Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 13: 923424, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35874724

RESUMEN

Innate lymphoid cells (ILCs) develop from ILC progenitors in the bone marrow. Various ILC precursors (ILCPs) with different ILC subset lineage potentials have been identified based on the expression of cell surface markers and ILC-associated key transcription factor reporter genes. This study characterized an interleukin (IL)-7Rα+IL-18Rα+ ILC progenitor population in the mouse bone marrow with multi-ILC lineage potential on the clonal level. Single-cell gene expression analysis revealed the heterogeneity of this population and identified several subpopulations with specific ILC subset-biased gene expression profiles. The role of IL-18 signaling in the regulation of IL-18Rα+ ILC progenitors and ILC development was further investigated using Il18- and Il18r1-deficient mice, in vitro differentiation assay, and adoptive transfer model. IL-18/IL-18R-mediated signal was found to not be required for early stages of ILC development. While Il18r1-/- lymphoid progenitors were able to generate all ILC subsets in vitro and in vivo like the wild-type counterpart, increased IL-18 level, as often occurred during infection or under stress, suppressed the growth of ILCP/ILC in an IL-18Ra-dependent manner via inhibiting proliferation and inducing apoptosis.


Asunto(s)
Inmunidad Innata , Subunidad alfa del Receptor de Interleucina-18 , Interleucina-18 , Linfocitos , Animales , Diferenciación Celular , Interleucina-18/metabolismo , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Linfocitos/metabolismo , Ratones , Análisis de la Célula Individual
2.
Front Immunol ; 12: 780804, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069552

RESUMEN

Objectives: Currently, cardiovascular risk associated with COVID-19 has been brought to people's attention, but the mechanism is not clear. The aim of this study is to elucidate the mechanisms based on multiple omics data. Methodology: Weighted gene co-expression network analysis (WGCNA) was used to identify key pathways. Combination analysis with aneurysm and atherosclerosis related pathways, hypoxia induced factor-1 (HIF-1) signaling were identified as key pathways of the increased cardiovascular risk associated with COVID-19. ScMLnet algorithm based on scRNA-seq was used to explore the regulation of HIF-1 pathway by intercellular communication. Proteomic analysis was used to detect the regulatory mechanisms between IL18 and HIF-1 signaling pathway. Pseudo time locus analysis was used to study the regulation of HIF1 signaling pathway in macrophages and vascular smooth muscle cells (VSMC) phenotypic transformation. The Virtual Inference of protein-activity by Enriched Regulon (VIPER) analysis was used to study the activity of regulatory proteins. Epigenetic analysis based on methylation revealed epigenetic changes in PBMC after SARS-CoV-2 infection. Potential therapeutic compounds were explored by using Cmap algorithm. Results: HIF-1 signaling pathway is a common key pathway for aneurysms, atherosclerosis and SARS-CoV-2 infection. Intercellular communication analysis showed that macrophage-derived interleukin-18 (IL-18) activates the HIF-1 signaling pathway through IL18R1. Proteomic analysis showed that IL18/IL18R1 promote NF-κB entry into the nucleus, and activated the HIF-1 signaling pathway. Macrophage-derived IL18 promoted the M1 polarization of macrophages and the syntactic phenotype transformation of VSMCs. MAP2K1 mediates the functional regulation of HIF-1 signaling pathway in various cell types. Epigenetic changes in PBMC after COVID-19 infection are characterized by activation of the type I interferon pathway. MEK inhibitors are the promising compounds for the treatment of HIF-1 overactivation. Conclusions: The IL18/IL18R1/HIF1A axis is expected to be an therapeutic target for cardiovascular protection after SARS-CoV-2 infection. MEK inhibitors may be an choice for cardiovascular protection after SARS-COV-2 infection.


Asunto(s)
Aneurisma/etiología , Aneurisma/metabolismo , Aterosclerosis/etiología , Aterosclerosis/metabolismo , COVID-19/sangre , COVID-19/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-18/metabolismo , SARS-CoV-2 , Transducción de Señal , Aneurisma/patología , Aterosclerosis/patología , COVID-19/virología , Estudios de Casos y Controles , Células Cultivadas , Epigénesis Genética , Humanos , Interferón Tipo I/metabolismo , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Proteómica/métodos , RNA-Seq/métodos , Factores de Riesgo , Análisis de la Célula Individual/métodos
3.
Inflamm Res ; 69(5): 497-507, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32193584

RESUMEN

BACKGROUND: Fibrosis in multiple organs increases with age. Circulating fibrocytes are bone-marrow-derived mesenchymal progenitors that contribute to heart, lung, and kidney fibrosis under the diseased conditions. Whether circulating fibrocytes contribute to aging-related fibrosis is very limited. METHODS AND RESULTS: We measured the proportion and differentiation of circulating fibrocytes (CD45+/CD34+/collagen I+) from elders (n = 12) and adults (n = 12) using flow cytometry. Differentiated fibrocytes in the culture dishes were isolated and microarray was performed. The percentage of circulating fibrocytes in elders (1.95 ± 0.43%) was comparable to that in the adults (1.71 ± 0.38%). Cultured fibrocytes displayed enhanced potential of differentiation in the elder group (67.91 ± 5.88%) vs the adult group (44.03 ± 7.98%). In addition, expression of fibroblast activation markers and cell migratory ability were also increased in differentiated fibrocytes from elders. Microarray analysis revealed that differentiated fibrocytes from elders expressed high level of interleukin-18 (IL-18) receptor 1 (IL-18R1). Furthermore, we found IL-18 was elevated in the plasma of elders and IL-18/IL-18R1 was shown to promote fibrocyte differentiation. CONCLUSION: Circulating fibrocytes from elders had an enhanced capacity to differentiate into myofibroblasts, and might contribute to age-dependent fibrosis. Age-dependent increment of differentiation at least in part arose from their enhanced expression of IL-18R1. Inhibiting fibrocyte differentiation might be useful as an adjuvant treatment to delay the fibrosis process in aging population.


Asunto(s)
Envejecimiento , Fibroblastos/citología , Subunidad alfa del Receptor de Interleucina-18 , Interleucina-18 , Adolescente , Adulto , Anciano , Diferenciación Celular , Colágeno Tipo I/genética , Colágeno Tipo II/genética , Citocinas/metabolismo , Femenino , Citometría de Flujo , Humanos , Interleucina-18/metabolismo , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Masculino , Persona de Mediana Edad , Adulto Joven
4.
Proc Natl Acad Sci U S A ; 116(43): 21659-21665, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31591201

RESUMEN

Autism spectrum disorder (ASD) does not have a distinct pathogenesis or effective treatment. Increasing evidence supports the presence of immune dysfunction and inflammation in the brains of children with ASD. In this report, we present data that gene expression of the antiinflammatory cytokine IL-37, as well as of the proinflammatory cytokines IL-18 and TNF, is increased in the amygdala and dorsolateral prefrontal cortex of children with ASD as compared to non-ASD controls. Gene expression of IL-18R, which is a receptor for both IL-18 and IL-37, is also increased in the same brain areas of children with ASD. Interestingly, gene expression of the NTR3/sortilin receptor is reduced in the amygdala and dorsolateral prefrontal cortex. Pretreatment of cultured human microglia from normal adult brains with human recombinant IL-37 (1 to 100 ng/mL) inhibits neurotensin (NT)-stimulated secretion and gene expression of IL-1ß and CXCL8. Another key finding is that NT, as well as the proinflammatory cytokines IL-1ß and TNF increase IL-37 gene expression in cultured human microglia. The data presented here highlight the connection between inflammation and ASD, supporting the development of IL-37 as a potential therapeutic agent of ASD.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Trastorno del Espectro Autista/metabolismo , Interleucina-1/metabolismo , Microglía/metabolismo , Neurotensina/metabolismo , Corteza Prefrontal/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Células Cultivadas , Niño , Humanos , Interleucina-18/metabolismo , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-8/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
5.
Nat Immunol ; 20(8): 1004-1011, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31263280

RESUMEN

Innate lymphoid cells (ILCs) are tissue-resident sentinels that are essential for early host protection from pathogens at initial sites of infection. However, whether pathogen-derived antigens directly modulate the responses of tissue-resident ILCs has remained unclear. In the present study, it was found that liver-resident type 1 ILCs (ILC1s) expanded locally and persisted after the resolution of infection with mouse cytomegalovirus (MCMV). ILC1s acquired stable transcriptional, epigenetic and phenotypic changes a month after the resolution of MCMV infection, and showed an enhanced protective effector response to secondary challenge with MCMV consistent with a memory lymphocyte response. Memory ILC1 responses were dependent on the MCMV-encoded glycoprotein m12, and were independent of bystander activation by proinflammatory cytokines after heterologous infection. Thus, liver ILC1s acquire adaptive features in an MCMV-specific manner.


Asunto(s)
Memoria Inmunológica/inmunología , Hígado/inmunología , Linfocitos/inmunología , Glicoproteínas de Membrana/inmunología , Muromegalovirus/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Inmunidad Innata/inmunología , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Hígado/citología , Ratones
6.
Cell Physiol Biochem ; 45(3): 1034-1050, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29439249

RESUMEN

BACKGROUND/AIMS: Recently, studies have shown that interleukin-37 (IL-37) is involved in atherosclerosis-related diseases. However, the regulatory mechanisms of IL-37 in atherosclerosis remain unknown. This study aims to determine the role of IL-37 in atherosclerosis and to investigate the underlying mechanisms involved. METHODS: IL-37 expression in human atherosclerotic plaques was detected by immunohistochemical staining and real-time reverse transcription polymerase chain reaction (RT-PCR). Oil Red O staining was used to measure the size of plaques. Cell apoptosis in vitro and in vivo was tested by flow cytometric analysis and terminal deoxynucleotidyl-transferase mediated dUTP nick-end labeling (TUNEL) staining, respectively. Protein expression levels of IL-37, IL-18Rα and p-Smad3 were measured by Weston blotting. RESULTS: Immunohistochemical staining revealed that IL-37 was highly expressed in human atherosclerotic plaques. Intracellular cytokine staining revealed that infiltrated CD4+ T lymphocytes and vascular smooth muscle cells (VSMCs), but not macrophages, were the major sources of IL-37. Mice that overexpressed IL-37 exhibited significant improvements in their atherosclerotic burden, as demonstrated by reduced plaque size, increased collagen levels, and reduced numbers of apoptotic cells in vivo. Subsequently, mechanistic studies showed that IL-37 played an anti-atherosclerotic role, at least partially, through reducing inflammation by promoting the differentiation of the T helper cell anti-inflammatory phenotype, and through increasing plaque stability by decreasing matrix metalloproteinase (MMP)-2/13-mediated degradation of collagen and inhibiting VSMCs apoptosis. CONCLUSION: IL-37 may be a novel potential therapeutic target in patients with atherosclerotic heart disease.


Asunto(s)
Interleucina-1/metabolismo , Placa Aterosclerótica/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apoptosis/efectos de los fármacos , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/prevención & control , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Citocinas/análisis , Humanos , Peróxido de Hidrógeno/toxicidad , Interleucina-1/genética , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/inmunología , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteína smad3/deficiencia , Proteína smad3/genética
7.
J Innate Immun ; 10(1): 56-69, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29176319

RESUMEN

Signaling by the interleukin-36 receptor (IL-36R) is linked to inflammatory diseases such as psoriasis. However, the regulation of IL-36R signaling is poorly understood. Activation of IL-36R signaling in cultured cells results in an increased polyubiquitination of the receptor subunit, IL-1Rrp2. Treatment with deubiquitinases shows that the receptor subunit of IL-36R, IL-1Rrp2, is primarily polyubiquitinated at the K63 position, which is associated with endocytic trafficking and signal transduction. A minor amount of ubiquitination is at the K48 position that is associated with protein degradation. A focused siRNA screen identified RNF125, an E3 ubiquitin ligase, to ubiquitinate IL-1Rrp2 upon activation of IL-36R signaling while not affecting the activated IL-1 receptor. Knockdown of RNF125 decreases signal transduction by the IL-36R. Overexpression of RNF125 in HEK293T cells activates IL-36R signaling and increases the ubiquitination of IL-1Rrp2 and its subsequent turnover. RNF125 can coimmunoprecipitate with the IL-36R, and it traffics with IL-1Rrp2 from the cell surface to lysosomes. Mutations of Lys568 and Lys569 in the C-terminal tail of IL-1Rrp2 decrease ubiquitination by RNF125 and increase the steady-state levels of IL-1Rrp2. These results demonstrate that RNF125 has multiple regulatory roles in the signaling, trafficking, and turnover of the IL-36R.


Asunto(s)
Inflamación/inmunología , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Lisosomas/metabolismo , Psoriasis/inmunología , Receptores de Interleucina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Endocitosis , Células HEK293 , Humanos , Subunidad alfa del Receptor de Interleucina-18/genética , Mutación/genética , Transporte de Proteínas , ARN Interferente Pequeño/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
Int J Mol Sci ; 18(12)2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29261164

RESUMEN

The proinflammatory cytokine interleukin (IL)-18 is an important mediator of the organ failure induced by endotoxemia. IL-18 (known as an interferon-gamma (IFN-γ) inducing factor), and other inflammatory cytokines have important roles in lipopolysaccharide (LPS)-induced acute kidney injury (AKI). We investigated the effect of inflammatory cytokines and Toll-like receptor 4 (TLR4) expression, an event that is accompanied by an influx of monocytes, including CD4⁺ T cells and antigen-presenting cells (APCs) in IL-18Rα knockout (KO) mice and wild-type (WT) mice after LPS injection. In the acute advanced phase, the IL-18Rα KO mice showed a higher survival rate and a suppressed increase of blood urea nitrogen, increased levels of proinflammatory cytokines such as IFN-γ and IL-18, the infiltration of CD4⁺ T cells and the expression of kidney injury molecule-1 as an AKI marker. In that phase, the renal mRNA expression of the M1 macrophage phenotype and C-C chemokine receptor type 7 as the maturation marker of dendritic cells (DCs) was also significantly decreased in the IL-18Rα KO mice, although there were small numbers of F4/80⁺ cells and DCs in the kidney. Conversely, there were no significant differences in the expressions of mRNA and protein TLR4 after LPS injection between the WT and IL-18Rα KO groups. Our results demonstrated that the IL-18Rα-mediated signaling pathway plays critical roles in CD4⁺ T cells and APCs and responded more quickly to IFN-γ and IL-18 than TLR4 stimulation in the pathogenesis of LPS-induced AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Transducción de Señal , Lesión Renal Aguda/etiología , Animales , Células Presentadoras de Antígenos/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Interferón gamma/sangre , Interleucina-18/sangre , Subunidad alfa del Receptor de Interleucina-18/genética , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores CCR7/genética , Receptores CCR7/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
9.
Elife ; 62017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28895840

RESUMEN

MyD88 is the main adaptor molecule for TLR and IL-1R family members. Here, we demonstrated that T-cell intrinsic MyD88 signaling is required for proliferation, protection from apoptosis and expression of activation/memory genes during infection with the intracellular parasite Trypanosoma cruzi, as evidenced by transcriptome and cytometry analyses in mixed bone-marrow (BM) chimeras. The lack of direct IL-18R signaling in T cells, but not of IL-1R, phenocopied the absence of the MyD88 pathway, indicating that IL-18R is a critical MyD88-upstream pathway involved in the establishment of the Th1 response against an in vivo infection, a presently controvert subject. Accordingly, Il18r1-/- mice display lower levels of Th1 cells and are highly susceptible to infection, but can be rescued from mortality by the adoptive transfer of WT CD4+ T cells. Our findings establish the T-cell intrinsic IL-18R/MyD88 pathway as a crucial element for induction of cognate Th1 responses against an important human pathogen.


Asunto(s)
Enfermedad de Chagas/inmunología , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Células TH1/inmunología , Células TH1/parasitología , Trypanosoma cruzi/inmunología , Traslado Adoptivo , Animales , Enfermedad de Chagas/terapia , Modelos Animales de Enfermedad , Citometría de Flujo , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia
10.
Rheumatology (Oxford) ; 55(12): 2220-2229, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27567100

RESUMEN

OBJECTIVES: The IL-1 family member IL-37 was recently characterized as a fundamental inhibitor of innate inflammation. We investigated the effects of recombinant IL-37 in joint inflammation and joint pathology in a mouse model of arthritis. In addition, we explored the potential for therapeutic use in human joint inflammation. METHODS: Wild-type mice were treated systemically with a recombinant form of the naturally occurring human IL-37, and then the knee joints were injected with streptococcal cell wall fragments; joint inflammation, synovial cytokine concentrations and histology were evaluated after 24 h. Mice deficient in the IL-1 family decoy receptor IL-1R8 were treated in a similar manner. The effects of IL-37 treatment were also assessed in a model of streptococcal cell wall-induced systemic inflammation. Changes in IL37 and IL1R8 gene expression were evaluated in the synovia of patients with rheumatoid arthritis. RESULTS: In wild-type mice, low doses (40 µg/kg) of IL-37 suppressed joint inflammation by 51.7% (P < 0.001) and significantly decreased synovial IL-1ß by 84%, IL-6 by 73%, TNF-α by 33%, chemokine (C-X-C motif) ligand 1 by 58%, Chemokine (C-C motif) ligand 3 or macrophage inflammatory protein 1-alpha by 64%, IL-1α by 40% and MPO by 60%. These reductions were associated with a lower recruitment of neutrophils into the joint. The anti-inflammatory properties of IL-37 were dependent on the presence of IL-1R8, also in streptococcal cell wall-induced peritonitis. We found that gene expression of IL1R8, but not IL37, is markedly increased in the synovia of patients with rheumatoid arthritis. CONCLUSION: IL-37 emerges as a key suppressor of joint and systemic inflammation. These findings indicate a rationale for using recombinant IL-37 in the treatment of arthritis.


Asunto(s)
Antirreumáticos/farmacología , Artritis Experimental/tratamiento farmacológico , Interleucina-1/farmacología , Animales , Pared Celular , Citocinas/metabolismo , Humanos , Interleucina-1/metabolismo , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Masculino , Ratones Endogámicos C57BL , Peritonitis/tratamiento farmacológico , Proteínas Recombinantes/farmacología , Streptococcus , Membrana Sinovial/química , Sinovitis/tratamiento farmacológico
11.
J Biol Chem ; 291(32): 16597-609, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27307043

RESUMEN

Signal transduction by the IL-36 receptor (IL-36R) is linked to several human diseases. However, the structure and function of the IL-36R is not well understood. A molecular model of the IL-36R complex was generated and a cell-based reporter assay was established to assess the signal transduction of recombinant subunits of the IL-36R. Mutational analyses and functional assays have identified residues of the receptor subunit IL-1Rrp2 needed for cytokine recognition, stable protein expression, disulfide bond formation and glycosylation that are critical for signal transduction. We also observed that, overexpression of ectodomain (ECD) of Il-1Rrp2 or IL-1RAcP exhibited dominant-negative effect on IL-36R signaling. The presence of IL-36 cytokine significantly increased the interaction of IL-1Rrp2 ECD with the co-receptor IL-1RAcP. Finally, we found that single nucleotide polymorphism A471T in the Toll-interleukin 1 receptor domain (TIR) of the IL-1Rrp2 that is present in ∼2% of the human population, down-regulated IL-36R signaling by a decrease of interaction with IL-1RAcP.


Asunto(s)
Proteína Accesoria del Receptor de Interleucina-1 , Subunidad alfa del Receptor de Interleucina-18 , Polimorfismo Genético , Células HEK293 , Humanos , Proteína Accesoria del Receptor de Interleucina-1/química , Proteína Accesoria del Receptor de Interleucina-1/genética , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Subunidad alfa del Receptor de Interleucina-18/química , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Dominios Proteicos , Transducción de Señal , Relación Estructura-Actividad
12.
Cell ; 163(6): 1444-56, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26638073

RESUMEN

The intestinal mucosal barrier controlling the resident microbiome is dependent on a protective mucus layer generated by goblet cells, impairment of which is a hallmark of the inflammatory bowel disease, ulcerative colitis. Here, we show that IL-18 is critical in driving the pathologic breakdown of barrier integrity in a model of colitis. Deletion of Il18 or its receptor Il18r1 in intestinal epithelial cells (Δ/EC) conferred protection from colitis and mucosal damage in mice. In contrast, deletion of the IL-18 negative regulator Il18bp resulted in severe colitis associated with loss of mature goblet cells. Colitis and goblet cell loss were rescued in Il18bp(-/-);Il18r(Δ/EC) mice, demonstrating that colitis severity is controlled at the level of IL-18 signaling in intestinal epithelial cells. IL-18 inhibited goblet cell maturation by regulating the transcriptional program instructing goblet cell development. These results inform on the mechanism of goblet cell dysfunction that underlies the pathology of ulcerative colitis.


Asunto(s)
Colitis Ulcerosa/patología , Colitis Ulcerosa/fisiopatología , Interleucina-18/inmunología , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/metabolismo , Sulfato de Dextran , Células Endoteliales/metabolismo , Células Epiteliales/citología , Femenino , Células Caliciformes/metabolismo , Células Caliciformes/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Mucosa Intestinal/fisiopatología , Masculino , Ratones , Transducción de Señal
13.
Oncol Rep ; 34(4): 2031-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26252430

RESUMEN

Graft-versus-host disease (GVHD) is a devastating complication of hematopoietic stem cell transplantation (HSCT), and is characterized by systemic inflammation and tissue damage in multiple organs, such as the liver and small intestine. Interleukin-18 (IL-18), an important pro-inflammatory cytokine, is elevated during the course of acute GVHD (aGVHD), and is associated with the severe clinical manifestations of the disease. The biological activity of IL-18 is based on its interaction with the IL-18 receptor (IL-18R) expressed in a variety of cells. The aim of this study was to assess whether blocking the interaction of IL-18 with IL-18R by the anti-IL­18Rα antibody could attenuate the severity of aGVHD. We used a well-established mouse bone marrow transplantation (BMT) model (B6→BALB/c) to block the IL-18/IL-18R interaction by a neutralizing monoclonal antibody (mAb) against murine IL-18Rα. Administration of anti-IL-18Rα mAb had a significant protective effect on the clinical and pathologic manifestations of aGVHD, resulting in a markedly improved survival rate, modified inflammatory response and decreased tissue damage. Interfering with IL-18/IL-18R interaction affected levels of Th1, Th2 and Th17 subsets in the peripheral blood of the aGVHD animals. Additionally, it led to decreased tissue expression of IL-18 and apoptosis-associated molecules (Fas and FasL), and lower phosphorylation levels of p38MAPK in the liver and small intestine. These changes coincided with the decrease in cell apoptosis in aGVHD target organs. Thus, anti­IL-18Rα therapy may, therefore, represent a new therapeutic interference approach for treating aGVHD.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Subunidad alfa del Receptor de Interleucina-18/antagonistas & inhibidores , Interleucina-18/metabolismo , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Intestino Delgado/metabolismo , Hígado/metabolismo , Masculino , Ratones , Análisis de Supervivencia , Linfocitos T Colaboradores-Inductores/metabolismo
14.
Clin Immunol ; 157(2): 103-13, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25638412

RESUMEN

IL-18 is induced in food allergy and EoE is food allergen-induced disease. Therefore, we tested the hypothesis whether IL-18 is involved in food allergen-induced EoE pathogenesis. Accordingly, we examined normal SPT+ and SPT- EoE patient blood and biopsy samples for IL-18, IL-18Rα, ICAM and VCAM expression. Herein, we show increased IL-18 level is highly significant in food allergen SPT+ compared to SPT- EoE patients. We also report that IL-18Rα+ cells and mRNA levels are induced in the esophageal biopsies of EoE patients and blood IL-18 levels correlate with esophageal eosinophilia (P<0.01). Additionally, we report that the levels of esophageal eosinophil and mast cells correlate with ICAM expression in human EoE. Mechanistically, we show that IL-18 in vitro stimulates iNKT cells and endothelial cells and induce eosinophil active cytokines IL-5 and IL-13. We provide the evidence that IL-18 is critical cytokine involved in activation of iNKT cells and ICAM in promoting human EoE.


Asunto(s)
Esofagitis Eosinofílica/inmunología , Esófago/inmunología , Hipersensibilidad a los Alimentos/inmunología , Molécula 1 de Adhesión Intercelular/genética , Subunidad alfa del Receptor de Interleucina-18/inmunología , Interleucina-18/inmunología , Células T Asesinas Naturales/inmunología , ARN Mensajero/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Adolescente , Estudios de Casos y Controles , Línea Celular , Niño , Preescolar , Esofagitis Eosinofílica/etiología , Esofagitis Eosinofílica/genética , Esófago/metabolismo , Esófago/patología , Femenino , Hipersensibilidad a los Alimentos/complicaciones , Hipersensibilidad a los Alimentos/genética , Humanos , Lactante , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-13/metabolismo , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-5/genética , Interleucina-5/inmunología , Interleucina-5/metabolismo , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Pruebas Cutáneas , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
Allergy ; 70(4): 366-73, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25557042

RESUMEN

BACKGROUND: Interleukin (IL) 37 has been described as a negative regulator of innate immunity, as it reduces the activation and cytokine production of different innate immune cells. Recently, results from the CLARA childhood asthma cohort suggested an implication of IL-37 for human asthma pathogenesis. This study aimed to investigate the effects of IL-37 on allergic airway inflammation in a mouse model of experimental asthma. METHODS: Peripheral blood mononuclear cells (PBMCs) of children were cultured for 48 h (anti-CD3/anti-CD28 stimulation or unstimulated), and IL-37 concentrations in supernatants were determined. Wild-type, IL-18Rα-deficient ((-/-) ), and SIGIRR(-/-) C57BL/6 mice were sensitized to ovalbumin (OVA) and challenged with OVA aerosol to induce acute experimental asthma, and IL-37 was applied intranasally prior to each OVA challenge. Airway hyper-responsiveness (AHR), airway inflammation, cytokine levels in broncho-alveolar lavage fluid, and mucus production were determined. RESULTS: IL-37 production of human PBMCs was significantly lower in allergic asthmatics vs healthy children. In wild-type mice, intranasal administration of IL-37 ablated allergic airway inflammation as well as cytokine production and subsequently diminished the hallmarks of experimental asthma including mucus hyperproduction and AHR. In contrast, local application of IL-37 produced none of these effects in mice lacking either IL18Rα or SIGIRR/IL-1R8. CONCLUSIONS: This study demonstrates that IL-37 is able to ablate a TH2 cell-directed allergic inflammatory response and the hallmarks of experimental asthma in mice, suggesting that IL-37 may be critical for asthma pathogenesis. Furthermore, these data suggest a mode of action of IL-37 that involves IL18Rα as well as the orphan receptor SIGIRR/IL-1R8.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-1/metabolismo , Receptores de Interleucina-1/metabolismo , Hipersensibilidad Respiratoria/metabolismo , Adolescente , Animales , Asma/genética , Asma/inmunología , Asma/patología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Niño , Preescolar , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1/sangre , Subunidad alfa del Receptor de Interleucina-18/genética , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Noqueados , Receptores de Interleucina-1/genética , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/patología
16.
J Acquir Immune Defic Syndr ; 67(3): 254-7, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25314247

RESUMEN

: The importance of interleukin (IL)-18 in mediating immune activation during HIV infection has recently emerged. IL-18 activity is regulated by its receptor (IL-18R), formed by an α and a ß chain, the IL-18-binding protein, and the newly identified shorter isoforms of both IL-18R chains. We evaluated gene expression of the IL-18/IL-18R system in peripheral blood mononuclear cells from HIV+ patients. Compared with healthy donors, IL-18 expression decreased in patients with primary infection. The IL-18Rα short transcript expression was strongly upregulated by successful highly active antiretroviral therapy. HIV progression and its treatment can influence the expression of different components of the complex IL-18/IL-18R system.


Asunto(s)
Terapia Antirretroviral Altamente Activa , Infecciones por VIH/tratamiento farmacológico , VIH-1 , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Adulto , Análisis de Varianza , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Estudios de Casos y Controles , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Humanos , Interleucina-18/metabolismo , Leucocitos Mononucleares/metabolismo , ARN Mensajero/análisis , ARN Viral/análisis
17.
FEBS Lett ; 588(21): 3838-43, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25261253

RESUMEN

Interleukin 18 (IL-18), a member of the IL-1 family of cytokines, is an important regulator of innate and acquired immune responses. It signals through its ligand-binding primary receptor IL-18Rα and accessory receptor IL-18Rß. Here we report the crystal structure of IL-18 with the ectodomain of IL-18Rα, which reveals the structural basis for their specific recognition. It confirms that surface charge complementarity determines the ligand-binding specificity of primary receptors in the IL-1 receptor family. We suggest that IL-18 signaling complex adopts an architecture similar to other agonistic cytokines and propose a general ligand-receptor assembly and activation model for the IL-1 family.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-18/química , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-18/química , Interleucina-18/metabolismo , Animales , Cristalografía por Rayos X , Humanos , Interleucina-1/metabolismo , Ligandos , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Células Sf9 , Spodoptera , Especificidad por Sustrato , Propiedades de Superficie
18.
Mediators Inflamm ; 2014: 914954, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24963217

RESUMEN

Proinflammatory cytokine IL-18 has been shown to be elevated in the sera of ovarian carcinoma patients. The aim of the study was to examine the levels and cellular origin of IL-18, IL-18 binding protein, and IL-18 receptor in normal and cancerous ovarian tissues. Ovarian tissue samples were examined by immunohistochemical staining for IL-18, IL-18BP, and IL-18R and mRNA of these cytokines was analyzed with semiquantitative PT-PCR. IL-18 levels were significantly higher in cancerous ovarian tissues (P = 0.0007), IL-18BP levels were significantly higher in normal ovarian tissues (P = 0.04), and the ratio of IL-18/IL-18BP was significantly higher in cancerous ovarian tissues (P = 0.036). Cancerous ovarian tissues expressed significantly higher IL-18 mRNA levels (P = 0.025), while there was no difference in the expression of IL-18BP mRNA and IL-18R mRNA between cancerous and normal ovarian tissues. IL-18 and IL-18BP were expressed dominantly in the epithelial cells of both cancerous and normal ovarian tissues, while IL-18R was expressed dominantly in the epithelial cells of cancerous ovarian tissues but expressed similarly in the epithelial and stromal cells of normal cancerous tissues. This study indicates a possible role of IL-18, IL-18BP, and IL-18R in the pathogenesis of epithelial ovarian carcinoma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-18/metabolismo , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , ARN Mensajero/metabolismo
19.
Immunology ; 142(1): 140-50, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24843874

RESUMEN

Natural killer (NK) cells contribute to the effector phase of vaccine-induced adaptive immune responses, secreting cytokines and releasing cytotoxic granules. The proportion of responding NK cells varies between individuals and by vaccine, suggesting that functionally discrete subsets of NK cells with different activation requirements may be involved. Here, we have used responses to individual components of the DTP vaccine [tetanus toxoid (TT), diphtheria toxoid (DT), whole cell inactivated pertussis] to characterize the NK cell subsets involved in interleukin-2-dependent recall responses. Culture with TT, DT or pertussis induced NK cell CD25 expression and interferon-γ production in previously vaccinated individuals. Responses were the most robust against whole cell pertussis, with responses to TT being particularly low. Functional analysis of discrete NK cell subsets revealed that transition from CD56bright to CD56dim correlated with increased responsiveness to CD16 cross-linking, whereas increasing CD57 expression correlated with a loss of responsiveness to cytokines. A higher frequency of CD56dim CD57− NK cells expressed CD25 and interferon-γ following stimulation with vaccine antigen compared with CD56dim CD57+ NK cells and made the largest overall contribution to this response. CD56dim CD57int NK cells represent an intermediate functional phenotype in response to vaccine-induced and receptor-mediated stimuli. These findings have implications for the ability of NK cells to contribute to the effector response after vaccination and for vaccine-induced immunity in older individuals.


Asunto(s)
Inmunidad Adaptativa , Antígenos Bacterianos/inmunología , Antígenos CD57/metabolismo , Vacuna contra Difteria, Tétanos y Tos Ferina/inmunología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Adulto , Anciano , Biomarcadores/metabolismo , Antígeno CD56/metabolismo , Células Cultivadas , Toxoide Diftérico/inmunología , Proteínas Ligadas a GPI/metabolismo , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Células Asesinas Naturales/clasificación , Células Asesinas Naturales/metabolismo , Subgrupos Linfocitarios/clasificación , Subgrupos Linfocitarios/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Persona de Mediana Edad , Vacuna contra la Tos Ferina/inmunología , Fenotipo , Receptores de IgG/metabolismo , Receptores de Interleucina-12/metabolismo , Toxoide Tetánico/inmunología , Adulto Joven
20.
Semin Immunol ; 25(6): 466-8, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24275599

RESUMEN

IL-37 was formerly termed IL-1 family member 7. The cytokine was discovered by in silico research of human databases. Although there are no genes in the databases with an open reading frame for a murine homologue for IL-37, human IL-37 is functional in the mouse. Like others members of the IL-1 family, IL-37 lacks a signal peptide. The precursor form of IL-37 has a caspase-1 site, but the role of caspase-1 in the processing and secretion of IL-37 has not been documented with certainty. IL-37 is similar to IL-1α and IL-33, in that the cytokine is found in the nucleus where, like IL-1α and IL-33, functions in transcription. Translocation of IL-37 to the nucleus likely involves SMAD3, which is a component of the TGFß anti-inflammatory signaling pathway. Also similar to IL-1α and IL-33, with loss of membrane integrity upon cell death, the IL-37 precursor exits from the cell where it binds to the IL-18 receptor alpha chain. However, this binding results in reduced inflammation. Without a murine form of IL-37, deletion studies were carried out with specific siRNA. In human monocytes deficient in IL-37, LPS and IL-1ß induced cytokines increased 2-3 fold, suggesting that endogenous IL-37 serves as a break on inflammation. Indeed, in mice expressing human IL-37, inflammation is reduced in models of LPS shock, chemical colitis, cardiac ischemia and contact dermatitis.


Asunto(s)
Inflamación/inmunología , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Interleucina-1/inmunología , Secuencia de Aminoácidos , Animales , Caspasa 1/metabolismo , Núcleo Celular/metabolismo , Colitis/inmunología , Humanos , Interleucina-1/genética , Interleucina-18/genética , Macrófagos/inmunología , Ratones , Isoformas de Proteínas/genética , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...