Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 38(2): 110209, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021099

RESUMEN

Innate-like T cells, including invariant natural killer T cells, mucosal-associated invariant T cells, and γδ T cells, are present in various barrier tissues, including the lung, where they carry out protective responses during infections. Here, we investigate their roles during pulmonary pneumococcal infection. Following infection, innate-like T cells rapidly increase in lung tissue, in part through recruitment, but T cell antigen receptor activation and cytokine production occur mostly in interleukin-17-producing NKT17 and γδ T cells. NKT17 cells are preferentially located within lung tissue prior to infection, as are CD103+ dendritic cells, which are important both for antigen presentation to NKT17 cells and γδ T cell activation. Whereas interleukin-17-producing γδ T cells are numerous, granulocyte-macrophage colony-stimulating factor is exclusive to NKT17 cells and is required for optimal protection. These studies demonstrate how particular cellular interactions and responses of functional subsets of innate-like T cells contribute to protection from pathogenic lung infection.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Animales , Línea Celular , Células Dendríticas/inmunología , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-17/inmunología , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones Neumocócicas/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Streptococcus pneumoniae/inmunología
2.
Mucosal Immunol ; 14(3): 679-690, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33568785

RESUMEN

Inflammatory bowel disease is characterized by an exacerbated intestinal immune response, but the critical mechanisms regulating immune activation remain incompletely understood. We previously reported that the TNF-superfamily molecule TNFSF14 (LIGHT) is required for preventing severe disease in mouse models of colitis. In addition, deletion of lymphotoxin beta receptor (LTßR), which binds LIGHT, also led to aggravated colitis pathogenesis. Here, we aimed to determine the cell type(s) requiring LTßR and the mechanism critical for exacerbation of colitis. Specific deletion of LTßR in neutrophils (LTßRΔN), but not in several other cell types, was sufficient to induce aggravated colitis and colonic neutrophil accumulation. Mechanistically, RNA-Seq analysis revealed LIGHT-induced suppression of cellular metabolism, and mitochondrial function, that was dependent on LTßR. Functional studies confirmed increased mitochondrial mass and activity, associated with excessive mitochondrial ROS production and elevated glycolysis at steady-state and during colitis. Targeting these metabolic changes rescued exacerbated disease severity. Our results demonstrate that LIGHT signals to LTßR on neutrophils to suppress metabolic activation and thereby prevents exacerbated immune pathogenesis during colitis.


Asunto(s)
Colitis/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Receptor beta de Linfotoxina/metabolismo , Mitocondrias/metabolismo , Neutrófilos/metabolismo , Activación Metabólica , Animales , Sulfato de Dextran , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
3.
J Invest Dermatol ; 140(1): 132-142.e3, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31260672

RESUMEN

The cell adhesion molecule E-cadherin is a major component of adherens junctions and marks Langerhans cells (LC), the only dendritic cell (DC) population of the epidermis. LC form a dense network and attach themselves to the surrounding keratinocytes via homophilic E-cadherin binding. LC activation, mobilization, and migration require a reduction in LC E-cadherin expression. To determine whether E-cadherin plays a role in regulating LC homeostasis and function, we generated CD11c-specific E-cadherin knockout mice (CD11c-Ecaddel). In the absence of E-cadherin-mediated cell adhesion, LC numbers remained stable and similar as in control mice, even in aged animals. Intriguingly, E-cadherin-deficient LC displayed a dramatically changed morphology characterized by a more rounded cell body and fewer dendrites than wild-type cells. Nevertheless, maturation and migration of LC lacking E-cadherin was not altered, neither under steady-state nor inflammatory conditions. Accordingly, CD11c-Ecaddel and control mice developed comparable contact hypersensitivity reactions and imiquimod-triggered psoriatic skin inflammation, indicating that E-cadherin on LC does not influence their ability to orchestrate T cell-mediated immunity. In conclusion, our data demonstrate that E-cadherin is dispensable to maintain LC in the epidermis and does not regulate LC maturation, migration, and function.


Asunto(s)
Cadherinas/metabolismo , Dermatitis por Contacto/inmunología , Epidermis/fisiología , Células de Langerhans/fisiología , Psoriasis/inmunología , Animales , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Cadherinas/genética , Diferenciación Celular , Movimiento Celular , Forma de la Célula , Células Cultivadas , Dermatitis por Contacto/genética , Modelos Animales de Enfermedad , Homeostasis , Humanos , Imiquimod , Ratones , Ratones Noqueados , Psoriasis/genética
4.
Front Immunol ; 9: 2585, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30524422

RESUMEN

Over 1.5 million individuals in the United States are afflicted with inflammatory bowel disease (IBD). While the progression of IBD is multifactorial, chronic, unresolved inflammation certainly plays a key role. Additionally, while multiple immune mediators have been shown to affect pathogenesis, a comprehensive understanding of disease progression is lacking. Previous work has demonstrated that a member of the TNF superfamily, TNFSF14 (LIGHT), which is pro-inflammatory in several contexts, surprisingly plays an important role in protection from inflammation in mouse models of colitis, with LIGHT deficient mice having more severe disease pathogenesis. However, LIGHT is a single member of a complex signaling network. It signals through multiple receptors, including herpes virus entry mediator (HVEM) and lymphotoxin beta receptor (LTßR); these two receptors in turn can bind to other ligands. It remains unknown which receptors and competing ligands can mediate or counteract the outcome of LIGHT-signaling during colitis. Here we demonstrate that LIGHT signaling through LTßR, rather than HVEM, plays a critical role in the progression of DSS-induced colitis, as LTßR deficient mice exhibit a more severe disease phenotype. Further, mice deficient in LTαß do not exhibit differential colitis progression compared to WT mice. However, deletion of both LIGHT and LTαß, but not deletion of both LTαß and LTßR, resulted in a reversal of the adverse effects associated with the loss of LIGHT. In sum, the LIGHT/LTαß/LTßR signaling network contributes to DSS colitis, but there may be additional receptors or indirect effects, and therefore, the relationships between these receptors and ligands remains enigmatic.


Asunto(s)
Colitis/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
Cell Host Microbe ; 24(2): 249-260.e4, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-30092201

RESUMEN

Innate lymphoid cells (ILCs) are important regulators of early infection at mucosal barriers. ILCs are divided into three groups based on expression profiles, and are activated by cytokines and neuropeptides. Yet, it remains unknown if ILCs integrate other signals in providing protection. We show that signaling through herpes virus entry mediator (HVEM), a member of the tumor necrosis factor (TNF) receptor superfamily, in ILC3 is important for host defense against oral infection with the bacterial pathogen Yersinia enterocolitica. HVEM stimulates protective interferon-γ (IFN-γ) secretion from ILCs, and mice with HVEM-deficient ILC3 exhibit reduced IFN-γ production, higher bacterial burdens and increased mortality. In addition, IFN-γ production is critical as adoptive transfer of wild-type but not IFN-γ-deficient ILC3 can restore protection to mice lacking ILCs. We identify the TNF superfamily member, LIGHT, as the ligand inducing HVEM signals in ILCs. Thus HVEM signaling mediated by LIGHT plays a critical role in regulating ILC3-derived IFN-γ production for protection following infection. VIDEO ABSTRACT.


Asunto(s)
Infecciones por Enterobacteriaceae/prevención & control , Interferón gamma/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Traslado Adoptivo , Adulto , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/metabolismo , Transporte de Proteínas , Receptores CCR6/genética , Receptores CCR6/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Bazo/microbiología , Bazo/patología , Yersinia enterocolitica/patogenicidad
6.
J Exp Med ; 215(2): 415-422, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29339444

RESUMEN

Dermatitis is often associated with an allergic reaction characterized by excessive type 2 responses leading to epidermal acanthosis, hyperkeratosis, and dermal inflammation. Although factors like IL-4, IL-13, and thymic stromal lymphopoietin (TSLP) are thought to be instrumental for the development of this type of skin disorder, other cytokines may be critical. Here, we show that the tumor necrosis factor (TNF) superfamily protein LIGHT (homologous to lymphotoxin, exhibits inducible expression, and competes with HSV glycoprotein D for binding to HVEM, a receptor expressed on T lymphocytes) is required for experimental atopic dermatitis, and LIGHT directly controls keratinocyte hyperplasia, and production of periostin, a matricellular protein that contributes to the clinical features of atopic dermatitis as well as other skin diseases such as scleroderma. Mice with a conditional deletion of the LIGHT receptor HVEM (herpesvirus entry mediator) in keratinocytes phenocopied LIGHT-deficient mice in exhibiting reduced epidermal thickening and dermal collagen deposition in a model of atopic dermatitis driven by house dust mite allergen. LIGHT signaling through HVEM in human epidermal keratinocytes directly induced proliferation and periostin expression, and both keratinocyte-specific deletion of HVEM or antibody blocking of LIGHT-HVEM interactions after disease onset prevented expression of periostin and limited atopic dermatitis symptoms. Developing reagents that neutralize LIGHT-HVEM signaling might be useful for therapeutic intervention in skin diseases where periostin is a central feature.


Asunto(s)
Dermatitis Atópica/metabolismo , Queratinocitos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Antígenos Dermatofagoides/efectos adversos , Moléculas de Adhesión Celular/metabolismo , Proliferación Celular , Dermatitis Atópica/etiología , Dermatitis Atópica/inmunología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Queratinocitos/inmunología , Queratinocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Miembro 14 de Receptores del Factor de Necrosis Tumoral/deficiencia , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
7.
Nat Commun ; 7: 13696, 2016 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-27982078

RESUMEN

Asthma has multiple features, including airway hyperreactivity, inflammation and remodelling. The TNF superfamily member TNFSF14 (LIGHT), via interactions with the receptor TNFRSF14 (HVEM), can support TH2 cell generation and longevity and promote airway remodelling in mouse models of asthma, but the mechanisms by which TNFSF14 functions in this setting are incompletely understood. Here we find that mouse and human mast cells (MCs) express TNFRSF14 and that TNFSF14:TNFRSF14 interactions can enhance IgE-mediated MC signalling and mediator production. In mouse models of asthma, TNFRSF14 blockade with a neutralizing antibody administered after antigen sensitization, or genetic deletion of Tnfrsf14, diminishes plasma levels of antigen-specific IgG1 and IgE antibodies, airway hyperreactivity, airway inflammation and airway remodelling. Finally, by analysing two types of genetically MC-deficient mice after engrafting MCs that either do or do not express TNFRSF14, we show that TNFRSF14 expression on MCs significantly contributes to the development of multiple features of asthma pathology.


Asunto(s)
Asma/inducido químicamente , Asma/metabolismo , Mastocitos/fisiología , Receptores de IgE/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Anticuerpos , Antígenos Dermatofagoides/inmunología , Antígenos Dermatofagoides/toxicidad , Asma/patología , Líquido del Lavado Bronquioalveolar/citología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Inmunoglobulina E , Inmunoglobulina G , Ratones , Ratones Noqueados , Ovalbúmina/inmunología , Ovalbúmina/toxicidad , Receptores de IgE/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética
8.
Mediators Inflamm ; 2016: 9765238, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27578924

RESUMEN

Inflammatory bowel disease (IBD) is characterized by an impairment of the integrity of the mucosal epithelial barrier, which causes exacerbated inflammation of the intestine. The intestinal barrier is formed by different specialized epithelial cells, which separate the intestinal lumen from the lamina propria. In addition to its crucial role in protecting the body from invading pathogens, the intestinal epithelium contributes to intestinal homeostasis by its biochemical properties and communication to underlying immune cells. Innate lymphoid cells (ILCs) are a recently described population of lymphocytes that have been implicated in both mucosal homeostasis and inflammation. Recent findings indicate a critical feedback loop in which damaged epithelium activates these innate immune cells to restore epithelial barrier function. This review will focus on the signalling pathways between damaged epithelium and ILCs involved in repair of the epithelial barrier and tissue homeostasis and the relationship of these processes with the control of IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Linfocitos/fisiología , Animales , Humanos , Mucosa Intestinal/patología , Linfocitos/metabolismo
9.
J Invest Dermatol ; 136(11): 2260-2269, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27430407

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor involved in adaptive cell functions, and it is highly active in the epidermis. AhR ligands can accelerate keratinocyte differentiation, but the precise role of AhR in the skin barrier is unknown. Our study showed that transepidermal water loss, a parameter of skin barrier integrity, is high in AhR-deficient mice. Experiments with conditionally AhR-deficient mouse lines identified keratinocytes as the primary cell population responsible for high transepidermal water loss. Electron microscopy showed weaker intercellular connectivity in the epidermis of keratinocytes in AhR-knockout mice, and gene expression analysis identified many barrier-associated genes as AhR targets. Moreover, AhR-deficient mice had higher interindividual differences in their microbiome. Interestingly, removing AhR ligands from the diet of wild-type mice mimicked AhR deficiency with respect to the impaired barrier; conversely, re-addition of the plant-derived ligand indole-3-carbinol rescued the barrier deficiency even in aged mice. Our results suggest that functional AhR expression is critical for skin barrier integrity and that AhR represents a molecular target for the development of therapeutic approaches for skin barrier diseases, including by dietary intervention.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , ADN/genética , Regulación de la Expresión Génica , Queratinocitos/metabolismo , Receptores de Hidrocarburo de Aril/genética , Enfermedades de la Piel/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Queratinocitos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Receptores de Hidrocarburo de Aril/biosíntesis , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología
10.
J Immunol ; 195(3): 924-33, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26078271

RESUMEN

In this article, we characterize a novel Ag for invariant NKT (iNKT) cells capable of producing an especially robust Th1 response. This glycosphingolipid, DB06-1, is similar in chemical structure to the well-studied α-galactosylceramide (αGalCer), with the only change being a single atom: the substitution of a carbonyl oxygen with a sulfur atom. Although DB06-1 is not a more effective Ag in vitro, the small chemical change has a marked impact on the ability of this lipid Ag to stimulate iNKT cells in vivo, with increased IFN-γ production at 24 h compared with αGalCer, increased IL-12, and increased activation of NK cells to produce IFN-γ. These changes are correlated with an enhanced ability of DB06-1 to load in the CD1d molecules expressed by dendritic cells in vivo. Moreover, structural studies suggest a tighter fit into the CD1d binding groove by DB06-1 compared with αGalCer. Surprisingly, when iNKT cells previously exposed to DB06-1 are restimulated weeks later, they have greatly increased IL-10 production. Therefore, our data are consistent with a model whereby augmented and or prolonged presentation of a glycolipid Ag leads to increased activation of NK cells and a Th1-skewed immune response, which may result, in part, from enhanced loading into CD1d. Furthermore, our data suggest that strong antigenic stimulation in vivo may lead to the expansion of IL-10-producing iNKT cells, which could counteract the benefits of increased early IFN-γ production.


Asunto(s)
Galactosilceramidas/inmunología , Glicoesfingolípidos/inmunología , Interferón gamma/biosíntesis , Activación de Linfocitos/inmunología , Células T Asesinas Naturales/inmunología , Células TH1/inmunología , Animales , Antígenos CD1d/inmunología , Sitios de Unión/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Galactosilceramidas/química , Glicoesfingolípidos/química , Humanos , Interleucina-10/biosíntesis , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica/inmunología
11.
Gastroenterology ; 146(7): 1752-62.e4, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24560868

RESUMEN

BACKGROUND & AIMS: The pathogenesis of inflammatory bowel disease (IBD) is associated with a dysregulated mucosal immune response. Expression of the tumor necrosis factor (TNF) superfamily member 14 (TNFSF14, also known as LIGHT [homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes]) on T cells is involved in their activation; transgenic expression of LIGHT on T cells in mice promotes inflammation in multiple organs, including intestine. We investigated the roles for LIGHT in recovery from intestinal inflammation in mice. METHODS: We studied the role of LIGHT in intestinal inflammation using Tnfsf14(-/-) and wild-type mice. Colitis was induced by transfer of CD4(+)CD45RB(high) T cells into Rag1(-/-) or Tnfsf14(-/-)Rag1(-/-) mice, or by administration of dextran sulfate sodium to Tnfsf14(-/-) or wild-type C57BL/6J mice. Mice were weighed, colon tissues were collected and measured, and histology analyses were performed. We measured infiltrating cell populations and expression of cytokines, chemokines, and LIGHT. RESULTS: After administration of dextran sulfate sodium, Tnfsf14(-/-) mice developed more severe colitis than controls, based on their reduced survival, accelerated loss of body weight, and histologic scores. LIGHT protected mice from colitis via the lymphotoxin ß receptor and was expressed mainly by myeloid cells in the colon. Colons of Tnfsf14(-/-) mice also had increased accumulation of innate immune cells and higher levels of cytokines than colons from control mice. LIGHT, therefore, appears to regulate inflammation in the colon. CONCLUSIONS: Tnfsf14(-/-) mice develop more severe colitis than control mice. LIGHT signals through the lymphotoxin ß receptor in the colon to regulate the innate immune response and mediate recovery from intestinal inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Colitis/metabolismo , Colon/metabolismo , Mucosa Intestinal/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/trasplante , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Colitis/prevención & control , Colon/inmunología , Colon/patología , Citocinas/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Antígenos Comunes de Leucocito/metabolismo , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Tiempo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Pérdida de Peso
12.
J Leukoc Biol ; 95(1): 179-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24023259

RESUMEN

Whereas CD8⁺ T cells are essential for anti-tumor immunity, tumors often evade CD8⁺ T cell surveillance by immunosuppression. As the initiators of antigen-specific immune responses, DCs are likely to play a central role in regulating the balance between immunity and tolerance to tumor antigens and are specialized in their ability to cross-present exogenous tumor antigens on MHC class I molecules to initiate CD8⁺ T cell immunity. However, it remains unclear whether and how tumors modulate DC functions to suppress CD8⁺ T cell responses. We have shown previously that ß-catenin signaling in DCs promotes DC-mediated CD8⁺ T cell tolerance. Here, we tested the hypothesis that ß-catenin in DCs mediates tumor-induced suppression of CD8⁺ T cell immunity by inhibiting the ability of DCs in cross-priming. ß-Catenin was activated in DCs by multiple tumors in vivo and in vitro. B16 melanoma-bearing mice, when vaccinated with DC-targeting anti-DEC-205 mAb fused with tumor antigens, exhibited dampened CD8⁺ immunity, similar to DC-ß-catenin(active) mice. DCs from DC-ß-catenin(active) and tumor-bearing mice were deficient in cross-priming, and antigen-specific CD8⁺ T cells primed in these mice resulted in dampened CD8⁺ memory responses. Importantly, DC-ß-catenin⁻/⁻ mice completely abrogate tumor-mediated inhibition of cross-priming, suggesting that tumor-induced inhibition of cross-priming is dependent on ß-catenin. Finally, enhancing cross-priming at the priming or recall phase rescued ß-catenin-suppressed CD8⁺ immunity in DC-ß-catenin(active) and tumor-bearing mice. Thus, ß-catenin-mediated inhibition of cross-priming represents a new and potentially general mechanism that tumors employ to achieve immunosuppression.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Reactividad Cruzada/inmunología , Tolerancia Inmunológica , Neoplasias/inmunología , Neoplasias/metabolismo , beta Catenina/metabolismo , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Memoria Inmunológica , Melanoma Experimental , Ratones , Ratones Noqueados , Neoplasias/genética
13.
Blood ; 123(2): 217-27, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24092934

RESUMEN

Langerhans cells (LCs) are dendritic cells (DCs) residing in epithelia, where they critically regulate immunity and tolerance. The p14 adaptor molecule is part of the late endosomal/LAMTOR (lysosomal adaptor and mitogen-activated protein kinase and mammalian target of rapamycin [mTOR] activator/regulator) complex, thereby contributing to the signal transduction of the extracellular signaling-regulated kinase (ERK) and the mTOR cascade. Furthermore, p14 represents an important regulator for endosomal sorting processes within the cell. Mutated, dysfunctional p14 leads to a human immunodeficiency disorder with endosomal/lysosomal defects in immune cells. Because p14 participates in the regulation of endosomal trafficking, growth factor signaling, and cell proliferation, we investigated the role of p14 in mouse DCs/LCs using a conditional knockout mouse model. p14-deficient animals displayed a virtually complete loss of LCs in the epidermis early after birth due to impaired proliferation and increased apoptosis of LCs. Repopulation analysis after application of contact sensitizer leads to the recruitment of a transient LC population, predominantly consisting of short-term LCs. The underlying molecular mechanism involves the p14-mediated disruption of the LAMTOR complex which results in the malfunction of both ERK and mTOR signal pathways. Hence, we conclude that p14 acts as a novel and essential regulator of LC homeostasis in vivo.


Asunto(s)
Endosomas/metabolismo , Homeostasis , Células de Langerhans/metabolismo , Proteínas/genética , Proteínas/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/genética , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Mitosis/genética , Complejos Multiproteicos/metabolismo , Transducción de Señal , Piel/inmunología , Piel/metabolismo , Piel/patología , Serina-Treonina Quinasas TOR/metabolismo
14.
J Exp Med ; 210(9): 1657-64, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23940255

RESUMEN

Langerhans cells (LCs) constitute a network of immune sentinels in the skin epidermis that is seeded during embryogenesis. Whereas the development of LCs has been extensively studied, much less is known about the homeostatic renewal of adult LCs in "nonmanipulated" animals. Here, we present a new multicolor fluorescent fate mapping system and quantification approach to investigate adult LC homeostasis. This novel approach enables us to propose and provide evidence for a model in which the adult epidermal LC network is not formed by mature coequal LCs endowed with proliferative capabilities, but rather constituted by adjacent proliferative units composed of "dividing" LCs and their terminally differentiated daughter cells. Altogether, our results demonstrate the general utility of our novel fate-mapping system to follow cell population dynamics in vivo and to establish an alternative model for LC homeostasis.


Asunto(s)
Linaje de la Célula , Técnicas Citológicas/métodos , Homeostasis , Células de Langerhans/patología , Animales , Color , Imagenología Tridimensional , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
Proc Natl Acad Sci U S A ; 110(26): 10723-8, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23754427

RESUMEN

Psoriasis is an autoinflammatory skin disease of unknown etiology. Topical application of Aldara cream containing the Toll-like receptor (TLR)7 agonist Imiquimod (IMQ) onto patients induces flares of psoriasis. Likewise, in mice IMQ triggers pathological changes closely resembling psoriatic plaque formation. Key cytokines like IL-23 and type-I IFN (IFN-I), both being produced mainly by dendritic cells (DCs), have been implicated in psoriasis. Although plasmacytoid DCs (pDCs) are the main source of IFNα and thought to initiate disease, conventional DCs (cDCs) appear to maintain the psoriatic lesions. Any role of cDCs during lesion formation remains elusive. Here, we report that selective activation of TLR7 signaling specifically in CD11c(+) DCs was sufficient to induce psoriasiform skin disease in mice. Intriguingly, both pDCs and the IFN-I pathway were dispensable for the development of local skin inflammation. Selective TLR7 triggering of Langerin(+) DCs resulted in attenuated disease, whereas their depletion did not alter the severity of skin lesions. Moreover, after IMQ-painting, IL-23 was exclusively produced by Langerin(neg) DCs in vivo. In conclusion, TLR7-activated Langerin(neg) cDCs trigger psoriatic plaque formation via IL-23-mediated activation of innate IL-17/IL-22-producing lymphocytes, independently of pDCs or IFN-I. These results suggest therapeutic targeting of IL-23 production by cDCs to refine current treatment strategies for psoriasis.


Asunto(s)
Antígenos de Superficie/genética , Interleucina-23/biosíntesis , Células de Langerhans/inmunología , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Lectinas de Unión a Manosa/deficiencia , Lectinas de Unión a Manosa/genética , Psoriasis/inmunología , Aminoquinolinas/administración & dosificación , Animales , Antígenos de Superficie/biosíntesis , Modelos Animales de Enfermedad , Imiquimod , Células de Langerhans/efectos de los fármacos , Lectinas Tipo C/biosíntesis , Lectinas de Unión a Manosa/biosíntesis , Glicoproteínas de Membrana/agonistas , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Psoriasis/etiología , Psoriasis/patología , Receptor Toll-Like 7/agonistas
16.
Nature ; 488(7410): 222-5, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22801499

RESUMEN

The herpes virus entry mediator (HVEM), a member of the tumour-necrosis factor receptor family, has diverse functions, augmenting or inhibiting the immune response. HVEM was recently reported as a colitis risk locus in patients, and in a mouse model of colitis we demonstrated an anti-inflammatory role for HVEM, but its mechanism of action in the mucosal immune system was unknown. Here we report an important role for epithelial HVEM in innate mucosal defence against pathogenic bacteria. HVEM enhances immune responses by NF-κB-inducing kinase-dependent Stat3 activation, which promotes the epithelial expression of genes important for immunity. During intestinal Citrobacter rodentium infection, a mouse model for enteropathogenic Escherichia coli infection, Hvem−/− mice showed decreased Stat3 activation, impaired responses in the colon, higher bacterial burdens and increased mortality. We identified the immunoglobulin superfamily molecule CD160 (refs 7 and 8), expressed predominantly by innate-like intraepithelial lymphocytes, as the ligand engaging epithelial HVEM for host protection. Likewise, in pulmonary Streptococcus pneumoniae infection, HVEM is also required for host defence. Our results pinpoint HVEM as an important orchestrator of mucosal immunity, integrating signals from innate lymphocytes to induce optimal epithelial Stat3 activation, which indicates that targeting HVEM with agonists could improve host defence.


Asunto(s)
Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Inmunidad Mucosa/inmunología , Membrana Mucosa/inmunología , Membrana Mucosa/microbiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Carga Bacteriana , Línea Celular , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enteropatógena , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Infecciones por Escherichia coli , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Intestinos/inmunología , Intestinos/microbiología , Ligandos , Pulmón/inmunología , Pulmón/microbiología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/metabolismo , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/deficiencia , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Factor de Transcripción STAT3/metabolismo , Streptococcus pneumoniae/inmunología , Tasa de Supervivencia , Quinasa de Factor Nuclear kappa B
17.
J Immunol ; 187(10): 5069-76, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21998450

RESUMEN

The critical role of Langerhans cells (LC) in contact hypersensitivity (CHS) was recently questioned in studies using different LC-depletion mouse models. On one hand, inducible ablation of LC led to diminished ear swelling, suggesting functional redundancy between LC and (Langerin(+)) dermal dendritic cells (DC). On the other hand, constitutive or acute depletion of LC resulted in an enhanced reaction, supporting a regulatory role of LC in CHS. To address this controversy by conditional gene targeting, we generated Langerin-Cre knockin mice. Breeding these mice to a Cre-reporter strain demonstrated robust and specific DNA recombination in LC, as well as other Langerin(+) tissue DC. In agreement with the vital requirement of TGF-ß signaling for LC development, crossing Langerin-Cre to mice homozygous for a loxP-flanked TGF-ßR1 allele resulted in permanent LC deficiency, whereas the homeostasis of dermal Langerin(+) DC was unaffected. In the absence of LC, induction of CHS in these Langerin(+) DC-specific TGF-ßR1-deficient mice elicited decreased ear swelling compared with controls. This novel approach provided further evidence against a regulatory function of LC in CHS. Moreover, these Langerin-Cre mice represent a unique and powerful tool to dissect the role and molecular control of Langerin(+) DC populations beyond LC.


Asunto(s)
Antígenos CD/genética , Dermatitis Alérgica por Contacto/inmunología , Eliminación de Gen , Integrasas/genética , Células de Langerhans/inmunología , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Lectinas de Unión a Manosa/deficiencia , Lectinas de Unión a Manosa/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Animales , Antígenos CD/biosíntesis , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Células Dendríticas/patología , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/inmunología , Técnicas de Sustitución del Gen/métodos , Marcación de Gen , Células de Langerhans/enzimología , Células de Langerhans/patología , Lectinas Tipo C/biosíntesis , Lectinas de Unión a Manosa/biosíntesis , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología
18.
J Immunol ; 187(6): 3104-10, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21844385

RESUMEN

An immunoregulatory role of aryl hydrocarbon receptor (AhR) has been shown in conventional αß and γδ T cells, but its function in skin γδ T cells (dendritic epidermal T cells [DETC]) is unknown. In this study, we demonstrate that DETC express AhR in wild-type mice, and are specifically absent in the epidermis of AhR-deficient mice (AhR-KO). We show that DETC precursors are generated in the thymus and home to the skin. Proliferation of DETC in the skin was impaired in AhR-KO mice, resulting in a >90% loss compared with wild type. Surprisingly, DETC were not replaced by αß T cells or conventional γδ T cells, suggesting a limited time frame for seeding this niche. We found that DETC from AhR-KO mice failed to express the receptor tyrosine kinase c-Kit, a known growth factor for γδ T cells in the gut. Moreover, we found that c-kit is a direct target of AhR, and propose that AhR-dependent c-Kit expression is potentially involved in DETC homeostasis. DETC are a major source of GM-CSF in the skin. Recently, we had shown that impaired Langerhans cell maturation in AhR-KO is related to low GM-CSF levels. Our findings suggest that the DETCs are necessary for LC maturation, and provide insights into a novel role for AhR in the maintenance of skin-specific γδ T cells, and its consequences for the skin immune network.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Epidermis/inmunología , Homeostasis/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Linfocitos T/inmunología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Separación Celular , Células Epidérmicas , Epidermis/metabolismo , Femenino , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...