Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 35: 53-84, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-27912316

RESUMO

Helper T (Th) cell subsets direct immune responses by producing signature cytokines. Th2 cells produce IL-4, IL-5, and IL-13, which are important in humoral immunity and protection from helminth infection and are central to the pathogenesis of many allergic inflammatory diseases. Molecular analysis of Th2 cell differentiation and maintenance of function has led to recent discoveries that have refined our understanding of Th2 cell biology. Epigenetic regulation of Gata3 expression by chromatin remodeling complexes such as Polycomb and Trithorax is crucial for maintaining Th2 cell identity. In the context of allergic diseases, memory-type pathogenic Th2 cells have been identified in both mice and humans. To better understand these disease-driving cell populations, we have developed a model called the pathogenic Th population disease induction model. The concept of defined subsets of pathogenic Th cells may spur new, effective strategies for treating intractable chronic inflammatory disorders.


Assuntos
Helmintíase/imunologia , Hipersensibilidade/imunologia , Células Th2/imunologia , Animais , Diferenciação Celular , Modelos Animais de Doenças , Epigênese Genética , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Imunidade Humoral , Memória Imunológica , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Camundongos , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo
2.
Nat Immunol ; 19(12): 1427-1440, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30374131

RESUMO

Multipotent progenitor cells confirm their T cell-lineage identity in the CD4-CD8- double-negative (DN) pro-T cell DN2 stages, when expression of the essential transcription factor Bcl11b begins. In vivo and in vitro stage-specific deletions globally identified Bcl11b-controlled target genes in pro-T cells. Proteomics analysis revealed that Bcl11b associated with multiple cofactors and that its direct action was needed to recruit those cofactors to selective target sites. Regions near functionally regulated target genes showed enrichment for those sites of Bcl11b-dependent recruitment of cofactors, and deletion of individual cofactors relieved the repression of many genes normally repressed by Bcl11b. Runx1 collaborated with Bcl11b most frequently for both activation and repression. In parallel, Bcl11b indirectly regulated a subset of target genes by a gene network circuit via the transcription inhibitor Id2 (encoded by Id2) and transcription factor PLZF (encoded by Zbtb16); Id2 and Zbtb16 were directly repressed by Bcl11b, and Id2 and PLZF controlled distinct alternative programs. Thus, our study defines the molecular basis of direct and indirect Bcl11b actions that promote T cell identity and block alternative potentials.


Assuntos
Linfopoese/imunologia , Células Precursoras de Linfócitos T/imunologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/biossíntese , Proteínas Repressoras/imunologia , Proteínas Supressoras de Tumor/imunologia , Animais , Diferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Proteína 2 Inibidora de Diferenciação/biossíntese , Proteína 2 Inibidora de Diferenciação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Precursoras de Linfócitos T/citologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/imunologia
3.
Immunity ; 48(6): 1119-1134.e7, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29924977

RESUMO

Transcription factors normally regulate gene expression through their action at sites where they bind to DNA. However, the balance of activating and repressive functions that a transcription factor can mediate is not completely understood. Here, we showed that the transcription factor PU.1 regulated gene expression in early T cell development both by recruiting partner transcription factors to its own binding sites and by depleting them from the binding sites that they preferred when PU.1 was absent. The removal of partner factors Satb1 and Runx1 occurred primarily from sites where PU.1 itself did not bind. Genes linked to sites of partner factor "theft" were enriched for genes that PU.1 represses despite lack of binding, both in a model cell line system and in normal T cell development. Thus, system-level competitive recruitment dynamics permit PU.1 to affect gene expression both through its own target sites and through action at a distance.


Assuntos
Diferenciação Celular/imunologia , Regulação da Expressão Gênica/imunologia , Linfopoese/fisiologia , Proteínas Proto-Oncogênicas/imunologia , Linfócitos T/imunologia , Transativadores/imunologia , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Linfopoese/imunologia , Proteínas de Ligação à Região de Interação com a Matriz/imunologia , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
4.
Nat Immunol ; 13(8): 778-86, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22751141

RESUMO

Sox4 is a transcription factor that regulates various developmental processes. Here we show that Sox4 was induced by TGF-ß and negatively regulated the transcription factor GATA-3, the master regulator of function of T helper type 2 (T(H)2) cells, by two distinct mechanisms. First, Sox4 bound directly to GATA-3, preventing its binding to GATA-3 consensus DNA sequences. Second, Sox4 bound to the promoter region of the gene encoding interleukin 5 (IL-5), a T(H)2 cytokine, and prevented binding of GATA-3 to this promoter. T(H)2 cell-driven airway inflammation was modulated by alterations in Sox4 expression. Thus, Sox4 acted as a downstream target of TGF-ß to inhibit GATA-3 function, T(H)2 differentiation and T(H)2 cell-mediated inflammation.


Assuntos
Fator de Transcrição GATA3/metabolismo , Fatores de Transcrição SOXC/genética , Fatores de Transcrição SOXC/metabolismo , Células Th2/citologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Proteínas de Ligação a DNA/antagonistas & inibidores , Fator de Transcrição GATA3/biossíntese , Interferon gama/biossíntese , Interleucina-5/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pneumonia/imunologia , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Células Th2/imunologia , Células Th2/metabolismo , Fator de Crescimento Transformador beta/genética
5.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33479171

RESUMO

Runt domain-related (Runx) transcription factors are essential for early T cell development in mice from uncommitted to committed stages. Single and double Runx knockouts via Cas9 show that target genes responding to Runx activity are not solely controlled by the dominant factor, Runx1. Instead, Runx1 and Runx3 are coexpressed in single cells; bind to highly overlapping genomic sites; and have redundant, collaborative functions regulating genes pivotal for T cell development. Despite stable combined expression levels across pro-T cell development, Runx1 and Runx3 preferentially activate and repress genes that change expression dynamically during lineage commitment, mostly activating T-lineage genes and repressing multipotent progenitor genes. Furthermore, most Runx target genes are sensitive to Runx perturbation only at one stage and often respond to Runx more for expression transitions than for maintenance. Contributing to this highly stage-dependent gene regulation function, Runx1 and Runx3 extensively shift their binding sites during commitment. Functionally distinct Runx occupancy sites associated with stage-specific activation or repression are also distinguished by different patterns of partner factor cobinding. Finally, Runx occupancies change coordinately at numerous clustered sites around positively or negatively regulated targets during commitment. This multisite binding behavior may contribute to a developmental "ratchet" mechanism making commitment irreversible.


Assuntos
Linhagem da Célula/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Células Precursoras de Linfócitos T/imunologia , Linfócitos T/imunologia , Transcriptoma , Animais , Diferenciação Celular , Linhagem da Célula/genética , Subunidade alfa 2 de Fator de Ligação ao Core/imunologia , Subunidade alfa 3 de Fator de Ligação ao Core/imunologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Subunidade alfa de Receptor de Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Masculino , Camundongos , Células Precursoras de Linfócitos T/citologia , Cultura Primária de Células , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Linfócitos T/classificação , Linfócitos T/citologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/imunologia
6.
J Biol Chem ; 298(11): 102506, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36126774

RESUMO

Hematopoietic stem and progenitor cells can differentiate into all types of blood cells. Regulatory mechanisms underlying pluripotency in progenitors, such as the ability of lymphoid progenitor cells to differentiate into T-lineage, remain unclear. We have previously reported that LIM domain only 2 (Lmo2), a bridging factor in large transcriptional complexes, is essential to retain the ability of lymphoid progenitors to differentiate into T-lineage. However, biochemical characterization of Lmo2 protein complexes in physiological hematopoietic progenitors remains obscure. Here, we identified approximately 600 Lmo2-interacting molecules in a lymphoid progenitor cell line by two-step affinity purification with LC-MS/MS analysis. Zinc finger and BTB domain containing 1 (Zbtb1) and CBFA2/RUNX1 partner transcriptional corepressor 3 (Cbfa2t3) were found to be the functionally important binding partners of Lmo2. We determined CRISPR/Cas9-mediated acute disruption of Zbtb1 or Cbfa2t3 in the lymphoid progenitor or bone marrow-derived primary hematopoietic progenitor cells causes significant defects in the initiation of T-cell development when Notch signaling is activated. Our transcriptome analysis of Zbtb1- or Cbfa2t3-deficient lymphoid progenitors revealed that Tcf7 was a common target for both factors. Additionally, ChIP-seq analysis showed that Lmo2, Zbtb1, and Cbfa2t3 cobind to the Tcf7 upstream enhancer region, which is occupied by the Notch intracellular domain/RBPJ transcriptional complex after Notch stimulation, in lymphoid progenitors. Moreover, transduction with Tcf7 restored the defect in the T-lineage potential of Zbtb1-deficient lymphoid progenitors. Thus, in lymphoid progenitors, the Lmo2/Zbtb1/Cbfa2t3 complex directly binds to the Tcf7 locus and maintains responsiveness to the Notch-mediated inductive signaling to facilitate T-lineage differentiation.


Assuntos
Células Progenitoras Linfoides , Fatores de Transcrição , Células Progenitoras Linfoides/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Cromatografia Líquida , Espectrometria de Massas em Tandem , Células-Tronco Hematopoéticas/metabolismo , Diferenciação Celular , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo
7.
Bioessays ; 43(5): e2000345, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33624856

RESUMO

Establishment of cell lineage identity from multipotent progenitors is controlled by cooperative actions of lineage-specific and stably expressed transcription factors, combined with input from environmental signals. Lineage-specific master transcription factors activate and repress gene expression by recruiting consistently expressed transcription factors and chromatin modifiers to their target loci. Recent technical advances in genome-wide and multi-omics analysis have shed light on unexpected mechanisms that underlie more complicated actions of transcription factors in cell fate decisions. In this review, we discuss functional dynamics of stably expressed and continuously required factors, Notch and Runx family members, throughout developmental stages of early T cell development in the thymus. Pre- and post-commitment stage-specific transcription factors induce dynamic redeployment of Notch and Runx binding genomic regions. Thus, together with stage-specific transcription factors, shared transcription factors across distinct developmental stages regulate acquisition of T lineage identity.


Assuntos
Genoma , Fatores de Transcrição , Diferenciação Celular , Linhagem da Célula/genética , Linfócitos T , Fatores de Transcrição/genética
8.
Int Immunol ; 33(9): 469-478, 2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34147033

RESUMO

The intracellular fragment of Notch1, a mediator of Notch signaling that is frequently detected in thymic immigrants, is critical for specifying T-cell fate in the thymus, where Delta-like 4 (Dll4) functions as a Notch ligand on the epithelium. However, as such Notch signaling has not been detected in mature T cells, how Notch signaling contributes to their response in secondary lymphoid organs has not yet been fully defined. Here, we detected the marked expression of Dll4 on the stromal cells and the active fragment of Notch1 (Notch1 intracellular domain, N1ICD) in CD4+ T cells in the follicles of Peyer's patches (PPs). In addition, N1ICD-bearing T cells were found in the T-cell zone of PPs, especially in the transcription factor Foxp3+ regulatory T (Treg) cells, with slight expression of Dll4 on the stromal cells. These fragments disappeared in Dll4-deficient conditions. It was also found that Notch1- and Notch2-deficient T cells preferentially differentiated into Treg cells in PPs, but not CXCR5+PD-1+ follicular helper T (Tfh) cells. Moreover, these phenotypes were also observed in chimeric mice reconstituted with the control and T-cell-specific Notch1/2-deficient bone marrow or Treg cells. These results demonstrated that Dll4-mediated Notch signaling in PPs is required for the efficient appearance of Tfh cells in a Treg cell-prone environment, which is common among the gut-associated lymphoid tissues, and is critical for the generation of Tfh-mediated germinal center B cells.


Assuntos
Nódulos Linfáticos Agregados/imunologia , Receptores Notch/imunologia , Células T Auxiliares Foliculares/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Linfócitos B/imunologia , Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/imunologia , Centro Germinativo/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia
9.
Int Immunol ; 33(2): 107-118, 2021 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-32909612

RESUMO

AMBRA1 (activating molecule in Beclin1-regulated autophagy) is a member of the BECN1 (BECLIN1) protein complex, and it plays a role in autophagy, cell death, tumorigenesis and proliferation. We recently reported that on T-cell receptor (TCR) stimulation, AMBRA1 controlled both autophagy and the cell cycle with metabolic regulation. Accumulating evidence has shown that autophagy and metabolic control are pivotal for T-cell activation, clonal expansion and effector/memory cell fate decision. However, it is unknown whether AMBRA1 is involved in T-cell function under physiological conditions. We found that T cells in Ambra1-conditional knockout (cKO) mice induced an exacerbated graft versus host response when they were transplanted into allogeneic BALB/c mice. Furthermore, Ambra1-deficient T cells showed increased proliferation and cytotoxic capability toward specific antigens in response to in vivo stimulation using allogeneic spleen cells. This enhanced immune response mainly contributed to naive T-cell hyperactivity. The T-cell hyperactivity observed in this study was similar to those in some metabolic factor-deficient mice, but not those in other pro-autophagic factor-deficient mice. Under the static condition, however, naive T cells were reduced in Ambra1-cKO mice, the same as in pro-autophagic factor-deficient mice. Collectively, these results suggested that AMBRA1 was involved in regulating T cell-mediated immune responses through autophagy-dependent and -independent mechanisms.

11.
Immunity ; 39(5): 819-32, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24238339

RESUMO

After antigen encounter by CD4(+) T cells, polarizing cytokines induce the expression of master regulators that control differentiation. Inactivation of the histone methyltransferase Ezh2 was found to specifically enhance T helper 1 (Th1) and Th2 cell differentiation and plasticity. Ezh2 directly bound and facilitated correct expression of Tbx21 and Gata3 in differentiating Th1 and Th2 cells, accompanied by substantial trimethylation at lysine 27 of histone 3 (H3K27me3). In addition, Ezh2 deficiency resulted in spontaneous generation of discrete IFN-γ and Th2 cytokine-producing populations in nonpolarizing cultures, and under these conditions IFN-γ expression was largely dependent on enhanced expression of the transcription factor Eomesodermin. In vivo, loss of Ezh2 caused increased pathology in a model of allergic asthma and resulted in progressive accumulation of memory phenotype Th2 cells. This study establishes a functional link between Ezh2 and transcriptional regulation of lineage-specifying genes in terminally differentiated CD4(+) T cells.


Assuntos
Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/fisiologia , Complexo Repressor Polycomb 2/fisiologia , Subpopulações de Linfócitos T/citologia , Células Th1/citologia , Células Th2/citologia , Animais , Asma/genética , Asma/imunologia , Asma/patologia , Diferenciação Celular , Células Cultivadas/citologia , Células Cultivadas/imunologia , Células Cultivadas/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Fator de Transcrição GATA3/metabolismo , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/deficiência , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Memória Imunológica , Testes de Liberação de Interferon-gama , Linfocinas/biossíntese , Linfocinas/genética , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Complexo Repressor Polycomb 2/química , Complexo Repressor Polycomb 2/deficiência , Complexo Repressor Polycomb 2/genética , Processamento de Proteína Pós-Traducional , Deleção de Sequência , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Subpopulações de Linfócitos T/imunologia , Células Th1/imunologia , Células Th2/imunologia
12.
Genome Res ; 28(10): 1508-1519, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30171019

RESUMO

SPI1 (also known as PU.1) is a dominant but transient regulator in early T-cell precursors and a potent transcriptional controller of developmentally important pro-T-cell genes. Before T-lineage commitment, open chromatin is frequently occupied by PU.1, and many PU.1 sites lose accessibility when PU.1 is later down-regulated. Pioneering activity of PU.1 was tested in this developmentally dynamic context by quantitating the relationships between PU.1 occupancy and site quality and accessibility as PU.1 levels naturally declined in pro-T-cell development and by using stage-specific gain- and loss-of-function perturbations to relate binding to effects on target genes. PU.1 could bind closed genomic sites, but rapidly opened many of them, despite the absence of its frequent collaborator, CEBPA. RUNX motifs and RUNX1 binding were often linked to PU.1 at open sites, but highly expressed PU.1 could bind its sites without RUNX1. The dynamic properties of PU.1 engagements implied that PU.1 binding affinity and concentration determine its occupancy choices, but with quantitative trade-offs for occupancy between site sequence quality and stage-dependent site accessibility in chromatin. At nonpromoter sites, PU.1 binding criteria were more stringent than at promoters, and PU.1 was also much more effective as a transcriptional regulator at nonpromoter sites where local chromatin accessibility depended on the presence of PU.1. Notably, closed chromatin presented a qualitative barrier to occupancy by the PU.1 DNA-binding domain alone. Thus, effective pioneering at closed chromatin sites also depends on requirements beyond site recognition, served by non-DNA-binding domains of PU.1.


Assuntos
Cromatina/química , Cromatina/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Regiões Promotoras Genéticas , Análise de Sequência de RNA
13.
Immunity ; 35(5): 733-45, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22118525

RESUMO

The regulation of memory CD4(+) helper T (Th) cell function, such as polarized cytokine production, remains unclear. Here we show that memory T helper 2 (Th2) cells are divided into four subpopulations by CD62L and CXCR3 expression. All four subpopulations produced interleukin-4 (IL-4) and IL-13, whereas only the CD62L(lo)CXCR3(lo) population produced IL-5 accompanied by increased H3-K4 methylation at the Il5 gene locus. The transcription factor Eomesodermin (encoded by Eomes) was highly expressed in memory Th2 cells, whereas its expression was selectively downregulated in the IL-5-producing cells. Il5 expression was enhanced in Eomes-deficient cells, and Eomesodermin was shown to interact with the transcription factor GATA3, preventing GATA3 binding to the Il5 promoter. Memory Th2 cell-dependent airway inflammation was attenuated in the absence of the CD62L(lo)CXCR3(lo) population but was enhanced by Eomes-deficient memory Th2 cells. Thus, IL-5 production in memory Th2 cells is regulated by Eomesodermin via the inhibition of GATA3 activity.


Assuntos
Fator de Transcrição GATA3/metabolismo , Memória Imunológica/imunologia , Interleucina-5/biossíntese , Proteínas com Domínio T/metabolismo , Células Th2/imunologia , Animais , Células Cultivadas , Fator de Transcrição GATA3/antagonistas & inibidores , Expressão Gênica , Inflamação/imunologia , Selectina L/metabolismo , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Regiões Promotoras Genéticas , Receptores CXCR3/metabolismo , Sistema Respiratório/imunologia , Proteínas com Domínio T/genética , Células Th2/metabolismo , Transcrição Gênica
14.
Proc Natl Acad Sci U S A ; 114(23): 5800-5807, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28584128

RESUMO

T-cell development from hematopoietic progenitors depends on multiple transcription factors, mobilized and modulated by intrathymic Notch signaling. Key aspects of T-cell specification network architecture have been illuminated through recent reports defining roles of transcription factors PU.1, GATA-3, and E2A, their interactions with Notch signaling, and roles of Runx1, TCF-1, and Hes1, providing bases for a comprehensively updated model of the T-cell specification gene regulatory network presented herein. However, the role of lineage commitment factor Bcl11b has been unclear. We use self-organizing maps on 63 RNA-seq datasets from normal and perturbed T-cell development to identify functional targets of Bcl11b during commitment and relate them to other regulomes. We show that both activation and repression target genes can be bound by Bcl11b in vivo, and that Bcl11b effects overlap with E2A-dependent effects. The newly clarified role of Bcl11b distinguishes discrete components of commitment, resolving how innate lymphoid, myeloid, and dendritic, and B-cell fate alternatives are excluded by different mechanisms.


Assuntos
Diferenciação Celular/genética , Redes Reguladoras de Genes , Proteínas Repressoras/fisiologia , Linfócitos T/citologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Notch , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
15.
Genes Dev ; 26(13): 1445-58, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22713603

RESUMO

The transcription factors Nanog and Gata6 are critical to specify the epiblast versus primitive endoderm (PrE) lineages. However, little is known about the mechanisms that regulate the protein stability and activity of these factors in the developing embryo. Here we uncover an early developmental function for the Polycomb group member Bmi1 in supporting PrE lineage formation through Gata6 protein stabilization. We show that Bmi1 is enriched in the extraembryonic (endoderm [XEN] and trophectodermal stem [TS]) compartment and repressed by Nanog in pluripotent embryonic stem (ES) cells. In vivo, Bmi1 overlaps with the nascent Gata6 and Nanog protein from the eight-cell stage onward before it preferentially cosegregates with Gata6 in PrE progenitors. Mechanistically, we demonstrate that Bmi1 interacts with Gata6 in a Ring finger-dependent manner to confer protection against Gata6 ubiquitination and proteasomal degradation. A direct role for Bmi1 in cell fate allocation is established by loss-of-function experiments in chimeric embryoid bodies. We thus propose a novel regulatory pathway by which Bmi1 action on Gata6 stability could alter the balance between Gata6 and Nanog protein levels to introduce a bias toward a PrE identity in a cell-autonomous manner.


Assuntos
Endoderma/metabolismo , Fator de Transcrição GATA6/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem da Célula , Endoderma/citologia , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteína Homeobox Nanog , Proteínas Nucleares/genética , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Transcrição Gênica
16.
Curr Top Membr ; 83: 77-106, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31196611

RESUMO

We have previously identified the interaction between mammalian V-ATPase a2-subunit isoform and cytohesin-2 (CTH2) and studied molecular details of binding between these proteins. In particular, we found that six peptides derived from the N-terminal cytosolic domain of a2 subunit (a2N1-402) are involved in interaction with CTH2 (Merkulova, Bakulina, Thaker, Grüber, & Marshansky, 2010). However, the actual 3D binding interface was not determined in that study due to the lack of high-resolution structural information about a-subunits of V-ATPase. Here, using a combination of homology modeling and NMR analysis, we generated the structural model of complete a2N1-402 and uncovered the CTH2-binding interface. First, using the crystal-structure of the bacterial M. rubber Icyt-subunit of A-ATPase as a template (Srinivasan, Vyas, Baker, & Quiocho, 2011), we built a homology model of mammalian a2N1-352 fragment. Next, we combined it with the determined NMR structures of peptides a2N368-395 and a2N386-402 of the C-terminal section of a2N1-402. The complete molecular model of a2N1-402 revealed that six CTH2 interacting peptides are clustered in the distal and proximal lobe sub-domains of a2N1-402. Our data indicate that the proximal lobe sub-domain is the major interacting site with the Sec7 domain of first CTH2 protein, while the distal lobe sub-domain of a2N1-402 interacts with the PH-domain of second CTH2. Indeed, using Sec7/Arf-GEF activity assay we experimentally confirmed our model. The interface formed by peptides a2N1-17 and a2N35-49 is involved in specific interaction with Sec7 domain and regulation of GEF activity. These data are critical for understanding of the cross-talk between V-ATPase and CTH2 as well as for the rational drug design to regulate their function.


Assuntos
Desenho de Fármacos , Proteínas Ativadoras de GTPase/química , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Sequência de Aminoácidos , Animais , Bactérias , Sítios de Ligação , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
17.
Biochem Biophys Res Commun ; 506(1): 27-32, 2018 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-30336981

RESUMO

Invariant natural killer T (iNKT) cells exhibit potent antitumor effects upon activation by recognizing a specific glycolipid antigen. We previously performed phase I-II clinical studies to utilize iNKT cells using α-galactosylceramide-pulsed dendritic cells and identified leukotriene B4 12-hydroxydehydrogenase (LTB4DH) as a biomarker highly expressed in T cells derived from non-small cell lung cancer (NSCLC) patients who showed prolonged survival in respond to the iNKT cell immunotherapy. Because LTB4DH expression correlated with prolonged survival of NSCLC patients, we considered LTB4DH to play a role in iNKT cell immunotherapy. We herein demonstrate that the overexpression of LTB4DH in CD4+ or CD8+ T cells increases interferon-γ production and tumoricidal activity in the presence of prostaglandin E2. Moreover, the expression of granzyme a, granzyme b, and perforin mRNA was increased in LTB4DH-overexpressing cells.


Assuntos
Oxirredutases do Álcool/genética , Carcinoma Pulmonar de Células não Pequenas/terapia , Células Dendríticas/imunologia , Galactosilceramidas/farmacologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/terapia , Oxirredutases do Álcool/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/transplante , Dinoprostona/imunologia , Dinoprostona/metabolismo , Granzimas/genética , Granzimas/imunologia , Humanos , Imunoterapia/métodos , Interferon gama/genética , Interferon gama/imunologia , Células K562 , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/mortalidade , Células T Matadoras Naturais/efeitos dos fármacos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/patologia , Perforina/genética , Perforina/imunologia , Cultura Primária de Células , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Transdução de Sinais , Análise de Sobrevida
18.
J Biol Chem ; 290(21): 13095-103, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25861992

RESUMO

Gata3 acts as a master regulator for T helper 2 (Th2) cell differentiation by inducing chromatin remodeling of the Th2 cytokine loci, accelerating Th2 cell proliferation, and repressing Th1 cell differentiation. Gata3 also directly transactivates the interleukin-5 (Il5) gene via additional mechanisms that have not been fully elucidated. We herein identified a mechanism whereby the methylation of Gata3 at Arg-261 regulates the transcriptional activation of the Il5 gene in Th2 cells. Although the methylation-mimicking Gata3 mutant retained the ability to induce IL-4 and repress IFNγ production, the IL-5 production was selectively impaired. We also demonstrated that heat shock protein (Hsp) 60 strongly associates with the methylation-mimicking Gata3 mutant and negatively regulates elongation of the Il5 transcript by RNA polymerase II. Thus, arginine methylation appears to play a pivotal role in the organization of Gata3 complexes and the target gene specificity of Gata3.


Assuntos
Arginina/genética , Metilação de DNA , Fator de Transcrição GATA3/genética , Interleucina-5/genética , Células Th2/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Animais , Arginina/metabolismo , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Chaperonina 60/antagonistas & inibidores , Chaperonina 60/genética , Chaperonina 60/metabolismo , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Citocinas/genética , Citocinas/metabolismo , Citometria de Fluxo , Imunofluorescência , Fator de Transcrição GATA3/metabolismo , Imunoprecipitação , Interleucina-5/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia
19.
Proc Natl Acad Sci U S A ; 110(12): 4691-6, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23471993

RESUMO

GATA binding protein 3 (Gata3) is a GATA family transcription factor that controls differentiation of naïve CD4 T cells into T helper 2 (Th2) cells. However, it is unknown how Gata3 simultaneously activates Th2-specific genes while repressing those of other Th lineages. Here we show that chromodomain helicase DNA-binding protein 4 (Chd4) forms a complex with Gata3 in Th2 cells that both activates Th2 cytokine transcription and represses the Th1 cytokine IFN-γ. We define a Gata3/Chd4/p300 transcriptional activation complex at the Th2 cytokine loci and a Gata3/Chd4-nucleosome remodeling histone deacetylase repression complex at the Tbx21 locus in Th2 cells. We also demonstrate a physiological role for Chd4 in Th2-dependent inflammation in an in vivo model of asthmatic inflammation. Thus, Gata3/Chd4 forms functionally distinct complexes, which mediate both positive and negative gene regulation to facilitate Th2 cell differentiation.


Assuntos
Diferenciação Celular/imunologia , DNA Helicases/metabolismo , Fator de Transcrição GATA3/imunologia , Complexos Multiproteicos/imunologia , Células Th2/imunologia , Transcrição Gênica/imunologia , Animais , Asma/genética , Asma/imunologia , Asma/patologia , DNA Helicases/genética , Modelos Animais de Doenças , Fator de Transcrição GATA3/genética , Loci Gênicos/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interferon gama/genética , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Complexos Multiproteicos/genética , Nucleossomos/genética , Nucleossomos/imunologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Células Th2/patologia , Transcrição Gênica/genética
20.
Proc Natl Acad Sci U S A ; 110(46): 18626-31, 2013 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-24167278

RESUMO

GATA-binding protein 3 (Gata3) controls the differentiation of naive CD4 T cells into T helper 2 (Th2) cells by induction of chromatin remodeling of the Th2 cytokine gene loci, direct transactivation of Il5 and Il13 genes, and inhibition of Ifng. Gata3 also facilitates Th2 cell proliferation via additional mechanisms that are far less well understood. We herein found that Gata3 associates with RuvB-like protein 2 (Ruvbl2) and represses the expression of a CDK inhibitor, cyclin-dependent kinase inhibitor 2c (Cdkn2c) to facilitate the proliferation of Th2 cells. Gata3 directly bound to the Cdkn2c locus in an Ruvbl2-dependent manner. The defect in the proliferation of Gata3-deficient Th2 cells is rescued by the knockdown of Cdkn2c, indicating that Cdkn2c is a key molecule involved in the Gata3-mediated induction of Th2 cell proliferation. Ruvbl2-knockdown Th2 cells showed decreased antigen-induced expansion and caused less airway inflammation in vivo. We therefore have identified a functional Gata3/Ruvbl2 complex that regulates the proliferation of differentiating Th2 cells through the repression of a CDK inhibitor, Cdkn2c.


Assuntos
Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p18/metabolismo , DNA Helicases/imunologia , Fator de Transcrição GATA3/imunologia , Regulação da Expressão Gênica/imunologia , Complexos Multiproteicos/imunologia , Células Th2/citologia , ATPases Associadas a Diversas Atividades Celulares , Animais , Bromodesoxiuridina , Imunoprecipitação da Cromatina , Primers do DNA/genética , Técnicas de Silenciamento de Genes , Immunoblotting , Imunoprecipitação , Luciferases , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA