15 were identified as the cytokines responsible for CD8(+) cytotoxic T cell lineage specification in vivo. Additionally, we found that small numbers of aberrant CD8(+) T cells expressing Runx3d could arise without γ(c) signaling, but these cells were developmentally arrested before expressing cytotoxic lineage genes. Thus, γ(c)-transduced cytokine signals are required for cytotoxic lineage specification in the thymus and for inducing the differentiation of MHC-I-selected thymocytes into functionally mature T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-7/metabolism , Receptors, Cytokine/genetics , Thymus Gland/cytology , Animals , Flow Cytometry , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-15/genetics , Receptors, Interleukin-7/genetics
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.
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
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 ; 177(10): 6613-25, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082573

ABSTRACT

TCR signals drive thymocyte development, but it remains controversial what impact, if any, the intensity of those signals have on T cell differentiation in the thymus. In this study, we assess the impact of CD8 coreceptor signal strength on positive selection and CD4/CD8 lineage choice using novel gene knockin mice in which the endogenous CD8alpha gene has been re-engineered to encode the stronger signaling cytoplasmic tail of CD4, with the re-engineered CD8alpha gene referred to as CD8.4. We found that stronger signaling CD8.4 coreceptors specifically improved the efficiency of CD8-dependent positive selection and quantitatively increased the number of MHC class I (MHC-I)-specific thymocytes signaled to differentiate into CD8+ T cells, even for thymocytes expressing a single, transgenic TCR. Importantly, however, stronger signaling CD8.4 coreceptors did not alter the CD8 lineage choice of any MHC-I-specific thymocytes, even MHC-I-specific thymocytes expressing the high-affinity F5 transgenic TCR. This study documents in a physiologic in vivo model that coreceptor signal strength alters TCR-signaling thresholds for positive selection and so is a major determinant of the CD4:CD8 ratio, but it does not influence CD4/CD8 lineage choice.


Subject(s)
CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Models, Immunological , Signal Transduction/immunology , Animals , CD4 Antigens/biosynthesis , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/biosynthesis , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Female , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Signal Transduction/genetics , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...