Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci Alliance ; 6(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37193606

RESUMO

The genome organizer, special AT-rich binding protein-1 (SATB1), functions to globally regulate gene networks during primary T cell development and plays a pivotal role in lineage specification in CD4+ helper-, CD8+ cytotoxic-, and FOXP3+ regulatory-T cell subsets. However, it remains unclear how Satb1 gene expression is controlled, particularly in effector T cell function. Here, by using a novel reporter mouse strain expressing SATB1-Venus and genome editing, we have identified a cis-regulatory enhancer, essential for maintaining Satb1 expression specifically in TH2 cells. This enhancer is occupied by STAT6 and interacts with Satb1 promoters through chromatin looping in TH2 cells. Reduction of Satb1 expression, by the lack of this enhancer, resulted in elevated IL-5 expression in TH2 cells. In addition, we found that Satb1 is induced in activated group 2 innate lymphoid cells (ILC2s) through this enhancer. Collectively, these results provide novel insights into how Satb1 expression is regulated in TH2 cells and ILC2s during type 2 immune responses.


Assuntos
Proteínas de Ligação à Região de Interação com a Matriz , Animais , Camundongos , Proteínas de Ligação à Região de Interação com a Matriz/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Imunidade Inata , Linfócitos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Diferenciação Celular
2.
J Immunol ; 210(11): 1728-1739, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37074186

RESUMO

Posttranslational modification, such as phosphorylation, is an important biological event that modulates and diversifies protein function. Bcl11b protein is a zinc-finger transcription factor that plays a crucial role in early T cell development and the segregation of T cell subsets. Bcl11b possesses at least 25 serine/threonine (S/T) residues that can be phosphorylated upon TCR stimulation. To understand the physiological relevance of the phosphorylation on Bcl11b protein, we replaced S/T residues with alanine (A) by targeting murine Bcl11b gene in embryonic stem cells. By combinational targeting of exons 2 and 4 in the Bcl11b gene, we generated a mouse strain, Bcl11b-phosphorylation site mutation mice, in which 23 S/T residues were replaced with A residues. Such extensive manipulation left only five putative phosphorylated residues, two of which were specific for mutant protein, and resulted in reduced amounts of Bcl11b protein. However, primary T cell development in the thymus, as well as the maintenance of peripheral T cells, remained intact even after loss of major physiological phosphorylation. In addition, in vitro differentiation of CD4+ naive T cells into effector Th cell subsets-Th1, Th2, Th17, and regulatory T-was comparable between wild-type and Bcl11b-phosphorylation site mutation mice. These findings indicate that the physiological phosphorylation on major 23 S/T residues in Bcl11b is dispensable for Bcl11b functions in early T cell development and effector Th cell differentiation.


Assuntos
Proteínas Repressoras , Proteínas Supressoras de Tumor , Animais , Camundongos , Fosforilação , Proteínas Repressoras/genética , Proteínas Supressoras de Tumor/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Diferenciação Celular , Processamento de Proteína Pós-Traducional , Serina/genética , Serina/metabolismo , Treonina/genética , Treonina/metabolismo
3.
Life Sci Alliance ; 3(2)2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31980555

RESUMO

Temporal down-regulation of the CD8 co-receptor after receiving positive-selection signals has been proposed to serve as an important determinant to segregate helper versus cytotoxic lineages by generating differences in the duration of TCR signaling between MHC-I and MHC-II selected thymocytes. By contrast, little is known about whether CD8 also modulates TCR signaling engaged by the non-classical MHC-I-like molecule, CD1d, during development of invariant natural killer T (iNKT) cells. Here, we show that constitutive transgenic CD8 expression resulted in enhanced differentiation of innate memory-like CD8+ thymocytes in both a cell-intrinsic and cell-extrinsic manner, the latter being accomplished by an increase in the IL-4-producing iNKT2 subset. Skewed iNKT2 differentiation requires cysteine residues in the intracellular domain of CD8α that are essential for transmitting cellular signaling. Collectively, these findings shed a new light on the relevance of CD8 down-regulation in shaping the balance of iNKT-cell subsets by modulating TCR signaling.


Assuntos
Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/genética , Imunidade Inata , Células T Matadoras Naturais/imunologia , Animais , Antígenos CD8/genética , Diferenciação Celular/imunologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/genética , Timócitos/imunologia , Transfecção
4.
Front Immunol ; 10: 409, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30915074

RESUMO

CD8 expression in T lymphocytes is tightly regulated by the activity of at least six Cd8 enhancers (E8I-E8VI), however their complex developmental stage-, subset-, and lineage-specific interplays are incompletely understood. Here we analyzed ATAC-seq data on the Immunological Genome Project database and identified a similar developmental regulation of chromatin accessibility of a subregion of E8I, designated E8I-core, and of E8VI. Loss of E8I-core led to a similar reduction in CD8 expression in naïve CD8+ T cells and in IELs as observed in E8I-/- mice, demonstrating that we identified the core enhancer region of E8I. While E8VI-/- mice displayed a mild reduction in CD8 expression levels on CD8SP thymocytes and peripheral CD8+ T cells, CD8 levels were further reduced upon combined deletion of E8I-core and E8VI. Moreover, activated E8I-core-/-E8VI-/- CD8+ T cells lost CD8 expression to a greater degree than E8I-core-/- and E8VI-/- CD8+ T cells, suggesting that the combined activity of both enhancers is required for establishment and maintenance of CD8 expression before and after TCR activation. Finally, we observed a severe reduction of CD4 CTLs among the TCRß+CD4+ IEL population in E8I-core-/- but not E8VI-/- mice. Such a reduction was not observed in Cd8a-/- mice, indicating that E8I-core controls the generation of CD4 CTLs independently of its role in Cd8a gene regulation. Further, the combined deletion of E8I-core and E8VI restored CD4 CTL subsets, suggesting an antagonistic function of E8VI in the generation of CD4 CTLs. Together, our study demonstrates a complex utilization and interplay of E8I-core and E8VI in regulating CD8 expression in cytotoxic lineage T cells and in IELs. Moreover, we revealed a novel E8I-mediated regulatory mechanism controlling the generation of intestinal CD4 CTLs.


Assuntos
Antígenos CD8/biossíntese , Regulação da Expressão Gênica/imunologia , Mucosa Intestinal/imunologia , Linfócitos Intraepiteliais/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos Intraepiteliais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Citotóxicos/metabolismo
5.
Nat Commun ; 9(1): 3593, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30185787

RESUMO

An intronic silencer, S4, in the Cd4 gene has been shown to be responsible for the helper-lineage-specific expression of CD4; S4 requires Runx complex binding to exert its silencer function against the enhancer-mediated Cd4 activation by modulating the epigenetic state of the Cd4 gene. Here we identify a late-acting maturation enhancer. Bcl11b plays essential roles for activation of both the early-acting proximal enhancer and maturation enhancer of Cd4. Notably, Runx complexes suppress these enhancers by distinct mechanisms. Whereas repression of the proximal enhancer depends on the S4 silencer, the maturation enhancer is repressed by Runx in the absence of S4. Moreover, ThPOK, known to antagonize S4-mediated Cd4 repression, assists Runx complexes to restrain maturation enhancer activation. Distinct modes of S4 silencer action upon distinct enhancers thus unravel a pathway that restricts CD4 expression to helper-lineage cells by silencer-independent and Runx-dependent repression of maturation enhancer activity in cytotoxic-lineage cells.


Assuntos
Antígenos CD4/genética , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas Repressoras/metabolismo , Linfócitos T Auxiliares-Indutores/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Antígenos CD4/metabolismo , Células Cultivadas , Quimera , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Íntrons/genética , Camundongos , Camundongos Transgênicos , Proteínas Repressoras/genética , Elementos Silenciadores Transcricionais/fisiologia , Linfócitos T Citotóxicos/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética
6.
Data Brief ; 17: 1180-1183, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29876477

RESUMO

The data presented here are related to the research article entitled "Loss of Eed leads to lineage instability and increased CD8 expression of mouse CD4+ T cells upon TGFß signaling" [1]. The cited research article investigates the molecular mechanism of CD8α upregulation observed in Eed-deficient (∆Eed) CD4+ T cells upon activation in the presence of TGFß. This data report describes the effect of retinoic acid (RA) and/or anti-interferon-gamma (IFNγ) antibody supplementation on up-regulation of CD8α and Foxp3 in ∆Eed CD4+ T cells, the effect of dose or timing of TGFß treatment on CD4+ T cell identity of ∆Eed, adding further information regarding the conditions that induces CD8α, and mRNA expression changes of genes encoding polycomb repressive complex 2 (PRC2) subunits by TGFß treatment.

7.
Proc Natl Acad Sci U S A ; 115(13): 3440-3445, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29531042

RESUMO

During differentiation of CD4+CD8+ double-positive (DP) thymocytes into the CD4-CD8+ single-positive (CD8SP) thymocytes committed to the cytotoxic T cell lineage, Cd8a transcription is temporally terminated after positive selection and is subsequently reinitiated, a process known as coreceptor reversal. Despite the identification of a transcriptional enhancer in the Cd8a gene that directs reporter transgene expression specifically in CD8SP thymocytes, the molecular mechanisms controlling reactivation of the Cd8a gene are not fully understood. Here, we show that, after positive selection, hCD2 reporter expression from the Cd8a locus, which was generated by insertion of hCD2 cDNA into the first exon of the Cd8a gene, requires the incorporation of intron sequences into the hCD2 transcript. The presence of polyadenylation signals after hCD2 cDNA inhibited hCD2 expression in mature CD8+ T cells, whereas hCD2 expression in DP thymocytes recapitulated the Cd8a expression. Incorporation of the endogenous short intron structure and heterologous intron structure of the Cd4 locus restored hCD2 expression in mature CD8+ T cells in a variegated manner. Interestingly, stage-specific DNA demethylation was impaired in Cd8a reporter alleles that failed to express hCD2 in CD8+ T cells, and intron sequences lacking RNA splicing signals still restored hCD2 expression. These observations indicate that "intron-mediated enhancement" is involved in a stage-specific reactivation of the Cd8a locus harboring hCD2 cDNA. However, the Cd8a gene was transcribed in mature CD8+ T cells, albeit at a lower level, from a mutant Cd8a locus lacking intron structures, suggesting that protein-coding sequences in transcripts affect sensitivity to intron-mediated enhancement.


Assuntos
Antígenos CD4/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Íntrons , Linfócitos T Citotóxicos/metabolismo , Timócitos/metabolismo , Animais , Antígenos CD4/genética , Linfócitos T CD4-Positivos/citologia , Antígenos CD8/genética , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular , Células Cultivadas , Camundongos , Splicing de RNA , Timócitos/citologia
8.
Mol Immunol ; 94: 140-152, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29310022

RESUMO

Tri-methylation of lysine 27 on histone H3 (H3K27me3) is a repressive epigenetic modification catalyzed by polycomb repressive complex 2 (PRC2) that is required for proper cell fate determination as well as cellular function. Numerous studies have been performed to elucidate the role of PRC2 in T-cell differentiation and function; however, its role in the regulation of T-helper (Th) subset differentiation and identity has not been fully explored. Here, we report that Eed, an essential subunit of PRC2, is crucial to maintain the identity of CD4+ T cells under TGFß-induced regulatory T cell (Treg)-polarizing conditions. Mouse CD4+ T cells lacking Eed exhibited unstable CD4 expression upon TCR stimulation in vitro. Helper lineage instability was further augmented by Treg-polarizing conditions, leading to the immense up-regulation of CD8α as well as other molecules, resembling CD4+ CD8αα+ intraepithelial lymphocyte (DP-IEL) differentiation. Genetic studies suggested that the altered balance between transcription factors T-bet, Runx3, and Th-POK underlies the induction of the DP-IEL-like phenotype in Eed-deficient CD4+ cells. Furthermore, comparison to Th1- and Th17-polarizing conditions indicated that cooperation between Smad3 and the T-bet-Runx3 axis facilitated by the loss of H3K27me3 is crucial for phenotype induction. Collectively, our results provide insight into the molecular mechanism that maintains and regulates the proper cellular response upon TGFß signaling in CD4+ T cells.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Antígenos CD8/genética , Linhagem da Célula/genética , Complexo Repressor Polycomb 2/genética , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos CD8/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Células Cultivadas , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Transdução de Sinais/imunologia
9.
J Exp Med ; 215(2): 595-610, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29343500

RESUMO

Multipotent hematopoietic progenitors must acquire thymus-homing capacity to initiate T lymphocyte development. Despite its importance, the transcriptional program underlying this process remains elusive. Cbfß forms transcription factor complexes with Runx proteins, and here we show that Cbfß2, encoded by an RNA splice variant of the Cbfb gene, is essential for extrathymic differentiation of T cell progenitors. Furthermore, Cbfß2 endows extrathymic progenitors with thymus-homing capacity by inducing expression of the principal thymus-homing receptor, Ccr9. This occurs via direct binding of Cbfß2 to cell type-specific enhancers, as is observed in Rorγt induction during differentiation of lymphoid tissue inducer cells by activation of an intronic enhancer. As in mice, an alternative splicing event in zebrafish generates a Cbfß2-specific mRNA, important for ccr9 expression. Thus, despite phylogenetically and ontogenetically variable sites of origin of T cell progenitors, their robust thymus-homing capacity is ensured by an evolutionarily conserved mechanism emerging from functional diversification of Runx transcription factor complexes by acquisition of a novel splice variant.


Assuntos
Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/imunologia , Células Precursoras de Linfócitos T/citologia , Células Precursoras de Linfócitos T/imunologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/imunologia , Processamento Alternativo , Animais , Diferenciação Celular , Linhagem da Célula , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/deficiência , Elementos Facilitadores Genéticos , Evolução Molecular , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Knockout , Camundongos Mutantes , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , RNA Mensageiro/genética , Receptores CCR/genética , Receptores CCR/imunologia , Especificidade da Espécie , Timo/citologia , Timo/embriologia , Timo/imunologia , Peixe-Zebra , Proteínas de Peixe-Zebra/deficiência
10.
BMC Dev Biol ; 17(1): 14, 2017 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-29047338

RESUMO

BACKGROUND: The Runt-related transcription factors (Runx) are a family of evolutionarily conserved transcriptional regulators that play multiple roles in the developmental control of various cell types. Among the three mammalian Runx proteins, Runx1 is essential for definitive hematopoiesis and its dysfunction leads to human leukemogenesis. There are two promoters, distal (P1) and proximal (P2), in the Runx1 gene, which produce two Runx1 isoforms with distinct N-terminal amino acid sequences, P1-Runx1 and P2-Runx1. However, it remains unclear whether P2-Runx specific N-terminal sequence have any specific function for Runx1 protein. RESULTS: To address the function of the P2-Runx1 isoform, we established novel mutant mouse models in which the translational initiation AUG (+1) codon for P2-Runx1 isoform was modulated. We found that a truncated P2-Runx1 isoform is translated from a downstream non-canonical AUG codon. Importantly, the truncated P2-Runx1 isoform is sufficient to support primary hematopoiesis, even in the absence of the P1-Runx1 isoform. Furthermore, the truncated P2-Runx1 isoform was able to restore defect in basophil development caused by loss of the P1-Runx1 isoform. The truncated P2-Runx1 isoform was more stable than the canonical P2-Runx1 isoform. CONCLUSIONS: Our results demonstrate that the N-terminal sequences specific for P2-Runx1 are dispensable for Runx1 function, and likely serve as a de-stabilization module to regulate Runx1 production.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/química , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/genética , Hematopoese/genética , Hematopoese/fisiologia , Immunoblotting , Camundongos , Camundongos Mutantes , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
11.
Nat Commun ; 8(1): 702, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28951542

RESUMO

T-lineage committed precursor thymocytes are screened by a fate-determination process mediated via T cell receptor (TCR) signals for differentiation into distinct lineages. However, it remains unclear whether any antecedent event is required to couple TCR signals with the transcriptional program governing lineage decisions. Here we show that Bcl11b, known as a T-lineage commitment factor, is essential for proper expression of ThPOK and Runx3, central regulators for the CD4-helper/CD8-cytotoxic lineage choice. Loss of Bcl11b results in random expression of these factors and, thereby, lineage scrambling that is disconnected from TCR restriction by MHC. Initial Thpok repression by Bcl11b prior to the pre-selection stage is independent of a known silencer for Thpok, and requires the last zinc-finger motif in Bcl11b protein, which by contrast is dispensable for T-lineage commitment. Collectively, our findings shed new light on the function of Bcl11b in priming lineage-specifying genes to integrate TCR signals into subsequent transcriptional regulatory mechanisms.CD4 and CD8 T cells develop in the thymus with their transcription programs controlled by ThPOK and Runx3, respectively. Here the authors show that a pre-commitment event modulated by the transcription factor, Bcl11b, is required for the proper expression of ThPOK and Runx3 and correct CD4/CD8 lineage commitment.


Assuntos
Diferenciação Celular/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Proteínas Repressoras/genética , Linfócitos T Citotóxicos/citologia , Linfócitos T Auxiliares-Indutores/citologia , Timócitos/citologia , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Animais , Linhagem da Célula , Regulação da Expressão Gênica , Camundongos , Receptores de Antígenos de Linfócitos T/genética
12.
J Exp Med ; 214(10): 2933-2946, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28814567

RESUMO

The mouse Langerhans cell (LC) network is established through the differentiation of embryonic LC precursors. BMP7 and TGFß1 initiate cellular signaling that is essential for inducing LC differentiation and preserving LCs in a quiescent state, respectively. Here we show that loss of Cbfß2, one of two RNA splice variants of the Cbfb gene, results in long-term persistence of embryonic LC precursors after their developmental arrest at the transition into the EpCAM+ stage. This phenotype is caused by selective loss of BMP7-mediated signaling essential for LC differentiation, whereas TGFßR signaling is intact, maintaining cells in a quiescent state. Transgenic Cbfß2 expression at the neonatal stage, but not at the adult stage, restored differentiation from Cbfß2-deficient LC precursors. Loss of developmental potential in skin-residential precursor cells was accompanied by diminished BMP7-BMPR1A signaling. Collectively, our results reveal an essential requirement for the Cbfß2 variant in LC differentiation and provide novel insight into how the establishment and homeostasis of the LC network is regulated.


Assuntos
Subunidade beta de Fator de Ligação ao Core/deficiência , Células de Langerhans/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , Proteína Morfogenética Óssea 7/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Diferenciação Celular/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia
13.
Cell Rep ; 19(6): 1176-1188, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28494867

RESUMO

T cell receptor (TCR) signaling by MHC class I and II induces thymocytes to acquire cytotoxic and helper fates via the induction of Runx3 and ThPOK transcription factors, respectively. The mechanisms by which TCR signaling is translated into transcriptional programs for each cell fate remain elusive. Here, we show that, in post-selection thymocytes, a genome organizer, SATB1, activates genes for lineage-specifying factors, including ThPOK, Runx3, CD4, CD8, and Treg factor Foxp3, via regulating enhancers in these genes in a locus-specific manner. Indeed, SATB1-deficient thymocytes are partially re-directed into inappropriate T lineages after both MHC class I- and II-mediated selection, and they fail to generate NKT and Treg subsets. Despite its essential role in activating enhancers for the gene encoding ThPOK in TCR-signaled thymocytes, SATB1 becomes dispensable for maintaining ThPOK in CD4+ T cells. Collectively, our findings demonstrate that SATB1 shapes the primary T cell pool by directing lineage-specific transcriptional programs in the thymus.


Assuntos
Linfopoese , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Subpopulações de Linfócitos T/citologia , Animais , Antígenos CD4/genética , Antígenos CD4/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linhagem da Célula , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Elementos Facilitadores Genéticos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos , Subpopulações de Linfócitos T/metabolismo , Timo/citologia , Timo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional
14.
Sci Rep ; 7: 41351, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28150718

RESUMO

A TCRß enhancer, known as the Eß enhancer, plays a critical role in V(D)J recombination and transcription of the Tcrb gene. However, the coordinated action of trans-acting factors in the activation of Eß during T cell development remains uncharacterized. Here, we characterized the roles of Runx complexes in the regulation of the Eß function. A single mutation at one of the two Runx binding motifs within the Eß severely impaired Tcrb activation at the initiation phase in immature thymocytes. However, TCRß expression level in mature thymocytes that developed under such a single Runx site mutation was similar to that of the control. In contrast, mutations at two Runx motifs eliminated Eß activity, demonstrating that Runx complex binding is essential to initiate Eß activation. In cells expressing Tcrb harboring rearranged V(D)J structure, Runx complexes are dispensable to maintain TCRß expression, whereas Eß itself is continuously required for TCRß expression. These findings imply that Runx complexes are essential for Eß activation at the initiation phase, but are not necessary for maintaining Eß activity at later developmental stages. Collectively, our results indicate that the requirements of trans-acting factor for Eß activity are differentially regulated, depending on the developmental stage and cellular activation status.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Animais , Deleção de Genes , Células Germinativas/metabolismo , Cinética , Ativação Linfocitária/genética , Camundongos , Mutação/genética , Ligação Proteica , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Recombinação Genética , Transcrição Gênica
15.
EMBO J ; 32(8): 1183-94, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23481257

RESUMO

CD4(+) helper and CD8(+) cytotoxic T cells differentiate from common precursors in the thymus after T-cell receptor (TCR)-mediated selection. Commitment to the helper lineage depends on persistent TCR signals and expression of the ThPOK transcription factor, whereas a ThPOK cis-regulatory element, ThPOK silencer, represses Thpok gene expression during commitment to the cytotoxic lineage. Here, we show that silencer-mediated alterations of chromatin structures in cytotoxic-lineage thymocytes establish a repressive state that is epigenetically inherited in peripheral CD8(+) T cells even after removal of the silencer. When silencer activity is enhanced in helper-lineage cells, by increasing its copy number, a similar heritable Thpok silencing occurs. Epigenetic locking of the Thpok locus may therefore be an independent event from commitment to the cytotoxic lineage. These findings imply that long-lasting TCR signals are needed to establish stable Thpok expression activity to commit to helper T-cell fate and that full commitment to the helper lineage requires persistent reversal of silencer activity during a particular time window.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Timo/imunologia , Fatores de Transcrição/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Fatores de Tempo
16.
Nat Immunol ; 14(3): 281-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23334788

RESUMO

TCRαß thymocytes differentiate into either CD8αß(+) cytotoxic T lymphocytes or CD4(+) helper T cells. This functional dichotomy is controlled by key transcription factors, including the helper T cell master regulator ThPOK, which suppresses the cytolytic program in major histocompatibility complex (MHC) class II-restricted CD4(+) thymocytes. ThPOK continues to repress genes of the CD8 lineage in mature CD4(+) T cells, even as they differentiate into effector helper T cell subsets. Here we found that the helper T cell fate was not fixed and that mature, antigen-stimulated CD4(+) T cells terminated expression of the gene encoding ThPOK and reactivated genes of the CD8 lineage. This unexpected plasticity resulted in the post-thymic termination of the helper T cell program and the functional differentiation of distinct MHC class II-restricted CD4(+) cytotoxic T lymphocytes.


Assuntos
Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Citrobacter rodentium/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Proteínas de Homeodomínio/genética , Interleucina-7/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/metabolismo , Timócitos/metabolismo
17.
J Immunol ; 190(4): 1397-401, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23315079

RESUMO

Differentiation of MHC class II-selected thymocytes toward the CD4(+) helper lineage depends on function of the transcription factor ThPOK, whose expression is repressed in CD8(+) cytotoxic lineage cells by a transcriptional silencer activity within the distal regulatory element (DRE) in the Thpok gene. Interestingly, the DRE also functions as a transcriptional enhancer. However, how the DRE exerts such dual functionality remains obscure. In this study, we dissected the DRE and identified DNA sequences specifically responsible for enhancer activity, and designated this as the thymic enhancer. Removal of the thymic enhancer from the murine Thpok locus resulted in inefficient ThPOK induction, thereby inducing a redirection toward alternative CD8(+) cytotoxic lineage in a proportion of MHC class II-selected cells, even when they express monoclonal MHC class II-restricted transgenic TCR. Thus, regulation of contiguous but separable sequences with opposite function in the DRE plays an important role in precise coupling of TCR signaling with the selection process of two opposite lineages.


Assuntos
Elementos Facilitadores Genéticos/imunologia , Elementos Silenciadores Transcricionais/imunologia , Fatores de Transcrição/genética , Ativação Transcricional/imunologia , Animais , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Elementos Facilitadores Genéticos/genética , Genes Reporter , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência de DNA , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Timo/citologia , Timo/imunologia , Timo/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/deficiência
18.
Blood ; 115(15): 3042-50, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20139099

RESUMO

The transcription factor Runx1 is a pivotal regulator of definitive hematopoiesis in mouse ontogeny. Vertebrate Runx1 is transcribed from 2 promoters, the distal P1 and proximal P2, which provide a paradigm of the complex transcriptional and translational control of Runx1 function. However, very little is known about the biologic relevance of alternative Runx1 promoter usage in definitive hematopoietic cell emergence. Here we report that both promoters are active at the very onset of definitive hematopoiesis, with a skewing toward the P2. Moreover, functional and morphologic analysis of a novel P1-null and an attenuated P2 mouse model revealed that although both promoters play important nonredundant roles in the emergence of definitive hematopoietic cells, the proximal P2 was most critically required for this. The nature of the observed phenotypes is indicative of a differential contribution of the P1 and P2 promoters to the control of overall Runx1 levels, where and when this is most critically required. In addition, the dynamic expression of P1-Runx1 and P2-Runx1 points at a requirement for Runx1 early in development, when the P2 is still the prevalent promoter in the emerging hemogenic endothelium and/or first committed hematopoietic cells.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Hematopoese/genética , Regiões Promotoras Genéticas/genética , Envelhecimento/genética , Alelos , Animais , Aorta/metabolismo , Aorta/patologia , Células da Medula Óssea/metabolismo , Caderinas/metabolismo , Agregação Celular , Contagem de Células , Ensaio de Unidades Formadoras de Colônias , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Perda do Embrião/genética , Perda do Embrião/patologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Loci Gênicos/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Modelos Genéticos , Mutação/genética
19.
Nat Immunol ; 9(10): 1113-21, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18776907

RESUMO

CD4 and the transcription factor ThPOK are essential for the differentiation of major histocompatibility complex class II-restricted thymocytes into the helper T cell lineage; their genes (Cd4 and Zbtb7b (called 'ThPOK' here)) are repressed by transcriptional silencer elements in cytotoxic T cells. The molecular mechanisms regulating expression of these genes during helper T cell lineage differentiation remain unknown. Here we showed that inefficient upregulation of ThPOK, induced by removal of the proximal enhancer from the ThPOK locus, resulted in the transdifferentiation of helper lineage-specified cells into the cytotoxic T cell lineage. Furthermore, direct antagonism by ThPOK of the Cd4 and ThPOK silencers generated two regulatory loops that initially inhibited Cd4 downregulation and later stabilized ThPOK expression. Our results show how an initial lineage-specification signal can be amplified and stabilized during the lineage-commitment process.


Assuntos
Transdiferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Elementos Silenciadores Transcricionais/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Fatores de Transcrição/genética , Animais , Antígenos CD4/imunologia , Antígenos CD4/metabolismo , Antígenos CD8/imunologia , Antígenos CD8/metabolismo , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Citometria de Fluxo , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Timo/citologia , Timo/imunologia , Transfecção
20.
Science ; 319(5864): 822-5, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18258917

RESUMO

Mouse CD4+CD8+ double-positive (DP) thymocytes differentiate into CD4+ helper-lineage cells upon expression of the transcription factor Th-POK but commit to the CD8+ cytotoxic lineage in its absence. We report the redirected differentiation of class I-restricted thymocytes into CD4+CD8- helper-like T cells upon loss of Runx transcription factor complexes. A Runx-binding sequence within the Th-POK locus acts as a transcriptional silencer that is essential for Th-POK repression and for development of CD8+ T cells. Thus, Th-POK expression and genetic programming for T helper cell development are actively inhibited by Runx-dependent silencer activity, allowing for cytotoxic T cell differentiation. Identification of the transcription factors network in CD4 and CD8 lineage choice provides insight into how distinct T cell subsets are developed for regulating the adaptive immune system.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subunidade alfa 3 de Fator de Ligação ao Core/fisiologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Imunoprecipitação da Cromatina , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Elementos Silenciadores Transcricionais , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...