101
|
Ruiz-Miyazawa KW, Staurengo-Ferrari L, Mizokami SS, Domiciano TP, Vicentini FTMC, Camilios-Neto D, Pavanelli WR, Pinge-Filho P, Amaral FA, Teixeira MM, Casagrande R, Verri WA. Quercetin inhibits gout arthritis in mice: induction of an opioid-dependent regulation of inflammasome. Inflammopharmacology 2017; 25:10.1007/s10787-017-0356-x. [PMID: 28508104 DOI: 10.1007/s10787-017-0356-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/26/2017] [Indexed: 02/07/2023]
Abstract
We investigated the anti-inflammatory and analgesic effects of quercetin in monosodium urate crystals (MSU)-induced gout arthritis, and the sensitivity of quercetin effects to naloxone, an opioid receptor antagonist. Mice were treated with quercetin, and mechanical hyperalgesia was assessed at 1-24 h after MSU injection. In vivo, leukocyte recruitment, cytokine levels, oxidative stress, NFκB activation, and gp91phox and inflammasome components (NLRP3, ASC, Pro-caspase-1, and Pro-IL-1β) mRNA expression by qPCR were determined in the knee joints at 24 h after MSU injection. Inflammasome activation was determined, in vitro, in lipopolysaccharide-primed macrophages challenged with MSU. Quercetin inhibited MSU-induced mechanical hyperalgesia, leukocyte recruitment, TNFα and IL-1β production, superoxide anion production, inflammasome activation, decrease of antioxidants levels, NFκB activation, and inflammasome components mRNA expression. Naloxone pre-treatment prevented all the inhibitory effects of quercetin over MSU-induced gout arthritis. These results demonstrate that quercetin exerts analgesic and anti-inflammatory effect in the MSU-induced arthritis in a naloxone-sensitive manner.
Collapse
Affiliation(s)
- Kenji W Ruiz-Miyazawa
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil
| | - Larissa Staurengo-Ferrari
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil
| | - Sandra S Mizokami
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil
| | - Talita P Domiciano
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil
| | - Fabiana T M C Vicentini
- Farmacore Biotecnologia LTDA, Rua Edson Souto, 728, Lagoinha, 14095-250, Ribeirão Preto, São Paulo, Brazil
| | - Doumit Camilios-Neto
- Departamento de Bioquímica e Biotecnologia, Centro de Ciências Exatas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011, Londrina, Paraná, 86057-970, Brazil
| | - Wander R Pavanelli
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil
| | - Phileno Pinge-Filho
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil
| | - Flávio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Laboratório de Imunofarmacologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Laboratório de Imunofarmacologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Healthy Sciences Center, Londrina State University, Av. Robert Koch, 60, Londrina, Paraná, CEP 86038-350, Brazil
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil.
- Departamento de Patologia, Universidade Estadual de Londrina, Rod. Celso Garcia Cid KM480 PR445, Cx Postal 10.011, Londrina, Paraná, CEP 86057-970, Brazil.
| |
Collapse
|
102
|
Liu HJ, Pan XX, Liu BQ, Gui X, Hu L, Jiang CY, Han Y, Fan YX, Tang YL, Liu WT. Grape seed-derived procyanidins alleviate gout pain via NLRP3 inflammasome suppression. J Neuroinflammation 2017; 14:74. [PMID: 28376889 PMCID: PMC5381065 DOI: 10.1186/s12974-017-0849-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/22/2017] [Indexed: 01/08/2023] Open
Abstract
Background Gout is one of the common inflammatory arthritis which affects many people for inflicting unbearable pain. Macrophage-mediated inflammation plays an important role in gout. The uptake of monosodium urate (MSU) crystals by macrophages can lead to activation of NOD-like receptors containing a PYD 3 (NLRP3) inflammasome, thus accelerating interleukin (IL)-1β production. Reactive oxygen species (ROS) promoted development of the inflammatory process through NLRP3 inflammasome. Our study aimed to find a food-derived compound to attenuate gout pain via the specific inhibition of the NLRP3 inflammasome in macrophages. Methods CD-1 mice were used to evaluate the degree of pain and the swelling dimension of joints after an intra-articular (IA) MSU injection in the ankle. The murine macrophage cell line Raw 264.7 was used to investigate the effects of procyanidins and the mechanism underlying such effects. Histological analysis was used to measure the infiltration of inflammatory cells. ROS produced from Raw 264.7 cells were evaluated by flow cytometry. Cell signaling was measured by Western blot assay and immunofluorescence. Results Procyanidins significantly attenuated gout pain and suppressed ankle swelling. Procyanidins also inhibited MSU-induced activation of the NLRP3 inflammasome and increase of IL-1β. Furthermore, procyanidins decreased ROS levels in Raw 264.7 cells. Conclusions Suppression of the NLRP3 inflammasome in macrophages contributes to the amelioration of gout pain by procyanidins. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0849-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hai-Jiao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China.,Department of Pharmacology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, People's Republic of China
| | - Xiu-Xiu Pan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Bing-Qian Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xuan Gui
- Department of Pharmacy, Sir Run Run Shaw Hospital Affiliated to Nanjing Medical University, Jiangsu, 211166, People's Republic of China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Chun-Yi Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yuan Han
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Yi-Xin Fan
- Department of Pharmacology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, People's Republic of China
| | - Yu-Lin Tang
- Department of Pharmacy, Sir Run Run Shaw Hospital Affiliated to Nanjing Medical University, Jiangsu, 211166, People's Republic of China
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China.
| |
Collapse
|
103
|
Galvão I, Vago JP, Barroso LC, Tavares LP, Queiroz-Junior CM, Costa VV, Carneiro FS, Ferreira TP, Silva PMR, Amaral FA, Sousa LP, Teixeira MM. Annexin A1 promotes timely resolution of inflammation in murine gout. Eur J Immunol 2017; 47:585-596. [PMID: 27995621 DOI: 10.1002/eji.201646551] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/27/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022]
Abstract
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-1β, and CXCL1 production in periarticular joint. Posttreatment with Ac2-26 decreased neutrophil accumulation, IL-1β, 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.
Collapse
Affiliation(s)
- Izabela Galvão
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana P Vago
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Livia C Barroso
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P Tavares
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda S Carneiro
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiana P Ferreira
- Laboratório de Inflamação, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | - Patricia M R Silva
- Laboratório de Inflamação, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | - Flávio A Amaral
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
104
|
Mitsios A, Arampatzioglou A, Arelaki S, Mitroulis I, Ritis K. NETopathies? Unraveling the Dark Side of Old Diseases through Neutrophils. Front Immunol 2017; 7:678. [PMID: 28123386 PMCID: PMC5225098 DOI: 10.3389/fimmu.2016.00678] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/21/2016] [Indexed: 12/14/2022] Open
Abstract
Neutrophil extracellular traps (NETs) were initially described as an antimicrobial mechanism of neutrophils. Over the last decade, several lines of evidence support the involvement of NETs in a plethora of pathological conditions. Clinical and experimental data indicate that NET release constitutes a shared mechanism, which is involved in a different degree in various manifestations of non-infectious diseases. Even though the backbone of NETs is similar, there are differences in their protein load in different diseases, which represent alterations in neutrophil protein expression in distinct disorder-specific microenvironments. The characterization of NET protein load in different NET-driven disorders could be of significant diagnostic and/or therapeutic value. Additionally, it will provide further evidence for the role of NETs in disease pathogenesis, and it will enable the characterization of disorders in which neutrophils and NET-dependent inflammation are of critical importance.
Collapse
Affiliation(s)
- Alexandros Mitsios
- Laboratory of Molecular Hematology, Democritus University of Thrace , Alexandroupolis , Greece
| | | | - Stella Arelaki
- Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece; Department of Pathology, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Ioannis Mitroulis
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine Technische Universität Dresden , Dresden , Germany
| | - Konstantinos Ritis
- Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece; First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
105
|
Liu X, Chia E, Shaw O, Martin WJ, Harper J. Rapid CCL2 Release by Membrane Stromal Cells Initiates Monosodium Urate Crystal-Induced Monocyte Recruitment in a Peritoneal Model of Gouty Inflammation. EUR J INFLAMM 2017. [DOI: 10.1177/1721727x1201000201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- X. Liu
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - E. Chia
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - O.M. Shaw
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - W-J. Martin
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - J.L. Harper
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
106
|
Dinesh P, Rasool M. Berberine, an isoquinoline alkaloid suppresses TXNIP mediated NLRP3 inflammasome activation in MSU crystal stimulated RAW 264.7 macrophages through the upregulation of Nrf2 transcription factor and alleviates MSU crystal induced inflammation in rats. Int Immunopharmacol 2017; 44:26-37. [PMID: 28068647 DOI: 10.1016/j.intimp.2016.12.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/11/2016] [Accepted: 12/26/2016] [Indexed: 12/18/2022]
Abstract
The current study was designed to investigate the therapeutic potential of berberine on monosodium urate (MSU) crystal stimulated RAW 264.7 macrophages and in MSU crystal induced rats. Our results indicate that berberine (25, 50 and 75μM) suppressed the levels of pro-inflammatory cytokines (interleukin-1beta (IL-1β) and tumor necrosis factor alpha (TNFα)) and intracellular reactive oxygen species in MSU crystal stimulated RAW 264.7 macrophages. The mRNA expression levels of IL-1β, caspase 1, nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3), thioredoxin interacting protein (TXNIP) and kelch-like ECH-associated protein 1 (Keap1) were found downregulated with the upregulation of nuclear factor erythroid-2-related factor 2 (Nrf2) transcription factor and its associated anti-oxidant enzymes: Heme oxygenase I (HO-1), superoxide dismutase (SOD1), glutathione peroxidase (GPx), NADPH quinone oxidoreductase-1 (NQO1) and catalase (CAT) in MSU crystal stimulated RAW 264.7 macrophages upon berberine treatment. Subsequently, western blot analysis revealed that berberine decreased the protein expression of IL-1β and caspase 1 and increased Nrf2 expression in RAW 264.7 macrophages. Immunofluorescence analysis also explored increased expression of Nrf2 in MSU crystal stimulated RAW 264.7 macrophages by berberine treatment. In addition, the paw edema, pain score, pro-inflammatory cytokines (IL-1β and TNFα) and articular elastase activity were found significantly reduced in berberine (50mg/kgb·wt) administered MSU crystal-induced rats. Conclusively, our current findings suggest that berberine may represent as a potential candidate for the treatment of gouty arthritis by suppressing inflammatory mediators and activating Nrf2 anti-oxidant pathway.
Collapse
Affiliation(s)
- Palani Dinesh
- Immunopathology Lab, School of Bio Sciences and Technology, VIT University, Vellore 632 014, Tamil Nadu, India
| | - MahaboobKhan Rasool
- Immunopathology Lab, School of Bio Sciences and Technology, VIT University, Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
107
|
Liu L, Xue Y, Zhu Y, Xuan D, Yang X, Liang M, Wang J, Zhu X, Zhang J, Zou H. Interleukin 37 limits monosodium urate crystal-induced innate immune responses in human and murine models of gout. Arthritis Res Ther 2016; 18:268. [PMID: 27863506 PMCID: PMC5116141 DOI: 10.1186/s13075-016-1167-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 10/27/2016] [Indexed: 12/22/2022] Open
Abstract
Background Interleukin (IL)-37 has emerged as a fundamental inhibitor of innate immunity. Acute gout is a self-limiting inflammatory response to monosodium urate (MSU) crystals. In the current study, we assessed the preventive and therapeutic effect of recombinant human IL-37 (rhIL-37) in human and murine gout models. Methods We investigated the expression of IL-37 in patients with active and inactive gouty arthritis and assessed the effect of rhIL-37 in human and murine gout models: a human monocyte cell line (THP-1) and human synovial cells (containing macrophage-like and fibroblast-like synoviocytes) exposed to MSU crystals, a peritoneal murine model of gout and a murine gouty arthritis model. After inhibition of Mer receptor tyrosine kinase (Mertk), levels of IL-1β, IL-8 and chemokine (C-C motif) ligand 2 (CCL-2) were detected by ELISA and expression of mammalian homologs of the drosophila Mad gene 3 (Smad), suppressor of cytokine signaling 3 (SOCS3), NACHT-LRR-PYD-containing protein 3 (NLRP3), and IL-8R of THP-1 were assessed by qPCR and western blot to explore the molecular mechanisms. Results Our studies strongly indicated that rhIL-37 played a potent immunosuppressive role in the pathogenesis of experimental gout models both in vitro and in vivo, by downregulating proinflammatory cytokines and chemokines, markedly reducing neutrophil and monocyte recruitment, and mitigating pathological joint inflammation. In our studies, rhIL-37 suppressed MSU-induced innate immune responses by enhancing expression of Smad3 and IL-1R8 to trigger multiple intracellular switches to block inflammation, including inhibition of NLRP3 and activation of SOCS3. Mertk signaling participated in rhIL-37 inhibitory pathways in gout models. By inhibition of Mertk, the anti-inflammatory effect of rhIL-37 was partly abrogated, and IL-1R8, Smad3 and SOCS3 expression were suppressed, whereas NLRP3 expression was reactivated. Conclusions Our studies reveal that IL-37 limits runaway inflammation initiated by MSU crystal-induced immune responses, partly in a Mertk-dependent fashion. Thus, rhIL-37 has both preventive and therapeutic effects in gouty arthritis.
Collapse
Affiliation(s)
- Lei Liu
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China
| | - Yingfeng Zhu
- Department of Pathology, North Huashan Hospital, Fudan University, 108# Luqiang Road, Baoshan District, Shanghai, China
| | - Dandan Xuan
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China
| | - Xue Yang
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China
| | - Minrui Liang
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China
| | - Juan Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China
| | - Jiong Zhang
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China. .,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China. .,Institute of Rheumatology, Immunology and Allergy, Fudan University, 12# Wulumuqi Road, Jingan District, Shanghai, China.
| |
Collapse
|
108
|
Abstract
Gout is the most common inflammatory arthritis in adults in the Western world. Characterized by hyperuricemia and the effects of acute and chronic inflammation in joints and bursa, gout leads to an agonizing, chronically painful arthritis. Arthritis can also be accompanied by urate nephropathy and subcutaneous urate deposits (tophi). Exciting new developments in the last decade have brought back the focus on this interesting, crystal-induced chronic inflammatory condition. New insights include the role of NALP3 inflammasome-induced inflammation in acute gout, the characterization of diagnostic signs on ultrasound and dual-energy computed tomography imaging modalities, the recognition of target serum urate less than 6 mg/day as the goal for urate-lowering therapies, and evidence-based treatment guidelines. A better understanding of disease mechanisms has enabled drug discovery - three new urate-lowering drugs have been approved in the last decade, with several more in the pipeline. We now recognize the important role that environment and genetics play in the causation of gout. A focus on the cardiac, renal, and metabolic comorbidities of gout will help translational research and discovery over the next decade.
Collapse
Affiliation(s)
- Jasvinder A Singh
- Birmingham VA Medical Center, Birmingham, AL, USA.
- Department of Medicine at the School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Division of Epidemiology at the School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
109
|
Detection of Anti-Type II Collagen Antibodies in Patients With Chronic Gouty Arthritis. J Clin Rheumatol 2016; 22:360-3. [DOI: 10.1097/rhu.0000000000000438] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
110
|
Jhang JJ, Lu CC, Yen GC. Epigallocatechin gallate inhibits urate crystals-induced peritoneal inflammation in C57BL/6 mice. Mol Nutr Food Res 2016; 60:2297-2303. [DOI: 10.1002/mnfr.201600106] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/18/2016] [Accepted: 05/20/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Jhih-Jia Jhang
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| | - Chi-Cheng Lu
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| |
Collapse
|
111
|
Enhanced p62 Is Responsible for Mitochondrial Pathway-Dependent Apoptosis and Interleukin-1β Production at the Early Phase by Monosodium Urate Crystals in Murine Macrophage. Inflammation 2016; 39:1603-16. [DOI: 10.1007/s10753-016-0387-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
112
|
Cavalcanti NG, Marques CDL, Lins E Lins TU, Pereira MC, Rêgo MJBDM, Duarte ALBP, Pitta IDR, Pitta MGDR. Cytokine Profile in Gout: Inflammation Driven by IL-6 and IL-18? Immunol Invest 2016; 45:383-95. [PMID: 27219123 DOI: 10.3109/08820139.2016.1153651] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Gout is considered to be an autoinflammatory disease and the presence of monosodium urate (MSU) crystals stimulates activation of NPRL3 inflammasome and subsequently caspase-1, generating production of active IL-1β and IL-18. However, the association between serum cytokines levels and clinical manifestations of the disease is not yet well understood. We evaluated the serum profile of proinflammatory cytokines (IL-1β, IL-6, IL-8, IL-17A, IL-18, IL-22, and IL-23) and described their relationship with clinical and laboratory data. METHODOLOGY Thirty-nine male patients with gout (GG) were assessed for clinical and laboratory variables and cytokine levels were measured by ELISA. For the purposes of comparison, 34 males with no previous history of arthritis were also included in the study (CG). RESULTS Seventeen participants (43%) exhibited active arthritis on evaluation. Levels of IL-18 were significantly higher in patients in relation to the CG (p = 0.0013). No statistically significant differences were found between the GG and CG for the other measured cytokines. There was a moderate correlation between IL-18 and ESR (R = 0.43, p = 0.0073), CRP (R = 0.47, p = 0.0025), and serum levels of IL-6 (R = 0.36, p = 0.023). An association was observed between serum levels of IL-6 and the presence of tophi (p = 0.005) and deformities (p = 0.0008), as well as a correlation between this cytokine and ESR (R = 0.41, p = 0.011) and CRP (R = 0.48, p = 0.02). CONCLUSIONS IL-18 is associated with inflammatory activity in gout, as well as with IL-6 levels, while IL-6 is associated with clinical and laboratory activity, the presence of tophi and articular deformities, and may be a prognostic marker of this pathology.
Collapse
Affiliation(s)
- Nara Gualberto Cavalcanti
- a Rheumatology Department , Hospital das Clínicas da Universidade Federal de Pernambuco (UFPE) , Recife , Brazil
| | - Cláudia Diniz Lopes Marques
- a Rheumatology Department , Hospital das Clínicas da Universidade Federal de Pernambuco (UFPE) , Recife , Brazil
| | - Thiago Ubiratan Lins E Lins
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisa em Inovação Suely Galdino (NUPIT-SG), Centro de Ciências Biológicas, Universidade Federal de Pernambuco , Recife , Brazil
| | - Michelly Cristiny Pereira
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisa em Inovação Suely Galdino (NUPIT-SG), Centro de Ciências Biológicas, Universidade Federal de Pernambuco , Recife , Brazil
| | - Moacyr Jesus Barreto de Melo Rêgo
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisa em Inovação Suely Galdino (NUPIT-SG), Centro de Ciências Biológicas, Universidade Federal de Pernambuco , Recife , Brazil
| | | | - Ivan da Rocha Pitta
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisa em Inovação Suely Galdino (NUPIT-SG), Centro de Ciências Biológicas, Universidade Federal de Pernambuco , Recife , Brazil
| | - Maira Galdino da Rocha Pitta
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Núcleo de Pesquisa em Inovação Suely Galdino (NUPIT-SG), Centro de Ciências Biológicas, Universidade Federal de Pernambuco , Recife , Brazil
| |
Collapse
|
113
|
Gicquel T, Robert S, Victoni T, Lagente V. [The NLRP3 inflammasome: Physiopathology and therapeutic application]. Presse Med 2016; 45:438-46. [PMID: 26880081 DOI: 10.1016/j.lpm.2016.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/30/2015] [Accepted: 01/11/2016] [Indexed: 11/19/2022] Open
Abstract
The innate immune system constitutes the first line of host defense against pathogens. "Nonself", such as exogenous particles or pathogens, triggers an inflammatory response. Inflammasomes are molecular platforms activated upon cellular infection or stress that trigger the maturation of proinflammatory cytokines such as IL-1β. Activation of the NLRP3 inflammasome pathway, the most extensively studied, appears to be the corner stone of many inflammatory diseases, including Crohn's disease, rheumatoid arthritis and gout. Cryopyrine-associated periodic syndromes (CAPS) are NLRP3 inflammasome-associated diseases. Canakinumab (Ilaris(®)) is the only drug approved for CAPS treatment in France. Targeted therapy against NLRP3 inflammasome and IL-1β might be the new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Thomas Gicquel
- CHU de Rennes, laboratoire de toxicologie biologique et médicolégale, 35033 Rennes, France; Université Rennes 1, faculté de pharmacie, 35043 Rennes, France; Inserm, UMR991 « foie, métabolismes et cancer », 35043 Rennes, France.
| | - Sacha Robert
- Université Rennes 1, faculté de pharmacie, 35043 Rennes, France; Inserm, UMR991 « foie, métabolismes et cancer », 35043 Rennes, France
| | - Tatiana Victoni
- Université Rennes 1, faculté de pharmacie, 35043 Rennes, France; Inserm, UMR991 « foie, métabolismes et cancer », 35043 Rennes, France
| | - Vincent Lagente
- Université Rennes 1, faculté de pharmacie, 35043 Rennes, France; Inserm, UMR991 « foie, métabolismes et cancer », 35043 Rennes, France
| |
Collapse
|
114
|
Galvão I, Dias ACF, Tavares LD, Rodrigues IPS, Queiroz-Junior CM, Costa VV, Reis AC, Ribeiro Oliveira RD, Louzada-Junior P, Souza DG, Leng L, Bucala R, Sousa LP, Bozza MT, Teixeira MM, Amaral FA. Macrophage migration inhibitory factor drives neutrophil accumulation by facilitating IL-1β production in a murine model of acute gout. J Leukoc Biol 2016; 99:1035-43. [PMID: 26868525 DOI: 10.1189/jlb.3ma0915-418r] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/25/2016] [Indexed: 12/25/2022] Open
Abstract
This study evaluated the role of macrophage migration inhibitory factor in inflammation caused by monosodium urate crystals. The concentration of macrophage migration inhibitory factor was increased in synovial fluid of patients with acute gout, and there was a positive correlation between intra-articular macrophage migration inhibitory factor and IL-1β concentrations. In mice, the injection of monosodium urate crystals into the knee joint increased the levels of macrophage migration inhibitory factor in macrophages and in inflamed tissue. The injection of recombinant macrophage migration inhibitory factor into the joint of mice reproduced the inflammatory response observed in acute gout, including histologic changes, the recruitment of neutrophils, and increased levels of IL-1β and CXCL1. Importantly, the accumulation of neutrophils and the amount IL-1β in the joints were reduced in macrophage migration inhibitory factor-deficient mice when injected with monosodium urate crystals. We observed a similar effect when we blocked macrophage migration inhibitory factor with (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid or anti-macrophage migration inhibitory factor. In addition, the blockade of IL-1R and CXCR2 reduced recombinant macrophage migration inhibitory factor-induced neutrophil recruitment. Mechanistically, recombinant macrophage migration inhibitory factor is important for the synthesis of il1β mRNA in vivo and in isolated macrophages. Altogether, macrophage migration inhibitory factor promotes neutrophil accumulation and is important for IL-1β production, which are 2 crucial events contributing to the pathogenesis of acute gout.
Collapse
Affiliation(s)
- Izabela Galvão
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Carolina Fialho Dias
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Livia Duarte Tavares
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Irla Paula Stopa Rodrigues
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alesandra Corte Reis
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Paulo Louzada-Junior
- Division of Rheumatology, School of Medicine of Ribeirao Preto, Universidade de São Paulo, Brazil
| | - Daniele Glória Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lin Leng
- Department of Medicine/Rheumatology, Yale University School of Medicine, The Anlyan Center, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Medicine/Rheumatology, Yale University School of Medicine, The Anlyan Center, New Haven, Connecticut, USA
| | - Lirlândia Pires Sousa
- Department of Clinical Analysis and Toxicological, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Torres Bozza
- Laboratory of Inflammation and Immunity, Department of Immunology, Institute of Microbiology, Universidade Federal do Rio de Janeiro, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio Almeida Amaral
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
115
|
Fattori V, Amaral FA, Verri WA. Neutrophils and arthritis: Role in disease and pharmacological perspectives. Pharmacol Res 2016; 112:84-98. [PMID: 26826283 DOI: 10.1016/j.phrs.2016.01.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/25/2022]
Abstract
The inflammatory response in the joint can induce an intense accumulation of leukocytes in the tissue that frequently results in severe local damage and loss of function. Neutrophils are essential cells to combat many pathogens, but their arsenal can contribute or aggravate articular inflammation. Here we summarized some aspects of neutrophil biology, their role in inflammation and indicated how the modulation of neutrophil functions could be useful for the treatment of different forms of arthritis.
Collapse
Affiliation(s)
- Victor Fattori
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | - Flavio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Laboratório de Imunofarmacologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Paraná, Brazil.
| |
Collapse
|
116
|
Shi YC, Lin KS, Jhai YF, Lee BH, Han Y, Cui Z, Hsu WH, Wu SC. Miracle Fruit (Synsepalum dulcificum) Exhibits as a Novel Anti-Hyperuricaemia Agent. Molecules 2016; 21:140. [PMID: 26821007 PMCID: PMC6274476 DOI: 10.3390/molecules21020140] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 01/15/2016] [Accepted: 01/20/2016] [Indexed: 01/20/2023] Open
Abstract
Miracle fruit (Synsepalum dulcificum) belongs to the Sapotaceae family. It can change flavors on taste buds, transforming acidic tastes to sweet. We evaluated various miracle fruit extracts, including water, butanol, ethyl acetate (EA), and hexane fractions, to determine its antioxidant effects. These extracts isolated from miracle fruit exerted potential for reduction of uric acid and inhibited xanthine oxidase activity in vitro and in monosodiumurate (MSU)-treated RAW264.7 macrophages. Moreover, we also found that the butanol extracts of miracle fruit attenuated oxonic acid potassium salt-induced hyperuricaemia in ICR mice by lowering serum uric acid levels and activating hepatic xanthine oxidase. These effects were equal to those of allopurinol, suggesting that the butanol extract of miracle fruit could be developed as a novel anti-hyperuricaemia agent or health food.
Collapse
Affiliation(s)
- Yeu-Ching Shi
- Taiwan Indigena Botanica Co., Ltd., Taipei 11458, Taiwan.
| | - Kai-Sian Lin
- Department of Food Sciences, National Chiayi University, Chiayi 60004, Taiwan.
| | - Yi-Fen Jhai
- Department of Food Sciences, National Chiayi University, Chiayi 60004, Taiwan.
| | - Bao-Hong Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11042, Taiwan.
- Department of Traditional Chinese Medicine, Taipei Medical University Hospital, Taipei 10042, Taiwan.
| | - Yifan Han
- Department of Oral Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Zhibin Cui
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA.
| | - Wei-Hsuan Hsu
- Biochemical Process Technology Department, Center of Excellence for Drug Development, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 30058, Taiwan.
| | - She-Ching Wu
- Department of Food Sciences, National Chiayi University, Chiayi 60004, Taiwan.
| |
Collapse
|
117
|
Morin, a Bioflavonoid Suppresses Monosodium Urate Crystal-Induced Inflammatory Immune Response in RAW 264.7 Macrophages through the Inhibition of Inflammatory Mediators, Intracellular ROS Levels and NF-κB Activation. PLoS One 2015; 10:e0145093. [PMID: 26709520 PMCID: PMC4692533 DOI: 10.1371/journal.pone.0145093] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/28/2015] [Indexed: 12/16/2022] Open
Abstract
Our previous studies had reported that morin, a bioflavanoid exhibited potent anti-inflammatory effect against adjuvant-induced arthritic rats. In this current study, we investigated the anti-inflammatory mechanism of morin against monosodium urate crystal (MSU)-induced inflammation in RAW 264.7 macrophage cells, an in vitro model for acute gouty arthritis. For comparison purpose, colchicine was used as a reference drug. We have observed that morin (100–300 μM) treatment significantly suppressed the levels of inflammatory cytokines (TNF-α, IL-1β, IL-6, MCP-1 and VEGF), inflammatory mediators (NO and PEG2), and lysosomal enzymes (acid phosphatase, β-galactosidase, N-acetyl glucosamindase and cathepsin D) in MSU-crystals stimulated macrophage cells. The mRNA expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and MCP-1), inflammatory enzymes (iNOS and COX-2), and NF-κBp65 was found downregulated in MSU crystal stimulated macrophage cells by morin treatment, however, the mRNA expression of hypoxanthine phospho ribosyl transferse (HPRT) was found to be increased. The flow cytometry analysis revealed that morin treatment decreased intracellular reactive oxygen species levels in MSU crystal stimulated macrophage cells. The western blot analysis clearly showed that morin mainly exerts its anti-inflammatory effects by inhibiting the MSU crystal-induced COX-2 and TNF-α protein expression through the inactivation of NF-κB signaling pathway in RAW 264.7 macrophage cells similar to that of BAY 11–7082 (IκB kinase inhibitor). Our results collectively suggest that morin can be a potential therapeutic agent for inflammatory disorders like acute gouty arthritis.
Collapse
|
118
|
Innate Immunity and Inflammation Post-Stroke: An α7-Nicotinic Agonist Perspective. Int J Mol Sci 2015; 16:29029-46. [PMID: 26690125 PMCID: PMC4691088 DOI: 10.3390/ijms161226141] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/24/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023] Open
Abstract
Stroke is one of the leading causes of death and long-term disability, with limited treatment options available. Inflammation contributes to damage tissue in the central nervous system across a broad range of neuropathologies, including Alzheimer's disease, pain, Schizophrenia, and stroke. While the immune system plays an important role in contributing to brain damage produced by ischemia, the damaged brain, in turn, can exert a powerful immune-suppressive effect that promotes infections and threatens the survival of stroke patients. Recently the cholinergic anti-inflammatory pathway, in particular its modulation using α7-nicotinic acetylcholine receptor (α7-nAChR) ligands, has shown potential as a strategy to dampen the inflammatory response and facilitate functional recovery in stroke patients. Here we discuss the current literature on stroke-induced inflammation and the effects of α7-nAChR modulators on innate immune cells.
Collapse
|
119
|
Amaral FA, Bastos LFS, Oliveira THC, Dias ACF, Oliveira VLS, Tavares LD, Costa VV, Galvão I, Soriani FM, Szymkowski DE, Ryffel B, Souza DG, Teixeira MM. Transmembrane TNF-α is sufficient for articular inflammation and hypernociception in a mouse model of gout. Eur J Immunol 2015; 46:204-11. [PMID: 26449770 DOI: 10.1002/eji.201545798] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 09/10/2015] [Accepted: 10/01/2015] [Indexed: 11/09/2022]
Abstract
Gout manifests as recurrent episodes of acute joint inflammation and pain due to the deposition of monosodium urate (MSU) crystals within the affected tissue in a process dependent on NLRP3 inflammasome activation. The synthesis, activation, and release of IL-1β are crucial for MSU-induced inflammation. The current study evaluated the mechanism by which TNF-α contributed to MSU-induced inflammation. Male C57BL/6J or transgenic mice were used in this study and inflammation was induced by the injection of MSU crystals into the joint. TNF-α was markedly increased in the joint after the injection of MSU. There was inhibition in the infiltration of neutrophils, production of CXCL1 and IL-1β, and decreased hypernociception in mice deficient for TNF-α or its receptors. Pharmacological blockade of TNF-α with Etanercept or pentoxyfylline produced similar results. Mechanistically, TNF-α blockade resulted in lower amounts of IL-1β protein and pro-IL-1β mRNA transcripts in joints. Gene-modified mice that express only transmembrane TNF-α had an inflammatory response similar to that of WT mice and blockade of soluble TNF-α (XPro™1595) did not decrease MSU-induced inflammation. In conclusion, TNF-α drives expression of pro-IL-1β mRNA and IL-1β protein in experimental gout and that its transmembrane form is sufficient to trigger MSU-induced inflammation in mice.
Collapse
Affiliation(s)
- Flávio A Amaral
- Immunopharmacology Lab, Department of Biochemistry and Immunology, Federal University of Minas, Gerais, Brazil
| | - Leandro F S Bastos
- Immunopharmacology Lab, Department of Biochemistry and Immunology, Federal University of Minas, Gerais, Brazil
| | - Thiago H C Oliveira
- Immunopharmacology Lab, Department of Biochemistry and Immunology, Federal University of Minas, Gerais, Brazil
| | - Ana C F Dias
- Department of Microbiology, Federal University of Minas, Gerais, Brazil
| | - Vívian L S Oliveira
- Immunopharmacology Lab, Department of Biochemistry and Immunology, Federal University of Minas, Gerais, Brazil
| | - Lívia D Tavares
- Department of Microbiology, Federal University of Minas, Gerais, Brazil
| | - Vivian V Costa
- Department of Microbiology, Federal University of Minas, Gerais, Brazil
| | - Izabela Galvão
- Immunopharmacology Lab, Department of Biochemistry and Immunology, Federal University of Minas, Gerais, Brazil
| | | | | | - Bernhard Ryffel
- Molecular Immunology and Embryology, Centre National de la Recherche Scientifique, Orléans, France
| | - Danielle G Souza
- Department of Microbiology, Federal University of Minas, Gerais, Brazil
| | - Mauro M Teixeira
- Immunopharmacology Lab, Department of Biochemistry and Immunology, Federal University of Minas, Gerais, Brazil
| |
Collapse
|
120
|
Jhang JJ, Lu CC, Ho CY, Cheng YT, Yen GC. Protective Effects of Catechin against Monosodium Urate-Induced Inflammation through the Modulation of NLRP3 Inflammasome Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7343-7352. [PMID: 26234731 DOI: 10.1021/acs.jafc.5b02605] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Gouty inflammation results from the stimulation of monosodium urate (MSU). Interleukin-1β (IL-1β) secretion is the primary clinical manifestation of MSU attack, and MSU activates IL-1β through a nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome. This study investigated the protective effect and underlying mechanism of naturally occurring phenolic compounds on MSU-induced inflammation in vivo and in vitro. A screening of phenolic compounds revealed that gallic acid and catechin exhibited the most potent free radical scavenging activities. Subcutaneous injection of gallic acid or catechin significantly reduced MSU-induced IL-1β and IL-6 secretion in C57BL/6 mice. However, only catechin inhibited MSU-induced IL-1β secretion and NLRP3 inflammasome activation in MSU-challenged THP-1 cells. MSU-triggered mitochondrial reactive oxygen species (MtROS) production and intracellular calcium levels were significantly decreased by treatment with catechin in THP-1 cells. Catechin treatment also up-regulated Bcl-2 levels and restored MSU-induced mitochondrial transmembrane potential impairment. These results indicate that the protective effects of catechin on MSU-induced IL-1β secretion are associated with modulation of mitochondrial damage. It also suggests that catechin has the potential to protect gout attack by modulation of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Jhih-Jia Jhang
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Chi-Cheng Lu
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Cheng-Ying Ho
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Yu-Ting Cheng
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University , 250 Kuokuang Road, Taichung 40227, Taiwan
- Agricultural Biotechnology Center, National Chung Hsing University , Taichung 40227, Taiwan
| |
Collapse
|
121
|
Kim ND, Luster AD. The role of tissue resident cells in neutrophil recruitment. Trends Immunol 2015; 36:547-55. [PMID: 26297103 DOI: 10.1016/j.it.2015.07.007] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/14/2015] [Accepted: 07/17/2015] [Indexed: 01/09/2023]
Abstract
Neutrophils are first responders of the immune system, rapidly migrating into affected tissues in response to injury or infection. To effectively call in this first line of defense, strategically placed cells within the vasculature and tissue respond to noxious stimuli by sending out coordinated signals that recruit neutrophils. Regulation of organ-specific neutrophil entry occurs at two levels. First, the vasculature supplying the organ provides cues for neutrophil egress out of the bloodstream in a manner dependent upon its unique cellular composition and architectural features. Second, resident immune cells and stromal cells within the organ send coordinated signals that guide neutrophils to their final destination. Here, we review recent findings that highlight the importance of these tissue-specific responses in the regulation of neutrophil recruitment and the initiation and resolution of inflammation.
Collapse
Affiliation(s)
- Nancy D Kim
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andrew D Luster
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
122
|
Greaney AJ, Maier NK, Leppla SH, Moayeri M. Sulforaphane inhibits multiple inflammasomes through an Nrf2-independent mechanism. J Leukoc Biol 2015; 99:189-99. [PMID: 26269198 DOI: 10.1189/jlb.3a0415-155rr] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/28/2015] [Indexed: 12/18/2022] Open
Abstract
The inflammasomes are intracellular complexes that have an important role in cytosolic innate immune sensing and pathogen defense. Inflammasome sensors detect a diversity of intracellular microbial ligands and endogenous danger signals and activate caspase-1, thus initiating maturation and release of the proinflammatory cytokines interleukin-1β and interleukin-18. These events, although crucial to the innate immune response, have also been linked to the pathology of several inflammatory and autoimmune disorders. The natural isothiocyanate sulforaphane, present in broccoli sprouts and available as a dietary supplement, has gained attention for its antioxidant, anti-inflammatory, and chemopreventive properties. We discovered that sulforaphane inhibits caspase-1 autoproteolytic activation and interleukin-1β maturation and secretion downstream of the nucleotide-binding oligomerization domain-like receptor leucine-rich repeat proteins NLRP1 and NLRP3, NLR family apoptosis inhibitory protein 5/NLR family caspase-1 recruitment domain-containing protein 4 (NAIP5/NLRC4), and absent in melanoma 2 (AIM2) inflammasome receptors. Sulforaphane does not inhibit the inflammasome by direct modification of active caspase-1 and its mechanism is not dependent on protein degradation by the proteasome or de novo protein synthesis. Furthermore, sulforaphane-mediated inhibition of the inflammasomes is independent of the transcription factor nuclear factor erythroid-derived 2-like factor 2 (Nrf2) and the antioxidant response-element pathway, to which many of the antioxidant and anti-inflammatory effects of sulforaphane have been attributed. Sulforaphane was also found to inhibit cell recruitment to the peritoneum and interleukin-1β secretion in an in vivo peritonitis model of acute gout and to reverse NLRP1-mediated murine resistance to Bacillus anthracis spore infection. These findings demonstrate that sulforaphane inhibits the inflammasomes through a novel mechanism and contributes to our understanding of the beneficial effects of sulforaphane.
Collapse
Affiliation(s)
- Allison J Greaney
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Nolan K Maier
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Mahtab Moayeri
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
123
|
Vieira AT, Macia L, Galvão I, Martins FS, Canesso MCC, Amaral FA, Garcia CC, Maslowski KM, De Leon E, Shim D, Nicoli JR, Harper JL, Teixeira MM, Mackay CR. A Role for Gut Microbiota and the Metabolite-Sensing Receptor GPR43 in a Murine Model of Gout. Arthritis Rheumatol 2015; 67:1646-56. [PMID: 25914377 DOI: 10.1002/art.39107] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 03/03/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Host-microbial interactions are central in health and disease. Monosodium urate monohydrate (MSU) crystals cause gout by activating the NLRP3 inflammasome, leading to interleukin-1β (IL-1β) production and neutrophil recruitment. This study was undertaken to investigate the relevance of gut microbiota, acetate, and the metabolite-sensing receptor GPR43 in regulating inflammation in a murine model of gout. METHODS Gout was induced by the injection of MSU crystals into the knee joints of mice. Macrophages from the various animals were stimulated to determine inflammasome activation and production of reactive oxygen species (ROS). RESULTS Injection of MSU crystals caused joint inflammation, as seen by neutrophil influx, hypernociception, and production of IL-1β and CXCL1. These parameters were greatly decreased in germ-free mice, mice treated with antibiotics, and GPR-43-deficient mice. Recolonization or administration of acetate to germ-free mice restored inflammation in response to injection of MSU crystals. In vitro, macrophages produced ROS and assembled the inflammasome when stimulated with MSU. Macrophages from germ-free animals produced little ROS, and there was little inflammasome assembly. Similar results were observed in macrophages from GPR-43-deficient mice. Treatment of germ-free mice with acetate restored in vitro responsiveness of macrophages to MSU crystals. CONCLUSION In the absence of microbiota, there is decreased production of short-chain fatty acids that are necessary for adequate inflammasome assembly and IL-1β production in a manner that is at least partially dependent on GPR43. These results clearly show that the commensal microbiota shapes the host's ability to respond to an inflammasome-dependent acute inflammatory stimulus outside the gut.
Collapse
Affiliation(s)
- Angélica T Vieira
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil, and Monash University, Clayton, Victoria, Australia
| | | | - Izabela Galvão
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Flávio A Amaral
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Kendle M Maslowski
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Ellen De Leon
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Doris Shim
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | | | | | | | | |
Collapse
|
124
|
Cumpelik A, Ankli B, Zecher D, Schifferli JA. Neutrophil microvesicles resolve gout by inhibiting C5a-mediated priming of the inflammasome. Ann Rheum Dis 2015; 75:1236-45. [PMID: 26245757 PMCID: PMC4893114 DOI: 10.1136/annrheumdis-2015-207338] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 07/14/2015] [Indexed: 01/01/2023]
Abstract
Objectives Gout is a highly inflammatory but self-limiting joint disease induced by the precipitation of monosodium urate (MSU) crystals. While it is well established that inflammasome activation by MSU mediates acute inflammation, little is known about the mechanism controlling its spontaneous resolution. The aim of this study was to analyse the role of neutrophil-derived microvesicles (PMN-Ecto) in the resolution of acute gout. Methods PMN-Ecto were studied in a murine model of MSU-induced peritonitis using C57BL/6, MerTK−/− and C5aR−/− mice. The peritoneal compartment was assessed for the number of infiltrating neutrophils (PMN), neutrophil microvesicles (PMN-Ecto), cytokines (interleukin-1β, TGFβ) and complement factors (C5a). Human PMN-Ecto were isolated from exudates of patients undergoing an acute gouty attack and functionally tested in vitro. Results C5a generated after the injection of MSU primed the inflammasome for IL-1β release. Neutrophils infiltrating the peritoneum in response to C5a released phosphatidylserine (PS)-positive PMN-Ecto early on in the course of inflammation. These PMN-Ecto in turn suppressed C5a priming of the inflammasome and consequently inhibited IL-1β release and neutrophil influx. PMN-Ecto-mediated suppression required surface expression of the PS-receptor MerTK and could be reproduced using PS-expressing liposomes. In addition, ectosomes triggered the release of TGFβ independent of MerTK. TGFβ, however, was not sufficient to control acute MSU-driven inflammation in vivo. Finally, PMN-Ecto from joint aspirates of patients with gouty arthritis had similar anti-inflammatory properties. Conclusions PMN-Ecto-mediated control of inflammasome-driven inflammation is a compelling concept of autoregulation initiated early on during PMN activation in gout.
Collapse
Affiliation(s)
- Arun Cumpelik
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland Department of Medicine, University Hospital Basel, Basel, Switzerland
| | - Barbara Ankli
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - Daniel Zecher
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland Department of Medicine, University Hospital Basel, Basel, Switzerland Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Jürg A Schifferli
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland Department of Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
125
|
Effects of RuPeng15 Powder (RPP15) on Monosodium Urate Crystal-Induced Gouty Arthritis in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015. [PMID: 26221174 PMCID: PMC4499391 DOI: 10.1155/2015/527019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RuPeng15 Powder (RPP15) is a herbal multicompound remedy that originates from traditional Tibetan medicine and possesses antigout, anti-inflammatory, and antihyperuricemic properties based on the traditional conceptions. The present study was undertaken to evaluate the therapeutic effect of PRP15 in rat gouty arthritis induced by monosodium urate (MSU) crystals. In the present study, we found that treatment with RPP15 (0.4, 0.8, and 1.2 g/kg) in rats with gouty arthritis induced by MSU crystals significantly attenuated the knee swelling. Histomorphometric and immunohistochemistry analyses revealed that MSU-induced inflammatory cell infiltration and the elevated expressions of nuclear transcription factor-κB p65 (NF-κB p65) in synovial tissues were significantly inhibited, and enzyme-linked immunosorbent assay (ELISA) result showed that MSU-induced high levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-8 (IL-8) in synovial fluid were reduced by treatment with RPP15 (0.4, 0.8, and 1.2 g/kg). We conclude that RPP15 may be a promising candidate for the development of a new treatment for gout and its activity of antigout may be partially related to inhibiting TNF-α, IL-1β, IL-8, and NF-κB p65 expression in the synovial tissues.
Collapse
|
126
|
Zheng SC, Zhu XX, Xue Y, Zhang LH, Zou HJ, Qiu JH, Liu Q. Role of the NLRP3 inflammasome in the transient release of IL-1β induced by monosodium urate crystals in human fibroblast-like synoviocytes. JOURNAL OF INFLAMMATION-LONDON 2015; 12:30. [PMID: 25897296 PMCID: PMC4403983 DOI: 10.1186/s12950-015-0070-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 03/12/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND To investigate whether monosodium urate (MSU) crystals induce interleukin (IL)-1β in human fibroblast-like synoviocytes (FLS), and whether the NLRP3 inflammasome is involved in the inflammatory mechanism. METHODS Human FLS isolated from explants of synovial tissue were stimulated with MSU crystals (0.001 to 0.5 mg/ml) for different time course (6 hours to 48 hours). The expressions of IL-1β, IL-6, TNF-α and NLRP3 were evaluated with ELISA, Western blot and quantitative real-time PCR. RESULTS Exposure of FLS to MSU crystals transiently induced a significant increase in IL-1β expression in culture medium with a peak at 6 h. The mRNA level of IL-1β in the FLS cells had a similar pattern at this time point. Changes in IL-6 and TNF-α expression were not observed. Simultaneously, intercellular pro-IL-1β was detected at 6 h. Furthermore, MSU crystals also induced NLRP3 mRNA and protein expression at 6 h to 48 h after MSU treatment. CONCLUSIONS MSU crystals directly increased IL-1β and intercellular NLRP3 expression in FLS cells. It is suggested that the NLRP3 inflammasome may be associated with IL-1β in FLS treated with MSU. Altogether, MSU could induce production and release of IL-1β through the NLRP3 inflammasome in human synoviocytes.
Collapse
Affiliation(s)
- Shu-Cong Zheng
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Xiao-Xia Zhu
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yu Xue
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Li-Hong Zhang
- Department of Anatomy, Histology and Embryology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai, 200032 PR China
| | - He-Jian Zou
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Jian-Hua Qiu
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.,Division of Emergency Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA USA
| | - Qiong Liu
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.,Department of Anatomy, Histology and Embryology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai, 200032 PR China.,Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, 138 Yixueyuan Road, Shanghai, 200032 PR China
| |
Collapse
|
127
|
Yen JH, Lin LC, Chen MC, Sarang Z, Leong PY, Chang IC, Hsu JD, Chen JH, Hsieh YF, Pallai A, Köröskényi K, Szondy Z, Tsay GJ. The metastatic tumor antigen 1-transglutaminase-2 pathway is involved in self-limitation of monosodium urate crystal-induced inflammation by upregulating TGF-β1. Arthritis Res Ther 2015; 17:65. [PMID: 25889736 PMCID: PMC4422600 DOI: 10.1186/s13075-015-0592-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 02/26/2015] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Transglutaminase 2 (TG2), a protein crosslinking enzyme with multiple biochemical functions, has been connected to various inflammatory processes. In this study, the involvement of TG2 in monosodium urate (MSU) crystal-induced inflammation was studied. METHODS Immunohistochemistry, reverse transcription-polymerase chain reaction (RT-PCR) were performed to detect TG2 expression in synovial fluid mononuclear cells (SFMCs) and synovial tissue from patients with gouty arthritis. MSU crystal-exposed RAW264.7 mouse macrophages were analyzed for interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), transforming growth factor β1 (TGF-β1) and TG2 expression by RT-PCR and enzyme-linked immunosorbent assay (ELISA). TG2 small interfering (si)-RNA-mediated silencing and overexpression in RAW264.7 cells were used to evaluate the involvement of TG2 in resolving MSU crystal-induced inflammation. The role of metastatic tumor antigen 1 (MTA1), a master chromatin modifier, was investigated by MTA1 si-RNA-mediated knockdown. In addition, the inflammatory responses were followed in wild type and TG2 null mice after being challenged with MSU crystals in an in vivo peritonitis model. RESULTS TG2 expression was up-regulated in the synovium tissue and SFMCs from patients with gouty arthritis. The levels of MTA1, TG2, TGF-β1, IL-1β and TNF-α mRNAs were consistently increased in MSU crystal-stimulated RAW264.7 cells. si-MTA1 impaired the basal, as well as the MSU crystal-induced expression of TG2 and TGF-β1, but increased that of IL-1β and TNF-α. TG2 overexpression dramatically suppressed MSU crystal-induced IL-1β and TNF-α, but significantly enhanced the TGF-β1 production. Neutralizing TGF-β antibodies or inhibition of the crosslinking activity of TG2 attenuated these effects. On the contrary, loss of TG2 resulted in a reduced TGF-β, but in an increased IL-1β and TNF-α production in MSU crystal-stimulated RAW264.7 cells and mouse embryonic fibroblasts (MEFs). MSU crystal-stimulated IL-1β production was Janus kinase 2 (JAK2)-signaling dependent and TG2-induced TGF-β suppressed the activity of it. Finally, TG2-deficient mice exhibited hyper inflammatory responses after being challenged with MSU crystals in an in vivo peritonitis model. CONCLUSIONS These findings reveal an inherent regulatory role of the MTA1-TG2 pathway in the self-limitation of MSU crystal-induced inflammation via positively regulating the levels of active TGF-β1 in macrophages that opposes the MSU crystal-induced JAK2-dependent pro-inflammatory cytokine formation.
Collapse
Affiliation(s)
- Jia-Hau Yen
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.
| | - Ling-Chung Lin
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.
| | - Meng-Chi Chen
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.
| | - Zsolt Sarang
- Division of Dental Biochemistry, Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Debrecen, Hungary.
| | - Pui-Ying Leong
- Department of Internal Medicine, Chung Shan Medical University Hospital, No. 110, Section 1, Chien-Kuo N. Road, Taichung, Taiwan.
| | - I-Chang Chang
- Department of Orthopedic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Jeng-Dong Hsu
- Department of Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Jiunn-Horng Chen
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.
| | - Yu-Fan Hsieh
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.
| | - Anna Pallai
- Division of Dental Biochemistry, Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Debrecen, Hungary.
| | - Krisztina Köröskényi
- Division of Dental Biochemistry, Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Debrecen, Hungary.
| | - Zsuzsa Szondy
- Division of Dental Biochemistry, Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Debrecen, Hungary.
| | - Gregory J Tsay
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.
- Department of Internal Medicine, Chung Shan Medical University Hospital, No. 110, Section 1, Chien-Kuo N. Road, Taichung, Taiwan.
| |
Collapse
|
128
|
Maier NK, Leppla SH, Moayeri M. The cyclopentenone prostaglandin 15d-PGJ2 inhibits the NLRP1 and NLRP3 inflammasomes. THE JOURNAL OF IMMUNOLOGY 2015; 194:2776-85. [PMID: 25681332 DOI: 10.4049/jimmunol.1401611] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammasomes are cytosolic protein complexes that respond to diverse danger signals by activating caspase-1. The sensor components of the inflammasome, often proteins of the nucleotide-binding oligomerization domain-like receptor (NLR) family, detect stress, danger stimuli, and pathogen-associated molecular patterns. We report that the eicosanoid 15-deoxy-Δ(12,14)-PGJ2 (15d-PGJ2) and related cyclopentenone PGs inhibit caspase-1 activation by the NLR family leucine-rich repeat protein (NLRP)1 and NLRP3 inflammasomes. This inhibition was independent of the well-characterized role of 15d-PGJ2 as a peroxisome proliferator receptor-γ agonist, its activation of NF erythroid 2-related factor 2, or its anti-inflammatory function as an inhibitor of NF-κB. Instead, 15d-PGJ2 prevents the autoproteolytic activation of caspase-1 and the maturation of IL-1β through induction of a cellular state inhibitory to caspase-1 proteolytic function. The eicosanoid does not directly modify or inactivate the caspase-1 enzyme. Rather, inhibition is dependent on de novo protein synthesis. In a mouse peritonitis model of gout, using monosodium urate crystals to activate NLRP3, 15d-PGJ2 caused a significant inhibition of cell recruitment and associated IL-1β release. Furthermore, in a murine anthrax infection model, 15d-PGJ2 reversed anthrax lethal toxin-mediated NLRP1-dependent resistance. The findings reported in this study suggest a novel mechanism for the anti-inflammatory properties of the cyclopentenone PGs through inhibition of caspase-1 and the inflammasome.
Collapse
Affiliation(s)
- Nolan K Maier
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Stephen H Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
129
|
Etiology and pathogenesis of gout. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00187-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
130
|
Cyclic GMP-dependent protein kinase II is necessary for macrophage M1 polarization and phagocytosis via toll-like receptor 2. J Mol Med (Berl) 2014; 93:523-33. [PMID: 25475742 DOI: 10.1007/s00109-014-1236-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/18/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
Abstract
UNLABELLED Cyclic GMP-dependent protein kinase II (cGKII; PRKG2) phosphorylates a variety of biological targets and has been identified as a gout-susceptible gene. However, the regulatory role of cGKII in triggering gout disease has yet to be clarified. Thus, we plan to explore the specific function of cGKII in macrophages related to gout disease. By using cGKII gene knockdown method, we detected macrophage M1/M2 polarization, phagocytosis, and their responses to stimulation by monosodium urate (MSU). cGKII was highly expressed in M1 phenotype, but not in M2, and cGKII knockdown significantly inhibited macrophage M1 polarization by decreasing M1 chemokine markers (CXCL10 and CCL2) and downregulating phagocytosis function. We further identified that cGKII-associated phagocytosis was mediated by upregulating toll-like receptor 2 (TLR2) expression, but not by TLR4. Mimicking gout condition by MSU treatments, we found that MSU alone induced cGKII and TLR2 expression with increased M1 polarization markers and phagocytosis activity. It means that cGKII knockdown significantly inhibited this MSU-induced cGKII-TLR2-phagocytosis axis. Our study showed that cGKII plays a key role in M1 polarization, especially in TLR2-mediated phagocytosis under MSU exposure. The findings provide evidence for the possible role of cGKII as an inflammation exciter in gout disease. KEY MESSAGE Gout-susceptible gene cGKII is necessary for macrophage M1 polarization. cGKII regulates M1 phagocytosis function via TLR2. Monosodium urate treatments increase cGKII expression and related function. This study reveals the role of cGKII in enhancing gouty inflammatory responses.
Collapse
|
131
|
Reber LL, Marichal T, Sokolove J, Starkl P, Gaudenzio N, Iwakura Y, Karasuyama H, Schwartz LB, Robinson WH, Tsai M, Galli SJ. Contribution of mast cell-derived interleukin-1β to uric acid crystal-induced acute arthritis in mice. Arthritis Rheumatol 2014; 66:2881-91. [PMID: 24943488 DOI: 10.1002/art.38747] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 06/10/2014] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Gouty arthritis is caused by the precipitation of monosodium urate monohydrate (MSU) crystals in the joints. While it has been reported that mast cells (MCs) infiltrate gouty tophi, little is known about the actual roles of MCs during acute attacks of gout. This study was undertaken to assess the role of MCs in a mouse model of MSU crystal-induced acute arthritis. METHODS We assessed the effects of intraarticular (IA) injection of MSU crystals in various strains of mice with constitutive or inducible MC deficiency or in mice lacking interleukin-1β (IL-1β) or other elements of innate immunity. We also assessed the response to IA injection of MSU crystals in genetically MC-deficient mice after IA engraftment of wild-type or IL-1β(-/-) bone marrow-derived cultured MCs. RESULTS MCs were found to augment acute tissue swelling following IA injection of MSU crystals in mice. IL-1β production by MCs contributed importantly to MSU crystal-induced tissue swelling, particularly during its early stages. Selective depletion of synovial MCs was able to diminish MSU crystal-induced acute inflammation in the joints. CONCLUSION Our findings identify a previously unrecognized role of MCs and MC-derived IL-1β in the early stages of MSU crystal-induced acute arthritis in mice.
Collapse
|
132
|
Shaw OM, Steiger S, Liu X, Hamilton JA, Harper JL. Brief report: Granulocyte-macrophage colony-stimulating factor drives monosodium urate monohydrate crystal-induced inflammatory macrophage differentiation and NLRP3 inflammasome up-regulation in an in vivo mouse model. Arthritis Rheumatol 2014; 66:2423-8. [PMID: 24910235 DOI: 10.1002/art.38730] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 05/28/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To determine the role of granulocyte-macrophage colony-stimulating factor (GM-CSF) in the differentiation of inflammatory macrophages in an in vivo model of monosodium urate monohydrate (MSU) crystal-induced inflammation. METHODS C57BL/6J mice were treated with either clodronate liposomes to deplete peritoneal macrophages or GM-CSF antibody and were then challenged by intraperitoneal injection of MSU crystals. Peritoneal lavage fluid was collected, and cellular infiltration was determined by flow cytometry. Purified resident and MSU crystal-recruited monocyte/macrophages were stimulated ex vivo with MSU crystals. The interleukin-1β (IL-1β) levels in lavage fluids and ex vivo assay supernatants were measured. GM-CSF-derived and macrophage colony-stimulating factor (M-CSF)-derived macrophages were generated in vitro from bone marrow cells. Protein expression of IL-1β, caspase 1, NLRP3, and ASC by in vitro- and in vivo-generated monocyte/macrophages was analyzed by Western blotting. RESULTS Depletion of resident macrophages lowered MSU crystal-induced IL-1β and GM-CSF levels in vivo as well as IL-1β production by MSU crystal-recruited monocytes stimulated ex vivo. GM-CSF neutralization in vivo decreased MSU crystal-induced IL-1β levels and neutrophil infiltration. MSU crystal-recruited monocyte/macrophages from GM-CSF-neutralized mice expressed lower levels of the macrophage marker CD115 and produced less IL-1β following ex vivo stimulation. These monocytes exhibited decreased expression of NLRP3, pro/active IL-1β, and pro/active caspase 1. In vitro-derived GM-CSF-differentiated macrophages expressed higher levels of NLRP3, pro/active IL-1β, and pro/active caspase 1 compared to M-CSF-differentiated macrophages. CONCLUSION GM-CSF plays a key role in the differentiation of MSU crystal-recruited monocytes into proinflammatory macrophages. GM-CSF production may therefore contribute to the exacerbation of inflammation in gout.
Collapse
Affiliation(s)
- Odette M Shaw
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | | | | | | |
Collapse
|
133
|
Dalbeth N, Lauterio TJ, Wolfe HR. Mechanism of action of colchicine in the treatment of gout. Clin Ther 2014; 36:1465-79. [PMID: 25151572 DOI: 10.1016/j.clinthera.2014.07.017] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/07/2014] [Accepted: 07/22/2014] [Indexed: 12/25/2022]
Abstract
PURPOSE The aims of this article were to systematically review the literature about the mechanism of action of colchicine in the multimodal pathology of acute inflammation associated with gout and to consider the clinical utility of colchicine in other chronic inflammatory diseases. METHODS The English-language literature on PubMed was searched for articles published between 1990 and October 2013, with a cross-reference to citations across all years. Relevant articles pertaining to the mechanism of action of colchicine and the clinical applications of colchicine in gout and other inflammatory conditions were identified and reviewed. FINDINGS The molecular pathology of acute inflammation associated with gouty arthritis involves several concurrent pathways triggered by a variety of interactions between monosodium urate crystals and the surface of cells. Colchicine modulates multiple pro- and antiinflammatory pathways associated with gouty arthritis. Colchicine prevents microtubule assembly and thereby disrupts inflammasome activation, microtubule-based inflammatory cell chemotaxis, generation of leukotrienes and cytokines, and phagocytosis. Many of these cellular processes can be found in other diseases involving chronic inflammation. The multimodal mechanism of action of colchicine suggests potential efficacy of colchicine in other comorbid conditions associated with gout, such as osteoarthritis and cardiovascular disease. IMPLICATIONS Colchicine has multiple mechanisms of action that affect inflammatory processes and result in its utility for treating and preventing acute gout flare. Other chronic inflammatory diseases that invoke these molecular pathways may represent new therapeutic applications for colchicine.
Collapse
Affiliation(s)
- Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand.
| | | | | |
Collapse
|
134
|
Shaw OM, Pool B, Dalbeth N, Harper JL. The effect of diet-induced obesity on the inflammatory phenotype of non-adipose-resident macrophages in an in vivo model of gout. Rheumatology (Oxford) 2014; 53:1901-5. [DOI: 10.1093/rheumatology/keu174] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
135
|
Activation of the NLRP3 inflammasome is not a feature of all particulate vaccine adjuvants. Immunol Cell Biol 2014; 92:535-42. [PMID: 24687021 DOI: 10.1038/icb.2014.21] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 12/28/2022]
Abstract
Particulate vaccine formulations, designed to improve the delivery of antigens to antigen-presenting cells (APCs) and to stimulate an immune response, have been shown to activate the NLRP3 inflammasome. This leads to the processing and secretion of interleukin (IL)-1β, which supports the recruitment of pro-inflammatory immune cells into the tissue and can therefore be beneficial for vaccine potency. Recent work suggested that this may be a common mechanism of action for all particulate formulations. The aim of this study was to investigate whether the activation of the NLRP3 inflammasome was common to many delivery systems. We prepared polymer-based chitosan nanoparticles (CNPs), lipid-based cubosomes, a water in oil emulsion of incomplete Freund's adjuvant (IFA) and alum formulations and examined inflammasome activation in vitro using murine bone-marrow-derived dendritic cells and human peripheral blood mononuclear cells and in vivo in mice. The formulations differed in their morphology, size and zeta-potential. Only the positively charged particles (CNPs and alum) were able to activate the inflammasome and increase the secretion of IL-1β. A decrease in the activation of the inflammasome with these particulates was observed when cathepsin B-mediated effects were blocked, implying a role of lysosomal rupture in the activation process. These findings demonstrate a role for the surface charge of particulates in the activation of the NLRP3 inflammasome, which should be considered when designing a novel vaccine formulation.
Collapse
|
136
|
Stenberg Å, Karlsson A, Feuk-Lagerstedt E, Christenson K, Bylund J, Oldenborg A, Vesterlund L, Matozaki T, Sehlin J, Oldenborg PA. Signal regulatory protein alpha is present in several neutrophil granule populations and is rapidly mobilized to the cell surface to negatively fine-tune neutrophil accumulation in inflammation. J Innate Immun 2014; 6:553-60. [PMID: 24516072 DOI: 10.1159/000357820] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/09/2013] [Indexed: 01/13/2023] Open
Abstract
Signal regulatory protein alpha (SIRPα) is a cell surface glycoprotein with inhibitory functions, which may regulate neutrophil transmigration. SIRPα is mobilized to the neutrophil surface from specific granules, gelatinase granules, and secretory vesicles following inflammatory activation in vitro and in vivo. The lack of SIRPα signaling and the ability to upregulate SIRPα to the cell surface promote neutrophil accumulation during inflammation in vivo.
Collapse
Affiliation(s)
- Åsa Stenberg
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Maier NK, Crown D, Liu J, Leppla SH, Moayeri M. Arsenic trioxide and other arsenical compounds inhibit the NLRP1, NLRP3, and NAIP5/NLRC4 inflammasomes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:763-70. [PMID: 24337744 PMCID: PMC3884817 DOI: 10.4049/jimmunol.1301434] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inflammasomes are large cytoplasmic multiprotein complexes that activate caspase-1 in response to diverse intracellular danger signals. Inflammasome components termed nucleotide-binding oligomerization domain-like receptor (NLR) proteins act as sensors for pathogen-associated molecular patterns, stress, or danger stimuli. We discovered that arsenicals, including arsenic trioxide and sodium arsenite, inhibited activation of the NLRP1, NLRP3, and NAIP5/NLRC4 inflammasomes by their respective activating signals, anthrax lethal toxin, nigericin, and flagellin. These compounds prevented the autoproteolytic activation of caspase-1 and the processing and secretion of IL-1β from macrophages. Inhibition was independent of protein synthesis induction, proteasome-mediated protein breakdown, or kinase signaling pathways. Arsenic trioxide and sodium arsenite did not directly modify or inhibit the activity of preactivated recombinant caspase-1. Rather, they induced a cellular state inhibitory to both the autoproteolytic and substrate cleavage activities of caspase-1, which was reversed by the reactive oxygen species scavenger N-acetylcysteine but not by reducing agents or NO pathway inhibitors. Arsenicals provided protection against NLRP1-dependent anthrax lethal toxin-mediated cell death and prevented NLRP3-dependent neutrophil recruitment in a monosodium urate crystal inflammatory murine peritonitis model. These findings suggest a novel role in inhibition of the innate immune response for arsenical compounds that have been used as therapeutics for a few hundred years.
Collapse
Affiliation(s)
- Nolan K. Maier
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Devorah Crown
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jie Liu
- Center for Molecular Medicine, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen H. Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
138
|
Deronic A, Helmersson S, Leanderson T, Ivars F. The quinoline-3-carboxamide paquinimod (ABR-215757) reduces leukocyte recruitment during sterile inflammation: leukocyte- and context-specific effects. Int Immunopharmacol 2013; 18:290-7. [PMID: 24370393 DOI: 10.1016/j.intimp.2013.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/03/2013] [Accepted: 12/09/2013] [Indexed: 12/30/2022]
Abstract
Quinoline-3-carboxamides (Q-compounds) are currently in clinical development for both autoimmune disease and cancer. We have previously shown that the Q-compound paquinimod (ABR-215757) significantly ameliorates disease symptoms in several mouse models of human inflammatory disease. Considering that recruitment of inflammatory cells into tissue is a common denominator of these models, we have in this report investigated whether paquinimod would interfere with cell accumulation during sterile peritoneal inflammation. To mimic the cell recruitment elicited by tissue injury, we used necrotic cells to induce the acute inflammatory response. We show that per oral treatment with paquinimod significantly reduced the accumulation of Ly6C(hi) inflammatory monocytes and eosinophils, but not neutrophils, in this model, and that this correlated with reduced number of such cells also in the omentum. Treatment also reduced the accumulation of these cell populations at a subcutaneous site of inflammation. In alum-induced inflammation, however, neutrophils were the dominant cell population and paquinimod failed to reduce the accumulation of inflammatory cells. Taken together, our results indicate that paquinimod selectively inhibits cell recruitment during acute sterile inflammation, but that this effect is context-dependent. These data have important implications for the understanding of the mechanism of action of Q-compounds in both pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Adnan Deronic
- Immunology Group, Section for Immunology, Department of Experimental Medical Science, Lund University, Sweden.
| | - Sofia Helmersson
- Immunology Group, Section for Immunology, Department of Experimental Medical Science, Lund University, Sweden.
| | - Tomas Leanderson
- Immunology Group, Section for Immunology, Department of Experimental Medical Science, Lund University, Sweden.
| | - Fredrik Ivars
- Immunology Group, Section for Immunology, Department of Experimental Medical Science, Lund University, Sweden.
| |
Collapse
|
139
|
Abstract
Innate immune response pathways and metabolic pathways are evolutionarily conserved throughout species and are fundamental to survival. As such, the regulation of whole-body and cellular metabolism is intimately integrated with immune responses. However, the introduction of new variables to this delicate evolutionarily conserved physiological interaction can lead to deleterious consequences for organisms as a result of inappropriate immune responses. In recent decades, the prevalence and incidence of metabolic diseases associated with obesity have dramatically increased worldwide. As a recently acquired human characteristic, obesity has exposed the critical role of innate immune pathways in multiple metabolic pathophysiological processes. Here, we review recent evidence that highlights inflammasomes as critical sensors of metabolic perturbations in multiple tissues and their role in the progression of highly prevalent metabolic diseases.
Collapse
Affiliation(s)
- Jorge Henao-Mejia
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520;
| | | | | | | |
Collapse
|
140
|
Zandi-Nejad K, Takakura A, Jurewicz M, Chandraker AK, Offermanns S, Mount D, Abdi R. The role of HCA2 (GPR109A) in regulating macrophage function. FASEB J 2013; 27:4366-74. [PMID: 23882124 DOI: 10.1096/fj.12-223933] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We investigated the novel role of HCA2 (GPR109A) and its ligand nicotinic acid in regulating macrophage function. Hca2 expression in the RAW264.7 murine macrophage cell line is strongly induced by LPS treatment and correlates with the expression of TNF-α. Treatment with 300 μM nicotinic acid (reported EC50 3 μM, peak plasma concentration 50-300 μM), significantly inhibited TNF-α, IL-6, IL-12p40, and IL-1β production (P<0.05) in LPS (1 ng/ml)-stimulated wild-type murine bone marrow-derived macrophages (BMMs) but failed to do so in Hca2(-/-) BMMs. Treatment with nicotinic acid reduced nuclear factor κB (NF-κB) activation levels by 43% (P<0.03) in wild-type BMMs 6 h after LPS stimulation but not in Hca2(-/-) BMMs. Nicotinic acid significantly inhibited wild-type BMM chemotaxis (P<0.001), but had no effect on the chemotaxis of Hca2(-/-) BMMs. A significant increase in low-density lipoprotein uptake by both wild-type (P<0.006) and Hca2(-/-) BMMs (P<0.03) in response to LPS was observed, which was significantly suppressed by nicotinic acid in wild-type BMMs (P<0.04) but not in Hca2(-/-) BMMs. Our results suggest that the nicotinic acid-HCA2 axis is a novel negative regulator of macrophage activation.
Collapse
Affiliation(s)
- Kambiz Zandi-Nejad
- 2Division of Nephrology, 330 Brookline Ave., E/RN 304, Boston, MA 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
141
|
Mechanistic aspects of inflammation and clinical management of inflammation in acute gouty arthritis. J Clin Rheumatol 2013; 19:19-29. [PMID: 23319019 DOI: 10.1097/rhu.0b013e31827d8790] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been recently demonstrated that interleukin 1β (IL-1β) plays a central role in monosodium urate crystal-induced inflammation and that the NALP3 inflammasome plays a major role in IL-1β production. These discoveries have offered new insights into the pathogenesis of acute gouty arthritis. In this review, we discuss the molecular mechanisms by which monosodium urate crystals induce acute inflammation and examine the mechanisms of action (MOAs) of traditional anti-inflammatory drugs (e.g., nonsteroidal anti-inflammatory drugs, colchicine, and glucocorticoids) and biologic agents (e.g., the IL-1β antagonists anakinra, rilonacept, and canakinumab) to understand how their MOAs contribute to their safety profiles. Traditional anti-inflammatory agents may act on the IL-1β pathway at some level; however, their MOAs are broad-ranging, unspecific, and biologically complex. This lack of specificity may explain the range of systemic adverse effects associated with them. The therapeutic margins of nonsteroidal anti-inflammatory drugs, colchicine, and glucocorticoids are particularly low in elderly patients and in patients with cardiovascular, metabolic, or renal comorbidities that are frequently associated with gouty arthritis. In contrast, the IL-1β antagonists act on very specific targets of inflammation, which may decrease the potential for systemic adverse effects, although infrequent but serious adverse events (including infection and administration reactions) have been reported. Because these IL-1β antagonists target an early event immediately downstream from NALP3 inflammasome activation, they may provide effective alternatives to traditional agents with minimal systemic adverse effects. Results of ongoing trials of IL-1β antagonists will likely provide clarification of their potential role in the management of acute gouty arthritis.
Collapse
|
142
|
Steiger S, Harper JL. Neutrophil cannibalism triggers transforming growth factor β1 production and self regulation of neutrophil inflammatory function in monosodium urate monohydrate crystal-induced inflammation in mice. ACTA ACUST UNITED AC 2013; 65:815-23. [DOI: 10.1002/art.37822] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 12/04/2012] [Indexed: 01/16/2023]
|
143
|
Neutrophils, IL-1β, and gout: is there a link? Semin Immunopathol 2013; 35:501-12. [DOI: 10.1007/s00281-013-0361-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 01/09/2013] [Indexed: 01/09/2023]
|
144
|
|
145
|
Grainger R, McLaughlin RJ, Harrison AA, Harper JL. Hyperuricaemia elevates circulating CCL2 levels and primes monocyte trafficking in subjects with inter-critical gout. Rheumatology (Oxford) 2012. [PMID: 23204548 DOI: 10.1093/rheumatology/kes326] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To investigate the effect of hyperuricaemia on serum chemokine (C-C motif) ligand 2 (CCL2) levels and blood monocytes in people with gout. METHODS Whole blood was collected from subjects with a history of acute or chronic gout but not currently experiencing an attack of gout, subjects with asymptomatic hyperuricaemia and healthy individuals with normouricaemia. Serum concentrations of CCL2 were measured by bead array and levels of CD14(+)/CD11b(+) blood monocytes determined by flow cytometry. RESULTS Subjects with gout and asymptomatic hyperuricaemia had higher serum levels of CCL2 and showed an increase in the percentage of circulating CD14(+) monocytes compared with subjects with normouricaemia. CONCLUSION Hyperuricaemia causes elevated serum CCL2 levels and increased monocyte recruitment that may be driven by soluble uric acid-induced CCL2 production. Hyperuricaemia may initiate subclinical priming of circulating blood monocytes for adhesion and trafficking during a gout attack.
Collapse
Affiliation(s)
- Rebecca Grainger
- Malaghan Institute of Medical Research, PO Box 7060, Wellington 6242, New Zealand
| | | | | | | |
Collapse
|
146
|
Burns CM, Wortmann RL. Latest evidence on gout management: what the clinician needs to know. Ther Adv Chronic Dis 2012; 3:271-86. [PMID: 23342241 PMCID: PMC3539261 DOI: 10.1177/2040622312462056] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Until recently, the last drug approved for the treatment of gout by the United States Food and Drug Administration was allopurinol in 1966. Since 2008, two new drugs for the treatment of gout, febuxostat and pegloticase, have been approved in the US. Febuxostat has been approved in the EU and pegloticase approval is anticipated. A new single-ingredient colchicine preparation is available in the US, and the treatment recommendations for the use of colchicine in acute gout have evolved, now favoring a low-dose regimen. Several other exciting drugs are in development. Herein, we review some of basic principles in the diagnosis and staging of gout. We then examine current treatment principles, with particular attention to febuxostat and pegloticase, offering suggestions as to where they might fit into a modern therapeutic algorithm for gout treatment. We then present available data on several exciting new agents in development, including interleukin-1 inhibitors, and relate them to advances in our understanding of gout pathogenesis. We conclude with some important nonpharmacologic principles for optimal management of this ancient and eminently treatable disease. Dedicated gout research, going on quietly in the background of other breathtaking advances in rheumatology, is now paying off. This comes at a time when the number of patients affected by gout continues to rise, mainly due to an epidemic of obesity. An effort to improve lifestyle choices as a society and better management of the disease by clinicians should have a positive impact on gout incidence and outcome in our lifetimes.
Collapse
Affiliation(s)
- Christopher M Burns
- Dartmouth Medical School, Rheumatology, One Medical Center Drive, Lebanon, NH 03768, USA
| | | |
Collapse
|
147
|
Amaral FA, Costa VV, Tavares LD, Sachs D, Coelho FM, Fagundes CT, Soriani FM, Silveira TN, Cunha LD, Zamboni DS, Quesniaux V, Peres RS, Cunha TM, Cunha FQ, Ryffel B, Souza DG, Teixeira MM. NLRP3 inflammasome-mediated neutrophil recruitment and hypernociception depend on leukotriene B(4) in a murine model of gout. ACTA ACUST UNITED AC 2012; 64:474-84. [PMID: 21952942 DOI: 10.1002/art.33355] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Deposition of monosodium urate monohydrate (MSU) crystals in the joints promotes an intense inflammatory response and joint dysfunction. This study evaluated the role of the NLRP3 inflammasome and 5-lipoxygenase (5-LOX)-derived leukotriene B(4) (LTB(4) ) in driving tissue inflammation and hypernociception in a murine model of gout. METHODS Gout was induced by injecting MSU crystals into the joints of mice. Wild-type mice and mice deficient in NLRP3, ASC, caspase 1, interleukin-1β (IL-1β), IL-1 receptor type I (IL-1RI), IL-18R, myeloid differentiation factor 88 (MyD88), or 5-LOX were used. Evaluations were performed to assess neutrophil influx, LTB(4) activity, cytokine (IL-1β, CXCL1) production (by enzyme-linked immunosorbent assay), synovial microvasculature cell adhesion (by intravital microscopy), and hypernociception. Cleaved caspase 1 and production of reactive oxygen species (ROS) were analyzed in macrophages by Western blotting and fluorometric assay, respectively. RESULTS Injection of MSU crystals into the knee joints of mice induced neutrophil influx and neutrophil-dependent hypernociception. MSU crystal-induced neutrophil influx was CXCR2-dependent and relied on the induction of CXCL1 in an NLRP3/ASC/caspase 1/IL-1β/MyD88-dependent manner. LTB(4) was produced rapidly after injection of MSU crystals, and this was necessary for caspase 1-dependent IL-1β production and consequent release of CXCR2-acting chemokines in vivo. In vitro, macrophages produced LTB(4) after MSU crystal injection, and LTB(4) was relevant in the MSU crystal-induced maturation of IL-1β. Mechanistically, LTB(4) drove MSU crystal-induced production of ROS and ROS-dependent activation of the NLRP3 inflammasome. CONCLUSION These results reveal the role of the NLRP3 inflammasome in mediating MSU crystal-induced inflammation and dysfunction of the joints, and highlight a previously unrecognized role of LTB(4) in driving NLRP3 inflammasome activation in response to MSU crystals, both in vitro and in vivo.
Collapse
|
148
|
The Role of NLR-related Protein 3 Inflammasome in Host Defense and Inflammatory Diseases. Int Neurourol J 2012; 16:2-12. [PMID: 22500248 PMCID: PMC3321399 DOI: 10.5213/inj.2012.16.1.2] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 03/20/2012] [Indexed: 12/22/2022] Open
Abstract
Among a number of innate receptors, the nucleotide-binding domain leucine-rich repeat containing (NLR) nucleotide oligomerization domain (NOD)-like receptor families are involved in the recognition of cytosolic pathogen- or danger-associated molecules. Activation of these specific sets of receptors leads to the assembly of a multiprotein complex, the inflammasome, leading to the activation of caspase-1 and maturation of the cytokines interleukin (IL)-1β, IL-18, and IL-33. Among NLRs, NLR-related protein 3 (NLRP3) is one of the best-characterized receptors that activates the inflammasome. There is no doubt that NLRP3 inflammasome activation is important for host defense and effective pathogen clearance against fungal, bacterial, and viral infection. In addition, mounting evidence indicates that the NLRP3 inflammasome plays a role in a variety of inflammatory diseases, including gout, atherosclerosis, and type II diabetes, as well as under conditions of cellular stress or injury. Here, we review recent advances in our understanding of the role of the NLRP3 inflammasome in host defense and various inflammatory diseases.
Collapse
|
149
|
Sakamaki I, Inai K, Tsutani H. Safety of intradermal injection of monosodium urate crystals as a vaccine carrier in volunteers. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2012; 30:1077-84. [PMID: 22132960 DOI: 10.1080/15257770.2011.597368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Monosodium urate (MSU) crystals are known to induce gouty arthritis, but also evoke specific cell immunity and work as an adjuvant by delivering several kinds of binding proteins, including idiotypic cancer vaccine peptides into dendritic cells. To investigate the potency of MSU crystals as a cancer vaccine carrier in vivo, this preclinical study examined whether intradermal injection of MSU crystals was safe for healthy adults. Subjects comprised 12 volunteers. Four different dose levels of MSU crystals were injected as follows: 2 μg (n = 3), 20 μg (n = 3), 200 μg (n = 3), or 2000 μg (n = 3). At 24 hours after administration, documented erythema was seen around the injection site in a dose-dependent manner, particularly in all adults with MSU dose ≥200 μg. However, redness was limited to the grade I level of the National Cancer Institute toxicity criteria. Serum uric acid levels did not show any change before and after injection. Moreover, neither gouty arthritis nor tophi developed in any volunteers, indicating that intradermal injection of MSU crystals did not induce systemic inflammation at the doses that evoked significant local inflammation. These findings suggest that intradermal injection of MSU crystals is fundamentally safe and should be made available for clinical trials using MSU-crystal-conjugated cancer vaccines.
Collapse
Affiliation(s)
- Ippei Sakamaki
- Division of Clinical Research, National Hospital Organization Awara Hospital, Fukui, Japan
| | | | | |
Collapse
|
150
|
Mitroulis I, Kambas K, Chrysanthopoulou A, Skendros P, Apostolidou E, Kourtzelis I, Drosos GI, Boumpas DT, Ritis K. Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout. PLoS One 2011; 6:e29318. [PMID: 22195044 PMCID: PMC3241704 DOI: 10.1371/journal.pone.0029318] [Citation(s) in RCA: 283] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 11/24/2011] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Gout is a prevalent inflammatory arthritis affecting 1-2% of adults characterized by activation of innate immune cells by monosodium urate (MSU) crystals resulting in the secretion of interleukin-1β (IL-1β). Since neutrophils play a major role in gout we sought to determine whether their activation may involve the formation of proinflammatory neutrophil extracellular traps (NETs) in relation to autophagy and IL-1β. METHODOLOGY/PRINCIPAL FINDINGS Synovial fluid neutrophils from six patients with gout crisis and peripheral blood neutrophils from six patients with acute gout and six control subjects were isolated. MSU crystals, as well as synovial fluid or serum obtained from patients with acute gout, were used for the treatment of control neutrophils. NET formation was assessed using immunofluorescence microscopy. MSU crystals or synovial fluid or serum from patients induced NET formation in control neutrophils. Importantly, NET production was observed in neutrophils isolated from synovial fluid or peripheral blood from patients with acute gout. NETs contained the alarmin high mobility group box 1 (HMGB1) supporting their pro-inflammatory potential. Inhibition of phosphatidylinositol 3-kinase signaling or phagolysosomal fusion prevented NET formation, implicating autophagy in this process. NET formation was driven at least in part by IL-1β as demonstrated by experiments involving IL-1β and its inhibitor anakinra. CONCLUSIONS/SIGNIFICANCE These findings document for the first time that activation of neutrophils in gout is associated with the formation of proinflammatory NETs and links this process to both autophagy and IL-1β. Modulation of the autophagic machinery may represent an additional therapeutic study in crystalline arthritides.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Konstantinos Kambas
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Akrivi Chrysanthopoulou
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Panagiotis Skendros
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Eirini Apostolidou
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Ioannis Kourtzelis
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Georgios I. Drosos
- Department of Orthopaedic Surgery, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Dimitrios T. Boumpas
- Department of Rheumatology, Clinical Immunology and Allergy, University Hospital, Medical School, University of Crete, Heraklion, Greece
| | - Konstantinos Ritis
- First Department of Internal Medicine, Democritus University of Thrace, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
- * E-mail:
| |
Collapse
|