Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(10): 1316-1326, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34531562

RESUMEN

Environmental allergens, including fungi, insects and mites, trigger type 2 immunity; however, the innate sensing mechanisms and initial signaling events remain unclear. Herein, we demonstrate that allergens trigger RIPK1-caspase 8 ripoptosome activation in epithelial cells. The active caspase 8 subsequently engages caspases 3 and 7, which directly mediate intracellular maturation and release of IL-33, a pro-atopy, innate immunity, alarmin cytokine. Mature IL-33 maintained functional interaction with the cognate ST2 receptor and elicited potent pro-atopy inflammatory activity in vitro and in vivo. Inhibiting caspase 8 pharmacologically and deleting murine Il33 and Casp8 each attenuated allergic inflammation in vivo. Clinical data substantiated ripoptosome activation and IL-33 maturation as likely contributors to human allergic inflammation. Our findings reveal an epithelial barrier, allergen-sensing mechanism that converges on the ripoptosome as an intracellular molecular signaling platform, triggering type 2 innate immune responses. These findings have significant implications for understanding and treating human allergic diseases.


Asunto(s)
Alérgenos/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Adolescente , Animales , Caspasa 8/inmunología , Línea Celular , Línea Celular Tumoral , Niño , Preescolar , Citocinas/inmunología , Células Epiteliales/inmunología , Femenino , Células HEK293 , Humanos , Hipersensibilidad/inmunología , Interleucina-33/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
2.
Immunity ; 57(4): 632-648, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599163

RESUMEN

One of the most significant conceptual advances in immunology in recent history is the recognition that signals from the innate immune system are required for induction of adaptive immune responses. Two breakthroughs were critical in establishing this paradigm: the identification of dendritic cells (DCs) as the cellular link between innate and adaptive immunity and the discovery of pattern recognition receptors (PRRs) as a molecular link that controls innate immune activation as well as DC function. Here, we recount the key events leading to these discoveries and discuss our current understanding of how PRRs shape adaptive immune responses, both indirectly through control of DC function and directly through control of lymphocyte function. In this context, we provide a conceptual framework for how variation in the signals generated by PRR activation, in DCs or other cell types, can influence T cell differentiation and shape the ensuing adaptive immune response.


Asunto(s)
Células Dendríticas , Inmunidad Innata , Inmunidad Adaptativa , Receptores de Reconocimiento de Patrones/metabolismo , Activación de Linfocitos
3.
Nat Immunol ; 21(1): 65-74, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31848486

RESUMEN

The cytokine interleukin (IL)-1ß is a key mediator of antimicrobial immunity as well as autoimmune inflammation. Production of IL-1ß requires transcription by innate immune receptor signaling and maturational cleavage by inflammasomes. Whether this mechanism applies to IL-1ß production seen in T cell-driven autoimmune diseases remains unclear. Here, we describe an inflammasome-independent pathway of IL-1ß production that was triggered upon cognate interactions between effector CD4+ T cells and mononuclear phagocytes (MPs). The cytokine TNF produced by activated CD4+ T cells engaged its receptor TNFR on MPs, leading to pro-IL-1ß synthesis. Membrane-bound FasL, expressed by CD4+ T cells, activated death receptor Fas signaling in MPs, resulting in caspase-8-dependent pro-IL-1ß cleavage. The T cell-instructed IL-1ß resulted in systemic inflammation, whereas absence of TNFR or Fas signaling protected mice from CD4+ T cell-driven autoimmunity. The TNFR-Fas-caspase-8-dependent pathway provides a mechanistic explanation for IL-1ß production and its consequences in CD4+ T cell-driven autoimmune pathology.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Inflamación/patología , Interleucina-1beta/metabolismo , Células Mieloides/metabolismo , Animales , Caspasa 1/genética , Caspasa 8/metabolismo , Células Cultivadas , Células Dendríticas/inmunología , Proteína Ligando Fas/metabolismo , Inmunidad Innata/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
4.
Proc Natl Acad Sci U S A ; 121(34): e2401658121, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39136987

RESUMEN

Alloreactive memory T cells have been implicated as central drivers of transplant rejection. Perplexingly, innate cytokines, such as IL-6, IL-1ß, and IL-12, are also associated with rejection of organ transplants. However, the pathways of innate immune activation in allogeneic transplantation are unclear. While the role of microbial and cell death products has been previously described, we identified alloreactive memory CD4 T cells as the primary triggers of innate inflammation. Memory CD4 T cells engaged MHC II-mismatched dendritic cells (DCs), leading to the production of innate inflammatory cytokines. This innate inflammation was independent of several pattern recognition receptors and was primarily driven by TNF superfamily ligands expressed by alloreactive memory CD4 T cells. Blocking of CD40L and TNFα resulted in dampened inflammation, and mice genetically deficient in these molecules exhibited prolonged survival of cardiac allografts. Furthermore, myeloid cell and CD8 T cell infiltration into cardiac transplants was compromised in both CD40L- and TNFα-deficient recipients. Strikingly, we found that priming of naive alloreactive CD8 T cells was dependent on licensing of DCs by memory CD4 T cells. This study unravels the key mechanisms by which alloreactive memory CD4 T cells contribute to destructive pathology and transplant rejection.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Células Dendríticas , Rechazo de Injerto , Trasplante de Corazón , Inmunidad Innata , Inflamación , Animales , Rechazo de Injerto/inmunología , Ratones , Células Dendríticas/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD4-Positivos/inmunología , Inflamación/inmunología , Inmunidad Innata/inmunología , Ratones Endogámicos C57BL , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Células T de Memoria/inmunología , Ratones Noqueados , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Citocinas/metabolismo , Citocinas/inmunología
5.
Immunity ; 45(2): 267-79, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27496733

RESUMEN

Toll-like receptors (TLRs) and other pattern-recognition receptors (PRRs) sense microbial ligands and initiate signaling to induce inflammatory responses. Although the quality of inflammatory responses is influenced by internalization of TLRs, the role of endosomal maturation in clearing receptors and terminating inflammatory responses is not well understood. Here, we report that Drosophila and mammalian Vps33B proteins play critical roles in the maturation of phagosomes and endosomes following microbial recognition. Vps33B was necessary for clearance of endosomes containing internalized PRRs, failure of which resulted in enhanced signaling and expression of inflammatory mediators. Lack of Vps33B had no effect on trafficking of endosomes containing non-microbial cargo. These findings indicate that Vps33B function is critical for determining the fate of signaling endosomes formed following PRR activation. Exaggerated inflammatory responses dictated by persistence of receptors in aberrant endosomal compartments could therefore contribute to symptoms of ARC syndrome, a disease linked to loss of Vps33B.


Asunto(s)
Artrogriposis/inmunología , Colestasis/inmunología , Proteínas de Drosophila/metabolismo , Endosomas/metabolismo , Infecciones por Escherichia coli/inmunología , Inflamación/inmunología , Macrófagos/fisiología , Insuficiencia Renal/inmunología , Proteínas de Transporte Vesicular/metabolismo , Animales , Animales Modificados Genéticamente , Artrogriposis/genética , Células Cultivadas , Colestasis/genética , Drosophila , Proteínas de Drosophila/genética , Técnicas de Inactivación de Genes , Ratones , Transporte de Proteínas , ARN Interferente Pequeño/genética , Insuficiencia Renal/genética , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Proteínas de Transporte Vesicular/genética
6.
Proc Natl Acad Sci U S A ; 117(48): 30628-30638, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33199625

RESUMEN

Macrophages respond to microbial ligands and various noxious cues by initiating an inflammatory response aimed at eliminating the original pathogenic insult. Transition of macrophages from a proinflammatory state to a reparative state, however, is vital for resolution of inflammation and return to homeostasis. The molecular players governing this transition remain poorly defined. Here, we find that the reparative macrophage transition is dictated by B-cell adapter for PI3K (BCAP). Mice harboring a macrophage-specific deletion of BCAP fail to recover from and succumb to dextran sulfate sodium-induced colitis due to prolonged intestinal inflammation and impaired tissue repair. Following microbial stimulation, gene expression in WT macrophages switches from an early inflammatory signature to a late reparative signature, a process that is hampered in BCAP-deficient macrophages. We find that absence of BCAP hinders inactivation of FOXO1 and GSK3ß, which contributes to their enhanced inflammatory state. BCAP deficiency also results in defective aerobic glycolysis and reduced lactate production. This translates into reduced histone lactylation and decreased expression of reparative macrophage genes. Thus, our results reveal BCAP to be a critical cell-intrinsic switch that regulates transition of inflammatory macrophages to reparative macrophages by imprinting epigenetic changes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Histonas/metabolismo , Macrófagos/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Animales , Ratones , Procesamiento Proteico-Postraduccional
7.
Traffic ; 21(9): 578-589, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32677257

RESUMEN

Loss of the arthrogryposis-renal dysfunction-cholestasis (ARC) syndrome-linked Vps33B protein results in exaggerated inflammatory responses upon activation of receptors of the innate immune system in both vertebrates and flies. However, little is known about the signaling elements downstream of these receptors that are critical for the hypersensitivity of Vps33B mutants. Here, we show that p38b MAP kinase contributes to the enhanced inflammatory responses in flies lacking Vps33B. Loss of p38b mitogen-activated protein kinase (MAPK) reduces enhanced inflammatory responses and prolongs the survival of infected Vps33B deficient flies. The function of p38 MAPK is not limited to its proinflammatory effects downstream of the PGRP-LC receptor as p38 also modulates endosomal trafficking of PGRP-LC and phagocytosis of bacteria. Expression of constitutively active p38b MAPK, but not dominant negative p38b MAPK enhances accumulation of endocytosed PGRP-LC receptors or phagocytosed bacteria within cells. Moreover, p38 MAPK is required for induction of macropinocytosis, an alternate pathway for the downregulation of immune receptors. Together, our data indicate that p38 MAPK activates multiple pathways that can contribute to the dysregulation of innate immune signaling in ARC syndrome.


Asunto(s)
Artrogriposis , Colestasis , Dípteros , Animales , Dípteros/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos , Transporte de Proteínas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Immunol ; 204(10): 2651-2660, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32238461

RESUMEN

Preterm birth (PTB) is a major cause of neonatal mortality and morbidity, often triggered by chorioamnionitis or intrauterine inflammation (IUI) with or without infection. Recently, there has been a strong association of IL-1 with PTB. We hypothesized that IL-1R-associated kinase 1 (IRAK1), a key signaling mediator in the TLR/IL-1 pathway, plays a critical role in PTB. In human fetal membranes (FM) collected immediately after birth from women delivering preterm, p-IRAK1 was significantly increased in all the layers of FM with chorioamnionitis, compared with no-chorioamnionitis subjects. In a preterm rhesus macaque model of IUI given intra-amniotic LPS, induction of p-IRAK1 and downstream proinflammatory signaling mediators were seen in the FM. In a C57BL/6J wild-type PTB mouse model of IUI given intrauterine LPS, an IRAK1 inhibitor significantly decreased PTB and increased live birth in a dose-dependent manner. Furthermore, IRAK1 knockout mice were protected from LPS-induced PTB, which was seen in wild-type controls. Activation of IRAK1 was maintained by K63-mediated ubiquitination in preterm FM of humans with chorioamnionitis and rhesus and mouse IUI models. Mechanistically, IRAK1 induced PTB in the mouse model of IUI by upregulating expression of COX-2. Thus, our data from human, rhesus, and mouse demonstrates a critical role IRAK1 in IUI and inflammation-associated PTB and suggest it as potential therapeutic target in IUI-induced PTB.


Asunto(s)
Membranas Extraembrionarias/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Nacimiento Prematuro/metabolismo , Útero/inmunología , Adulto , Animales , Corioamnionitis , Modelos Animales de Enfermedad , Membranas Extraembrionarias/patología , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Lipopolisacáridos/inmunología , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Nacimiento Prematuro/inmunología , Adulto Joven
9.
Immunity ; 35(6): 1010-22, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22137454

RESUMEN

Activation of pattern recognition receptors on dendritic cells (DCs) and macrophages leads to secretion of cytokines that control differentiation of CD4(+) T cells. The current understanding is that interleukin-6 (IL-6) in combination with transforming growth factor-ß (TGF-ß) leads to generation of T helper 17 (Th17) lineage cells. Here, we have discovered that the cytokine requirements for Th17 cell polarization depend on the site of priming. Although IL-6 played a critical role in Th17 cell lineage priming in the skin and mucosal tissues, it was not required for Th17 cell priming in the spleen. In contrast, IL-1 played an irreplaceable role for priming of Th17 lineage cells in all tissues. Importantly, we have demonstrated that IL-6-independent and -dependent pathways of Th17 cell differentiation are guided by DCs residing in various tissues. These results reveal fundamental differences by which the systemic, mucosal, and cutaneous immune systems guide Th17 cell lineage commitment.


Asunto(s)
Diferenciación Celular/inmunología , Citocinas/fisiología , Células Th17/citología , Células Th17/inmunología , Animales , Antígenos/inmunología , Antígenos CD/metabolismo , Linaje de la Célula , Células Dendríticas/inmunología , Cadenas alfa de Integrinas/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal
10.
J Immunol ; 198(10): 3791-3800, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28483987

RESUMEN

Activation of cells in the adaptive immune system is a highly orchestrated process dictated by multiples cues from the innate immune system. Although the fundamental principles of innate control of adaptive immunity are well established, it is not fully understood how innate cells integrate qualitative pathogenic information to generate tailored protective adaptive immune responses. In this review, we discuss complexities involved in the innate control of adaptive immunity that extend beyond TCR engagement, costimulation, and priming cytokine production but are critical for the generation of protective T cell immunity.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Transducción de Señal , Linfocitos T/inmunología , Animales , Citocinas/inmunología , Humanos , Ratones , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA