Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 429
Filtrar
1.
Int Immunopharmacol ; 113(Pt A): 109375, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36461592

RESUMEN

BACKGROUND: Recent studies have uncovered that hyperuricemia (HUA) leads to cognitive deficits, which are accompanied by neuronal damage and neuroinflammation. Here, we aim to explore the role of methyltransferase-like 3 (METTL3) in HUA-mediated neuronal apoptosis and microglial inflammation. METHODS: A HUA mouse model was constructed. The spatial memory ability of the mice was assessed by the Morris water maze experiment (MWM), and neuronal apoptosis was analyzed by the TdT-mediated dUTP nick end labeling (TUNEL) assay. Besides, enzyme-linked immunosorbent assay (ELISA) was utilized to measure the contents of inflammatory factors (IL-1ß, IL-6, and TNF-α) and oxidative stress markers (MDA, SOD, and CAT) in the serum of mice. In vitro, the mouse hippocampal neuron (HT22) and microglia (BV2) were treated with uric acid (UA). Flow cytometry was applied to analyze HT22 and BV2 cell apoptosis, and ELISA was conducted to observe neuroinflammation and oxidative stress. In addition, the expression of MyD88, p-NF-κB, NF-κB, NLRP3, ASC and Caspase1 was determined by Western blot. RESULTS: METTL3 and miR-124-3p were down-regulated, while the MyD88-NF-κB pathway was activated in the HUA mouse model. UA treatment induced neuronal apoptosis in HT22 and stimulated microglial activation in BV2. Overexpressing METTL3 alleviated HT22 neuronal apoptosis and resisted the release of inflammatory cytokines and oxidative stress mediators in BV2 cells. METTL3 repressed MyD88-NF-κB and NLRP3-ASC-Caspase1 inflammasome. In addition, METTL3 overexpression enhanced miR-124-3p expression, while METTL3 knockdown aggravated HT22 cell apoptosis and BV2 cell overactivation. CONCLUSION: METTL3 improves neuronal apoptosis and microglial activation in the HUA model by choking the MyD88/NF-κB pathway and up-regulating miR-124-3p.


Asunto(s)
Disfunción Cognitiva , Hiperuricemia , Inflamasomas , Metiltransferasas , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Células Cultivadas , Disfunción Cognitiva/etiología , Disfunción Cognitiva/genética , Disfunción Cognitiva/inmunología , Modelos Animales de Enfermedad , Hiperuricemia/complicaciones , Hiperuricemia/genética , Hiperuricemia/inmunología , Inflamasomas/genética , Inflamasomas/inmunología , Metiltransferasas/genética , Metiltransferasas/inmunología , MicroARNs/genética , MicroARNs/inmunología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/inmunología , Sistema Nervioso/fisiopatología , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/genética , Enfermedades Neuroinflamatorias/inmunología , FN-kappa B , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Ácido Úrico/administración & dosificación , Ácido Úrico/efectos adversos , Ácido Úrico/farmacología
2.
Front Immunol ; 13: 934264, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35844548

RESUMEN

Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), responsible for COVID-19, has caused a global pandemic. Observational studies revealed a condition, herein called as Long-COVID syndrome (PC), that affects both moderately and severely infected patients, reducing quality-of-life. The mechanism/s underlying the onset of fibrotic-like changes in PC are still not well defined. The goal of this study was to understand the involvement of the Absent in melanoma-2 (AIM2) inflammasome in PC-associated lung fibrosis-like changes revealed by chest CT scans. Peripheral blood mononuclear cells (PBMCs) obtained from PC patients who did not develop signs of lung fibrosis were not responsive to AIM2 activation by Poly dA:dT. In sharp contrast, PBMCs from PC patients with signs of lung fibrosis were highly responsive to AIM2 activation, which induced the release of IL-1α, IFN-α and TGF-ß. The recognition of Poly dA:dT was not due to the activation of cyclic GMP-AMP (cGAMP) synthase, a stimulator of interferon response (cGAS-STING) pathways, implying a role for AIM2 in PC conditions. The release of IFN-α was caspase-1- and caspase-4-dependent when AIM2 was triggered. Instead, the release of pro-inflammatory IL-1α and pro-fibrogenic TGF-ß were inflammasome independent because the inhibition of caspase-1 and caspase-4 did not alter the levels of the two cytokines. Moreover, the responsiveness of AIM2 correlated with higher expression of the receptor in circulating CD14+ cells in PBMCs from patients with signs of lung fibrosis.


Asunto(s)
COVID-19 , Proteínas de Unión al ADN , Fibrosis Pulmonar , COVID-19/sangre , COVID-19/inmunología , COVID-19/patología , Proteínas Portadoras , Caspasa 1/inmunología , Proteínas de Unión al ADN/sangre , Proteínas de Unión al ADN/inmunología , Humanos , Inflamasomas/sangre , Inflamasomas/inmunología , Interferón-alfa/metabolismo , Leucocitos Mononucleares/inmunología , Fibrosis Pulmonar/sangre , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/virología , SARS-CoV-2 , Factor de Crecimiento Transformador beta/metabolismo , Síndrome Post Agudo de COVID-19
3.
Eur J Pediatr ; 181(8): 3093-3101, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35705877

RESUMEN

The exact immunological mechanisms of post infectious bronchiolitis obliterans (PIBO) in childhood are not fully known. It has been shown that the inflammasome and IL-18 pathway play important roles in the pathogenesis of lung fibrosis. We aimed to investigate the role of caspase-1, IL-18, and IL-18 components in PIBO. From January to May 2020, children with PIBO, children with history of influenza infection without PIBO, and healthy children were asked to participate in the study in three pediatric pulmonology centers. Serum caspase-1, IL-18, IL-18BP, IL-18R, and INF-γ levels were measured by ELISA and compared between the 3 groups. There were 21 children in the PIBO group, 16 children in the influenza group, and 39 children in the healthy control group. No differences in terms of age and gender between the 3 groups were found. IL-18 and IL-18BP levels were higher in the healthy control group (p = 0.018, p = 0.005, respectively). IL-18R was higher in the PIBO group (p = 0.001) and caspase-1 was higher in the PIBO and influenza group than the healthy control group (p = 0.002). IFN-γ levels did not differ between the 3 groups. IL-18BP/IL-18 was higher in the influenza group than the PIBO group and the healthy control group (p = 0.003). CONCLUSIONS: Caspase-1 level was increased in patients with PIBO which suggests that inflammasome activation may have a role in fibrosis; however, IL-18 level was found to be low. Mediators other than IL-18 may be involved in the inflammatory pathway in PIBO. Further immunological studies investigating inflammasome pathway are needed for PIBO with chronic inflammation. WHAT IS KNOWN: • Post infectious bronchiolitis obliterans (PIBO) is a rare, severe chronic lung disease during childhood which is associated with inflammation and fibrosis which lead to partial or complete luminal obstruction especially in small airways. • The exact immunological mechanisms of PIBO in childhood are not fully known. WHAT IS NEW: • Inflammasome activation persists even years after acute infection and may play a role in fibrosis in PIBO. • Mediators other than IL-18 may be involved in these inflammatory pathway.


Asunto(s)
Bronquiolitis Obliterante , Caspasa 1 , Interleucina-18 , Bronquiolitis Obliterante/sangre , Bronquiolitis Obliterante/etiología , Bronquiolitis Obliterante/genética , Bronquiolitis Obliterante/inmunología , Estudios de Casos y Controles , Caspasa 1/sangre , Caspasa 1/genética , Caspasa 1/inmunología , Niño , Fibrosis/sangre , Fibrosis/genética , Fibrosis/inmunología , Humanos , Inflamasomas/inmunología , Inflamación/sangre , Inflamación/genética , Inflamación/inmunología , Gripe Humana/sangre , Gripe Humana/complicaciones , Gripe Humana/genética , Gripe Humana/inmunología , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Interleucina-18/sangre , Interleucina-18/genética , Interleucina-18/inmunología
4.
Front Immunol ; 13: 810582, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154128

RESUMEN

Neuroinflammation has been proven to exert an important effect on brain injury after intracerebral hemorrhage (ICH). Previous studies reported that Didymin possessed anti-inflammatory properties after acute hepatic injury, hyperglycemia-induced endothelial dysfunction, and death. However, the role of Didymin in microglial pyroptosis and neuroinflammation after ICH is unclear. The current study aimed to investigate the effect of Didymin on neuroinflammation mediated by microglial pyroptosis in mouse models of ICH and shed some light on the underlying mechanisms. In this study, we observed that Didymin treatment remarkably improved neurobehavioral performance and decreased BBB disruption and brain water content. Microglial activation and neutrophil infiltration in the peri-hematoma tissue after ICH were strikingly mitigated by Didymin as well. At the molecular level, administration of Didymin significantly unregulated the expression of Rkip and downregulated the expression of pyroptotic molecules and inflammatory cytokines such as Nlrp3 inflammasome, GSDMD, caspase-1, and mature IL-1ß, TNF-α, and MPO after ICH. Besides, Didymin treatment decreased the number of Caspase-1-positive microglia and GSDMD-positive microglia after ICH. Inversely, Locostatin, an Rkip-specific inhibitor, significantly abolished the anti-pyroptosis and anti-neuroinflammation effects of Didymin. Moreover, Rkip binding with Asc could interrupt the activation and assembly of the inflammasome. Mechanistically, inhibition of Caspase-1 by VX-765 attenuated brain injury and suppressed microglial pyroptosis and neuroinflammation by downregulation of GSDMD, mature IL-1ß, TNF-α, and MPO based on Locostatin-treated ICH. Taken together, Didymin alleviated microglial pyroptosis and neuroinflammation, at least in part through the Asc/Caspase-1/GSDMD pathway via upregulating Rkip expression after ICH. Therefore, Didymin may be a potential agent to attenuate neuroinflammation via its anti-pyroptosis effect after ICH.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/inmunología , Caspasa 1/inmunología , Microglía/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Proteínas de Unión a Fosfato/antagonistas & inhibidores , Proteínas de Unión a Fosfatidiletanolamina/inmunología , Proteínas Citotóxicas Formadoras de Poros/antagonistas & inhibidores , Animales , Células Cultivadas , Hemorragia Cerebral , Flavonoides/farmacología , Glicósidos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Enfermedades Neuroinflamatorias/inmunología , Proteínas de Unión a Fosfato/inmunología , Proteínas Citotóxicas Formadoras de Poros/inmunología
5.
Ann N Y Acad Sci ; 1508(1): 92-104, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34671981

RESUMEN

Recently, interleukin-38 (IL-38) was identified as an important anti-inflammatory and immunosuppressive factor, but its functional role in temporomandibular joint (TMJ) inflammation remains unknown. This study aimed to elucidate how IL-38 affects chondrocytes and the underlying mechanism that contributes to anti-inflammatory processes in the TMJ. Western blotting, quantitative real-time PCR, enzyme-linked immunosorbent assay, and immunofluorescence analysis were used to verify that IL-38 has anti-inflammatory effects on chondrocytes, and the related key pathways were analyzed by western blotting. SiRNA-IL-38, siRNA-NLRP3, and MCC950 were used to investigate the mechanism underlying the anti-inflammatory effects of IL-38. Inflammation models were induced by injection of complete Freund's adjuvant in TMJ with mouse recombinant IL-38 in in vivo studies. Histological and immunohistochemical analyses were used to investigate histological changes in the cartilage. The results showed that IL-38 inhibited the expression of inflammatory cytokines and MMPs. IL-38 limited inflammation by inhibiting the expression of MAPKs/NF-κB and the NLRP3/caspase-1 pathway. In vivo, IL-38 reduced chondrocyte inflammation and limited cartilage degeneration. This study shows for the first time that IL-38 plays a protective role in TMJ cartilage. IL-38 exerts anti-inflammatory effects through the NLRP3/caspase-1 pathway and may be a promising agent for treating TMJ inflammation.


Asunto(s)
Caspasa 1/inmunología , Inflamación/inmunología , Interleucina-1/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Osteoartritis/inmunología , Articulación Temporomandibular/inmunología , Animales , Cartílago/inmunología , Masculino , Ratones
6.
Scand J Immunol ; 95(2): e13124, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34861056

RESUMEN

Cells encounter continuous challenges due to tissue insult caused by endogenous and/or exogenous stimuli. Among the mechanisms set in place to counterbalance the tissue insult, innate immunity is always at the forefront. Cells of innate immunity efficiently recognize the 'danger signals' via a specialized set of membrane-bound receptors known as Toll-like receptors. Once this interaction is established, toll-like receptor passes on the responsibility to cytosolic NOD-like receptors through a cascade of signalling pathways. Subsequently, NOD-like receptors assemble to a specialized multiprotein intracellular complex, that is inflammasome. Inflammasome activates Caspase-1 and Gasdermin-D which initiate pyroptotic cell death in the affected tissue by two simultaneous mechanisms. Being a protease, caspase-1 cleaves and activates pro-inflammatory cytokines IL-1ß and IL-18. On the other hand, Gasdermin-D causes proteolytic cleavage which forms a pore in the cell membrane. This review highlights the molecular events ranging from recognition of stimuli to pyroptosis. The review is also an attempt to discuss the mechanisms of the most specific experimental NLRP3 inhibitors.


Asunto(s)
Membrana Celular/metabolismo , Inmunidad Innata/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/inmunología , Receptores Toll-Like/metabolismo , Alarminas/metabolismo , Caspasa 1/inmunología , Inhibidores Enzimáticos/farmacología , Humanos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Proteínas de Unión a Fosfato/inmunología , Transducción de Señal/inmunología
7.
Front Immunol ; 12: 761345, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34950139

RESUMEN

Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to infection that often results in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). An emerging mechanism of sepsis-induced ARDS involves neutrophils/macrophages undergoing cell death, releasing nuclear histones to cause tissue damage that exacerbates pulmonary injury. While published studies focus on unmodified histones, little is known about the role of citrullinated histone H3 (CitH3) in the pathogenesis of sepsis and ALI. In this study, we found that levels of CitH3 were elevated in the patients with sepsis-induced ARDS and correlated to PaO2/FiO2 in septic patients. Systematic administration of CitH3 peptide in mice provoked Caspase-1 activation in the lung tissue and caused ALI. Neutralization of CitH3 with monoclonal antibody improved survival and attenuated ALI in a mouse sepsis model. Furthermore, we demonstrated that CitH3 induces ALI through activating Caspase-1 dependent inflammasome in bone marrow derived macrophages and bone marrow derived dendritic cells. Our study suggests that CitH3 is an important mediator of inflammation and mortality during sepsis-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Histonas/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Sepsis/inmunología , Lesión Pulmonar Aguda/etiología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Caspasa 1/inmunología , Células Cultivadas , Citrulinación , Células Dendríticas/inmunología , Humanos , Inflamasomas/inmunología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Péptidos/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Síndrome de Dificultad Respiratoria/etiología , Sepsis/complicaciones
8.
Nat Commun ; 12(1): 6699, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795266

RESUMEN

Candida albicans is the most common cause of fungal sepsis. Inhibition of inflammasome activity confers resistance to polymicrobial and LPS-induced sepsis; however, inflammasome signaling appears to protect against C. albicans infection, so inflammasome inhibitors are not clinically useful for candidiasis. Here we show disruption of GSDMD, a known inflammasome target and key pyroptotic cell death mediator, paradoxically alleviates candidiasis, improving outcomes and survival of Candida-infected mice. Mechanistically, C. albicans hijacked the canonical inflammasome-GSDMD axis-mediated pyroptosis to promote their escape from macrophages, deploying hyphae and candidalysin, a pore-forming toxin expressed by hyphae. GSDMD inhibition alleviated candidiasis by preventing C. albicans escape from macrophages while maintaining inflammasome-dependent but GSDMD-independent IL-1ß production for anti-fungal host defenses. This study demonstrates key functions for GSDMD in Candida's escape from host immunity in vitro and in vivo and suggests that GSDMD may be a potential therapeutic target in C. albicans-induced sepsis.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Inflamasomas/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Macrófagos/inmunología , Proteínas de Unión a Fosfato/inmunología , Animales , Candida albicans/fisiología , Candidiasis/genética , Candidiasis/microbiología , Caspasa 1/genética , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Femenino , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Estimación de Kaplan-Meier , Riñón/inmunología , Riñón/metabolismo , Riñón/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo
9.
Aging (Albany NY) ; 13(16): 20534-20551, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34432650

RESUMEN

OBJECTIVE: The NOD-like receptor protein 3 (NOD-like receptor protein 3, NLRP3) inflammasome is associated with many physiological processes related to aging. We investigated whether NLRP3 inflammasome activation contributes to the pathogenesis of cardiocytes aging dissected the underlying mechanism. METHODS: H9c2 cells were treated with different concentrations of D-galactose (D-gal, 0, 2, 10 and 50 g/L) for 24 hours. The cytochemical staining, flow cytometry and fluorescence microscope analysis were employed to detect the ß-galactosidase (ß-gal) activity. Western blot analysis was used to detect the age-associated proteins (P53, P21) and NLRP3 inflammasome proteins [NLRP3, apoptosis-associated speck-like protein (ASC)]. Confocal fluorescent images were applied to capture the colocalization of NLRP3 and caspase-1. Intracellular reactive oxygen species (ROS) was measured using 2'7'-dichlorodihydrofluorescein diacetate (DCFH-DA) by flow cytometry and visualized using a fluorescence microscope. The IL-1ß, IL-18 and lactate dehydrogenase (LDH) release were also detected. RESULTS: D-gal induced-H9c2 cells caused cardiocytes' aging changes (ß-gal staining, CellEvent™ Senescence Green staining, P53, P21) in a concentration-dependent manner. NLRP3 inflammasomes were activated, IL-1ß, IL-18 and LDH release and ROS generation were increased in the cardiocytes aging progress. When MCC950 inhibited NLRP3 inflammasomes, it attenuated the cardiocytes aging, yet the ROS generation was similar. Inhibition of ROS by NAC attenuated cardiocytes aging and inhibited the NLRP3 inflammasome activation at the same time. NLRP3 inflammasome activation by nigericin-induced cardiocytes cells aging progress. CONCLUSIONS: NLRP3 inflammasome activation contributes to the pathogenesis of cardiocytes aging, and ROS generation may serve as a potential mechanism by which NLRP3 inflammasome is activated.


Asunto(s)
Envejecimiento/inmunología , Inflamasomas/inmunología , Miocitos Cardíacos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Envejecimiento/genética , Animales , Caspasa 1/genética , Caspasa 1/inmunología , Senescencia Celular , Humanos , Inflamasomas/genética , Miocitos Cardíacos/citología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Ratas , Especies Reactivas de Oxígeno/inmunología
10.
Exp Cell Res ; 406(1): 112738, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34270981

RESUMEN

We determined the role of time in adipose-derived stem/stromal cell (ASC) response to a model inflammatory environment. ASCs and other mesenchymal stem/stromal cells exhibit immune plasticity. We evaluated the persistence of pro- and anti-inflammatory phenotypes for ASCs exposed to a sustained or pulse inflammatory stimulus. Using qPCR, flow cytometry, and immunocytochemistry, we monitored the temporal expression and up-regulation patterns of a pro-inflammatory gene (caspase 1), a pleiotropic gene/protein (interleukin 6, IL-6), and an anti-inflammatory gene/protein (indoleamine 2, 3-dioxygenase, IDO1) after exposing ASCs to the cytokines tumor necrosis factor-α and interferon-γ. In response to sustained cytokine stimulation, we discovered that time played a role in the balance of pro- and anti-inflammatory ASC phenotypes. IL-6 was present at all time points for both cytokine-stimulated and non-stimulated conditions, whereas IDO1 was heterogeneously up-regulated in stimulated conditions at later time points. After a pulse stimulus, ASC immunoresponse remained consistent for 96-168 h. As a final measure of immune plasticity, we cultured cytokine-stimulated ASCs with blood-derived macrophages to observe macrophage polarization. While the presence of ASCs altered macrophage phenotype, there was no dependency on the length of ASC cytokine exposure time.


Asunto(s)
Caspasa 1/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interferón gamma/farmacología , Interleucina-6/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Tejido Adiposo/citología , Tejido Adiposo/inmunología , Caspasa 1/inmunología , Diferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , Regulación de la Expresión Génica , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interleucina-6/inmunología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Cultivo Primario de Células , Transducción de Señal , Factores de Tiempo
11.
EMBO J ; 40(18): e108249, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34296442

RESUMEN

SARS-CoV-2 is an emerging coronavirus that causes dysfunctions in multiple human cells and tissues. Studies have looked at the entry of SARS-CoV-2 into host cells mediated by the viral spike protein and human receptor ACE2. However, less is known about the cellular immune responses triggered by SARS-CoV-2 viral proteins. Here, we show that the nucleocapsid of SARS-CoV-2 inhibits host pyroptosis by blocking Gasdermin D (GSDMD) cleavage. SARS-CoV-2-infected monocytes show enhanced cellular interleukin-1ß (IL-1ß) expression, but reduced IL-1ß secretion. While SARS-CoV-2 infection promotes activation of the NLRP3 inflammasome and caspase-1, GSDMD cleavage and pyroptosis are inhibited in infected human monocytes. SARS-CoV-2 nucleocapsid protein associates with GSDMD in cells and inhibits GSDMD cleavage in vitro and in vivo. The nucleocapsid binds the GSDMD linker region and hinders GSDMD processing by caspase-1. These insights into how SARS-CoV-2 antagonizes cellular inflammatory responses may open new avenues for treating COVID-19 in the future.


Asunto(s)
COVID-19/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Nucleocápside/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Piroptosis/fisiología , SARS-CoV-2/metabolismo , Enzima Convertidora de Angiotensina 2/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ratones , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células THP-1
12.
Nat Commun ; 12(1): 2713, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976225

RESUMEN

Interleukin-1ß (IL-1ß) is activated by inflammasome-associated caspase-1 in rare autoinflammatory conditions and in a variety of other inflammatory diseases. Therefore, IL-1ß activity must be fine-tuned to enable anti-microbial responses whilst limiting collateral damage. Here, we show that precursor IL-1ß is rapidly turned over by the proteasome and this correlates with its decoration by K11-linked, K63-linked and K48-linked ubiquitin chains. The ubiquitylation of IL-1ß is not just a degradation signal triggered by inflammasome priming and activating stimuli, but also limits IL-1ß cleavage by caspase-1. IL-1ß K133 is modified by ubiquitin and forms a salt bridge with IL-1ß D129. Loss of IL-1ß K133 ubiquitylation, or disruption of the K133:D129 electrostatic interaction, stabilizes IL-1ß. Accordingly, Il1bK133R/K133R mice have increased levels of precursor IL-1ß upon inflammasome priming and increased production of bioactive IL-1ß, both in vitro and in response to LPS injection. These findings identify mechanisms that can limit IL-1ß activity and safeguard against damaging inflammation.


Asunto(s)
Caspasa 1/genética , Inflamasomas/genética , Interleucina-1beta/genética , Complejo de la Endopetidasa Proteasomal/genética , Procesamiento Proteico-Postraduccional , Animales , Caspasa 1/inmunología , Células HEK293 , Humanos , Inflamasomas/inmunología , Inflamación , Interleucina-1beta/inmunología , Lipopolisacáridos/administración & dosificación , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Cultivo Primario de Células , Complejo de la Endopetidasa Proteasomal/inmunología , Proteolisis , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Ubiquitina/genética , Ubiquitina/inmunología , Ubiquitinación
13.
Arch Insect Biochem Physiol ; 107(3): e21793, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33949719

RESUMEN

Apoptosis, as one kind of innate immune system, is involved in host response against pathogens innovation. Caspases play a vital role in the execution stage of host cell apoptosis. It has been reported that Bmcaspase-1 (Bmcas-1) has a close relationship with Bombyx mori nucleopolyhedrovirus (BmNPV) infection for its differentially expressed patterns after viral infection. However, its underlying response mechanism is still unclear. The significant differential expression of Bmcas-1 in different tissues of differentially resistant strains revealed its vital role in BmNPV infection. To further validate its role in BmNPV infection, budded virus (BV)-eGFP was analyzed after knockdown and overexpression of Bmcas-1 by small interfering RNA and the pIZT-mCherry vector, respectively. The reproduction of BV-eGFP obviously increased at 72 h after knockdown of Bmcas-1, and decreased after overexpression in BmN cells. Moreover, the conserved functional domain of Cas-1 among different species and the closed evolutionary relationship of Cas-1 in Lepidoptera hinted that Bmcas-1 might be associated with apoptosis, and this was also validated by the apoptosis inducer, Silvestrol, and the inhibitor, Z-DEVD-FMK. Therefore, Bmcas-1 plays an essential antiviral role by activating apoptosis, and this result lays a fundament for clarifying the molecular mechanism of silkworm in response against BmNPV infection and breeding of resistant strains.


Asunto(s)
Apoptosis , Bombyx/virología , Caspasa 1/metabolismo , Interacciones Huésped-Patógeno/inmunología , Nucleopoliedrovirus/inmunología , Animales , Bombyx/enzimología , Bombyx/inmunología , Caspasa 1/inmunología , Proteínas Fluorescentes Verdes
14.
Cell Rep ; 35(2): 108998, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852854

RESUMEN

Cellular inflammasome activation causes caspase-1 cleavage of the pore-forming protein gasdermin D (GSDMD) with subsequent pyroptotic cell death and cytokine release. Here, we clarify the ambiguous role of the related family member gasdermin E (GSDME) in this process. Inflammasome stimulation in GSDMD-deficient cells led to apoptotic caspase cleavage of GSDME. Endogenous GSDME activation permitted sublytic, continuous interleukin-1ß (IL-1ß) release and membrane leakage, even in GSDMD-sufficient cells, whereas ectopic expression led to pyroptosis with GSDME oligomerization and complete liberation of IL-1ß akin to GSDMD pyroptosis. We find that NLRP3 and NLRP1 inflammasomes ultimately rely concurrently on both gasdermins for IL-1ß processing and release separately from their ability to induce cell lysis. Our study thus identifies GSDME as a conduit for IL-1ß release independent of its ability to cause cell death.


Asunto(s)
Inflamasomas/genética , Interleucina-1beta/genética , Macrófagos/inmunología , Proteínas de Unión a Fosfato/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Piroptosis/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Caspasa 3/genética , Caspasa 3/inmunología , Línea Celular Transformada , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteínas de Unión a Fosfato/deficiencia , Proteínas de Unión a Fosfato/inmunología , Proteínas Citotóxicas Formadoras de Poros/deficiencia , Proteínas Citotóxicas Formadoras de Poros/inmunología , Piroptosis/efectos de los fármacos , Piroptosis/inmunología , Salmonella typhimurium/química , Salmonella typhimurium/patogenicidad , Transducción de Señal , Células THP-1
15.
Int Immunopharmacol ; 95: 107529, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33744777

RESUMEN

Sepsis is a systemic inflammatory response syndrome resulted from severe infection. Excessive inflammation response plays an important role in sepsis-induced acute lung injury (ALI). Loganin is an iridoid glycoside isolated from Corni fructus and exerts an anti-inflammatory effect in multiple inflammatory diseases; however, the role of loganin in sepsis-induced ALI remains unknown. In the current study, the cecal ligation and puncture (CLP)-induced murine sepsis model was constructed to investigate the anti-inflammatory property of loganin in sepsis-induced ALI. Lipopolysaccharide (LPS)-treated Raw 264.7 cells and primary murine peritoneal macrophages were established to further explore underlying mechanism of loganin. Results showed that intragastrical administration of loganin significantly increased murine survival, reduced the alveolar structure damage and inflammatory cell infiltration. Loganin suppressed the release of the M1 macrophage-associated pro-inflammatory cytokines and induced the activation of M2-type anti-inflammatory cytokines. Besides, loganin dramatically inhibited NLRP3 inflammasome-mediated caspase-1 activation and subsequent IL-1ß secretion. Further in vitro studies confirmed that loganin efficiently inhibited M1 macrophage polarization and NLRP3 inflammasome activation by blocking the extra-cellular signal-regulated kinase (ERK) and nuclear factor-kappa B (NF-κB) pathways. Taken together, the anti-inflammatory effect of loganin in sepsis-induced ALI was associated with the ERK and NF-κB pathway-mediated macrophage polarization and NLRP3 inflammasome activation. Our study offers a favorable mechanistic basis to support the therapeutic potential of loganin in anti-inflammatory diseases, such as sepsis-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Inflamasomas/antagonistas & inhibidores , Iridoides/uso terapéutico , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Sepsis/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/farmacología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Caspasa 1/inmunología , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Iridoides/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Células RAW 264.7 , Sepsis/complicaciones , Sepsis/inmunología , Sepsis/patología
16.
Int Immunopharmacol ; 94: 107503, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33647825

RESUMEN

Our previous studies have implicated Caspase-1 signaling in driving the proinflammatory state of acute graft versus host disease (aGVHD). Therefore, we aimed to elucidate the mechanism of Caspase-1 in in murine models of aGVHD through specific inhibition of its activity with the decoy peptide Ac-YVAD-CMK. We transplanted bone marrow from donor C57BL/6 (H-2b) mice into recipient BALB/c (H-2Kd) mice and randomized the recipients into the following treatment cohorts: (1) allogeneic hematopoietic stem cell transplantation and splenic cell infusion control (PBS group); (2) low dose Ac-YVAD-CMK (AC low group); (3) and high dose Ac-YVAD-CMK (AC high group). Indeed, we observed that Caspase-1 inhibition by Ac-YVAD-CMK ameliorated pathological damage and inflammation in the liver, lungs, and colon elicited by aGVHD. This was associated with reduced mortality secondary to aGVHD. Mechanistically, we found that Caspase-1 inhibition modulated donor T cell expansion, restored the balance of Th1/Th17/Treg subsets, and markedly decreased serum levels and aGVHD target organ mRNA expression of IL-1ß, IL-18, and HMGB1. Thus, we demonstrate that inhibition of Caspase-1 by Ac-YVAD-CMK mitigates murine aGVHD by regulating Th1/Th17/Treg balance and attenuating its characteristic proinflammatory state.


Asunto(s)
Clorometilcetonas de Aminoácidos/uso terapéutico , Caspasa 1/inmunología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Clorometilcetonas de Aminoácidos/farmacología , Animales , Colon/efectos de los fármacos , Colon/inmunología , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Proteína HMGB1/sangre , Proteína HMGB1/genética , Proteína HMGB1/inmunología , Interleucina-18/sangre , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/sangre , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Hígado/efectos de los fármacos , Hígado/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología
17.
Front Immunol ; 12: 603416, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692782

RESUMEN

Diabetic kidney disease (DKD) is a major cause of chronic kidney disease (CKD) in many developed and developing countries. Pyroptosis is a recently discovered form of programmed cell death (PCD). With progress in research on DKD, researchers have become increasingly interested in elucidating the role of pyroptosis in DKD pathogenesis. This review focuses on the three pathways of pyroptosis generation: the canonical inflammasome, non-canonical inflammasome, and caspase-3-mediated inflammasome pathways. The molecular and pathophysiological mechanisms of the pyroptosis-related inflammasome pathway in the development of DKD are summarized. Activation of the diabetes-mediated pyroptosis-related inflammasomes, such as nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), Toll-like receptor 4 (TLR4), caspase-1, interleukin (IL)-1ß, and the IL-18 axis, plays an essential role in DKD lesions. By inhibiting activation of the TLR4 and NLRP3 inflammasomes, the production of caspase-1, IL-1ß, and IL-18 is inhibited, thereby improving the pathological changes associated with DKD. Studies using high-glucose-induced cell models, high-fat diet/streptozotocin-induced DKD animal models, and human biopsies will help determine the spatial and temporal expression of DKD inflammatory components. Recent studies have confirmed the relationship between the pyroptosis-related inflammasome pathway and kidney disease. However, these studies are relatively superficial at present, and the mechanism needs further elucidation. Linking these findings with disease activity and prognosis would provide new ideas for DKD research.


Asunto(s)
Nefropatías Diabéticas/inmunología , Inflamasomas/inmunología , Piroptosis/inmunología , Insuficiencia Renal Crónica/inmunología , Animales , Caspasa 1/inmunología , Citocinas/inmunología , Nefropatías Diabéticas/patología , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Insuficiencia Renal Crónica/patología , Receptor Toll-Like 4/inmunología
18.
Int Immunopharmacol ; 94: 107496, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33639565

RESUMEN

OBJECTIVES: Cathepsin B (CTSB), nod-like receptor family pyrin domain-containing 3 (NLRP3), and caspase-1 play an important role in the development of Acute Pancreatitis (AP). Besides, the relationship between the proteins remains poorly understood. In addition, whereas previous studies have found caspase-1 activation in AP, pyroptosis, a caspase-1 induced cell death mode, has never been proposed and proved in AP. METHODS: We induced AP in mice by intraperitoneal injection of cerulein. Mice in the inhibitor group of CTSB were pretreated with injection of CA-074me, while mice in the inhibitor group of caspase-1 were of Ac-YVAD-CHO, 1 h earlier. We evaluated the inflammation of the pancreas and the detected expression of activated CTSB, NLRP3, ASC, caspase-1p20, IL-1ß and IL-18. TUNEL staining was used to detect acinar cell death. RESULTS: The inflammation of the pancreas in the two inhibitor groups was significantly reduced compared with that in the AP group. We observed that CA-074me not only inhibits CTSB, but also suppresses the expression and activity of NLRP3, ASC and caspase-1. We found that CA-074me further inhibits the downstream event of caspase-1, including pro-inflammatory cytokine secretion and pyroptosis. Whereas Ac-YVAD-CHO inhibited caspase-1 and decreased pro-inflammatory cytokine secretion and pyroptosis, it did not down-regulate the expression and activity ofCTSB, NLRP3 and ASC. CONCLUSION: The results indicate that CTSB may aggravate AP by activating the NLRP3 inflammasome and promoting Caspase-1-induced pyroptosis. These provide clues about the pathophysiological mechanisms of AP, shedding light on new ideas and potential targets for the prevention and treatment of AP.


Asunto(s)
Catepsina B/inmunología , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Pancreatitis/inmunología , Animales , Caspasa 1/inmunología , Ceruletida , Interleucina-1beta/sangre , Masculino , Ratones Endogámicos C57BL , Páncreas/inmunología , Páncreas/patología , Pancreatitis/sangre , Pancreatitis/inducido químicamente , Pancreatitis/patología , Piroptosis
19.
Cell Commun Signal ; 19(1): 13, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33573688

RESUMEN

BACKGROUND: Epimedii Folium (EF) is commonly used for treating bone fractures and joint diseases, but the potential hepatotoxicity of EF limits its clinical application. Our previous study confirms that EF could lead to idiosyncratic drug-induced liver injury (IDILI) and hepatocyte apoptosis, but the mechanism remains unknown. Studies have shown that NLRP3 inflammasome plays an important role in the development of various inflammatory diseases such as IDILI. Specific stimulus-induced NLRP3 inflammasome activation may has been a key strategy for lead to liver injury. Therefore, main compounds derived from EF were chosen to test whether the ingredients in EF could activate the NLRP3 inflammasome and to induce IDILI. METHODS: Bone-marrow-derived macrophages (BMDMs) were treated with Icariside I, and then stimulated with inflammasome stimuli and assayed for the production of caspase-1 and interleukin 1ß (IL-1ß) and the release of lactate dehydrogenase (LDH). Determination of intracellular potassium, ASC oligomerization as well as reactive oxygen species (ROS) production were used to evaluate the stimulative mechanism of Icariside I on inflammasome activation. Mouse models of NLRP3 diseases were used to test whether Icariside I has hepatocyte apoptosis effects and promoted NLRP3 inflammasome activation in vivo. RESULTS: Icariside I specifically enhances NLRP3 inflammasome activation triggered by ATP or nigericin but not SiO2, poly(I:C) or cytosolic LPS. Additionally, Icariside I does not alter the activation of NLRC4 and AIM2 inflammasomes. Mechanically, Icariside I alone does not induce mitochondrial reactive oxygen species (mtROS), which is one of the critical upstream events of NLRP3 inflammasome activation; however, Icariside I increases mtROS production induced by ATP or nigericin but not SiO2. Importantly, Icariside I leads to liver injury and NLRP3 inflammasome activation in an LPS-mediated susceptibility mouse model of IDILI, but the effect of Icariside I is absent in the LPS-mediated mouse model pretreated with MCC950, which is used to mimic knockdown of NLRP3 inflammasome activation. CONCLUSIONS: Our study reveals that Icariside I specifically facilitates ATP or nigericin-induced NLRP3 inflammasome activation and causes idiosyncratic hepatotoxicity. The findings suggest that Icariside I or EF should be avoided in patients with diseases related to ATP or nigericin-induced NLRP3 inflammasome activation, which may be risk factors for IDILI. Video abstract.


Asunto(s)
Adenosina Trifosfato , Enfermedad Hepática Inducida por Sustancias y Drogas , Flavonas/toxicidad , Inflamasomas/inmunología , L-Lactato Deshidrogenasa/toxicidad , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Nigericina , Umbeliferonas/toxicidad , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Caspasa 1/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Femenino , Furanos/farmacología , Indenos/farmacología , Inflamasomas/antagonistas & inhibidores , Interleucina-1beta/sangre , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/farmacología , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...