Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
1.
Biomed Pharmacother ; 174: 116548, 2024 May.
Article En | MEDLINE | ID: mdl-38599064

BACKGROUND: Various heart diseases ultimately lead to chronic heart failure (CHF). In CHF, the inflammatory response is associated with pyroptosis, which is mediated by the NOD-like receptor protein 3 (NLRP3) inflammasome. Fu Xin decoction (FXD) is commonly used in clinical practice to treat CHF and improve inflammatory conditions. However, the specific pharmacological mechanisms of action for FXD in these processes have yet to be fully understood. PURPOSE: The objective of this study was to examine the protective mechanism of FXT against CHF, both in H9c2 cells and mice. METHOD: A CHF mouse model was established, and the effect of FXD was observed via gavage. Cardiac function was evaluated using echocardiography, while serum BNP and LDH levels were analyzed to assess the severity of CHF. Hematoxylin and eosin staining (H&E) and Masson staining were performed to evaluate myocardial pathological changes, and TdT-mediated dUTP Nick-End Labeling staining was used to detect DNA damage. Additionally, doxorubicin was utilized to induce myocardial cell injury in H9c2 cells, establishing a relevant model. CCK8 was used to observe cell viability and detect LDH levels in the cell supernatant. Subsequently, the expression of pyroptosis-related proteins was detected using immunohistochemistry, immunofluorescence, and western blotting. Finally, the pharmacological mechanism of FXD against CHF was further validated by treating H9c2 cells with an NLRP3 activator and inducing NLRP3 overexpression. RESULT: According to current research findings, echocardiography demonstrated a significant improvement of cardiac function by FXD, accompanied by reduced levels of BNP and LDH, indicating the amelioration of cardiac injury in CHF mice. FXD exhibited the ability to diminish serum CRP and MCP inflammatory markers in CHF mice. The results of HE and Masson staining analyses revealed a significant reduction in pathological damage of the heart tissue following FXD treatment. The CCK8 assay demonstrated the ability of FXD to enhance H9c2 cell viability, improve cell morphology, decrease LDH levels in the cell supernatant, and alleviate cell damage. Immunohistochemistry, Western blotting, and immunofluorescence staining substantiated the inhibitory effect of FXD on the NLRP3/caspase-1/GSDMD pyroptosis signaling pathway in both CHF and H9c2 cell injury models. Ultimately, the administration of the NLRP3 activator (Nigericin) and the overexpression of NLRP3 counteract the effects of FXD on cardiac protection and pyroptosis inhibition in vitro. CONCLUSION: FXD exhibits a cardioprotective effect, improving CHF and alleviating pyroptosis by inhibiting the NLRP3/caspase-1/GSDMD pathway.


Drugs, Chinese Herbal , Heart Failure , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Animals , Mice , Caspase 1/drug effects , Caspase 1/metabolism , Cell Line , Chronic Disease , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Gasdermins/drug effects , Gasdermins/metabolism , Heart Failure/drug therapy , Heart Failure/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphate-Binding Proteins/metabolism , Pyroptosis/drug effects , Signal Transduction/drug effects
2.
Environ Sci Pollut Res Int ; 31(13): 19844-19855, 2024 Mar.
Article En | MEDLINE | ID: mdl-38367109

It is widespread of endemic fluorosis in China, and the exposure of excessive fluoride will cause nervous system disease and activate microglia. However, the mechanism of the damage is not clear. It is well-known that NLRP3/Caspase-1/GSDMD pathway, a classic pyroptosis pathway, is widely involved in the occurrence and development of nervous system-related diseases, infectious diseases, and atherosclerotic diseases. This research aimed to explore the molecular mechanism of sodium fluoride on inflammation and pyroptosis in BV2 microglia based on the NLRP3/Caspase-1/GSDMD signaling pathway. BV2 microglia was treated with sodium fluoride at the dose of 0.25, 1, and 2 mmol/L for 24, 48, and 72 h, respectively. Cell viability, cell morphology, lactate dehydrogenase content, and related proteins and genes were examined to investigate if sodium fluoride caused damage to BV2 microglia through the pyroptosis pathway. Dithiolam (5 µmol/L), a pyroptosis inhibitor, was added for further verification. NaF could induced BV2 cells injury in a dose-dependent fashion through disrupting the integrity of cell membranes and increasing IL-1ß via upregulating NLRP3, Caspase-1, and its downstream protein GSDMD. Disulfiram could improve these changes caused by NaF. In conclusion, our results suggested that NLRP3/Caspase-1/GSDMD-mediated classical pyroptosis pathway was involved in fluoride-induced BV2 microglia damage.


Fluorides , Microglia , Sodium Fluoride , Caspase 1/drug effects , Caspase 1/metabolism , Fluorides/toxicity , Microglia/drug effects , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sodium Fluoride/toxicity , Gasdermins/drug effects , Gasdermins/metabolism , Animals , Mice
3.
Toxins (Basel) ; 14(2)2022 02 18.
Article En | MEDLINE | ID: mdl-35202178

The Gram-negative, opportunistic pathogen Pseudomonas aeruginosa utilizes a type III secretion system to inject exoenzyme effectors into a target host cell. Of the four best-studied exoenzymes, ExoU causes rapid cell damage and death. ExoU is a phospholipase A2 (PLA2) that hydrolyses host cell membranes, and P. aeruginosa strains expressing ExoU are associated with poor outcomes in critically ill patients with pneumonia. While the effects of ExoU on lung epithelial and immune cells are well studied, a role for ExoU in disrupting lung endothelial cell function has only recently emerged. Lung endothelial cells maintain a barrier to fluid and protein flux into tissue and airspaces and regulate inflammation. Herein, we describe a pulmonary microvascular endothelial cell (PMVEC) culture infection model to examine the effects of ExoU. Using characterized P. aeruginosa strains and primary clinical isolates, we show that strains expressing ExoU disrupt PMVEC barrier function by causing substantial PMVEC damage and lysis, in a PLA2-dependent manner. In addition, we show that strains expressing ExoU activate the pro-inflammatory caspase-1, in a PLA2-dependent manner. Considering the important roles for mitochondria and oxidative stress in regulating inflammatory responses, we next examined the effects of ExoU on reactive oxygen species production. Infection of PMVECs with P. aeruginosa strains expressing ExoU triggered a robust oxidative stress compared to strains expressing other exoenzyme effectors. We also provide evidence that, intriguingly, ExoU PLA2 activity was detectable in mitochondria and mitochondria-associated membrane fractions isolated from P. aeruginosa-infected PMVECs. Interestingly, ExoU-mediated activation of caspase-1 was partially inhibited by reactive oxygen species scavengers. Together, these data suggest ExoU exerts pleiotropic effects on PMVEC function during P. aeruginosa infection that may inhibit endothelial barrier and inflammatory functions.


Bacterial Proteins/toxicity , Caspase 1/drug effects , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/physiopathology , Endothelial Cells/drug effects , Pseudomonas Infections/physiopathology , Pseudomonas aeruginosa/genetics , Caspase 1/metabolism , Genetic Variation , Genotype , Humans , Inflammation/chemically induced , Inflammation/physiopathology , Pseudomonas Infections/genetics
4.
Int J Mol Sci ; 23(3)2022 Feb 06.
Article En | MEDLINE | ID: mdl-35163769

The inflammatory protease caspase-1 is associated with the release of cytokines. An excessive number of cytokines (a "cytokine storm") is a dangerous consequence of COVID-19 infection and has been indicated as being among the causes of death by COVID-19. The anti-inflammatory drug colchicine (which is reported in the literature to be a caspase-1 inhibitor) and the corticosteroid drugs, dexamethasone and methylprednisolone, are among the most effective active compounds for COVID-19 treatment. The SERM raloxifene has also been used as a repurposed drug in COVID-19 therapy. In this study, inhibition of caspase-1 by these four compounds was analyzed using computational methods. Our aim was to see if the inhibition of caspase-1, an important biomolecule in the inflammatory response that triggers cytokine release, could shed light on how these drugs help to alleviate excessive cytokine production. We also measured the antioxidant activities of dexamethasone and colchicine when scavenging the superoxide radical using cyclic voltammetry methods. The experimental findings are associated with caspase-1 active site affinity towards these compounds. In evaluating our computational and experimental results, we here formulate a mechanism for caspase-1 inhibition by these drugs, which involves the active site amino acid Cys285 residue and is mediated by a transfer of protons, involving His237 and Ser339. It is proposed that the molecular moiety targeted by all of these drugs is a carbonyl group which establishes a S(Cys285)-C(carbonyl) covalent bond.


Anti-Inflammatory Agents/pharmacology , COVID-19 Drug Treatment , Caspase 1/drug effects , Caspase Inhibitors/pharmacology , Coronavirus 3C Proteases/drug effects , Anti-Inflammatory Agents/chemistry , COVID-19/metabolism , Caspase 1/chemistry , Caspase 1/metabolism , Caspase Inhibitors/chemistry , Colchicine/chemistry , Colchicine/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , Dexamethasone/pharmacology , Humans , Models, Molecular , Molecular Docking Simulation , Pentacyclic Triterpenes/pharmacology , Protein Interaction Domains and Motifs , Raloxifene Hydrochloride/chemistry , Raloxifene Hydrochloride/pharmacology , Viral Protease Inhibitors/chemistry , Viral Protease Inhibitors/pharmacology
5.
Int Immunopharmacol ; 102: 108405, 2022 Jan.
Article En | MEDLINE | ID: mdl-34865993

Inflammatory bowel disease (IBD) is an autoimmune disease involving intestinal tissue. IBD activates a series of cell death pathways. Pyroptosis is recently identified as a critical cell death pathway in IBD associated with the activation of caspase-1. VX765 is a caspase-1 inhibitor that can be converted to VRT-043198 in vivo. This study was designed to explore the therapeutic effect of VX765 on colitis using a dextran sulfate sodium (DSS)-induced colitis model in mice. In this research, the caspase-1 inhibitor on inflammatory, pyroptosis, apoptosis, macrophage activation, and intestinal barrier were investigated. We found that administration of VX765 attenuated body weight loss, colonic shortening, and colonic pathological injury in mice. Our study also revealed a therapeutic effect of VX765 on colitis in a dose-dependent manner. VX765 inhibited pyroptosis by curbing the Caspase-1/GSDMD pathway and its downstream key inflammatory cytokines--IL-1ß and IL-18. These results indicated that VX765 might have a dose-dependent therapeutic effect on DSS-induced colitis in mice.


Caspase 1/metabolism , Caspase Inhibitors/therapeutic use , Colitis/drug therapy , Dipeptides/therapeutic use , Pyroptosis/drug effects , para-Aminobenzoates/therapeutic use , Animals , Blotting, Western , Caspase 1/drug effects , Colitis/chemically induced , Dextran Sulfate/pharmacology , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
6.
Eur J Med Chem ; 229: 114002, 2022 Feb 05.
Article En | MEDLINE | ID: mdl-34823899

Compounds targeting the inflammasome-caspase-1 pathway could be of use for the treatment of inflammation and inflammatory diseases. Previous caspase-1 inhibitors were in great majority covalent inhibitors and failed in clinical trials. Using a mixed modelling, computational screening, synthesis and in vitro testing approach, we identified a novel class of non-covalent caspase-1 non cytotoxic inhibitors which are able to inhibit IL-1ß release in activated macrophages in the low µM range, in line with the best activities observed for the known covalent inhibitors. Our compounds could form the basis of further optimization towards potent drugs for the treatment of inflammation and inflammatory disorders including also dysregulated inflammation in Covid 19.


Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Autoimmune Diseases/drug therapy , Caspase 1/drug effects , Inflammasomes/drug effects , Inflammation/drug therapy , Serpins/chemical synthesis , Serpins/pharmacology , Tetrazoles/chemical synthesis , Tetrazoles/therapeutic use , Viral Proteins/chemical synthesis , Viral Proteins/pharmacology , COVID-19 , Cell Division/drug effects , Drug Design , Drug Evaluation, Preclinical , Humans , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Tetrazoles/pharmacology , U937 Cells
7.
Front Immunol ; 12: 720877, 2021.
Article En | MEDLINE | ID: mdl-34867948

Objectives: Combination therapy with mycophenolate mofetil, tacrolimus and steroids are effective in achieving complete remission in lupus nephritis (LN). Combination therapy uniquely downregulated caspase-1 compared with monotherapies, which can cleave gasdermin D (GSDMD) and was recently identified as the pyroptosis executioner. We therefore investigated whether combination therapy enabled the suppression of caspase-1/GSDMD-mediated pyroptosis in LN. Methods: Expression and activation of GSDMD were detected in kidney specimens of the human and mouse with LN using immunohistochemical staining and immunoblotting. Primary podocytes isolated from MRL/lpr mice were incubated with LPS+ATP, and pretreated with monotherapy or combination therapy. Inhibition of caspase-1/GSDMD-induced pyroptosis by combination therapy were assessed in MRL/lpr mice and human specimens. Pyroptosis was examined using a FAM caspase-1 kit and flow cytometry. The correlation between pyroptosis in peripheral blood and the systemic lupus erythematosus disease activity index (SLEDAI) was analyzed. Results: Kidney tissue specimens from LN patients and mice exhibited greatly increased expression levels and cleavage of GSDMD. In cultured podocytes, combination treatment significantly suppressed the activation of NLRP3 and caspase-1 and reduced GSDMD N-terminal levels. Combination therapy repressed disease progression through inhibition of caspase-1/GSDMD-mediated pyroptosis in both humans and MRL/lpr mice. Caspase-1/PI positive cell numbers in peripheral blood were positively correlated with SLE-DAI. LN patients with complete remission and partial remission had remarkably reduced caspase-1/PI positive cell numbers compared to baseline. Ac-FLTD-CMK, a GSDMD-derived inhibitor, prevented the development of LN. Conclusion: Combination therapy suppressed caspase-1/GSDMD-mediated pyroptosis in vitro and in vivo and reduced disease progression.


Caspase Inhibitors/administration & dosage , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lupus Nephritis/drug therapy , Phosphate-Binding Proteins/antagonists & inhibitors , Adolescent , Adult , Aged , Animals , Calcineurin Inhibitors/administration & dosage , Caspase 1/drug effects , Cells, Cultured , Cohort Studies , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Middle Aged , Mycophenolic Acid/administration & dosage , Podocytes/drug effects , Podocytes/metabolism , Podocytes/pathology , Prednisone/administration & dosage , Pyroptosis/drug effects , Tacrolimus/administration & dosage , Young Adult
8.
Biomed Pharmacother ; 141: 111835, 2021 Sep.
Article En | MEDLINE | ID: mdl-34146852

Thymic stromal lymphopoietin (TSLP) produced by mast cells is involved in allergic inflammation pathogenesis. Chloroquine (CQ) is known to be an anti-malarial drug; however, additional protective functions of CQ have been discovered. This study aims to clarify an anti-inflammatory effect of CQ through modulating TSLP levels using an in vitro model of phorbol myristate acetate (PMA) + A23187-activated human mast cell line (HMC-1) and an in vivo model of PMA-irritated ear edema. CQ treatment reduced the production and mRNA expression levels of TSLP in activated HMC-1 cells. CQ down-regulated caspase-1 (CASP1), MAPKs, and NF-κB levels enhanced by stimulation with PMA + A23187. Moreover, ear thickness in ear edema was suppressed following CQ treatment. CQ decreased CASP1 and NF-κB levels in the ear tissue. TSLP levels in the ear tissue and serum were reduced following CQ treatment. Collectively, the above findings elucidate that CQ inhibits the pro-inflammatory mechanisms of TSLP via the down-regulation of distinct intracellular signaling cascade in mast cells. Therefore, CQ may have protective roles against TSLP-mediated inflammatory disorders.


Caspase 1/drug effects , Caspase Inhibitors/pharmacology , Chloroquine/pharmacology , Cytokines/biosynthesis , Mast Cells/drug effects , Signal Transduction/drug effects , Stromal Cells/metabolism , Thymus Gland/metabolism , Animals , Calcimycin/pharmacology , Cell Line , Ear Diseases/drug therapy , Edema/drug therapy , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred ICR , NF-kappa B/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stromal Cells/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Thymus Gland/drug effects , Thymic Stromal Lymphopoietin
9.
Exp Cell Res ; 405(2): 112648, 2021 08 15.
Article En | MEDLINE | ID: mdl-34119493

The acute inflammatory stimulation occurring after a bone fracture regulates the repair and healing of local bone injury; however, under certain conditions, pyroptosis may occur in osteoblasts, which affects osteoblast proliferation and differentiation, thereby affecting the growth, development and morphological changes of bone tissue. The aim of the present study was to examine the effect of the pyroptosis inhibitor necrosulfonamide (NSA) on the proliferation and differentiation of osteoblasts and elucidate the underlying mechanism. The results revealed that NSA reversed the effects of ATP/lipopolysaccharide (LPS) on cell viability and pyroptosis, and on the mRNA and protein expression of pyroptosis-related genes. It also suppressed the secretion of IL-6, TNF-α and IL-1ß and reversed the effects of ATP/LPS on the activity of ALP and the mRNA expression of differentiation-related genes in osteoblasts. The fact that overexpression of caspase-1, gasdermin D (GSDMD) and NLRP3 abolished the effects of NSA on the viability and pyroptosis of osteoblasts, as well as the mRNA expression of differentiation-related genes and the activity of ALP in osteoblasts, indicated that NSA promoted the proliferation and differentiation of osteoblasts by inhibiting the NLRP3/caspase-1/GSDMD pyroptosis pathway. The present study provides proof supporting the potential application of NSA for improving the function of osteoblasts in fracture repair and indicates the value of the NLRP3/caspase-1/GSDMD pyroptosis pathway as a pharmaceutical target.


Acrylamides/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Osteoblasts/drug effects , Pyroptosis/drug effects , Sulfonamides/pharmacology , Caspase 1/drug effects , Caspase 1/metabolism , Cell Survival/drug effects , Humans , Lipopolysaccharides/pharmacology
10.
Brain Res Bull ; 174: 11-21, 2021 09.
Article En | MEDLINE | ID: mdl-33991606

Dexmedetomidine (Dex) has been suggested to exert a protective function in ischemic brain injury. In this study, we aimed to elucidate the intrinsic mechanisms of Dex in regulating microglia pyroptosis in ischemic brain injury via the purinergic 2X7 receptor (P2X7R)/NLRP3/Caspase-1 signaling pathway. First, permanent middle cerebral artery occlusion (p-MCAO) rat model was established, followed by the measurement of behavioral deficit, neuronal injury, the volume of brain edema and the infarct size. Dex treatment was suggested to alleviate the neurological deficits in p-MCAO rats and reduce the brain water content and infarct size. Additionally, rat microglia were cultured in vitro and a model of oxygen and glucose (OGD) was established. Microglia cell activity and ultrastructure were detected. Dex could increase cell activity and reduce LDH activity, partially reversing the changes in cell morphology. Furthermore, the activation of P2X7R/NLRP3/Caspase-1 pathway was tested. The obtained findings indicated Dex suppressed microglial pyroptosis by inhibiting the P2X7R/NLRP3/Caspase-1 pathway. Inhibition of P2X7R or NLRP3 could inhibit Caspase-1 p10 expression, improve cell activity, and reduce LDH activity. The same result was verified in vivo experiments. This study indicated that Dex inhibited microglia pyroptosis by blocking the P2X7R/NLRP3/Caspase-1 pathway, thus playing a protective role against ischemic brain injury.


Adrenergic alpha-2 Receptor Agonists/pharmacology , Brain Ischemia/prevention & control , Caspase 1/drug effects , Dexmedetomidine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Neuroprotective Agents/pharmacology , Receptors, Purinergic P2X7/drug effects , Signal Transduction/drug effects , Animals , Brain Chemistry/drug effects , Brain Edema/prevention & control , Brain Ischemia/psychology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/psychology , Male , Microglia/metabolism , Microglia/pathology , Pyroptosis/drug effects , Rats, Sprague-Dawley
11.
Int J Cosmet Sci ; 43(3): 283-291, 2021 Jun.
Article En | MEDLINE | ID: mdl-33559194

BACKGROUND: Cannabidiol is a plant-derived cannabinoid that has been suggested to have several human health benefits including potential anti-inflammatory effects. It is now common to find various forms of Cannabidiol, most often referred to as CBD, in nutritional supplements and topical treatments. The mechanisms by which CBD can influence inflammatory pathways in the body, and more particularly in the skin, are presently still unclear. It is known that CBD will bind to cannabinoid receptors, CB1 and CB2, in the body and recent work has shown that in keratinocytes, CBD can regulate inflammation through transcriptional regulation involving the NFÆ™ß nuclear pathways. The fact that CBD operates through the NFÆ™ß pathways suggests that, perhaps, the molecule may influence the expression of active caspase-1 through NLRP inflammasome-mediated pathways. METHODS: Recently, work has published demonstrating that Normal Human Epidermal Keratinocytes (NHEKs) can be activated by UVB and ATP to express active caspase-1 via NLRP inflammasome-mediated pathways. There was a strong interest to see whether highly purified Cannabidiol Isolate (>99% purity) might function to control release of active caspase-1 by testing of NHEKs using the previously described models. In addition, NHEKs expression of non-NLRP inflammasome-induced inflammation markers including IL-6, IL-8 and PGE2 was examined in UVB-activated NHEKs. RESULTS: It was found that purified Cannabidiol Isolate did not influence active caspase-1 release in either UVB or ATP-activated NHEKs suggesting the molecule does not influence the NLRP inflammasome pathways. In addition, it was surprisingly found that the Cannabidiol Isolate did not impact the expression of additional UVB-activated non-NLRP inflammatory markers. CONCLUSIONS: Data presented suggest that if Cannabidiol functions as an anti-inflammatory, it does so through pathways not associated with either the NLRP inflammasome-mediated expression of caspase-1 or through the more commonly known expression of interleukin or prostaglandin inflammatory pathways.


CONTEXTE: Le cannabidiol est un cannabinoïde d'origine végétale considéré comme bénéfique pour la santé humaine et présentant notamment des effets anti-inflammatoires potentiels. Il est désormais courant de trouver diverses formes de cannabidiol dans les suppléments alimentaires et les traitements topiques. Les mécanismes par lesquels le cannabidiol peut influencer les voies inflammatoires dans l'organisme, et plus particulièrement dans la peau sont actuellement encore flous. On sait que le cannabidiol se lie aux récepteurs cannabinoïdes, CB1 et CB2 dans l'organisme et des travaux récents ont montré que dans les kératinocytes, le cannabidiol peut réguler l'inflammation par régulation transcriptionnelle impliquant les voies nucléaires NF-ƙß. Le fait que le cannabidiol fonctionne par le biais des voies NF-Æ™ß laisse à penser que la molécule peut influencer l'expression de la Caspase-1 active à travers les voies médiées par l'inflammasome NLRP. MÉTHODES: Récemment, des travaux ont été publiés démontrant que les kératinocytes épidermiques humains normaux (Normal Human Epidermal Keratinocytes, NHEK) peuvent être activés par les UVB et l'ATP pour exprimer la Caspase-1 active à travers les voies médiées par l'inflammasome NLRP. On cherchait surtout à savoir si l'isolat de cannabidiol hautement purifié (pureté > 99 %) pouvait fonctionner pour contrôler la libération de Caspase-1 active en analysant les NHEK à l'aide des modèles décrits précédemment. En outre, l'expression des NHEK des marqueurs de l'inflammation induits par l'inflammasome non-NLRP, notamment : IL-6, IL-8 et la PGE2 ont été examinées dans des NHEK activées par les UVB. RÉSULTATS: Il a été constaté que l'isolat de cannabidiol purifié n'influençait pas la libération active de Caspase-1 dans les NHEK activées par les UVB ou l'ATP, ce qui suggère que la molécule n'influence pas les voies de l'inflammasome NLRP. En outre, il a été surprenant de constater que l'isolat de cannabidiol n'avait pas d'impact sur l'expression des marqueurs inflammatoires non-NLRP activés par les UVB supplémentaires. CONCLUSIONS: Les données présentées suggèrent que si le cannabidiol fonctionne comme un anti-inflammatoire, il le fait par des voies non associées à l'expression de la Caspase-1 médiée par l'inflammasome NLRP ou par l'expression plus connue des voies inflammatoires de l'interleukine ou de la prostaglandine.


Adenosine Triphosphate/metabolism , Cannabidiol/pharmacology , Caspase 1/drug effects , Cytokines/metabolism , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Keratinocytes/radiation effects , NLR Proteins/metabolism , Ultraviolet Rays , Cannabidiol/isolation & purification , Humans , Keratinocytes/metabolism
12.
Aging (Albany NY) ; 13(3): 3763-3778, 2021 01 10.
Article En | MEDLINE | ID: mdl-33461169

Dl-3-n-butylphthalide (NBP) has been widely used to treat ischemic stroke in China. To investigate the mechanisms underlying NBP activity, we established a permanent middle cerebral artery occlusion (pMCAO) rat model and injected the rats with 4 mg/kg/d NBP for nine days. We then assessed neuroinflammation, neovascularization and nerve regeneration within the brain. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry imaging (MALDI-TOF MSI) was used to determine the phospholipid distribution, while laser ablation-inductively coupled plasma mass spectrometry imaging (LA-ICP MSI) was used to measure Foxp3, Ki-67 and pCREB levels in the brain. Immunohistochemistry was used to investigate the expression of NLR family pyrin domain containing 3 (NLRP3) and its inflammatory products, caspase-1 and interleukin-1ß, in brain tissues. NBP attenuated ischemic damage and ameliorated neurological deficits in rats with pMCAO. In the ischemic brain region, NBP reduced phosphatidylethanolamine (18:0), NLRP3, caspase-1 and interleukin-1ß levels, but increased levels of Foxp3, Ki-67, pCREB and several phospholipids. In molecular docking analyses, NBP bound to NLRP3, interleukin-1ß, caspase-1, Foxp3, and Ki-67. These results demonstrate that NBP reduces neuroinflammation in brain tissues and promotes nerve and blood vessel regeneration, thus protecting neuromorphology and function.


Benzofurans/pharmacology , Brain/drug effects , Forkhead Transcription Factors/drug effects , Ischemic Stroke/metabolism , Ki-67 Antigen/drug effects , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain/metabolism , Brain/pathology , Caspase 1/drug effects , Caspase 1/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Forkhead Transcription Factors/metabolism , Infarction, Middle Cerebral Artery , Inflammation/metabolism , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Ki-67 Antigen/metabolism , Molecular Docking Simulation , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Regeneration/drug effects , Phospholipids/metabolism , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Neuroreport ; 32(3): 188-197, 2021 02 03.
Article En | MEDLINE | ID: mdl-33470761

Pyroptosis has been reported to contribute to the traumatic brain injury (TBI) process. Ac-FLTD-CMK is a newly synthesized pyroptosis inhibitor. However, whether Ac-FLTD-CMK inhibits pyroptosis and plays a neuroprotective role after TBI is unknown. The present study aimed to determine the effects of Ac-FLTD-CMK on TBI in a mouse model. Male C57BL/6 mice were randomly divided into sham, TBI + vehicle, and TBI + Ac-FLTD-CMK groups. TBI was induced using a weight-drop apparatus. Intraventricular injection of Ac-FLTD-CMK was performed 30 min after TBI. Caspase-1, caspase-11, gasdermin-D (GSDMD), and caspase-3 expression in the peri-contusional cortex were assessed by western blotting. Interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) expression in the peri-contusional cortex were measured using ELISA. Behavioral experiments, brain water content, Evans blue extravasation, lactate dehydrogenase (LDH) release, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining were also performed. The results showed that Ac-FLTD-CMK administration significantly downregulated caspase-1 p20, caspase-11 p20, GSDMD N-terminal, IL-1ß, and IL-18 expression; reduced LDH release; alleviated neuronal death; attenuated brain edema and blood-brain barrier damage; and improved neurobehavioral function. These findings indicate that Ac-FLTD-CMK treatment suppresses pyroptosis and protects mice against TBI.


Brain Contusion/metabolism , Brain/drug effects , Caspase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Pyroptosis/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Injuries, Traumatic/metabolism , Caspase 1/drug effects , Caspase 1/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Caspases, Initiator/drug effects , Caspases, Initiator/metabolism , Disease Models, Animal , Interleukin-18/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Open Field Test , Phosphate-Binding Proteins/drug effects , Phosphate-Binding Proteins/metabolism , Rotarod Performance Test
14.
J Med Chem ; 64(1): 101-122, 2021 01 14.
Article En | MEDLINE | ID: mdl-33351619

Inhibiting the NLRP3 inflammasome mediates inflammation in an extensive number of preclinical models. As excitement in this field has grown, several companies have recently initiated testing of direct NLRP3 inhibitors in the clinic. At the same time, the NLRP3 inflammasome is part of a larger pro-inflammatory pathway, whose modulation is also being explored. Multiple targets in this pathway are already impinged upon by molecules that have been through clinical trials. These data, informed by the growing mechanistic understanding of the NLRP3 inflammasome in the preclinical space, provide a rich backdrop to assess the current state of the field. Here we explore attempts to inhibit the NLRP3 inflammasome in light of clinical and preclinical data around efficacy and safety.


Drug Delivery Systems , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Caspase 1/drug effects , Clinical Trials as Topic , Humans , Inflammation/drug therapy , Interleukin-1beta/antagonists & inhibitors , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Protein Binding , Receptors, Interleukin-1/antagonists & inhibitors , Signal Transduction
15.
Aging (Albany NY) ; 13(1): 865-876, 2020 12 01.
Article En | MEDLINE | ID: mdl-33260147

The activation of pyroptosis is an important feature of renal ischemia/reperfusion (rI/R)-induced acute lung injury (ALI). Propofol, a general anesthetic, is known to inhibit inflammation in I/R-induced ALI. We investigated whether propofol could suppress pyroptosis during rI/R-induced ALI by upregulating sirtuin 1 (SIRT1). We generated an in vivo model of rI/R-induced ALI by applying microvascular clamps to the renal pedicles of rats for 45 min. Pathological studies revealed that rI/R provoked substantial lung injury and inflammatory cell infiltration. The rI/R stimulus markedly activated pyroptotic proteins such as NLRP3, ASC, caspase 1, interleukin-1ß and interleukin-18 in the lungs, but reduced the mRNA and protein levels of SIRT1. Propofol treatment greatly inhibited rI/R-induced lung injury and pyroptosis, whereas it elevated SIRT1 expression. Treatment with the selective SIRT1 inhibitor nicotinamide reversed the protective effects of propofol during rI/R-induced ALI. Analogous defensive properties of propofol were detected in vitro in rat alveolar macrophages incubated with serum from the rI/R rat model. These findings indicate that propofol attenuates rI/R-induced ALI by suppressing pyroptosis, possibly by upregulating SIRT1 in the lungs.


Acute Lung Injury/genetics , Anesthetics, Intravenous/pharmacology , Kidney/blood supply , Lung/drug effects , Propofol/pharmacology , Pyroptosis/drug effects , Reperfusion Injury/genetics , Sirtuin 1/drug effects , Acute Lung Injury/metabolism , Animals , CARD Signaling Adaptor Proteins/drug effects , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/drug effects , Caspase 1/metabolism , Cell Line , Interleukin-18/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Lung/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Niacinamide/pharmacology , Pyroptosis/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Reperfusion Injury/metabolism , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Sirtuin 1/metabolism
16.
Int Immunopharmacol ; 88: 106872, 2020 Nov.
Article En | MEDLINE | ID: mdl-32769069

Receptor activator of NF-κB ligand (RANKL) as an osteoclast differentiation factor induces inflammatory reactions via production of thymic stromal lymphopoietin (TSLP). Epigallocatechin gallate (EGCG) is the major and the most active compound in green tea and has anti-inflammatory, anti-cancer, anti-oxidant, and neuroprotective effects. However, the effect and molecular mechanisms of EGCG are still unknown in RANKL-induced inflammatory reactions. Here we investigated the immuno-regulatory effects and its molecular mechanisms of epigallocatechin gallate (EGCG) in RANKL-stimulated human mast cell line, HMC-1 cells. In this study, EGCG prevented expression of PI3 Kinase and phosphorylation of mitogen-activated protein (MAP) Kinases in RANKL-stimulated HMC-1 cells. EGCG prevented caspase-1 activity and decreased transcriptional activity of nuclear factor (NF)-κB by suppressing inhibitory protein κBα phosphorylation in RANKL-stimulated HMC-1 cells. EGCG has been shown to prevent production and mRNA expression of TSLP, interleukin (IL)-1ß, IL-6, and IL-8 by RANKL without cytotoxicity. Furthermore, EGCG prevented degranulation of mast cell in RANKL-stimulated HMC-1 cells. Overall, these results suggest that EGCG acts as a natural agent for preventing and treating RANKL-mediated inflammatory diseases by targeting PI3 Kinase, MAP Kinase, caspase-1, and NF-κB signaling cascade in mast cells.


Catechin/analogs & derivatives , Inflammation/metabolism , Mast Cells/drug effects , RANK Ligand/antagonists & inhibitors , Signal Transduction/drug effects , Caspase 1/drug effects , Caspase 1/metabolism , Catechin/pharmacology , Cell Line , Cell Survival/drug effects , Cytokines/drug effects , Cytokines/metabolism , Elafin/drug effects , Elafin/metabolism , Histamine/metabolism , Humans , Inflammation/chemically induced , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Mast Cells/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , RANK Ligand/adverse effects , Thymic Stromal Lymphopoietin
17.
Biomed Pharmacother ; 130: 110521, 2020 Oct.
Article En | MEDLINE | ID: mdl-32717631

OBJECTIVE: This study aimed to assess the effect and mechanism of SS31 on cisplatin-induced acute kidney injury (CP-AKI) both in vivo and in vitro. METHOD: Male mices and HK-2 cells were treated using cisplatin to establish models of CP-AKI. 32 C57BL/6 mices were randomly divided into four groups (control group, CP group, CP + normal saline group, CP + SS-31 group). Cisplatin was intraperitoneally injected once to the mice (25 mg/kg). SS31 was administrated for 10 days at dosages of 10 mg/kg per day. Kidney histological changes and level of reactive oxygen species(ROS) were detected. In vitro studies, HK-2 cells were incubated with cisplatin (50 u M) or combimed with SS-31(100 u M), the level of mitochondrial ROS, apoptosis rate and the the expression of NLRP3, Caspase-1 and IL-1ß were tested. RESULTS: Renal tubulointerstitial apoptosis and oxidative stress were significantly increased in CP-AKI mice. Cisplatin caused elevation of serum creatinine (Scr), blood urea nitrogen (BUN) levels and enhanced IL-1ß, caspase1 and NLRP3 expression, the electron microscopy examination showed mitochondria cristae swelling, mitochondrial spheres and partial ridge breakdown in renal tubular cell of CP-AKI mice. SS31 treatment could effectively suppress mitochondrial ROS, ameliorate these lesions and decrease the expression of NLRP3, IL-1ß and Caspase1. In vitro studies, SS31 could restored the level of mitochondrial ROS and downregulate apoptosis rate in HK-2 cells, moreover, the elevated expression of NLRP3, IL-1ß and Caspase-1were restored. CONCLUSION: SS31 could protect CP-AKI in mices, which might be due to an anti-oxidative and anti-apoptotic action via regulating mitochondrial ROS-NLRP3 pathway. NLRP3 inflammasome might be considered as a novel therapeutic target of CP-AKI.


Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Antineoplastic Agents/toxicity , Cisplatin/antagonists & inhibitors , Cisplatin/toxicity , Mitochondria/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Oligopeptides/therapeutic use , Reactive Oxygen Species , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Caspase 1/drug effects , Cell Line , Humans , Inflammasomes/drug effects , Interleukin-1beta/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/metabolism
18.
Alcohol Clin Exp Res ; 44(8): 1585-1597, 2020 08.
Article En | MEDLINE | ID: mdl-32524615

BACKGROUND: Alcohol use disorders affect millions of people worldwide, and there is growing evidence that excessive alcohol intake causes severe damage to the brain of both humans and animals. Numerous studies on chronic alcohol exposure in animal models have identified that many functional impairments are associated with the hippocampus, which is a structure exhibiting substantial vulnerability to alcohol exposure. However, the precise mechanisms that lead to structural and functional impairments of the hippocampus are poorly understood. Herein, we report a novel cell death type, namely pyroptosis, which accounts for alcohol neurotoxicity in mice. METHODS: For this study, we used an in vivo model to induce alcohol-related neurotoxicity in the hippocampus. Adult male C57BL/6 mice were treated with 95% alcohol vapor either alone or in combination with selective cannabinoid receptor antagonists or agonists, and VX765 (Belnacasan), which is a selective caspase-1 inhibitor. RESULTS: Alcohol-induced in vivo pyroptosis occurs because of an increase in the levels of pyroptotic proteins such as nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3), caspase-1, gasdermin D (GSDMD), and amplified inflammatory response. Our results indicated that VX765 suppressed the expression of caspase-1 and inhibited the maturation of the proinflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. Additionally, chronic alcohol intake created an imbalance in the endocannabinoid system and regulated 2 cannabinoid receptors (CB1R and CB2R) in the hippocampus. Specific antagonists of CB1R (AM251 and AM281) significantly ameliorated alcohol-induced pyroptosis signaling and inactivated the inflammatory response. CONCLUSIONS: Alcohol induces hippocampal pyroptosis, which leads to neurotoxicity, thereby indicating that pyroptosis may be an essential pathway involved in chronic alcohol-induced hippocampal neurotoxicity. Furthermore, cannabinoid receptors are regulated during this process, which suggests promising therapeutic strategies against alcohol-induced neurotoxicity through pharmacologic inhibition of CB1R.


Alcohol-Induced Disorders, Nervous System/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Pyroptosis/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Cannabinoid Receptor Agonists/pharmacology , Caspase 1/drug effects , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Dipeptides/pharmacology , Inflammation , Interleukin-18/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Morpholines/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurotoxicity Syndromes , Phosphate-Binding Proteins/drug effects , Phosphate-Binding Proteins/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , para-Aminobenzoates/pharmacology
19.
Alcohol Clin Exp Res ; 44(6): 1300-1311, 2020 06.
Article En | MEDLINE | ID: mdl-32282939

BACKGROUND: Activation of NLRP3 in liver macrophages contributes to alcohol-associated liver disease (ALD). Molecular chaperone heat shock protein (HSP) 90 facilitates NLRP3 inflammasome activity during infections and inflammatory diseases. We previously reported that HSP90 is induced in ALD and regulates proinflammatory cytokines, tumor necrosis factor alpha, and IL-6. Whether HSP90 affects IL-1ß and IL-18 regulated by NLRP3 inflammasome in ALD is unknown. Here, we hypothesize that HSP90 modulated NLRP3 inflammasome activity and affects IL-1ß and IL-18 secretion in ALD. METHODS: The expression of HSP90AA1 and NLRP3 inflammasome genes was evaluated in human alcoholic livers and in mouse model of ALD. The importance of HSP90 on NLRP3 inflammasome activation in ALD was evaluated by administering HSP90 inhibitor, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) to mice subjected to ALD, and in vitro to bone marrow-derived macrophages (BMDM) stimulated with LPS and ATP. The effect of activation of HSF1/HSPA1A axis during HSP90 inhibition or direct activation during heat shock of BMDMs on NLRP3 activity and secretion of downstream cytokines was evaluated. RESULTS: We found positive correlation between induction of HSP90 and NLRP3 inflammasome genes in human alcoholic cirrhotic livers. Administration of 17-DMAG in mouse model of ALD significantly down-regulated NLRP3 inflammasome-mediated caspase-1 (CASP-1) activity and cytokine secretion, with reduction in ALD. 17-DMAG-mediated decrease in NLRP3 was restricted to liver macrophages. Using BMDMs, we show that inhibition of HSP90 prevented CASP-1 activity, and Gasdermin D (GSDMD) cleavage, important in release of active IL-1ß and IL-18. Interestingly, activation of the heat shock factor 1 (HSF1)/HSPA1A axis, either during HSP90 inhibition or by heat shock, decreased NLRP3 inflammasome activity and reduced secretion of cytokines. CONCLUSION: Our studies indicate that inhibition of HSP90 and activation of HSF1/HSPA1A reduce IL-1ß and IL-18 via decrease in NLRP3/CASP-1 and GSDMD activity in ALD.


Liver Diseases, Alcoholic/genetics , Adult , Aged , Animals , Benzoquinones/pharmacology , Caspase 1/drug effects , Caspase 1/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors/metabolism , Humans , In Vitro Techniques , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lactams, Macrocyclic/pharmacology , Liver Cirrhosis, Alcoholic/genetics , Liver Cirrhosis, Alcoholic/metabolism , Liver Diseases, Alcoholic/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasm Proteins , RNA, Messenger/metabolism , Young Adult
20.
Arthritis Rheumatol ; 72(7): 1192-1202, 2020 07.
Article En | MEDLINE | ID: mdl-32134203

OBJECTIVE: The NLRP3 inflammasome is closely linked to the pathophysiology of a wide range of inflammatory diseases. This study was undertaken to identify small molecules that directly bind to NLRP3 in order to develop pharmacologic interventions for NLRP3-related diseases. METHODS: A structure-based virtual screening analysis was performed with ~62,800 compounds to select efficient NLRP3 inhibitors. The production of caspase 1-p10 and interleukin-1ß (IL-1ß) was measured by immunoblotting and enzyme-linked immunosorbent assay to examine NLRP3 inflammasome activation. Two gouty arthritis models and an air pouch inflammation model induced by monosodium urate monohydrate (MSU) crystal injection were used for in vivo experiments. Primary synovial fluid cells from gout patients were used to determine the relevance of NLRP3 inflammasome inhibition in human gout. RESULTS: Beta-carotene (provitamin A) suppressed the NLRP3 inflammasome activation induced by various activators, including MSU crystals, in mouse bone marrow-derived primary macrophages (P < 0.05). Surface plasmon resonance analysis demonstrated the direct binding of ß-carotene to the pyrin domain (PYD) of NLRP3 (KD = 3.41 × 10-6 ). Molecular modeling and mutation assays revealed the interaction mode between ß-carotene and the NLRP3 PYD. Inflammatory symptoms induced by MSU crystals were attenuated by oral administration of ß-carotene in gouty arthritis mouse models (P < 0.05), correlating with its suppressive effects on the NLRP3 inflammasome in inflamed tissues. Furthermore, ß-carotene reduced IL-1ß secretion from human synovial fluid cells isolated from gout patients (P < 0.05), showing its inhibitory efficacy in human gout. CONCLUSION: Our results present ß-carotene as a selective and direct inhibitor of NLRP3, and the binding of ß-carotene to NLRP3 PYD as a novel pharmacologic strategy to combat NLRP3 inflammasome-driven diseases, including gouty arthritis.


Arthritis, Gouty/immunology , Inflammasomes/antagonists & inhibitors , Inflammation/immunology , Macrophages/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Provitamins/pharmacology , beta Carotene/pharmacology , Animals , Caspase 1/drug effects , Caspase 1/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gout/immunology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Macrophages/immunology , Mice , Molecular Docking Simulation , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyrin Domain , Surface Plasmon Resonance , Synovial Fluid/cytology
...