Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 266
Filter
1.
Int Immunopharmacol ; 138: 112598, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38981223

ABSTRACT

Euphorbia L. is a traditionally used herb and contains many newly identified compounds with novel chemical structures. Euphorbia factor L2 (EFL2), a diterpenoid derived from Euphorbia seeds, is reported to alleviate acute lung injury and arthritis by exerting anti-inflammatory effects. In this study, we aimed to test the therapeutic benefit and mechanisms of EFL2 in NLRP3 inflammasome-mediated gouty models and identified the potential molecular mechanism. A cell-based system was used to test the specific inhibitory effect of EFL2 on NLRP3-related inflammation. The gouty arthritis model and an air pouch inflammation model induced by monosodium urate monohydrate (MSU) crystals were used for in vivo experiments. Nlrp3-/- mice and in vitro studies were used for mechanistic exploration. Virtual molecular docking and biophysical assays were performed to identify the direct binding and regulatory target of EFL2. The inhibitory effect of EFL2 on inflammatory cell infiltration was determined by flow cytometry in vivo. The mechanism by which EFL2 activates the NLRP3 inflammasome signaling pathway was evaluated by immunological experiment and transmission electron microscopy. In vitro, EFL2 specifically reduced NLRP3 inflammasome-mediated IL-1ß production and alleviated MSU crystal-induced arthritis, as well as inflammatory cell infiltration. EFL2 downregulated NF-κB phosphorylation and NLRP3 inflammasome expression by binding to glucocorticoid receptors. Moreover, EFL2 could specifically suppress the lysosome damage-mediated NLRP3 inflammasome activation process. It is expected that this work may be useful to accelerate the development of anti-inflammatory drugs originated from traditional herbs and improve therapeutics in gout and its complications.


Subject(s)
Anti-Inflammatory Agents , Euphorbia , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Humans , Male , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Gouty/drug therapy , Arthritis, Gouty/immunology , Arthritis, Gouty/metabolism , Arthritis, Gouty/chemically induced , Disease Models, Animal , Diterpenes/pharmacology , Diterpenes/therapeutic use , Euphorbia/chemistry , Gout/drug therapy , Gout/immunology , Gout/pathology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/drug effects , Uric Acid
2.
J Control Release ; 372: 862-873, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906421

ABSTRACT

Improving the activity of uricase and lowering its immunogenicity remain significant challenges in the enzyme replacement management of hyperuricemia and related inflammatory diseases. Herein, an immunogenicity-masking strategy based on engineered red blood cells (RBCs) was developed for effective uricase delivery against both hyperuricemia and gout. The dynamic membrane of RBCs enabled high resistance to protease inactivation and hydrogen peroxide accumulation. Benefiting from these advantages, a single infusion of RBC-loaded uricase (Uri@RBC) performed prolonged blood circulation and sustained hyperuricemia management. Importantly, RBCs masked the immunogenicity of uricase, leading to the maintenance of UA-lowering performance after repeated infusion through reduced antibody-mediated macrophage clearance. In an acute gout model, Uri@RBC profoundly alleviated joint edema and inflammation with minimal systemic toxicity. This study supports the employment of immunogenicity-masking tools for efficient and safe enzyme delivery, and this strategy may be leveraged to improve the usefulness of enzyme replacement therapies for managing a wide range of inflammatory diseases.


Subject(s)
Erythrocytes , Gout , Hyperuricemia , Urate Oxidase , Urate Oxidase/administration & dosage , Urate Oxidase/therapeutic use , Urate Oxidase/immunology , Hyperuricemia/drug therapy , Hyperuricemia/immunology , Animals , Gout/immunology , Erythrocytes/immunology , Male , Humans , Uric Acid/blood , Mice , Mice, Inbred C57BL
3.
Clin Rheumatol ; 43(6): 2027-2034, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38625643

ABSTRACT

OBJECTIVES: Gout is characterized by hyperuricemia and recurrent inflammatory episodes caused by intra-articular crystal deposition of monosodium urate (MSU). There is a clear relationship between gout and metabolic syndrome. Recent evidence indicates that perforin plays a role in regulating glucose homeostasis and provides protection in diet-induced non-alcoholic steatohepatitis models. However, the impact of perforin on immune inflammation in gout remains unclear. METHODS: We induced acute gout models in both wild-type (WT) mice and Prf1null mice by administering intra-articular injections of MSU crystals. We compared the ankle joint swelling and the histological score between the two groups. Furthermore, we investigated underlying mechanisms through in vitro co-culture experiments involving CD8 T cells and macrophages. RESULTS: In this study, Prf1null mice showed significantly more pronounced ankle swelling with increased inflammatory cell infiltrations compared with WT mice 24 h after local MSU injection. Moreover, MSU-induced Prf1null mice exhibited increased accumulation of CD8 T cells but not NK cells. Perforin-deficient CD8 T cells displayed reduced cytotoxicity towards bone marrow-derived M0 and M1 macrophages and promoted TNF-α secretion from macrophage. CONCLUSIONS: Perforin from CD8 T cells limits joint inflammation in mice with acute gout by downregulating macrophage-mediated inflammation. Key Points • Perforin deficiency increased swelling in the ankle joints of mice upon MSU injection. • Perforin deficiency is associated with increased immune cell recruitment and severe joint damage in gout. • Perforin regulated CD8 T cell accumulation in gout and promoted CD8 T cell cytotoxicity towards M0 and M1 macrophages. • CD8 T cell-derived perforin regulated pro-inflammatory cytokine secretion of macrophage.


Subject(s)
CD8-Positive T-Lymphocytes , Disease Models, Animal , Gout , Inflammation , Macrophages , Perforin , Uric Acid , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , Macrophages/metabolism , Macrophages/immunology , Perforin/metabolism , Gout/immunology , Gout/metabolism , Mice, Inbred C57BL , Mice, Knockout , Male , Tumor Necrosis Factor-alpha/metabolism , Pore Forming Cytotoxic Proteins
4.
Biol Res Nurs ; 25(2): 267-281, 2023 04.
Article in English | MEDLINE | ID: mdl-36207143

ABSTRACT

BACKGROUND: Gout is triggered by high urate levels and causes inflammation, pain, and an impaired quality of life. Immersion in water at 20-30°C reduces inflammation and pain in arthritis. Yet, relationships of immersion in water at 20-30°C with urate levels and the nucleotide-binding domain (NOD)-like receptor protein 1 (NLRP1) inflammasome have never been clarified. OBJECTIVES: We aimed to investigate the effects of immersion in water at 20-30°C on urate levels, the NLRP1 inflammasome, pain, and quality of life among acute gout patients. METHODS: A community-based randomized control trial design was used with 2 parallel-intervention groups: immersion in water at 20-30°C (20 min/day for 4 weeks) group and a control group. In total, 76 eligible participants in Tomohon City, Indonesia, were assigned using block randomization. We analyze the results (coef. ß) and 95% confidence intervals (CIs) using a generalized estimating equation model. We analyzed mediating effects using a path analysis. RESULTS: Significant pain alleviation (ß = -2.06 [95% CI = -2.67∼-1.45]; ß = -2.42 [95% CI = -2.97∼-1.87]) and improved quality of life (ß = 5.34 [95% CI = 3.12-7.57]; ß = 9.93 [95% CI = 7.02-12.83]) were detected at 2 and 4 weeks of follow-up compared to the pre-test and control group. Urate levels (ß = -0.34 [95% CI = -0.52∼-0.16]) were reduced at the 2-week follow-up, but there was no significant change in the NLRP1 inflammasome compared to the pre-test and control group after immersion in water at 20-30°C. Both the NLRP1 inflammasome (ß = -0.48 [95% CI = -0.63∼-0.34]); water 0.01) and urate levels (ß = -0.11 [95% CI = -0.24∼-0.03]; p < 0.01) had partial indirect (mediating) effects on the link between immersion in water at 20-30°C and pain at the 4-week follow-up. CONCLUSIONS: Immersion in water at 20-30°C significantly decreased pain and increased the quality of life. Immersion in water at 20-30°C mediated NLRP1 and urate levels to decrease pain, although it had no significant effect on the NLRP1 inflammasome concentration after 4 weeks of follow-up and reduced urate levels only at 2 weeks after immersion in water at 20-30°C.


Subject(s)
Gout , Inflammasomes , Inflammation , Pain Management , Pain , Humans , Gout/complications , Gout/genetics , Gout/immunology , Gout/therapy , Immersion , Indonesia , Inflammasomes/genetics , Inflammasomes/immunology , Inflammation/genetics , Inflammation/immunology , Pain/genetics , Pain/immunology , Pain Management/methods , Quality of Life , Temperature , Uric Acid/adverse effects , Uric Acid/analysis , Water , Biomarkers
5.
Blood ; 140(10): 1054-1056, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36074537
6.
Front Immunol ; 13: 1065739, 2022.
Article in English | MEDLINE | ID: mdl-36591268

ABSTRACT

Objective: Hyperuricemia and gout have become gradually more common. The effect of serum urate on organism aging and systematic inflammation is not determined. This study aims to evaluate whether serum urate is causally associated with cellular aging markers and serum inflammation markers. Methods: A Mendelian randomization study was performed on summary-level data from the largest published genome-wide association studies. Single nucleotide polymorphisms with a genome-wide significance level were selected as instrumental variables for leukocyte telomere length (LTL), and serum soluble makers of inflammation (CRP, IL-6, TNF-α, and IGF-1). Standard inverse variance weighted (IVW) method was used as the primary statistical method. The weighted median, MR-Egger regression, and MR-PRESSO methods were used for sensitivity analysis. Results: An inverse causal association of genetically predicted serum urate levels and LTL was found using IVW method (OR: 0.96, 95%CI 0.95, 0.97; ß=-0.040; SE=0.0072; P=4.37×10-8). The association was also supported by MR results using MR-Egger method and weighted median method. The MR-PRESSO analysis and leave-one-out sensitivity analysis supported the robustness of the combined results. In terms of other aging-related serum biomarkers, there was no evidence supporting a causal effect of serum urate on CRP, IL-6, TNF-α, or IGF-1 levels. Conclusions: Serum urate levels are negatively associated with telomere length but are not associated with serum soluble indicators of inflammation. Telomere length may be a critical marker that reflects urate-related organismal aging and may be a mechanism in the age-related pathologies and mortality caused by hyperuricemia.


Subject(s)
Gout , Hyperuricemia , Inflammation , Telomere , Uric Acid , Humans , Biological Specimen Banks , Biomarkers/blood , Genome-Wide Association Study , Hyperuricemia/blood , Hyperuricemia/genetics , Hyperuricemia/immunology , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Insulin-Like Growth Factor I , Interleukin-6 , Telomere/genetics , Telomere/immunology , Tumor Necrosis Factor-alpha , United Kingdom , Uric Acid/blood , Uric Acid/immunology , Gout/blood , Gout/immunology
7.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884486

ABSTRACT

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.


Subject(s)
Arthritis/pathology , Fibroblasts/pathology , Gout/pathology , Lupus Erythematosus, Systemic/pathology , Phospholipases A1/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Synoviocytes/pathology , Arthritis/genetics , Arthritis/immunology , Arthritis/metabolism , Case-Control Studies , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Gout/genetics , Gout/immunology , Gout/metabolism , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Male , Phospholipases A1/genetics , Phosphoric Diester Hydrolases/genetics , Receptors, Lysophosphatidic Acid/genetics , Synovial Fluid/immunology , Synovial Fluid/metabolism , Synoviocytes/immunology , Synoviocytes/metabolism
8.
Pathology ; 53(7): 824-829, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34657735

ABSTRACT

Gout is a form of arthritis, resulting from an inflammatory reaction to the deposition of monosodium urate (MSU) crystals in the synovial fluid of the joint space. It is characterised by periods of acute inflammation in the affected joint, or joints (known as gout flares), separated by asymptomatic periods. There seems to be substantial overlap between environmental triggers of gout flares and common environmental modifiers (diet, pharmaceuticals, and stress) of epigenetic markers (DNA methylation, histone modifications, and ncRNA). Very few studies have looked at whether environment is influencing gout through epigenetic mechanisms. The pathogenesis of gouty inflammation is well understood but understanding the variation of response to hyperuricaemia in terms of gout flare initiation is less well known. In this review, we will examine the potential of epigenomics in understanding how gout flares may occur, both in terms of development of hyperuricaemia and the inflammatory response. Looking at the epigenome and its intersection with lifestyle could help identify new targets and strategies for effective management of gout flares.


Subject(s)
Epigenome , Epigenomics , Gout/genetics , Hyperuricemia , Inflammation , Gout/immunology , Gout/pathology , Humans , Life Style , Symptom Flare Up
9.
Ann Rheum Dis ; 80(12): 1604-1614, 2021 12.
Article in English | MEDLINE | ID: mdl-34663597

ABSTRACT

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.


Subject(s)
Arthralgia/metabolism , Arthritis/metabolism , Crystal Arthropathies/metabolism , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nociceptors/metabolism , TRPV Cation Channels/genetics , Adult , Animals , Arthralgia/immunology , Arthritis/immunology , Arthritis, Gouty/immunology , Arthritis, Gouty/metabolism , Crystal Arthropathies/immunology , Gout/immunology , Gout/metabolism , Humans , Inflammasomes/immunology , Inflammation , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Male , Mice , Middle Aged , Optical Imaging , Patch-Clamp Techniques , Synovial Membrane/cytology , THP-1 Cells , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Uric Acid
10.
Int J Rheum Dis ; 24(10): 1257-1265, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34390315

ABSTRACT

INTRODUCTION: The NLR family pyrin domain containing 3 (NLRP3) signaling pathway has an important role in inflammation mediated by monosodium urate crystals in gout, and the characterization of single nucleotide polymorphisms (SNPs) have helped to recognize disease susceptibility. OBJECTIVE: The aim of this review is to provide an overview of the potential role of the inflammasome gene SNPs as a susceptibility factor for gout, discussing the current evidence available. METHODS: This review analyzes the relevant literature in the field of inflammasome SNPs and gout published in the last 10 years. The systematic research was performed in 16 articles, including both the SNPs associated and those not associated with gout, with the goal to have a complete overview. RESULTS: Sixty-nine SNPs from 10 different genes have been reported in the literature. Of these, 13 SNPs present association with gout susceptibility in different populations, while 56 have been established as not being associated with the disease. CONCLUSIONS: This review is a summary of the potential role of inflammasome gene SNPs and their association with gout risk, all of them related with NLRP3 inflammasome signaling, suggesting these polymorphisms are susceptibility candidates and genetic markers for gout. From the 69 SNPs analyzed in the literature, 13 of them have been associated with gout as follows: NLRP3 (rs3806268 and rs10754558), CARD8 (rs2043211), TLR4 (rs2149356), CD14 (rs2569190), IL-1ß (rs1143623), P2RX7 (rs2230911, rs1653624, rs7958316 and rs17525809) and PPARGC1B (rs45520937, rs10491360 and rs7712296) in different populations.


Subject(s)
Gout/genetics , Inflammasomes/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Polymorphism, Single Nucleotide , Genetic Association Studies , Genetic Predisposition to Disease , Gout/diagnosis , Gout/immunology , Humans , Phenotype , Risk Assessment , Risk Factors
11.
Int Immunopharmacol ; 97: 107819, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34098486

ABSTRACT

Tanshinones, the active ingredients derived from the roots of Salvia miltiorrhiza, have been widely used as traditional medicinal herbs for treating human diseases. Although tanshinones showed anti-inflammatory effects in many studies, large knowledge gaps remain regarding their underlying mechanisms. Here, we identified 15 tanshinones that suppressed the activation of NLRP3 inflammasome and studied their structure-activity relationships. Three tanshinones (tanshinone IIA, isocryptotanshinone, and dihydrotanshinone I) reduced mitochondrial reactive-oxygen species production in lipopolysaccharide (LPS)/nigericin-stimulated macrophages and correlated with altered mitochondrial membrane potentials, mitochondria complexes activities, and adenosine triphosphate and protonated-nicotinamide adenine dinucleotide production. The tanshinones may confer mitochondrial protection by promoting autophagy and the AMP-activated protein kinase pathway. Importantly, our findings demonstrate that dihydrotanshinone I improved the survival of mice with LPS shock and ameliorated inflammatory responses in septic and gouty animals. Our results suggest a potential pharmacological mechanism whereby tanshinones can effectively treat inflammatory diseases, such as septic and gouty inflammation.


Subject(s)
Abietanes/pharmacology , Furans/pharmacology , Gout/drug therapy , Inflammasomes/antagonists & inhibitors , Phenanthrenes/pharmacology , Quinones/pharmacology , Shock, Septic/drug therapy , AMP-Activated Protein Kinases/metabolism , Abietanes/therapeutic use , Animals , Autophagy/drug effects , Autophagy/immunology , Disease Models, Animal , Female , Furans/therapeutic use , Gout/chemically induced , Gout/immunology , Gout/pathology , Humans , Inflammasomes/metabolism , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phenanthrenes/therapeutic use , Quinones/therapeutic use , Rats , Reactive Oxygen Species/metabolism , Shock, Septic/immunology , Shock, Septic/pathology , Uric Acid/administration & dosage , Uric Acid/toxicity
12.
Rheumatol Int ; 41(8): 1503-1507, 2021 08.
Article in English | MEDLINE | ID: mdl-34089357

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19/virology , Colchicine/therapeutic use , Gout/drug therapy , SARS-CoV-2/pathogenicity , COVID-19/diagnosis , COVID-19/immunology , Cytokines/blood , Gout/diagnosis , Gout/immunology , Host-Pathogen Interactions , Humans , Inflammation Mediators/blood , Male , Middle Aged , SARS-CoV-2/immunology
13.
Immunology ; 164(2): 305-317, 2021 10.
Article in English | MEDLINE | ID: mdl-34002852

ABSTRACT

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.


Subject(s)
Antigens, CD/immunology , Apoptosis/immunology , Inflammation/immunology , Neutrophils/immunology , Receptors, Immunologic/immunology , Uric Acid/immunology , Animals , Cells, Cultured , Gout/immunology , Humans , Interleukin-1beta/immunology , Joints/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C
14.
Front Immunol ; 12: 638676, 2021.
Article in English | MEDLINE | ID: mdl-33746978

ABSTRACT

Gout is a common inflammatory arthritis caused by the deposition of monosodium urate (MSU) crystals in the joints. This activates the macrophages into a proinflammatory state by inducing NLRP3-dependent interleukin-1ß (IL-1ß) secretion, resulting in neutrophil recruitment. Soluble decoy receptor 3 (DcR3) is an immune modulator and can exert biological functions via decoy and non-decoy actions. Previously, we showed that DcR3 suppresses lipopolysaccharides (LPS)- and virus-induced inflammatory responses in the macrophages and promotes the macrophages into the M2 phenotype. In this study, we clarified the actions of DcR3 and its non-decoy action motif heparin sulfate proteoglycan (HSPG) binding domain (HBD) in the MSU crystal-induced NLRP3 inflammasome activation in the macrophages and in mice. In bone marrow-derived macrophages, THP-1 and U937 cells, we found that the MSU crystal-induced secretion of IL-1ß and activation of NLRP3 were suppressed by both DcR3.Fc and HBD.Fc. The suppression of the MSU-induced NLRP3 inflammasome activation is accompanied by the inhibition of lysosomal rupture, mitochondrial production of the reactive oxygen species (ROS), expression of cathepsins, and activity of cathepsin B, without affecting the crystal uptake and the expression of NLRP3 or pro-IL-1ß. In the air pouch mice model of gout, MSU induced less amounts of IL-1ß and chemokines secretion, an increased M2/M1 macrophage ratio, and a reduction of neutrophil recruitment in DcR3-transgenic mice, which expresses DcR3 in myeloid cells. Similarly, the mice intravenously treated with DcR3.Fc or HBD.Fc displayed less inflammation response. These findings indicate that HBD of DcR3 can reduce MSU crystal-induced NLRP3 inflammasome activation via modulation of mitochondrial and lysosomal functions. Therefore, we, for the first time, demonstrate a new therapeutic potential of DcR3 for the treatment of gout.


Subject(s)
Gout/immunology , Inflammasomes/metabolism , Lysosomes/metabolism , Macrophages/immunology , Neutrophils/immunology , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Animals , Disease Models, Animal , Humans , Interleukin-1beta/metabolism , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophil Infiltration , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/genetics , THP-1 Cells , Uric Acid/metabolism
15.
PLoS One ; 16(3): e0248564, 2021.
Article in English | MEDLINE | ID: mdl-33725018

ABSTRACT

BACKGROUND AND OBJECTIVE: Stroke is a major contributor to the global burden of disease. Although numerous modifiable risk factors (RF) for stroke have been identified, some remain unexplained. Increasing studies have investigated stroke risk in arthritis, but their results are inconsistent. We aimed to synthesize, quantify, and compare the risk of stroke for the major types of arthritis in cohort studies by using a systematic review and meta-analysis approach. METHODS: We searched Chinese and English databases to identify relevant studies from inception to April 30, 2020. Only studies adjusting at least for age and sex were included. We calculated pooled effect estimates for relative risk (RR) and 95% confidence interval (CI) and identified potential sources of heterogeneity and publication bias. RESULTS: A total of 1,348 articles were retrieved, and after an preliminary screening of titles and abstracts, 69 were reviewed for full text, and finally, 32 met the criteria for meta-analysis. Stroke risk in arthritis was significantly increased in studies adjusting for age and sex (RR = 1.36, 95% CI: 1.27-1.46) and for at least one traditional risk factor (RR = 1.40, 95% CI: 1.28-1.54). The results of studies stratified by stroke subtype were consistent with the main finding (ischemic stroke: RR = 1.53, 95% CI: 1.32-1.78; hemorrhagic stroke: RR = 1.45, 95% CI: 1.15-1.84). In subgroup analysis by arthritis type, stroke risk was significantly increased in rheumatoid arthritis (RR = 1.38, 95% CI: 1.29-1.48), ankylosing spondylitis (RR = 1.49, 95% CI: 1.25-1.77), psoriatic arthritis (RR = 1.33, 95% CI: 1.22-1.45), and gout (RR = 1.40, 95% CI: 1.13-1.73) but not osteoarthritis (RR = 1.03, 95% CI: 0.91-1.16). Age and sex subgroup analyses indicated that stroke risk was similar by sex (women: RR = 1.47, 95% CI: 1.31-1.66; men: RR = 1.44, 95% CI: 1.28-1.61); risk was higher with younger age (<45 years) (RR = 1.46, 95% CI: 1.17-1.82) than older age (≥65 years) (RR = 1.17, 95% CI: 1.08-1.26). CONCLUSIONS: Stroke risk was increased in multiple arthritis and similar between ischemic and hemorrhagic stroke. Young patients with arthritis had the highest risk.


Subject(s)
Arthritis, Psoriatic/epidemiology , Arthritis, Rheumatoid/epidemiology , Gout/epidemiology , Spondylitis, Ankylosing/epidemiology , Stroke/epidemiology , Adult , Age Factors , Aged , Arthritis, Psoriatic/complications , Arthritis, Psoriatic/immunology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/immunology , Cohort Studies , Female , Gout/complications , Gout/immunology , Humans , Male , Risk Factors , Sex Factors , Spondylitis, Ankylosing/complications , Spondylitis, Ankylosing/immunology , Stroke/immunology
16.
Arthritis Rheumatol ; 73(8): 1523-1532, 2021 08.
Article in English | MEDLINE | ID: mdl-33750034

ABSTRACT

OBJECTIVE: Pegloticase is used for the treatment of severe gout, but its use is limited by immunogenicity. This study was undertaken to evaluate whether mycophenolate mofetil (MMF) prolongs the efficacy of pegloticase. METHODS: Participants were randomized 3:1 to receive 1,000 mg MMF twice daily or placebo for 14 weeks, starting 2 weeks before receiving pegloticase and continuing while receiving intravenous pegloticase 8 mg biweekly for 12 weeks. Participants then received pegloticase alone from week 12 to week 24. The primary end points were the proportion of patients who sustained a serum urate level of ≤6 mg/dl at 12 weeks and the rate of adverse events (AEs). Secondary end points included 24-week durability of serum urate level ≤6 mg/dl. Fisher's exact test and Wilcoxon's 2-sample test were used for analyses, along with Kaplan-Meier estimates and log rank tests. RESULTS: A total of 32 participants received ≥1 dose of pegloticase. Participants were predominantly men (88%), with a mean age of 55.2 years, mean gout duration of 13.4 years, and mean baseline serum urate level of 9.2 mg/dl. At 12 weeks, a serum urate level of ≤6 mg/dl was achieved in 19 (86%) of 22 participants in the MMF arm compared to 4 (40%) of 10 in the placebo arm (P = 0.01). At week 24, the serum urate level was ≤6 mg/dl in 68% of MMF-treated patients versus 30% of placebo-treated patients (P = 0.06), and rates of AEs were similar between groups, with more infusion reactions occurring in the placebo arm (30% versus 0%). CONCLUSION: Our findings indicate that MMF therapy with pegloticase is well tolerated and shows a clinically meaningful improvement in targeted serum urate level of ≤6 mg/dl at 12 and 24 weeks. This study suggests an innovative approach to pegloticase therapy in gout.


Subject(s)
Adaptive Immunity/drug effects , Gout Suppressants/administration & dosage , Gout/drug therapy , Mycophenolic Acid/administration & dosage , Polyethylene Glycols/administration & dosage , Urate Oxidase/administration & dosage , Double-Blind Method , Drug Therapy, Combination , Female , Gout/immunology , Gout Suppressants/immunology , Humans , Male , Middle Aged , Mycophenolic Acid/immunology , Proof of Concept Study , Treatment Outcome , Urate Oxidase/immunology
17.
Cytokine ; 141: 155468, 2021 05.
Article in English | MEDLINE | ID: mdl-33647713

ABSTRACT

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.


Subject(s)
Gout/blood , Inflammation Mediators/blood , Interleukin-1/blood , Adult , Aged , Female , Gout/diagnostic imaging , Gout/immunology , Humans , Inflammation Mediators/immunology , Interleukin-1/immunology , Male , Middle Aged
18.
Front Immunol ; 12: 777522, 2021.
Article in English | MEDLINE | ID: mdl-35069549

ABSTRACT

Background: Si-Miao-San (SMS) is a well-known traditional Chinese medicine. This study aims to evaluate the anti-inflammatory effects of SMS on gouty arthritis and its potential mechanism of action. Methods: The effects and mechanism of SMS were evaluated in monosodium urate (MSU)-treated mice or macrophages. The expression of cytokines and PI3K/Akt was analyzed using real-time PCR and Western blotting analyses. Macrophage polarization was assessed with immunofluorescence assays, real-time PCR, and Western blotting. Mass spectrometry was used to screen the active ingredients of SMS. Results: Pretreatment with SMS ameliorated MSU-induced acute gouty arthritis in mice with increased PI3K/Akt activation and M2 macrophage polarization in the joint tissues. In vitro, SMS treatment significantly inhibited MSU-triggered inflammatory response, increased p-Akt and Arg-1 expression in macrophages, and promoted M2 macrophage polarization. These effects of SMS were inhibited when PI3K/Akt activation was blocked by LY294002 in the macrophages. Moreover, SMS significantly reduced serum uric acid levels in the hyperuricemia mice. Using mass spectrometry, the plant hormones ecdysone and estrone were detected as the potentially effective ingredients of SMS. Conclusion: SMS ameliorated MSU-induced gouty arthritis and inhibited hyperuricemia. The anti-inflammatory mechanism of SMS may exert anti-inflammatory effects by promoting M2 polarization via PI3K/Akt signaling. Ecdysone and estrone might be the potentially effective ingredients of SMS. This research may provide evidence for the application of SMS in the treatment of gout.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Gout , Macrophages , Uric Acid , Animals , Chromones/pharmacology , Gout/drug therapy , Gout/immunology , Gout/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , THP-1 Cells , Uric Acid/immunology , Uric Acid/metabolism
19.
Front Immunol ; 12: 771677, 2021.
Article in English | MEDLINE | ID: mdl-34992596

ABSTRACT

Objectives: To compare phagocytic activities of monocytes in peripheral blood mononuclear cells (PBMCs) from acute gout patients and normal subjects, examine monosodium urate monohydrate (MSU) crystal-induced IL-1ß secretion ± recombinant human proteoglycan 4 (rhPRG4) or interleukin-1 receptor antagonist (IL-1RA), and study the anti-inflammatory mechanism of rhPRG4 in MSU stimulated monocytes. Methods: Acute gout PBMCs were collected from patients in the Emergency Department and normal PBMCs were obtained from a commercial source. Monocytes in PBMCs were identified by flow cytometry. PBMCs were primed with Pam3CSK4 (1µg/mL) for 24h and phagocytic activation of monocytes was determined using fluorescently labeled latex beads. MSU (200µg/mL) stimulated IL-1ß secretion was determined by ELISA. Reactive oxygen species (ROS) generation in monocytes was determined fluorometrically. PBMCs were incubated with IL-1RA (250ng/mL) or rhPRG4 (200µg/mL) and bead phagocytosis by monocytes was determined. THP-1 monocytes were treated with MSU crystals ± rhPRG4 and cellular levels of NLRP3 protein, pro-IL-1ß, secreted IL-1ß, and activities of caspase-1 and protein phosphatase-2A (PP2A) were quantified. The peritoneal influx of inflammatory and anti-inflammatory monocytes and neutrophils in Prg4 deficient mice was studied and the impact of rhPRG4 on immune cell trafficking was assessed. Results: Enhanced phagocytic activation of gout monocytes under basal conditions (p<0.001) was associated with ROS generation and MSU stimulated IL-1ß secretion (p<0.05). rhPRG4 reduced bead phagocytosis by normal and gout monocytes compared to IL-1RA and both treatments were efficacious in reducing IL-1ß secretion (p<0.05). rhPRG4 reduced pro-IL-1ß content, caspase-1 activity, conversion of pro-IL-1ß to mature IL-1ß and restored PP2A activity in monocytes (p<0.05). PP2A inhibition reversed rhPRG4's effects on pro-IL-1ß and mature IL-1ß in MSU stimulated monocytes. Neutrophils accumulated in peritoneal cavities of Prg4 deficient mice (p<0.01) and rhPRG4 treatment reduced neutrophil accumulation and enhanced anti-inflammatory monocyte influx (p<0.05). Conclusions: MSU phagocytosis was higher in gout monocytes resulting in higher ROS and IL-1ß secretion. rhPRG4 reduced monocyte phagocytic activation to a greater extent than IL-1RA and reduced IL-1ß secretion. The anti-inflammatory activity of rhPRG4 in monocytes is partially mediated by PP2A, and in vivo, PRG4 plays a role in regulating the trafficking of immune cells into the site of a gout flare.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Gout/drug therapy , Interleukin-1beta/immunology , Proteoglycans/therapeutic use , Acetylcysteine/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Anti-Inflammatory Agents/pharmacology , Female , Free Radical Scavengers/pharmacology , Gout/immunology , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Knockout , Middle Aged , Neutrophils/drug effects , Neutrophils/immunology , Phagocytosis/drug effects , Protein Phosphatase 2/immunology , Proteoglycans/genetics , Proteoglycans/pharmacology , Reactive Oxygen Species/immunology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , THP-1 Cells , Uric Acid
20.
Mod Rheumatol ; 31(1): 270-275, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32148148

ABSTRACT

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.


Subject(s)
Azetidines/pharmacology , Gout/immunology , Inflammasomes/metabolism , Neutrophils/drug effects , Purines/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Uric Acid/metabolism , Cells, Cultured , Humans , Immunity, Innate , Interleukin-6/metabolism , Neutrophils/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL