Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int Immunopharmacol ; 129: 111656, 2024 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-38340422

RESUMEN

Geraniin, a chemical component of the traditional Chinese medicine geranii herba, possesses anti-inflammatory and anti-oxidative activities. However, its anti-inflammatory role in managing NLRP3 inflammasome and pyroptosis remains to be elucidated. To investigate the anti-inflammation mechanism of geraniin, LPS-primed macrophages were incubated with classical activators of NLRP3 inflammasome (such as ATP, Nigericin, or MSU crystals), and MSU crystals were injected into the ankle joints of mice to establish an acute gouty arthritis model. The propidium iodide (PI) staining results showed that geraniin could restrain cell death in the ATP- or nigericin-stimulated bone marrow-derived macrophages (BMDMs). Geraniin decreased the release of lactate dehydrogenase (LDH) and interleukin (IL)-1ß from cytoplasm to cell supernatant. Geraniin also inhibited the expression of caspase-1 p20, IL-1ß in cell supernatant and N-terminal of gasdermin D (GSDMD-NT) while blocking the oligomerization of ASC to form speck. The inhibitory effects of geraniin on caspase-1 p20, IL-1ß, GSDMD-NT, and ASC speck were not observed in NLRP3 knockout (NLRP3-/-) BMDMs. Hence, the resistance of geraniin to inflammasome and pyroptosis was contingent upon NLRP3 presence. Geraniin reduced reactive oxygen species (ROS) production and maintained mitochondrial membrane potential while preventing interaction between ASC and NLRP3 protein. Additionally, geraniin diminished MSU crystal-induced mouse ankle joint swelling and IL-1ß expression. Geraniin blocked the recruitment of neutrophils and macrophages to the synovium of joints. Our results demonstrate that geraniin prevents the assembly of ASC and NLRP3 through its antioxidant effect, thereby inhibiting inflammasome activation, pyroptosis, and IL-1ß release to provide potential insights for gouty arthritis targeted therapy.


Asunto(s)
Artritis Gotosa , Glucósidos , Taninos Hidrolizables , Inflamasomas , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Artritis Gotosa/inducido químicamente , Piroptosis , Nigericina/farmacología , Macrófagos , Antiinflamatorios/efectos adversos , Adenosina Trifosfato/metabolismo , Caspasas/metabolismo , Interleucina-1beta/metabolismo
2.
Proc Natl Acad Sci U S A ; 121(2): e2309579121, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38175865

RESUMEN

Nigericin, an ionophore derived from Streptomyces hygroscopicus, is arguably the most commonly used tool compound to study the NLRP3 inflammasome. Recent findings, however, showed that nigericin also activates the NLRP1 inflammasome in human keratinocytes. In this study, we resolve the mechanistic basis of nigericin-driven NLRP1 inflammasome activation. In multiple nonhematopoietic cell types, nigericin rapidly and specifically inhibits the elongation stage of the ribosome cycle by depleting cytosolic potassium ions. This activates the ribotoxic stress response (RSR) sensor kinase ZAKα, p38, and JNK, as well as the hyperphosphorylation of the NLRP1 linker domain. As a result, nigericin-induced pyroptosis in human keratinocytes is blocked by extracellular potassium supplementation, ZAKα knockout, or pharmacologic inhibitors of ZAKα and p38 kinase activities. By surveying a panel of ionophores, we show that electroneutrality of ion movement is essential to activate ZAKα-driven RSR and a greater extent of K+ depletion is necessary to activate ZAKα-NLRP1 than NLRP3. These findings resolve the mechanism by which nigericin activates NLRP1 in nonhematopoietic cell types and demonstrate an unexpected connection between RSR, perturbations of potassium ion flux, and innate immunity.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nigericina/farmacología , Potasio/metabolismo , Inmunidad Innata , Ionóforos , Proteínas NLR
3.
Int Immunopharmacol ; 121: 110423, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37331291

RESUMEN

Eleutheroside E, a major natural bioactive compound in Acanthopanax senticosus (Rupr.etMaxim.) Harms, possesses anti-oxidative, anti-fatigue, anti-inflammatory, anti-bacterial and immunoregulatory effects. High-altitude hypobaric hypoxia affects blood flow and oxygen utilisation, resulting in severe heart injury that cannot be reversed, thereby eventually causing or exacerbating high-altitude heart disease and heart failure. The purpose of this study was to determine the cardioprotective effects of eleutheroside E against high-altitude-induced heart injury (HAHI), and to study the mechanisms by which this happens. A hypobaric hypoxia chamber was used in the study to simulate hypobaric hypoxia at the high altitude of 6000 m. 42 male rats were randomly assigned to 6 equal groups and pre-treated with saline, eleutheroside E 100 mg/kg, eleutheroside E 50 mg/kg, or nigericin 4 mg/kg. Eleutheroside E exhibited significant dose-dependent effects on a rat model of HAHI by suppressing inflammation and pyroptosis. Eleutheroside E downregulated the expressions of brain natriuretic peptide (BNP), creatine kinase isoenzymes (CK-MB) and lactic dehydrogenase (LDH). Moreover, The ECG also showed eleutheroside E improved the changes in QT interval, corrected QT interval, QRS interval and heart rate. Eleutheroside E remarkably suppressed the expressions of NLRP3/caspase-1-related proteins and pro-inflammatory factors in heart tissue of the model rats. Nigericin, known as an agonist of NLRP3 inflammasome-mediated pyroptosis, reversed the effects of eleutheroside E. Eleutheroside E prevented HAHI and inhibited inflammation and pyroptosis via the NLRP3/caspase-1 signalling pathway. Taken together, eleutheroside E is a prospective, effective, safe and inexpensive agent that can be used to treat HAHI.


Asunto(s)
Eleutherococcus , Lesiones Cardíacas , Masculino , Ratas , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Caspasa 1/metabolismo , Altitud , Nigericina/farmacología , Estudios Prospectivos , Antiinflamatorios/farmacología , Inflamación , Hipoxia
4.
Cell Death Dis ; 13(4): 372, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440074

RESUMEN

As a widely acknowledged FDA-approved dietary supplement or over-the-counter medicines, dehydroepiandrosterone (DHEA) exerts anti-inflammatory and immunomodulatory function. Pyroptosis is an important form of programmed cell death (PCD), and which acts a key role in the body's anti-infection and inflammatory responses. But the effects and mechanisms of DHEA on pyroptosis remain unclear. Here, we found that DHEA inhibited the NLRP3 inflammasome components expression by blocking inflammatory signals in lipopolysaccharide (LPS)-primed macrophages, and prevented the bacterial toxin nigericin (Nig)-induced NLRP3 inflammasome assembly. However, DHEA exacerbated NLRP3-independent cell death in Nig-treated inflammatory macrophages. During this process, DHEA induced the abnormal autophagy, which reflected as the blocking of autophagic flux and the accumulation of autophagy receptor p62 (SQSTM1) protein. In addition, DHEA caused a burst of reactive oxygen species (ROS) and activated extracellular signal-regulated kinase (ERK) phosphorylation in LPS plus Nig-stimulated macrophages but not in LPS-treated macrophages. Mechanistically, the present study certified that the activation of G protein-coupled estrogen receptor (GPER) signal mediated the cell death induced by DHEA in Nig-stimulated inflammatory macrophages, as GPER specific inhibitor G15 alleviated the abnormal autophagy and ultimately prevented the gasdermin D (GSDMD)-mediated pyroptosis induced by DHEA. Collectively, DHEA can exacerbate Nig-induced abnormal autophagy and pyroptosis via activation of GPER in LPS-primed macrophages, which prompts us the potential application value of DHEA in anti-infection or anti-tumor immunity.


Asunto(s)
Lipopolisacáridos , Piroptosis , Autofagia , Deshidroepiandrosterona/farmacología , Inflamasomas/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nigericina/farmacología , Receptores Acoplados a Proteínas G/metabolismo
5.
Sci Rep ; 11(1): 15319, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321581

RESUMEN

Inhibition of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome has recently emerged as a promising therapeutic target for several inflammatory diseases. After priming and activation by inflammation triggers, NLRP3 forms a complex with apoptosis-associated speck-like protein containing a CARD domain (ASC) followed by formation of the active inflammasome. Identification of inhibitors of NLRP3 activation requires a well-validated primary high-throughput assay followed by the deployment of a screening cascade of assays enabling studies of structure-activity relationship, compound selectivity and efficacy in disease models. We optimized a NLRP3-dependent fluorescent tagged ASC speck formation assay in murine immortalized bone marrow-derived macrophages and utilized it to screen a compound library of 81,000 small molecules. Our high-content screening assay yielded robust assay metrics and identified a number of inhibitors of NLRP3-dependent ASC speck formation, including compounds targeting HSP90, JAK and IKK-ß. Additional assays to investigate inflammasome priming or activation, NLRP3 downstream effectors such as caspase-1, IL-1ß and pyroptosis form the basis of a screening cascade to identify NLRP3 inflammasome inhibitors in drug discovery programs.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Inflamasomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/biosíntesis , Células Cultivadas , Dimetilsulfóxido/farmacología , Descubrimiento de Drogas , Furanos/farmacología , Genes Reporteros , Indenos/farmacología , Interleucina-1beta/biosíntesis , Lipopolisacáridos/farmacología , Ratones , Nigericina/farmacología , Fenotipo , Piroptosis/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas , Sulfonamidas/farmacología
6.
Mol Neurobiol ; 58(9): 4460-4476, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34021869

RESUMEN

Neurological disorders have been demonstrated to be associated with mitochondrial dysfunction. This impairment may lead to oxidative stress and neuroinflammation, specifically promoted by NLRP3 expression. Açaí (Euterpe oleracea Mart.) has been studied in this field, since it presents important biological activities. We investigated açaí extract's anti-neuroinflammatory capacity, through NLRP3 inflammasome modulation. Microglia (EOC 13.31) were exposed to LPS and nigericin, as agents of inflammatory induction, and treated with açaí extract. Additionally, we used lithium (Li) as an anti-inflammatory control. Three different experiment models were conducted: (1) isolated NLRP3 priming and activation signals; (2) combined NLRP3 priming and activation signals followed by açaí extract as a therapeutic agent; and (3) combined NLRP3 priming and activation signals with açaí extract as a preventive agent. Cells exposed to 0.1 µg/mL of LPS presented high proliferation and increased levels of NO, and ROS, while 0.1 µg/mL of açaí extract was capable to reduce cellular proliferation and recover levels of NO and ROS. Primed and activated cells presented increased levels of NLRP3, caspase-1, and IL-1ß, while açaí, Li, and orientin treatments reversed this impairment. We found that açaí, Li, and orientin were effective prophylactic treatments. Preventative treatment with Li and orientin was unable to avoid overexpression of IL-1ß compared to the positive control. However, orientin downregulated NLRP3 and caspase-1. Lastly, primed and activated cells impaired ATP production, which was prevented by pre-treatment with açaí, Li, and orientin. In conclusion, we suggest that açaí could be a potential agent to treat or prevent neuropsychiatric diseases related to neuroinflammation.


Asunto(s)
Antiinflamatorios/farmacología , Euterpe , Microglía/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Caspasa 1/metabolismo , Proliferación Celular/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Nigericina/farmacología , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Cell Rep ; 34(10): 108826, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33691121

RESUMEN

A major pathway for proinflammatory protein release by macrophages is inflammasome-mediated pyroptotic cell death. As conventional secretion, unconventional secretion, and cell death are executed simultaneously, however, the cellular mechanisms regulating this complex paracrine program remain incompletely understood. Here, we devise a quantitative proteomics strategy to define the cellular exit route for each protein by pharmacological and genetic dissection of cellular checkpoints regulating protein release. We report the release of hundreds of proteins during pyroptosis, predominantly due to cell lysis. They comprise constitutively expressed and transcriptionally induced proteins derived from the cytoplasm and specific intracellular organelles. Many low-molecular-weight proteins including the cytokine interleukin-1ß, alarmins, and lysosomal-cargo proteins exit cells in the absence of cell lysis. Cytokines and alarmins are released in an endoplasmic reticulum (ER)-Golgi-dependent manner as free proteins rather than by extracellular vesicles. Our work provides an experimental framework for the dissection of cellular exit pathways and a resource for pyroptotic protein release.


Asunto(s)
Alarminas/análisis , Citocinas/análisis , Proteómica/métodos , Piroptosis , Adenosina Trifosfato/farmacología , Alarminas/metabolismo , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión , Citocinas/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Humanos , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nigericina/farmacología , Espectrometría de Masas en Tándem
8.
Front Immunol ; 12: 632606, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679781

RESUMEN

Taraxasterol (TAS) is an active ingredient of Dandelion (Taraxacum mongolicum Hand. -Mazz.), a medicinal plant that has long been used in China for treatment of inflammatory disorders. But the underlying mechanism for its therapeutic effects on inflammatory disorders is not completely clear. Inflammasome activation is a critical step of innate immune response to infection and aseptic inflammation. Among the various types of inflammasome sensors that has been reported, NLR family pyrin domain containing 3 (NLRP3) is implicated in various inflammatory diseases and therefore has been most extensively studied. In this study, we aimed to explore whether TAS could influence NLPR3 inflammasome activation in macrophages. The results showed that TAS dose-dependently suppressed the activation of caspase-1 in lipopolysaccharide (LPS)-primed murine primary macrophages upon nigericin treatment, resulting in reduced mature interleukin-1ß (IL-1ß) release and gasdermin D (GSDMD) cleavage. TAS greatly reduced ASC speck formation upon the stimulation of nigericin or extracellular ATP. Consistent with reduced cleavage of GSDMD, nigericin-induced pyroptosis was alleviated by TAS. Interestingly, TAS time-dependently suppressed the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) and mTORC2 signaling induced by LPS priming. Like TAS, both INK-128 (inhibiting both mTORC1 and mTORC2) and rapamycin (inhibiting mTORC1 only) also inhibited NLRP3 inflammasome activation, though their effects on mTOR signaling were different. Moreover, TAS treatment alleviated mitochondrial damage by nigericin and improved mouse survival from bacterial infection, accompanied by reduced IL-1ß levels in vivo. Collectively, by inhibiting the NLRP3 inflammasome activation, TAS displayed anti-inflammatory effects likely through regulation of the mTOR signaling in macrophages, highlighting a potential action mechanism for the anti-inflammatory activity of Dandelion in treating inflammation-related disorders, which warrants further clinical investigation.


Asunto(s)
Inflamasomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Esteroles/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Triterpenos/farmacología , Animales , Antiinflamatorios/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Proteínas Adaptadoras de Señalización CARD/metabolismo , Inflamasomas/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Nigericina/farmacología , Esteroles/uso terapéutico , Análisis de Supervivencia , Triterpenos/uso terapéutico
9.
Cell Mol Life Sci ; 78(6): 3073-3085, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33231721

RESUMEN

Inflammasomes are large immune multiprotein complexes that tightly regulate the production of the pro-inflammatory cytokines, being dependent on cell regulatory volume mechanisms. Aquaporins (AQPs) are protein channels that facilitate the transport of water and glycerol (aquaglyceroporins) through membranes, essential for cell volume regulation. Although these membrane proteins are highly expressed in monocytes and macrophages, their role in the inflammatory process is still unclear. Here, we investigated the role of aquaglyceroporin AQP3 in NLRP3-inflammasome activation by complementary approaches based either on shRNA silencing or on AQP3 selective inhibition. The latter has been achieved using a reported potent gold-based inhibitor, Auphen. AQP3 inhibition or silencing partially blocked LPS-priming and decreased production of IL-6, proIL-1ß, and TNF-α, suggesting the possible involvement of AQP3 in macrophage priming by Toll-like receptor 4 engagement. Moreover, AQP3-dependent cell reswelling increased IL-1ß release through caspase-1 activation. NLRP3-inflammasome activation induced by reswelling, nigericin, and ATP was also blocked when AQP3 was inhibited or silenced. Altogether, these data point towards AQPs as potential players in the setting of the inflammatory response.


Asunto(s)
Acuaporina 3/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Acuaporina 3/antagonistas & inhibidores , Acuaporina 3/genética , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular , Citocinas/metabolismo , Glicerol/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Nigericina/farmacología , Compuestos Orgánicos de Oro/química , Compuestos Orgánicos de Oro/metabolismo , Potasio/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
FEBS Open Bio ; 10(8): 1482-1491, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32428336

RESUMEN

Sophora flavescens is used as a traditional herbal medicine to modulate inflammatory responses. However, little is known about the impact of (-)-maackiain, a compound derived from S. flavescens, on the activation of inflammasome/caspase-1, a key factor in interleukin-1ß (IL-1ß) processing. Here, we report that (-)-maackiain potently amplified caspase-1 cleavage in macrophages in response to nigericin (Nig). In macrophages primed with either lipopolysaccharide or monophosphoryl lipid A, Nig-mediated caspase-1 cleavage was also markedly promoted by (-)-maackiain. Notably, (-)-maackiain induced the production of vimentin, an essential mediator for the activation of the NOD-, LRR-, and pyrin domain-containing protein 3 inflammasome, thereby contributing to promotion of the formation of the inflammasome complex to activate caspase-1. Taken together, our data suggest that (-)-maackiain exerts an immunostimulatory effect by promoting IL-1ß production via activation of the inflammasome/caspase-1 pathway. Thus, the potent inflammasome-activating effect of (-)-maackiain may be clinically useful as an acute immune-stimulating agent.


Asunto(s)
Inflamasomas/efectos de los fármacos , Interleucina-1beta/biosíntesis , Extractos Vegetales/farmacología , Pterocarpanos/farmacología , Sophora/química , Animales , Células Cultivadas , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Nigericina/farmacología , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Pterocarpanos/química , Pterocarpanos/aislamiento & purificación
11.
Cell Immunol ; 349: 104046, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32057354

RESUMEN

Uncontrolled activation of NLRP3 inflammasome initiates a series of human inflammatory diseases. Targeting NLRP3 inflammasome has attracted considerable attention in developing potential therapeutic interventions. Here, we reported that dehydrocostus lactone (DCL), a main component of Saussurea lappa from the traditional Chinese medicine, inhibited NLRP3 inflammasome-mediated caspase-1 activation and subsequent interleukin (IL)-1ß production in primary mouse macrophages and human peripheral blood mononuclear cells and exerted an inhibitory effect on NLRP3-driven inflammation. Mechanistically, DCL significantly blocked the ASC oligomerization, which is essential for the assembly of activated inflammasome. Importantly, in vivo experiments showed that DCL reduced IL-1ß secretion and peritoneal neutrophils recruitment in LPS-mediated inflammation mouse model, which is demonstrated to be NLRP3 dependent. These results suggest that DCL is a potent pharmacological inhibitor of NLRP3 inflammasome and may be developed as a therapeutic drug for treating NLRP3-associated diseases.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Inflamasomas/efectos de los fármacos , Inflamación/prevención & control , Lactonas/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Sesquiterpenos/farmacología , Adulto , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/fisiología , Caspasa 1/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/fisiología , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Interleucina-1beta/biosíntesis , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Nigericina/farmacología , Poli I-C/farmacología , Polimerizacion/efectos de los fármacos , Organismos Libres de Patógenos Específicos , Ácido Úrico/farmacología
12.
Front Immunol ; 10: 2130, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31616405

RESUMEN

Occupational exposure to respirable crystalline silica (cSiO2) has been etiologically linked to human autoimmunity. Intranasal instillation with cSiO2 triggers profuse inflammation in the lung and onset of autoimmunity in lupus-prone mice; however, dietary supplementation with the omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) abrogates these responses. Inflammasome activation, IL-1 cytokine release, and death in alveolar macrophages following cSiO2 exposure are early and critical events that likely contribute to triggering premature autoimmune pathogenesis by this particle. Here we tested the hypothesis that DHA suppresses cSiO2-induced NLRP3 inflammasome activation, IL-1 cytokine release, and cell death in the macrophage. The model used was the murine macrophage RAW 264.7 cell line stably transfected with the inflammasome adapter protein ASC (RAW-ASC). Following priming with LPS, both the canonical activator nigericin and cSiO2 elicited robust inflammasome activation in RAW-ASC cells, as reflected by IL-1ß release and caspase-1 activation. These responses were greatly diminished or absent in wild-type RAW cells. In contrast to IL-1ß, cSiO2 induced IL-1α release in both RAW-ASC and to a lesser extent in RAW-WT cells after LPS priming. cSiO2-driven effects in RAW-ASC cells were confirmed in bone-marrow derived macrophages. Pre-incubating RAW-ASC cells with 10 and 25 µM DHA for 24 h enriched this fatty acid in the phospholipids by 15- and 25-fold, respectively, at the expense of oleic acid. DHA pre-incubation suppressed inflammasome activation and release of IL-1ß and IL-1α by nigericin, cSiO2, and two other crystals - monosodium urate and alum. DHA's suppressive effects were linked to inhibition of LPS-induced Nlrp3, Il1b, and Il1a transcription, potentially through the activation of PPARγ. Finally, nigericin-induced death was inflammasome-dependent, indicative of pyroptosis, and could be inhibited by DHA pretreatment. In contrast, cSiO2-induced death was inflammasome-independent and not inhibited by DHA. Taken together, these findings indicate that DHA suppresses cSiO2-induced inflammasome activation and IL-1 cytokine release in macrophages by acting at the level of priming, but was not protective against cSiO2-induced cell death.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Inflamasomas/efectos de los fármacos , Interleucina-1/metabolismo , Dióxido de Silicio/farmacología , Animales , Línea Celular , Células HEK293 , Humanos , Inflamasomas/metabolismo , Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nigericina/farmacología , Células RAW 264.7
13.
Molecules ; 24(9)2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-31052362

RESUMEN

Four new sesquiterpenoids (1-4) and six known sesquiterpenoids (5-10), were isolated from the EtOAc phase of the ethanolic extract of Ainsliaea yunnanensis. Their structures were established by spectroscopic methods, including 1-D, 2-D NMR and HPLC-MS. All compounds were tested for their anti-inflammatory effect by the inhibition of the activity of NLRP3 inflammasome by blocking the self-slicing of pro-caspase-1, which is induced by nigericin, then the secretion of mature IL-1ß, mediated by caspase-1, was suppressed. Unfortunately none of the compounds showed an anti-inflammatory effect.


Asunto(s)
Antiinflamatorios/química , Asteraceae/química , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sesquiterpenos/química , Antiinflamatorios/farmacología , Caspasa 1/metabolismo , Línea Celular , Cromatografía Líquida de Alta Presión , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/metabolismo , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Estructura Molecular , Nigericina/farmacología , Extractos Vegetales/química , Extractos Vegetales/farmacología , Sesquiterpenos/farmacología
14.
Cell Death Dis ; 10(3): 190, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30804327

RESUMEN

Fas-associated death domain (FADD) is a key adaptor molecule involved in numerous physiological processes including cell death, proliferation, innate immunity and inflammation. Therefore, changes in FADD expression have dramatic cellular consequences. In mice and humans, FADD regulation can occur through protein secretion. However, the molecular mechanisms accounting for human FADD secretion were still unknown. Here we report that canonical, non-canonical, but not alternative, NLRP3 inflammasome activation in human monocytes/macrophages induced FADD secretion. NLRP3 inflammasome activation by the bacterial toxin nigericin led to the proinflammatory interleukin-1ß (IL-1ß) release and to the induction of cell death by pyroptosis. However, we showed that FADD secretion could occur in absence of increased IL-1ß release and pyroptosis and, reciprocally, that IL-1ß release and pyroptosis could occur in absence of FADD secretion. Especially, FADD, but not IL-1ß, secretion following NLRP3 inflammasome activation required extracellular glucose. Thus, FADD secretion was an active process distinct from unspecific release of proteins during pyroptosis. This FADD secretion process required K+ efflux, NLRP3 sensor, ASC adaptor and CASPASE-1 molecule. Moreover, we identified FADD as a leaderless protein unconventionally secreted through microvesicle shedding, but not exosome release. Finally, we established human soluble FADD as a new marker of joint inflammation in gout and rheumatoid arthritis, two rheumatic diseases involving the NLRP3 inflammasome. Whether soluble FADD could be an actor in these diseases remains to be determined. Nevertheless, our results advance our understanding of the mechanisms contributing to the regulation of the FADD protein expression in human cells.


Asunto(s)
Artritis Reumatoide/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Gota/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/efectos de los fármacos , Animales , Caspasa 1/metabolismo , Línea Celular Tumoral , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/ultraestructura , Proteína de Dominio de Muerte Asociada a Fas/genética , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nigericina/farmacología , Canales de Potasio/metabolismo
15.
Int J Mol Sci ; 19(3)2018 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-29495403

RESUMEN

Sei-hai-to (TJ-90, Qing Fei Tang), a Chinese traditional medicine, increases ciliary beat frequency (CBF) and ciliary bend angle (CBA) mediated via cAMP (3',5'-cyclic adenosine monophosphate) accumulation modulated by Ca2+-activated phosphodiesterase 1 (PDE1A). A high concentration of TJ-90 (≥40 µg/mL) induced two types of CBF increases, a transient increase (an initial increase, followed by a decrease) and a sustained increase without any decline, while it only sustained the CBA increase. Upon inhibiting increases in intracellular Ca2+ concentration ([Ca2+]i) by 10 µM BAPTA-AM (Ca2+-chelator, 1,2-Bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester) or Ca2+/calmodulin-dependent PDE1 by 8MmIBMX (a selective PDE1 inhibitor), TJ-90 (400 µg/mL) induced only the sustained CBF increase without any transient CBF increase. The two types of the CBF increase (the transient increase and the sustained increase) induced by TJ-90 (≥40 µg/mL) were mimicked by the stimulation with both procaterol (100 pM) and ionomycin (500 nM). Thus, TJ-90 stimulates small increases in the intracellular cAMP concentration ([cAMP]i) and [Ca2+]i in airway ciliary cells of mice. These small increases in [cAMP]i and [Ca2+]i cause inducing a transient CBF increase or a sustained CBF increase in an airway ciliary cells, depending on the dominant signal, Ca2+-signal, or cAMP-signal.


Asunto(s)
Calcio/metabolismo , Cilios/efectos de los fármacos , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Medicamentos Herbarios Chinos/farmacología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Femenino , Ratones , Nigericina/análogos & derivados , Nigericina/farmacología , Procaterol/farmacología
16.
Microbiol Immunol ; 62(1): 44-54, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29193282

RESUMEN

The inflammasome is a multiprotein signaling complex that mediates inflammatory innate immune responses through caspase 1 activation and subsequent IL-1ß secretion. However, because its aberrant activation often leads to inflammatory diseases, targeting the inflammasome holds promise for the treatment of inflammation-related diseases. In this study, it was found that a hot-water extract of Sanguisorba officinalis (HSO) suppresses inflammasome activation triggered by adenosine 5'-triphosphate, nigericin, microbial pathogens, and double stranded DNA in bone marrow-derived macrophages. HSO was found to significantly suppress IL-1ß production in a dose-dependent manner; this effect correlated well with small amounts of caspase 1 and little ASC pyroptosome formation in HSO-treated cells. The anti-inflammatory activity of HSO was further confirmed in a mouse model of endotoxin-induced septic shock. Oral administration of HSO reduced IL-1ß titers in the serum and peritoneal cavity, increasing the survival rate. Taken together, our results suggest that HSO is an inhibits inflammasome activation through nucleotide-binding domain and leucine-rich repeat pyrin domain 3, nucleotide-binding domain and leucine-rich repeat caspase recruitment domain 4 and absent in melanoma 2 pathways, and may be useful for treatment of inflammasome-mediated diseases.


Asunto(s)
Inflamasomas/efectos de los fármacos , Extractos Vegetales/antagonistas & inhibidores , Sanguisorba/química , Choque Séptico/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Caspasa 1/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Endotoxinas/efectos adversos , Femenino , Medicina de Hierbas , Inflamación , Interleucina-1beta/sangre , Interleucina-1beta/metabolismo , Lipopolisacáridos/efectos adversos , Listeria monocytogenes/patogenicidad , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas NLR/metabolismo , Nigericina/farmacología , Extractos Vegetales/administración & dosificación , Raíces de Plantas/química , República de Corea , Salmonella typhimurium/patogenicidad , Choque Séptico/microbiología , Tasa de Supervivencia
17.
PLoS One ; 11(7): e0159826, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27467658

RESUMEN

Shikonin is a highly lipophilic naphtoquinone found in the roots of Lithospermum erythrorhizon used for its pleiotropic effects in traditional Chinese medicine. Based on its reported antipyretic and anti-inflammatory properties, we investigated whether shikonin suppresses the activation of NLRP3 inflammasome. Inflammasomes are cytosolic protein complexes that serve as scaffolds for recruitment and activation of caspase-1, which, in turn, results in cleavage and secretion of proinflammatory cytokines IL-1ß and IL-18. NLRP3 inflammasome activation involves two steps: priming, i.e. the activation of NF-κB pathway, and inflammasome assembly. While shikonin has previously been reported to suppress the priming step, we demonstrated that shikonin also inhibits the second step of inflammasome activation induced by soluble and particulate NLRP3 instigators in primed immortalized murine bone marrow-derived macrophages. Shikonin decreased NLRP3 inflammasome activation in response to nigericin more potently than acetylshikonin. Our results showed that shikonin also inhibits AIM2 inflammasome activation by double stranded DNA. Shikonin inhibited ASC speck formation and caspase-1 activation in murine macrophages and suppressed the activity of isolated caspase-1, demonstrating that it directly targets caspase-1. Complexing shikonin with ß-lactoglobulin reduced its toxicity while preserving the inhibitory effect on NLRP3 inflammasome activation, suggesting that shikonin with improved bioavailability might be interesting for therapeutic applications in inflammasome-mediated conditions.


Asunto(s)
Caspasa 1/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Línea Celular Transformada/metabolismo , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Naftoquinonas/farmacología , Animales , Proteínas de Unión al ADN , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Nigericina/farmacología
18.
Biochim Biophys Acta ; 1813(8): 1483-94, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21601596

RESUMEN

BACKGROUND: A novel family of intracellular Ca(2+)-release channels termed two-pore channels (TPCs) has been presented as the receptors of NAADP (nicotinic acid adenine dinucleotide phosphate), the most potent Ca(2+) mobilizing intracellular messenger. TPCs have been shown to be exclusively localized to the endolysosomal system mediating NAADP-evoked Ca(2+) release from the acidic compartments. OBJECTIVES: The present study is aimed to investigate NAADP-mediated Ca(2+) release from intracellular stores in the megakaryoblastic cell line MEG01. METHODS: Changes in cytosolic and intraluminal free Ca(2+) concentrations were registered by fluorimetry using fura-2 and fura-ff, respectively; TPC expression was detected by PCR. RESULTS: Treatment of MEG01 cells with the H(+)/K(+) ionophore nigericin or the V-type H(+)-ATPase selective inhibitor bafilomycin A1 revealed the presence of acidic Ca(2+) stores in these cells, sensitive to the SERCA inhibitor 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ). NAADP releases Ca(2+) from acidic lysosomal-like Ca(2+) stores in MEG01 cells probably mediated by the activation of TPC1 and TPC2 as demonstrated by TPC1 and TPC2 expression silencing and overexpression. Ca(2+) efflux from the acidic lysosomal-like Ca(2+) stores or the endoplasmic reticulum (ER) results in ryanodine-sensitive activation of Ca(2+)-induced Ca(2+) release (CICR) from the complementary Ca(2+) compartment. CONCLUSION: Our results show for the first time NAADP-evoked Ca(2+) release from acidic compartments through the activation of TPC1 and TPC2, and CICR, in a megakaryoblastic cell line.


Asunto(s)
Señalización del Calcio/fisiología , Células Progenitoras de Megacariocitos/metabolismo , NADP/análogos & derivados , Secuencia de Bases , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Compartimento Celular , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Concentración de Iones de Hidrógeno , Hidroquinonas/farmacología , Ionóforos/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Macrólidos/farmacología , Células Progenitoras de Megacariocitos/efectos de los fármacos , NADP/metabolismo , Nigericina/farmacología , ARN Interferente Pequeño/genética , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores
19.
J Bioenerg Biomembr ; 42(5): 387-93, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20734224

RESUMEN

Fast responding voltage sensitive dyes, RH421 and di-4-ASPBS, were used to study the electrogenic properties of plant plasma membrane proton pumps on sealed plasma membrane vesicles extracted by two-phase partitioning from Beta vulgaris and Avena sativa cv Swan root material. Fluorescence spectroscopy in the presence of the dye RH421 (10.8 nM) was sufficiently sensitive to detect electrogenic activity of the extracted plant vesicles. The dye detection system could detect inhibition of electrogenic activity of vesicles by vanadate (75 µM) and stimulation by nigericin (0.5 µM). The newly developed dye di-4-ASPBS was less sensitive to detecting the electrogenic proton pump activity. This study represents an important innovation in plant biophysics as this class of fast responding voltage sensitive dyes have never to our knowledge been used to study electrogenic proton pump activity derived from plant membranes and represents a novel approach for carrying out such studies.


Asunto(s)
Avena/enzimología , Beta vulgaris/enzimología , Membrana Celular/metabolismo , Potenciales de la Membrana/fisiología , Raíces de Plantas/enzimología , ATPasas de Translocación de Protón/metabolismo , Avena/fisiología , Beta vulgaris/fisiología , Biofisica , Cinética , Estructura Molecular , Nigericina/farmacología , Fosfatos/metabolismo , Raíces de Plantas/fisiología , Compuestos de Piridinio/química , Espectrometría de Fluorescencia , Estirenos/química , Vanadatos/farmacología , Imagen de Colorante Sensible al Voltaje
20.
Neurochem Int ; 43(1): 9-17, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12605878

RESUMEN

The pH fluctuations have been often interpreted as an insufficient regulation or as a consequence of the onset of pathological events, such as ischemia, in which a significant decrease in pH levels occurs. Neurotransmitter release appears to be affected by pH drop significantly. In this study, we investigated the effect of an extracellular and an intracellular acidification on tritiated dopamine release ([3H]-DA release), from superfused rat hypothalamic synaptosomes. When compared to basal release, extracellular acidification, due to a reduction in the external pH of the nominally carbonic-free superfusion media, provoked a significant increase in [3H]-DA release that showed a sensitiveness to calcium omission. Intraterminal acidification, obtained blocking the Na(+)/H(+) exchanger by 5-(N-ethyl-N-isopropyl)-amiloride (EIPA) and 5-(N,N-dimethyl)-amiloride (DMA), induced a significant increase in [3H]-DA outflow which occurred in a calcium-dependent manner (80% inhibition in absence of calcium from superfusion media). To further promote an intraterminal acidification through a H(+) inner accumulation, the proton ionophore nigericin was used. At every dose employed (10 microM), this compound induced a significant increase in [3H]-DA outflow, compared to basal release. Nigericin-evoked [3H]-DA release showed a 50% decrease when calcium was omitted from superfusion media. When BAPTA-AM, a chelator of intracellular calcium, was added, nigericin-evoked [3H]-DA was completely abolished. These data indicate that [3H]-DA release can be induced by extracellular acidification due to a lowering of external pH and by an intraterminal acidification due to an internal proton accumulation. The mechanism that can trigger this exocytotic process appears to depend on calcium presence, and in particular, on an increased intraterminal calcium availability.


Asunto(s)
Amilorida/análogos & derivados , Calcio/fisiología , Dopamina/metabolismo , Hipotálamo/metabolismo , Sinaptosomas/metabolismo , Amilorida/farmacología , Animales , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Concentración de Iones de Hidrógeno , Hipotálamo/efectos de los fármacos , Técnicas In Vitro , Ionóforos/farmacología , Masculino , Fármacos Neuroprotectores/farmacología , Nigericina/farmacología , Cloruro de Potasio/farmacología , Protones , Ratas , Ratas Wistar , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Sinaptosomas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA