Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Annu Rev Immunol ; 34: 173-202, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-26772211

RESUMEN

The formation and accumulation of crystalline material in tissues is a hallmark of many metabolic and inflammatory conditions. The discovery that the phase transition of physiologically soluble substances to their crystalline forms can be detected by the immune system and activate innate immune pathways has revolutionized our understanding of how crystals cause inflammation. It is now appreciated that crystals are part of the pathogenesis of numerous diseases, including gout, silicosis, asbestosis, and atherosclerosis. In this review we discuss current knowledge of the complex mechanisms of crystal formation in diseased tissues and their interplay with the nutrients, metabolites, and immune cells that account for crystal-induced inflammation.


Asunto(s)
Asbestosis/inmunología , Aterosclerosis/inmunología , Cristalización , Gota/inmunología , Inmunidad Innata , Inflamación/metabolismo , Silicosis/inmunología , Animales , Humanos , Interleucina-1/metabolismo , Nanotecnología , Transición de Fase
2.
Nat Immunol ; 15(8): 727-37, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24952505

RESUMEN

Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1ß (IL-1ß) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1ß. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1ß in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.


Asunto(s)
Apoptosis/inmunología , Caspasa 1/inmunología , Proteínas del Citoesqueleto/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Anticuerpos/inmunología , Proteínas Reguladoras de la Apoptosis , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Caspasa 1/genética , Inhibidores de Caspasas/farmacología , Comunicación Celular/inmunología , Proteínas del Citoesqueleto/genética , Humanos , Inflamasomas/inmunología , Lisosomas/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Fagocitosis/inmunología , Priones/química , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Transducción de Señal/inmunología
3.
Nature ; 552(7685): 355-361, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29293211

RESUMEN

The spreading of pathology within and between brain areas is a hallmark of neurodegenerative disorders. In patients with Alzheimer's disease, deposition of amyloid-ß is accompanied by activation of the innate immune system and involves inflammasome-dependent formation of ASC specks in microglia. ASC specks released by microglia bind rapidly to amyloid-ß and increase the formation of amyloid-ß oligomers and aggregates, acting as an inflammation-driven cross-seed for amyloid-ß pathology. Here we show that intrahippocampal injection of ASC specks resulted in spreading of amyloid-ß pathology in transgenic double-mutant APPSwePSEN1dE9 mice. By contrast, homogenates from brains of APPSwePSEN1dE9 mice failed to induce seeding and spreading of amyloid-ß pathology in ASC-deficient APPSwePSEN1dE9 mice. Moreover, co-application of an anti-ASC antibody blocked the increase in amyloid-ß pathology in APPSwePSEN1dE9 mice. These findings support the concept that inflammasome activation is connected to seeding and spreading of amyloid-ß pathology in patients with Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Microglía/metabolismo , Agregación Patológica de Proteínas , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Anticuerpos/farmacología , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/inmunología , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Presenilina-1/deficiencia , Presenilina-1/genética , Dominios Proteicos , Memoria Espacial/fisiología
4.
Immunol Rev ; 281(1): 74-87, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29247990

RESUMEN

Inflammasomes are the central signaling hubs of the inflammatory response. They process cytosolic evidence of infection, cell damage, or metabolic disturbances, and elicit a pro-inflammatory response mediated by members of the interleukin-1 family of cytokines and pyroptotoic cell death. On the molecular level, this is accomplished by the sensor-nucleated recruitment and oligomerization of the adapter protein ASC. Once a tunable threshold is reached, cooperative assembly of ASC into linear filaments and their condensation into macromolecular ASC specks promotes an all-or-none response. These structures are highly regulated and provide a unique signaling platform or compartment to control the activity of caspase-1 and likely other effectors. Emerging evidence indicates that ASC specks are also released from inflammasome-activated cells and accumulate in inflamed tissues, where they can continue to mature cytokines or be internalized by surrounding cells to further nucleate ASC specks in their cytosol. Little is known about the mechanisms governing ASC speck release, uptake, and endosomal escape, as well as its contribution to inflammation and disease. Here, we describe the different outcomes of inflammasome activation and discuss the potential function of extracellular ASC specks. We highlight gaps in our understanding of this central process of inflammation, which may have direct consequences on the modulation of host responses and chronic inflammation.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Enfermedades Transmisibles/inmunología , Enfermedades del Sistema Inmune/inmunología , Inmunoterapia/métodos , Inflamasomas/metabolismo , Inflamación/metabolismo , Interleucina-1/metabolismo , Animales , Autoinmunidad , Caspasa 1/metabolismo , Enfermedades Transmisibles/terapia , Espacio Extracelular , Humanos , Enfermedades del Sistema Inmune/terapia , Complejos Multiproteicos/metabolismo , Multimerización de Proteína , Transducción de Señal
5.
J Immunol ; 202(2): 550-558, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30559319

RESUMEN

Charcot-Leyden crystals (CLCs) are Galectin-10 protein crystals that can form after eosinophils degranulate. CLCs can appear and persist in tissues from patients with eosinophilic disorders, such as asthma, allergic reactions, and fungal and helminthic infections. Despite abundant reports of their occurrence in human disease, the inflammatory potential of CLCs has remained unknown. In this article, we show that CLCs induce the release of the proinflammatory cytokine IL-1ß upon their phagocytosis by primary human macrophages in vitro. Chemical inhibition and small interfering RNA knockdown of NLRP3 in primary human macrophages abrogated their IL-1ß response to CLCs. Using C57BL/6 ASC-mCitrine transgenic inflammasome reporter mice, we showed that the instillation of CLCs into the lungs promoted the assembly of ASC complexes in infiltrating immune cells (neutrophils and inflammatory monocytes) and resulted in IL-1ß accumulation into the bronchoalveolar lavage fluid. Our findings reveal that CLCs are recognized by the NLRP3 inflammasome, which may sustain inflammation that follows eosinophilic inflammatory processes.


Asunto(s)
Eosinófilos/fisiología , Galectinas/metabolismo , Inflamasomas/metabolismo , Inflamación/inmunología , Pulmón/fisiología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Degranulación de la Célula , Células Cultivadas , Cristalización , Galectinas/química , Humanos , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fagocitosis , Cultivo Primario de Células , ARN Interferente Pequeño/genética
6.
Cell Commun Signal ; 18(1): 141, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32894139

RESUMEN

BACKGROUND: Low molecular weight carrageenan (Cg) is a seaweed-derived sulfated polysaccharide widely used as inflammatory stimulus in preclinical studies. However, the molecular mechanisms of Cg-induced inflammation are not fully elucidated. The present study aimed to investigate the molecular basis involved in Cg-induced macrophages activation and cytokines production. METHODS: Primary culture of mouse peritoneal macrophages were stimulated with Kappa Cg. The supernatant and cell lysate were used for ELISA, western blotting, immunofluorescence. Cg-induced mouse colitis was also developed. RESULTS: Here we show that Cg activates peritoneal macrophages to produce pro-inflammatory cytokines such as TNF and IL-1ß. While Cg-induced TNF production/secretion depends on TLR4/MyD88 signaling, the production of pro-IL-1ß relies on TLR4/TRIF/SYK/reactive oxygen species (ROS) signaling pathway. The maturation of pro-IL1ß into IL-1ß is dependent on canonical NLRP3 inflammasome activation via Pannexin-1/P2X7/K+ efflux signaling. In vivo, Cg-induced colitis was reduced in mice in the absence of NLRP3 inflammasome components. CONCLUSIONS: In conclusion, we unravel a critical role of the NLRP3 inflammasome in Cg-induced pro-inflammatory cytokines production and colitis, which is an important discovery on the pro-inflammatory properties of this sulfated polysaccharide for pre-clinical studies. Video abstract Carrageenan (Cg) is one the most used flogistic stimulus in preclinical studies. Nevertheless, the molecular basis of Cg-induced inflammation is not totally elucidated. Herein, Lopes et al. unraveled the molecular basis for Cg-induced macrophages production of biological active IL-1ß. The Cg-stimulated macrophages produces pro-IL-1ß depends on TLR4/TRIF/Syk/ROS, whereas its processing into mature IL-1ß is dependent on the canonical NLRP3 inflammasome.


Asunto(s)
Carragenina/inmunología , Citocinas/inmunología , Activación de Macrófagos , Macrófagos Peritoneales/inmunología , Animales , Células Cultivadas , Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Factor de Necrosis Tumoral alfa/inmunología
8.
J Mol Cell Cardiol ; 104: 43-52, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28143713

RESUMEN

BACKGROUND: Vascular smooth muscle cell (VSMC) proliferation is of importance in the pathogenesis of vascular diseases such as restenosis or atherosclerosis. Endothelial microparticles (EMPs) regulate function and phenotype of target endothelial cells (ECs), but their influence on VSMC biology is unknown. We aim to investigate the role of EMPs in the regulation of vascular smooth muscle cell (VSMC) proliferation and vascular remodeling. METHODS AND RESULTS: Systemic treatment of mice with EMPs after vascular injury reduced neointima formation in vivo. In vitro, EMP uptake in VSMCs diminished VSMC proliferation and migration, both pivotal steps in neointima formation. To explore the underlying mechanisms, Taqman microRNA-array was performed and miR-126-3p was identified as the predominantly expressed miR in EMPs. Confocal microscopy revealed an EMP-mediated miR-126 transfer into recipient VSMCs. Expression of miR-126 target protein LRP6, regulating VSMC proliferation, was reduced in VSMCs after EMP treatment. Importantly, genetic regulation of miR-126 in EMPs showed a miR-126-dependent inhibition of LRP6 expression, VSMC proliferation and neointima formation in vitro and in vivo, suggesting a crucial role of miR-126 in EMP-mediated neointima formation reduction. Finally, analysis of miR-126 expression in circulating MPs in 176 patients with coronary artery disease revealed a reduced PCI rate in patients with high miR-126 expression level, supporting a central role for MP-incorporated miR-126 in vascular remodelling. CONCLUSION: EMPs reduce VSMC proliferation, migration and subsequent neointima formation by delivering functional miR-126 into recipient VSMCs.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Células Endoteliales/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Anciano , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Transporte Biológico , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Persona de Mediana Edad , Neointima/patología , Interferencia de ARN
9.
Kidney Int ; 90(3): 525-39, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27262364

RESUMEN

Intrarenal crystal formation activates the Nlrp3 inflammasome in myeloid cells and triggers a profound inflammatory response. Here, we studied whether a specific inhibitor of the Nlrp3 inflammasome, CP-456,773, can prevent kidney fibrosis in a murine model of crystal nephropathy induced by diets rich in oxalate or adenine. Inflammasome activation in renal dendritic cells and the resulting interleukin (IL)-1ß and IL-18 production were markedly reduced by CP-456,773 treatment both ex vivo and in vivo. We directly visualized intrarenal inflammasome activation and its inhibition by CP-456,773 in vivo by adoptive transfer of bone marrow cells transduced with interleukin-1ß-Gaussia luciferase, a proteolytic luciferase-based reporter for inflammasome activation, into irradiated mice. CP-456,773 treatment strongly attenuated kidney fibrosis when given early in the genesis of crystal nephropathy, but was unable to reverse established crystal-induced fibrosis. The urinary IL-18 concentration appeared to be a useful noninvasive biomarker for renal inflammasome activation. Finally, NLRP3 inhibition did not compromise adaptive immune responses as previously reported for the global inhibition of IL-1 signaling. Thus, early NLRP3 inhibition by CP-456,773 may be an effective treatment for crystal nephropathy. Use of iGLuc transfected cells introduces a novel imaging technique for inflammasome activation in mice.


Asunto(s)
Células Dendríticas/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Inflamasomas/efectos de los fármacos , Riñón/patología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Nefritis/tratamiento farmacológico , Sulfonas/uso terapéutico , Adenina/efectos adversos , Traslado Adoptivo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Furanos , Humanos , Inmunohistoquímica , Indenos , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Riñón/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nefritis/inducido químicamente , Oxalatos/efectos adversos , Cultivo Primario de Células , Transducción de Señal , Sulfonamidas
10.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G724-G733, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27634010

RESUMEN

Nonalcoholic steatohepatitis (NASH), especially as part of the metabolic syndrome (MS), is an increasing burden in Western countries. Statins are already used in MS and seem to be beneficial in liver diseases. The aim of this study was to investigate the molecular mechanisms underlying pleiotropic effects on small GTPases of statins in NASH. NASH within MS was induced in 12-wk-old apoE-/- mice after 7 wk of Western diet (NASH mice). Small GTPases were inhibited by activated simvastatin (SMV), NSC23766 (NSC), or Clostridium sordellii lethal toxin (LT) by using subcutaneous osmotic minipumps. Hepatic steatosis, inflammation, and fibrosis were assessed by histology, Western blot, and RT-PCR measurements of cholesterol and hydroxyproline content. SMV treatment significantly decreased hepatic inflammation and fibrosis, but had no significant effect on steatosis and hepatic cholesterol content in NASH. SMV blunted fibrosis due to inhibition of both RhoA/Rho kinase and Ras/ERK pathways. Interestingly, inhibition of RAC1 and Ras (by LT) failed to decrease fibrosis to the same extent. Inhibition of RAC1 (by NSC) showed no significant effect at all. Inhibition of RhoA and Ras downstream signaling by statins is responsible for the beneficial hepatic effects in NASH.


Asunto(s)
Colesterol/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Simvastatina/uso terapéutico , Proteínas ras/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Dieta Occidental , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hígado/metabolismo , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología
11.
PLoS Pathog ; 10(1): e1003885, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24453977

RESUMEN

Cyclic paroxysm and high fever are hallmarks of malaria and are associated with high levels of pyrogenic cytokines, including IL-1ß. In this report, we describe a signature for the expression of inflammasome-related genes and caspase-1 activation in malaria. Indeed, when we infected mice, Plasmodium infection was sufficient to promote MyD88-mediated caspase-1 activation, dependent on IFN-γ-priming and the expression of inflammasome components ASC, P2X7R, NLRP3 and/or NLRP12. Pro-IL-1ß expression required a second stimulation with LPS and was also dependent on IFN-γ-priming and functional TNFR1. As a consequence of Plasmodium-induced caspase-1 activation, mice produced extremely high levels of IL-1ß upon a second microbial stimulus, and became hypersensitive to septic shock. Therapeutic intervention with IL-1 receptor antagonist prevented bacterial-induced lethality in rodents. Similar to mice, we observed a significantly increased frequency of circulating CD14(+)CD16(-)Caspase-1(+) and CD14(dim)CD16(+)Caspase-1(+) monocytes in peripheral blood mononuclear cells from febrile malaria patients. These cells readily produced large amounts of IL-1ß after stimulation with LPS. Furthermore, we observed the presence of inflammasome complexes in monocytes from malaria patients containing either NLRP3 or NLRP12 pyroptosomes. We conclude that NLRP12/NLRP3-dependent activation of caspase-1 is likely to be a key event in mediating systemic production of IL-1ß and hypersensitivity to secondary bacterial infection during malaria.


Asunto(s)
Infecciones Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Malaria Vivax/microbiología , Plasmodium chabaudi/metabolismo , Plasmodium vivax/metabolismo , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Femenino , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Malaria Vivax/inmunología , Malaria Vivax/metabolismo , Malaria Vivax/patología , Masculino , Ratones , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Proteína con Dominio Pirina 3 de la Familia NLR , Plasmodium chabaudi/inmunología , Plasmodium vivax/inmunología , Choque Séptico/genética , Choque Séptico/inmunología , Choque Séptico/metabolismo , Choque Séptico/patología
12.
Cardiovasc Diabetol ; 15: 49, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27005938

RESUMEN

BACKGROUND: Circulating microRNAs (miRs) are differentially regulated and selectively packaged into microparticles (MPs). We evaluated whether diabetes mellitus alters circulating vascular and endothelial MP-incorporated miRs expression levels. METHODS AND RESULTS: Circulating MPs were isolated from 135 patients with or without diabetes mellitus type II and characterized using flow cytometer and electron microscope. Nine miRs involved in the regulation of vascular performance-miR-126, miR-222, miR-let7d, miR-21, miR-30, miR-92a, miR-139, miR-199a and miR-26a-were quantified in circulating MPs by reverse transcription polymerase chain reaction. Among those, miR-126 and miR-26a were significantly reduced in diabetic patients compared to non-diabetic patients. Patients with low miR-26a and miR-126 levels were at higher risk for a concomitant coronary artery disease. MP-sorting experiments showed that endothelial cells were the major cell sources of MPs containing miR-126 and miR-26a, respectively. Finally, in accordance with our clinical results, in vitro experiments revealed that hyperglycemia reduces the packaging of miR-126 and miR-26a into EMPs. CONCLUSION: Diabetes mellitus significantly alters the expression of vascular endothelial miRs in circulating endothelial MPs with potential implications on vascular heath.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Diabetes Mellitus Tipo 2/genética , Células Endoteliales/metabolismo , MicroARNs/genética , Anciano , Glucemia/metabolismo , Estudios de Casos y Controles , Micropartículas Derivadas de Células/ultraestructura , Células Cultivadas , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Células Endoteliales/ultraestructura , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Marcadores Genéticos , Humanos , Masculino , MicroARNs/sangre , Microscopía Electrónica , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Circulation ; 128(18): 2026-38, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24014835

RESUMEN

BACKGROUND: Repair of the endothelium after vascular injury is crucial for preserving endothelial integrity and preventing the development of vascular disease. The underlying mechanisms of endothelial cell repair are largely unknown. We sought to investigate whether endothelial microparticles (EMPs), released from apoptotic endothelial cells (ECs), influence EC repair. METHODS AND RESULTS: Systemic treatment of mice with EMPs after electric denudation of the endothelium accelerated reendothelialization in vivo. In vitro experiments revealed that EMP uptake in ECs promotes EC migration and proliferation, both critical steps in endothelial repair. To dissect the underlying mechanisms, Taqman microRNA array was performed, and microRNA (miR)-126 was identified as the predominantly expressed miR in EMPs. The following experiments demonstrated that miR-126 was transported into recipient human coronary artery endothelial cells by EMPs and functionally regulated the target protein sprouty-related, EVH1 domain-containing protein 1 (SPRED1). Knockdown of miR-126 in EMPs abrogated EMP-mediated effects on human coronary artery endothelial cell migration and proliferation in vitro and reendothelialization in vivo. Interestingly, after simulating diabetic conditions, EMPs derived from glucose-treated ECs contained significantly lower amounts of miR-126 and showed reduced endothelial repair capacity in vitro and in vivo. Finally, expression analysis of miR-126 in circulating microparticles from 176 patients with stable coronary artery disease with and without diabetes mellitus revealed a significantly reduced miR-126 expression in circulating microparticles from diabetic patients. CONCLUSIONS: Endothelial microparticles promote vascular endothelial repair by delivering functional miR-126 into recipient cells. In pathological hyperglycemic conditions, EMP-mediated miR-126-induced EC repair is altered.


Asunto(s)
Micropartículas Derivadas de Células/fisiología , Vasos Coronarios/fisiología , Células Endoteliales/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Proteínas Represoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/fisiopatología , Movimiento Celular/fisiología , Proliferación Celular , Micropartículas Derivadas de Células/patología , Células Cultivadas , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/fisiopatología , Vasos Coronarios/lesiones , Vasos Coronarios/patología , Células Endoteliales/patología , Glucosa/toxicidad , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/patología , Ratones Endogámicos C57BL , Cicatrización de Heridas/fisiología
14.
Proc Natl Acad Sci U S A ; 108(9): 3689-94, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21303985

RESUMEN

Excessive release of proinflammatory cytokines by innate immune cells is an important component of the pathogenic basis of malaria. Proinflammatory cytokines are a direct output of Toll-like receptor (TLR) activation during microbial infection. Thus, interference with TLR function is likely to render a better clinical outcome by preventing their aberrant activation and the excessive release of inflammatory mediators. Herein, we describe the protective effect and mechanism of action of E6446, a synthetic antagonist of nucleic acid-sensing TLRs, on experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA. We show that in vitro, low doses of E6446 specifically inhibited the activation of human and mouse TLR9. Tenfold higher concentrations of this compound also inhibited the human TLR8 response to single-stranded RNA. In vivo, therapy with E6446 diminished the activation of TLR9 and prevented the exacerbated cytokine response observed during acute Plasmodium infection. Furthermore, severe signs of ECM, such as limb paralysis, brain vascular leak, and death, were all prevented by oral treatment with E6446. Hence, we provide evidence that supports the involvement of nucleic acid-sensing TLRs in malaria pathogenesis and that interference with the activation of these receptors is a promising strategy to prevent deleterious inflammatory responses that mediate pathogenesis and severity of malaria.


Asunto(s)
Hidrocarburos Aromáticos/farmacología , Malaria Cerebral/prevención & control , Malaria Cerebral/terapia , Ácidos Nucleicos/metabolismo , Receptores Toll-Like/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Humanos , Hidrocarburos Aromáticos/química , Inflamación/complicaciones , Inflamación/patología , Lipopolisacáridos/farmacología , Malaria Cerebral/inducido químicamente , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium chabaudi/efectos de los fármacos , Plasmodium chabaudi/fisiología , Choque Séptico/inducido químicamente , Choque Séptico/complicaciones , Receptores Toll-Like/metabolismo
15.
Nat Rev Rheumatol ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886559

RESUMEN

Kawasaki disease, a systemic vasculitis that affects young children and can result in coronary artery aneurysms, is the leading cause of acquired heart disease among children. A hallmark of Kawasaki disease is increased blood platelet counts and platelet activation, which is associated with an increased risk of developing resistance to intravenous immunoglobulin and coronary artery aneurysms. Platelets and their releasate, including granules, microparticles, microRNAs and transcription factors, can influence innate immunity, enhance inflammation and contribute to vascular remodelling. Growing evidence indicates that platelets also interact with immune and non-immune cells to regulate inflammation. Platelets boost NLRP3 inflammasome activation and IL-1ß production by human immune cells by releasing soluble mediators. Activated platelets form aggregates with leukocytes, such as monocytes and neutrophils, enhancing numerous functions of these cells and promoting thrombosis and inflammation. Leukocyte-platelet aggregates are increased in children with Kawasaki disease during the acute phase of the disease and can be used as biomarkers for disease severity. Here we review the role of platelets in Kawasaki disease and discuss progress in understanding the immune-effector role of platelets in amplifying inflammation related to Kawasaki disease vasculitis and therapeutic strategies targeting platelets or platelet-derived molecules.

16.
Br J Pharmacol ; 181(8): 1308-1323, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37990806

RESUMEN

BACKGROUND AND PURPOSE: Sepsis-surviving adult individuals commonly develop immunosuppression and increased susceptibility to secondary infections, an outcome mediated by the axis IL-33/ILC2s/M2 macrophages/Tregs. Nonetheless, the long-term immune consequences of paediatric sepsis are indeterminate. We sought to investigate the role of age in the genesis of immunosuppression following sepsis. EXPERIMENTAL APPROACH: Here, we compared the frequency of Tregs, the activation of the IL-33/ILC2s axis in M2 macrophages and the DNA methylation of epithelial lung cells from post-septic infant and adult mice. Likewise, sepsis-surviving mice were inoculated intranasally with Pseudomonas aeruginosa or by subcutaneous inoculation of the B16 melanoma cell line. Finally, blood samples from sepsis-surviving patients were collected and the concentration of IL-33 and Tregs frequency were assessed. KEY RESULTS: In contrast to 6-week-old mice, 2-week-old mice were resistant to secondary infection and did not show impairment in tumour controls upon melanoma challenge. Mechanistically, increased IL-33 levels, Tregs expansion, and activation of ILC2s and M2-macrophages were observed in 6-week-old but not 2-week-old post-septic mice. Moreover, impaired IL-33 production in 2-week-old post-septic mice was associated with increased DNA methylation in lung epithelial cells. Notably, IL-33 treatment boosted the expansion of Tregs and induced immunosuppression in 2-week-old mice. Clinically, adults but not paediatric post-septic patients exhibited higher counts of Tregs and seral IL-33 levels. CONCLUSION AND IMPLICATIONS: These findings demonstrate a crucial and age-dependent role for IL-33 in post-sepsis immunosuppression. Thus, a better understanding of this process may lead to differential treatments for adult and paediatric sepsis.


Asunto(s)
Interleucina-33 , Sepsis , Humanos , Ratones , Animales , Niño , Inmunidad Innata , Linfocitos/metabolismo , Linfocitos/patología , Terapia de Inmunosupresión
17.
EMBO Mol Med ; 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38977927

RESUMEN

In humans, blood Classical CD14+ monocytes contribute to host defense by secreting large amounts of pro-inflammatory cytokines. Their aberrant activity causes hyper-inflammation and life-threatening cytokine storms, while dysfunctional monocytes are associated with 'immunoparalysis', a state of immune hypo responsiveness and reduced pro-inflammatory gene expression, predisposing individuals to opportunistic infections. Understanding how monocyte functions are regulated is critical to prevent these harmful outcomes. We reveal platelets' vital role in the pro-inflammatory cytokine responses of human monocytes. Naturally low platelet counts in patients with immune thrombocytopenia or removal of platelets from healthy monocytes result in monocyte immunoparalysis, marked by impaired cytokine response to immune challenge and weakened host defense transcriptional programs. Remarkably, supplementing monocytes with fresh platelets reverses these conditions. We discovered that platelets serve as reservoirs of key cytokine transcription regulators, such as NF-κB and MAPK p38, and pinpointed the enrichment of platelet NF-κB2 in human monocytes by proteomics. Platelets proportionally restore impaired cytokine production in human monocytes lacking MAPK p38α, NF-κB p65, and NF-κB2. We uncovered a vesicle-mediated platelet-monocyte-propagation of inflammatory transcription regulators, positioning platelets as central checkpoints in monocyte inflammation.

18.
Arterioscler Thromb Vasc Biol ; 32(8): 1925-35, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22701020

RESUMEN

OBJECTIVE: Endothelial microparticles (EMP) are released from activated or apoptotic cells, but their effect on target cells and the exact way of incorporation are largely unknown. We sought to determine the uptake mechanism and the biological effect of EMP on endothelial and endothelial-regenerating cells. METHODS AND RESULTS: EMP were generated from starved endothelial cells and isolated by ultracentrifugation. Caspase 3 activity assay and terminal deoxynucleotidyl transferase dUTP nick end labeling assay showed that EMP protect target endothelial cells against apoptosis in a dose-dependent manner. Proteomic analysis was performed to identify molecules contained in EMP, which might be involved in EMP uptake. Expression of annexin I in EMP was found and confirmed by Western blot, whereas the corresponding receptor phosphatidylserine receptor was present on endothelial target cells. Silencing either annexin I on EMP or phosphatidylserine receptor on target cells using small interfering RNA showed that the uptake of EMP by human coronary artery endothelial cells is annexin I/phosphatidylserine receptor dependent. Annexin I-downregulated EMP abrogated the EMP-mediated protection against apoptosis of endothelial target cells. p38 activation was found to mediate camptothecin-induced apoptosis. Finally, human coronary artery endothelial cells pretreated with EMP inhibited camptothecin-induced p38 activation. CONCLUSIONS: EMP are incorporated by endothelial cells in an annexin I/phosphatidylserine receptor-dependent manner and protect target cells against apoptosis. Inhibition of p38 activity is involved in EMP-mediated protection against apoptosis.


Asunto(s)
Anexina A1/fisiología , Apoptosis , Micropartículas Derivadas de Células/fisiología , Células Endoteliales/fisiología , Receptores de Superficie Celular/fisiología , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , Células Cultivadas , Humanos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
19.
Methods Mol Biol ; 2696: 73-92, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37578716

RESUMEN

Inflammasomes are crucial sentinels of the innate immune system that sense clues of infection, cellular stress, or metabolic imbalances. Upon activation, the inflammasome sensor (e.g., NLRP3) recruits the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC). ASC rapidly oligomerizes to form a micron-sized structure termed "ASC speck." These are crucial for the activation of caspase-1 and downstream inflammatory signals released following a specific form of lytic cell death called pyroptosis. Hence, due to their considerably large size, ASC specks can be easily visualized by microscopy as a simple upstream readout for inflammasome activation. Here, we provide three detailed protocols for imaging ASC specks: (1) live-cell imaging of macrophage cell lines expressing a fluorescent protein fusion form of ASC, (2) imaging of human primary cells using immunofluorescence staining of endogenous ASC, and (3) visualization and quantification of specks on a single-cell level using imaging flow cytometry.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Inflamasomas , Humanos , Inflamasomas/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Apoptosis , Caspasa 1/metabolismo , Microscopía Confocal , Técnica del Anticuerpo Fluorescente , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
20.
JCI Insight ; 8(14)2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37279077

RESUMEN

Kawasaki disease (KD) is the leading cause of acquired heart disease among children. Increased platelet counts and activation are observed during the course of KD, and elevated platelet counts are associated with higher risks of developing intravenous immunoglobulin resistance and coronary artery aneurysms. However, the role of platelets in KD pathogenesis remains unclear. Here, we analyzed transcriptomics data generated from the whole blood of patients with KD and discovered changes in the expression of platelet-related genes during acute KD. In the Lactobacillus casei cell wall extract (LCWE) murine model of KD vasculitis, LCWE injection increased platelet counts and the formation of monocyte-platelet aggregates (MPAs), upregulated the concentration of soluble P-selectin, and increased circulating thrombopoietin and interleukin 6 (IL-6). Furthermore, platelet counts correlated with the severity of cardiovascular inflammation. Genetic depletion of platelets (Mpl-/- mice) or treatment with an anti-CD42b antibody significantly reduced LCWE-induced cardiovascular lesions. Furthermore, in the mouse model, platelets promoted vascular inflammation via the formation of MPAs, which likely amplified IL-1B production. Altogether, our results indicate that platelet activation exacerbates the development of cardiovascular lesions in a murine model of KD vasculitis. These findings enhance our understanding of KD vasculitis pathogenesis and highlight MPAs, which are known to enhance IL-1B production, as a potential therapeutic target for this disorder.


Asunto(s)
Síndrome Mucocutáneo Linfonodular , Vasculitis , Animales , Ratones , Síndrome Mucocutáneo Linfonodular/genética , Síndrome Mucocutáneo Linfonodular/tratamiento farmacológico , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Inflamación
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda