Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
iScience ; 26(11): 108134, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37867943

RESUMEN

AIM2 is an interferon-inducible HIN-200 protein family member and is well-documented for its roles in innate immune responses as a DNA sensor. Recent studies have highlighted AIM2's function on regulatory T cells (Treg) and follicular T cells (Tfh). However, its involvement in Th17 cell differentiation remains unclear. This study reveals that AIM2 promotes Th17 cell differentiation. AIM2 deficiency decreases IL-17A production and downregulates key Th17 associated proteins (RORγt, IL-1R1, IL-23R). AIM2 is located in the nucleus of Th17 cells, where it interacts with RORγt, enhancing its binding to the Il17a promoter. The absence of AIM2 hinders naive CD4 T cells from differentiating into functional Th17 cells and from inducing colitis in Rag1-/- mice. This study uncovers AIM2's role as a regulator of Th17 cell transcriptional programming, highlighting its potential as a therapeutic target for Th17 cell-mediated inflammatory diseases.

2.
J Clin Invest ; 133(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37856217

RESUMEN

A20 is a ubiquitin-modifying protein that negatively regulates NF-κB signaling. Mutations in A20/TNFAIP3 are associated with a variety of autoimmune diseases, including multiple sclerosis (MS). We found that deletion of A20 in central nervous system (CNS) endothelial cells (ECs) enhances experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. A20ΔCNS-EC mice showed increased numbers of CNS-infiltrating immune cells during neuroinflammation and in the steady state. While the integrity of the blood-brain barrier (BBB) was not impaired, we observed a strong activation of CNS-ECs in these mice, with dramatically increased levels of the adhesion molecules ICAM-1 and VCAM-1. We discovered ICOSL to be expressed by A20-deficient CNS-ECs, which we found to function as adhesion molecules. Silencing of ICOSL in CNS microvascular ECs partly reversed the phenotype of A20ΔCNS-EC mice without reaching statistical significance and delayed the onset of EAE symptoms in WT mice. In addition, blocking of ICOSL on primary mouse brain microvascular ECs impaired the adhesion of T cells in vitro. Taken together, we propose that CNS EC-ICOSL contributes to the firm adhesion of T cells to the BBB, promoting their entry into the CNS and eventually driving neuroinflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Enfermedades Neuroinflamatorias , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Animales , Ratones , Barrera Hematoencefálica/metabolismo , Sistema Nervioso Central/metabolismo , Células Endoteliales/metabolismo , Ratones Endogámicos C57BL , Esclerosis Múltiple/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Linfocitos T/metabolismo , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
3.
Cells ; 11(14)2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35883631

RESUMEN

Heterodimeric ß2 integrin surface receptors (CD11a-d/CD18) are specifically expressed by leukocytes that contribute to pathogen uptake, cell migration, immunological synapse formation and cell signaling. In humans, the loss of CD18 expression results in leukocyte adhesion deficiency syndrome (LAD-)1, largely characterized by recurrent severe infections. All available mouse models display the constitutive and ubiquitous knockout of either α or the common ß2 (CD18) subunit, which hampers the analysis of the cell type-specific role of ß2 integrins in vivo. To overcome this limitation, we generated a CD18 gene floxed mouse strain. Offspring generated from crossing with CD11c-Cre mice displayed the efficient knockdown of ß2 integrins, specifically in dendritic cells (DCs). Stimulated ß2-integrin-deficient splenic DCs showed enhanced cytokine production and the concomitantly elevated activity of signal transducers and activators of transcription (STAT) 1, 3 and 5, as well as the impaired expression of suppressor of cytokine signaling (SOCS) 2-6 as assessed in bone marrow-derived (BM) DCs. Paradoxically, these BMDCs also showed the attenuated expression of genes involved in inflammatory signaling. In line, in experimental autoimmune encephalomyelitis mice with a conditional DC-specific ß2 integrin knockdown presented with a delayed onset and milder course of disease, associated with lower frequencies of T helper cell populations (Th)1/Th17 in the inflamed spinal cord. Altogether, our mouse model may prove to be a valuable tool to study the leukocyte-specific functions of ß2 integrins in vivo.


Asunto(s)
Antígenos CD18 , Células Dendríticas , Encefalomielitis Autoinmune Experimental , Inflamación , Animales , Antígenos CD18/genética , Antígenos CD18/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Encefalomielitis , Encefalomielitis Autoinmune Experimental/genética , Expresión Génica , Inflamación/genética , Síndrome de Deficiencia de Adhesión del Leucocito , Ratones
4.
Cell Mol Immunol ; 18(1): 182-193, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31988493

RESUMEN

The vast majority of Foxp3+ regulatory T cells (Tregs) are generated in the thymus, and several factors, such as cytokines and unique thymic antigen-presenting cells, are known to contribute to the development of these thymus-derived Tregs (tTregs). Here, we report the existence of a specific subset of Foxp3+ Tregs within the thymus that is characterized by the expression of IL-1R2, which is a decoy receptor for the inflammatory cytokine IL-1. Detailed flow cytometric analysis of the thymocytes from Foxp3hCD2xRAG1GFP reporter mice revealed that the IL-1R2+ Tregs are mainly RAG1GFP- and CCR6+CCR7-, demonstrating that these Tregs are recirculating cells entering the thymus from the periphery and that they have an activated phenotype. In the spleen, the majority of IL-1R2+ Tregs express neuropilin-1 (Nrp-1) and Helios, suggesting a thymic origin for these Tregs. Interestingly, among all tissues studied, the highest frequency of IL-1R2+ Tregs was observed in the thymus, indicating preferential recruitment of this Treg subset by the thymus. Using fetal thymic organ cultures (FTOCs), we demonstrated that increased concentrations of exogenous IL-1ß blocked intrathymic Treg development, resulting in a decreased frequency of CD25+Foxp3+ tTregs and an accumulation of CD25+Foxp3- Treg precursors. Interestingly, the addition of IL-1R2+ Tregs, but not IL-1R2- Tregs, to reaggregated thymic organ cultures (RTOCs) abrogated the IL-1ß-mediated blockade, demonstrating that these recirculating IL-1R2+ Tregs can quench IL-1 signaling in the thymus and thereby maintain thymic Treg development even under inflammatory conditions.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/fisiología , Inflamación/inmunología , Receptores Tipo II de Interleucina-1/metabolismo , Linfocitos T Reguladores/citología , Timocitos/citología , Animales , Citocinas/metabolismo , Proteínas de Homeodominio/fisiología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Timocitos/inmunología , Timocitos/metabolismo
5.
J Immunol ; 204(4): 747-751, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31924653

RESUMEN

IL-6 binds to the IL-6R α-chain (IL-6Rα) and signals via the signal transducer gp130. Recently, IL-6 was found to also bind to the cell surface glycoprotein CD5, which would then engage gp130 in the absence of IL-6Rα. However, the biological relevance of this alternative pathway is under debate. In this study, we developed a mouse model, in which murine IL-6 is overexpressed in a CD11c-Cre-dependent manner. Transgenic mice developed a lethal immune dysregulation syndrome with increased numbers of Ly-6G+ neutrophils and Ly-6Chi monocytes/macrophages. IL-6 overexpression promoted activation of CD4+ T cells while suppressing CD5+ B-1a cell development. However, additional ablation of IL-6Rα protected IL-6-overexpressing mice from IL-6-triggered inflammation and fully phenocopied IL-6Rα-deficient mice without IL-6 overexpression. Mechanistically, IL-6Rα deficiency completely prevented downstream activation of STAT3 in response to IL-6. Altogether, our data clarify that IL-6Rα is the only biologically relevant receptor for IL-6 in mice.


Asunto(s)
Interleucina-6/inmunología , Receptores de Interleucina-6/inmunología , Transducción de Señal/inmunología , Animales , Ratones , Ratones Transgénicos
6.
J Mol Med (Berl) ; 96(10): 983-992, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30109367

RESUMEN

The pleiotropic cytokine IL-1 mediates its biological functions via association with the signaling receptor IL-1R1. Despite an apparent simplicity in IL-1 signaling activation, multiple negative regulators have been identified. The decoy receptor IL-1R2 (also known as CD121b) can suppress IL-1 maturation, sequester its active forms or hinder the signaling complex assembly. IL-1R2 is differentially expressed among numerous cell types and displays cis- and trans- modes of action. In this review, we link different forms of IL-1R2 (membrane-bound (mIL-1R2), secreted (sIL-1R2), shedded (shIL-1R2), cytoplasmic, and intracellular domain (IL-1R2ICD) restricted) with their ability to interfere with IL-1, thereby regulating immune responses. We also discuss the intriguing possible function of IL-1R2 as a transcriptional regulator. Finally, we summarize the known impact of IL-1R2 in disease pathogenesis and discuss its potential role in treatment of inflammatory conditions.


Asunto(s)
Interleucina-1/metabolismo , Receptores Tipo II de Interleucina-1/metabolismo , Animales , Humanos , Transducción de Señal
7.
EMBO J ; 36(1): 102-115, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-27827809

RESUMEN

Interleukin-1 (IL-1) is implicated in numerous pathologies, including multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). However, the exact mechanism by which IL-1 is involved in the generation of pathogenic T cells and in disease development remains largely unknown. We found that following EAE induction, pertussis toxin administration leads to IL-1 receptor type 1 (IL-1R1)-dependent IL-1ß expression by myeloid cells in the draining lymph nodes. This myeloid-derived IL-1ß did not vitally contribute to the generation and plasticity of Th17 cells, but rather promoted the expansion of a GM-CSF+ Th17 cell subset, thereby enhancing its encephalitogenic potential. Lack of expansion of GM-CSF-producing Th17 cells led to ameliorated disease in mice deficient for IL-1R1 specifically in T cells. Importantly, pathogenicity of IL-1R1-deficient T cells was fully restored by IL-23 polarization and expansion in vitro Therefore, our data demonstrate that IL-1 functions as a mitogenic mediator of encephalitogenic Th17 cells rather than qualitative inducer of their generation.


Asunto(s)
Proliferación Celular , Encefalomielitis Autoinmune Experimental/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/análisis , Interleucina-1/metabolismo , Células Th17/química , Células Th17/fisiología , Animales , Ratones , Toxina del Pertussis/administración & dosificación , Toxina del Pertussis/toxicidad
8.
PLoS One ; 11(8): e0161505, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27551932

RESUMEN

Interleukin-1 (IL-1) plays a crucial role in numerous inflammatory diseases via action on its only known signaling IL-1 receptor type 1 (IL-1R1). To investigate the role of IL-1 signaling in selected cell types, we generated a new mouse strain in which exon 5 of the Il1r1 gene is flanked by loxP sites. Crossing of these mice with CD4-Cre transgenic mice resulted in IL-1R1 loss of function specifically in T cells. These mice, termed IL-1R1ΔT, displayed normal development under steady state conditions. Importantly, isolated CD4 positive T cells retained their capacity to differentiate toward Th1 or Th17 cell lineages in vitro, and strongly proliferated in cultures supplemented with either anti-CD3/CD28 or Concanavalin A, but, as predicted, were completely unresponsive to IL-1ß administration. Furthermore, IL-1R1ΔT mice were protected from gut inflammation in the anti-CD3 treatment model, due to dramatically reduced frequencies and absolute numbers of IL-17A and interferon (IFN)-γ producing cells. Taken together, our data shows the necessity of intact IL-1 signaling for survival and expansion of CD4 T cells that were developed in an otherwise IL-1 sufficient environment.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/genética , Citocinas/biosíntesis , Receptores Tipo I de Interleucina-1/deficiencia , Subgrupos de Linfocitos T/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Linfocitos B/metabolismo , Biomarcadores , Complejo CD3/inmunología , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Marcación de Gen , Inmunofenotipificación , Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Activación de Linfocitos , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...