Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Front Immunol ; 13: 958616, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35983034

RESUMEN

Upon antigen recognition by the T cell receptor (TCR), a complex signaling network orchestrated by protein-tyrosine kinases (PTKs) and protein-tyrosine phosphatases (PTPs) regulates the transmission of the extracellular signal to the nucleus. The role of the PTPs Src-homology 2 (SH2) domain-containing phosphatase 1 (SHP1, Ptpn6) and Src-homology 2 (SH2) domain-containing phosphatase 2 (SHP2, Ptpn11) have been studied in various cell types including T cells. Whereas SHP1 acts as an essential negative regulator of the proximal steps in T cell signalling, the role of SHP2 in T cell activation is still a matter of debate. Here, we analyzed the role of the constitutively active SHP2-D61Y-mutant in T cell activation using knock-in mice expressing the mutant form Ptpn11D61Y in T cells. We observed reduced numbers of CD8+ and increased numbers of CD4+ T cells in the bone marrow and spleen of young and aged SHP2-D61Y-mutant mice as well as in Influenza A Virus (IAV)-infected mice compared to controls. In addition, we found elevated frequencies of effector memory CD8+ T cells and an upregulation of the programmed cell death protein 1 (PD-1)-receptor on both CD4+ and CD8+ T cells. Functional analysis of SHP2-D61Y-mutated T cells revealed an induction of late apoptosis/necrosis, a reduced proliferation and altered signaling upon TCR stimulation. However, the ability of D61Y-mutant mice to clear viral infection was not affected. In conclusion, our data indicate an important regulatory role of SHP2 in T cell function, where the effect is determined by the kinetics of SHP2 phosphatase activity and differs in the presence of the permanently active and the temporally regulated phosphatase. Due to interaction of SHP2 with the PD-1-receptor targeting the protein-tyrosine phosphatase might be a valuable tool to enhance T cell activities in immunotherapy.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células de Memoria Inmunológica , Activación de Linfocitos , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Dominios Homologos src
2.
Front Immunol ; 13: 870720, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711460

RESUMEN

Background: Allergic asthma is a chronic disease and medical treatment often fails to fully control the disease in the long term, leading to a great need for new therapeutic approaches. Immunoproteasome inhibition impairs T helper cell function and is effective in many (auto-) inflammatory settings but its effect on allergic airway inflammation is unknown. Methods: Immunoproteasome expression was analyzed in in vitro polarized T helper cell subsets. To study Th2 cells in vivo acute allergic airway inflammation was induced in GATIR (GATA-3-vYFP reporter) mice using ovalbumin and house dust mite extract. Mice were treated with the immunoproteasome inhibitor ONX 0914 or vehicle during the challenge phase and the induction of airway inflammation was analyzed. Results: In vitro polarized T helper cell subsets (Th1, Th2, Th17, and Treg) express high levels of immunoproteasome subunits. GATIR mice proved to be a useful tool for identification of Th2 cells. Immunoproteasome inhibition reduced the Th2 response in both airway inflammation models. Furthermore, T cell activation and antigen-specific cytokine secretion was impaired and a reduced infiltration of eosinophils and professional antigen-presenting cells into the lung and the bronchoalveolar space was observed in the ovalbumin model. Conclusion: These results show the importance of the immunoproteasome in Th2 cells and airway inflammation. Our data provides first insight into the potential of using immunoproteasome inhibition to target the aberrant Th2 response, e.g. in allergic airway inflammation.


Asunto(s)
Asma , Células Th2 , Animales , Asma/tratamiento farmacológico , Asma/metabolismo , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ratones , Ovalbúmina/farmacología , Células Th17 , Células Th2/metabolismo
3.
Eur J Immunol ; 52(3): 404-417, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34918350

RESUMEN

BOB.1/OBF.1 is a lymphocyte-specific transcriptional co-activator of octamer-dependent transcription. It regulates the expression of genes important for lymphocyte physiology together with the Oct-1 and Oct-2 transcription factors. So far, BOB.1/OBF.1 has been studied in conventional knockout mice, whereby a function of BOB.1/OBF.1 in B but also in T cells was described. The main characteristic of BOB.1/OBF.1-deficient mice is the complete absence of germinal centers. However, it is entirely unsolved at which stage of B-cell development BOB.1/OBF.1 expression is essential for germinal center formation. Still, it is not known whether defects observed late in B-cell development of BOB.1/OBF.1-deficient mice are merely a consequence of defective early B-cell development. To answer the question, whether BOB.1/OBF.1 expression is required before or during the process of germinal center formation, we established a mouse system, which allows the conditional deletion of BOB.1/OBF.1 at different stages of B-cell development. Our data reveal a requirement for BOB.1/OBF.1 during both early antigen-independent and late antigen-dependent B-cell development, and further a requirement for efficient germinal center reaction during complete B-cell ontogeny. By specifically deleting BOB.1/OBF.1 in germinal center B cells, we provide evidence that the failure to form germinal centers is a germinal center B-cell intrinsic defect and not exclusively a consequence of defective early B-cell maturation.


Asunto(s)
Linfocitos B , Centro Germinal , Transactivadores/metabolismo , Animales , Linfocitos B/metabolismo , Diferenciación Celular , Activación de Linfocitos , Ratones , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción/metabolismo
4.
J Immunol ; 204(9): 2600-2611, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32213568

RESUMEN

Accurately tuned expression levels of the transcription factor GATA-3 are crucial at several stages of T cell and innate lymphoid cell development and differentiation. Moreover, several lines of evidence suggest that Gata3 expression might provide a reliable molecular marker for the identification of elusive progenitor cell subsets at the earliest stages of T lineage commitment. To be able to faithfully monitor Gata3 expression noninvasively at the single-cell level, we have generated a novel strain of knock-in reporter mice, termed GATIR, by inserting an expression cassette encoding a bright fluorescent marker into the 3'-untranslated region of the endogenous Gata3 locus. Importantly, in contrast to three previously published strains of Gata3 reporter mice, GATIR mice preserve physiological Gata3 expression on the targeted allele. In this study, we show that GATIR mice faithfully reflect endogenous Gata3 expression without disturbing the development of GATA-3-dependent lymphoid cell populations. We further show that GATIR mice provide an ideal tool for noninvasive monitoring of Th2 polarization and straightforward identification of innate lymphoid cell 2 progenitor populations. Finally, as our reporter is non-gene-destructive, GATIR mice can be bred to homozygosity, not feasible with previously published strains of Gata3 reporter mice harboring disrupted alleles. The availability of hetero- and homozygous Gata3 reporter mice with an exceptionally bright fluorescent marker, allowed us to visualize allelic Gata3 expression in individual cells simply by flow cytometry. The unambiguous results obtained provide compelling evidence against previously postulated monoallelic Gata3 expression in early T lineage and hematopoietic stem cell subsets.


Asunto(s)
Factor de Transcripción GATA3/genética , Genes Reporteros/genética , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/inmunología , Alelos , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Femenino , Citometría de Flujo/métodos , Colorantes Fluorescentes/metabolismo , Factor de Transcripción GATA3/inmunología , Técnicas de Sustitución del Gen/métodos , Genes Reporteros/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Linfocitos/inmunología , Células Progenitoras Linfoides/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología
5.
Front Immunol ; 10: 2158, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31572375

RESUMEN

In normal conditions gut homeostasis is maintained by the suppressive activity of regulatory T cells (Tregs), characterized by the expression of the transcription factor FoxP3. In human inflammatory bowel disease, which is believed to be the consequence of the loss of tolerance toward antigens normally contained in the gut lumen, Tregs have been found to be increased and functionally active, thus pointing against their possible role in the pathogenesis of this immune-mediated disease. Though, in inflammatory conditions, Tregs have been shown to upregulate the T helper (Th) type 1-related transcription factor Tbet and to express the pro-inflammatory cytokine IFNγ, thus suggesting that at a certain point of the inflammatory process, Tregs might contribute to inflammation rather than suppress it. Starting from the observation that Tregs isolated from the lamina propria of active but not inactive IBD patients or uninflamed controls express Tbet and IFNγ, we investigated the functional role of Th1-like Tregs in the dextran sulfate model of colitis. As observed in human IBD, Th1-like Tregs were upregulated in the inflamed lamina propria of treated mice and the expression of Tbet and IFNγ in Tregs preceded the accumulation of conventional Th1 cells. By using a Treg-specific Tbet conditional knockout, we demonstrated that Tbet expression in Tregs is required for the development of colitis. Indeed, Tbet knockout mice developed milder colitis and showed an impaired Th1 immune response. In these mice not only the Tbet deficient Tregs but also the Tbet proficient conventional T cells showed reduced IFNγ expression. However, Tbet deficiency did not affect the Tregs suppressive capacity in vitro and in vivo in the adoptive transfer model of colitis. In conclusion here we show that Tbet expression by Tregs sustains the early phase of the Th1-mediated inflammatory response in the gut.


Asunto(s)
Colitis/inmunología , Regulación de la Expresión Génica/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Proteínas de Dominio T Box/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Animales , Colitis/patología , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/patología , Masculino , Ratones , Linfocitos T Reguladores/patología , Células TH1/patología
6.
J Immunol ; 203(4): 783-788, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31243089

RESUMEN

TGF-ß-activated kinase 1 (TAK1) is known to play vital roles for innate and adaptive immunity; however, little is known about its potential role in limiting biological responses such as inflammation. In this study, we report that macrophage TAK1 participates in negatively regulating inflammation by restraining proinflammatory cell death. Macrophages from TAK1-deficient mice underwent cell death in response to LPS and poly(I:C), which took place in a manner dependent on TLR/TRIF-induced active Caspase8-mediated cleavage of gasdermin D, known as an executioner of pyroptosis. Likewise, TNF-α induced Caspase8-dependent gasdermin D processing following cell death in TAK1-deficient macrophages. Importantly, we demonstrated that this type of proinflammatory macrophage death is linked to susceptibility to septic shock in mice lacking TAK1 in macrophages in a TNF-α-independent fashion. Taken together, our data revealed that TAK1 acts as a signaling checkpoint to protect macrophages from unique proinflammatory cell death, ensuring the maintenance of innate immune homeostasis.


Asunto(s)
Inflamación/inmunología , Quinasas Quinasa Quinasa PAM/inmunología , Macrófagos/inmunología , Animales , Muerte Celular/inmunología , Inmunidad Innata/fisiología , Inflamación/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Cell Rep ; 27(3): 762-775.e5, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995475

RESUMEN

Direct bacterial recognition by innate receptors is crucial for bacterial clearance. Here, we show that the IgA receptor CD89 is a major innate receptor that directly binds bacteria independently of its cognate ligands IgA and c-reactive protein (CRP). This binding is only partially inhibited by serum IgA and induces bacterial phagocytosis by CD11c+ dendritic cells and monocytes and/or macrophages, suggesting a physiological role in innate host defense. Blood phagocytes from common variable immunodeficiency patients bind, internalize, and kill bacteria in a CD89-dependent manner, confirming the IgA independence of this mechanism. In vivo, CD89 transgenic mice are protected in two different models of sepsis: a model of pneumonia and the cecal ligation and puncture (CLP) polymicrobial model of infection. These data identify CD89 as a first-line innate receptor for bacterial clearance before adaptive responses can be mounted. Fc receptors may emerge as a class of innate receptors for various bacteria with pleiotropic roles.


Asunto(s)
Antígenos CD/metabolismo , Escherichia coli/fisiología , Receptores Fc/metabolismo , Sepsis/prevención & control , Streptococcus pneumoniae/fisiología , Animales , Antígenos CD/genética , Proteína C-Reactiva/metabolismo , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Huésped Inmunocomprometido , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Fagocitosis , Neumonía/mortalidad , Neumonía/patología , Receptores Fc/genética , Sepsis/inmunología
8.
J Invest Dermatol ; 139(6): 1274-1283, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30670308

RESUMEN

Atopic dermatitis is a chronic inflammatory skin disease with persistent pruritus. To clarify its molecular mechanism, it is important to establish a mouse model similar to the phenotypes of atopic dermatitis patients, particularly in exhibiting scratching behavior. Ikk2, a component of the IκB kinase complex, exerts pro-inflammatory responses, whereas its deficiency in keratinocytes paradoxically causes skin inflammation. In this study, we sought to generate a mouse model exhibiting skin inflammation by which dermal fibroblasts lack Ikk2 expression and evaluate whether cutaneous inflammatory phenotypes are similar to those of atopic dermatitis patients. To generate Ikk2-deficient mice (Nestincre;Ikk2FL/FL) in which Ikk2 is deleted in dermal fibroblasts, we crossed female Ikk2FL/FL mice to male Nestincre;Ikk2FL/+mice. These mice spontaneously developed skin inflammation limited to the face, with the appearance of Ikk2-deficient fibroblasts in the facial skin. These mice showed phenotypes similar to those of atopic dermatitis patients, including scratching behaviors, which are resistant to immunosuppressive or molecularly targeted drugs. These findings suggest that the Nestincre;Ikk2FL/FL mouse is an atopic dermatitis model that will be useful in clarifying atopic dermatitis pathogenesis and in developing a novel therapeutic agent for atopic dermatitis symptoms.


Asunto(s)
Dermatitis Atópica/genética , Dermis/inmunología , Modelos Animales de Enfermedad , Quinasa I-kappa B/genética , Prurito/genética , Animales , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Dermis/citología , Dermis/patología , Femenino , Fibroblastos/inmunología , Fibroblastos/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Nestina/genética , Prurito/inmunología
9.
Stem Cell Res ; 31: 83-94, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30031967

RESUMEN

Developmental pluripotency associated factor 4 (Dppa4) is a highly specific marker of pluripotent cells, and is also overexpressed in certain cancers, but its function in either of these contexts is poorly understood. In this study, we use ChIP-Seq to identify Dppa4 binding genome-wide in three distinct cell types: mouse embryonic stem cells (mESC), embryonal carcinoma cells, and 3T3 fibroblasts ectopically expressing Dppa4. We find a core set of Dppa4 binding sites shared across cell types, and also a substantial number of sites unique to each cell type. Across cell types Dppa4 shows a preference for binding to regions with active chromatin signatures, and can influence chromatin modifications at target genes. In 3T3 fibroblasts with enforced Dppa4 expression, Dppa4 represses the cell cycle inhibitor Cdkn2c and activates Ets family transcription factor Etv4, leading to alterations in the cell cycle that likely contribute to the oncogenic phenotype. Dppa4 also directly regulates Etv4 in mESC but represses it in this context, and binds with Oct4 to a set of shared targets that are largely independent of Sox2 and Nanog, indicating that Dppa4 functions independently of the core pluripotency network in stem cells. Together these data provide novel insights into Dppa4 function in both pluripotent and oncogenic contexts.


Asunto(s)
Células Madre de Carcinoma Embrionario/fisiología , Proteínas Nucleares/genética , Células Madre Pluripotentes/fisiología , Células 3T3 , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animales , Proliferación Celular/fisiología , Cromatina/genética , Cromatina/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre de Carcinoma Embrionario/citología , Células Madre de Carcinoma Embrionario/metabolismo , Regulación de la Expresión Génica , Genómica/métodos , Humanos , Ratones , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Transfección
10.
Cancer Immunol Res ; 6(9): 1082-1092, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29991500

RESUMEN

Chronic inflammation drives colitis-associated colorectal cancer (CAC) in inflammatory bowel disease (IBD). FoxP3+ regulatory T cells (Treg) coexpressing the Th17-related transcription factor RORγt accumulate in the lamina propria of IBD patients, where they are thought to represent an intermediate stage of development toward a Th17 proinflammatory phenotype. However, the role of these cells in CAC is unknown. RORγt+FoxP3+ cells were investigated in human samples of CAC, and their phenotypic stability and function were investigated in an azoxymethane/dextran sulfate sodium model of CAC using Treg fate-mapping reporter and Treg-specific RORγt conditional knockout mice. Tumor development and the intratumoral inflammatory milieu were characterized in these mice. The functional role of CTLA-4 expressed by Tregs and FoxO3 in dendritic cells (DC) was studied in vitro and in vivo by siRNA-silencing experiments. RORγt expression identified a phenotypically stable population of tumor-infiltrating Tregs in humans and mice. Conditional RORγt knockout mice showed reduced tumor incidence, and dysplastic cells exhibited low Ki67 expression and STAT3 activation. Tumor-infiltrating DCs produced less IL6, a cytokine that triggers STAT3-dependent proliferative signals in neoplastic cells. RORγt-deficient Tregs isolated from tumors overexpressed CTLA-4 and induced DCs to have elevated expression of the transcription factor FoxO3, thus reducing IL6 expression. Finally, in vivo silencing of FoxO3 obtained by siRNA microinjection in the tumors of RORγt-deficient mice restored IL6 expression and tumor growth. These data demonstrate that RORγt expressed by tumor-infiltrating Tregs sustains tumor growth by leaving IL6 expression in DCs unchecked. Cancer Immunol Res; 6(9); 1082-92. ©2018 AACR.


Asunto(s)
Colitis/complicaciones , Neoplasias Colorrectales/inmunología , Células Dendríticas/inmunología , Interleucina-6/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Linfocitos T Reguladores/inmunología , Animales , Azoximetano , Antígeno CTLA-4/genética , Colitis/inducido químicamente , Neoplasias Colorrectales/etiología , Sulfato de Dextran , Proteína Forkhead Box O3/genética , Silenciador del Gen , Humanos , Inflamación , Interleucina-6/genética , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño
11.
Nature ; 545(7652): 98-102, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28445461

RESUMEN

The relative contribution of the effector molecules produced by T cells to tumour rejection is unclear, but interferon-γ (IFNγ) is critical in most of the analysed models. Although IFNγ can impede tumour growth by acting directly on cancer cells, it must also act on the tumour stroma for effective rejection of large, established tumours. However, which stroma cells respond to IFNγ and by which mechanism IFNγ contributes to tumour rejection through stromal targeting have remained unknown. Here we use a model of IFNγ induction and an IFNγ-GFP fusion protein in large, vascularized tumours growing in mice that express the IFNγ receptor exclusively in defined cell types. Responsiveness to IFNγ by myeloid cells and other haematopoietic cells, including T cells or fibroblasts, was not sufficient for IFNγ-induced tumour regression, whereas responsiveness of endothelial cells to IFNγ was necessary and sufficient. Intravital microscopy revealed IFNγ-induced regression of the tumour vasculature, resulting in arrest of blood flow and subsequent collapse of tumours, similar to non-haemorrhagic necrosis in ischaemia and unlike haemorrhagic necrosis induced by tumour necrosis factor. The early events of IFNγ-induced tumour ischaemia resemble non-apoptotic blood vessel regression during development, wound healing or IFNγ-mediated, pregnancy-induced remodelling of uterine arteries. A better mechanistic understanding of how solid tumours are rejected may aid the design of more effective protocols for adoptive T-cell therapy.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Hipoxia de la Célula/inmunología , Interferón gamma/inmunología , Isquemia/inmunología , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Remodelación Vascular , Animales , Vasos Sanguíneos/inmunología , Vasos Sanguíneos/metabolismo , Línea Celular Tumoral , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Interferón gamma/biosíntesis , Microscopía Intravital , Isquemia/metabolismo , Isquemia/patología , Masculino , Ratones , Necrosis , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Interferón/metabolismo , Células del Estroma/inmunología , Células del Estroma/metabolismo , Especificidad por Sustrato , Cicatrización de Heridas , Receptor de Interferón gamma
14.
Cell Stem Cell ; 19(6): 752-767, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27641306

RESUMEN

Mouse mutants with an impaired DNA damage response frequently exhibit a set of remarkably similar defects in the HSPC compartment that are of largely unknown molecular basis. Using Mixed-Lineage-Leukemia-5 (Mll5)-deficient mice as prototypical examples, we have identified a mechanistic pathway linking DNA damage and HSPC malfunction. We show that Mll5 deficiency results in accumulation of DNA damage and reactive oxygen species (ROS) in HSPCs. Reduction of ROS efficiently reverses hematopoietic defects, establishing ROS as a major cause of impaired HSPC function. The Ink4a/Arf locus also contributes to HSPC phenotypes, at least in part via promotion of ROS. Strikingly, toxic ROS levels in Mll5-/- mice are critically dependent on type 1 interferon (IFN-1) signaling, which triggers mitochondrial accumulation of full-length Bid. Genetic inactivation of Bid diminishes ROS levels and reverses HSPC defects in Mll5-/- mice. Overall, therefore, our findings highlight an unexpected IFN-1 > Bid > ROS pathway underlying DNA damage-associated HSPC malfunction.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Daño del ADN , Células Madre Hematopoyéticas/metabolismo , Interferón Tipo I/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Administración Oral , Animales , Animales Recién Nacidos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Sitios Genéticos , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina , Espacio Intracelular/metabolismo , Ratones , Poli I-C/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores de Interferón/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia
15.
Immunol Cell Biol ; 93(4): 417-23, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25533288

RESUMEN

Costimulatory signals by CD28 are critical for thymic regulatory T-cell (Treg) development. To determine the functional relevance of CD28 for peripheral Treg post thymic selection, we crossed the widely used Forkhead box protein 3 (Foxp3)-CreYFP mice to mice bearing a conditional Cd28 allele. Treg-specific CD28 deficiency provoked a severe autoimmune syndrome as a result of a strong disadvantage in competitive fitness and proliferation of CD28-deficient Tregs. By contrast, Treg survival and lineage integrity were not affected by the lack of CD28. This data demonstrate that, even after the initial induction requirement, Treg maintain a higher dependency on CD28 signalling than conventional T cells for homeostasis. In addition, we found the Foxp3-CreYFP allele to be a hypomorph, with reduced Foxp3 protein levels. Furthermore, we report here the stochastic activity of the Foxp3-CreYFP allele in non-Tregs, sufficient to recombine some conditional alleles (including Cd28) but not others (including R26-RFP). This hypomorphism and 'leaky' expression of the Foxp3-CreYFP allele should be considered when analysing the conditionally mutated Treg.


Asunto(s)
Antígenos CD28/metabolismo , Factores de Transcripción Forkhead/metabolismo , Subgrupos de Linfocitos T/fisiología , Linfocitos T Reguladores/fisiología , Animales , Autoinmunidad/genética , Antígenos CD28/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Supervivencia Celular/genética , Selección Clonal Mediada por Antígenos/genética , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Ratones , Ratones Transgénicos , Transducción de Señal/genética
16.
Immunity ; 41(6): 934-46, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526308

RESUMEN

Gradations in extracellular regulated kinase (ERK) signaling have been implicated in essentially every developmental checkpoint or differentiation process encountered by lymphocytes. Yet, despite intensive effort, the molecular basis by which differences in ERK activation specify alternative cell fates remains poorly understood. We report here that differential ERK signaling controls lymphoid-fate specification through an alternative mode of action. While ERK phosphorylates most substrates, such as RSK, by targeting them through its D-domain, this well-studied mode of ERK action was dispensable for development of γδ T cells. Instead, development of γδ T cells was dependent upon an alternative mode of action mediated by the DEF-binding pocket (DBP) of ERK. This domain enabled ERK to bind a distinct and select set of proteins required for specification of the γδ fate. These data provide the first in vivo demonstration for the role of DBP-mediated interactions in orchestrating alternate ERK-dependent developmental outcomes.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Estabilidad Proteica , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal/genética , Especificidad por Sustrato/genética
17.
Nature ; 509(7501): 465-70, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24828041

RESUMEN

Cell competition is an emerging principle underlying selection for cellular fitness during development and disease. Competition may be relevant for cancer, but an experimental link between defects in competition and tumorigenesis is elusive. In the thymus, T lymphocytes develop from precursors that are constantly replaced by bone-marrow-derived progenitors. Here we show that in mice this turnover is regulated by natural cell competition between 'young' bone-marrow-derived and 'old' thymus-resident progenitors that, although genetically identical, execute differential gene expression programs. Disruption of cell competition leads to progenitor self-renewal, upregulation of Hmga1, transformation, and T-cell acute lymphoblastic leukaemia (T-ALL) resembling the human disease in pathology, genomic lesions, leukaemia-associated transcripts, and activating mutations in Notch1. Hence, cell competition is a tumour suppressor mechanism in the thymus. Failure to select fit progenitors through cell competition may explain leukaemia in X-linked severe combined immune deficiency patients who showed thymus-autonomous T-cell development after therapy with gene-corrected autologous progenitors.


Asunto(s)
Transformación Celular Neoplásica , Células Madre Hematopoyéticas/citología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Timo/citología , Animales , División Celular , Movimiento Celular , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas HMGA/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch1/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T/patología , Timo/patología , Transcriptoma/genética
18.
J Immunol ; 192(6): 2585-92, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24516201

RESUMEN

Understanding the cellular dynamics of Aire-expressing lineage(s) among medullary thymic epithelial cells (AEL-mTECs) is essential for gaining insight into the roles of Aire in establishment of self-tolerance. In this study, we monitored the maturation program of AEL-mTECs by temporal lineage tracing, in which bacterial artificial chromosome transgenic mice expressing tamoxifen-inducible Cre recombinase under control of the Aire regulatory element were crossed with reporter strains. We estimated that the half-life of AEL-mTECs subsequent to Aire expression was ∼7-8 d, which was much longer than that reported previously, owing to the existence of a post-Aire stage. We found that loss of Aire did not alter the overall lifespan of AEL-mTECs, inconsistent with the previous notion that Aire expression in medullary thymic epithelial cells (mTECs) might result in their apoptosis for efficient cross-presentation of self-antigens expressed by AEL-mTECs. In contrast, Aire was required for the full maturation program of AEL-mTECs, as exemplified by the lack of physiological downregulation of CD80 during the post-Aire stage in Aire-deficient mice, thus accounting for the abnormally increased CD80(high) mTECs seen in such mice. Of interest, increased CD80(high) mTECs in Aire-deficient mice were not mTEC autonomous and were dependent on cross-talk with thymocytes. These results further support the roles of Aire in the differentiation program of AEL-mTECs.


Asunto(s)
Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Células Epiteliales/inmunología , Factores de Transcripción/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Autoantígenos/inmunología , Autoantígenos/metabolismo , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Células Epiteliales/metabolismo , Citometría de Flujo , Inmunohistoquímica , Cinética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Timocitos/citología , Timocitos/inmunología , Timocitos/metabolismo , Timo/citología , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína AIRE
19.
Proc Natl Acad Sci U S A ; 111(10): 3805-10, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24567410

RESUMEN

mTOR is an evolutionarily conserved kinase that plays a critical role in sensing and responding to environmental determinants. Recent studies have shown that fine-tuning of the activity of mTOR complexes contributes to organogenesis and tumorigenesis. Although rapamycin, an allosteric mTOR inhibitor, is an effective immunosuppressant, the precise roles of mTOR complexes in early T-cell development remain unclear. Here we show that mTORC1 plays a critical role in the development of both early T-cell progenitors and leukemia. Deletion of Raptor, an essential component of mTORC1, produced defects in the earliest development of T-cell progenitors in vivo and in vitro. Deficiency of Raptor resulted in cell cycle abnormalities in early T-cell progenitors that were associated with instability of the Cyclin D2/D3-CDK6 complexes; deficiency of Rictor, an mTORC2 component, did not have the same effect, indicating that mTORC1 and -2 control T-cell development in different ways. In a model of myeloproliferative neoplasm and T-cell acute lymphoblastic leukemia (T-ALL) evoked by Kras activation, Raptor deficiency dramatically inhibited the cell cycle in oncogenic Kras-expressing T-cell progenitors, but not myeloid progenitors, and specifically prevented the development of T-ALL. Although rapamycin treatment significantly prolonged the survival of recipient mice bearing T-ALL cells, rapamycin-insensitive leukemia cells continued to propagate in vivo. In contrast, Raptor deficiency in the T-ALL model resulted in cell cycle arrest and efficient eradication of leukemia. Thus, understanding the cell-context-dependent role of mTORC1 illustrates the potential importance of mTOR signals as therapeutic targets.


Asunto(s)
Linfopoyesis/fisiología , Modelos Inmunológicos , Complejos Multiproteicos/fisiología , Células Precursoras de Linfocitos T/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Proteínas Portadoras/metabolismo , Ciclo Celular/inmunología , Ciclo Celular/fisiología , Cartilla de ADN , Citometría de Flujo , Perfilación de la Expresión Génica , Immunoblotting , Inmunohistoquímica , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Complejos Multiproteicos/deficiencia , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Reguladora Asociada a mTOR , Transducción de Señal/fisiología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/deficiencia
20.
PLoS One ; 8(9): e74639, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24058613

RESUMEN

Regulatory T cells (Tregs) maintain immune homeostasis by limiting inflammatory responses. TRAF6 plays a key role in the regulation of innate and adaptive immunity by mediating signals from various receptors including the T-cell receptor (TCR). T cell-specific deletion of TRAF6 has been shown to induce multiorgan inflammatory disease, but the role of TRAF6 in Tregs remains to be investigated. Here, we generated Treg-specific TRAF6-deficient mice using Foxp3-Cre and TRAF6-flox mice. Treg-specific TRAF6-deficient (cKO) mice developed allergic skin diseases, arthritis, lymphadenopathy and hyper IgE phenotypes. Although TRAF6-deficient Tregs possess similar in vitro suppression activity compared to wild-type Tregs, TRAF6-deficient Tregs did not suppress colitis in lymphopenic mice very efficiently due to reduced number of Foxp3-positive cells. In addition, the fraction of TRAF6-deficient Tregs was reduced compared with wild-type Tregs in female cKO mice without inflammation. Moreover, adoptive transfer of Foxp3 (+) Tregs into Rag2(-/-) mice revealed that TRAF6-deficient Tregs converted into Foxp3(-) cells more rapidly than WT Tregs under lymphopenic conditions. Fate-mapping analysis also revealed that conversion of Tregs from Foxp3(+) to Foxp3(-) (exFoxp3 cells) was accelerated in TRAF6-deficient Tregs. These data indicate that TRAF6 in Tregs plays important roles in the maintenance of Foxp3 in Tregs and in the suppression of pathogenic Th2 type conversion of Tregs.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T Reguladores/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Células Th2/inmunología , Animales , Proliferación Celular , Femenino , Factores de Transcripción Forkhead/metabolismo , Integrasas/metabolismo , Interleucina-2/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Fenotipo , Factor de Transcripción STAT5/metabolismo , Factor 6 Asociado a Receptor de TNF/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA