Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Inflamm Res ; 73(7): 1173-1184, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38739197

RESUMEN

OBJECTIVE AND DESIGN: To elucidate Sirt1's role in gouty arthritis inflammation and its potential mechanisms. MATERIAL: Constructed murine models of gouty arthritis and conducted THP-1 cell experiments. TREATMENT: 1 mg of MSU crystals injected into mice ankle joints for a 72-h intervention. After a 3-h pre-treatment with Sirt1-specific inhibitor (EX527) and agonist (SRT2104), inflammation was induced for 21 h using lipopolysaccharide (LPS) plus MSU crystals. METHODS: We assessed gouty arthritis severity through joint inflammation index, swelling, and hematoxylin and eosin (H&E) staining, and measured CD68 mononuclear macrophages and Sirt1 expression in synovial tissue via immunohistochemistry. ELISA, NO assay, RT-qPCR, Flow cytometry, and Western blot were utilized to examine macrophage inflammatory factors, polarization, reactive oxygen species(ROS), MAPK/NF-κB/AP-1 and Nrf2/HO-1 pathways proteins. RESULTS: Significant joint swelling, synovial tissue edema, and inflammatory cell infiltration were observed. CD68 mononuclear macrophages and Sirt1 expression were elevated in synovium. Sirt1 activation decreased inflammatory factors, M1 polarization, and ROS generation. Sirt1 activation reduced p38/JNK phosphorylation, thereby inhibiting downstream NF-κB p65/AP-1 and enhancing Nrf2/HO-1, thus suppressing inflammation. CONCLUSIONS: Sirt1 alleviates M1 macrophage polarization and inflammation in gouty arthritis by inhibiting the MAPK/NF-κB/AP-1 pathway and activating the Nrf2/HO-1 pathway. Thus, activating Sirt1 may provide a new therapeutic target for gouty arthritis.


Asunto(s)
Artritis Gotosa , Hemo-Oxigenasa 1 , Macrófagos , Factor 2 Relacionado con NF-E2 , FN-kappa B , Sirtuina 1 , Factor de Transcripción AP-1 , Animales , Artritis Gotosa/tratamiento farmacológico , Artritis Gotosa/metabolismo , Artritis Gotosa/inmunología , Sirtuina 1/metabolismo , Sirtuina 1/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/inmunología , Factor 2 Relacionado con NF-E2/metabolismo , Humanos , Masculino , FN-kappa B/metabolismo , Hemo-Oxigenasa 1/metabolismo , Ratones , Factor de Transcripción AP-1/metabolismo , Células THP-1 , Ratones Endogámicos C57BL , Inflamación , Transducción de Señal/efectos de los fármacos , Lipopolisacáridos/farmacología , Carbazoles , Proteínas de la Membrana
2.
Immunol Rev ; 294(1): 92-105, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31853991

RESUMEN

Trained immunity is a process in which innate immune cells undergo functional reprogramming in response to pathogens or damage-associated molecules leading to an enhanced non-specific immune response to subsequent stimulation. While this capacity to respond more strongly to stimuli is beneficial for host defense, in some circumstances it can lead to maladaptive programming and chronic inflammation. Gout is characterized by persistent low-grade inflammation and is associated with an increased number of comorbidities. Hyperuricemia is the main risk factor for gout and is linked to the development of comorbidities. Several experimental studies have shown that urate can mechanistically alter the inflammatory capacity of myeloid cells, while observational studies have indicated an association of hyperuricemia to a wide spectrum of common adult inflammatory diseases. In this review, we argue that hyperuricemia is a main culprit in the development of the long-term systemic inflammation seen in gout. We revisit existing evidence for urate-induced transcriptional and epigenetic reprogramming that could lead to an altered functional state of circulating monocytes consisting in enhanced responsiveness and maladaptive immune responses. By discussing specific functional adaptations of monocytes and macrophages induced by soluble urate or monosodium urate crystals and their contribution to inflammation in vitro and in vivo, we further enforce that urate is a metabolite that can induce innate immune memory and we discuss future research and possible new therapeutic approaches for gout and its comorbidities.


Asunto(s)
Artritis Gotosa/inmunología , Hiperuricemia/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Ácido Úrico/metabolismo , Animales , Reprogramación Celular , Epigénesis Genética , Humanos , Inmunidad Innata , Memoria Inmunológica
3.
J Immunol ; 205(3): 789-800, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32561569

RESUMEN

Although monosodium urate (MSU) crystals are known to trigger inflammation, published data on soluble uric acid (sUA) in this context are discrepant. We hypothesized that diverse sUA preparation methods account for this discrepancy and that an animal model with clinically relevant levels of asymptomatic hyperuricemia and gouty arthritis can ultimately clarify this issue. To test this, we cultured human monocytes with different sUA preparation solutions and found that solubilizing uric acid (UA) by prewarming created erroneous results because of UA microcrystal contaminants triggering IL-1ß release. Solubilizing UA with NaOH avoided this artifact, and this microcrystal-free preparation suppressed LPS- or MSU crystal-induced monocyte activation, a process depending on the intracellular uptake of sUA via the urate transporter SLC2A9/GLUT9. CD14+ monocytes isolated from hyperuricemic patients were less responsive to inflammatory stimuli compared with monocytes from healthy individuals. Treatment with plasma from hyperuricemic patients impaired the inflammatory function of CD14+ monocytes, an effect fully reversible by removing sUA from hyperuricemic plasma. Moreover, Alb-creERT2;Glut9 lox/lox mice with hyperuricemia (serum UA of 9-11 mg/dl) showed a suppressed inflammatory response to MSU crystals compared with Glut9 lox/lox controls without hyperuricemia. Taken together, we unravel a technical explanation for discrepancies in the published literature on immune effects of sUA and identify hyperuricemia as an intrinsic suppressor of innate immunity, in which sUA modulates the capacity of monocytes to respond to danger signals. Thus, sUA is not only a substrate for the formation of MSU crystals but also an intrinsic inhibitor of MSU crystal-induced tissue inflammation.


Asunto(s)
Artritis Gotosa/inmunología , Hiperuricemia/inmunología , Monocitos/inmunología , Ácido Úrico/toxicidad , Animales , Artritis Gotosa/inducido químicamente , Artritis Gotosa/genética , Artritis Gotosa/patología , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/inmunología , Humanos , Hiperuricemia/inducido químicamente , Hiperuricemia/genética , Hiperuricemia/patología , Inflamación , Ratones , Ratones Transgénicos , Monocitos/patología , Solubilidad
4.
Ann Rheum Dis ; 80(12): 1604-1614, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34663597

RESUMEN

Crystal structures activate innate immune cells, especially macrophages and initiate inflammatory responses. We aimed to understand the role of the mechanosensitive TRPV4 channel in crystal-induced inflammation. Real-time RT-PCR, RNAscope in situ hybridisation, and Trpv4eGFP mice were used to examine TRPV4 expression and whole-cell patch-clamp recording and live-cell Ca2+ imaging were used to study TRPV4 function in mouse synovial macrophages and human peripheral blood mononuclear cells (PBMCs). Both genetic deletion and pharmacological inhibition approaches were used to investigate the role of TRPV4 in NLRP3 inflammasome activation induced by diverse crystals in vitro and in mouse models of crystal-induced pain and inflammation in vivo. TRPV4 was functionally expressed by synovial macrophages and human PBMCs and TRPV4 expression was upregulated by stimulation with monosodium urate (MSU) crystals and in human PBMCs from patients with acute gout flares. MSU crystal-induced gouty arthritis were significantly reduced by either genetic ablation or pharmacological inhibition of TRPV4 function. Mechanistically, TRPV4 mediated the activation of NLRP3 inflammasome by diverse crystalline materials but not non-crystalline NLRP3 inflammasome activators, driving the production of inflammatory cytokine interleukin-1ß which elicited TRPV4-dependent inflammatory responses in vivo. Moreover, chemical ablation of the TRPV1-expressing nociceptors significantly attenuated the MSU crystal-induced gouty arthritis. In conclusion, TRPV4 is a common mediator of inflammatory responses induced by diverse crystals through NLRP3 inflammasome activation in macrophages. TRPV4-expressing resident macrophages are critically involved in MSU crystal-induced gouty arthritis. A neuroimmune interaction between the TRPV1-expressing nociceptors and the TRPV4-expressing synovial macrophages contributes to the generation of acute gout flares.


Asunto(s)
Artralgia/metabolismo , Artritis/metabolismo , Artropatías por Depósito de Cristales/metabolismo , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Nociceptores/metabolismo , Canales Catiónicos TRPV/genética , Adulto , Animales , Artralgia/inmunología , Artritis/inmunología , Artritis Gotosa/inmunología , Artritis Gotosa/metabolismo , Artropatías por Depósito de Cristales/inmunología , Gota/inmunología , Gota/metabolismo , Humanos , Inflamasomas/inmunología , Inflamación , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Imagen Óptica , Técnicas de Placa-Clamp , Membrana Sinovial/citología , Células THP-1 , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/metabolismo , Ácido Úrico
5.
Mol Pharm ; 18(2): 667-678, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32579365

RESUMEN

Gasdermin D (GSDMD) plays a causal role in NOD-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis eruption, which has been regarded as a potential therapeutic target for pyroptosis-related diseases including acute gouty arthritis. In the present study, the synthesized PEI-Chol (cholesterol grafted polyethylenimine) was assembled with GSDMD small interfering RNA (siRNA) to form PEI-Chol/siGSDMD polyplexes, which provided high transfection efficiency for siRNA-mediated GSDMD knockdown. Then we evaluated the effect of GSDMD siRNA-loaded PEI-Chol on inflammatory cascades in bone-marrow-derived macrophages (BMDMs) and acute gouty arthritis animal models under MSU exposure. When accompanied by pyroptosis blockade and decreased release of interleukin-1 beta (IL-1ß), NLRP3 inflammasome activation was also suppressed by GSDMD knockdown in vivo and in vitro. Moreover, in MSU-induced acute gouty arthritis mice, blocking GSDMD with siRNA significantly improved ankle swelling and inflammatory infiltration observed in histopathological analysis. Furthermore, investigation using a mouse air pouch model verified the effect of siGSDMD-loaded PEI-Chol on pyroptosis of recruited macrophages and related signaling pathways in response to MSU. These novel findings exhibited that GSDMD knockdown relieved acute gouty arthritis through inhibiting pyroptosis, providing a possible therapeutic approach for MSU-induced acute gouty arthritis molecular therapy using PEI-Chol as a nucleic acid delivery carrier.


