Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 176
Filtrer
1.
Arthritis Res Ther ; 26(1): 119, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38863059

RÉSUMÉ

OBJECTIVE: In gout, monosodium urate crystals are taken up by macrophages, triggering the activation of the NLRP3 inflammasome and the maturation of IL-1ß. This study aimed to investigate the role of integrin CD11b in inflammasome activation in macrophages stimulated by MSU. METHODS: BMDM from WT and CD11b KO mice were stimulated in vitro with MSU crystals. Cellular supernatants were collected to assess the expression of the inflammatory cytokines by enzyme-linked immunosorbent assay and western blot methods. The role of integrin CD11b in MSU-induced gouty arthritis in vivo was investigated by intra-articular injection of MSU crystals. Real-time extracellular acidification rate and oxygen consumption rate of BMDMs were measured by Seahorse Extracellular Flux Analyzer. RESULTS: We demonstrate that CD11b-deficient mice developed exacerbated gouty arthritis with increased recruitment of leukocytes in the joint and higher IL-1ß levels in the sera. In macrophages, genetic deletion of CD11b induced a shift of macrophage metabolism from oxidative phosphorylation to glycolysis, thus decreasing the overall generation of intracellular ATP. Upon MSU stimulation, CD11b-deficient macrophages showed an exacerbated secretion of IL-1ß. Treating wild-type macrophages with a CD11b agonist, LA1, inhibited MSU-induced release of IL-1ß in vitro and attenuated the severity of experimental gouty arthritis. Importantly, LA1, was also effective in human cells as it inhibited MSU-induced release of IL-1ß by peripheral blood mononuclear cells from healthy donors. CONCLUSION: Our data identified the CD11b integrin as a principal cell membrane receptor that modulates NLRP3 inflammasome activation by MSU crystal in macrophages, which could be a potential therapeutic target to treat gouty arthritis in human patients.


Sujet(s)
Goutte articulaire , Antigènes CD11b , Inflammasomes , Macrophages , Souris de lignée C57BL , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine , Acide urique , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Macrophages/métabolisme , Antigènes CD11b/métabolisme , Inflammasomes/métabolisme , Goutte articulaire/induit chimiquement , Goutte articulaire/métabolisme , Souris , Mâle
2.
Front Biosci (Landmark Ed) ; 29(6): 222, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38940057

RÉSUMÉ

BACKGROUND: Persistent hyperuricemia can lead to the generation and deposition of monosodium urate (MSU) crystals. This can trigger gouty arthritis (GA), which in turn induces inflammation. Activation of the Nod-like receptor pyrin domain containing 3 (NLRP3) inflammasome plays a critical role in the onset and progression of GA. Autophagy may have a dual effect on GA with regard to the NLRP3 inflammasome. Therefore, the present study aimed to gain a deeper comprehension of the interaction between autophagy and NLRP3 inflammasome activation is imperative for developing more efficacious treatments for GA. METHODS: Peripheral blood monocytes (PBMCs) were first isolated from GA patients and healthy controls and underwent bulk RNA sequencing analysis. Overexpression and knockdown of dual specificity phosphatase 1 (DUSP1) was performed in THP-1 monocytes to investigate its role in the immune response and mitochondrial damage. The luciferase assay and Western blot analysis were used to study the interaction between autophagy and NLRP3 inflammasome activation. RESULTS: Bulk RNA sequencing analysis showed significant upregulation of DUSP1 expression in PBMCs from GA patients compared to healthy controls. This result was subsequently verified by reverse transcription quantitative polymerase chain reaction (RT-qPCR). DUSP1 expression in human THP-1 monocytes was also shown to increase after MSU treatment. Downregulation of DUSP1 expression increased the secretion of inflammatory cytokines after MSU treatment, whereas the overexpression of DUSP1 decreased the secretion levels. Lipopolysaccharides (LPS) combined with adenosine-triphosphate (ATP) led to mitochondrial damage, which was rescued by overexpressing DUSP1. DUSP1 overexpression further increased the level of autophagy following MSU treatment, whereas downregulation of DUSP1 decreased autophagy. Treatment with the autophagy inhibitor 3-Methyladenine (3-MA) restored inflammatory cytokine secretion levels in the DUSP1 overexpression group. MSU caused pronounced pathological ankle swelling in vivo. However, DUSP1 overexpression significantly mitigated this phenotype, accompanied by significant downregulation of inflammatory cytokine secretion levels in the joint tissues. CONCLUSIONS: This study revealed a novel function and mechanism for DUSP1 in promoting autophagy to mitigate the MSU-induced immune response in GA. This finding suggests potential diagnostic biomarkers and anti-inflammatory targets for more effective GA therapy.


Sujet(s)
Goutte articulaire , Autophagie , Dual Specificity Phosphatase 1 , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Acide urique , Humains , Autophagie/effets des médicaments et des substances chimiques , Dual Specificity Phosphatase 1/génétique , Dual Specificity Phosphatase 1/métabolisme , Goutte articulaire/génétique , Goutte articulaire/métabolisme , Goutte articulaire/immunologie , Goutte articulaire/induit chimiquement , Acide urique/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Inflammasomes/métabolisme , Inflammasomes/immunologie , Cellules THP-1 , Mâle , Monocytes/métabolisme , Monocytes/immunologie , Monocytes/effets des médicaments et des substances chimiques , Études cas-témoins , Femelle , Agranulocytes/métabolisme , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/immunologie , Adulte d'âge moyen
3.
Mol Med ; 30(1): 67, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773379

RÉSUMÉ

BACKGROUND: Gouty arthritis (GA) is characterized by monosodium urate (MSU) crystal accumulation that instigates NLRP3-mediated pyroptosis; however, the underlying regulatory mechanisms have yet to be fully elucidated. The present research endeavors to elucidate the regulatory mechanisms underpinning this MSU-induced pyroptotic cascade in GA. METHODS: J774 cells were exposed to lipopolysaccharide and MSU crystals to establish in vitro GA models, whereas C57BL/6 J male mice received MSU crystal injections to mimic in vivo GA conditions. Gene and protein expression levels were evaluated using real-time quantitative PCR, Western blotting, and immunohistochemical assays. Inflammatory markers were quantified via enzyme-linked immunosorbent assays. Pyroptosis was evaluated using immunofluorescence staining for caspase-1 and flow cytometry with caspase-1/propidium iodide staining. The interaction between MDM2 and PPARγ was analyzed through co-immunoprecipitation assays, whereas the interaction between BRD4 and the MDM2 promoter was examined using chromatin immunoprecipitation and dual-luciferase reporter assays. Mouse joint tissues were histopathologically evaluated using hematoxylin and eosin staining. RESULTS: In GA, PPARγ was downregulated, whereas its overexpression mitigated NLRP3 inflammasome activation and pyroptosis. MDM2, which was upregulated in GA, destabilized PPARγ through the ubiquitin-proteasome degradation pathway, whereas its silencing attenuated NLRP3 activation by elevating PPARγ levels. Concurrently, BRD4 was elevated in GA and exacerbated NLRP3 activation and pyroptosis by transcriptionally upregulating MDM2, thereby promoting PPARγ degradation. In vivo experiments showed that BRD4 silencing ameliorated GA through this MDM2-PPARγ-pyroptosis axis. CONCLUSION: BRD4 promotes inflammation and pyroptosis in GA through MDM2-mediated PPARγ degradation, underscoring the therapeutic potential of targeting this pathway in GA management.


Sujet(s)
Goutte articulaire , Récepteur PPAR gamma , Protéines proto-oncogènes c-mdm2 , Pyroptose , Facteurs de transcription , Animaux , Mâle , Souris , Goutte articulaire/métabolisme , Goutte articulaire/génétique , Goutte articulaire/anatomopathologie , Goutte articulaire/induit chimiquement , Protéines contenant un bromodomaine , Lignée cellulaire , Modèles animaux de maladie humaine , Inflammasomes/métabolisme , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéines nucléaires , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Protéolyse , Protéines proto-oncogènes c-mdm2/métabolisme , Protéines proto-oncogènes c-mdm2/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Acide urique/métabolisme
4.
Arthritis Res Ther ; 26(1): 96, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38711064

RÉSUMÉ

BACKGROUND: Gout is caused by monosodium urate (MSU) crystals deposition to trigger immune response. A recent study suggested that inhibition of Class I Histone deacetylases (HDACs) can significantly reduce MSU crystals-induced inflammation. However, which one of HDACs members in response to MSU crystals was still unknown. Here, we investigated the roles of HDAC3 in MSU crystals-induced gouty inflammation. METHODS: Macrophage specific HDAC3 knockout (KO) mice were used to investigate inflammatory profiles of gout in mouse models in vivo, including ankle arthritis, foot pad arthritis and subcutaneous air pouch model. In the in vitro experiments, bone marrow-derived macrophages (BMDMs) from mice were treated with MSU crystals to assess cytokines, potential target gene and protein. RESULTS: Deficiency of HDAC3 in macrophage not only reduced MSU-induced foot pad and ankle joint swelling but also decreased neutrophils trafficking and IL-1ß release in air pouch models. In addition, the levels of inflammatory genes related to TLR2/4/NF-κB/IL-6/STAT3 signaling pathway were significantly decreased in BMDMs from HDAC3 KO mice after MSU treatment. Moreover, RGFP966, selective inhibitor of HDAC3, inhibited IL-6 and TNF-α production in BMDMs treated with MSU crystals. Besides, HDAC3 deficiency shifted gene expression from pro-inflammatory macrophage (M1) to anti-inflammatory macrophage (M2) in BMDMs after MSU challenge. CONCLUSIONS: Deficiency of HDAC3 in macrophage alleviates MSU crystals-induced gouty inflammation through inhibition of TLR2/4 driven IL-6/STAT3 signaling pathway, suggesting that HDAC3 could contribute to a potential therapeutic target of gout.


Sujet(s)
Acrylamides , Goutte , Histone deacetylases , Macrophages , Souris de lignée C57BL , Souris knockout , Phénylènediamines , Acide urique , Animaux , Acide urique/toxicité , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Histone deacetylases/déficit , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Goutte/métabolisme , Goutte/anatomopathologie , Souris , Inflammation/métabolisme , Inflammation/induit chimiquement , Mâle , Goutte articulaire/induit chimiquement , Goutte articulaire/métabolisme , Goutte articulaire/anatomopathologie , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques
5.
Inflammopharmacology ; 32(3): 1929-1940, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38556563

RÉSUMÉ

Gout is a metabolic condition characterized by the accumulation of urate crystals in the synovial joints. These crystal depositions result in joint swelling and increased concentration of serum uric acid in blood. The commercially available drugs lower serum uric acid levels and reduce inflammation, but these standard therapies have many side effects. This study aimed to investigate anti-gout and anti-inflammatory properties of curcumin nanoparticles (CNPs). For this purpose, CNPs were prepared by dissolving curcumin into dichloromethane. Then, gout was induced by injecting monosodium urate crystals (MSU) in the ankle joint and in the intra-peritoneal cavity which caused ankle swelling and increased blood uric acid levels. CNPs in different concentrations (5, 10, and 20 ppm) and allopurinol were orally administered. The MSU crystals increased the xanthine oxidase levels both in serum and the liver. Moreover, MSU crystals increased the serum levels of interleukin 1ß, interleukin-6, tumor necrosis factor-alpha, liver function tests markers, renal function tests markers, and lipid profiles. However, the administration of CNPs decreased the levels of all these variables. CNPs increased the serum high-density lipoprotein and interleukin-10 levels. Moreover, CNPs also reduced ankle swelling significantly. Hence, the levels of xanthine oxidase, uric acid and ankle swelling were reduced significantly by oral administration of CNPs. Our findings indicate that CNPs through their anti-inflammatory properties significantly alleviate gouty arthritis. Thus, the study concluded that CNPs can be developed as an efficient anti-gout agent with minimal side effects.


Sujet(s)
Anti-inflammatoires , Goutte articulaire , Curcumine , Souris de lignée BALB C , Nanoparticules , Acide urique , Animaux , Curcumine/pharmacologie , Curcumine/administration et posologie , Acide urique/sang , Goutte articulaire/traitement médicamenteux , Goutte articulaire/induit chimiquement , Souris , Nanoparticules/administration et posologie , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/administration et posologie , Mâle , Xanthine oxidase/métabolisme , Antigoutteux/pharmacologie , Antigoutteux/administration et posologie , Inflammation/traitement médicamenteux , Inflammation/induit chimiquement
6.
Chem Biodivers ; 21(5): e202400448, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38498112

RÉSUMÉ

Citronella and Nutmeg are two common spices used for seasoning and medicinal purposes, both of which have significant economic value. This study aimed to investigate whether Citronella essential oil and Nutmeg essential oil (NEO) can ameliorate monosodium urate (MSU)-induced gouty arthritis in rats and the potential mechanisms. The results showed that CEO and NEO reduced swelling and redness at joint sites, inhibited neutrophil infiltration, and limited proinflammatory mediator secretion in mice with MSU-induced gouty arthritis. Based on the results of network pharmacology, molecular docking, and western blotting, CEO and NEO may exert anti-gouty arthritis effects by reducing the expression of reactive oxygen species and oxidative stress and downregulating the phosphorylation of the PI3K/AKT/mTOR signaling pathway, thereby inhibiting the production of the NLRP3 inflammasome and inhibiting the production of inflammatory cytokines. Therefore, these two essential oils show potential for use as adjuvant treatments for gouty arthritis in specific aromatherapy products or food seasonings.


Sujet(s)
Goutte articulaire , Protéine-3 de la famille des NLR contenant un domaine pyrine , Huile essentielle , Stress oxydatif , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Sérine-thréonine kinases TOR , Huile essentielle/pharmacologie , Huile essentielle/composition chimique , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/antagonistes et inhibiteurs , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Stress oxydatif/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Phosphatidylinositol 3-kinases/métabolisme , Souris , Goutte articulaire/traitement médicamenteux , Goutte articulaire/induit chimiquement , Goutte articulaire/métabolisme , Rats , Mâle , Myristica/composition chimique , Acide urique/métabolisme , Simulation de docking moléculaire , Rat Sprague-Dawley
7.
Cell Mol Biol (Noisy-le-grand) ; 70(2): 205-211, 2024 Feb 29.
Article de Anglais | MEDLINE | ID: mdl-38430024

RÉSUMÉ

Gouty arthritis (GA) is an inflammatory disease caused by the deposition of monosodium urate (MSU) crystals into joints. Tetrandrine (TET) is a bisbenzylisoquinoline alkaloid extracted from the root of Stephania tetrandra and can exert an anti-inflammatory function in different diseases. Nevertheless, the specific function of TET in GA remains unclear. We established the GA mouse model by MSU injection into joints of mice. Paw volume and gait score were detected for measuring the degree of joint swelling and the situation of joint dysfunction. Western blot were utilized to test the alterations of M1-related factors (IL-6, IL-1ß, TNF-α, IL-12, and iNOS) and M2-related factors (Mgl1, Mgl2, Pgc1-ß, Arg-1, and IL-10). The activity of NF-κB p65 in tissues was determined. The interaction of NF-κB p65 and Lcp1 was measured by ChIP and luciferase reporter assay. Lcp1 KO mice were utilized to detect the effect of Lcp1 depletion on GA process. TET treatment markedly suppressed MSU-induced joint swelling, joint dysfunction, and joint injury in GA mice. TET can also reduce inflammatory reactions in MUS-induced mice. Furthermore, we proved that TET facilitated M2 macrophage polarization and inhibited M1 macrophage polarization in GA mice. In addition, TET was found to inhibit NF-κB activity and NF-κB-mediated Lcp1 expression. Lcp1 knockdown can improve joint injury and promote M2 macrophage polarization in GA mice, while this effect was further enhanced by TET. TET alleviates inflammation and facilitates macrophage M2 polarization in GA by NF-κB-mediated Lcp1.


Sujet(s)
Goutte articulaire , Benzylisoquinoléines , Goutte articulaire/traitement médicamenteux , Goutte articulaire/induit chimiquement , Goutte articulaire/métabolisme , Benzylisoquinoléines/effets indésirables , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Macrophages , Facteur de transcription NF-kappa B/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Acide urique/effets indésirables , Acide urique/métabolisme , Animaux , Souris
8.
Zhongguo Zhong Yao Za Zhi ; 49(1): 224-231, 2024 Jan.
Article de Chinois | MEDLINE | ID: mdl-38403355

RÉSUMÉ

This study aims to reveal the effect of acteoside on gouty arthritis(GA) in rats based on liver metabolomics. The ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry(UPLC-Q-TOF-MS) was employed to search for the potential biomarkers and metabolic pathways. SD rats were randomly assigned into blank, model, colchicine(0.3 mg·kg~(-1)), and high-, medium-, low-dose(200, 100, and 50 mg·kg~(-1), respectively) acteoside groups(n=7). The rats were administrated once a day for 7 continuous days. Monosodium urate(MSU) was used to induce GA model in rats during administration. The degree of joint swelling and pathological changes of synovial tissue in rats were observed, and the levels of interleukin(IL)-1ß, IL-18 and tumor necrosis factor(TNF)-α in the synovial tissue of rats were measured. UPLC-Q-TOF-MS was employed to collect rat liver data, and Progenesis QI and EZ info were used for data analysis. Human Metabolomics Database(HMDB) and Kyoto Encyclopedia of Genes and Genomes(KEGG) were employed to predict the potential biomarkers and metabolic pathways. The results showed that acteoside alleviated joint swelling, reduced synovial tissue damage, and lowered the levels of inflammatory cytokines in GA rats. A total of 19 common biomarkers were identified, 17 of which can be regulated by acteoside. Seven metabolic pathways were enriched, such as glycerophospholipid metabolism, linoleic acid metabolism, and taurine and hypotaurine metabolism, among which glycerophospholipid metabolism was strongly disturbed. The metabolomics analysis suggested that acteoside may down-regulate the expression of inflammatory cytokines and alleviate the symptoms of GA rats by regulating glycerophospholipid metabolism, linoleic acid metabolism, and taurine and hypotaurine metabolism. The findings provide a reference for future research and development of acteoside.


Sujet(s)
Goutte articulaire , Glucosides , Polyphénols , Taurine/analogues et dérivés , Humains , Rats , Animaux , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Acide linoléique , Rat Sprague-Dawley , Métabolomique , Foie/métabolisme , Cytokines , Marqueurs biologiques/métabolisme , Glycérophospholipides , Chromatographie en phase liquide à haute performance
9.
Int Immunopharmacol ; 129: 111656, 2024 Mar 10.
Article de Anglais | MEDLINE | ID: mdl-38340422

RÉSUMÉ

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.


Sujet(s)
Goutte articulaire , Glucosides , Tanins hydrolysables , Inflammasomes , Souris , Animaux , Inflammasomes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Goutte articulaire/induit chimiquement , Pyroptose , Nigéricine/pharmacologie , Macrophages , Anti-inflammatoires/effets indésirables , Adénosine triphosphate/métabolisme , Caspases/métabolisme , Interleukine-1 bêta/métabolisme
10.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 1015-1023, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-37555854

RÉSUMÉ

Evodiamine (EVO) was tested on acute gouty arthritis rats to investigate its anti-inflammatory effect. Seventy-two male Sprague-Dawley (SD) rats were randomly assigned into the control, model, high, medium, and low dose of EVO groups and colchicine group. The ankle swelling degrees were measured at 2 h, 6 h, and 24 h following sodium urate injection into ankle joint. Histopathological examination was performed 24 h after injection. Reactive oxygen species (ROS) content in the ankle joint was detected using chemical fluorescence. Serum interleukin-1ß (IL-1ß), interleukin-18 (IL-18), and tumor necrosis factor-α (TNF-α) content were determined by ELISA. Serum xanthine oxidase (XOD), superoxide dismutase (SOD), and malondialdehyde (MDA) were determined by spectrophotometry. The expressions of thioredoxin-interacting protein (TXNIP), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), pro-caspase-1, caspase-1, and apoptosis-related spot like protein (ASC) in synovium were detected by Western blot. Evodiamine alleviated the ankle swelling of the affected foot in gouty arthritis rats and reduced inflammatory cell infiltration in joint synovial tissue. Evodiamine also decreased the content of serum inflammatory factors including IL-1ß, IL-18, and TNF-α, and increased serum SOD activity, while it decreased serum XOD, MDA activity, and ROS level. Moreover, evodiamine downregulated the protein expression levels of TXNIP, NLRP3, pro-caspase-1, cleaved caspae-1, and ASC. The mechanism of EVO in treating gouty arthritis is associated with the inhibition of NLRP3 inflammasome by regulating the ROS/TXNIP/NLRP3 signaling pathway.


Sujet(s)
Goutte articulaire , Protéine-3 de la famille des NLR contenant un domaine pyrine , Quinazolines , Rats , Mâle , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Goutte articulaire/métabolisme , Interleukine-18/usage thérapeutique , Espèces réactives de l'oxygène/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Caspase-1/métabolisme , Rat Sprague-Dawley , Superoxide dismutase , Protéines du cycle cellulaire/usage thérapeutique
11.
Curr Pharm Biotechnol ; 25(3): 350-364, 2024.
Article de Anglais | MEDLINE | ID: mdl-37078349

RÉSUMÉ

BACKGROUND: Gouty arthritis (GA) is a common form of inflammatory arthritis caused by intra-articular deposition of monosodium urate (MSU) crystals; however, there is a tremendous lack of safe and effective therapy in the clinic. OBJECTIVE: The goal of this work was to investigate a novel leflunomide analogue, N-(2,4- dihydroxyphenyl)-5-methyl-1,2-oxazole-3-carboxamide (UTLOH-4e), for its potential to prevent/ treat gouty arthritis. METHODS: In this study, the anti-inflammatory activity of UTLOH-4e was evaluated by MSUinduced GA model in vivo and in vitro, and the molecular docking test was applied to estimate the affinity of UTLOH-4e/UTL-5g/b for MAPKs, NF-κB, and NLRP3. RESULTS: In vitro, UTLOH-4e (1~100 µM) treatment inhibited the inflammatory reaction with no obvious cytotoxicity in PMA-induced THP-1 macrophages exposed to MSU crystals for 24 h, involving the prominent decreased production and gene expression of IL-1ß, TNF-α, and IL-6. Western blot analyses demonstrated that UTLOH-4e (1~100 µM) significantly suppressed the activation of NLRP3 inflammasomes, NF-κB, and MAPK pathways. Furthermore, the data from the experiment on gouty rats induced by intra-articular injection of MSU crystal confirmed that UTLOH-4e markedly ameliorated rat paw swelling, articular synovium inflammation and reduced the concentration of IL-1ß and TNF-α in serum through down-regulating NLRP3 protein expression. CONCLUSION: These results manifested that UTLOH-4e ameliorates GA induced by MSU crystals, which contributes to the modulation of NF-κB/ NLRP3 signaling pathway, suggesting that UTLOH- 4e is a promising and potent drug candidate for the prevention and treatment of gouty arthritis.


Sujet(s)
Goutte articulaire , Rats , Animaux , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Goutte articulaire/métabolisme , Acide urique/effets indésirables , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Léflunomide/effets indésirables , Facteur de nécrose tumorale alpha/métabolisme , Simulation de docking moléculaire , Inflammation/métabolisme , Inflammasomes/métabolisme , Transduction du signal
12.
J Ethnopharmacol ; 319(Pt 3): 117313, 2024 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-37924998

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: DaiTongXiao (DTX) is a traditional Chinese Dai folk formulation utilized for gouty arthritis treatment, with substantial evidence supporting its anti-inflammatory properties. The NLRP3 inflammasome disorder is tightly linked to the development of many inflammatory diseases. AIM OF THE STUDY: To elucidate the therapeutic efficacy of DTX in gouty arthritis and reveal its potential underlying mechanism. MATERIALS AND METHODS: The primary active constituents in DTX were determined through ultraviolet spectrophotometry and gas chromatography. Rats underwent induction with monosodium urate (MSU), followed by treatment of J774A.1 cells with adenosine triphosphate (ATP) activation and lipopolysaccharide (LPS) induction and the subsequent culture in Dulbecco's modified Eagle's medium. The degree of foot joint swelling in rats was assessed, and ankle joints were evaluated through H&E staining. Enzyme-linked immunosorbent assay was performed to measure the levels of interleukin (IL)-1ß, IL-6, IL-8, and tumor necrosis factor (TNF)-α in both serum and cells. Reverse transcription-polymerase chain reaction (RT-PCR) was performed to determine the relative mRNA expression levels of NLRP3, ASC, Caspase-1, and NF-κB in J774A.1 macrophages. The expression of NLRP3, ASC, Caspase-1, and NF-κB was examined by western blotting. RESULTS: DTX could alleviate MSU-induced joint swelling in rats, as evidenced by a reduction in joint inflammation. Moreover, DTX effectively enhanced the survival rate of J774A.1 cells following LPS induction and ATP activation. Furthermore, DTX significantly reduced IL-1ß, IL-6, IL-8, and TNF-α levels in both cell culture medium and rat serum. RT-PCR results revealed that DTX notably downregulated the mRNA expression levels of NLRP3, ASC, Caspase-1, and NF-κB in J774A.1 cells. Additionally, DTX downregulated NLRP3, ASC, NF-κB, and Caspase-1 expression in the joint tissue. CONCLUSIONS: DTX exerts a significant anti-gouty arthritis effect, with its mechanism being tightly linked to the NLRP3 inflammatory signaling pathway. This pathway may be modulated by inhibiting IL-1ß differentiation and maturation by downregulating NLRP3, ASC, Caspase-1, and NF-κB protein expression. This, in turn, leads to a reduction in the release of IL-6, IL-8, and TNF-α, ultimately impeding gouty arthritis progression.


Sujet(s)
Goutte articulaire , Rats , Animaux , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Goutte articulaire/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6 , Lipopolysaccharides , Interleukine-8 , Transduction du signal , Inflammasomes/métabolisme , Acide urique , Caspase-1/métabolisme , Oedème , Adénosine triphosphate , ARN messager
13.
RMD Open ; 9(4)2023 11.
Article de Anglais | MEDLINE | ID: mdl-37973536

RÉSUMÉ

OBJECTIVES: Gout, as the most prevalent form of inflammatory arthritis, necessitates the use of animal models to investigate the molecular mechanisms involved in its development. Therefore, our objective was to develop a novel chronic mouse model of gout that more closely mimics the progression of gout in humans. METHODS: A novel chronic mouse model of gout was established by a simple method, which does not require high technical proficiency, predominantly involves daily intraperitoneal injections of potassium oxonate for approximately 4 months, combined with a high fat-diet and injections of acetic acid into the hind paws to facilitate the formation of monosodium urate (MSU). Arthritis scores and paw oedema were assessed, behavioural tests were conducted, and histopathological and imaging evaluations of the arthritic paw joints were performed. RESULTS: After 4 months of induction, mice in the model group exhibited noticeable increases in arthritis severity, joint and cartilage damage, as well as bone erosion. Gomori's methenamine silver stain revealed the presence of MSU crystal deposition or tophi in the paw joints or ankle joints of up to 37.9% of the model mice (11 out of 29 mice). Moreover, treatment with benzbromarone effectively prevented the further development of gout or tophi formation in model mice. CONCLUSIONS: Our model more accurately replicates the pathological features of gouty arthritis compared with gout induced by MSU crystal injections. Therefore, it is particularly suitable for further investigations into the pathogenesis of gout and also serves as a valuable platform for screening potential antigout agents.


Sujet(s)
Goutte articulaire , Goutte , Humains , Souris , Animaux , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Goutte articulaire/anatomopathologie , Goutte/traitement médicamenteux , Acide urique , Antigoutteux/effets indésirables , Modèles animaux de maladie humaine
14.
Einstein (Sao Paulo) ; 21: eAO0465, 2023.
Article de Anglais | MEDLINE | ID: mdl-37909651

RÉSUMÉ

OBJECTIVE: Gouty arthritis is characterized by painful inflammation due to the deposition of monosodium urate crystals in joint tissues. Despite available treatments, many patients experience ineffective management and adverse effects. This study evaluated a manual therapy protocol involving passive joint mobilization at the peak of inflammation in a gouty arthritis model using functional and inflammatory parameters. METHODS: Twenty male Wistar rats, 12 weeks old, were divided into two groups (n=10 each): Gouty Arthritis and Control Groups, which were further subdivided into treated and untreated groups (n=5 each). The Gouty Arthritis Group received intraarticular knee injection of 50µL of monosodium urate crystals, while the Control Group received 50µL of phosphate buffered saline. The treatment involved a 9-minutes session of grade III joint mobilization (according to Maitland). Nociception, grip strength, and edema were evaluated before induction (EV0), 7 hours after assessment (EV1), immediately after treatment (EV2), and 1 hour after treatment (EV3). The animals were euthanized, and synovial fluid was collected to analyze leukocyte migration. RESULTS: The model mimicked the signs of the Gouty Arthritis Group, with a decrease in the threshold of nociception and strength and an increase in edema and leukocyte count. The mobilization protocol significantly increased the nociceptive threshold and grip strength and reduced edema; however, it did not reverse the increase in leukocyte count. CONCLUSION: Our results suggest that mobilization promotes analgesia and may modulate the inflammatory process owing to reduced edema and subtle attenuation of cell migration, which contributes to strength gain.


Sujet(s)
Goutte articulaire , Humains , Rats , Animaux , Mâle , Goutte articulaire/induit chimiquement , Goutte articulaire/thérapie , Goutte articulaire/métabolisme , Acide urique , Rat Wistar , Inflammation , Douleur , Oedème
15.
Int J Rheum Dis ; 26(12): 2478-2488, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37860923

RÉSUMÉ

INTRODUCTION: This study aimed to compare the efficacy of non-loading versus loading low-dose colchicine in patients with acute crystal-associated arthritis. MATERIALS AND METHODS: All in-patients who were admitted to Chiang Mai University Hospital with non-arthritis disease and developed acute crystal-associated arthritis during admission (within 48 h after arthritis onset) were invited to join this study. The patients were randomized into two groups. Patients in Group I (non-loading group) and Group II (loading group) received colchicine at 1.2 and 2.4 mg in the first 24 h, respectively. The primary outcome was the patients' pain response at 24 h after treatment. RESULTS: Of 80 patients, 49 were acute gouty arthritis, and 31 acute calcium pyrophosphate (CPP) arthritis. The mean [95% CI] pain score was no different between Groups I and II at the baseline level (6.46[5.72-7.19] vs. 6.654[5.85-7.44], p = .867) and at 24 h (3.13[2.43-3.82] vs. 3.18[2.42-3.93], p = .907). The proportion of patients with ≥50% pain reduction was not different (57.50% vs. 55.00%, p = .822). Sensitivity analysis among patients with a baseline pain score of ≥4 showed the same pattern of response. Mild diarrhea was common and comparable in both groups. Subgroup analysis according to renal function (eGFR < 60 vs. ≥60 mL/min/1.73 m2 ) or type of crystals (acute gouty arthritis vs. acute CPP arthritis) also showed the same pattern of response. CONCLUSION: Non-loading low-dose colchicine was as effective as loading low-dose colchicine in patients with acute crystal-associated arthritis, regardless of renal function or type of crystals.


Sujet(s)
Goutte articulaire , Colchicine , Humains , Goutte articulaire/diagnostic , Goutte articulaire/traitement médicamenteux , Goutte articulaire/induit chimiquement , Colchicine/effets indésirables , Douleur/induit chimiquement , Plan de recherche , Méthode en double aveugle
16.
Front Endocrinol (Lausanne) ; 14: 1071630, 2023.
Article de Anglais | MEDLINE | ID: mdl-37810893

RÉSUMÉ

Neutrophil extracellular traps (NETs) are composed of chromatin filaments coated with granular and cytosolic proteins, which contribute to the pathogenesis and progression of immune-related diseases. NETs are frequently observed in gouty arthritis, but the related mechanisms remain poorly understood. The aim of our study was to systematically elucidate the molecular mechanisms of self-remitting effects in gouty arthritis, and the causative relationship between neutrophil autophagy and NETs. The air pouch and paw edema model were used to simulate gouty arthritis in mice. Neutrophil infiltration and the formation of NETs were found in gouty arthritis. Interestingly, monosodium urate (MSU) crystals could induce the formation of NETs, degrade inflammatory factors, and alleviate the inflammatory response in gouty arthritis. In addition, MSU crystals resulted in profound molecular alterations in neutrophils using RNA-seq analysis, including autophagy activation. MSU crystals could activate neutrophil autophagy in vitro, and autophagy activators and inhibitors could regulate the formation of NETs. Furthermore, we explored the mechanism of autophagy-induced NETs. Autophagy related protein 7 (ATG7) produced by neutrophils stimulated with MSU crystals worked synergistically with p53 to enter the nucleus, promoting peptidyl arginine deiminase 4 (PAD4) expression, and inducing the formation of NETs. Finally, we substantiated that neutrophil autophagy regulates the severity of gouty arthritis via the formation of NETs in PAD4 -/- mice. Our results indicated that the autophagy of neutrophils regulates the formation of NETs and degrades inflammatory factors. Regulating autophagy and interfering with the formation of NETs represents a potential therapeutic approach against gouty arthritis during clinical practice.


Sujet(s)
Goutte articulaire , Pièges extracellulaires , Souris , Animaux , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Goutte articulaire/métabolisme , Granulocytes neutrophiles/anatomopathologie , Pièges extracellulaires/métabolisme , Acide urique , Inflammation/anatomopathologie
17.
Int J Rheum Dis ; 26(11): 2233-2239, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37715329

RÉSUMÉ

AIM: According to reports, long non-coding RNAs (lncRNAs) are involved in the regulation of many inflammatory diseases. Here, our main purpose was to ascertain the expression data of lncRNA SNHG14 in acute gouty arthritis (AGA) and to explore its possible mechanism in the regulation of AGA. METHOD: Reverse transcription quantitative polymerase chain reaction technology was supplied to detect the lncRNA SNHG14 expression. A receiver operating characteristics curve was drawn to estimate the accuracy of lncRNA SNHG14 in AGA diagnosis. An in vitro AGA cell model was constructed by inducing THP-1 cells with monosodium urate (MSU). The concentrations of inflammatory factors such as interleukin-1ß, interleukin-6, and tumor necrosis factor-α were measured by enzyme-linked immunosorbent assay. The luciferase reporter gene was used to verify the relationship between lncRNA SNHG14 and miR-223-3p. RESULTS: In clinical analysis, the levels of serum lncRNA SNHG14 in AGA patients were significantly higher than those in the control group. Abnormally elevated lncRNA SNHG14 has high sensitivity and specificity for AGA diagnosis. In in vitro cell experiments, silencing lncRNA SNHG14 inhibited the inflammatory response of THP-1 cells stimulated by MSU, and the luciferase reporter gene proved that lncRNA SNHG14 could bind to miR-223-3p. In addition, the level of miR-223-3p declined in AGA patients and the AGA cell model. Overexpression of miR-223-3p is helpful to alleviate an MSU-induced inflammatory response. CONCLUSION: In the AGA cell model, lncRNA SNHG14, as an miR-223-3p sponge, induces a cellular inflammatory response by controlling the level of miR-223-3p, so aggravating the disease progress of AGA.


Sujet(s)
Goutte articulaire , microARN , ARN long non codant , Humains , Goutte articulaire/induit chimiquement , Goutte articulaire/génétique , Goutte articulaire/métabolisme , ARN long non codant/génétique , microARN/génétique , microARN/métabolisme , Acide urique , Luciferases
18.
Cell Mol Biol (Noisy-le-grand) ; 69(7): 28-34, 2023 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-37715438

RÉSUMÉ

Gouty arthritis is an acute and chronic joint inflammatory joint disease characterized by the deposition of monosodium urate (MSU) crystals in joints and periarticular tissues. Resveratrol (3, 5, 4-trihydroxy-trans-stilbene, RV), a natural polyphenolic compound, has anti-inflammatory and antioxidant properties. The purpose of this study was to investigate the effect of resveratrol on rats with gouty arthritis and its molecular mechanism. THP-1-derived macrophages were induced by lipopolysaccharide (LPS) and MSU to create an in vitro gout cell inflammation model, and rats were injected with MSU crystals into the right ankle joint for an in vivo acute gouty arthritis model. We investigated the anti-inflammatory properties of resveratrol using these in vitro and in vitro models. Our findings suggested that resveratrol effectively reduced ankle swelling and synovial inflammation in a dose-dependent manner in rats with acute gouty arthritis, with almost the same effect as colchicine treatment. In MSU-treated THP-1-derived macrophages, resveratrol inhibited NLRP3 inflammasome activation and IL-1ß secretion. Furthermore, resveratrol and the HIF-1α inhibitor PX478 both inhibited the expression of the NLRP3 inflammasome, IL-1ß, and HIF-1α. This study demonstrated that resveratrol significantly improved the symptoms of acute gouty arthritis and its potential mechanism may be IL-1ß reduction via HIF-1α modulation and inhibition of NLRP3 inflammasome activation. Our study might offer a novel sight for the treatment of gouty arthritis.


Sujet(s)
Goutte articulaire , Resvératrol , Animaux , Rats , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Inflammasomes , Inflammation , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Resvératrol/pharmacologie , Resvératrol/usage thérapeutique
19.
Reumatismo ; 75(3)2023 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-37721349

RÉSUMÉ

Acute gouty arthritis is a recognized complication of hyperuricemia and one of the most common forms of inflammatory arthritis in adults. Drug-induced hyperuricemia is increasingly prevalent in clinical practice. Diuretics, antitubercular medications, and immunosuppressants are the common drugs associated with hyperuricemia. Oral isotretinoin is the drug of choice for different forms of severe acne and is rarely associated with hyperuricemia. We present the case of a 30-year-old male with severe acne vulgaris who was prescribed isotretinoin and later presented with acute gout. The patient developed hyperuricemia and swelling of the right first metatarsophalangeal joint within two months of isotretinoin commencement. There was a second episode of similar joint swelling three months later, parallel to the isotretinoin rechallenge. The dose of isotretinoin was reduced with the addition of febuxostat. The patient did not develop further episodes and remained symptom-free without urate-lowering therapy.


Sujet(s)
Acné juvénile , Goutte articulaire , Hyperuricémie , Adulte , Humains , Mâle , Goutte articulaire/induit chimiquement , Goutte articulaire/traitement médicamenteux , Isotrétinoïne/effets indésirables , Hyperuricémie/induit chimiquement , Hyperuricémie/traitement médicamenteux , Acné juvénile/traitement médicamenteux , Fébuxostat/usage thérapeutique
20.
J Nat Prod ; 86(9): 2091-2101, 2023 09 22.
Article de Anglais | MEDLINE | ID: mdl-37625387

RÉSUMÉ

In the present study, a natural product database of compounds associated with herbs traditionally verified to treat gout/hyperuricemia/arthritis was constructed. 3D-shape and docking-based virtual screening was conducted. To identify potential xanthine oxidase (XOD) inhibitors in the database, eight compounds with commercial availability were identified as high 3D-shape similarity with febuxostat (1), a known XOD inhibitor. Docking was used to further predict the possible interactions between XOD and these compounds. Moracin C (2), moracin D (3), and isoformononetin (8) exhibited higher docking scores and binding energies than other compounds. In vitro, 2 inhibited XOD with an IC50 value of 0.25 ± 0.14 µM, which is similar to that of 1 (0.16 ± 0.08 µM). In a hyperuricemic mouse model, 5-20 mg/kg 2 exhibited satisfying urate-lowering and XOD inhibitory effects. Compound 2 also exhibited antiarthritis activities. In RAW264.7 cells, 2 at 1-10 µM inhibited the expression of IL-1ß and TNF-α induced by MSU. In an acute gouty arthritis model in SD rats, 5-20 mg/kg 2 significantly alleviated the toe swelling, inflammatory response, and dysfunction disorder caused by monosodium urate (MSU). Compound 2 inhibited serum IL-1ß and TNF-α cytokines as well as reduced the expression of the NLRP3/ASC/caspase-1 inflammasome in joints. In summary, 2 was an effective compound for the treatment of hyperuricemia/gouty arthritis.


Sujet(s)
Goutte articulaire , Hyperuricémie , Souris , Rats , Animaux , Hyperuricémie/traitement médicamenteux , Hyperuricémie/métabolisme , Facteur de nécrose tumorale alpha , Rat Sprague-Dawley , Goutte articulaire/traitement médicamenteux , Goutte articulaire/induit chimiquement , Goutte articulaire/métabolisme , Acide urique/effets indésirables , Antienzymes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...