Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 264
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 34: 173-202, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-26772211

RESUMEN

The formation and accumulation of crystalline material in tissues is a hallmark of many metabolic and inflammatory conditions. The discovery that the phase transition of physiologically soluble substances to their crystalline forms can be detected by the immune system and activate innate immune pathways has revolutionized our understanding of how crystals cause inflammation. It is now appreciated that crystals are part of the pathogenesis of numerous diseases, including gout, silicosis, asbestosis, and atherosclerosis. In this review we discuss current knowledge of the complex mechanisms of crystal formation in diseased tissues and their interplay with the nutrients, metabolites, and immune cells that account for crystal-induced inflammation.


Asunto(s)
Asbestosis/inmunología , Aterosclerosis/inmunología , Cristalización , Gota/inmunología , Inmunidad Innata , Inflamación/metabolismo , Silicosis/inmunología , Animales , Humanos , Interleucina-1/metabolismo , Nanotecnología , Transición de Fase
2.
J Immunol ; 205(12): 3300-3310, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33199537

RESUMEN

Gout is a painful arthritic inflammatory disease caused by buildup of monosodium urate (MSU) crystals in the joints. Colchicine, a microtubule-depolymerizing agent that is used in prophylaxis and treatment of acute gout flare, alleviates the painful inflammatory response to MSU crystals. Using i.p. and intra-articular mouse models of gout-like inflammation, we found that GEF-H1/GEF-H1/AHRGEF2, a microtubule-associated Rho-GEF, was necessary for the inhibitory effect of colchicine on neutrophil recruitment. GEF-H1 was required for neutrophil polarization in response to colchicine, characterized by uropod formation, accumulation of F-actin and myosin L chain at the leading edge, and accumulation of phosphorylated myosin L chain, flotillin-2, and P-selectin glycoprotein ligand-1 (PSGL-1) in the uropod. Wild-type neutrophils that were pre-exposed to colchicine failed to roll or accumulate on activated endothelial monolayers, whereas GEF-H1 knockout (GEF-H1-/-) neutrophils were unaffected by treatment with colchicine. In vivo, colchicine blocked MSU-induced recruitment of neutrophils to the peritoneum and the synovium in wild-type mice, but not in GEF-H1-/- mice. Inhibition of macrophage IL-1ß production by colchicine was independent of GEF-H1, supporting a neutrophil-intrinsic mode of action. Our results suggest that the anti-inflammatory effects of colchicine in acute gout-like inflammation can be accounted for by inhibition of neutrophil-rolling interactions with the inflamed vasculature and occurs through GEF-H1-dependent neutrophil stimulation by colchicine. These results contribute to our understanding of the therapeutic action of colchicine, and could inform the application of this drug in other conditions.


Asunto(s)
Colchicina/farmacología , Gota , Rodamiento de Leucocito , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos , Factores de Intercambio de Guanina Nucleótido Rho/inmunología , Actinas/genética , Actinas/inmunología , Animales , Modelos Animales de Enfermedad , Gota/tratamiento farmacológico , Gota/genética , Gota/inmunología , Gota/patología , Rodamiento de Leucocito/efectos de los fármacos , Rodamiento de Leucocito/genética , Rodamiento de Leucocito/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Cadenas Ligeras de Miosina , Neutrófilos/inmunología , Neutrófilos/patología , Factores de Intercambio de Guanina Nucleótido Rho/genética
3.
Mol Cell ; 54(2): 297-308, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24766894

RESUMEN

Modern medical and hygienic practices have greatly improved human health and longevity; however, increased human life span occurs concomitantly with the emergence of metabolic and age-related diseases. Studies over the past decade have strongly linked host inflammatory responses to the etiology of several metabolic diseases including atherosclerosis, type 2 diabetes (T2D), obesity, and gout. A common immunological factor to these diseases is the activation of the inflammasome and release of proinflammatory cytokines that promote disease progression. Here we review the molecular mechanism(s) of inflammasome activation in response to metabolic damage-associated molecular patterns (DAMPs) and discuss potential targets for therapeutic intervention.


Asunto(s)
Inflamasomas/fisiología , Enfermedades Metabólicas/genética , Modelos Inmunológicos , Animales , Aterosclerosis/genética , Aterosclerosis/inmunología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Gota/genética , Gota/inmunología , Humanos , Enfermedades Metabólicas/inmunología , Ratones , Obesidad/genética , Obesidad/inmunología
4.
Immunology ; 164(2): 305-317, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34002852

RESUMEN

Gout is an inflammatory disease triggered by deposition of monosodium urate (MSU) crystals in the joints, resulting in high neutrophil influx and pain. Here, we studied the role of the inhibitory receptor CD300a in the resolution process in a murine model of gout. We found increased CD300a expression on neutrophils emigrated to the joint. When compared to WT mice, CD300a-/- mice had persistent neutrophil influx till 24 hr after MSU injection. This was associated with increased concentration of IL-1ß and greater tissue damage in the joints of CD300a-/- mice. There was an increase in the percentage of apoptotic neutrophils in the synovial lavage of WT mice, as compared to CD300a-/- mice. This difference was reflected in the decline of efferocytic events in the synovial cavity of CD300a-/- mice 24 hr after MSU injection. A CD300a agonistic antibody was shown, for the first time, to increase apoptosis of human neutrophils, and this was associated with cleavage of caspase-8. In conclusion, our results reveal an important role of CD300a in the control of leucocyte infiltration, IL-1ß production and caspase-8 cleavage in neutrophils, contributing to the resolution of inflammation triggered by MSU injection.


Asunto(s)
Antígenos CD/inmunología , Apoptosis/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Receptores Inmunológicos/inmunología , Ácido Úrico/inmunología , Animales , Células Cultivadas , Gota/inmunología , Humanos , Interleucina-1beta/inmunología , Articulaciones/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C
5.
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
6.
Rheumatology (Oxford) ; 60(6): 2783-2790, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33188698

RESUMEN

OBJECTIVE: To investigate the association between neutrophil activation and cardiovascular risk in gout patients. We hypothesize that neutrophil activation mediates inflammation and therefore takes part in atherogenesis in gout patients. METHOD: Patient data were collected from 75 consecutive gout patients participating in the Reade gout cohort Amsterdam. Levels of neutrophil extracellular traps (NETs) and neutrophil activation (calprotectin and peroxidase activity) were analysed by ELISA and fluorimetry in plasma and compared with healthy controls. Markers of neutrophil activation were related to clinical markers of cardiovascular risk, including BMI, smoking, blood pressure, lipid profile and 10 year risk of cardiovascular mortality (EU-SCORE). RESULTS: Increased levels of NETs were found in gout patients, although increased levels were not associated with cardiovascular risk. However, markers of neutrophil activation, including peroxidase activity correlated with waist:hip ratio (ß = 0.33, P < 0.001), cholesterol ratio (ß = 0.46, P < 0.005) and triglycerides (ß = 0.60, P < 0.001) as well as the 10 year risk of cardiovascular mortality (ß = 0.44, P = 0.001). Calprotectin levels were elevated in hypertension (P = 0.005) and diabetes (P = 0.02). Finally, gout patients with high levels of both peroxidase and calprotectin ('neutrophil activation signature') had a markedly elevated cardiovascular risk score (P = 0.001), with 68% of the patients having high cardiovascular risk (odds ratio 2.9, P = 0.03). CONCLUSION: We demonstrated elevated levels of neutrophil activation markers, MPO and calprotectin in gout patients as compared with healthy controls. Of note, neutrophil activation markers were associated with several risk factors for cardiovascular disease, including hyperlipidaemia, hypertension and diabetes. Finally, the presence of a neutrophil activation signature was strongly associated with an increased 10 year risk of cardiovascular mortality. Further studies are needed to determine whether gout-specific factors and/or cardiovascular risk factors contribute to the elevated neutrophil activation observed in these patients.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Trampas Extracelulares , Gota/complicaciones , Activación Neutrófila , Índice de Masa Corporal , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/mortalidad , Estudios de Casos y Controles , Femenino , Gota/sangre , Gota/inmunología , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Hipertensión/sangre , Hipertensión/complicaciones , Complejo de Antígeno L1 de Leucocito/sangre , Lípidos/sangre , Masculino , Persona de Mediana Edad , Peroxidasa/sangre , Fumar/efectos adversos , Relación Cintura-Cadera
7.
Cytokine ; 141: 155468, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33647713

RESUMEN

INTRODUCTION: Interleukin-37(IL-37), a natural inhibitor of innate immunity, has been identified to protect against various inflammatory diseases, including monosodium urate (MSU)-induced inflammation. However, the association of IL-37 with clinical indexes and pro-inflammatory mediators in gout patients remains unclear. The aim of this study was to determine IL-37 level in hyperuricemia and gout patients with or without tophus, and to investigate the correlations of IL-37 with clinical indexs such as Uric Acid (UA), CRP(C-reactive protein), Creatinine Clearance Rate (Ccr), Erythrocyte Sedimentation Rate (ESR) and so on, as well as with the pro-inflammatory mediators in serum including Interleukin-1ß(IL-1ß), Interleukin-6(IL-6) and Interleukin-18(IL-18) from gout patients. METHODOLOGY: The serum levels of IL-37, IL-1ß, IL-6 and IL-18 levels in serum of gout patients were determined by ELISA; the correlations between IL-37 and clinical values or pro-inflammatory mediators in serum of gout were analyzed by Spearman correlation test. RESULTS: The serum levels of IL-37 were higher in active gout patients than inactive gout patients and HCs, especially in active gout patients with tophus. No significant difference was observed in serum IL-37 levels between hyperuricemia and normal controls. IL-1ß, IL-6 and IL-18 levels were significant elevated in gout patients with tophus than those without tophus; Serum IL-37 were positively correlated with CRP and ESR, as well as with IL-1ß, IL-6 and IL-18, negatively correlated with Ccr, and not correlated with UA, creatinine (Cr) and triglyceride (TG) in gout patients. CONCLUSIONS: IL-37 increased in gout patients positively associated CRP and ESR, as well as with proinflammatory mediators IL-1ß, IL-6, IL-18, the presence of tophus and chronic kidney disease in gout. It may be a novel marker for predicting this pathology.


Asunto(s)
Gota/sangre , Mediadores de Inflamación/sangre , Interleucina-1/sangre , Adulto , Anciano , Femenino , Gota/diagnóstico por imagen , Gota/inmunología , Humanos , Mediadores de Inflamación/inmunología , Interleucina-1/inmunología , Masculino , Persona de Mediana Edad
8.
Immunol Invest ; 50(8): 884-890, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32633162

RESUMEN

We describe the case of a 42 year old, healthy patient with Covid-19 who despite improvement in his respiratory symptoms developed a mild to moderate cytokine release syndrome (CRS) and an associated monoarticular gout flare. Since the patient refused admission to the hospital and had stable vital signs, we chose to treat him with a safe anti-inflammatory and non-immunosuppressive therapy. To hit two birds with one stone, we considered colchicine, as it has systemic anti-inflammatory effects and is also effective in gout flare. Unexpectedly, 48 hours after treatment, not only did his ongoing fever and toe pain disappear, he also had significant improvements in his general state of health and all his inflammatory markers including fibrinogen, ferritin, D-dimer, and IL-6 levels normalized. To our knowledge, the use of colchicine in Covid-19 and CRS has not been reported. This observation merits the consideration of colchicine as a safe, inexpensive and oral medication for the treatment of mild to moderate CRS in Covid-19 patients. More importantly, in Covid-19 patients with early lung involvement colchicine may be an appropriate candidate to prevent CRS in adjunction with routine antiviral agents. Indeed, multicenter, randomized controlled studies are required to evaluate the benefits of this therapy.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Colchicina/administración & dosificación , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Gota/tratamiento farmacológico , Administración Oral , Adulto , COVID-19/complicaciones , COVID-19/inmunología , COVID-19/virología , Síndrome de Liberación de Citoquinas/diagnóstico , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Gota/diagnóstico , Gota/inmunología , Gota/virología , Humanos , Masculino , SARS-CoV-2/inmunología , Resultado del Tratamiento
9.
Rheumatol Int ; 41(8): 1503-1507, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34089357

RESUMEN

Current data demonstrated that severe cases of coronavirus-disease-19 (COVID-19) require treatment with antiviral therapy, dexamethasone, supportive care, as well as some anti-rheumatic drugs, among them, cytokine inhibitors and colchicine. Colchicine is an anti-inflammatory drug that is being used in rheumatology for many years to treat mostly gout, calcium pyrophosphate deposition disease, and Familial Mediterranean Fever. Here, we present for the first time, two patients suffering from gout being treated with colchicine, who were affected from severe acute respiratory coronavirus-2 (SARS-CoV-2) syndrome. Both patients presented with mild symptoms of COVID-19 expressed with myalgias, arthralgias, and sore throat, while laboratory investigations showed only high acute phase reactants. Four weeks later, both patients were free of symptoms with negative SARS-CoV-2 tests and without any complications. To our knowledge, there are no other studies of gout arthritis and SARS-CoV-2 infection published so far. Thus, our preliminary conclusion is that chronic use of colchicine may mitigate the clinical picture and disease course of COVID-19 in gout arthritis patients. Further studies with a large number of patients are needed to confirm the above beneficial effect of colchicine.


Asunto(s)
Antiinflamatorios/uso terapéutico , COVID-19/virología , Colchicina/uso terapéutico , Gota/tratamiento farmacológico , SARS-CoV-2/patogenicidad , COVID-19/diagnóstico , COVID-19/inmunología , Citocinas/sangre , Gota/diagnóstico , Gota/inmunología , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , SARS-CoV-2/inmunología
10.
Int J Mol Sci ; 22(23)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34884486

RESUMEN

Lysophosphatidylserine (lysoPS) is known to regulate immune cell functions. Phospholipase A1 member A (PLA1A) can generate this bioactive lipid through hydrolysis of sn-1 fatty acids on phosphatidylserine (PS). PLA1A has been associated with cancer metastasis, asthma, as well as acute coronary syndrome. However, the functions of PLA1A in the development of systemic autoimmune rheumatic diseases remain elusive. To investigate the possible implication of PLA1A during rheumatic diseases, we monitored PLA1A in synovial fluids from patients with rheumatoid arthritis and plasma of early-diagnosed arthritis (EA) patients and clinically stable systemic lupus erythematosus (SLE) patients. We used human primary fibroblast-like synoviocytes (FLSs) to evaluate the PLA1A-induced biological responses. Our results highlighted that the plasma concentrations of PLA1A in EA and SLE patients were elevated compared to healthy donors. High concentrations of PLA1A were also detected in synovial fluids from rheumatoid arthritis patients compared to those from osteoarthritis (OA) and gout patients. The origin of PLA1A in FLSs and the arthritic joints remained unknown, as healthy human primary FLSs does not express the PLA1A transcript. Besides, the addition of recombinant PLA1A stimulated cultured human primary FLSs to secrete IL-8. Preincubation with heparin, autotaxin (ATX) inhibitor HA130 or lysophosphatidic acid (LPA) receptor antagonist Ki16425 reduced PLA1A-induced-secretion of IL-8. Our data suggested that FLS-associated PLA1A cleaves membrane-exposed PS into lysoPS, which is subsequently converted to LPA by ATX. Since primary FLSs do not express any lysoPS receptors, the data suggested PLA1A-mediated pro-inflammatory responses through the ATX-LPA receptor signaling axis.


Asunto(s)
Artritis/patología , Fibroblastos/patología , Gota/patología , Lupus Eritematoso Sistémico/patología , Fosfolipasas A1/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Sinoviocitos/patología , Artritis/genética , Artritis/inmunología , Artritis/metabolismo , Estudios de Casos y Controles , Femenino , Fibroblastos/inmunología , Fibroblastos/metabolismo , Gota/genética , Gota/inmunología , Gota/metabolismo , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Masculino , Fosfolipasas A1/genética , Hidrolasas Diéster Fosfóricas/genética , Receptores del Ácido Lisofosfatídico/genética , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Sinoviocitos/inmunología , Sinoviocitos/metabolismo
11.
Mod Rheumatol ; 31(1): 270-275, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32148148

RESUMEN

OBJECTIVES: Gout is an inflammatory arthropathy caused by the deposition of monosodium urate (MSU). The synthesis and release of IL-1ß is crucial for MSU-induced synovial inflammation. The aim of the present study was to investigate the mechanism of MSU crystal-induced autoinflammatory processes. METHODS: In vitro studies were used to evaluate the role of IL-6 in inflammasome activation in human neutrophils cultured with MSU crystals. Human neutrophils were stimulated with MSU in the presence or absence of IL-6 priming to determine NLRP3 inflammasome activation and subsequent cleaved caspase-1 induction or IL-1ß production. RESULTS: IL-6 or MSU stimulation alone did not result in the efficient IL-1ß production from human neutrophils. However, MSU stimulation induced marked IL-1ß production from IL-6-primed neutrophils. Pretreatment with baricitinib, which blocks IL-6 receptor signaling, prevented MSU-induced cleaved caspase-1 or IL-1ß induction in IL-6-primed neutrophils. Tocilizumab pretreatment also inhibited MSU-mediated IL-1ß production from IL-6-primed neutrophils. CONCLUSION: Priming of human neutrophils with IL-6 promotes uric acid-mediated IL-1ß secretion in the absence of microbial stimulation. These results suggest that an endogenous cytokine, IL-6, is involved in MSU-mediated NLRP3 inflammasome activation and subsequent IL-1ß production from innate immune cells and has a crucial role in MSU crystal-induced synovial inflammation. These findings provide insights into uric acid-mediated autoinflammation in the innate immune system.


Asunto(s)
Azetidinas/farmacología , Gota/inmunología , Inflamasomas/metabolismo , Neutrófilos/efectos de los fármacos , Purinas/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Ácido Úrico/metabolismo , Células Cultivadas , Humanos , Inmunidad Innata , Interleucina-6/metabolismo , Neutrófilos/inmunología , Transducción de Señal
12.
Immunology ; 160(1): 78-89, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32107769

RESUMEN

Annexins are well-known Ca2+ phospholipid-binding proteins, which have a wide variety of cellular functions. The role of annexin A1 (AnxA1) in the innate immune system has focused mainly on the anti-inflammatory and proresolving properties through its binding to the formyl-peptide receptor 2 (FPR2)/ALX receptor. However, studies suggesting an intracellular role of AnxA1 are emerging. In this study, we aimed to understand the role of AnxA1 for interleukin (IL)-1ß release in response to activators of the nucleotide-binding domain leucine-rich repeat (NLR) and pyrin domain containing receptor 3 (NLRP3) inflammasome. Using AnxA1 knockout mice, we observed that AnxA1 is required for IL-1ß release in vivo and in vitro. These effects were due to reduction of transcriptional levels of IL-1ß, NLRP3 and caspase-1, a step called NLRP3 priming. Moreover, we demonstrate that AnxA1 co-localize and directly bind to NLRP3, suggesting the role of AnxA1 in inflammasome activation is independent of its anti-inflammatory role via FPR2. Therefore, AnxA1 regulates NLRP3 inflammasome priming and activation in a FPR2-independent manner.


Asunto(s)
Anexina A1/metabolismo , Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Administración Intranasal , Animales , Cartílago Articular , Caspasa 1/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Gota/inducido químicamente , Gota/inmunología , Gota/patología , Humanos , Inflamasomas/metabolismo , Inyecciones Intraarticulares , Pulmón/inmunología , Pulmón/patología , Macrófagos , Masculino , Ratones , Ratones Noqueados , Cultivo Primario de Células , Unión Proteica/inmunología , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/toxicidad , Silicosis/inmunología , Silicosis/patología , Transcripción Genética/inmunología , Ácido Úrico/administración & dosificación , Ácido Úrico/toxicidad
13.
Ann Rheum Dis ; 79(11): 1506-1514, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32699039

RESUMEN

OBJECTIVE: Macrophage activation by monosodium urate (MSU) and calcium pyrophosphate (CPP) crystals mediates an interleukin (IL)-1ß-dependent inflammation during gout and pseudo-gout flare, respectively. Since metabolic reprogramming of macrophages goes along with inflammatory responses dependently on stimuli and tissue environment, we aimed to decipher the role of glycolysis and oxidative phosphorylation in the IL-1ß-induced microcrystal response. METHODS: Briefly, an in vitro study (metabolomics and real-time extracellular flux analysis) on MSU and CPP crystal-stimulated macrophages was performed to demonstrate the metabolic phenotype of macrophages. Then, the role of aerobic glycolysis in IL-1ß production was evaluated, as well in vitro as in vivo using 18F-fluorodeoxyglucose positron emission tomography imaging and glucose uptake assay, and molecular approach of glucose transporter 1 (GLUT1) inhibition. RESULTS: We observed that MSU and CPP crystals led to a metabolic rewiring toward the aerobic glycolysis pathway explained by an increase in GLUT1 plasma membrane expression and glucose uptake on macrophages. Also, neutrophils isolated from human synovial fluid during gout flare expressed GLUT1 at their plasma membrane more frequently than neutrophils isolated from bloodstream. Both glucose deprivation and treatment with either 2-deoxyglucose or GLUT1 inhibitor suppressed crystal-induced NLRP3 activation and IL-1ß production, and microcrystal inflammation in vivo. CONCLUSION: In conclusion, we demonstrated that GLUT1-mediated glucose uptake is instrumental during the inflammatory IL-1ß response induced by MSU and CPP crystals. These findings open new therapeutic paths to modulate crystal-related inflammation.


Asunto(s)
Pirofosfato de Calcio , Gota/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Ácido Úrico , Animales , Pirofosfato de Calcio/inmunología , Pirofosfato de Calcio/metabolismo , Pirofosfato de Calcio/farmacología , Transportador de Glucosa de Tipo 1/inmunología , Transportador de Glucosa de Tipo 1/metabolismo , Glucólisis/efectos de los fármacos , Glucólisis/fisiología , Gota/inmunología , Humanos , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácido Úrico/inmunología , Ácido Úrico/metabolismo , Ácido Úrico/farmacología
14.
Ann Rheum Dis ; 79(4): 536-544, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32114511

RESUMEN

OBJECTIVE: Gout is characterised by severe interleukin (IL)-1-mediated joint inflammation induced by monosodium urate crystals. Since IL-37 is a pivotal anti-inflammatory cytokine suppressing the activity of IL-1, we conducted genetic and functional studies aimed at elucidating the role of IL-37 in the pathogenesis and treatment of gout. METHODS: Variant identification was performed by DNA sequencing of all coding bases of IL37 using molecular inversion probe-based resequencing (discovery cohort: gout n=675, controls n=520) and TaqMan genotyping (validation cohort: gout n=2202, controls n=2295). Predictive modelling of the effects of rare variants on protein structure was followed by in vitro experiments evaluating the impact on protein function. Treatment with recombinant IL-37 was evaluated in vitro and in vivo in a mouse model of gout. RESULTS: We identified four rare variants in IL37 in six of the discovery gout patients; p.(A144P), p.(G174Dfs*16), p.(C181*) and p.(N182S), whereas none emerged in healthy controls (Fisher's exact p-value=0.043). All variants clustered in the functional domain of IL-37 in exon 5 (p-value=5.71×10-5). Predictive modelling and functional studies confirmed loss of anti-inflammatory functions and we substantiated the therapeutic potential of recombinant IL-37 in the treatment of gouty inflammation. Furthermore, the carrier status of p.(N182S)(rs752113534) was associated with increased risk (OR=1.81, p-value=0.031) of developing gout in hyperuricaemic individuals of Polynesian ancestry. CONCLUSION: Here, we provide genetic as well as mechanistic evidence for the role of IL-37 in the pathogenesis of gout, and highlight the therapeutic potential of recombinant IL-37 for the treatment of gouty arthritis.


Asunto(s)
Gota/genética , Interleucina-1/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Gota/inmunología , Humanos , Técnicas In Vitro , Interleucina-1/inmunología , Interleucina-1/farmacología , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Interleucina-8/efectos de los fármacos , Interleucina-8/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Ratones , Persona de Mediana Edad , Nativos de Hawái y Otras Islas del Pacífico/genética , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Polimorfismo Genético , Proteínas Recombinantes/farmacología , Ácido Úrico/inmunología , Ácido Úrico/farmacología , Población Blanca/genética
15.
Ann Rheum Dis ; 78(11): 1601-1604, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31366470

RESUMEN

OBJECTIVES: The recombinant zoster vaccine (RZV) containing a strong non-aluminium adjuvant is associated with increased risk of gout flares, presumably via NLRP3 inflammasome activation. We tested the possibility that other vaccines may also be associated with gout flares. METHODS: We conducted an online case-crossover study of patients with gout to examine the association between vaccination and gout flares. We collected information through the Internet on exposures to potential risk factors, including vaccinations, during 2-day hazard periods prior to gout flare and 2-day control periods without a flare. Conditional logistic regression was used to adjust for covariates. RESULTS: There were 517 participants with gout (mean age 55 years, 79% male) who experienced gout flares during follow-up. There were 28 vaccinations during 990 hazard periods and 21 vaccinations during 1407 control periods. Vaccination was associated with twofold higher odds of gout flare (adjusted OR 1.99; 95% CI 1.01 to 3.89). CONCLUSION: Our findings suggest vaccines other than RZV are associated with increased odds of gout flares, potentially through a shared pathogenetic mechanism like NLRP3 inflammasome. However, the absolute magnitude of increased odds of gout flares with vaccinations remains small and must be interpreted within the context of the overwhelming benefits of vaccinations.


Asunto(s)
Gota/inmunología , Factores Inmunológicos/efectos adversos , Vacunación/efectos adversos , Vacunas/efectos adversos , Estudios Cruzados , Femenino , Humanos , Factores Inmunológicos/inmunología , Modelos Logísticos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Brote de los Síntomas , Vacunas/inmunología
16.
17.
Arch Biochem Biophys ; 670: 94-103, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31255694

RESUMEN

Chaetocin is a fungal metabolite that possesses a potential anti-inflammatory activity. Acute gout is a self-limiting inflammatory response to monosodium urate (MSU) crystals. However, the effect of cheatocin on gout has not been elucidated. In the study, we found that chaetocin could decrease MSU induced IL-1ß secretion in bone marrow derived macrophages by several mechanisms, including inhibiting the activation of NLRP3 inflammasome. Chaetocin negatively regulated apoptosis-associated speck-like protein with a CARD domain oligomerization, and caspase-1 processing, key events during inflammasome activation. Furthermore, chaetocin restrain expressions of Hypoxia-inducible factor-1α and Hexokinase 2, mediators of glycolysis, which necessary for synthesis of pro-IL-1ß during inflammasome priming. In vivo, chaetocin ameliorate MSU-induced arthritis, which showed as reduced local swelling and inflammatory cell infiltration. In MSU-induced peritonitis model, the peritoneal macrophages of chaetocin-pretreated mice showed significantly decreased mRNA levels of HIF-1α and NLRP3 related genes. These findings suggested that chaetocin has a potent anti-inflammatory effect against gout. More importantly, it is proposed that the inhibiting of glycolysis pathway would be a new avenue for the treatment of gout flare and other IL-1ß related diseases.


Asunto(s)
Gota/tratamiento farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Gota/inmunología , Gota/metabolismo , Interleucina-1beta/biosíntesis , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Piperazinas/farmacología , Piperazinas/uso terapéutico
18.
Pharmacol Res ; 147: 104351, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315067

RESUMEN

Gout is a paradigm of acute, self-limiting inflammation caused by the deposition of monosodium urate (MSU) crystals within intra-and/or peri-articular areas, leading to excruciating pain, joint swelling and stiffness. The infiltration of leukocytes drives the inflammatory response and remains an attractive target for therapeutic intervention. In this context, emerging evidence supports the view that systemic differentiation of Th17 cells and their in situ infiltration as one of the potential mechanisms by which these cells, and their main product IL-17, causes damage to target tissues. To test if IL-17 was having a detrimental role in gouty onset and progression we targeted this cytokine, using a neutralizing antibody strategy, in an experimental model of gout. Joint inflammation was induced in CD-1 mice by the intra-articular (i.a.) administration of MSU crystals (200 µg/20 µl). Animals from IL-17Ab-treated groups received 1, 3 and 10 µg (i.a.) in 20 µl of neutralizing antibody after MSU crystals administration. Thereafter, joints were scored macroscopically, and knee joint oedema determined with a caliper. Histological analysis, myeloperoxidase assay and western blots analysis for COX-2/mPGEs-1/IL-17R pathway were conducted at 18 h (peak of inflammation) to evaluate leukocytes infiltration and activation, followed by the analysis, in situ, of pro/anti-inflammatory cytokines and chemokines. Flow cytometry was also used to evaluate the modulation of infiltrated inflammatory monocytes and systemic Th17 and Treg profile. Treatment with IL-17Ab revealed a dose-dependent reduction of joint inflammation scores with maximal inhibition at 10 µg. The neutralizing antibody was also able to significantly reduce leukocytes infiltration and MPO activity as well the expression of JE, IL-1α, IL-1ß, IL-16, IL-17, C5a, BLC and, with a less extent IP-10, Rantes, KC, TIMP-1, SDF-1 and metalloproteinases in inflamed tissues. Biochemical analysis also revealed that IL-17Ab treatment modulated COX-2/mPGEs-1 pathway (and related PGE2 production) without interfering with IL-17R expression. Furthermore, flow cytometry analysis highlighted a selective modulation of infiltrating inflammatory monocytes (B220-/GR1hi-F480hi/CD115+) and circulating Th17, but not Treg, cells after IL-17Ab treatment. Collectively the results of this study report for the first time, that i.a. injection of MSU crystals stimulates in vivo production of Th17 cells and Th17-related inflammatory cyto-chemokines. In addition, we have demonstrated that the administration of a neutralizing antibody against IL-17 attenuates joint symptoms, swelling and leukocytes infiltration to the inflamed tissue, possibly providing a new strategy for the treatment of gouty inflammation and/or arthritis.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Gota/inmunología , Interleucina-17/inmunología , Ácido Úrico , Animales , Edema/inmunología , Edema/patología , Gota/patología , Inflamación/inmunología , Inflamación/patología , Inyecciones Intraarticulares , Articulación de la Rodilla/inmunología , Articulación de la Rodilla/patología , Masculino , Ratones
19.
J Immunol ; 198(1): 428-442, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27903742

RESUMEN

Human neutrophils (polymorphonuclear leukocytes [PMNs]) generate inflammatory responses within the joints of gout patients upon encountering monosodium urate (MSU) crystals. Neutrophil extracellular traps (NETs) are found abundantly in the synovial fluid of gout patients. The detailed mechanism of MSU crystal-induced NET formation remains unknown. Our goal was to shed light on possible roles of purinergic signaling and neutrophil migration in mediating NET formation induced by MSU crystals. Interaction of human neutrophils with MSU crystals was evaluated by high-throughput live imaging using confocal microscopy. We quantitated NET levels in gout synovial fluid supernatants and detected enzymatically active neutrophil primary granule enzymes, myeloperoxidase, and human neutrophil elastase. Suramin and PPADS, general P2Y receptor blockers, and MRS2578, an inhibitor of the purinergic P2Y6 receptor, blocked NET formation triggered by MSU crystals. AR-C25118925XX (P2Y2 antagonist) did not inhibit MSU crystal-stimulated NET release. Live imaging of PMNs showed that MRS2578 represses neutrophil migration and blocked characteristic formation of MSU crystal-NET aggregates called aggregated NETs. Interestingly, the store-operated calcium entry channel inhibitor (SK&F96365) also reduced MSU crystal-induced NET release. Our results indicate that the P2Y6/store-operated calcium entry/IL-8 axis is involved in MSU crystal-induced aggregated NET formation, but MRS2578 could have additional effects affecting PMN migration. The work presented in the present study could lead to a better understanding of gouty joint inflammation and help improve the treatment and care of gout patients.


Asunto(s)
Trampas Extracelulares/efectos de los fármacos , Isotiocianatos/farmacología , Activación Neutrófila/efectos de los fármacos , Receptores Purinérgicos P2/metabolismo , Tiourea/análogos & derivados , Ácido Úrico/inmunología , Quimiotaxis de Leucocito/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Trampas Extracelulares/inmunología , Técnica del Anticuerpo Fluorescente , Gota/inmunología , Gota/metabolismo , Gota/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas In Vitro , Microscopía Confocal , Activación Neutrófila/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Tiourea/farmacología
20.
Eur J Immunol ; 47(3): 585-596, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27995621

RESUMEN

Gout is a self-limited inflammatory disease caused by deposition of monosodium urate (MSU) crystals in the joints. Resolution of inflammation is an active process leading to restoration of tissue homeostasis. Here, we studied the role of Annexin A1 (AnxA1), a glucocorticoid-regulated protein that has anti-inflammatory and proresolving actions, in resolution of acute gouty inflammation. Injection of MSU crystals in the knee joint of mice induced inflammation that was associated with expression of AnxA1 during the resolving phase of inflammation. Neutralization of AnxA1 with antiserum or blockade of its receptor with BOC-1 (nonselective) or WRW4 (selective) prevented the spontaneous resolution of gout. There was greater neutrophil infiltration after challenge with MSU crystals in AnxA1 knockout mice (AnxA1-/- ) and delayed resolution associated to decreased neutrophil apoptosis and efferocytosis. Pretreatment of mice with AnxA1-active N-terminal peptide (Ac2-26 ) decreased neutrophil influx, IL-1ß, and CXCL1 production in periarticular joint. Posttreatment with Ac2-26 decreased neutrophil accumulation, IL-1ß, and hypernociception, and improved the articular histopathological score. Importantly, the therapeutic effects of Ac2-26 were associated with increased neutrophils apoptosis and shortened resolution intervals. In conclusion, AnxA1 plays a crucial role in the context of acute gouty inflammation by promoting timely resolution of inflammation.


Asunto(s)
Anexina A1/metabolismo , Antiinflamatorios/uso terapéutico , Gota/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Articulaciones/efectos de los fármacos , Neutrófilos/fisiología , Péptidos/uso terapéutico , Animales , Anexina A1/genética , Anexina A1/uso terapéutico , Anticuerpos Bloqueadores/administración & dosificación , Apoptosis/efectos de los fármacos , Apoptosis/genética , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Gota/inducido químicamente , Gota/inmunología , Humanos , Inflamación/inmunología , Articulaciones/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Oligopéptidos/administración & dosificación , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Ácido Úrico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA