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










Publication year range
2.
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
3.
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
4.
Front Immunol ; 13: 953160, 2022.
Article in English | MEDLINE | ID: mdl-35911724

ABSTRACT

Understanding the generation of an MHC-restricted T cell repertoire is the cornerstone of modern T cell immunology. The unique ability of αßT cells to only recognize peptide antigens presented by MHC molecules but not conformational antigens is referred to as MHC restriction. How MHC restriction is imposed on a very large T cell receptor (TCR) repertoire is still heavily debated. We recently proposed the selection model, which posits that newly re-arranged TCRs can structurally recognize a wide variety of antigens, ranging from peptides presented by MHC molecules to native proteins like cell surface markers. However, on a molecular level, the sequestration of the essential tyrosine kinase Lck by the coreceptors CD4 and CD8 allows only MHC-restricted TCRs to signal. In the absence of Lck sequestration, MHC-independent TCRs can signal and instruct the generation of mature αßT cells that can recognize native protein ligands. The selection model thus explains how only MHC-restricted TCRs can signal and survive thymic selection. In this review, we will discuss the genetic evidence that led to our selection model. We will summarize the selection mechanism and structural properties of MHC-independent TCRs and further discuss the various non-MHC ligands we have identified.


Subject(s)
Receptors, Antigen, T-Cell , T-Lymphocytes , Antigens/metabolism
5.
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
6.
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
7.
J Immunol ; 204(12): 3351-3359, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32321756

ABSTRACT

During normal T cell development in the thymus, αß TCRs signal immature thymocytes to differentiate into mature T cells by binding to peptide-MHC ligands together with CD4/CD8 coreceptors. Conversely, in MHC and CD4/CD8 coreceptor-deficient mice, the thymus generates mature T cells expressing MHC-independent TCRs that recognize native conformational epitopes rather than linear antigenic-peptides presented by MHC. To date, no structural information of MHC-independent TCRs is available, and their structural recognition of non-MHC ligand remains unknown. To our knowledge in this study, we determined the first structures of two murine MHC-independent TCRs (A11 and B12A) that bind with high nanomolar affinities to mouse adhesion receptor CD155. Solution binding demonstrated the Vαß-domain is responsible for MHC-independent B12A recognition of its ligand. Analysis of A11 and B12A sequences against various MHC-restricted and -independent TCR sequence repertoires showed that individual V-genes of A11 and B12A did not exhibit preference against MHC-restriction. Likewise, CDR3 alone did not discriminate against MHC binding, suggesting VDJ recombination together with Vα/Vß pairing determine their MHC-independent specificity for CD155. The structures of A11 and B12A TCR are nearly identical to those of MHC-restricted TCR, including the conformations of CDR1 and 2. Mutational analysis, together with negative-staining electron microscopy images, showed that the CDR regions of A11 and B12A recognized epitopes on D1 domain of CD155, a region also involved in CD155 binding to poliovirus and Tactile in human. Taken together, MHC-independent TCRs adopt canonical TCR structures to recognize native Ags, highlighting the importance of thymic selection in determining TCR ligand specificity.


Subject(s)
Major Histocompatibility Complex/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Virus/metabolism , Animals , HEK293 Cells , Humans , Ligands , Mice , Peptides/metabolism , Poliovirus/metabolism , Protein Binding , Protein Domains , Thymocytes/metabolism , V(D)J Recombination/physiology
8.
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
9.
Nat Commun ; 10(1): 1019, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30833553

ABSTRACT

The αß T cell receptor (TCR) repertoire on mature T cells is selected in the thymus, but the basis for thymic selection of MHC-restricted TCRs from a randomly generated pre-selection repertoire is not known. Here we perform comparative repertoire sequence analyses of pre-selection and post-selection TCR from multiple MHC-sufficient and MHC-deficient mouse strains, and find that MHC-restricted and MHC-independent TCRs are primarily distinguished by features in their non-germline CDR3 regions, with many pre-selection CDR3 sequences not compatible with MHC-binding. Thymic selection of MHC-independent TCR is largely unconstrained, but the selection of MHC-specific TCR is restricted by both CDR3 length and specific amino acid usage. MHC-restriction disfavors TCR with CDR3 longer than 13 amino acids, limits positively charged and hydrophobic amino acids in CDR3ß, and clonally deletes TCRs with cysteines in their CDR3 peptide-binding regions. Together, these MHC-imposed structural constraints form the basis to shape VDJ recombination sequences into MHC-restricted repertoires.


Subject(s)
Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Thymus Gland/immunology , Amino Acid Sequence , Animals , Complementarity Determining Regions/genetics , Lymphocyte Activation , Major Histocompatibility Complex/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/genetics , Sequence Analysis, Protein , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , V(D)J Recombination
10.
Nat Immunol ; 19(12): 1379-1390, 2018 12.
Article in English | MEDLINE | ID: mdl-30420628

ABSTRACT

The T cell antigen receptor (TCR) expressed on thymocytes interacts with self-peptide major histocompatibility complex (pMHC) ligands to signal apoptosis or survival. Here, we found that negative-selection ligands induced thymocytes to exert forces on the TCR and the co-receptor CD8 and formed cooperative TCR-pMHC-CD8 trimolecular 'catch bonds', whereas positive-selection ligands induced less sustained thymocyte forces on TCR and CD8 and formed shorter-lived, independent TCR-pMHC and pMHC-CD8 bimolecular 'slip bonds'. Catch bonds were not intrinsic to either the TCR-pMHC or the pMHC-CD8 arm of the trans (cross-junctional) heterodimer but resulted from coupling of the extracellular pMHC-CD8 interaction to the intracellular interaction of CD8 with TCR-CD3 via associated kinases to form a cis (lateral) heterodimer capable of inside-out signaling. We suggest that the coupled trans-cis heterodimeric interactions form a mechanotransduction loop that reinforces negative-selection signaling that is distinct from positive-selection signaling in the thymus.


Subject(s)
Mechanotransduction, Cellular/immunology , Receptors, Antigen, T-Cell/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Clonal Deletion/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Thymocytes/metabolism
11.
Nat Commun ; 9(1): 3749, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30218105

ABSTRACT

While CD69 may regulate thymocyte egress by inhibiting S1P1 expression, CD69 expression is not thought to be required for normal thymocyte development. Here we show that CD69 is in fact specifically required for the differentiation of mature NKT2 cells, which do not themselves express CD69. Mechanistically, CD69 expression is required on CD24+ PLZFhi innate precursors for their retention in the thymus and completion of their differentiation into mature NKT2 cells. By contrast, CD69-deficient CD24+ PLZFhi innate precursors express S1P1 and prematurely exit the thymus, while S1P1 inhibitor treatment of CD69-deficient mice retains CD24+ PLZFhi innate precursors in the thymus and restores NKT2 cell differentiation. Thus, CD69 prevents S1P1 expression on CD24+ PLZFhi innate precursor cells from aborting NKT2 differentiation in the thymus. This study reveals the importance of CD69 to prolong the thymic residency time of developing immature precursors for proper differentiation of a T cell subset.


Subject(s)
Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Lectins, C-Type/genetics , Lymphopoiesis/genetics , Natural Killer T-Cells/cytology , Receptors, Lysosphingolipid/genetics , T-Lymphocyte Subsets/cytology , Thymocytes/cytology , Animals , CD24 Antigen/metabolism , Cell Differentiation , Gene Expression Regulation , Mice , Mice, Knockout , Natural Killer T-Cells/metabolism , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Sphingosine-1-Phosphate Receptors , T-Lymphocyte Subsets/metabolism , Thymocytes/metabolism
12.
Cell Rep ; 24(1): 117-129, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29972774

ABSTRACT

T cell differentiation in the thymus proceeds in an ordered sequence of developmental events characterized by variable expression of CD4 and CD8 coreceptors. Here, we report that immature single-positive (ISP) thymocytes are molecularly distinct from all other T cell populations in the thymus in their expression of a gene profile that is dependent on the transcription factor BRD4. Conditional deletion of BRD4 at various stages of thymic differentiation reveals that BRD4 selectively regulates the further differentiation of ISPs by targeting cell cycle and metabolic pathways, but it does not affect the extensive proliferation that results in the generation of ISPs. These studies lead to the conclusion that the ISP subpopulation is not a hybrid transitional state but a molecularly distinct subpopulation that is selectively dependent on BRD4.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Nuclear Proteins/metabolism , Thymocytes/cytology , Transcription Factors/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Gene Deletion , Glycolysis , Mice, Knockout , Natural Killer T-Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/metabolism , Thymocytes/metabolism
13.
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
14.
Elife ; 62017 07 21.
Article in English | MEDLINE | ID: mdl-28731407

ABSTRACT

Diversity of T cell receptor (TCR) repertoires, generated by somatic DNA rearrangements, is central to immune system function. However, the level of sequence similarity of TCR repertoires within and between species has not been characterized. Using network analysis of high-throughput TCR sequencing data, we found that abundant CDR3-TCRß sequences were clustered within networks generated by sequence similarity. We discovered a substantial number of public CDR3-TCRß segments that were identical in mice and humans. These conserved public sequences were central within TCR sequence-similarity networks. Annotated TCR sequences, previously associated with self-specificities such as autoimmunity and cancer, were linked to network clusters. Mechanistically, CDR3 networks were promoted by MHC-mediated selection, and were reduced following immunization, immune checkpoint blockade or aging. Our findings provide a new view of T cell repertoire organization and physiology, and suggest that the immune system distributes its TCR sequences unevenly, attending to specific foci of reactivity.


Subject(s)
Conserved Sequence , Genetic Variation , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Cluster Analysis , Humans , Mice , Sequence Homology
15.
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
17.
Nat Immunol ; 16(10): 1069-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26301566

ABSTRACT

In the thymus, low-affinity T cell antigen receptor (TCR) engagement facilitates positive selection of a useful T cell repertoire. Here we report that TCR responsiveness of mature CD8(+) T cells is fine tuned by their affinity for positively selecting peptides in the thymus and that optimal TCR responsiveness requires positive selection on major histocompatibility complex class I-associated peptides produced by the thymoproteasome, which is specifically expressed in the thymic cortical epithelium. Thymoproteasome-independent positive selection of monoclonal CD8(+) T cells results in aberrant TCR responsiveness, homeostatic maintenance and immune responses to infection. These results demonstrate a novel aspect of positive selection, in which TCR affinity for positively selecting peptides produced by thymic epithelium determines the subsequent antigen responsiveness of mature CD8(+) T cells in the periphery.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Proteasome Endopeptidase Complex/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Cell Proliferation , Flow Cytometry , Mice , Peptides/immunology , Thymus Gland/enzymology
18.
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
19.
Sci Signal ; 7(340): ra83, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25161318

ABSTRACT

T cell receptor (TCR)-mediated inhibition of interleukin-7 (IL-7) signaling is important for lineage fate determination in the thymus and for T cell survival in the periphery because uninterrupted IL-7 signaling results in T cell death. The initial event in IL-7 signaling is the transactivation of Janus kinases 1 and 3 (Jak1 and Jak3), which are associated with the cytosolic tails of the IL-7 receptor α chain (IL-7Rα) and the γc subunit, the two cell surface proteins that constitute IL-7R. We found that Jak1 is a highly unstable protein with a half-life of only 1.5 hours, so that continuous Jak1 protein synthesis is required to maintain Jak1 protein in sufficient abundance to support IL-7 signaling. However, we also found that Jak1 protein synthesis was acutely reduced by TCR-responsive microRNAs in the miR-17 family, which targeted Jak1 mRNA (messenger RNA) to inhibit its translation. Thus, this study identifies a molecular mechanism by which TCR engagement acutely disrupts IL-7 signaling.


Subject(s)
Gene Expression Regulation/immunology , Janus Kinase 1/genetics , MicroRNAs/metabolism , RNA, Messenger/antagonists & inhibitors , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-7/metabolism , Signal Transduction/immunology , Animals , Blotting, Western , Flow Cytometry , Immunoprecipitation , Janus Kinase 1/biosynthesis , Luciferases , Mice , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-7/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...