Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Immunol ; 18(6): 633-641, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28459434

RESUMEN

Microglia and other tissue-resident macrophages within the central nervous system (CNS) have essential roles in neural development, inflammation and homeostasis. However, the molecular pathways underlying their development and function remain poorly understood. Here we report that mice deficient in NRROS, a myeloid-expressed transmembrane protein in the endoplasmic reticulum, develop spontaneous neurological disorders. NRROS-deficient (Nrros-/-) mice show defects in motor functions and die before 6 months of age. Nrros-/- mice display astrogliosis and lack normal CD11bhiCD45lo microglia, but they show no detectable demyelination or neuronal loss. Instead, perivascular macrophage-like myeloid cells populate the Nrros-/- CNS. Cx3cr1-driven deletion of Nrros shows its crucial role in microglial establishment during early embryonic stages. NRROS is required for normal expression of Sall1 and other microglial genes that are important for microglial development and function. Our study reveals a NRROS-mediated pathway that controls CNS-resident macrophage development and affects neurological function.


Asunto(s)
Astrocitos/metabolismo , Sistema Nervioso Central/embriología , Regulación del Desarrollo de la Expresión Génica , Microglía/metabolismo , Células Mieloides/metabolismo , Enfermedades del Sistema Nervioso/genética , Proteínas/genética , Animales , Astrocitos/citología , Western Blotting , Sistema Nervioso Central/citología , Citometría de Flujo , Inmunohistoquímica , Cojera Animal/genética , Proteínas de Unión a TGF-beta Latente , Locomoción , Macrófagos/citología , Macrófagos/metabolismo , Proteínas de la Membrana , Ratones , Ratones Noqueados , Microglía/citología , Células Mieloides/citología , Postura , Factores de Transcripción/genética , Incontinencia Urinaria/genética , Retención Urinaria/genética
2.
Immunity ; 42(2): 321-331, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25680273

RESUMEN

T helper 1 (Th1) cell-associated immunity exacerbates ileitis induced by oral Toxoplasma gondii infection. We show here that attenuated ileitis observed in interleukin-22 (IL-22)-deficient mice was associated with reduced production of Th1-cell-promoting IL-18. IL-22 not only augmented the expression of Il18 mRNA and inactive precursor protein (proIL-18) in intestinal epithelial cells after T. gondii or Citrobacter rodentium infection, but also maintained the homeostatic amount of proIL-18 in the ileum. IL-22, however, did not induce the processing to active IL-18, suggesting a two-step regulation of IL-18 in these cells. Although IL-18 exerted pathogenic functions during ileitis triggered by T. gondii, it was required for host defense against C. rodentium. Conversely, IL-18 was required for the expression of IL-22 in innate lymphoid cells (ILCs) upon T. gondii infection. Our results define IL-18 as an IL-22 target gene in epithelial cells and describe a complex mutual regulation of both cytokines during intestinal infection.


Asunto(s)
Infecciones por Enterobacteriaceae/inmunología , Interleucina-18/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Toxoplasmosis/inmunología , Animales , Células Cultivadas , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/microbiología , Células Epiteliales/inmunología , Ileítis/inmunología , Ileítis/microbiología , Ileítis/parasitología , Íleon/inmunología , Íleon/microbiología , Íleon/parasitología , Inflamación/inmunología , Interferón gamma/biosíntesis , Interleucina-18/biosíntesis , Interleucinas/genética , Mucosa Intestinal/microbiología , Mucosa Intestinal/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Técnicas de Cultivo de Órganos , ARN Mensajero/biosíntesis , Células TH1/inmunología , Toxoplasma/inmunología , Toxoplasmosis/parasitología , Regulación hacia Arriba , Interleucina-22
3.
Nat Immunol ; 12(10): 941-8, 2011 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-21874025

RESUMEN

Colonic patches (CLPs) and isolated lymphoid follicles (ILFs) are two main lymphoid structures in the colon. Lymphoid tissue-inducer cells (LTi cells) are indispensable for the development of ILFs. LTi cells also produce interleukin 17 (IL-17) and IL-22, signature cytokines secreted by IL-17-producing helper T cells. Here we report that IL-22 acted downstream of the lymphotoxin pathway and regulated the organization and maintenance of mature CLPs and ILFs in the colon during infection with Citrobacter rodentium. Lymphotoxin (LTα(1)ß(2)) regulated the production of IL-22 during infection with C. rodentium, but the lymphotoxin-like protein LIGHT did not. IL-22 signaling was sufficient to restore the organization of CLPs and ILFs and host defense against infection with C. rodentium in mice lacking lymphotoxin signals, which suggests that IL-22 connects the lymphotoxin pathway to mucosal epithelial defense mechanisms.


Asunto(s)
Citrobacter rodentium , Colon/inmunología , Infecciones por Enterobacteriaceae/inmunología , Interleucinas/fisiología , Tejido Linfoide/fisiología , Linfotoxina-alfa/fisiología , Animales , Colon/microbiología , Interleucina-23/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Interleucina-22
4.
Nat Immunol ; 12(12): 1238-45, 2011 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22001828

RESUMEN

Interleukin 22 (IL-22), which is produced by cells of the T(H)17 subset of helper T cells and other leukocytes, not only enhances proinflammatory innate defense mechanisms in epithelial cells but also provides crucial protection to tissues from damage caused by inflammation and infection. In T(H)17 cells, transforming growth factor-ß (TGF-ß) regulates IL-22 and IL-17 differently. IL-6 alone induces T cells to produce only IL-22, whereas the combination of IL-6 and high concentrations of TGF-ß results in the production of IL-17 but not IL-22 by T cells. Here we identify the transcription factor c-Maf, which is induced by TGF-ß, as a downstream repressor of Il22. We found that c-Maf bound to the Il22 promoter and was both necessary and sufficient for the TGF-ß-dependent suppression of IL-22 production in T(H)17 cells.


Asunto(s)
Interleucinas/biosíntesis , Proteínas Proto-Oncogénicas c-maf/metabolismo , Células Th17/inmunología , Factor de Crecimiento Transformador beta/farmacología , Animales , Secuencia de Bases , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Sitios de Unión/genética , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Interleucinas/genética , Ratones , Ratones Endogámicos BALB C , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-maf/genética , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Células Th17/efectos de los fármacos , Transcripción Genética , Interleucina-22
5.
J Immunol ; 202(7): 1935-1941, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30770417

RESUMEN

IL-17 family cytokines are critical to host defense responses at cutaneous and mucosal surfaces. Whereas IL-17A, IL-17F, and IL-17C induce overlapping inflammatory cascades to promote neutrophil-mediated immunity, IL-17E/IL-25 drives type 2 immune pathways and eosinophil activity. Genetic and pharmacological studies reveal the significant contribution these cytokines play in antimicrobial and autoimmune mechanisms. However, little is known about the related family member, IL-17B, with contrasting reports of both pro- and anti-inflammatory function in rodents. We demonstrate that in the human immune system, IL-17B is functionally similar to IL-25 and elicits type 2 cytokine secretion from innate type 2 lymphocytes, NKT, and CD4+ CRTH2+ Th2 cells. Like IL-25, this activity is dependent on the IL-17RA and IL-17RB receptor subunits. Furthermore, IL-17B can augment IL-33-driven type 2 responses. These data position IL-17B as a novel component in the regulation of human type 2 immunity.


Asunto(s)
Inmunidad Innata/inmunología , Interleucina-17/inmunología , Receptores de Interleucina-17/inmunología , Subgrupos de Linfocitos T/inmunología , Humanos , Inflamación/inmunología
6.
J Immunol ; 202(1): 183-193, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30510070

RESUMEN

Both common and rare genetic variants of laccase domain-containing 1 (LACC1, previously C13orf31) are associated with inflammatory bowel disease, leprosy, Behcet disease, and systemic juvenile idiopathic arthritis. However, the functional relevance of these variants is unclear. In this study, we use LACC1-deficient mice to gain insight into the role of LACC1 in regulating inflammation. Following oral administration of Citrobacter rodentium, LACC1 knockout (KO) mice had more severe colon lesions compared with wildtype (WT) controls. Immunization with collagen II, a collagen-induced arthritis (CIA) model, resulted in an accelerated onset of arthritis and significantly worse arthritis and inflammation in LACC1 KO mice. Similar results were obtained in a mannan-induced arthritis model. Serum and local TNF in CIA paws and C. rodentium colons were significantly increased in LACC1 KO mice compared with WT controls. The percentage of IL-17A-producing CD4+ T cells was elevated in LACC1 KO mice undergoing CIA as well as aged mice compared with WT controls. Neutralization of IL-17, but not TNF, prevented enhanced mannan-induced arthritis in LACC1 KO mice. These data provide new mechanistic insight into the function of LACC1 in regulating TNF and IL-17 during inflammatory responses. We hypothesize that these effects contribute to immune-driven pathologies observed in individuals carrying LACC1 variants.


Asunto(s)
Artritis Experimental/inmunología , Artritis Juvenil/inmunología , Citrobacter rodentium/fisiología , Infecciones por Enterobacteriaceae/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oxidorreductasas/metabolismo , Células Th17/inmunología , Alelos , Animales , Artritis Experimental/microbiología , Artritis Juvenil/genética , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/genética , Interleucina-17/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidorreductasas/genética , Polimorfismo Genético , Factores de Necrosis Tumoral/metabolismo
7.
Nature ; 514(7521): 237-41, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25119041

RESUMEN

The connection between an altered gut microbiota and metabolic disorders such as obesity, diabetes, and cardiovascular disease is well established. Defects in preserving the integrity of the mucosal barriers can result in systemic endotoxaemia that contributes to chronic low-grade inflammation, which further promotes the development of metabolic syndrome. Interleukin (IL)-22 exerts essential roles in eliciting antimicrobial immunity and maintaining mucosal barrier integrity within the intestine. Here we investigate the connection between IL-22 and metabolic disorders. We find that the induction of IL-22 from innate lymphoid cells and CD4(+) T cells is impaired in obese mice under various immune challenges, especially in the colon during infection with Citrobacter rodentium. While innate lymphoid cell populations are largely intact in obese mice, the upregulation of IL-23, a cytokine upstream of IL-22, is compromised during the infection. Consequently, these mice are susceptible to C. rodentium infection, and both exogenous IL-22 and IL-23 are able to restore the mucosal host defence. Importantly, we further unveil unexpected functions of IL-22 in regulating metabolism. Mice deficient in IL-22 receptor and fed with high-fat diet are prone to developing metabolic disorders. Strikingly, administration of exogenous IL-22 in genetically obese leptin-receptor-deficient (db/db) mice and mice fed with high-fat diet reverses many of the metabolic symptoms, including hyperglycaemia and insulin resistance. IL-22 shows diverse metabolic benefits, as it improves insulin sensitivity, preserves gut mucosal barrier and endocrine functions, decreases endotoxaemia and chronic inflammation, and regulates lipid metabolism in liver and adipose tissues. In summary, we identify the IL-22 pathway as a novel target for therapeutic intervention in metabolic diseases.


Asunto(s)
Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Inmunidad Mucosa , Interleucinas/inmunología , Interleucinas/metabolismo , Enfermedades Metabólicas/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Crónica , Citrobacter rodentium/efectos de los fármacos , Citrobacter rodentium/inmunología , Citrobacter rodentium/fisiología , Colon/efectos de los fármacos , Colon/inmunología , Colon/microbiología , Diabetes Mellitus/patología , Dieta Alta en Grasa , Femenino , Hiperglucemia/dietoterapia , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Inmunidad Mucosa/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Insulina/metabolismo , Resistencia a la Insulina , Interleucina-23/inmunología , Interleucina-23/metabolismo , Interleucina-23/farmacología , Interleucinas/farmacología , Interleucinas/uso terapéutico , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Enfermedades Metabólicas/dietoterapia , Enfermedades Metabólicas/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Receptores de Interleucina/deficiencia , Receptores de Interleucina/metabolismo , Receptores de Leptina/deficiencia , Receptores de Leptina/metabolismo , Interleucina-22
8.
Nature ; 509(7499): 235-9, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24739962

RESUMEN

Reactive oxygen species (ROS) produced by phagocytes are essential for host defence against bacterial and fungal infections. Individuals with defective ROS production machinery develop chronic granulomatous disease. Conversely, excessive ROS can cause collateral tissue damage during inflammatory processes and therefore needs to be tightly regulated. Here we describe a protein, we termed negative regulator of ROS (NRROS), which limits ROS generation by phagocytes during inflammatory responses. NRROS expression in phagocytes can be repressed by inflammatory signals. NRROS-deficient phagocytes produce increased ROS upon inflammatory challenges, and mice lacking NRROS in their phagocytes show enhanced bactericidal activity against Escherichia coli and Listeria monocytogenes. Conversely, these mice develop severe experimental autoimmune encephalomyelitis owing to oxidative tissue damage in the central nervous system. Mechanistically, NRROS is localized to the endoplasmic reticulum, where it directly interacts with nascent NOX2 (also known as gp91(phox) and encoded by Cybb) monomer, one of the membrane-bound subunits of the NADPH oxidase complex, and facilitates the degradation of NOX2 through the endoplasmic-reticulum-associated degradation pathway. Thus, NRROS provides a hitherto undefined mechanism for regulating ROS production--one that enables phagocytes to produce higher amounts of ROS, if required to control invading pathogens, while minimizing unwanted collateral tissue damage.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Escherichia coli/inmunología , Listeria monocytogenes/inmunología , Proteínas/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Animales , Autoinmunidad/genética , Células de la Médula Ósea/citología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/metabolismo , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Proteínas de Unión a TGF-beta Latente , Macrófagos/citología , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana , Ratones , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Fagocitos/citología , Fagocitos/inmunología , Fagocitos/metabolismo , Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo
9.
Microbiome ; 11(1): 47, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36894983

RESUMEN

BACKGROUND: IL-22 is induced by aryl hydrocarbon receptor (AhR) signaling and plays a critical role in gastrointestinal barrier function through effects on antimicrobial protein production, mucus secretion, and epithelial cell differentiation and proliferation, giving it the potential to modulate the microbiome through these direct and indirect effects. Furthermore, the microbiome can in turn influence IL-22 production through the synthesis of L-tryptophan (L-Trp)-derived AhR ligands, creating the prospect of a host-microbiome feedback loop. We evaluated the impact IL-22 may have on the gut microbiome and its ability to activate host AhR signaling by observing changes in gut microbiome composition, function, and AhR ligand production following exogenous IL-22 treatment in both mice and humans. RESULTS: Microbiome alterations were observed across the gastrointestinal tract of IL-22-treated mice, accompanied by an increased microbial functional capacity for L-Trp metabolism. Bacterially derived indole derivatives were increased in stool from IL-22-treated mice and correlated with increased fecal AhR activity. In humans, reduced fecal concentrations of indole derivatives in ulcerative colitis (UC) patients compared to healthy volunteers were accompanied by a trend towards reduced fecal AhR activity. Following exogenous IL-22 treatment in UC patients, both fecal AhR activity and concentrations of indole derivatives increased over time compared to placebo-treated UC patients. CONCLUSIONS: Overall, our findings indicate IL-22 shapes gut microbiome composition and function, which leads to increased AhR signaling and suggests exogenous IL-22 modulation of the microbiome may have functional significance in a disease setting. Video Abstract.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Animales , Ratones , Receptores de Hidrocarburo de Aril/metabolismo , Interleucinas , Indoles , Interleucina-22
10.
J Immunol ; 185(10): 6041-8, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20926799

RESUMEN

The trans presentation of IL-15 by cells expressing the specific high-affinity receptor α-chain (IL-15Rα) to cells expressing the signaling receptor ß-chain and γ-chain is essential for the generation and maintenance of CD8 memory T cells, NK cells, and NKT cells in an in vivo mouse system. We have also demonstrated in vitro that cell-surface IL-15Rα on cells expressing all the receptor components present IL-15 to receptor ß-chain/γ-chain coexpressed on the same cell surface (cis presentation). However, although mouse CD8 T cells express all the IL-15R components, they show no evidence of cis presentation. In this study, we demonstrate that increased expression of mouse IL-15Rα in mouse CD8 T cells by retrovirus-mediated gene transfer changes the ability of the T cell to use cis presentation on the cell surface, indicating that cis presentation requires high expression of mouse IL-15Rα on the cell surface. Using cell lines expressing human or mouse receptors, we demonstrate that cis presentation occurs more efficiently in the human receptor-ligand combination than in that of the mouse system. Moreover, we found that primary human CD8 T cells do not require trans presentation of human IL-15 in vitro. These findings raise the possibility that the maintenance and generation of memory CD8 T cells are achieved via distinct mechanisms in humans and mice. Therefore, careful study of the human immune system, rather than extrapolation from the murine model, is necessary to achieve more complete understanding of memory CD8 T cell development in humans.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Interleucina-15/metabolismo , Activación de Linfocitos/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Separación Celular , Citometría de Flujo , Humanos , Interleucina-15/química , Interleucina-15/inmunología , Subunidad alfa del Receptor de Interleucina-15/inmunología , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
11.
J Immunol ; 184(8): 4307-16, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20231694

RESUMEN

It has been suggested that IL-17RC forms a complex with IL-17RA to mediate the functions of IL-17A and IL-17F homodimers as well as IL-17A/F heterodimers. It is still unclear whether IL-17RC is absolutely required for the signaling of IL-17 cytokines in vivo. By using Il-17rc-deficient mice, we show that IL-17RC is essential for the signaling of IL-17A, IL-17F, and IL-17A/F both in vitro and in vivo. IL-17RC does not preassociate with IL-17RA on the cell surface; rather IL-17A can induce the formation of an IL-17RC and IL-17RA complex. This process is not dependent on the intracellular similar expression to fibroblast growth factor genes and IL-17Rs (SEFIR) domain of IL-17RC, but the SEFIR is essential in IL-17A signal transduction. Finally, Il-17rc(-/-) mice develop much milder disease in an experimental autoimmune encephalomyelitis model, supporting an essential role for IL-17RC in mediating immune-mediated CNS inflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Interleucina-17/fisiología , Receptores de Interleucina/fisiología , Transducción de Señal/inmunología , Animales , Línea Celular Transformada , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Técnicas de Cocultivo , Encefalomielitis Autoinmune Experimental/patología , Humanos , Interleucinas/fisiología , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Receptores de Interleucina/deficiencia , Receptores de Interleucina/genética
12.
Nat Commun ; 13(1): 6079, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36241643

RESUMEN

NOX2 is the prototypical member of the NADPH oxidase NOX superfamily and produces superoxide (O2•-), a key reactive oxygen species (ROS) that is essential in innate and adaptive immunity. Mutations that lead to deficiency in NOX2 activity correlate with increased susceptibility to bacterial and fungal infections, resulting in chronic granulomatous disease. The core of NOX2 is formed by a heterodimeric transmembrane complex composed of NOX2 (formerly gp91) and p22, but a detailed description of its structural architecture is lacking. Here, we present the structure of the human NOX2 core complex bound to a selective anti-NOX2 antibody fragment. The core complex reveals an intricate extracellular topology of NOX2, a four-transmembrane fold of the p22 subunit, and an extensive transmembrane interface which provides insights into NOX2 assembly and activation. Functional assays uncover an inhibitory activity of the 7G5 antibody mediated by internalization-dependent and internalization-independent mechanisms. Overall, our results provide insights into the NOX2 core complex architecture, disease-causing mutations, and potential avenues for selective NOX2 pharmacological modulation.


Asunto(s)
NADPH Oxidasas , Superóxidos , Humanos , Fragmentos de Inmunoglobulinas , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo
13.
Sci Immunol ; 6(59)2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33963061

RESUMEN

Repair of the intestinal epithelium is tightly regulated to maintain homeostasis. The response after epithelial damage needs to be local and proportional to the insult. How different types of damage are coupled to repair remains incompletely understood. We report that after distinct types of intestinal epithelial damage, IL-1R1 signaling in GREM1+ mesenchymal cells increases production of R-spondin 3 (RSPO3), a Wnt agonist required for intestinal stem cell self-renewal. In parallel, IL-1R1 signaling regulates IL-22 production by innate lymphoid cells and promotes epithelial hyperplasia and regeneration. Although the regulation of both RSPO3 and IL-22 is critical for epithelial recovery from Citrobacter rodentium infection, IL-1R1-dependent RSPO3 production by GREM1+ mesenchymal cells alone is sufficient and required for recovery after dextran sulfate sodium-induced colitis. These data demonstrate how IL-1R1-dependent signaling orchestrates distinct repair programs tailored to the type of injury sustained that are required to restore intestinal epithelial barrier function.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae/inmunología , Mucosa Intestinal/fisiología , Receptores Tipo I de Interleucina-1/inmunología , Animales , Células Cultivadas , Técnicas de Cocultivo , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Sulfato de Dextran , Células Epiteliales , Fibroblastos , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones Transgénicos , Organoides , Receptores Tipo I de Interleucina-1/genética , Regeneración , Transducción de Señal , Trombospondinas/inmunología , Interleucina-22
14.
J Clin Immunol ; 30(2): 185-95, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20177959

RESUMEN

INTRODUCTION: Accumulating evidence suggests that the interleukin (IL)-17 cytokines are major players in the immune response to foreign pathogens. In addition, the pathogeneses of a number of inflammatory diseases have been linked to uncontrolled expression of these cytokine pathways. DISCUSSION: Genetic and biochemical analyses have elucidated the cellular and molecular events triggered by these proteins during an inflammatory response. While significant efforts have been placed on understanding the functions of IL-17A, IL-17F, and IL-17E, the significance of the other family members, IL-17B-D, in inflammation remains to be determined. CONCLUSION: This review will focus on the cellular sources, target cell/receptors that are utilized by these cytokines to control pathogenesis, and the therapeutic potential of targeting these pathways to treat inflammatory disorders.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Interleucina-17/inmunología , Receptores de Interleucina-17/inmunología , Animales , Humanos , Péptidos y Proteínas de Señalización Intercelular/inmunología , Homología de Secuencia de Aminoácido , Células TH1/inmunología , Células Th2/inmunología
15.
Immunohorizons ; 2(5): 164-171, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31022698

RESUMEN

Intestinal epithelial cells form a physical barrier that is tightly regulated to control intestinal permeability. Proinflammatory cytokines, such as TNF-α, increase epithelial permeability through disruption of epithelial junctions. The regulation of the epithelial barrier in inflammatory gastrointestinal disease remains to be fully characterized. In this article, we show that the human inflammatory bowel disease genetic susceptibility gene C1ORF106 plays a key role in regulating gut epithelial permeability. C1ORF106 directly interacts with cytohesins to maintain functional epithelial cell junctions. C1orf106-deficient mice are hypersensitive to TNF-α-induced increase in epithelial permeability, and this is associated with increased diarrhea. This study identifies C1ORF106 as an epithelial cell junction protein, and the loss of C1ORF106 augments TNF-α-induced intestinal epithelial leakage and diarrhea that may play a critical role in the development of inflammatory bowel disease.


Asunto(s)
Proteínas Portadoras/genética , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Animales , Células CACO-2 , Proteínas Portadoras/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Células Epiteliales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Permeabilidad , Receptores Citoplasmáticos y Nucleares/metabolismo , Uniones Estrechas/genética , Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/genética
16.
Sci Signal ; 10(475)2017 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-28420753

RESUMEN

Tumor progression locus 2 (TPL2; also known as MAP3K8) is a mitogen-activated protein kinase (MAPK) kinase kinase (MAP3K) that phosphorylates the MAPK kinases MEK1 and MEK2 (MEK1/2), which, in turn, activate the MAPKs extracellular signal-regulated kinase 1 (ERK1) and ERK2 (ERK1/2) in macrophages stimulated through the interleukin-1 receptor (IL-1R), Toll-like receptors (TLRs), or the tumor necrosis factor receptor (TNFR). We describe a conserved and critical role for TPL2 in mediating the effector functions of neutrophils through the activation of the p38 MAPK signaling pathway. Gene expression profiling and functional studies of neutrophils and monocytes revealed a MEK1/2-independent branch point downstream of TPL2 in neutrophils. Biochemical analyses identified the MAPK kinases MEK3 and MEK6 and the MAPKs p38α and p38δ as downstream effectors of TPL2 in these cells. Genetic ablation of the catalytic activity of TPL2 or therapeutic intervention with a TPL2-specific inhibitor reduced the production of inflammatory mediators by neutrophils in response to stimulation with the TLR4 agonist lipopolysaccharide (LPS) in vitro, as well as in rodent models of inflammatory disease. Together, these data suggest that TPL2 is a drug target that activates not only MEK1/2-dependent but also MEK3/6-dependent signaling to promote inflammatory responses.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Activación Neutrófila , Neutrófilos/enzimología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Activación Enzimática , Inflamación/enzimología , Inflamación/genética , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , MAP Quinasa Quinasa 6/genética , MAP Quinasa Quinasa 6/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
17.
J Biol Chem ; 282(51): 37191-204, 2007 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17947230

RESUMEN

Interleukin (IL)-15 is a pleiotropic cytokine that plays a pivotal role in both innate and adaptive immunity. IL-15 is unique among cytokines due to its participation in a trans signaling mechanism in which IL-15 receptor alpha (IL-15Ralpha) from one subset of cells presents IL-15 to neighboring IL-2Rbeta/gammac-expressing cells. Here we present the crystal structure of IL-15 in complex with the sushi domain of IL-15Ralpha. The structure reveals that the alpha receptor-binding epitope of IL-15 adopts a unique conformation, which, together with amino acid substitutions, permits specific interactions with IL-15Ralpha that account for the exceptionally high affinity of the IL-15.IL-15Ralpha complex. Interestingly, analysis of the topology of IL-15 and IL-15Ralpha at the IL-15.IL-15Ralpha interface suggests that IL-15 should be capable of participating in a cis signaling mechanism similar to that of the related cytokine IL-2. Indeed, we present biochemical data demonstrating that IL-15 is capable of efficiently signaling in cis through IL-15Ralpha and IL-2Rbeta/gammac expressed on the surface of a single cell. Based on our data we propose that cis presentation of IL-15 may be important in certain biological contexts and that flexibility of IL-15Ralpha permits IL-15 and its three receptor components to be assembled identically at the ligand-receptor interface whether IL-15 is presented in cis or trans. Finally, we have gained insights into IL-15.IL-15Ralpha.IL-2Rbeta.gammac quaternary complex assembly through the use of molecular modeling.


Asunto(s)
Epítopos/química , Interleucina-15/química , Modelos Moleculares , Complejos Multiproteicos/química , Animales , Sitios de Unión/fisiología , Cristalografía por Rayos X , Epítopos/metabolismo , Subunidad gamma Común de Receptores de Interleucina/química , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Interleucina-15/metabolismo , Subunidad beta del Receptor de Interleucina-2/química , Subunidad beta del Receptor de Interleucina-2/metabolismo , Ratones , Complejos Multiproteicos/metabolismo , Estructura Cuaternaria de Proteína , Receptores de Interleucina-15/química , Receptores de Interleucina-15/metabolismo , Transducción de Señal/fisiología
18.
J Biol Chem ; 281(50): 38257-65, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17050525

RESUMEN

Vav proteins are multidomain signaling molecules critical for mediating signals downstream of several surface receptors, including the antigen receptors of T and B lymphocytes. The catalytic guanine nucleotide exchange factor (GEF) activity of the Vav Dbl homology (DH) domain is thought to be controlled by an intramolecular autoinhibitory mechanism involving an N-terminal extension and phosphorylation of tyrosine residues in the acidic region (AC). Here, we report that the sequences surrounding the Vav1 AC: Tyr(142), Tyr(160), and Tyr(174) are evolutionarily conserved, conform to consensus SH2 domain binding motifs, and bind several proteins implicated in TCR signaling, including Lck, PI3K p85alpha, and PLCgamma1, through direct interactions with their SH2 domains. In addition, the AC tyrosines regulate tyrosine phosphorylation of Vav1. We also show that Tyr(174) is required for the maintenance of TCR-signaling microclusters and for normal T cell development and activation. In this regard, our data demonstrate that while Vav1 Tyr(174) is essential for maintaining the inhibitory constraint of the DH domain in both developing and mature T cells, constitutively activated Vav GEF disrupts TCR-signaling microclusters and leads to defective T cell development and proliferation.


Asunto(s)
Activación de Linfocitos , Proteínas Proto-Oncogénicas c-vav/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/citología , Tirosina/fisiología , Proliferación Celular , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-vav/química , Dominios Homologos src
19.
Immunol Rev ; 202: 157-74, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15546392

RESUMEN

T-helper 1 cell (Th1) development participates in immunity to many pathogens in part by providing a source of interferon (IFN)-gamma that contributes numerous protective effects. The process of Th1 development involves signals provided by antigen-presenting cells and cytokines produced in response to pathogens, with IFN-gamma itself, interleukin (IL)-12, and IL-18 each promoting the process in some way. Despite the rapid progress into mechanisms of Th1 development in recent years, there are still a number of important unresolved issues in this area. The precise sequence of effector and cellular mechanisms represents a relatively recent avenue of research but is still the subject of current debate, as is the basis of mechanisms that may stabilize a Th1 response. Another unresolved issue is the role of type I IFNs in substituting for IL-12-mediated activation of signal transducer and activator of transcription 4 (Stat4) and induction of IFN-gamma in either murine or human T cells. It is now clear that Th1 cells acquire the property of being capable of nonantigen-dependent activation through the coordinate signaling of IL-12 and IL-18, but the precise order of intracellular signaling events and the uniqueness of this pathway's reliance on the p38 mitogen-activated protein kinase (MAPK) pathway are still issues in need of resolution. Finally, the process of verifying the effects of Stat4 mutations on functional responses has led to the recognition of an unexpected action of the STAT N-domain that may apply generally to other STAT proteins as well. None of these areas is static or resolved fully, and they likely will remain topics of rapid progress.


Asunto(s)
Diferenciación Celular/fisiología , Subgrupos de Linfocitos T/fisiología , Células TH1/fisiología , Animales , Diferenciación Celular/genética , Citocinas/fisiología , Dimerización , Humanos , Interferón gamma/fisiología , Ratones , Subgrupos de Linfocitos T/citología , Células TH1/citología , Factores de Transcripción/fisiología
20.
Nat Immunol ; 5(2): 208-15, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14704793

RESUMEN

The N-terminal protein interaction domain (N-domain) of the signal transducer and activator of transcription-4 (STAT4) is believed to stabilize interactions between two phosphorylated STAT4 dimers to form STAT4 tetramers. Here, we show that nonphosphorylated STAT4 dimers form in vivo before cytokine receptor-driven activation. Mutations in the N-domain dimerization interface abolished assembly of nonphosphorylated STAT4 dimers and prevented STAT4 phosphorylation mediated by cytokine receptors. In addition, N-domain dimerization occurred for other STAT family members but was homotypic in character. This implies a conserved role for N-domain dimerization, which might include influencing interactions with cytokine receptors, favoring homodimer formation or accelerating formation of the phosphorylated STAT dimer.


Asunto(s)
Citocinas/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/inmunología , Transactivadores/química , Transactivadores/inmunología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dimerización , Humanos , Técnicas In Vitro , Ratones , Modelos Moleculares , Mutación , Fosforilación , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Factor de Transcripción STAT4 , Transactivadores/genética , Transactivadores/metabolismo , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA