Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
2.
Science ; 382(6670): 534-541, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37917689

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos , Deleção Clonal , Tolerância Periférica , Timo , Animais , Camundongos , Linfócitos T CD8-Positivos/imunologia , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Timo/citologia , Timo/imunologia , Fatores de Transcrição/metabolismo , Masculino , Feminino
3.
Nat Immunol ; 24(4): 637-651, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36959291

RESUMO

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.


Assuntos
Deleção Clonal , Timócitos , Timócitos/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Timo/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Linfócitos T Reguladores/metabolismo
4.
Front Immunol ; 13: 953160, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35911724

RESUMO

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.


Assuntos
Receptores de Antígenos de Linfócitos T , Linfócitos T , Antígenos/metabolismo
5.
Nat Immunol ; 23(5): 731-742, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523960

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Diferenciação Celular , Linhagem da Célula/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Timo/metabolismo
6.
Front Immunol ; 11: 1216, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32612609

RESUMO

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.


Assuntos
Seleção Clonal Mediada por Antígeno , Complexo Principal de Histocompatibilidade/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Tolerância a Antígenos Próprios/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/fisiologia , Animais , Antígenos CD/metabolismo , Antígenos CD8/imunologia , Moléculas de Adesão Celular/metabolismo , Ligantes , Antígeno-1 Associado à Função Linfocitária/metabolismo , Complexo Principal de Histocompatibilidade/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores Virais/imunologia
7.
J Immunol ; 204(12): 3351-3359, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32321756

RESUMO

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.


Assuntos
Complexo Principal de Histocompatibilidade/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores Virais/metabolismo , Animais , Células HEK293 , Humanos , Ligantes , Camundongos , Peptídeos/metabolismo , Poliovirus/metabolismo , Ligação Proteica , Domínios Proteicos , Timócitos/metabolismo , Recombinação V(D)J/fisiologia
8.
J Exp Med ; 216(8): 1749-1761, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31201207

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Receptores CXCR4/metabolismo , Timócitos/metabolismo , Timo/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Antígenos CD8/metabolismo , Diferenciação Celular/genética , Quimiocina CXCL12/metabolismo , Células Epiteliais/metabolismo , Humanos , Linfopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores CXCR4/genética , Transdução de Sinais/genética
9.
Nat Commun ; 10(1): 1019, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30833553

RESUMO

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.


Assuntos
Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/imunologia , Timo/imunologia , Sequência de Aminoácidos , Animais , Regiões Determinantes de Complementaridade/genética , Ativação Linfocitária , Complexo Principal de Histocompatibilidade/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/genética , Análise de Sequência de Proteína , Linfócitos T/imunologia , Linfócitos T/metabolismo , Recombinação V(D)J
10.
Nat Immunol ; 19(12): 1379-1390, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30420628

RESUMO

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.


Assuntos
Mecanotransdução Celular/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Timócitos/imunologia , Timo/imunologia , Animais , Deleção Clonal/imunologia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Timócitos/metabolismo
11.
Nat Commun ; 9(1): 3749, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30218105

RESUMO

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.


Assuntos
Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/genética , Lectinas Tipo C/genética , Linfopoese/genética , Células T Matadoras Naturais/citologia , Receptores de Lisoesfingolipídeo/genética , Subpopulações de Linfócitos T/citologia , Timócitos/citologia , Animais , Antígeno CD24/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Receptores de Esfingosina-1-Fosfato , Subpopulações de Linfócitos T/metabolismo , Timócitos/metabolismo
12.
Cell Rep ; 24(1): 117-129, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29972774

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/citologia , Diferenciação Celular , Proteínas Nucleares/metabolismo , Timócitos/citologia , Fatores de Transcrição/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Deleção de Genes , Glicólise , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T Reguladores/metabolismo , Timócitos/metabolismo
13.
Nat Immunol ; 18(11): 1218-1227, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28945245

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linhagem da Célula/imunologia , Citocinas/imunologia , Timo/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/imunologia , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Citocinas/metabolismo , Citometria de Fluxo , Expressão Gênica/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Timócitos/imunologia , Timócitos/metabolismo , Timo/citologia , Timo/metabolismo
14.
Elife ; 62017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28731407

RESUMO

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.


Assuntos
Sequência Conservada , Variação Genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Animais , Análise por Conglomerados , Humanos , Camundongos , Homologia de Sequência
15.
Nat Immunol ; 17(12): 1415-1423, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27668801

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Seleção Clonal Mediada por Antígeno , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Timo/imunologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Citocinas/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Fatores de Transcrição
17.
Nat Immunol ; 16(10): 1069-76, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26301566

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Proliferação de Células , Citometria de Fluxo , Camundongos , Peptídeos/imunologia , Timo/enzimologia
18.
Nat Immunol ; 16(5): 517-24, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25848867

RESUMO

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.


Assuntos
Citocinas/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , MicroRNAs/metabolismo , Células T Matadoras Naturais/fisiologia , Timócitos/fisiologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Citotoxicidade Imunológica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica , Ligação Proteica , Processamento Pós-Transcricional do RNA , Tretinoína/metabolismo , Regulação para Cima , Vitamina D/metabolismo
19.
Sci Signal ; 7(340): ra83, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25161318

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/imunologia , Janus Quinase 1/genética , MicroRNAs/metabolismo , RNA Mensageiro/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Interleucina-7/metabolismo , Transdução de Sinais/imunologia , Animais , Western Blotting , Citometria de Fluxo , Imunoprecipitação , Janus Quinase 1/biossíntese , Luciferases , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interleucina-7/antagonistas & inibidores
20.
Nat Immunol ; 15(7): 638-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24880459

RESUMO

Lineage fate in the thymus is determined by mutually exclusive expression of the transcription factors ThPOK and Runx3, with ThPOK imposing the CD4(+) lineage fate and Runx3 promoting the CD8(+) lineage fate. While it is known that cytokine signals induce thymocytes to express Runx3, it is not known how ThPOK prevents thymocytes from expressing Runx3 and adopting the CD8(+) lineage fate, nor is it understood why ThPOK itself imposes the CD4(+) lineage fate on thymocytes. We now report that genes encoding members of the SOCS (suppressor of cytokine signaling) family are critical targets of ThPOK and that their induction by ThPOK represses Runx3 expression and promotes the CD4(+) lineage fate. Thus, induction of SOCS-encoding genes is the main mechanism by which ThPOK imposes the CD4(+) lineage fate in the thymus.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Linhagem da Célula , Subunidade alfa 3 de Fator de Ligação ao Core/fisiologia , Proteínas Supressoras da Sinalização de Citocina/genética , Fatores de Transcrição/fisiologia , Animais , Linfócitos T CD8-Positivos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA