Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 8(6)2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36787195

RESUMEN

Low Club Cell 16 kDa protein (CC16) plasma levels are linked to accelerated lung function decline in patients with chronic obstructive pulmonary disease (COPD). Cigarette smoke-exposed (CS-exposed) Cc16-/- mice have exaggerated COPD-like disease associated with increased NF-κB activation in their lungs. It is unclear whether CC16 augmentation can reverse exaggerated COPD in CS-exposed Cc16-/- mice and whether increased NF-κB activation contributes to the exaggerated COPD in CS-exposed Cc16-/- lungs. CS-exposed WT and Cc16-/- mice were treated with recombinant human CC16 (rhCC16) or an NF-κB inhibitor versus vehicle beginning at the midpoint of the exposures. COPD-like disease and NF-κB activation were measured in the lungs. RhCC16 limited the progression of emphysema, small airway fibrosis, and chronic bronchitis-like disease in WT and Cc16-/- mice partly by reducing pulmonary inflammation (reducing myeloid leukocytes and/or increasing regulatory T and/or B cells) and alveolar septal cell apoptosis, reducing NF-κB activation in CS-exposed Cc16-/- lungs, and rescuing the reduced Foxj1 expression in CS-exposed Cc16-/- lungs. IMD0354 treatment reduced exaggerated lung inflammation and rescued the reduced Foxj1 expression in CS-exposed Cc16-/- mice. RhCC16 treatment reduced NF-κB activation in luciferase reporter A549 cells. Thus, rhCC16 treatment limits COPD progression in CS-exposed Cc16-/- mice partly by inhibiting NF-κB activation and represents a potentially novel therapeutic approach for COPD.


Asunto(s)
Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Animales , Humanos , Ratones , Pulmón/metabolismo , FN-kappa B/metabolismo , Neumonía/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/metabolismo , Nicotiana
2.
Immunity ; 55(9): 1663-1679.e6, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36070768

RESUMEN

Interleukin-23 receptor plays a critical role in inducing inflammation and autoimmunity. Here, we report that Th1-like cells differentiated in vitro with IL-12 + IL-21 showed similar IL-23R expression to that of pathogenic Th17 cells using eGFP reporter mice. Fate mapping established that these cells did not transition through a Th17 cell state prior to becoming Th1-like cells, and we observed their emergence in vivo in the T cell adoptive transfer colitis model. Using IL-23R-deficient Th1-like cells, we demonstrated that IL-23R was required for the development of a highly colitogenic phenotype. Single-cell RNA sequencing analysis of intestinal T cells identified IL-23R-dependent genes in Th1-like cells that differed from those expressed in Th17 cells. The perturbation of one of these regulators (CD160) in Th1-like cells inhibited the induction of colitis. We thus uncouple IL-23R as a purely Th17 cell-specific factor and implicate IL-23R signaling as a pathogenic driver in Th1-like cells inducing tissue inflammation.


Asunto(s)
Colitis , Receptores de Interleucina , Animales , Inflamación/metabolismo , Interleucina-23/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Células TH1 , Células Th17
3.
Cell Rep ; 33(8): 108433, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33238123

RESUMEN

Interleukin-27 (IL-27) is an immunoregulatory cytokine that suppresses inflammation through multiple mechanisms, including induction of IL-10, but the transcriptional network mediating its diverse functions remains unclear. Combining temporal RNA profiling with computational algorithms, we predict 79 transcription factors induced by IL-27 in T cells. We validate 11 known and discover 5 positive (Cebpb, Fosl2, Tbx21, Hlx, and Atf3) and 2 negative (Irf9 and Irf8) Il10 regulators, generating an experimentally refined regulatory network for Il10. We report two central regulators, Prdm1 and Maf, that cooperatively drive the expression of signature genes induced by IL-27 in type 1 regulatory T cells, mediate IL-10 expression in all T helper cells, and determine the regulatory phenotype of colonic Foxp3+ regulatory T cells. Prdm1/Maf double-knockout mice develop spontaneous colitis, phenocopying ll10-deficient mice. Our work provides insights into IL-27-driven transcriptional networks and identifies two shared Il10 regulators that orchestrate immunoregulatory programs across T helper cell subsets.


Asunto(s)
Redes Reguladoras de Genes/genética , Interleucina-10/metabolismo , Interleucina-27/metabolismo , Células TH1/metabolismo , Animales , Humanos , Ratones , Ratones Noqueados
4.
Immunity ; 51(4): 696-708.e9, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31618654

RESUMEN

Signaling abnormalities in immune responses in the small intestine can trigger chronic type 2 inflammation involving interaction of multiple immune cell types. To systematically characterize this response, we analyzed 58,067 immune cells from the mouse small intestine by single-cell RNA sequencing (scRNA-seq) at steady state and after induction of a type 2 inflammatory reaction to ovalbumin (OVA). Computational analysis revealed broad shifts in both cell-type composition and cell programs in response to the inflammation, especially in group 2 innate lymphoid cells (ILC2s). Inflammation induced the expression of exon 5 of Calca, which encodes the alpha-calcitonin gene-related peptide (α-CGRP), in intestinal KLRG1+ ILC2s. α-CGRP antagonized KLRG1+ ILC2s proliferation but promoted IL-5 expression. Genetic perturbation of α-CGRP increased the proportion of intestinal KLRG1+ ILC2s. Our work highlights a model where α-CGRP-mediated neuronal signaling is critical for suppressing ILC2 expansion and maintaining homeostasis of the type 2 immune machinery.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Inflamación/inmunología , Intestinos/inmunología , Linfocitos/inmunología , Neuropéptidos/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Células Cultivadas , Biología Computacional , Inmunidad Innata , Interleucina-5/genética , Interleucina-5/metabolismo , Lectinas Tipo C/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neuropéptidos/genética , Receptores Inmunológicos/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Células Th2/inmunología , Transcriptoma , Regulación hacia Arriba
5.
Immunity ; 51(4): 709-723.e6, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31604686

RESUMEN

Neuroimmune interactions have emerged as critical modulators of allergic inflammation, and type 2 innate lymphoid cells (ILC2s) are an important cell type for mediating these interactions. Here, we show that ILC2s expressed both the neuropeptide calcitonin gene-related peptide (CGRP) and its receptor. CGRP potently inhibited alarmin-driven type 2 cytokine production and proliferation by lung ILC2s both in vitro and in vivo. CGRP induced marked changes in ILC2 expression programs in vivo and in vitro, attenuating alarmin-driven proliferative and effector responses. A distinct subset of ILCs scored highly for a CGRP-specific gene signature after in vivo alarmin stimulation, suggesting CGRP regulated this response. Finally, we observed increased ILC2 proliferation and type 2 cytokine production as well as exaggerated responses to alarmins in mice lacking the CGRP receptor. Together, these data indicate that endogenous CGRP is a critical negative regulator of ILC2 responses in vivo.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Linfocitos/fisiología , Neuropéptidos/metabolismo , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Alarminas/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Proliferación Celular , Células Cultivadas , Retroalimentación Fisiológica , Inmunidad Innata , Interleucina-33/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación , Neuropéptidos/genética , Receptores de Péptido Relacionado con el Gen de Calcitonina/genética , Transducción de Señal , Células Th2/inmunología
6.
JCI Insight ; 3(24)2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30568032

RESUMEN

Matrix metalloproteinase-9 (MMP-9) cleaves various proteins to regulate inflammatory and injury responses. However, MMP-9's activities during influenza A viral (IAV) infections are incompletely understood. Herein, plasma MMP-9 levels were increased in patients with pandemic H1N1 and seasonal IAV infections. MMP-9 lung levels were increased and localized to airway epithelial cells and leukocytes in H1N1-infected WT murine lungs. H1N1-infected Mmp-9-/- mice had lower mortality rates, reduced weight loss, lower lung viral titers, and reduced lung injury, along with lower E-cadherin shedding in bronchoalveolar lavage fluid (BALF) samples than WT mice. H1N1-infected Mmp-9-/- mice had an altered immune response to IAV with lower BALF PMN and macrophage counts, higher Th1-like CD4+ and CD8+ T cell subsets, lower T regulatory cell counts, reduced lung type I interferon levels, and higher lung interferon-γ levels. Mmp-9 bone marrow-chimera studies revealed that Mmp-9 deficiency in lung parenchymal cells protected mice from IAV-induced mortality. H1N1-infected Mmp-9-/- lung epithelial cells had lower viral titers than H1N1-infected WT cells in vitro. Thus, H1N1-infected Mmp-9-/- mice are protected from IAV-induced lung disease due to a more effective adaptive immune response to IAV and reduced epithelial barrier injury due partly to reduced E-cadherin shedding. Thus, we believe that MMP-9 is a novel therapeutic target for IAV infections.


Asunto(s)
Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/inmunología , Gripe Humana/prevención & control , Interferón Tipo I/metabolismo , Interferón gamma , Leucocitos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/prevención & control , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pérdida de Peso
7.
Immunol Rev ; 286(1): 53-73, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30294962

RESUMEN

Type 2 immunity against pathogens is tightly regulated to ensure appropriate inflammatory responses that clear infection and prevent excessive tissue damage. Recent research has shown that type 2 innate lymphoid cells (ILC2s) contribute to steady-state tissue integrity and exert tissue-specific functions. However, upon exposure to inflammatory stimuli, they also initiate and amplify type 2 inflammation by inducing mucus production, eosinophilia, and Th2 differentiation. In this review, we discuss the regulation of ILC2 activation by transcription factors and metabolic pathways, as well as by extrinsic signals such as cytokines, lipid mediators, hormones, and neuropeptides. We also review recent discoveries about ILC2 plasticity and heterogeneity in different tissues, as revealed partly through single-cell RNA sequencing of transcriptional responses to various stimuli. Understanding the tissue-specific pathways that regulate ILC2 diversity and function is a critical step in the development of potential therapies for allergic diseases.


Asunto(s)
Inflamación/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular , Citocinas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunidad Innata , Activación de Linfocitos , Análisis de la Célula Individual
8.
Nat Med ; 24(10): 1625-1626, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30013196

RESUMEN

In the version of this article initially published, the line graph showing TNF-α levels in Fig. 2d was inadvertently duplicated. A graph of IL-6 levels should be shown in place of the duplication.These results were also incorrectly described in the main text, which originally stated: "At an early time point of infection (6 h), RTX-treated mice showed higher induction of total inflammatory-protein levels in the bronchoalveolar lavage fluid (BALF) (Fig. 2c), as well as levels of the cytokines TNF-α and IL-6, and the chemokine CXCL-1 (Fig. 2d)". This should instead read: "At an early time point of infection (6 h), RTX-treated mice showed higher induction of total inflammatory-protein levels in the bronchoalveolar lavage fluid (BALF) (Fig. 2c), as well as levels of the cytokine TNF-α and the chemokine CXCL-1 (Fig. 2d)".In the supplementary information initially posted online, incorrect bar graphs were presented in Supplementary Fig. 1b (VG, TRPV1+ data, top panel) and Supplementary Fig. 4b (DRG, CGRP+ data, middle panel).

9.
Nature ; 558(7710): 454-459, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29899446

RESUMEN

The expression of co-inhibitory receptors, such as CTLA-4 and PD-1, on effector T cells is a key mechanism for ensuring immune homeostasis. Dysregulated expression of co-inhibitory receptors on CD4+ T cells promotes autoimmunity, whereas sustained overexpression on CD8+ T cells promotes T cell dysfunction or exhaustion, leading to impaired ability to clear chronic viral infections and diseases such as cancer1,2. Here, using RNA and protein expression profiling at single-cell resolution in mouse cells, we identify a module of co-inhibitory receptors that includes not only several known co-inhibitory receptors (PD-1, TIM-3, LAG-3 and TIGIT) but also many new surface receptors. We functionally validated two new co-inhibitory receptors, activated protein C receptor (PROCR) and podoplanin (PDPN). The module of co-inhibitory receptors is co-expressed in both CD4+ and CD8+ T cells and is part of a larger co-inhibitory gene program that is shared by non-responsive T cells in several physiological contexts and is driven by the immunoregulatory cytokine IL-27. Computational analysis identified the transcription factors PRDM1 and c-MAF as cooperative regulators of the co-inhibitory module, and this was validated experimentally. This molecular circuit underlies the co-expression of co-inhibitory receptors in T cells and identifies regulators of T cell function with the potential to control autoimmunity and tumour immunity.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Redes Reguladoras de Genes/genética , Melanoma/inmunología , Transcripción Genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Tolerancia Inmunológica/genética , Tolerancia Inmunológica/inmunología , Interleucina-27/inmunología , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Melanoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reproducibilidad de los Resultados
10.
Nat Med ; 24(4): 417-426, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29505031

RESUMEN

Lung-innervating nociceptor sensory neurons detect noxious or harmful stimuli and consequently protect organisms by mediating coughing, pain, and bronchoconstriction. However, the role of sensory neurons in pulmonary host defense is unclear. Here, we found that TRPV1+ nociceptors suppressed protective immunity against lethal Staphylococcus aureus pneumonia. Targeted TRPV1+-neuron ablation increased survival, cytokine induction, and lung bacterial clearance. Nociceptors suppressed the recruitment and surveillance of neutrophils, and altered lung γδ T cell numbers, which are necessary for immunity. Vagal ganglia TRPV1+ afferents mediated immunosuppression through release of the neuropeptide calcitonin gene-related peptide (CGRP). Targeting neuroimmunological signaling may be an effective approach to treat lung infections and bacterial pneumonia.


Asunto(s)
Infecciones Bacterianas/inmunología , Neutrófilos/metabolismo , Nociceptores/metabolismo , Neumonía/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Células Receptoras Sensoriales/metabolismo , Linfocitos T/inmunología , Animales , Infecciones Bacterianas/microbiología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Citocinas/metabolismo , Femenino , Interacciones Huésped-Patógeno/inmunología , Masculino , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Neumonía/microbiología , Neumonía/patología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/fisiología , Canales Catiónicos TRPV/metabolismo , Nervio Vago/metabolismo
12.
Nature ; 549(7672): 351-356, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28902842

RESUMEN

Type 2 innate lymphoid cells (ILC2s) both contribute to mucosal homeostasis and initiate pathologic inflammation in allergic asthma. However, the signals that direct ILC2s to promote homeostasis versus inflammation are unclear. To identify such molecular cues, we profiled mouse lung-resident ILCs using single-cell RNA sequencing at steady state and after in vivo stimulation with the alarmin cytokines IL-25 and IL-33. ILC2s were transcriptionally heterogeneous after activation, with subpopulations distinguished by expression of proliferative, homeostatic and effector genes. The neuropeptide receptor Nmur1 was preferentially expressed by ILC2s at steady state and after IL-25 stimulation. Neuromedin U (NMU), the ligand of NMUR1, activated ILC2s in vitro, and in vivo co-administration of NMU with IL-25 strongly amplified allergic inflammation. Loss of NMU-NMUR1 signalling reduced ILC2 frequency and effector function, and altered transcriptional programs following allergen challenge in vivo. Thus, NMUR1 signalling promotes inflammatory ILC2 responses, highlighting the importance of neuro-immune crosstalk in allergic inflammation at mucosal surfaces.


Asunto(s)
Hipersensibilidad/inmunología , Hipersensibilidad/patología , Inflamación/inmunología , Inflamación/patología , Pulmón/patología , Linfocitos/inmunología , Neuropéptidos/metabolismo , Animales , Femenino , Regulación de la Expresión Génica , Inmunidad Innata/inmunología , Interleucina-17/inmunología , Interleucina-33/inmunología , Ligandos , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Neurotransmisores/biosíntesis , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Transducción de Señal , Transcripción Genética
13.
Cell ; 167(7): 1669, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984714

RESUMEN

IL-17A both directly induces and synergizes with other cytokines to promote autoimmune tissue inflammation. Secukinumab and ixekizumab are monoclonal antibodies (mAb) that inhibit interleukin-17A. These two agents were recently approved for treatment of psoriasis, and secukinumab is also approved for treatment of two spondyloarthropathies, psoriatic arthritis and ankylosing spondylitis.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Interleucina-17/antagonistas & inhibidores , Psoriasis/tratamiento farmacológico , Humanos
14.
Neuron ; 87(2): 341-54, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26119026

RESUMEN

Lung nociceptors initiate cough and bronchoconstriction. To elucidate if these fibers also contribute to allergic airway inflammation, we stimulated lung nociceptors with capsaicin and observed increased neuropeptide release and immune cell infiltration. In contrast, ablating Nav1.8(+) sensory neurons or silencing them with QX-314, a charged sodium channel inhibitor that enters via large-pore ion channels to specifically block nociceptors, substantially reduced ovalbumin- or house-dust-mite-induced airway inflammation and bronchial hyperresponsiveness. We also discovered that IL-5, a cytokine produced by activated immune cells, acts directly on nociceptors to induce the release of vasoactive intestinal peptide (VIP). VIP then stimulates CD4(+) and resident innate lymphoid type 2 cells, creating an inflammatory signaling loop that promotes allergic inflammation. Our results indicate that nociceptors amplify pathological adaptive immune responses and that silencing these neurons with QX-314 interrupts this neuro-immune interplay, revealing a potential new therapeutic strategy for asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Nociceptores/fisiología , Hipersensibilidad Respiratoria/inmunología , Anestésicos Locales/farmacología , Animales , Animales Recién Nacidos , Capsaicina/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Adyuvante de Freund/toxicidad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interleucina-5/metabolismo , Lidocaína/análogos & derivados , Lidocaína/farmacología , Ratones , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Ovalbúmina/toxicidad , Hipersensibilidad Respiratoria/inducido químicamente , Factores de Tiempo , Péptido Intestinal Vasoactivo/metabolismo
15.
J Clin Invest ; 125(6): 2211-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25961452

RESUMEN

Cytokines play a critical role in controlling the differentiation of CD4 Th cells into distinct subsets, including IL-17-producing Th17 cells. Unfortunately, the incidence of a number of autoimmune diseases, particularly those in which the IL-23/IL-17 axis has been implicated, has risen in the last several decades, suggesting that environmental factors can promote autoimmunity. Here we review the role of cytokines in Th17 differentiation, particularly the role of IL-23 in promoting the differentiation of a pathogenic subset of Th17 cells that potently induce autoimmune tissue inflammation. Moreover, we highlight emerging data that indicate that environmental factors, including the intestinal microbiota and changes in diet, can alter normal cytokine regulation with potent effects on Th17 differentiation and thus promote autoimmunity, which has strong implications for human disease.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Diferenciación Celular/inmunología , Exposición a Riesgos Ambientales/efectos adversos , Intestinos , Microbiota/inmunología , Células Th17/inmunología , Animales , Enfermedades Autoinmunes/patología , Humanos , Interleucina-17/inmunología , Interleucina-23/inmunología , Intestinos/inmunología , Intestinos/microbiología , Células Th17/patología
16.
J Clin Invest ; 125(1): 129-40, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25415436

RESUMEN

Podoplanin (PDPN, also known as Gp38) is highly expressed on the surface of lymphatic endothelial cells, where it regulates development of lymphatic vessels. We have recently observed that PDPN is also expressed on effector T cells that infiltrate target tissues during autoimmune inflammation; however, the function of PDPN in T cells is largely unclear. Here, we demonstrated that global deletion of Pdpn results in exaggerated T cell responses and spontaneous experimental autoimmune encephalomyelitis (EAE) in mice with a susceptible genetic background. In contrast, T cell-specific overexpression of PDPN resulted in profound defects in IL-7-mediated T cell expansion and survival. Consequently, these animals exhibited a more rapid resolution of CNS inflammation, characterized by a reduced effector CD4+ T cell population in the CNS. Mice harboring a T cell-specific deletion of Pdpn developed exacerbated EAE, with increased accumulation of effector CD4+ T cells in the CNS. Transcriptional profiling of naturally occurring PDPN+ effector T cells in the CNS revealed increased expression of other inhibitory receptors, such as Pd1 and Tim3, and decreased expression of prosurvival factors, including Il7ra. Together, our data suggest that PDPN functions as an inhibitory molecule on T cells, thereby promoting tissue tolerance by limiting long-term survival and maintenance of CD4+ effector T cells in target organs.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Glicoproteínas de Membrana/fisiología , Animales , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Linfopenia/inmunología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados
17.
J Immunol ; 194(3): 863-7, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25539814

RESUMEN

Asthma is a chronic inflammatory disease that fails to resolve. Recently, a key role for type 2 innate lymphoid cells (ILC2s) was linked to asthma pathogenesis; however, mechanisms for ILC2 regulation remain to be determined. In this study, metabololipidomics of murine lungs identified temporal changes in endogenous maresin 1 (MaR1) during self-limited allergic inflammation. Exogenous MaR1 reduced lung inflammation and ILC2 expression of IL-5 and IL-13 and increased amphiregulin. MaR1 augmented de novo generation of regulatory T cells (Tregs), which interacted with ILC2s to markedly suppress cytokine production in a TGF-ß-dependent manner. Ab-mediated depletion of Tregs interrupted MaR1 control of ILC2 expression of IL-13 in vivo. Together, the findings uncover Tregs as potent regulators of ILC2 activation; MaR1 targets Tregs and ILC2s to restrain allergic lung inflammation, suggesting MaR1 as the basis for a new proresolving therapeutic approach to asthma and other chronic inflammatory diseases.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Inmunidad Innata , Activación de Linfocitos , Neumonía/inmunología , Neumonía/metabolismo , Subgrupos de Linfocitos T , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Inmunomodulación , Ratones , Ratones Transgénicos , Neumonía/terapia
18.
Immunity ; 40(4): 569-81, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24745333

RESUMEN

Foxp3(+) T regulatory (Treg) cells regulate immune responses and maintain self-tolerance. Recent work shows that Treg cells are comprised of many subpopulations with specialized regulatory functions. Here we identified Foxp3(+) T cells expressing the coinhibitory molecule TIGIT as a distinct Treg cell subset that specifically suppresses proinflammatory T helper 1 (Th1) and Th17 cell, but not Th2 cell responses. Transcriptional profiling characterized TIGIT(+) Treg cells as an activated Treg cell subset with high expression of Treg signature genes. Ligation of TIGIT on Treg cells induced expression of the effector molecule fibrinogen-like protein 2 (Fgl2), which promoted Treg-cell-mediated suppression of T effector cell proliferation. In addition, Fgl2 was necessary to prevent suppression of Th2 cytokine production in a model of allergic airway inflammation. TIGIT expression therefore identifies a Treg cell subset that demonstrates selectivity for suppression of Th1 and Th17 cell but not Th2 cell responses.


Asunto(s)
Fibrinógeno/metabolismo , Receptores Inmunológicos/metabolismo , Hipersensibilidad Respiratoria/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Eosinófilos/inmunología , Fibrinógeno/genética , Fibrinógeno/inmunología , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Terapia de Inmunosupresión , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Balance Th1 - Th2
19.
Proc Natl Acad Sci U S A ; 110(6): 2270-5, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341605

RESUMEN

Myeloid differentiation primary response protein 88 (MyD88) is classically known as an adaptor, linking TLR and IL-1R to downstream signaling pathways in the innate immune system. In addition to its role in innate immune cells, MyD88 has been shown to play an important role in T cells. How MyD88 regulates helper T-cell differentiation remains largely unknown, however. Here we demonstrate that MyD88 is an important regulator of IL-17-producing CD4(+) T helper cells (Th17) cell proliferation. MyD88-deficient CD4(+) T cells showed a defect in Th17 cell differentiation, but not in Th1 cell or Th2 cell differentiation. The impaired IL-17 production from MyD88-deficient CD4(+) T cells is not a result of defective RAR-related orphan receptor γt (RORγt) expression. Instead, MyD88 is essential for sustaining the mammalian target of rapamycin (mTOR) activation necessary to promote Th17 cell proliferation by linking IL-1 and IL-23 signaling. MyD88-deficient CD4(+) T cells showed impaired mTOR activation and, consequently, reduced Th17 cell proliferation. Importantly, the absence of MyD88 in T cells ameliorated disease in the experimental autoimmune encephalomyelitis model. Taken together, our results demonstrate that MyD88 has a dual function in Th17 cells by delivering IL-1 signaling during the early differentiation stage and integrating IL-23 signaling to the mTOR complex to expand committed Th17 cells.


Asunto(s)
Interleucina-1/metabolismo , Interleucina-23/metabolismo , Factor 88 de Diferenciación Mieloide/inmunología , Serina-Treonina Quinasas TOR/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Inmunidad Innata , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Células Th17/citología , Células Th17/metabolismo
20.
J Exp Med ; 200(7): 825-34, 2004 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-15452177

RESUMEN

The high affinity interleukin (IL)-15 receptor, IL-15Ralpha, is essential for supporting lymphoid homeostasis. To assess whether IL-15Ralpha's role in vivo is to trans present IL-15, we generated mixed bone marrow chimera from IL-15Ralpha- and IL-2/15Rbeta-deficient mice. We find that IL-15Ralpha-competent, IL-2/15Rbeta-deficient cells are able to support IL-15Ralpha-deficient natural killer (NK) and memory CD8+ T cells, thus ruling out secondary signals on these cells and demonstrating that IL-15Ralpha-mediated presentation of IL-15 in trans is the primary mechanism by which IL-15Ralpha functions in vivo. Surprisingly, using IL-15- and IL-15Ralpha-deficient mixed chimera, we also find that IL-15 and IL-15Ralpha must be expressed by the same cells to present IL-15 in trans, indicating that IL-15Ralpha is required on a cellular level for the elaboration of IL-15. These studies indicate that IL-15Ralpha defines homeostatic niches for NK and memory CD8+ T cells by controlling both the production and the presentation of IL-15 in trans to NK and CD8+ memory T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Homeostasis/inmunología , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Receptores de Interleucina-2/metabolismo , Animales , Células de la Médula Ósea , Citometría de Flujo , Memoria Inmunológica/inmunología , Interleucina-15/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , Quimera por Radiación , Receptores de Interleucina-15 , Receptores de Interleucina-2/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA