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










Base de datos
Intervalo de año de publicación
1.
Microvasc Res ; 139: 104277, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34752815

RESUMEN

Implantation of biomedical/synthetic devices to replace and/or repair biological tissues very often induces an adverse healing response (scarce angiogenesis, excessive collagen deposition) which is detrimental to implant functionality and integration to host tissue. Interleukin-33/ST2 axis (IL-33/ST2) has been shown to modulate angiogenic and remodeling processes in several types of injuries. However, its effects on these processes after implantation of synthetic matrix have not been reported. Using synthetic matrix of polyether-polyurethane implanted subcutaneously in mice lacking ST2 receptor (ST2/KO), we characterized neovascularization and matrix remodeling in the fibrovascular tissue induced by the implants. Tissue accumulation was increased inside and around the implants in KO implants relative to the wild type (WT). More intense proliferative activity, using CDC 47 marker, was observed in KO implants compared with that of WT implants. Angiogenesis, using two endothelial cell markers, Von Willebrand Factor (VWF) and vascular endothelial cell VE cadherin and hemoglobin content, increased in implants of KO mice relative to control WT. Remodeling of the newly formed fibrovascular tissue (soluble collagen and PicroSirius Red-stained histological sections) showed predominance of type 1 collagen in ST2-KO implants versus type 3 in control implants. The number of positive cells for caspase-3, apoptotic marker, decreased in ST2 group. Our findings evidenced a role of IL-33/ST2 axis in restraining blood vessel formation and regulating the pattern of matrix remodeling in the fibrovascular tissue induced by synthetic implants. Intervention in this cytokine complex holds potential to accelerate integration of biomaterial and host tissue by improving blood supply and matrix remodeling.


Asunto(s)
Matriz Extracelular/metabolismo , Reacción a Cuerpo Extraño/metabolismo , Mediadores de Inflamación/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Interleucina-33/metabolismo , Neovascularización Fisiológica , Tejido Subcutáneo/metabolismo , Cicatrización de Heridas , Animales , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Fibrosis , Reacción a Cuerpo Extraño/etiología , Reacción a Cuerpo Extraño/genética , Reacción a Cuerpo Extraño/patología , Eliminación de Gen , Proteína 1 Similar al Receptor de Interleucina-1/genética , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Polietilenglicoles , Poliuretanos , Transducción de Señal , Tejido Subcutáneo/patología , Tapones Quirúrgicos de Gaza , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Front Immunol ; 12: 657803, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815420

RESUMEN

The role of IL-33/ST2 signaling in cardiac allograft vasculopathy (CAV) is not fully addressed. Here, we investigated the role of IL-33/ST2 signaling in allograft or recipient in CAV respectively using MHC-mismatch murine chronic cardiac allograft rejection model. We found that recipients ST2 deficiency significantly exacerbated allograft vascular occlusion and fibrosis, accompanied by increased F4/80+ macrophages and CD3+ T cells infiltration in allografts. In contrast, allografts ST2 deficiency resulted in decreased infiltration of F4/80+ macrophages, CD3+ T cells and CD20+ B cells and thus alleviated vascular occlusion and fibrosis of allografts. These findings indicated that allografts or recipients ST2 deficiency oppositely affected cardiac allograft vasculopathy/fibrosis via differentially altering immune cells infiltration, which suggest that interrupting IL-33/ST2 signaling locally or systematically after heart transplantation leads different outcome.


Asunto(s)
Enfermedad Coronaria/etiología , Enfermedad Coronaria/patología , Trasplante de Corazón , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Leucocitos/patología , Aloinjertos , Animales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Enfermedad Coronaria/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Rechazo de Injerto , Supervivencia de Injerto , Trasplante de Corazón/efectos adversos , Trasplante de Corazón/métodos , Inmunohistoquímica , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Complicaciones Posoperatorias , Subgrupos de Linfocitos T/metabolismo
3.
Sci Rep ; 10(1): 6451, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32296080

RESUMEN

Current studies addressing the influence of interleukin-33 or its receptor (IL-33R/ST2) on development of atopic dermatitis-like inflammation in mice have reported conflicting results. We compared the response in single- and double-deficient IL-33-/-/ST2-/- C57BL/6J BomTac mice in the well-established calcipotriol-induced model of atopic dermatitis. All genotypes (groups of up to 14 mice) developed atopic dermatitis-like inflammation yet we observed no biologically relevant difference between groups in gross anatomy or ear thickness. Moreover, histological examination of skin revealed no differences in mononuclear leukocyte and granulocyte infiltration nor Th2 cytokine levels (IL-4 and IL-13). Finally, skin CD45+ cells and CD3+ cells were found at similar densities across all groups. Our findings indicate that lack of interleukin-33 and its receptor ST2 does not prevent the development of AD-like skin inflammation.


Asunto(s)
Dermatitis Atópica/inmunología , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Interleucina-33/deficiencia , Transducción de Señal/inmunología , Animales , Calcitriol/análogos & derivados , Calcitriol/inmunología , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/genética , Interleucina-33/inmunología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/genética , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología
4.
Respir Res ; 21(1): 98, 2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32326950

RESUMEN

BACKGROUND: Obesity augments pulmonary responses to ozone. We have reported that IL-33 contributes to these effects of obesity in db/db mice. The purpose of this study was to determine whether IL-33 also contributes to obesity-related changes in the response to ozone in mice with diet-induced obesity. METHODS: Male wildtype C57BL/6 mice and mice deficient in ST2, the IL-33 receptor, were placed on chow or high fat diets for 12 weeks from weaning. Because the microbiome has been implicated in obesity-related changes in the pulmonary response to ozone, mice were either housed with other mice of the same genotype (same housed) or with mice of the opposite genotype (cohoused). Cohousing transfers the gut microbiome from one mouse to its cagemates. RESULTS: Diet-induced increases in body mass were not affected by ST2 deficiency or cohousing. In same housed mice, ST2 deficiency reduced ozone-induced airway hyperresponsiveness and neutrophil recruitment in chow-fed but not HFD-fed mice even though ST2 deficiency reduced bronchoalveolar lavage IL-5 in both diet groups. In chow-fed mice, cohousing abolished ST2-related reductions in ozone-induced airway hyperresponsiveness and neutrophil recruitment, but in HFD-fed mice, no effect of cohousing on these responses to ozone was observed. In chow-fed mice, ST2 deficiency and cohousing caused changes in the gut microbiome. High fat diet-feeding caused marked changes in the gut microbiome and overrode both ST2-related and cohousing-related differences in the gut microbiome observed in chow-fed mice. CONCLUSION: Our data indicate a role for IL-33 in pulmonary responses to ozone in chow-fed but not high fat diet-fed mice and are consistent with the hypothesis that these diet-related differences in the role of IL-33 are the result of changes in the gut microbiome.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Interleucina-33/metabolismo , Pulmón/metabolismo , Obesidad/metabolismo , Ozono/toxicidad , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología
5.
Cell Rep ; 30(9): 2989-3003.e6, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130902

RESUMEN

We find that cardiac group 2 innate lymphoid cells (ILC2s) are essential for the development of IL-33-induced eosinophilic pericarditis. We show a pathogenic role for ILC2s in cardiac inflammation, in which ILC2s activated by IL-33 drive the development of eosinophilic pericarditis in collaboration with cardiac fibroblasts. ILCs, not T and B cells, are required for the development of pericarditis. ILC2s transferred to the heart of Rag2-/-Il2rg-/- mice restore their susceptibility to eosinophil infiltration. Moreover, ILC2s direct cardiac fibroblasts to produce eotaxin-1. We also find that eosinophils reside in the mediastinal cavity and that eosinophils transferred to the mediastinal cavity of eosinophil-deficient ΔdblGATA1 mice following IL-33 treatment migrate to the heart. Thus, the serous cavities may serve as a reservoir of cardiac-infiltrating eosinophils. In humans, patients with pericarditis show higher amounts of ILCs in pericardial fluid than do healthy controls and patients with other cardiac diseases. We demonstrate that ILCs play a critical role in pericarditis.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Pericarditis/inmunología , Animales , Movimiento Celular/efectos de los fármacos , Quimiocina CCL11/genética , Quimiocina CCL11/metabolismo , Susceptibilidad a Enfermedades , Eosinófilos/efectos de los fármacos , Eosinófilos/patología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/fisiopatología , Pruebas de Función Cardíaca/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/farmacología , Interleucina-5/metabolismo , Linfocitos/efectos de los fármacos , Masculino , Mediastino/patología , Ratones Endogámicos BALB C , Pericarditis/genética , Pericarditis/fisiopatología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
6.
Respir Res ; 20(1): 197, 2019 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455422

RESUMEN

BACKGROUND: Interleukin-33 is released in the airways following acute ozone exposure and has the ability to cause airway hyperresponsiveness, a defining feature of asthma. Ozone causes greater airway hyperresponsiveness in male than female mice. Moreover, sex differences in the gut microbiome account for sex differences in this response to ozone. The purpose of this study was to determine whether there were sex differences in the role of interleukin-33 in ozone-induced airway hyperresponsiveness and to examine the role of the microbiome in these events. METHODS: Wildtype mice and mice genetically deficient in ST2, the interleukin-33 receptor, were housed from weaning with either other mice of the same genotype and sex, or with mice of the same sex but opposite genotype. At 15 weeks of age, fecal pellets were harvested for 16S rRNA sequencing and the mice were then exposed to air or ozone. Airway responsiveness was measured and a bronchoalveolar lavage was performed 24 h after exposure. RESULTS: In same-housed mice, ozone-induced airway hyperresponsiveness was greater in male than female wildtype mice. ST2 deficiency reduced ozone-induced airway hyperresponsiveness in male but not female mice and abolished sex differences in the response to ozone. However, sex differences in the role of interleukin-33 were unrelated to type 2 cytokine release: ozone-induced increases in bronchoalveolar lavage interleukin-5 were greater in females than males and ST2 deficiency virtually abolished interleukin-5 in both sexes. Since gut microbiota contribute to sex differences in ozone-induced airway hyperresponsiveness, we examined the role of the microbiome in these ST2-dependent sex differences. To do so, we cohoused wildtype and ST2 deficient mice, a situation that allows for transfer of microbiota among cage-mates. Cohousing altered the gut microbial community structure, as indicated by 16S rRNA gene sequencing of fecal DNA and reversed the effect of ST2 deficiency on pulmonary responses to ozone in male mice. CONCLUSIONS: The data indicate that the interleukin-33 /ST2 pathway contributes to ozone-induced airway hyperresponsiveness in male mice and suggest that the role of interleukin-33 is mediated at the level of the gut microbiome.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Interleucina-33/metabolismo , Microbiota/efectos de los fármacos , Ozono/toxicidad , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/metabolismo , Animales , Femenino , Exposición por Inhalación/efectos adversos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/fisiología , Ozono/administración & dosificación
7.
J Immunol ; 202(10): 3053-3064, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30979817

RESUMEN

Acute graft-versus-host disease (aGVHD) hinders the efficacy of allogeneic hematopoietic cell transplantation (HCT). Plasma levels of soluble membrane-bound ST2 (ST2) are elevated in human and murine aGVHD and correlated to type 1 T cells response. ST2 signals through the adapter protein MyD88. The role of MyD88 in T cells during aGVHD has yet to be elucidated. We found that knocking out MyD88 in the donor T cells protected against aGVHD independent of IL-1R and TLR4 signaling in two murine HCT models. This protection was entirely driven by MyD88-/- CD4 T cells. Transplanting donor MyD88-/- conventional T cells (Tcons) with wild-type (WT) or MyD88-/- regulatory T cells (Tregs) lowered aGVHD severity and mortality. Transcriptome analysis of sorted MyD88-/- CD4 T cells from the intestine 10 d post-HCT showed lower levels of Il1rl1 (gene of ST2), Ifng, Csf2, Stat5, Batf, and Jak2 Transplanting donor ST2-/- Tcons with WT or ST2-/- Tregs showed a similar phenotype with what we observed when using donor MyD88-/- Tcons. Decreased ST2 was confirmed at the protein level with less secretion of soluble ST2 and more expression of ST2 compared with WT T cells. Our data suggest that Treg suppression from lack of MyD88 signaling in donor Tcons during alloreactivity uses the ST2 but not the IL-1R or TLR4 pathways, and ST2 represents a potential aGVHD therapeutic target sparing Tregs.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Factor 88 de Diferenciación Mieloide/deficiencia , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Enfermedad Aguda , Animales , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/genética , Linfocitos T Reguladores/patología , Trasplante Homólogo
8.
Arterioscler Thromb Vasc Biol ; 39(3): 446-458, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30651000

RESUMEN

Objective- Inflammation occurs during the progression of abdominal aortic aneurysm (AAA). IL (interleukin)-33 is a pleiotropic cytokine with multiple immunomodulatory effects, yet its role in AAA remains unknown. Approach and Results- Immunoblot, immunohistochemistry, and immunofluorescent staining revealed increased IL-33 expression in adventitia fibroblasts from mouse AAA lesions. Daily intraperitoneal administration of recombinant IL-33 or transgenic IL-33 expression ameliorated periaorta CaPO4 injury- and aortic elastase exposure-induced AAA in mice, as demonstrated by blunted aortic expansion, reduced aortic wall elastica fragmentation, enhanced AAA lesion collagen deposition, attenuated T-cell and macrophage infiltration, reduced inflammatory cytokine production, skewed M2 macrophage polarization, and reduced lesion MMP (matrix metalloproteinase) expression and cell apoptosis. Flow cytometry analysis, immunostaining, and immunoblot analysis showed that exogenous IL-33 increased CD4+Foxp3+ regulatory T cells in spleens, blood, and aortas in periaorta CaPO4-treated mice. Yet, ST2 deficiency muted these IL-33 activities. Regulatory T cells from IL-33-treated mice also showed significantly stronger activities in suppressing smooth muscle cell inflammatory cytokine and chemokine expression, macrophage MMP expression, and in increasing M2 macrophage polarization than those from vehicle-treated mice. In contrast, IL-33 failed to prevent AAA and lost its beneficial activities in CaPO4-treated mice after selective depletion of regulatory T cells. Conclusions- Together, this study established a role of IL-33 in protecting mice from AAA formation by enhancing ST2-dependent aortic and systemic regulatory T-cell expansion and their immunosuppressive activities.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Interleucina-33/fisiología , Linfocitos T Reguladores/efectos de los fármacos , Animales , Aorta/inmunología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/inmunología , Fosfatos de Calcio/toxicidad , Células Cultivadas , Citocinas/biosíntesis , Evaluación Preclínica de Medicamentos , Inyecciones Intraperitoneales , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/fisiología , Interleucina-33/genética , Interleucina-33/farmacología , Interleucina-33/uso terapéutico , Macrófagos/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Elastasa Pancreática/toxicidad , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Linfocitos T Reguladores/inmunología , Remodelación Vascular
9.
Immunology ; 155(4): 477-490, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30098206

RESUMEN

The excessive inflammation often present in patients with severe dengue infection is considered both a hallmark of disease and a target for potential treatments. Interleukin-33 (IL-33) is a pleiotropic cytokine with pro-inflammatory effects whose role in dengue has not been fully elucidated. We demonstrate that IL-33 plays a disease-exacerbating role during experimental dengue infection in immunocompetent mice. Mice infected with dengue virus serotype 2 (DENV2) produced high levels of IL-33. DENV2-infected mice treated with recombinant IL-33 developed markedly more severe disease compared with untreated mice as assessed by mortality, granulocytosis, liver damage and pro-inflammatory cytokine production. Conversely, ST2-/- mice (deficient in IL-33 receptor) infected with DENV2 developed significantly less severe disease compared with wild-type mice. Furthermore, the increased disease severity and the accompanying pathology induced by IL-33 during dengue infection were reversed by the simultaneous treatment with a CXCR2 receptor antagonist (DF2156A). Together, these results indicate that IL-33 plays a disease-exacerbating role in experimental dengue infection, probably driven by CXCR2-expressing cells, leading to elevated pro-inflammatory response-mediated pathology. Our results also indicate that IL-33 is a potential therapeutic target for dengue infection.


Asunto(s)
Virus del Dengue/inmunología , Interleucina-33/farmacología , Receptores de Interleucina-8B/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Animales , Dengue/inmunología , Dengue/virología , Progresión de la Enfermedad , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sulfonamidas/farmacología
10.
Inflammation ; 41(6): 2041-2051, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30069663

RESUMEN

IL-33/ST2 axis has been shown to exert both pro- and anti- effects in wound healing and tumor development. To further understand the role of this cytokine complex, we characterized comparatively the inflammatory component of a malignant tissue and non-malignant tissue in mice lacking ST2 receptor (ST2-KO). KO mice and their wild-type (WT) counterparts were either implanted subcutaneously with polyether-polyurethane sponge discs to induce non-malignant fibrovascular tissue growth or inoculated with 4T1 cells to induce mammary tumor. Loss of ST2 receptor in mice resulted in enhanced mammary tumor and fibrovascular tissue relative to the WT animals. The inflammatory parameters (MPO and NAG activities, levels of the cytokines CXCL1/KC, CCL2, TNF-α, TGF-ß1, and mast cell number) were differentially modulated in both tissues. In tumors, these parameters were, overall, lower compared with those in tumors of WT mice. In KO implants, CXCL1/KC and TNF-α levels increased; MPO, NAG, and CCL2 levels decreased relative to the WT implants. In addition, deletion of ST2 receptor inhibited mast cell recruitment but had no effect on TGF-ß1 levels in implants. Our study has shown antitumorigenic effect of ST2 in mammary tumor and this may be mediated by downregulation of pro-inflammatory cytokines (CXCL1/KC, CCL2, TNF-α, and TGF-ß1). Conversely, in the fibrovascular tissue, lack of ST2 receptor resulted in differential modulation of cytokine production. Differential signaling mechanisms may be activated by IL-33/ST2 axis to modulate cytokine production in malignant and non-malignant proliferative processes.


Asunto(s)
Citocinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Interleucina-33 , Ratones , Ratones Noqueados
11.
J Pathol ; 245(4): 399-409, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29722022

RESUMEN

Recent evidence supports that bone marrow (BM)-derived hematopoietic progenitor cells play an important role in lung injury and fibrosis. While these cells give rise to multiple cell types, the ST2 (Il1rl1)-expressing group 2 innate lymphoid cells (ILC2s) derived from BM progenitors have been implicated in tissue repair and remodeling, including in lung fibrosis. To further investigate the precise role of BM-derived ILC2s in the pathogenesis of fibrotic lung disease, their importance in the bleomycin-induced lung fibrosis model was evaluated by analyzing the effects of selective ST2 deficiency in the BM compartment. The results showed that while ST2-sufficient control mice exhibited activation of lung IL-33/ST2 signaling, ILC2 recruitment, IL-13 induction, and fibrosis, these responses were significantly diminished in ST2-deficient-BM chimera mice, with selective loss of ST2 expression only in the BM. This diminished response to bleomycin was similar to that seen in ST2 global knockout mice, suggesting the predominant importance of ST2 from the BM compartment. In wild-type mice, ILC2 recruitment to the lung was accompanied by a concomitant decrease in ST2+ BM cells. ST2-deficient BM cells were unresponsive to IL-33-induced ILC2 maturation. Finally, lineage-negative wild-type, but not ST2-deficient BM cells from bleomycin-treated mice stimulated lung fibroblast type I collagen expression, which was associated with elevated TGFß expression in the BM cells. Taken together, these findings suggested that the BM-derived ILC2s were recruited to fibrotic lung through the IL-33/ST2 pathway, and contributed to fibroblast activation to promote lung fibrosis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Inmunidad Innata , Inmunidad Mucosa , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Pulmón/metabolismo , Células Progenitoras Linfoides/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/metabolismo , Pulmón/inmunología , Pulmón/patología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Comunicación Paracrina , Fenotipo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
12.
J Am Soc Nephrol ; 29(4): 1272-1288, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29436517

RESUMEN

Inflammation is a prominent feature of ischemia-reperfusion injury (IRI), which is characterized by leukocyte infiltration and renal tubular injury. However, signals that initiate these events remain poorly understood. We examined the role of the nuclear alarmin IL-33 in tissue injury and innate immune response triggered by experimental kidney ischemia-reperfusion. In wild-type mice, we found that IL-33 was constitutively expressed throughout the kidney in peritubular and periglomerular spaces, mainly by microvascular endothelial cells, from which it was released immediately during IRI. Compared with wild-type mice, mice lacking IL-33 (IL-33Gt/Gt) exhibited reductions in early tubular cell injury and subsequent renal infiltration of IFN-γ/IL-17A-producing neutrophils, with preservation of renal functions. This protection associated with decreased renal recruitment of myeloid dendritic cells, natural killer (NK) cells, and invariant natural killer T (iNKT) cells, the latter of which were reported as deleterious in IRI. Increases in the level of circulating IL-12, a key IL-33 cofactor, and the expression of ST2, an IL-33-specific receptor, on the surface of iNKT cells preceded the IL-33- and iNKT cell-dependent phase of neutrophil infiltration. Furthermore, IL-33 directly targeted iNKT cells in vitro, inducing IFN-γ and IL-17A production. We propose that endogenous IL-33 is released as an alarmin and contributes to kidney IRI by promoting iNKT cell recruitment and cytokine production, resulting in neutrophil infiltration and activation at the injury site. Our findings show a novel molecular mediator contributing to innate immune cell recruitment induced by renal ischemia-reperfusion and may provide therapeutic insights into AKI associated with renal transplantation.


Asunto(s)
Alarminas/fisiología , Interleucina-33/fisiología , Riñón/irrigación sanguínea , Daño por Reperfusión/metabolismo , Alarminas/deficiencia , Alarminas/genética , Animales , Citocinas/biosíntesis , Citocinas/genética , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Interferón gamma/biosíntesis , Interferón gamma/genética , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/fisiología , Interleucina-12/sangre , Interleucina-17/biosíntesis , Interleucina-17/genética , Interleucina-33/biosíntesis , Interleucina-33/deficiencia , Interleucina-33/genética , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Neutrófila , Neutrófilos/inmunología , Daño por Reperfusión/inmunología
13.
Cell Mol Immunol ; 15(4): 388-398, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28194023

RESUMEN

Liver fibrosis is a consequence of chronic liver disease, causing morbidity and mortality. Interleukin-33 (IL-33) is a critical mediator of inflammation, which may be involved in the development of liver fibrosis. Here, we investigated the role of IL-33 in human patients and experimental bile-duct ligation (BDL)-induced fibrosis in mice. We report increased hepatic IL-33 expression in the murine BDL model of fibrosis and in surgical samples obtained from patients with liver fibrosis. Liver injury, inflammatory cell infiltration and fibrosis were reduced in the absence of the IL-33/ST2 receptor, and the activation of hepatic stellate cells (HSCs) was decreased in ST2-deficient mice. Recombinant IL-33 activated HSCs isolated from C57BL/6 mice, leading to the expression of IL-6, TGF-ß, α-SMA and collagen, which was abrogated in the absence of ST2 or by pharmacological inhibition of MAPK signaling. Finally, administration of recombinant IL-33 significantly increased hepatic inflammation in sham-operated BL6 mice but did not enhance BDL-induced hepatic inflammation and fibrosis. In conclusion, BDL-induced liver inflammation and fibrosis are dependent on ST2 signaling in HSCs, and therefore, the IL-33/ST2 pathway may be a potential therapeutic target in human patients with chronic hepatitis and liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Interleucina-33/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Adulto , Animales , Conductos Biliares/patología , Femenino , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Inflamación/patología , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Ligadura , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas Recombinantes/farmacología
14.
J Exp Med ; 215(1): 263-281, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29222107

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are effector cells within the mucosa and key participants in type 2 immune responses in the context of allergic inflammation and infection. ILC2s develop in the bone marrow from common lymphoid progenitor cells, but little is known about how ILC2s egress from the bone marrow for hematogenous trafficking. In this study, we identified a critical role for IL-33, a hallmark peripheral ILC2-activating cytokine, in promoting the egress of ILC2 lineage cells from the bone marrow. Mice lacking IL-33 signaling had normal development of ILC2s but retained significantly more ILC2 progenitors in the bone marrow via augmented expression of CXCR4. Intravenous injection of IL-33 or pulmonary fungal allergen challenge mobilized ILC2 progenitors to exit the bone marrow. Finally, IL-33 enhanced ILC2 trafficking to the lungs in a parabiosis mouse model of tissue disruption and repopulation. Collectively, these data demonstrate that IL-33 plays a critical role in promoting ILC2 egress from the bone marrow.


Asunto(s)
Células de la Médula Ósea/metabolismo , Inmunidad Innata , Interleucina-33/metabolismo , Subgrupos Linfocitarios/metabolismo , Alérgenos/inmunología , Animales , Antígenos Fúngicos/inmunología , Médula Ósea , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Recuento de Células , Diferenciación Celular , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Interleucina-33/genética , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Noqueados , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción de Señal
15.
J Neurotrauma ; 34(22): 3173-3182, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28490277

RESUMEN

Traumatic brain injury (TBI) is a devastating condition, often leading to life-long consequences for patients. Even though modern neurointensive care has improved functional and cognitive outcomes, efficient pharmacological therapies are still lacking. Targeting peripherally derived, or resident inflammatory, cells that are rapid responders to brain injury is promising, but complex, given that the contribution of inflammation to exacerbation versus improved recovery varies with time post-injury. The injury-induced inflammatory response is triggered by release of alarmins, and in the present study we asked whether interleukin-33 (IL-33), an injury-associated nuclear alarmin, is involved in TBI. Here, we used samples from human TBI microdialysate, tissue sections from human TBI, and mouse models of central nervous system injury and found that expression of IL-33 in the brain was elevated from nondetectable levels, reaching a maximum after 72 h in both human samples and mouse models. Astrocytes and oligodendrocytes were the main producers of IL-33. Post-TBI, brains of mice deficient in the IL-33 receptor, ST2, contained fewer microglia/macrophages in the injured region than wild-type mice and had an altered cytokine/chemokine profile in response to injury. These observations indicate that IL-33 plays a role in neuroinflammation with microglia/macrophages being cellular targets for this interleukin post-TBI.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/metabolismo , Interleucina-33/metabolismo , Macrófagos/inmunología , Microglía/inmunología , Oligodendroglía/metabolismo , Adolescente , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
16.
Nat Commun ; 8: 14919, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28374774

RESUMEN

Patients who survive sepsis can develop long-term immune dysfunction, with expansion of the regulatory T (Treg) cell population. However, how Treg cells proliferate in these patients is not clear. Here we show that IL-33 has a major function in the induction of this immunosuppression. Mice deficient in ST2 (IL-33R) develop attenuated immunosuppression in cases that survive sepsis, whereas treatment of naive wild-type mice with IL-33 induces immunosuppression. IL-33, released during tissue injury in sepsis, activates type 2 innate lymphoid cells, which promote polarization of M2 macrophages, thereby enhancing expansion of the Treg cell population via IL-10. Moreover, sepsis-surviving patients have more Treg cells, IL-33 and IL-10 in their peripheral blood. Our study suggests that targeting IL-33 may be an effective treatment for sepsis-induced immunosuppression.


Asunto(s)
Tolerancia Inmunológica/inmunología , Interleucina-33/inmunología , Sepsis/inmunología , Linfocitos T Reguladores/inmunología , Anciano , Animales , Femenino , Humanos , Tolerancia Inmunológica/genética , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Interleucina-33/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Sepsis/genética , Sepsis/metabolismo , Linfocitos T Reguladores/metabolismo
17.
PLoS One ; 11(8): e0161507, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27548066

RESUMEN

Immunomodulatory Foxp3+ regulatory T cells (Tregs) form a heterogeneous population consisting of subsets with different activation states, migratory properties and suppressive functions. Recently, expression of the IL-33 receptor ST2 was shown on Tregs in inflammatory settings. Here we report that ST2 expression identifies highly activated Tregs in mice even under homeostatic conditions. ST2+ Tregs preferentially accumulate at non-lymphoid sites, likely mediated by their high expression of several chemokine receptors facilitating tissue homing. ST2+ Tregs exhibit a Th2-biased character, expressing GATA-3 and producing the Th2 cytokines IL-5 and IL-13 -especially in response to IL-33. Yet, IL-33 is dispensable for the generation and maintenance of these cells in vivo. Furthermore, ST2+ Tregs are superior to ST2- Tregs in suppressing CD4+ T cell proliferation in vitro independent of IL-33. This higher suppressive capacity is partially mediated by enhanced production and activation of the anti-inflammatory cytokines IL-10 and TGFß. Thus, ST2 expression identifies a highly activated, strongly suppressive Treg subset preferentially located in non-lymphoid tissues. Here ST2+ Tregs may be well positioned to immediately react to IL-33 alarm signals. Their specific properties may render ST2+ Tregs useful targets for immunomodulatory therapies.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-10/inmunología , Interleucina-33/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Linaje de la Célula/inmunología , Proliferación Celular , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/inmunología , Regulación de la Expresión Génica , Inmunofenotipificación , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-10/genética , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-33/genética , Interleucina-5/genética , Interleucina-5/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Linfocitos T Reguladores/citología , Factor de Crecimiento Transformador beta/genética
18.
J Neuroinflammation ; 13(1): 159, 2016 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-27334012

RESUMEN

BACKGROUND: Viral encephalitis is a common cause of lethal infections in humans, and several different viruses are documented to be responsible. Rocio virus is a flavivirus that causes a severe lethal encephalitis syndrome in humans and also mice, providing an interesting model to study the CNS compartmentalized immune response. Interleukin 33 (IL-33), a member of the IL-1 family, is an immunomodulatory cytokine that is highly expressed in the CNS. However, the role of IL-33 on viral encephalitis remains unclear. Therefore, we aimed to explore how the IL-33/ST2 axis regulates the local immune response during Rocio virus infection. METHODS: Wild-type (WT), ST2 (ST2(-/-)), and nitric oxide synthase-deficient mice (iNOS(-/-)) and Stat6 (Stat6(-/-))-deficient mice were infected with different concentrations of the Rocio virus by intraperitoneal route, the cytokine mRNA level in CNS was analyzed by qPCR, and cellular immunophenotyping was performed on infected mice by the flow cytometry of isolated CNS mononuclear cells. RESULTS: We have shown that the mRNA expression of IL-33 and ST2 receptors is increased in the CNS of Rocio virus-infected WT mice and that ST2(-/-) mice showed increased susceptibility to infection. ST2 deficiency was correlated with increased tissue pathology, cellular infiltration, and tumor necrosis factor alpha (TNF-α) and interferon-gamma (IFN-γ) mRNA levels and higher viral load in the CNS, compared with wild-type mice. The increased Th1 cytokine levels released in the CNS acted on infiltrating macrophages, as evidenced by flow cytometry characterization of cellular infiltrates, inducing the expression of iNOS, contributing to brain injury. Moreover, iNOS(-/-) mice were more resistant to Rocio virus encephalitis, presenting a lower clinical score and reduced mortality rate, despite the increased tissue pathology. CONCLUSIONS: We provide evidences of a specific role for IL-33 receptor signaling in nitric oxide induction through local IFN-γ modulation, suggesting that nitric oxide overproduction might have an important role in the progression of experimental viral encephalitis.


Asunto(s)
Sistema Nervioso Central , Encefalitis Viral/patología , Interleucina-33/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Femenino , Infecciones por Flaviviridae/patología , Citometría de Flujo , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/genética , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/genética , Transducción de Señal/fisiología
19.
J Allergy Clin Immunol ; 138(1): 283-286, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26830114
20.
J Endod ; 41(3): 369-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25595464

RESUMEN

INTRODUCTION: ST2 is a member of the interleukin (IL)-1 receptor family, and IL-33 is its natural ligand. ST2 signaling promotes Th2 immune response in allergy, autoimmunity, and chronic inflammatory disorders, but its role in the pathogenesis of periapical lesions is unknown. The purpose of this study was to investigate whether ST2 gene deletion affects the development of experimentally induced periapical lesions in mice. METHODS: Pulps of mandibular molars from wild-type (WT) and ST2 knockout (ST2(-)/(-)) BALB/c mice were exposed and left open to the oral environment. After death, hemi-mandibles were isolated and prepared for histologic, immunohistochemical, and flow cytometric analysis. RESULTS: The expression of IL-33 and its receptor ST2 was higher in periapical lesions in WT mice compared with normal root apices (both P < .05). The increased periapical bone loss observed in ST2(-)/(-) mice was associated with enhanced influx of neutrophils, CD3+ CXCR3+ Th1 cells, and CD3+ CCR6+ Th17 cells and increased number of tartrate-resistant acid phosphatase+ osteoclasts (all P < .05). Furthermore, periapical lesions in ST2(-)/(-) mice contained increased percentages of T cells expressing interferon-γ, IL-17, tumor necrosis factor-α, and IL-6 (all P < .05). In comparison with WT mice, CD3+ receptor activator of nuclear factor kappa B ligand+ T cells were increased, whereas CD3+ osteoprotegerin+ T cells were decreased in the lesions of ST2(-)/(-) mice (both P < .05). CONCLUSIONS: ST2 deletion increases inflammatory bone loss in experimental periapical lesions in mice, which is associated with enhanced Th1/Th17 cell mediated periapical immune responses and increased osteoclastogenesis.


Asunto(s)
Huesos/patología , Eliminación de Gen , Inflamación/patología , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Tejido Periapical/patología , Animales , Recuento de Células , Pulpa Dental/patología , Citometría de Flujo , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Osteoclastos/metabolismo , Osteoclastos/patología , Fosfatasa Ácida Tartratorresistente/metabolismo , Células TH1/inmunología , Células Th17/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...