Asunto(s)
Artritis Gotosa/tratamiento farmacológico , Portadores de Fármacos/química , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas de Unión a Fosfato/antagonistas & inhibidores , Piroptosis/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Gotosa/inducido químicamente , Artritis Gotosa/inmunología , Artritis Gotosa/patología , Células Cultivadas , Colesterol , Técnicas de Silenciamiento del Gen/métodos , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Polietileneimina/química , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Ácido Úrico/administración & dosificación , Ácido Úrico/toxicidad
6.
J Immunol ; 202(6): 1807-1814, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30718300

RESUMEN

The purpose of this study was to investigate the role of pentraxin 3 (PTX3), a pivotal component of the innate immune system, in gout. Levels of PTX3 and IL-1ß in human samples were evaluated by ELISA. Development of murine gout was evaluated through the levels of cytokines (PTX3, CXCL1, and IL-1ß) and neutrophil recruitment into the joint cavity. Phagocytosis of monosodium urate (MSU) crystals and caspase-1 activation were determined by flow cytometer. Acute gout patients showed elevated concentration of PTX3 in plasma and synovial fluid as compared with healthy and osteoarthritic subjects. Moreover, there was a positive correlation between intra-articular PTX3 and IL-1ß levels. PTX3 was induced in the periarticular tissue of mice postinjection of MSU crystals. Importantly, Ptx3-deficient mice showed reduced inflammation in response to MSU crystal injection compared with wild-type mice, including reduction of neutrophil recruitment into the joint cavity and IL-1ß and CXCL1 production. Interestingly, addition of PTX3 in vitro enhanced MSU crystal phagocytosis by monocytes and resulted in higher production of IL-1ß by macrophages. This contribution of PTX3 to the phagocytosis of MSU crystals and consequent production of IL-1ß occurred through a mechanism mainly dependent on FcγRIII. Thus, our results suggest that PTX3 acts as a humoral pattern recognition molecule in gout facilitating MSU crystal phagocytosis and contributing to the pathogenesis of gouty arthritis.


Asunto(s)
Artritis Gotosa/inmunología , Proteína C-Reactiva/inmunología , Interleucina-1beta/inmunología , Fagocitosis/inmunología , Componente Amiloide P Sérico/inmunología , Ácido Úrico/inmunología , Animales , Artritis Gotosa/metabolismo , Artritis Gotosa/patología , Proteína C-Reactiva/metabolismo , Humanos , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Componente Amiloide P Sérico/metabolismo , Ácido Úrico/metabolismo
7.
J Immunol ; 202(9): 2728-2736, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30926645

RESUMEN

Gouty arthritis is an inflammatory disease that is triggered by abnormal uric acid metabolism, which is usually attributed to obesity, a risk factor of hyperuricemia and gout attack. A high level of leptin in plasma is a marker of individuals with obesity. Population studies show that leptin promotes obesity-related arthritis, such as osteoarthritis, but it is unknown whether leptin contributes to gouty arthritis, another form of obesity-related arthritis. Our present study showed that the levels of leptin and leptin receptor in patients with active gouty arthritis were elevated. Leptin facilitates the stimulation of human synoviocytes, mouse peritoneal macrophages, and HL-60 cells induced by monosodium urate, leading to higher levels of acute gout-related proinflammatory factors. Leptin obviously exacerbates the inflammation of monosodium urate-induced acute gouty arthritis in wild-type mice, whereas that in leptin-deficient C57BL6/Job/ob mice is markedly alleviated. The proinflammatory effect of leptin in acute gouty arthritis is partly mediated by mTORC1 signaling pathway. Our study reveals that leptin may serve as a novel prevention and treatment target in acute gouty arthritis.


Asunto(s)
Artritis Gotosa/inmunología , Leptina/inmunología , Sinoviocitos/inmunología , Ácido Úrico/toxicidad , Animales , Artritis Gotosa/tratamiento farmacológico , Artritis Gotosa/patología , Modelos Animales de Enfermedad , Femenino , Células HL-60 , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Leptina/efectos adversos , Leptina/farmacología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sinoviocitos/patología
8.
Inflammopharmacology ; 29(1): 35-47, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32918702

RESUMEN

BACKGROUND: Targeted anti-IL-1ß therapy may be a valuable option for the management of gouty arthritis. The present meta-analysis has evaluated the effect of canakinumab, an anti-IL-1ß monoclonal antibody in gouty arthritis. METHODS: A standard meta-analysis protocol was developed and after performing a comprehensive literature search in MEDLINE, Cochrane, and International Clinical Trial Registry Platform (ICTRP), reviewers assessed eligibility and extracted data from three relevant articles. A random-effects model was used to estimate the pooled effect size as the mean difference in Visual Analouge Scale (VAS) score, serum hsCRP, serum Amyloid A, and risk ratio for global assessment between the groups. Quality assessment was done using the risk of bias assessment tool and summary of findings was prepared using standard Cochrane methodology with GradePro GDT. RESULTS: Treatment with canakinumab showed a mean reduction of VAS score by 14.59 mm [95% CI - 19.42 to - 9.77], serum hsCRP by 15.36 mg/L [95% CI 1.62-29.11], serum Amyloid A by 67.18 mg/L [95% CI 17.06-117.31], and improvement in patient global assessment (RR = 1.478; 95% CI 1.29-1.67) and physician global assessment (RR = 1.44; 95% CI 1.28-1.61). The probability that future studies may have a mean difference in VAS score less than zero has been calculated to be 27.3% using a cumulative distribution function (CDF) calculator. CONCLUSION: This meta-analysis shows the beneficial effect of canakinumab over triamcinolone by reducing VAS score, serum hsCRP, serum amyloid A, and improvement in global assessments in acute gouty arthritis.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Artritis Gotosa/tratamiento farmacológico , Interleucina-1beta/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Artritis Gotosa/inmunología , Artritis Gotosa/fisiopatología , Proteína C-Reactiva/metabolismo , Humanos , Proteína Amiloide A Sérica/metabolismo , Resultado del Tratamiento , Triamcinolona/farmacología
9.
Mediators Inflamm ; 2020: 8298615, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32774151

RESUMEN

BACKGROUND: Cyr61 is considered a novel proinflammatory factor. Gouty arthritis (GA) is a self-limited inflammatory reaction caused by monosodium urate (MSU) crystals. In this study, we assessed the role of Cyr61 in the inflammatory process of GA. METHODS: We investigated the expression of Cyr61 in MSU-induced rat gout models and MSU-stimulated rat fibroblast-like synovial (FLS) cells. After inhibiting the expression of Cyr61, levels of IL-1ß, TNF-α, and IL-6 were detected by ELISA, qPCR, western blot, and immunohistochemical methods. We probed the downstream NF-κB signaling pathway using the NF-κB inhibitor PDTC, and levels of NF-κB and p-NF-κB were detected by western blot and qPCR. RESULTS: Our results demonstrate that Cyr61 plays a potent role in the formation of local inflammation in vitro and in vivo. Cyr61 was highly expressed in synovial tissues of gout models, and the expression of Cyr61 protein was also significantly increased in MSU-stimulated FLS cells. Cyr61 promoted MSU-induced acute inflammation via the NF-κB signaling pathway. CONCLUSIONS: Our study has revealed that Cyr61 is an important regulatory factor for the initiation of inflammation in GA. The high expression of Cyr61 protein can induce synovial cells to produce many inflammatory cytokines, such as IL-1ß, TNF-α, and IL-6, partly in an NF-κB-dependent manner. Thus, inhibition of Cyr61 could be a new target and strategy for the prevention and treatment of GA.


Asunto(s)
Artritis Gotosa/inmunología , Artritis Gotosa/metabolismo , Proteína 61 Rica en Cisteína/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Animales , Artritis Gotosa/genética , Western Blotting , Proteína 61 Rica en Cisteína/genética , Citocinas/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Técnicas In Vitro , Inflamación/inducido químicamente , Inflamación/genética , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Ácido Úrico/toxicidad
10.
Int J Mol Sci ; 21(24)2020 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371241

RESUMEN

Jakyakgamcho-Tang (JGT) is a traditional medicine used to treat muscular tension, spasm, and pain. Several studies have reported its clinical use as an anti-inflammatory and in gynaecological treatment. This study aimed to compare the anti-inflammatory effects of JGT according to extraction solvent, water (JGTW) and 30% EtOH (JGTE) on lipopolysaccharide (LPS)-stimulated macrophages and in mice with monosodium urate (MSU)-induced gouty arthritis. We evaluated the production of inflammatory mediators and cytokines and the expression of inducible nitric oxide (iNOS) and cyclooxygenase-2 (COX-2) in RAW 264.7 cells. We also examined oedema, pain, and inflammation in MSU-induced mice by measuring affected hind paw swelling, weight-bearing, pro-inflammatory cytokines levels, and myeloperoxidase (MPO) activity. In LPS-stimulated RAW264.7 cells, JGTW and JGTE significantly decreased prostaglandin (PG) E2(PGE2) production via suppressing COX-2 expression and cytokines interleukin-1ß and interleukin-6. Only JGTE reduced the production of NO and cytokines and the mRNA levels of iNOS and cytokines. In MSU-induced mice, JGTE and JGTW efficiently decreased paw swelling and attenuated joint pain. JGTE (200 and 300 mg/kg) effectively suppressed inflammation by downregulating pro-inflammatory cytokines (tumour necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6) and MPO activity, which were only slightly reduced by JGTW. Our data demonstrate the anti-inflammatory activity of JGT in macrophage and gouty arthritis animal models and show that JGTE is more effective than JGTW at lower concentrations.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Gotosa/tratamiento farmacológico , Edema/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Extractos Vegetales/farmacología , Solventes/química , Animales , Artritis Gotosa/inducido químicamente , Artritis Gotosa/inmunología , Artritis Gotosa/patología , Citocinas/metabolismo , Medicamentos Herbarios Chinos/química , Edema/inducido químicamente , Edema/inmunología , Edema/patología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL
11.
J Cell Physiol ; 234(4): 4081-4094, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30370562

RESUMEN

Dysregulation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is involved in many chronic inflammatory diseases, including gouty arthritis. Activation of the NLRP3 inflammasome requires priming and activation signals: the priming signal controls the expression of NLRP3 and interleukin (IL)-1ß precursor (proIL-1ß), while the activation signal leads to the assembly of the NLRP3 inflammasome and to caspase-1 activation. Here, we reported the effects of the alcoholic extract of Taiwanese green propolis (TGP) on the NLRP3 inflammasome in vitro and in vivo. TGP inhibited proIL-1ß expression by reducing nuclear factor kappa B activation and reactive oxygen species (ROS) production in lipopolysaccharide-activated macrophages. Additionally, TGP also suppressed the activation signal by reducing mitochondrial damage, ROS production, lysosomal rupture, c-Jun N-terminal kinases 1/2 phosphorylation and apoptosis-associated speck-like protein oligomerization. Furthermore, we found that TGP inhibited the NLRP3 inflammasome partially via autophagy induction. In the in vivo mouse model of uric acid crystal-induced peritonitis, TGP attenuated the peritoneal recruitment of neutrophils, and the levels of IL-1ß, active caspase-1, IL-6 and monocyte chemoattractant protein-1 in lavage fluids. As a proof of principle, in this study, we purified a known compound, propolin G, from TGP and identified this compound as a potential inhibitor of the NLRP3 inflammasome. Our results indicated that TGP might be useful for ameliorating gouty inflammation via inhibition of the NLRP3 inflammasome.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Gotosa/prevención & control , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Peritonitis/prevención & control , Própolis/farmacología , Animales , Artritis Gotosa/inmunología , Artritis Gotosa/metabolismo , Autofagia/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Estrés Oxidativo/efectos de los fármacos , Peritonitis/inmunología , Peritonitis/metabolismo , Transducción de Señal , Células THP-1
12.
Mediators Inflamm ; 2019: 5647074, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31885496

RESUMEN

Gout is a prevalent form of aseptic inflammation caused by the deposition of monosodium urate (MSU) crystals in joints or tissues. Triggering receptor expressed on myeloid cell-1 (TREM-1) is a superimmunoglobulin receptor expressed on innate immune cells including granulocytes, monocytes, and macrophages. TREM-1 serves as a link between innate immunity and adaptive immunity, playing a crucial role in regulating inflammation and immune response. The purpose of this study was to investigate the potential role of TREM-1 in THP-1 cells and peripheral blood mononuclear cells (PBMCs) from patients with gouty arthritis (GA). In the current study, we found that the mRNA and protein levels of TREM-1 increased in PBMCs from GA patients and soluble TREM-1 in plasma as well. In addition, an increased level of TREM-1 was observed in THP-1 treated with monosodium urate (MSU) in vitro, along with upregulation of proinflammatory cytokines. Moreover, upon specific inhibition of TREM-1, Toll-like receptor 4 (TLR-4), and myeloid differentiation factor 88 (MyD88), the levels of MyD88 and proinflammatory cytokines were decreased after MSU challenge in THP-1 cells. Interestingly, inhibition of TLR-4 could enhance the effect of TREM-1 inhibitor in MSU-induced inflammation. Taken together, our findings suggested that TREM-1 could accelerate MSU-induced acute inflammation. Inhibition of TREM-1 may provide a new strategy for alleviating acute gouty inflammation.


Asunto(s)
Artritis Gotosa/inmunología , Artritis Gotosa/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Adulto , Artritis Gotosa/genética , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inflamación/inducido químicamente , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Células THP-1 , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptor Activador Expresado en Células Mieloides 1/genética , Ácido Úrico/toxicidad
13.
Immunopharmacol Immunotoxicol ; 41(2): 277-284, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31084401

RESUMEN

Objectives: Gouty arthritis is caused by the deposition of monosodium urate (MSU) crystals in joints, which is associated with the rise of serum urate content. This study aims to investigate the therapeutic effect of Madecassoside on gouty arthritis and hyperuricemia. Methods: DBA/1 mice were intradermally injected with MSU to stimulate joint inflammation or intraperitoneally injected with MSU to trigger peritonitis. Moreover, ICR mice were exposed to potassium oxonate to stimulate hyperuricemia. Results: Madecassoside repressed MSU-triggered pad swelling, joint 99mTc uptake, and joint inflammation in DBA/1 mice with gouty arthritis. Neutrophil infiltration and IL-1ß & IL-6 & MCP-1 secretion was also alleviated in lavage fluids from DBA/1 mice with peritonitis due to Madecassoside treatment. Furthermore, Madecassoside decreased MSU-induced neutrophil cytosolic factor 1, caspase-1 and NLRP3 expression in mice with peritoneal inflammation. In hyperuricemic mice, Madecassoside improved renal dysfunction. Serum uric acid, BUN, and creatinine were down-regulated by Madecassoside. Conclusion: These findings indicate that Madecassoside has potential to ameliorate inflammation in both acute gouty arthritis model and peritonitis model, probably via regulating IL-1ß and NLRP3 expression. Practical point: Madecassoside also exhibited a urate-lowering effect and a renal protective effect in hyperuricemic mice.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Gotosa/tratamiento farmacológico , Hiperuricemia/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Triterpenos/farmacología , Animales , Artritis Gotosa/inducido químicamente , Artritis Gotosa/inmunología , Citocinas/inmunología , Hiperuricemia/inducido químicamente , Hiperuricemia/inmunología , Hiperuricemia/patología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones Endogámicos ICR , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/patología , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/patología , Ácido Úrico/toxicidad , omegacloroacetofenona
14.
Biochem Biophys Res Commun ; 497(3): 897-902, 2018 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-29476737

RESUMEN

Acute gouty arthritis (AGA) is one of the most common forms of auto-inflammatory arthritis. IL-17 is a key proinflammatory cytokine which has been implicated in several autoimmune diseases. However, to date little is known about the role of IL-17 in AGA. In the present study, we show that serum IL-17 levels are significantly elevated in AGA patients early in the onset of symptoms of gout, and decrease gradually as symptoms diminish. Correlation analysis indicated that IL-17 expression is not only positively correlated with disease activity, but is also correlated with serum levels of IL-1ß which plays a critical role in the differentiation of IL-17- γδT cells into IL-17+γδT cells. Flow cytometry analysis indicated that γδ T cells are a major source of IL-17 production during the early onset of AGA. We therefore identify IL-17 as a potential novel biomarker for AGA and suggest that targeting the γδ T cell/IL-17 immune axis is a potential strategy for treatment of acute flares of AGA.


Asunto(s)
Artritis Gotosa/sangre , Interleucina-17/sangre , Adulto , Artritis Gotosa/inmunología , Biomarcadores/sangre , Humanos , Interleucina-17/inmunología , Interleucina-1beta/sangre , Interleucina-1beta/inmunología , Masculino , Persona de Mediana Edad , Linfocitos T/citología , Linfocitos T/inmunología
15.
Curr Opin Rheumatol ; 29(2): 163-170, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27941389

RESUMEN

PURPOSE OF REVIEW: Gout is a common debilitating form of arthritis and despite our extensive knowledge on the pathogenesis its prevalence is still rising quickly. In the current review, we provide a concise overview of recent discoveries in factors tuning the inflammatory response to soluble uric acid and monosodium urate crystals. RECENT FINDINGS: It appears that soluble uric acid has a much larger role to play than just being a risk factor for gout. It may have widespread consequences for systemic inflammation and the development of metabolic syndrome. Additionally, a specific gout-related gut microbiome might not only provide us with a new diagnostic tool, but also highlights possible new therapeutic targets. Furthermore, several recent publications further elucidated the roles of mitochondrial dysfunction, production of reactive oxygen species, autophagy, and AMP-dependent protein kinase in monosodium urate-induced NLRP3 inflammasome activation. Finally, neutrophils have been shown to be involved in both the promotion and resolution of gouty inflammation. A new alpha-1-antitrypsin fusion protein may limit the proinflammatory effects of neutrophil-derived serine proteases. SUMMARY: Together, these studies provide us with many new insights in the pathogenesis of gout, important new treatment targets, and a rationale to further study the role of soluble uric acid in inflammatory diseases.


Asunto(s)
Artritis Gotosa/inmunología , Microbioma Gastrointestinal/inmunología , Hiperuricemia/inmunología , Ácido Úrico/inmunología , Autofagia/inmunología , Gota/inmunología , Humanos , Inflamasomas/inmunología , Inflamación , Mitocondrias/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Neutrófilos/inmunología , Estrés Oxidativo , Fosfotransferasas (Aceptor del Grupo Fosfato)/inmunología , Especies Reactivas de Oxígeno/inmunología
16.
BMC Complement Altern Med ; 17(1): 320, 2017 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-28623927

RESUMEN

BACKGROUND: The Gnaphalium pensylvanicum willd. is used in China as a folk medicine to treat anti-inflammatory, cough and rheumatism arthritis. The aim of this study was to evaluate the potential of the extract of G. pensylvanicum to treat hyperuricemia and acute gouty arthritis in animal model. METHODS: G. pensylvanicum extract was evaluated in an experimental model with potassium oxonate (PO) induced hyperuricemia in mice which was used to evaluate anti-hyperuricemia activity and xanthine oxidase (XO) inhibition. Therapies for acute gouty arthritis was also investigated on monosodium urate (MSU) crystal induced paw edema model. RESULTS: G. pensylvanicum extract showed activity in reducing serum uric acid (Sur) through effect renal glucose transporter 9 (GLUT9), organic anion transporter 1 (OAT1) and urate transporter 1 (URAT1) mainly and inhibited XO activity in vivo of mice with PO induced hyperuricemia. The extract of G. pensylvanicum also showed significant anti-inflammatory activity and reduced the paw swelling on MSU crystal-induced paw edema model. Meanwhile, 13 caffeoylquinic acid derivatives and 1 flavone were identified by UPLC-ESI-MS/MS as the main active component of G. pensylvanicum. CONCLUSIONS: The extract of G. pensylvanicum showed significant effect on evaluated models and therefore may be active agents for the treatment of hyperuricemia and acute gouty arthritis.


Asunto(s)
Antiinflamatorios/administración & dosificación , Artritis Gotosa/tratamiento farmacológico , Gnaphalium/química , Supresores de la Gota/administración & dosificación , Hiperuricemia/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Ácido Quínico/análogos & derivados , Animales , Antiinflamatorios/química , Artritis Gotosa/inmunología , Modelos Animales de Enfermedad , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Supresores de la Gota/química , Humanos , Hiperuricemia/genética , Hiperuricemia/inmunología , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones , Fitoterapia , Extractos Vegetales/química , Ácido Quínico/administración & dosificación , Ácido Quínico/química , Ácido Úrico/metabolismo
17.
Ann Rheum Dis ; 75(6): 1219-27, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26174021

RESUMEN

OBJECTIVES: In the present study, we generated a new protein, recombinant human alpha-1-anti-trypsin (AAT)-IgG1 Fc fusion protein (AAT-Fc), and evaluated its properties to suppress inflammation and interleukin (IL)-1ß in a mouse model of gouty arthritis. METHODS: A combination of monosodium urate (MSU) crystals and the fatty acid C16.0 (MSU/C16.0) was injected intra-articularly into the knee to induce gouty arthritis. Joint swelling, synovial cytokine production and histopathology were determined after 4 h. AAT-Fc was evaluated for inhibition of MSU/C16.0-induced IL-1ß release from human blood monocytes and for inhibition of extracellular IL-1ß precursor processing. RESULTS: AAT-Fc markedly suppressed MSU/C16.0-induced joint inflammation by 85-91% (p<0.001). Ex vivo production of IL-1ß and IL-6 from cultured synovia were similarly reduced (63% and 65%, respectively). The efficacy of 2.0 mg/kg AAT-Fc in reducing inflammation was comparable to 80 mg/kg of plasma-derived AAT. Injection of AAT-Fc into mice increased circulating levels of endogenous IL-1 receptor antagonist by fourfold. We also observed that joint swelling was reduced by 80%, cellular infiltration by 95% and synovial production of IL-1ß by 60% in transgenic mice expressing low levels of human AAT. In vitro, AAT-Fc reduced MSU/C16.0-induced release of IL-1ß from human blood monocytes and inhibited proteinase-3-mediated extracellular processing of the IL-1ß precursor into active IL-1ß. CONCLUSIONS: A single low dose of AAT-Fc is highly effective in reducing joint inflammation in this model of acute gouty arthritis. Considering the long-term safety of plasma-derived AAT use in humans, subcutaneous AAT-Fc emerges as a promising therapy for gout attacks.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Gotosa/tratamiento farmacológico , Supresores de la Gota/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Interleucina-1beta/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , alfa 1-Antitripsina/uso terapéutico , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Gotosa/inmunología , Artritis Gotosa/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Supresores de la Gota/administración & dosificación , Supresores de la Gota/farmacología , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/farmacología , Inyecciones Intraarticulares , Inyecciones Intraperitoneales , Interleucina-1beta/metabolismo , Receptores de Lipopolisacáridos/análisis , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/efectos de los fármacos , Monocitos/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacología , alfa 1-Antitripsina/administración & dosificación , alfa 1-Antitripsina/farmacología
18.
Scand J Rheumatol ; 45(5): 384-93, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27206713

RESUMEN

OBJECTIVES: Monosodium urate (MSU) crystal deposition in gouty joints promotes the release of inflammatory mediators, in particular interleukin (IL)-1ß. The induction of IL-1ß production by MSU crystals requires a co-stimulus. The objective of this study was to determine which part of the synovial fluid (SF) provides co-stimulation to MSU crystals to induce IL-1ß in macrophages. METHOD: The lipidic fraction (LF) and the protein fraction (PF) were isolated from the SF of patients with arthropathies. The PF was subfractionated according to different molecular weight (MW) ranges. THP-1 cells or human primary monocytes were stimulated with MSU crystals in the presence or absence of SF or SF fractions. IL-1ß and IL-8 production and IL-1ß mRNA expression were assessed by an enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qPCR). RESULTS: Exposure of monocytes/macrophages to MSU crystals alone induced the moderate release of IL-8 but not of IL-1ß. The production of IL-1ß required the presence of both SF from patients with inflammatory arthritis (SFi) and MSU crystals. SF from patients with non-inflammatory arthritis, that is patients with osteoarthritis (OA), did not affect the IL-1ß production but slightly enhanced the secretion of IL-8. Both MSU crystals and SFi were required for the induction of the IL-1ß transcript, which was not expressed in the presence of either stimulus alone. SFi fractionation demonstrated that the MSU crystal co-stimulus was contained in the PF of SFi with MW > 50 kDa but not in the LF. CONCLUSIONS: This study shows that the SF of inflammatory arthritis patients, including gout patients, contains proteins required for the induction of IL-1ß by MSU crystals in macrophages whereas lipids are not involved.


Asunto(s)
Artritis Gotosa/inmunología , Gota/inmunología , Interleucina-1beta/inmunología , Macrófagos/inmunología , Proteínas/inmunología , ARN Mensajero/metabolismo , Líquido Sinovial/inmunología , Ácido Úrico/inmunología , Artritis Gotosa/genética , Estudios de Casos y Controles , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Gota/genética , Humanos , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-8/inmunología , Osteoartritis/genética , Osteoartritis/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Líquido Sinovial/química
19.
J Clin Rheumatol ; 22(7): 360-3, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27660933

RESUMEN

OBJECTIVE: We hypothesized that chronic gouty arthritis patients would develop an immune response to type II collagen that would be revealed by the presence of anti-type II collagen (CII) antibodies in serum, which may in turn be involved in progression to non-remitting arthritis. METHODS: Chronic gouty arthritis was defined as crystal-confirmed gout in patients with no pain-free intercritical period, with or without the presence of tophi, who did not have clinical features of other forms of chronic arthritis. Age-matched gout patients suffering acute gouty attacks who had definite intercritical periods were selected as a control group. Four RA patients who had active disease were enrolled as a positive control group. Anti-CII antibodies were quantified in patient sera via ELISA using a human IgG anti-CII antibody assay kit. Correlations between anti-CII levels and clinical parameters were sought. RESULTS: Fifteen chronic gouty arthritis patients were identified. The anti-CII level was significantly higher among subjects with chronic gout compared to controls, but did not significantly differ in control gout patients during acute attacks and in the intercritical periods. Five patients with chronic gouty arthritis had anti-CII antibody levels higher than 200 AU/mL, whereas only one control gout patient exhibited this feature. Two of four patients with active RA had anti-CII antibody levels higher than 200 U/mL.Patients with tophi had significantly higher anti-CII levels than those without, whereas patients showing radiographic erosion tended to have higher anti-CII levels than those without. CONCLUSION: Patients with chronic gouty arthritis had significantly higher levels of anti-CII antibodies than control gout patients. Such antibody production would be triggered by initiation of cartilage damage but may also play a role in perpetuation of inflammation.


Asunto(s)
Artritis Gotosa/inmunología , Autoanticuerpos/inmunología , Colágeno Tipo II/inmunología , Formación de Anticuerpos/inmunología , Enfermedad Crónica , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Proyectos Piloto
20.
Curr Opin Rheumatol ; 27(2): 156-63, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25633244

RESUMEN

PURPOSE OF REVIEW: To give an overview of current evidence for interleukin (IL)-1 blockade in the management of gout. RECENT FINDINGS: Three IL-1 blockers are currently available for clinical use: anakinra, rilonacept and canakinumab. Recent studies have focused on drugs with a long half-life: rilonacept and canakinumab. For treatment of acute gouty arthritis, three randomized controlled trials (RCTs) showed efficacy of canakinumab with some safety concerns and one RCT failed to show efficacy of rilonacept. For prevention of gout flare when starting uric acid lowering therapy (ULT), four RCTs showed efficacy of rilonacept and one RCT showed efficacy of canakinumab. SUMMARY: There is sufficient evidence supporting the use of IL-1 blockers for treatment of acute gouty arthritis or for prevention of gout flares when starting ULT in selected patients, with contraindications or intolerance to conventional therapy. More data are needed to assess safety and to specify their use in routine practice.


Asunto(s)
Supresores de la Gota/uso terapéutico , Gota/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Interleucina-1/antagonistas & inhibidores , Enfermedad Aguda , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Artritis Gotosa/tratamiento farmacológico , Artritis Gotosa/inmunología , Gota/inmunología , Humanos , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Terapia Molecular Dirigida/métodos , Proteínas Recombinantes de Fusión/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA