Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Immunol ; 8(89): eadi9066, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37948511

RESUMEN

How CD4+ lineage gene expression is initiated in differentiating thymocytes remains poorly understood. Here, we show that the paralog transcription factors Zfp281 and Zfp148 control both this process and cytokine expression by T helper cell type 2 (TH2) effector cells. Genetic, single-cell, and spatial transcriptomic analyses showed that these factors promote the intrathymic CD4+ T cell differentiation of class II major histocompatibility complex (MHC II)-restricted thymocytes, including expression of the CD4+ lineage-committing factor Thpok. In peripheral T cells, Zfp281 and Zfp148 promoted chromatin opening at and expression of TH2 cytokine genes but not of the TH2 lineage-determining transcription factor Gata3. We found that Zfp281 interacts with Gata3 and is recruited to Gata3 genomic binding sites at loci encoding Thpok and TH2 cytokines. Thus, Zfp148 and Zfp281 collaborate with Gata3 to promote CD4+ T cell development and TH2 cell responses.


Asunto(s)
Linfocitos T CD4-Positivos , Factores de Transcripción , Animales , Ratones , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Methods Mol Biol ; 2580: 173-186, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36374457

RESUMEN

Expansion of T cell subsets in vitro is a valuable tool for exploration of effector function and differentiation. Here we provide protocols for in vitro differentiation of CD4 and CD8 T cell subsets from naïve T cells for functional studies.


Asunto(s)
Subgrupos de Linfocitos T , Células TH1 , Ratones , Animales , Diferenciación Celular , Células Th2
3.
J Exp Med ; 219(1)2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34792530

RESUMEN

During the immune response, CD4+ T cells differentiate into distinct effector subtypes, including follicular helper T (Tfh) cells that help B cells, and into memory cells. Tfh and memory cells are required for long-term immunity; both depend on the transcription factor Bcl6, raising the question whether they differentiate through similar mechanisms. Here, using single-cell RNA and ATAC sequencing, we show that virus-responding CD4+ T cells lacking both Bcl6 and Blimp1 can differentiate into cells with transcriptomic, chromatin accessibility, and functional attributes of memory cells but not of Tfh cells. Thus, Bcl6 promotes memory cell differentiation primarily through its repression of Blimp1. These findings demonstrate that distinct mechanisms underpin the differentiation of memory and Tfh CD4+ cells and define the Bcl6-Blimp1 axis as a potential target for promoting long-term memory T cell differentiation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Células T de Memoria/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Células T Auxiliares Foliculares/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Perfilación de la Expresión Génica/métodos , Células T de Memoria/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Células T Auxiliares Foliculares/metabolismo
4.
Immunity ; 51(3): 465-478.e6, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31422869

RESUMEN

The generation of high-affinity neutralizing antibodies, the objective of most vaccine strategies, occurs in B cells within germinal centers (GCs) and requires rate-limiting "help" from follicular helper CD4+ T (Tfh) cells. Although Tfh differentiation is an attribute of MHC II-restricted CD4+ T cells, the transcription factors driving Tfh differentiation, notably Bcl6, are not restricted to CD4+ T cells. Here, we identified a requirement for the CD4+-specific transcription factor Thpok during Tfh cell differentiation, GC formation, and antibody maturation. Thpok promoted Bcl6 expression and bound to a Thpok-responsive region in the first intron of Bcl6. Thpok also promoted the expression of Bcl6-independent genes, including the transcription factor Maf, which cooperated with Bcl6 to mediate the effect of Thpok on Tfh cell differentiation. Our findings identify a transcriptional program that links the CD4+ lineage with Tfh differentiation, a limiting factor for efficient B cell responses, and suggest avenues to optimize vaccine generation.


Asunto(s)
Diferenciación Celular/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Proteínas Proto-Oncogénicas c-maf/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Transcripción/inmunología , Transcripción Genética/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Centro Germinal/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL
5.
Immunity ; 50(1): 91-105.e4, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30638736

RESUMEN

Memory CD4+ T cells mediate long-term immunity, and their generation is a key objective of vaccination strategies. However, the transcriptional circuitry controlling the emergence of memory cells from early CD4+ antigen-responders remains poorly understood. Here, using single-cell RNA-seq to study the transcriptome of virus-specific CD4+ T cells, we identified a gene signature that distinguishes potential memory precursors from effector cells. We found that both that signature and the emergence of memory CD4+ T cells required the transcription factor Thpok. We further demonstrated that Thpok cell-intrinsically protected memory cells from a dysfunctional, effector-like transcriptional program, similar to but distinct from the exhaustion pattern of cells responding to chronic infection. Mechanistically, Thpok- bound genes encoding the transcription factors Blimp1 and Runx3 and acted by antagonizing their expression. Thus, a Thpok-dependent circuitry promotes both memory CD4+ T cells' differentiation and functional fitness, two previously unconnected critical attributes of adaptive immunity.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Subgrupos de Linfocitos T/fisiología , Factores de Transcripción/metabolismo , Animales , Antígenos Virales/inmunología , Diferenciación Celular , Células Cultivadas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Memoria Inmunológica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Unión Proteica , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Factores de Transcripción/genética , Transcriptoma
6.
Proc Natl Acad Sci U S A ; 114(50): 13236-13241, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180433

RESUMEN

CD8+ T cells are preprogrammed for cytotoxic differentiation in the thymus as they acquire expression of the transcription factor Runx3. However, a subset of effector CD8+ T cells (Tc17) produce IL-17 and fail to express cytotoxic genes. Here, we show that the transcription factors directing IL-17 production, STAT3 and RORγt, inhibit cytotoxicity despite persistent Runx3 expression. Cytotoxic gene repression did not require the transcription factor Thpok, which in CD4+ T cells restrains Runx3 functions and cytotoxicity; and STAT3 restrained cytotoxic gene expression in CD8+ T cells responding to viral infection in vivo. STAT3-induced RORγt represses cytotoxic genes by inhibiting the functions but not the expression of the "cytotoxic" transcription factors T-bet and Eomesodermin. Thus, the transcriptional circuitry directing IL-17 expression inhibits cytotoxic functions. However, by allowing expression of activators of the cytotoxic program, this inhibitory mechanism contributes to the instability of IL-17-producing T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica/genética , Factor de Transcripción STAT3/metabolismo , Animales , Células Cultivadas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo
7.
J Immunol ; 199(5): 1716-1728, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28754678

RESUMEN

The CD4+ lineage-specific transcription factor Thpok is required for intrathymic CD4+ T cell differentiation and, together with its homolog LRF, supports CD4+ T cell helper effector responses. However, it is not known whether these factors are needed for the regulatory T cell (Treg) arm of MHC class II responses. In this study, by inactivating in mice the genes encoding both factors in differentiated Tregs, we show that Thpok and LRF are redundantly required to maintain the size and functions of the postthymic Treg pool. They support IL-2-mediated gene expression and the functions of the Treg-specific factor Foxp3. Accordingly, Treg-specific disruption of Thpok and Lrf causes a lethal inflammatory syndrome similar to that resulting from Treg deficiency. Unlike in conventional T cells, Thpok and LRF functions in Tregs are not mediated by their repression of the transcription factor Runx3. Additionally, we found that Thpok is needed for the differentiation of thymic Treg precursors, an observation in line with the fact that Foxp3+ Tregs are CD4+ cells. Thus, a common Thpok-LRF node supports both helper and regulatory arms of MHC class II responses.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Linfocitos T Reguladores/inmunología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/microbiología , Factores de Transcripción/genética
8.
J Immunol ; 196(12): 4848-56, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27260768

RESUMEN

MHC-restricted CD4(+) and CD8(+) T cells are at the core of most adaptive immune responses. Although these cells carry distinct functions, they arise from a common precursor during thymic differentiation, in a developmental sequence that matches CD4 and CD8 expression and functional potential with MHC restriction. Although the transcriptional control of CD4(+)-CD8(+) lineage choice in the thymus is now better understood, less was known about what maintains the CD4(+) and CD8(+) lineage integrity of mature T cells. In this review, we discuss the mechanisms that establish in the thymus, and maintain in postthymic cells, the separation of these lineages. We focus on recent studies that address the mechanisms of epigenetic control of Cd4 expression and emphasize how maintaining a transcriptional circuitry nucleated around Thpok and Runx proteins, the key architects of CD4(+)-CD8(+) lineage commitment in the thymus, is critical for CD4(+) T cell helper functions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Timo/inmunología , Timo/metabolismo , Factores de Transcripción/metabolismo , Animales , Antígenos CD4/genética , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Diferenciación Celular , Linaje de la Célula , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Humanos , Ratones , Primates , Linfocitos T Colaboradores-Inductores/fisiología , Timo/citología , Factores de Transcripción/genética , Transcripción Genética
9.
Nucleic Acids Res ; 44(15): 7144-58, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27112571

RESUMEN

The activation of naïve B lymphocyte involves rapid and major changes in chromatin organization and gene expression; however, the complete repertoire of nuclear factors affecting these genomic changes is not known. We report that HMGN proteins, which bind to nucleosomes and affect chromatin structure and function, co-localize with, and maintain the intensity of DNase I hypersensitive sites genome wide, in resting but not in activated B cells. Transcription analyses of resting and activated B cells from wild-type and Hmgn(-/-) mice, show that loss of HMGNs dampens the magnitude of the transcriptional response and alters the pattern of gene expression during the course of B-cell activation; defense response genes are most affected at the onset of activation. Our study provides insights into the biological function of the ubiquitous HMGN chromatin binding proteins and into epigenetic processes that affect the fidelity of the transcriptional response during the activation of B cell lymphocytes.


Asunto(s)
Linfocitos B/metabolismo , Cromatina/genética , Cromatina/metabolismo , Regulación de la Expresión Génica , Proteínas HMGN/metabolismo , Activación de Linfocitos/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Desoxirribonucleasa I/metabolismo , Epigénesis Genética , Proteínas HMGN/deficiencia , Proteínas HMGN/genética , Proteína HMGN1/metabolismo , Proteína HMGN2/metabolismo , Masculino , Ratones , Nucleosomas/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Bazo/citología , Bazo/inmunología
10.
Methods Mol Biol ; 1323: 3-21, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26294394

RESUMEN

T lymphocytes (T cells) are essential for proper adaptive immune responses. They perform a variety of functions in defenses against pathogens, and notably control, positively or negatively, other cells involved in immune responses. T cells develop in the thymus from bone marrow-derived precursors. These precursors (thymocytes) proliferate, rearrange the genes encoding subunits of the T cell antigen receptor, which endow them with their unique antigen specificity, and undergo various degrees of pre-programming for their functions in immune responses. Thus, analyzing T cell development in the thymus is essential for understanding their functions in immune responses. In addition, the thymus constitutes an attractive experimental model to analyze mechanisms of cell proliferation, differentiation and survival, all of which are involved in thymocyte development. This chapter presents a quick overview of the key events characterizing intrathymic T cell development, as an introduction for readers entering this field of study.


Asunto(s)
Diferenciación Celular , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/fisiología , Timocitos/citología , Timocitos/fisiología , Animales , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos de Diferenciación de Linfocitos T/metabolismo , Selección Clonal Mediada por Antígenos , Humanos , Tolerancia Inmunológica , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal
11.
Methods Mol Biol ; 1323: 35-45, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26294396

RESUMEN

Genetics tools, and especially the ability to enforce, by transgenesis, or disrupt, by homologous recombination, gene expression in a cell-specific manner, have revolutionized the study of immunology and propelled the laboratory mouse as the main model to study immune responses. Perhaps more than any other aspect of immunology, the study of T cell development has benefited from these technologies. This brief chapter summarizes genetic tools specific to T cell development studies, focusing on mouse strains with lineage- and stage-specific expression of the Cre recombinase, or expressing unique antigen receptor specificities. It ends with a broader discussion of strategies to enforce ectopic lineage and stage-specific gene expression.


Asunto(s)
Diferenciación Celular/genética , Genómica/métodos , Linfocitos T/citología , Linfocitos T/fisiología , Animales , Linaje de la Célula/genética , Selección Clonal Mediada por Antígenos/genética , Selección Clonal Mediada por Antígenos/inmunología , Humanos , Receptores de Antígenos de Linfocitos T/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología
13.
Nat Commun ; 6: 8152, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26328764

RESUMEN

Although histone H3 lysine 27 trimethylation (H3K27Me3) is associated with gene silencing, whether H3K27Me3 demethylation affects transcription and cell differentiation in vivo has remained elusive. To investigate this, we conditionally inactivated the two H3K27Me3 demethylases, Jmjd3 and Utx, in non-dividing intrathymic CD4(+) T-cell precursors. Here we show that both enzymes redundantly promote H3K27Me3 removal at, and expression of, a specific subset of genes involved in terminal thymocyte differentiation, especially S1pr1, encoding a sphingosine-phosphate receptor required for thymocyte egress. Thymocyte expression of S1pr1 was not rescued in Jmjd3- and Utx-deficient male mice, which carry the catalytically inactive Utx homolog Uty, supporting the conclusion that it requires H3K27Me3 demethylase activity. These findings demonstrate that Jmjd3 and Utx are required for T-cell development, and point to a requirement for their H3K27Me3 demethylase activity in cell differentiation.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Histona Demetilasas/genética , Histona Demetilasas con Dominio de Jumonji/genética , Timocitos/citología , Animales , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Inmunoprecipitación de Cromatina , Citometría de Flujo , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Esfingosina-1-Fosfato , Timocitos/metabolismo
14.
Nat Immunol ; 15(10): 947-56, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25129370

RESUMEN

The transcription factor ThPOK promotes CD4(+) T cell differentiation in the thymus. Here, using a mouse strain that allows post-thymic gene deletion, we show that ThPOK maintains CD4(+) T lineage integrity and couples effector differentiation to environmental cues after antigenic stimulation. ThPOK preserved the integrity and amplitude of effector responses and was required for proper differentiation of types 1 and 2 helper T cells in vivo by restraining the expression and function of Runx3, a nuclear factor crucial for cytotoxic T cell differentiation. The transcription factor LRF acts redundantly with ThPOK to prevent the transdifferentiation of mature CD4(+) T cells into CD8(+) T cells. As such, the ThPOK-LRF transcriptional module was essential for CD4(+) T cell integrity and responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al ADN/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/inmunología
15.
Immunity ; 40(5): 681-91, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24792911

RESUMEN

Antigen specificity is critical in immune response and requires integration of antigen-specific signals with antigen-nonspecific signals such as those provided by cytokines. The mechanism integrating these pathways is incompletely understood. We report here that antigen-specific proliferative responses of CD4(+) T cells required downmodulation of tumor suppressor p53. In the absence of T cell receptor (TCR) signal, IL-2 induced sustained increase in p53 protein, which prevented proliferative responses despite strong signaling through the IL-2 receptor. In contrast, TCR signaling resulted in early termination of p53 protein expression by decreasing p53 mRNA as well as strong transcriptional induction of the p53-regulating protein Mdm2. Downmodulation of p53 in response to antigen stimulation was in fact critical for antigen-specific T cell proliferation, and preventing p53 degradation by inhibiting Mdm2 resulted in sustained p53 protein and prevented antigen-specific T cell proliferation. It is thus termination of p53 by TCR signaling that allows proliferative responses, enforcing antigen specificity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interleucina-2/inmunología , Proteínas Proto-Oncogénicas c-mdm2/genética , Receptores de Antígenos de Linfocitos T/inmunología , Proteína p53 Supresora de Tumor/genética , Animales , Especificidad de Anticuerpos/inmunología , Ciclo Celular/genética , Ciclo Celular/inmunología , Proliferación Celular , Células Cultivadas , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Receptores de Interleucina-2/inmunología , Transducción de Señal/inmunología , Transcripción Genética , Proteína p53 Supresora de Tumor/biosíntesis
17.
Proc Natl Acad Sci U S A ; 104(15): 6323-8, 2007 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-17405860

RESUMEN

The ATM (ataxia telangiectasia mutated) protein plays a central role in sensing and responding to DNA double-strand breaks. Lymphoid cells are unique in undergoing physiologic double-strand breaks in the processes of Ig class switch recombination and T or B cell receptor V(D)J recombination, and a role for ATM in these processes has been suggested by clinical observations in ataxia telangiectasia patients as well as in engineered mice with mutations in the Atm gene. We demonstrate here a defect in thymocyte maturation in ATM-deficient mice that is associated with decreased efficiency in V-J rearrangement of the endogenous T cell receptor (TCR)alpha locus, accompanied by increased frequency of unresolved TCR Jalpha coding end breaks. We also demonstrate that a functionally rearranged TCRalphabeta transgene is sufficient to restore thymocyte maturation, whereas increased thymocyte survival by bcl-2 cannot improve TCRalpha recombination and T cell development. These data indicate a direct role for ATM in TCR gene recombination in vivo that is critical for surface TCR expression in CD4(+)CD8(+) cells and for efficient thymocyte selection. We propose a unified model for the two major clinical characteristics of ATM deficiency, defective T cell maturation and increased genomic instability, frequently affecting the TCRalpha locus. In the absence of ATM, delayed TCRalpha coding joint formation results both in a reduction of alphabeta TCR-expressing immature cells, leading to inefficient thymocyte selection, and in accumulation of unstable open chromosomal DNA breaks, predisposing to TCRalpha locus-associated chromosomal abnormalities.


Asunto(s)
Proteínas de Ciclo Celular/inmunología , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/inmunología , Reordenamiento Génico de Linfocito T/genética , Proteínas Serina-Treonina Quinasas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Timo/citología , Proteínas Supresoras de Tumor/inmunología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Citometría de Flujo , Reordenamiento Génico de Linfocito T/inmunología , Inestabilidad Genómica/genética , Inestabilidad Genómica/inmunología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética
18.
J Immunol ; 174(8): 4657-61, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15814689

RESUMEN

Interaction of Fas with Fas ligand (FasL) is known to play a role in peripheral tolerance mediated by clonal deletion of Ag-specific T cells. We have assessed the requirement for Fas/FasL interactions during induction of tolerance to the fetus. Using H-Y-specific TCR transgenic mice, we have previously demonstrated that exposure of maternal T cells to H-Y expressed by male fetuses results in deletion of 50% of H-Y-specific maternal T cells. The remaining H-Y-specific T cells were hyporesponsive to H-Y as assayed by decreased proliferative ability and CTL activity. To determine whether Fas/FasL interactions contribute to induction of maternal T cell tolerance, responsiveness to fetal H-Y was assessed in H-Y-specific TCR transgenic pregnant females that were deficient in functional Fas or FasL. Surprisingly, both deletion and nondeletion components of tolerance were abrogated in TCR transgenic H-Y-specific lpr (Fas-deficient) or gld (FasL-deficient) pregnant females. Experiments further revealed that expression of FasL by the fetus, but not by the mother, is necessary and sufficient for both components of maternal T cell tolerance to fetal Ags. Fas interaction with fetal FasL is thus critical for both deletion and hyporesponsiveness of H-Y-reactive CD8+ T cells during pregnancy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Feto/inmunología , Antígeno H-Y/inmunología , Glicoproteínas de Membrana/metabolismo , Animales , Anergia Clonal , Proteína Ligando Fas , Femenino , Tolerancia Inmunológica , Ligandos , Masculino , Intercambio Materno-Fetal/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Embarazo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptor fas/metabolismo
19.
Eur J Immunol ; 35(2): 418-27, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15657954

RESUMEN

While the importance of the CD28/B7 costimulation pathway is well established for mature T cells, the role of CD28 in thymocyte selection is less well defined. The role of CD28 in both negative and positive selection was assessed using H-Y-specific TCR-transgenic (Tg) RAG-2-deficient (H-Yrag) mice. Negative selection in male H-Yrag mice was not affected by deficiency in CD28 or B7. Surprisingly, absence of CD28 or B7 in H-Yrag females resulted in increased numbers of CD8 single-positive (SP) thymocytes. The CD8 SP thymocytes found in these females were mature and functionally competent. Furthermore, double-positive (DP) thymocytes from CD28-knockout (CD28KO) or B7.1/B7.2 double-KO (B7DKO) females had higher levels of both CD5 and TCR than those from WT females, consistent with a stronger selecting signal. CD28KO H-Yrag fetal thymic organ cultures also had elevated numbers of thymic CD8 SP cells, reflecting increased thymic differentiation and not recirculation of peripheral T cells. Finally, increased selection of mature CD4 and CD8 SP T cells was observed in non-TCR-Tg CD28KO and B7DKO mice, indicating that this function of CD28-B7 interaction is not unique to a TCR-Tg model. Together these findings demonstrate a novel negative regulatory role for CD28 in inhibiting differentiation of SP thymocytes, probably through inhibition of thymic selection.


Asunto(s)
Antígeno B7-1/inmunología , Antígenos CD28/inmunología , Antígeno H-Y/inmunología , Timo/inmunología , Animales , Antígeno B7-1/genética , Antígenos CD28/genética , Antígenos CD5/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Timo/citología
20.
J Immunol ; 170(8): 3954-62, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12682222

RESUMEN

We have previously shown that transgenic (Tg) mice expressing either soluble or membrane-bound hen egg lysozyme (sHEL or mHEL, respectively) under control of the alphaA-crystallin promoter develop tolerance due to thymic expression of minuscule amounts of HEL. To further address the mechanisms by which this tolerance develops, we mated these two lines of Tg mice with the 3A9 line of HEL-specific TCR Tg mice, to produce double-Tg mice. Both lines of double-Tg mice showed deletion of HEL-specific T cells, demonstrated by reduction in numbers of these cells in the thymus and periphery, as well as by reduced proliferative response to HEL in vitro. In addition, the actual deletional process in thymi of the double-Tg mice was visualized in situ by the TUNEL assay and measured by binding of Annexin V. Notably, the apoptosis localized mainly in the thymic medulla, in line with the finding that the populations showing deletion and increased Annexin V binding consisted mainly of single- and double-positive thymocytes. Interestingly, the thymic deletional effect of sHEL was superior to that of mHEL in contrast to the opposite differential tolerogenic effects of these HEL forms on B cells specific to this Ag. Analysis of bone marrow chimeras indicates that both forms of HEL are produced by irradiation-resistant thymic stromal cells and the data suggest that sHEL is more effective in deleting 3A9 T cells due mainly to its higher accessibility to cross-presentation by dendritic APC.


Asunto(s)
Autoantígenos/biosíntesis , Células Epiteliales/inmunología , Proteínas de la Membrana/biosíntesis , Autotolerancia/inmunología , Subgrupos de Linfocitos T/inmunología , Timo/inmunología , Timo/metabolismo , Animales , Presentación de Antígeno/genética , Antígenos/biosíntesis , Antígenos/genética , Antígenos/inmunología , Apoptosis/genética , Apoptosis/inmunología , Autoantígenos/inmunología , Autoantígenos/metabolismo , Supresión Clonal/genética , Relación Dosis-Respuesta Inmunológica , Células Epiteliales/citología , Células Epiteliales/metabolismo , Ojo/anatomía & histología , Ojo/enzimología , Ojo/inmunología , Ojo/patología , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Proteínas del Ojo/inmunología , Inmunofenotipificación , Etiquetado Corte-Fin in Situ , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Muramidasa/biosíntesis , Muramidasa/genética , Muramidasa/inmunología , Autotolerancia/genética , Solubilidad , Células del Estroma/enzimología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Subgrupos de Linfocitos T/citología , Timo/citología , Timo/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...