1
|
Roman Y. Correlation of systemic immune inflammation and serum uric acid with gout: based on NHANES. Clin Rheumatol 2025:10.1007/s10067-025-07329-8. [PMID: 39821127 DOI: 10.1007/s10067-025-07329-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025]
Affiliation(s)
- Youssef Roman
- Department of Pharmacy Practice and Administrative Sciences, L.S. Skaggs College of Pharmacy, Idaho State University, 1311 E Central Dr, Meridian, ID, 83642, USA.
- Boise VA Medical Center, 500 W Fort St, Boise, ID, 83702, USA.
| |
Collapse
|
2
|
Yan J, Liu Y. Correlation of systemic immune inflammation and serum uric acid with gout: based on NHANES. Clin Rheumatol 2025; 44:425-432. [PMID: 39681768 DOI: 10.1007/s10067-024-07271-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 11/08/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
OBJECTIVES This study aimed to explore the relationship of systemic immune inflammation index (SII) and uric acid with gout using NHANES 2017-2018 data. METHOD Data were collected from the 2017-2018 circle of the NHANES database. SII was calculated based on the counts of lymphocytes (LC), neutrophils (NC), and platelets (PC). Data on gout patients were collected from questionnaires. Logistic regression analysis and subgroup analysis were used to explore the relationship between SII and gout. RESULTS A total of 4517 participants were included in the study, with 273 individuals in the gout group and 4244 in the non-gout group. Logistic regression analysis showed that SII and serum uric acid were higher in the gout group than those in the non-gout group. SII ≥ 511.8 and uric acid ≥ 7.0 mg/dL may be positively associated with gout. Further subgroup analysis revealed that the level of SII was positively associated with gout in the female group and the group with eGFR of 60-89.9 mL/min/1.73 m2. Serum uric acid was positively associated with gout in the age, hypertension, hyperglycemia, and male groups and the group with eGFR ≥ 60 mL/min/1.73 m2. CONCLUSIONS There are positive correlations of SII and serum uric acid with gout. Our study suggests that SII could be a novel, valuable, and feasible inflammatory marker for gout. Key Points • Gout is a joint disease associated with hyperuricemia. The mechanism of the disease involves multiple complex factors, including metabolic disorders and inflammation. • Systemic immune inflammation index (Sll), as an evaluation index of systemic inflammatory responses, can reflect the body's inflammatory changes when the immune system is continuously activated with chronic inflammation of joints and other tissues. • We used the NHANES database to explore the relationship of SII and uric acid with gout, providing a basis for the diagnosis of gout and prognostic assessment of gout patients.
Collapse
Affiliation(s)
- Jin Yan
- Department of Endocrinology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China.
| | - Yanmei Liu
- Department of Endocrinology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China
| |
Collapse
|
3
|
Sanchez C, Campeau A, Liu-Bryan R, Mikuls TR, O'Dell JR, Gonzalez DJ, Terkeltaub R. Effective xanthine oxidase inhibitor urate lowering therapy in gout is linked to an emergent serum protein interactome of complement and inflammation modulators. Sci Rep 2024; 14:24598. [PMID: 39426967 PMCID: PMC11490615 DOI: 10.1038/s41598-024-74154-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024] Open
Abstract
Urate-lowering treatment (ULT) to target with xanthine oxidase inhibitors (XOIs) paradoxically causes early increase in gouty arthritis flares. Because delayed reduction in flare burden is mechanistically unclear, we tested for ULT inflammation responsiveness markers. Unbiased proteomics analyzed blood samples (baseline, 48 weeks ULT) in two, independent ULT out trial cohorts (n = 19, n = 30). STRING-db and multivariate analyses supplemented determinations of altered proteins via Wilcoxon matched pairs signed rank testing in XOI ULT responders. Mechanistic studies characterized proteomes of cultured XOI-treated murine bone marrow macrophages (BMDMs). At 48 weeks ULT, serum urate normalized in all gout patients, and flares declined in association with significantly altered proteins (p < 0.05) in clustering and proteome networks in sera and peripheral blood mononuclear cells. Sera demonstrated altered complement activation and regulatory gene ontology biologic processes. In both cohorts, a treatment-emergent serum interactome included key gouty inflammation mediators (C5, IL-1B, CXCL8, IL6). Last, febuxostat treatment decreased complement activation biologic process proteins in cultured BMDMs. Reduced gout flares are linked with a XOI treatment-emergent serum protein interactome that includes inflammation regulators, associated with altered complement activation and regulatory biologic processes. Serum and leukocyte proteomics could help identify when gouty inflammatory processes begin to subside in response to ULT.Trial Registration: ClinicalTrials.gov Identifier NCT02579096, posted October 19, 2015.
Collapse
Affiliation(s)
- Concepcion Sanchez
- Department of Pharmacology, University of California, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, and Collaborative Center for Multiplexed Proteomics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Anaamika Campeau
- Department of Pharmacology, University of California, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, and Collaborative Center for Multiplexed Proteomics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ru Liu-Bryan
- Division of Rheumatology, Autoimmunity and Inflammation, Department of Medicine, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA
| | - Ted R Mikuls
- Department of Internal Medicine, University of Nebraska Medical Center, MSB 5544, 983331, Omaha, NE, 68198-3331, USA
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - James R O'Dell
- Department of Internal Medicine, University of Nebraska Medical Center, MSB 5544, 983331, Omaha, NE, 68198-3331, USA
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, and Collaborative Center for Multiplexed Proteomics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Robert Terkeltaub
- Division of Rheumatology, Autoimmunity and Inflammation, Department of Medicine, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
4
|
Barry A, Helget LN, Androsenko M, Wu H, Kramer B, Newcomb JA, Brophy MT, Davis-Karim A, England BR, Ferguson R, Pillinger MH, Neogi T, Palevsky PM, Merriman TR, O’Dell JR, Mikuls TR. Comparison of Gout Flares With the Initiation of Treat-to-Target Allopurinol and Febuxostat: A Post-Hoc Analysis of a Randomized Multicenter Trial. Arthritis Rheumatol 2024; 76:1552-1559. [PMID: 38925627 PMCID: PMC11421957 DOI: 10.1002/art.42927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/06/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Initiating urate-lowering therapy (ULT) in gout can precipitate arthritis flares. There have been limited comparisons of flare risk during the initiation and escalation of allopurinol and febuxostat, administered as a treat-to-target strategy with optimal anti-inflammatory prophylaxis. METHODS This was a post-hoc analysis of a 72-week randomized, double-blind, placebo-controlled, noninferiority trial comparing the efficacy of allopurinol and febuxostat. For this analysis, the occurrence of flares was examined during weeks 0 to 24 when ULT was initiated and titrated to a serum urate (sUA) goal of less than 6 mg/dl (<5 mg/dl if tophi). Flares were assessed at regular intervals through structured participant interviews. Predictors of flare, including treatment assignment, were examined using multivariable Cox proportional hazards regression. RESULTS Study participants (n = 940) were predominantly male (98.4%) and had a mean age of 62.1 years with approximately equal proportions receiving allopurinol or febuxostat. Mean baseline sUA was 8.5 mg/dl and all participants received anti-inflammatory prophylaxis (90% colchicine). In a multivariable model, there were no significant associations of ULT treatment (hazard ratio [HR] 1.17; febuxostat vs allopurinol), ULT-dose escalation (HR 1.18 vs no escalation), prophylaxis type, or individual comorbidity with flare and no evidence of ULT-dose escalation interaction. Factors independently associated with flare risk during ULT initiation/escalation included younger age, higher baseline sUA, and absence of tophi. CONCLUSION These results demonstrate that gout flare risk during the initiation and titration of allopurinol is similar to febuxostat when these agents are administered according to a treat-to-target strategy using gradual ULT-dose titration and best practice gout flare prophylaxis.
Collapse
Affiliation(s)
- Austin Barry
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Lindsay N. Helget
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Maria Androsenko
- VA Boston Cooperative Studies Program Coordinating Center, Boston, Massachusetts, USA
| | - Hongsheng Wu
- VA Boston Cooperative Studies Program Coordinating Center, Boston, Massachusetts, USA
- Babson College, Wellesley, Massachusetts, USA
| | - Bridget Kramer
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jeff A. Newcomb
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mary T. Brophy
- VA Boston Cooperative Studies Program Coordinating Center, Boston, Massachusetts, USA
- School of Medicine, VA Boston Health Care System, Boston University, Boston, Massachusetts, USA
| | - Anne Davis-Karim
- VA Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, New Mexico, USA
| | - Bryant R. England
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ryan Ferguson
- VA Boston Cooperative Studies Program Coordinating Center, Boston, Massachusetts, USA
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Michael H. Pillinger
- VA New York Harbor Health Care System, New York, New York, USA:
- New York University Grossman School of Medicine, New York, New York, USA
| | - Tuhina Neogi
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Paul M. Palevsky
- VA Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Tony R. Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James R. O’Dell
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ted R. Mikuls
- Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
5
|
Rodriguez-Navarro C, Elert K, Ibañez-Velasco A, Monasterio-Guillot L, Andres M, Sivera F, Pascual E, Ruiz-Agudo E. Unraveling the pathological biomineralization of monosodium urate crystals in gout patients. Commun Biol 2024; 7:828. [PMID: 38972919 PMCID: PMC11228021 DOI: 10.1038/s42003-024-06534-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
Crystallization of monosodium urate monohydrate (MSU) leads to painful gouty arthritis. Despite extensive research it is still unknown how this pathological biomineralization occurs, which hampers its prevention. Here we show how inflammatory MSU crystals form after a non-inflammatory amorphous precursor (AMSU) that nucleates heterogeneously on collagen fibrils from damaged articular cartilage of gout patients. This non-classical crystallization route imprints a nanogranular structure to biogenic acicular MSU crystals, which have smaller unit cell volume, lower microstrain, and higher crystallinity than synthetic MSU. These distinctive biosignatures are consistent with the template-promoted crystallization of biotic MSU crystals after AMSU at low supersaturation, and their slow growth over long periods of time (possibly years) in hyperuricemic gout patients. Our results help to better understand gout pathophysiology, underline the role of cartilage damage in promoting MSU crystallization, and suggest that there is a time-window to treat potential gouty patients before a critical amount of MSU has slowly formed as to trigger a gout flare.
Collapse
Grants
- PID2021.125305NB.I00 Ministry of Economy and Competitiveness | Agencia Estatal de Investigación (Spanish Agencia Estatal de Investigación)
- B-RNM-574-UGR20 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
- RNM-179 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
- UCE-PP2016-05 Universidad de Granada (University of Granada)
Collapse
Affiliation(s)
- Carlos Rodriguez-Navarro
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain.
| | - Kerstin Elert
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
- Escuela de Estudios Arabes, Consejo Superior de Investigaciones Científicas (EEA-CSIC), C. Chapiz 22, 18010, Granada, Spain
| | - Aurelia Ibañez-Velasco
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
| | - Luis Monasterio-Guillot
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
| | - Mariano Andres
- Department of Clinical Medicine, Miguel Hernandez University, CN 332 s/n, 03550, Alicante, Spain
- Department of Rheumatology, Dr. Balmis General University Hospital, Alicante Institute for Health and Biomedical Research, Av. Pintor Baeza s/n, 03010, Alicante, Spain
| | - Francisca Sivera
- Department of Clinical Medicine, Miguel Hernandez University, CN 332 s/n, 03550, Alicante, Spain
- Department of Rheumatology, Elda General University Hospital, Carretera Elda-Sax s/n, 03600, Elda, Spain
| | - Eliseo Pascual
- Department of Clinical Medicine, Miguel Hernandez University, CN 332 s/n, 03550, Alicante, Spain
- Department of Rheumatology, Dr. Balmis General University Hospital, Alicante Institute for Health and Biomedical Research, Av. Pintor Baeza s/n, 03010, Alicante, Spain
| | - Encarnación Ruiz-Agudo
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
| |
Collapse
|
6
|
Pesante BD, Salimi M, Miller WL, Young HL, Jenkins TC, Parry JA. The Effect of Crystal Arthropathy on the Diagnostic Criteria of Native Septic Arthritis. J Am Acad Orthop Surg 2024; 32:570-575. [PMID: 38652879 DOI: 10.5435/jaaos-d-23-00857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/28/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION Distinguishing between septic arthritis and crystal arthropathy flares can be challenging. The purpose of this study was to determine how the presence of synovial crystals affects the diagnostic criteria of septic arthritis. METHODS A retrospective review identified patients undergoing joint aspirations to rule out native septic arthritis. Differences between septic arthritis presenting with and without synovial crystals were analyzed. A receiver-operating characteristic curve was plotted for laboratory markers to determine the area under the curve, or diagnostic accuracy, for septic arthritis and to evaluate thresholds that maximized sensitivity and specificity. RESULTS There were 302 joint aspirations in 267 patients. Septic arthritis was diagnosed in 17.9% (54/302). Patients with synovial crystals were less likely to have septic arthritis (4.2% [5/119] vs. 26.8% [49/183], P < 0.0001). Septic arthritis in patients with no synovial crystals was associated with fever and a higher synovial white blood cell (WBC) count, synovial polymorphonuclear cell percentage (PMN%), serum WBC, and C-reactive protein (CRP) ( P < 0.05). Septic arthritis in patients with synovial crystals was only associated with inability to bear weight and a higher synovial WBC and CRP ( P < 0.05). Synovial PMN% was considered nondiagnostic of septic arthritis (area under the curve 0.56) in patients with crystals while synovial WBC and CRP had acceptable (0.76) and excellent (0.83) diagnostic utility, respectively. The WBC and CRP value thresholds that maximized sensitivity and specificity for septic arthritis were greater in patients with crystals (21,600 vs. 17,954 cells/μL and 125 vs. 69 mg/L, respectively). DISCUSSION The presence of synovial crystals reduced the likelihood of septic arthritis and altered the laboratory diagnostic criteria. PMN% was nondiagnostic in the setting of synovial crystals.
Collapse
Affiliation(s)
- Benjamin D Pesante
- From the Department of Orthopaedics (Pesante, Salimi, and Parry) and the Department of Medicine - Infectious Disease (Miller, Young, and Jenkins), Denver Health Medical Center, University of Colorado School of Medicine, Denver, CO
| | | | | | | | | | | |
Collapse
|
7
|
Wang T, Zhang C, Zhou M, Zhou H, Zhang X, Liu H, Bai M, Xu Y, Yang F, Zhu F, Hao Q, Zhang T, Song S, Qi H, Liu Y. CD8 T cell-derived perforin regulates macrophage-mediated inflammation in a murine model of gout. Clin Rheumatol 2024; 43:2027-2034. [PMID: 38625643 DOI: 10.1007/s10067-024-06964-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVES Gout is characterized by hyperuricemia and recurrent inflammatory episodes caused by intra-articular crystal deposition of monosodium urate (MSU). There is a clear relationship between gout and metabolic syndrome. Recent evidence indicates that perforin plays a role in regulating glucose homeostasis and provides protection in diet-induced non-alcoholic steatohepatitis models. However, the impact of perforin on immune inflammation in gout remains unclear. METHODS We induced acute gout models in both wild-type (WT) mice and Prf1null mice by administering intra-articular injections of MSU crystals. We compared the ankle joint swelling and the histological score between the two groups. Furthermore, we investigated underlying mechanisms through in vitro co-culture experiments involving CD8 T cells and macrophages. RESULTS In this study, Prf1null mice showed significantly more pronounced ankle swelling with increased inflammatory cell infiltrations compared with WT mice 24 h after local MSU injection. Moreover, MSU-induced Prf1null mice exhibited increased accumulation of CD8 T cells but not NK cells. Perforin-deficient CD8 T cells displayed reduced cytotoxicity towards bone marrow-derived M0 and M1 macrophages and promoted TNF-α secretion from macrophage. CONCLUSIONS Perforin from CD8 T cells limits joint inflammation in mice with acute gout by downregulating macrophage-mediated inflammation. Key Points • Perforin deficiency increased swelling in the ankle joints of mice upon MSU injection. • Perforin deficiency is associated with increased immune cell recruitment and severe joint damage in gout. • Perforin regulated CD8 T cell accumulation in gout and promoted CD8 T cell cytotoxicity towards M0 and M1 macrophages. • CD8 T cell-derived perforin regulated pro-inflammatory cytokine secretion of macrophage.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Chunpan Zhang
- Department of Infectious Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingzhu Zhou
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Hang Zhou
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Huilan Liu
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Mingxin Bai
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Yuetong Xu
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Fan Yang
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Fengyunzhi Zhu
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Qiyuan Hao
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Tong Zhang
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Shuju Song
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Haiyu Qi
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China
| | - Yanying Liu
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, 95th, Yongan Road, Beijing, 100050, China.
| |
Collapse
|
8
|
Ma Z, Zeng P, Feng H, Ni L. Network pharmacology and molecular docking to explore the treatment potential and molecular mechanism of Si-Miao decoction against gouty arthritis. Medicine (Baltimore) 2024; 103:e38221. [PMID: 39259129 PMCID: PMC11142817 DOI: 10.1097/md.0000000000038221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 09/12/2024] Open
Abstract
Gouty arthritis (GA) is a common metabolic rheumatological disease. Si-Miao decoction has therapeutic effects on GA. In our study, we investigated the mechanism of Si-Miao decoction against GA using network pharmacology and molecular docking analytical methods. The Traditional Chinese Medicine Systems Pharmacology Database was used as the basis for screening the main targets and agents of the Si-Miao decoction, and the Genecards, OMIM, and Drugbank databases were used to screen GA-related targets. They were analyzed using Venn with the drug targets to obtain the intersection targets. We used Cytoscape 3.9.1 to draw the "Drugs-Compounds-Targets" network and the String database for creative protein-protein interaction networks of target genes and filtered core targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes were used to analyze the core targets. Molecular docking was performed using AutoDockTools to predict the binding capacity between nuclear targets and active components in the Si-Miao decoction. A total of 50 chemically active components containing 53 common targets of Si-Miao decoction anti-GA and 53 potential drug target proteins were identified. Core targets, namely, TNF, STAT3, SRC, PPARG, TLR4, PTGS2, MMP9, RELA, TGFB1, and SIRT1, were obtained through PPI network analysis. GO and KEGG analyses showed that the mechanism of anti-GA in Si-Miao decoction may proceed by regulating biological processes such as inflammatory factor levels, cell proliferation, apoptosis, and lipid and glucose metabolism, and modulating the NOD-like receptor signaling pathway, IL-17 signaling pathway, TNF signaling pathway, NF-kappa B signaling pathway, and Toll-like receptor signaling pathway. We further screened the core targets, including PTGS2, MMP9, and PPAGR, as receptor proteins based on their degree value and molecular docking with the main active compounds in Si-Miao decoction, and found that baicalein had high affinity. In conclusion, Si-Miao decoction, through anti-inflammatory, apoptosis-regulating, and anti-oxidative stress action mechanisms in the treatment of GA.
Collapse
Affiliation(s)
- Zebing Ma
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Chinese Medicine, Changsha, China
| | - Peng Zeng
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Haibo Feng
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Lili Ni
- Orthopedics (Orthopedic Group), The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
9
|
Sanchez C, Campeau A, Liu-Bryan R, Mikuls TR, O'Dell JR, Gonzalez DJ, Terkeltaub R. Effective xanthine oxidase inhibitor urate lowering therapy in gout is linked to an emergent serum protein interactome of complement activation and inflammation modulators. RESEARCH SQUARE 2024:rs.3.rs-4278877. [PMID: 38766125 PMCID: PMC11100878 DOI: 10.21203/rs.3.rs-4278877/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background Urate-lowering treatment (ULT) to target with xanthine oxidase inhibitors (XOIs) paradoxically causes early increase in gouty arthritis flares. Because delayed reduction in flare burden is mechanistically unclear, we tested for ULT inflammation responsiveness markers. Methods Unbiased proteomics analyzed blood samples (baseline, 48 weeks ULT) in two, independent ULT out trial cohorts (n = 19, n = 30). STRING-db and multivariate analyses supplemented determinations of altered proteins via Wilcoxon matched pairs signed rank testing in XOI ULT responders. Mechanistic studies characterized proteomes of cultured XOI-treated murine bone marrow macrophages (BMDMs). Results At 48 weeks ULT, serum urate normalized in all gout patients, and flares declined, with significantly altered proteins (p < 0.05) in clustering and proteome networks in sera and peripheral blood mononuclear cells. Serum proteome changes included decreased complement C8 heterotrimer C8A and C8G chains and chemokine PPBP/CXCL7, and increased urate crystal phagocytosis inhibitor sCD44. In both cohorts, a treatment-emergent serum interactome included key gouty inflammation mediators (C5, IL-1B, CXCL8, IL6). Last, febuxostat inhibited complement activation pathway proteins in cultured BMDMs. Conclusions Reduced gout flares are kinked with a XOI-treatment emergent complement- and inflammation-regulatory serum protein interactome. Serum and leukocyte proteomes could help identify onset of anti-inflammatory responsiveness to ULT in gout. Trial registration ClinicalTrials.gov Identifier: NCT02579096, posted October 19, 2015.
Collapse
|
10
|
Alabarse PG, Oliveira P, Qin H, Yan T, Migaud M, Terkeltaub R, Liu-Bryan R. The NADase CD38 is a central regulator in gouty inflammation and a novel druggable therapeutic target. Inflamm Res 2024; 73:739-751. [PMID: 38493256 PMCID: PMC11058052 DOI: 10.1007/s00011-024-01863-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/23/2024] [Accepted: 02/13/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVES Cellular NAD+ declines in inflammatory states associated with increased activity of the leukocyte-expressed NADase CD38. In this study, we tested the potential role of therapeutically targeting CD38 and NAD+ in gout. METHODS We studied cultured mouse wild type and CD38 knockout (KO) murine bone marrow derived macrophages (BMDMs) stimulated by monosodium urate (MSU) crystals and used the air pouch gouty inflammation model. RESULTS MSU crystals induced CD38 in BMDMs in vitro, associated with NAD+ depletion, and IL-1β and CXCL1 release, effects reversed by pharmacologic CD38 inhibitors (apigenin, 78c). Mouse air pouch inflammatory responses to MSU crystals were blunted by CD38 KO and apigenin. Pharmacologic CD38 inhibition suppressed MSU crystal-induced NLRP3 inflammasome activation and increased anti-inflammatory SIRT3-SOD2 activity in macrophages. BMDM RNA-seq analysis of differentially expressed genes (DEGs) revealed CD38 to control multiple MSU crystal-modulated inflammation pathways. Top DEGs included the circadian rhythm modulator GRP176, and the metalloreductase STEAP4 that mediates iron homeostasis, and promotes oxidative stress and NF-κB activation when it is overexpressed. CONCLUSIONS CD38 and NAD+ depletion are druggable targets controlling the MSU crystal- induced inflammation program. Targeting CD38 and NAD+ are potentially novel selective molecular approaches to limit gouty arthritis.
Collapse
Affiliation(s)
- Paulo Gil Alabarse
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
| | - Patricia Oliveira
- University of California San Diego, La Jolla, San Diego, CA, USA
- The Janssen Pharmaceutical Companies of Johnson & Johnson, La Jolla, San Diego, CA, USA
| | - Huaping Qin
- University of California San Diego, La Jolla, San Diego, CA, USA
| | - Tiffany Yan
- University of California San Diego, La Jolla, San Diego, CA, USA
- Gritstone Bio, Emeryville, CA, USA
| | - Marie Migaud
- Department of Pharmacology, F. Whiddon College of Medicine, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Robert Terkeltaub
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- University of California San Diego, La Jolla, San Diego, CA, USA
| | - Ru Liu-Bryan
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA.
- University of California San Diego, La Jolla, San Diego, CA, USA.
| |
Collapse
|
11
|
Sanchez C, Campeau A, Liu-Bryan R, Mikuls T, O'Dell J, Gonzalez D, Terkeltaub R. Sustained xanthine oxidase inhibitor treat to target urate lowering therapy rewires a tight inflammation serum protein interactome. RESEARCH SQUARE 2024:rs.3.rs-3770277. [PMID: 38260556 PMCID: PMC10802734 DOI: 10.21203/rs.3.rs-3770277/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Effective xanthine oxidoreductase inhibition (XOI) urate-lowering treatment (ULT) to target significantly reduces gout flare burden and synovitis between 1-2 years therapy, without clearing all monosodium urate crystal deposits. Paradoxically, treat to target ULT is associated with increased flare activity for at least 1 year in duration on average, before gout flare burden decreases. Since XOI has anti-inflammatory effects, we tested for biomarkers of sustained, effective ULT that alters gouty inflammation. Methods We characterized the proteome of febuxostat-treated murine bone marrow macrophages. Blood samples (baseline and 48 weeks ULT) were analyzed by unbiased proteomics in febuxostat and allopurinol ULT responders from two, independent, racially and ethnically distinct comparative effectiveness trial cohorts (n=19, n=30). STRING-db and multivariate analyses supplemented determinations of significantly altered proteins via Wilcoxon matched pairs signed rank testing. Results The proteome of cultured IL-1b-stimulated macrophages revealed febuxostat-induced anti-inflammatory changes, including for classical and alternative pathway complement activation pathways. At 48 weeks ULT, with altered purine metabolism confirmed by serum metabolomics, serum urate dropped >30%, to normal (<6.8 mg/dL) in all the studied patients. Overall, flares declined from baseline. Treated gout patient sera and peripheral blood mononuclear cells (PBMCs) showed significantly altered proteins (p<0.05) in clustering and proteome networks. CRP was not a useful therapy response biomarker. By comparison, significant serum proteome changes included decreased complement C8 heterotrimer C8A and C8G chains essential for C5b-9 membrane attack complex assembly and function; increase in the NLRP3 inflammasome activation promoter vimentin; increased urate crystal phagocytosis inhibitor sCD44; increased gouty inflammation pro-resolving mediator TGFB1; decreased phagocyte-recruiting chemokine PPBP/CXCL7, and increased monocyte/macrophage-expressed keratin-related proteins (KRT9,14,16) further validated by PBMC proteomics. STRING-db analyses of significantly altered serum proteins from both cohorts revealed a tight interactome network including central mediators of gouty inflammation (eg, IL-1B, CXCL8, IL6, C5). Conclusions Rewiring of inflammation mediators in a tight serum protein interactome was a biomarker of sustained XOI-based ULT that effectively reduced serum urate and gout flares. Monitoring of the serum and PBMC proteome, including for changes in the complement pathway could help determine onset and targets of anti-inflammatory changes in response to effective, sustained XOI-based ULT.Trial Registration: ClinicalTrials.gov Identifier: NCT02579096.
Collapse
|
12
|
Wang Y, Xu Y, Tan J, Ye J, Cui W, Hou J, Liu P, Li J, Wang S, Zhao Q. Anti-inflammation is an important way that Qingre-Huazhuo-Jiangsuan recipe treats acute gouty arthritis. Front Pharmacol 2023; 14:1268641. [PMID: 37881185 PMCID: PMC10597652 DOI: 10.3389/fphar.2023.1268641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/21/2023] [Indexed: 10/27/2023] Open
Abstract
Background: Acute gouty arthritis (AGA) significantly impairs patients' quality of life. Currently, existing therapeutic agents exhibit definite efficacy but also lead to serious adverse reactions. Therefore, it is essential to develop highly efficient therapeutic agents with minimal adverse reactions, especially within traditional Chinese medicine (TCM). Additionally, food polyphenols have shown potential in treating various inflammatory diseases. The Qingre-Huazhuo-Jiangsuan-Recipe (QHJR), a modification of Si-Miao-San (SMS), has emerged as a TCM remedy for AGA with no reported side effects. Recent research has also highlighted a strong genetic link to gout. Methods: The TCM System Pharmacology (TCMSP) database was used to collect the main chemical components of QHJR and AGA-related targets for predicting the metabolites in QHJR. HPLC-Q-Orbitrap-MS was employed to identify the ingredients of QHJR. The collected metabolites were then used to construct a Drugs-Targets Network in Cytoscape software, ranked based on their "Degree" of significance. Differentially expressed genes (DEGs) were screened in the Gene Expression Omnibus (GEO) database using GEO2R online analysis. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed. The DEGs were utilized to construct a Protein-Protein Interaction (PPI) Network via the STRING database. In vivo experimental validation was conducted using colchicine, QHJR, rapamycin (RAPA), and 3-methyladenine (3-MA) as controls to observe QHJR's efficacy in AGA. Synovial tissues from rats were collected, and qRT-PCR and Western blot assays were employed to investigate Ampk-related factors (Ampk, mTOR, ULK1), autophagy-related factors (Atg5, Atg7, LC3, p62), and inflammatory-related factors (NLRP3). ELISA assays were performed to measure inflammatory-related factor levels (IL-6, IL-1β, TNF-α), and H&E staining was used to examine tissue histology. Results: Network analysis screened out a total of 94 metabolites in QHJR for AGA. HPLC-Q-Orbitrap-MS analysis identified 27 of these metabolites. Notably, five metabolites (Neochlorogenic acid, Caffeic acid, Berberine, Isoliquiritigenin, Formononetin) were not associated with any individual herbal component of QHJR in TCMSP database, while six metabolites (quercetin, luteolin, formononetin, naringenin, taxifolin, diosgenin) overlapped with the predicted results from the previous network analysis. Further network analysis highlighted key components, such as Caffeic acid, cis-resveratrol, Apigenin, and Isoliquiritigenin. Other studies have found that their treatment of AGA is achieved through reducing inflammation, consistent with this study, laying the foundation for the mechanism study of QHJR against AGA. PPI analysis identified TNF, IL-6, and IL-1β as hub genes. GO and KEGG analyses indicated that anti-inflammation was a key mechanism in AGA treatment. All methods demonstrated that inflammatory expression increased in the Model group but was reversed by QHJR. Additionally, autophagy-related expression increased following QHJR treatment. The study suggested that AMPKα and p-AMPKα1 proteins were insensitive to 3 MA and RAPA, implying that AMPK may not activate autophagy directly but through ULK1 and mTOR. Conclusion: In conclusion, this study confirms the effectiveness of QHJR, a modified formulation of SMS (a classic traditional Chinese medicine prescription for treating gout), against AGA. QHJR, as a TCM formula, offers advantages such as minimal safety concerns and potential long-term use. The study suggests that the mechanism by which QHJR treats AGA may involve the activation of the AMPK/mTOR/ULK1 pathway, thereby regulating autophagy levels, reducing inflammation, and alleviating AGA. These findings provide new therapeutic approaches and ideas for the clinical treatment of AGA.
Collapse
Affiliation(s)
- Yazhuo Wang
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Xu
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingrui Tan
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiaxue Ye
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Weizhen Cui
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Hou
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peiyu Liu
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jianwei Li
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shiyuan Wang
- Institute of Nursing, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingyang Zhao
- Institute of Nursing, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Feng Y, Sun H, Zhu R, Tao J, Su R, Sun Y, Wang D. Effects of alcohol on the symptoms of gouty arthritis and taxonomic structure of gut microbiota in C57BL/6 mice. Front Microbiol 2023; 14:1257701. [PMID: 37771709 PMCID: PMC10525330 DOI: 10.3389/fmicb.2023.1257701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/30/2023] [Indexed: 09/30/2023] Open
Abstract
Gout is an acute arthritis caused by the elevated levels of serum uric acid (UA), and its prevalence has been rapidly increasing. Alcohol abuse could lead to a series of health problems. Multiple pieces of evidence suggest that alcohol intake affects the development and progression of gout, while the gut microbiota plays an important role in the development of gout and the long-term alcohol consumption could affect the stability of the gut microbiota. This study aimed to explore the effects of alcohol intake at different concentrations on gouty arthritis based on the gut microbiota. We investigated the effects of different concentrations of alcohol on gouty arthritis in mouse models of acute gouty arthritis established by injection of monosodium urate (MSU) crystals into C57BL/6 mice. The results indicated that the high-alcohol consumption not only exacerbated joint swelling and pain, increased the levels of UA, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), but also showed dramatic effects on the composition and structure of the gut microbiota in gouty mice. Two key microorganisms, Parasutterella and Alistipes, could aggravate gout symptoms through lipopolysaccharide biosynthesis, riboflavin metabolism, phenylalanine metabolism, and arginine and proline metabolisms. In conclusion, our study suggested that high-concentrations of alcohol altered the gut microbiota structure in gouty mice induced by MSU crystals, which could exacerbate gouty symptoms by enhancing pro-inflammatory pathways.
Collapse
Affiliation(s)
- Yu Feng
- Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haihui Sun
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ruilou Zhu
- Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianxing Tao
- Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rui Su
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yundong Sun
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Microbiology, School of Basic Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dawei Wang
- Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
14
|
Zhou Q, Liu L, Sun H, Liu S. Relief of gouty arthritis in rats by total saponins from Dioscorea nipponica Makino through suppression of neutrophil extracellular trap formation via the PI3K/AKT/mTOR axis. Exp Ther Med 2023; 26:447. [PMID: 37614416 PMCID: PMC10443068 DOI: 10.3892/etm.2023.12146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/29/2023] [Indexed: 08/25/2023] Open
Abstract
Gouty arthritis (GA) is an inflammatory disorder that is associated with elevated serum levels of uric acid. Total saponins from Dioscorea nipponica Makino (TSDN) are a natural component that ameliorates inflammation while also decreasing uric acid levels. The aim of the present study was to unravel the mechanism of TSDN in gouty rats in regard to regulation of the formation of neutrophil extracellular traps (NETs) via the PI3K/AKT/mTOR axis. A total of 40 Wistar rats were divided into 4 groups: normal, model, TSDN and rapamycin groups. Reverse-transcription-quantitative PCR (RT-qPCR) and western blot analysis were used to assess the mRNA and protein expression levels of the PI3K/AKT/mTOR axis. The formation of NETs was detected by immunohistochemical and immunofluorescent methods. ELISA was used to measure the levels of IL-1β and TNF-α. RT-qPCR and western blotting demonstrated that TSDN compromised the mRNA and protein expression levels of activated protein kinase (AMPK) and mTOR, as well as the mRNA expression levels of AKT and PTEN. Furthermore, it increased the protein expression levels of phosphorylated (p-) PI3K, p-AKT and p-AMPK. Immunohistochemical and immunofluorescent analyses revealed that TSDN decreased the protein expression levels of neutrophil elastase, proteinase 3, cathepsin G, lactoferrin and myeloperoxidase, as well as the number of citrullinated histone 3+ cells. TSDN also reduced the release of IL-1β and TNF-α. Overall, the anti-inflammatory action of TSDN in gouty rats may be realized by suppressing the formation of NETs by regulating the PI3K/AKT/mTOR axis.
Collapse
Affiliation(s)
- Qi Zhou
- Research Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Lin Liu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Huijuan Sun
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Shumin Liu
- Technological Innovation Team of Basic Theory Study Research of Institution of Higher Education in Heilongjiang, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
15
|
Li M, Tian F, Guo J, Li X, Ma L, Jiang M, Zhao J. Therapeutic potential of Coptis chinensis for arthritis with underlying mechanisms. Front Pharmacol 2023; 14:1243820. [PMID: 37637408 PMCID: PMC10450980 DOI: 10.3389/fphar.2023.1243820] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Arthritis is a common degenerative disease of joints, which has become a public health problem affecting human health, but its pathogenesis is complex and cannot be eradicated. Coptis chinensis (CC) has a variety of active ingredients, is a natural antibacterial and anti-inflammatory drug. In which, berberine is its main effective ingredient, and has good therapeutic effects on rheumatoid arthritis (RA), osteoarthritis (OA), gouty arthritis (GA). RA, OA and GA are the three most common types of arthritis, but the relevant pathogenesis is not clear. Therefore, molecular mechanism and prevention and treatment of arthritis are the key issues to be paid attention to in clinical practice. In general, berberine, palmatine, coptisine, jatrorrhizine, magnoflorine and jatrorrhizine hydrochloride in CC play the role in treating arthritis by regulating Wnt1/β-catenin and PI3K/AKT/mTOR signaling pathways. In this review, active ingredients, targets and mechanism of CC in the treatment of arthritis were expounded, and we have further explained the potential role of AHR, CAV1, CRP, CXCL2, IRF1, SPP1, and IL-17 signaling pathway in the treatment of arthritis, and to provide a new idea for the clinical treatment of arthritis by CC.
Collapse
Affiliation(s)
- Mengyuan Li
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Fei Tian
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinling Guo
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Xiankuan Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Ma
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miaomiao Jiang
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Zhao
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
- Department of Geriatric, Fourth Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
Russell MD, Roddy E, Rutherford AI, Ellis B, Norton S, Douiri A, Gulliford MC, Cope AP, Galloway JB. Treat-to-target urate-lowering therapy and hospitalizations for gout: results from a nationwide cohort study in England. Rheumatology (Oxford) 2023; 62:2426-2434. [PMID: 36355461 PMCID: PMC10321109 DOI: 10.1093/rheumatology/keac638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/03/2022] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE To investigate associations between treat-to-target urate-lowering therapy (ULT) and hospitalizations for gout. METHODS Using linked Clinical Practice Research Datalink and NHS Digital Hospital Episode Statistics data, we described the incidence and timing of hospitalizations for flares in people with index gout diagnoses in England from 2004-2020. Using Cox proportional hazards and propensity models, we investigated associations between ULT initiation, serum urate target attainment, colchicine prophylaxis, and the risk of hospitalizations for gout. RESULTS Of 292 270 people with incident gout, 7719 (2.64%) had one or more hospitalizations for gout, with an incidence rate of 4.64 hospitalizations per 1000 person-years (95% CI 4.54, 4.73). There was an associated increased risk of hospitalizations within the first 6 months after ULT initiation, when compared with people who did not initiate ULT [adjusted Hazard Ratio (aHR) 4.54; 95% CI 3.70, 5.58; P < 0.001]. Hospitalizations did not differ significantly between people prescribed vs not prescribed colchicine prophylaxis in fully adjusted models. From 12 months after initiation, ULT associated with a reduced risk of hospitalizations (aHR 0.77; 95% CI 0.71, 0.83; P < 0.001). In ULT initiators, attainment of a serum urate <360 micromol/l within 12 months of initiation associated with a reduced risk of hospitalizations (aHR 0.57; 95% CI 0.49, 0.67; P < 0.001) when compared with people initiating ULT but not attaining this target. CONCLUSION ULT associates with an increased risk of hospitalizations within the first 6 months of initiation but reduces hospitalizations in the long term, particularly when serum urate targets are achieved.
Collapse
Affiliation(s)
- Mark D Russell
- Correspondence to: Mark D. Russell, Centre for Rheumatic Diseases, Weston Education Centre, King’s College London, 10 Cutcombe Road, London SE5 9RJ, UK. E-mail:
| | | | - Andrew I Rutherford
- Department of Rheumatology, King’s College Hospital NHS Foundation Trust, London, UK
| | - Benjamin Ellis
- Department of Rheumatology, Imperial College Healthcare NHS Foundation Trust, London, UK
| | - Sam Norton
- Centre for Rheumatic Diseases, King’s College London, London, UK
| | - Abdel Douiri
- School of Population Health and Environmental Sciences, King’s College London, London, UK
| | - Martin C Gulliford
- School of Population Health and Environmental Sciences, King’s College London, London, UK
| | - Andrew P Cope
- Centre for Rheumatic Diseases, King’s College London, London, UK
| | - James B Galloway
- Centre for Rheumatic Diseases, King’s College London, London, UK
| |
Collapse
|
17
|
Baxter B, Sanders S, Patel SA, Martin A, West M. Pegloticase in Uncontrolled Gout: The Infusion Nurse Perspective. JOURNAL OF INFUSION NURSING 2023; 46:223-231. [PMID: 37406337 PMCID: PMC10306337 DOI: 10.1097/nan.0000000000000510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Infused biologics, such as pegloticase, are a core component of managing uncontrolled gout, which is increasing in prevalence. Pegloticase is often the last line of therapy for patients with uncontrolled gout; therefore, achieving a successful course of treatment is critical. The infusion nurse's role in patient education, serum uric acid monitoring, and patient medication compliance is essential for ensuring patient safety and maximizing the number of patients who benefit from a full treatment course of pegloticase. Infusion nurses are on the front lines with patients and need to be educated on potential negative effects associated with the medications they infuse, such as infusion reactions, as well as risk management methods like patient screening and monitoring. Further, patient education provided by the infusion nurse plays a large role in empowering the patient to become their own advocate during pegloticase treatment. This educational overview includes a model patient case for pegloticase monotherapy, as well as one for pegloticase with immunomodulation and a step-by-step checklist for infusion nurses to refer to throughout the pegloticase infusion process. A video abstract is available for this article at http://links.lww.com/JIN/A105.
Collapse
Affiliation(s)
- Britni Baxter
- Altus Infusion/Houk Rheumatology, Little Rock, Arkansas (Ms Baxter); Premier Specialty Network, Columbia, Missouri (Ms Sanders); and Horizon Therapeutics PLC, Deerfield, Illinois (Ms Patel, Ms Martin, and Mr West)
| | - Shayla Sanders
- Altus Infusion/Houk Rheumatology, Little Rock, Arkansas (Ms Baxter); Premier Specialty Network, Columbia, Missouri (Ms Sanders); and Horizon Therapeutics PLC, Deerfield, Illinois (Ms Patel, Ms Martin, and Mr West)
| | - Shilpa A. Patel
- Altus Infusion/Houk Rheumatology, Little Rock, Arkansas (Ms Baxter); Premier Specialty Network, Columbia, Missouri (Ms Sanders); and Horizon Therapeutics PLC, Deerfield, Illinois (Ms Patel, Ms Martin, and Mr West)
| | - Andrea Martin
- Altus Infusion/Houk Rheumatology, Little Rock, Arkansas (Ms Baxter); Premier Specialty Network, Columbia, Missouri (Ms Sanders); and Horizon Therapeutics PLC, Deerfield, Illinois (Ms Patel, Ms Martin, and Mr West)
| | - Michael West
- Altus Infusion/Houk Rheumatology, Little Rock, Arkansas (Ms Baxter); Premier Specialty Network, Columbia, Missouri (Ms Sanders); and Horizon Therapeutics PLC, Deerfield, Illinois (Ms Patel, Ms Martin, and Mr West)
| |
Collapse
|
18
|
Zhang WZ. Uric acid en route to gout. Adv Clin Chem 2023; 116:209-275. [PMID: 37852720 DOI: 10.1016/bs.acc.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Gout and hyperuricemia (HU) have generated immense attention due to increased prevalence. Gout is a multifactorial metabolic and inflammatory disease that occurs when increased uric acid (UA) induce HU resulting in monosodium urate (MSU) crystal deposition in joints. However, gout pathogenesis does not always involve these events and HU does not always cause a gout flare. Treatment with UA-lowering therapeutics may not prevent or reduce the incidence of gout flare or gout-associated comorbidities. UA exhibits both pro- and anti-inflammation functions in gout pathogenesis. HU and gout share mechanistic and metabolic connections at a systematic level, as shown by studies on associated comorbidities. Recent studies on the interplay between UA, HU, MSU and gout as well as the development of HU and gout in association with metabolic syndromes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular, renal and cerebrovascular diseases are discussed. This review examines current and potential therapeutic regimens and illuminates the journey from disrupted UA to gout.
Collapse
Affiliation(s)
- Wei-Zheng Zhang
- VIDRL, The Peter Doherty Institute, Melbourne, VIC, Australia.
| |
Collapse
|
19
|
Popov D, Jain L, Alhilali M, Dalbeth N, Poulsen RC. Monosodium urate crystals alter the circadian clock in macrophages leading to loss of NLRP3 inflammasome repression: Implications for timing of the gout flare. FASEB J 2023; 37:e22940. [PMID: 37243314 DOI: 10.1096/fj.202202035r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/21/2023] [Accepted: 04/17/2023] [Indexed: 05/28/2023]
Abstract
Gout is caused by monosodium urate (MSU) crystal deposition within joints. This leads to acute episodes of inflammation ("gout flares") driven by NLRP3 inflammasome activation in macrophages. Gout flares are frequently present during late night/early morning. The reason for this timing is unclear. Recent evidence suggests the NLRP3 inflammasome is under circadian control. The purpose of this study was to determine whether MSU crystals cause changes in the circadian clock in macrophages leading to time-of-day differences in NLRP3 inflammasome activation. Levels of circadian clock components were measured in undifferentiated "monocytic" and PMA-differentiated "macrophagic" THP-1 cells cultured with/without MSU crystals. Caspase-1 activity was measured to assess NLRP3 inflammasome activity. MSU crystal exposure resulted in minimal effects on clock genes in THP-1 monocytes but BMAL1, CRY1, PER2, and REV-ERBα showed altered expression with reduced protein levels of BMAL1 and REV-ERBα in THP-1 macrophages. REV-ERBα activation or BMAL1 over-expression resulted in reduced MSU crystal-induced caspase-1 activity. BMAL1 knockdown resulted in a further increase in MSU crystal-induced caspase-1 activity, but only at times of day when BMAL1 levels were naturally high. MSU crystal-induced NLRP3 inflammasome activation was greatest at the time of day when BMAL1 levels were naturally low. MSU crystals alter the expression of circadian clock components in THP-1 macrophages leading to loss of BMAL1 and REV-ERBα-mediated repression of NLRP3 inflammasome activity and time-of-day differences in susceptibility to inflammasome activation. Our findings suggest that the nocturnal risk of gout flare is at least partially a consequence of altered circadian control of immune cell function.
Collapse
Affiliation(s)
- Dmitry Popov
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Lekha Jain
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Mariam Alhilali
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
20
|
Chen J, Zheng Y, Gong S, Zheng Z, Hu J, Ma L, Li X, Yu H. Mechanisms of theaflavins against gout and strategies for improving the bioavailability. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154782. [PMID: 36990009 DOI: 10.1016/j.phymed.2023.154782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/04/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Gout is a crystal related arthropathy caused by monosodium urate deposition. At present, the identification of appropriate treatments and new drugs to reduce serum uric acid levels and gout risk is a major research area. PURPOSE Theaflavins are naturally occurring compounds characterized by a benzodiazepine skeleton. The significant benefits of theaflavins have been well documented. A large number of studies have been carried out and excellent anti-gout results have been achieved in recent years. STUDY DESIGN A comprehensive analysis of the mechanism of the anti-gout effect of theaflavins is presented through a literature review and network pharmacology prediction, and strategies for increasing the bioavailability of theaflavins are summarized. METHODS In this review, the active components and pharmacological mechanisms of theaflavins in the treatment of gout were summarized, and the relationship between theaflavins and gout, the relevant components, and the potential mechanisms of anti-gout action were clarified by reviewing the literature on the anti-gout effects of theaflavins and network pharmacology. RESULTS Theaflavins exert anti-gout effects by down regulating the gene and protein expression of glucose transporter 9 (GLUT9) and uric acid transporter 1 (URAT1), while upregulating the mRNA expression levels of organic anion transporter 1 (OAT1), organic cation transporter N1 (OCTN1), organic cation transporters 1/2 (Oct1/2), and organic anion transporter 2 (OAT2). Network pharmacology prediction indicate that theaflavins can regulate the AGE-RAGE and cancer signaling pathways through ATP-binding cassette subfamily B member 1 (ABCB1), recombinant mitogen activated protein kinase 14 (MAPK14), telomerase reverse tranase (TERT), signal transducer and activator of transcription 1 (STAT1), matrix metalloproteinase 2 (MMP2), B-cell lymphoma-2 (BCL2), and matrix metalloproteinase 14 (MMP14) targets for anti-gout effects. CONCLUSION This review presents the mechanisms of anti-gout action of theaflavins and strategies for improving the bioavailability of theaflavins, as well as providing research strategies for anti-gout treatment measures and the development of novel anti-gout drugs.
Collapse
Affiliation(s)
- Jingzi Chen
- Chinese Medicine Rehabilitation Department, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Yanchao Zheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Sihan Gong
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Zhigang Zheng
- Wuxi Teaturn Bioengineering Co., Ltd., Wuxi 214000, China
| | - Jing Hu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Lin Ma
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China
| | - Xiankuan Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, China.
| | - Hongjian Yu
- Wuxi Teaturn Bioengineering Co., Ltd., Wuxi 214000, China.
| |
Collapse
|
21
|
Elsaid K, Merriman TR, Rossitto LA, Liu-Bryan R, Karsh J, Phipps-Green A, Jay GD, Elsayed S, Qadri M, Miner M, Cadzow M, Dambruoso TJ, Schmidt TA, Dalbeth N, Chhana A, Höglund J, Ghassemian M, Campeau A, Maltez N, Karlsson NG, Gonzalez DJ, Terkeltaub R. Amplification of Inflammation by Lubricin Deficiency Implicated in Incident, Erosive Gout Independent of Hyperuricemia. Arthritis Rheumatol 2023; 75:794-805. [PMID: 36457235 PMCID: PMC10191887 DOI: 10.1002/art.42413] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/26/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
OBJECTIVE In gout, hyperuricemia promotes urate crystal deposition, which stimulates the NLRP3 inflammasome and interleukin-1β (IL-1β)-mediated arthritis. Incident gout without background hyperuricemia is rarely reported. To identify hyperuricemia-independent mechanisms driving gout incidence and progression, we characterized erosive urate crystalline inflammatory arthritis in a young female patient with normouricemia diagnosed as having sufficient and weighted classification criteria for gout according to the American College of Rheumatology (ACR)/EULAR gout classification criteria (the proband). METHODS We conducted whole-genome sequencing, quantitative proteomics, whole-blood RNA-sequencing analysis using serum samples from the proband. We used a mouse model of IL-1β-induced knee synovitis to characterize proband candidate genes, biomarkers, and pathogenic mechanisms of gout. RESULTS Lubricin level was attenuated in human proband serum and associated with elevated acute-phase reactants and inflammatory whole-blood transcripts and transcriptional pathways. The proband had predicted damaging gene variants of NLRP3 and of inter-α trypsin inhibitor heavy chain 3, an inhibitor of lubricin-degrading cathepsin G. Changes in the proband's serum protein interactome network supported enhanced lubricin degradation, with cathepsin G activity increased relative to its inhibitors, SERPINB6 and thrombospondin 1. Activation of Toll-like receptor 2 (TLR-2) suppressed levels of lubricin mRNA and lubricin release in cultured human synovial fibroblasts (P < 0.01). Lubricin blunted urate crystal precipitation and IL-1β induction of xanthine oxidase and urate in cultured macrophages (P < 0.001). In lubricin-deficient mice, injection of IL-1β in knees increased xanthine oxidase-positive synovial resident M1 macrophages (P < 0.05). CONCLUSION Our findings linked normouricemic erosive gout to attenuated lubricin, with impaired control of cathepsin G activity, compounded by deleterious NLRP3 variants. Lubricin suppressed monosodium urate crystallization and blunted IL-1β-induced increases in xanthine oxidase and urate in macrophages. The collective activities of articular lubricin that could limit incident and erosive gouty arthritis independently of hyperuricemia are subject to disruption by inflammation, activated cathepsin G, and synovial fibroblast TLR-2 signaling.
Collapse
Affiliation(s)
- Khaled Elsaid
- Chapman University School of Pharmacy, Irvine, California
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, and Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Leigh-Ana Rossitto
- Department of Pharmacology, School of Medicine, and Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, California
| | - Ru Liu-Bryan
- VA San Diego Healthcare System, San Diego, and Department of Medicine, UC San Diego, La Jolla, California
| | - Jacob Karsh
- The Ottawa Hospital, Division of Rheumatology, University of Ottawa, Canada
| | | | - Gregory D Jay
- Department of Emergency Medicine, Alpert School of Medicine, and Division of Biomedical Engineering, School of Engineering, Brown University, Rhode, Island
| | - Sandy Elsayed
- Chapman University School of Pharmacy, Irvine, California
| | | | - Marin Miner
- VA San Diego Healthcare System, San Diego, California
| | - Murray Cadzow
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Talia J Dambruoso
- Division of Biomedical Engineering, School of Engineering, Brown University, Rhode, Island
| | - Tannin A Schmidt
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, Connecticut
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Ashika Chhana
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Jennifer Höglund
- Department of Medical Biochemistry, Institute for Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, Department of Chemistry/Biochemistry, UC San Diego
| | - Anaamika Campeau
- Department of Pharmacology, School of Medicine, and Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, California
| | - Nancy Maltez
- The Ottawa Hospital, Division of Rheumatology, University of Ottawa, Canada
| | - Niclas G Karlsson
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway, and Department of Medical Biochemistry, Institute for Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - David J Gonzalez
- Department of Pharmacology, School of Medicine, and Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Center for Multiplexed Proteomics, Program for Integrative Omics and Data Science in Disease Prevention and Therapeutics, UC San Diego, La Jolla, California
| | - Robert Terkeltaub
- VA San Diego Healthcare System and Department of Medicine, UC San Diego
| |
Collapse
|
22
|
Xu H, Zhang B, Chen Y, Zeng F, Wang W, Chen Z, Cao L, Shi J, Chen J, Zhu X, Xue Y, He R, Ji M, Hua Y. Type II collagen facilitates gouty arthritis by regulating MSU crystallisation and inflammatory cell recruitment. Ann Rheum Dis 2023; 82:416-427. [PMID: 36109143 DOI: 10.1136/ard-2022-222764] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/03/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Increasing evidence suggests that impaired cartilage is a substantial risk factor for the progression from hyperuricaemia to gout. Since the relationship between cartilage matrix protein and gout flares remains unclear, we investigated its role in monosodium urate (MSU) crystallisation and following inflammation. METHODS Briefly, we screened for cartilage matrix in synovial fluid from gouty arthritis patients with cartilage injuries. After identifying a correlation between crystals and matrix molecules, we conducted image analysis and classification of crystal phenotypes according to their morphology. We then evaluated the differences between the cartilage matrix protein-MSU complex and the pure MSU crystal in their interaction with immune cells and identified the related signalling pathway. RESULTS Type II collagen (CII) was found to be enriched around MSU crystals in synovial fluid after cartilage injury. Imaging analysis revealed that CII regulated the morphology of single crystals and the alignment of crystal bows in the co-crystalline system, leading to greater phagocytosis and oxidative stress in macrophages. Furthermore, CII upregulated MSU-induced chemokine and proinflammatory cytokine expression in macrophages, thereby promoting the recruitment of leucocytes. Mechanistically, CII enhanced MSU-mediated inflammation by activating the integrin β1(ITGB1)-dependent TLR2/4-NF-κB signal pathway. CONCLUSION Our study demonstrates that the release of CII and protein-crystal adsorption modifies the crystal profile and promotes the early immune response in MSU-mediated inflammation. These findings open up a new path for understanding the relationship between cartilage injuries and the early immune response in gout flares.
Collapse
Affiliation(s)
- HanLin Xu
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Bohan Zhang
- State Key Laboratory of Surface Physics and Department of Physics, Human Phenome Institute, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Yiwu Research Institute, Fudan University, Shanghai, China
| | - Yaxin Chen
- State Key Laboratory of Surface Physics and Department of Physics, Human Phenome Institute, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Yiwu Research Institute, Fudan University, Shanghai, China
| | - Fengzhen Zeng
- School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Wenjuan Wang
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Ziyi Chen
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Ling Cao
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Shi
- School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Jun Chen
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Rui He
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China .,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - MinBiao Ji
- State Key Laboratory of Surface Physics and Department of Physics, Human Phenome Institute, Academy for Engineering and Technology, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Yiwu Research Institute, Fudan University, Shanghai, China
| | - YingHui Hua
- Department of Sports Medicine, Huashan Hospital,Fudan University, Shanghai, China
| |
Collapse
|
23
|
Fang S, Gao Y, Fang Y, Sun J, Xie Z. Mechanism Underlying the Action of Berberine in the Treatment of Gouty Arthritis Based on Network Pharmacology. Nat Prod Commun 2023. [DOI: 10.1177/1934578x221143627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Introduction: Gouty arthritis (GA) is induced by a purine metabolism disorder and monosodium urate (MSU) crystal-related inflammation. Berberine (BBR), extracted from Coptis chinensis, ameliorates MSU-induced GA. However, the mechanisms of BBR against GA remain to be fully elucidated. This study aimed to identify the key targets and pathways mediating the effects of BBR against GA using network pharmacology. Methods: BBR and GA targets were obtained from several databases, and the network of BBR-GA common targets was visualized using Cytoscape software. Protein–protein interaction (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the STRING and Database for Annotation, Visualization, and Integrated Discovery databases, respectively. Animal experiments were performed to determine the outcomes of the BBB intervention. The serum levels of IL-1β, IL-8, and IL-6 were detected using enzyme-linked immunosorbent assay. Results: Thirty-three common targets (including NF-κB, RelA, MAPK1, IL-6, and IL-1β) of BBR and GA were identified, and a network of common targets between BBR and GA was constructed. PPI analysis demonstrated that IL-1β, IL-6, TNF, MAPK, and RelA are key targets with high degree values. GO and KEGG pathway analyses revealed the involvement of inflammation-related biological processes and signaling pathways, such as the NF-κB, MAPK, and TNF signaling pathways. Animal experiments demonstrated that the uric acid, IL-1β, IL-6, and IL-8 serum levels were significantly lower in the BBR group compared with those in hyperuricemic rats. Conclusions: Using systematic network analysis, potential targets mediating the effects of BBR on GA were detected. The pathways and inflammatory factors involved were identified using in vivo experiments, thus providing a reference for further basic research and clinical applications of BBR in the treatment of GA.
Collapse
Affiliation(s)
- Shan Fang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yan Gao
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuan Fang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Sun
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Xie
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Wang X, Long H, Chen M, Zhou Z, Wu Q, Xu S, Li G, Lu Z. Modified Baihu decoction therapeutically remodels gut microbiota to inhibit acute gouty arthritis. Front Physiol 2022; 13:1023453. [PMID: 36589463 PMCID: PMC9798006 DOI: 10.3389/fphys.2022.1023453] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Acute gouty arthritis (AGA) is the most common first symptom of gout, and the development of gout as a metabolic and immune inflammatory disease is also correlated with the gut microbiota. However, the mechanism of the effect of changes in the gut microbiota on AGA remains unclear. The intestinal flora can not only affect purine metabolism or regulate inflammation, but also influence the therapeutic effect of drugs on AGA. The aim of this study was to investigate the exact mechanism of modified Baihu decoction (MBD) in the treatment of AGA and whether it is related to the regulation of the structure of the intestinal flora. Methods: On the 21st day of MBD administration by continuous gavage, a rat acute gouty arthritis model was constructed using sodium urate (0.1 mL/rat, 50 mg/mL), and the ankle joint swelling was measured before and 4 h, 8 h, 24 h, and 48 h after the injection of sodium urate. After 48 h of sodium urate injection, serum, liver, kidney, ankle synovial tissue and feces were collected from rats. The collected samples were examined and analyzed using H&E, Elisa, Immunohistochemistry, Histopathology, 16S rDNA, and Biochemical analysis. To investigate the mechanism of MBD to alleviate AGA using pro-inflammatory factors and intestinal flora. Results: MBD (5.84, 35 g/kg) was administered orally to AGA rats and diclofenac sodium tablets (DS-tablets) were used as standard treatment control. Serum biochemical assessment confirmed that MBD is a safe drug for the treatment of AGA. In addition, our findings confirmed that MBD relieved AGA-related symptoms, such as toe swelling. Lowering serum levels of uric acid, IL-1β, and TGF-β1 immunohistochemical results also confirmed that MBD reduced the expression of inflammatory elements such as IL-1β, NLRP3, ASC, and Caspase-1 in synovial tissue.Furthermore, compared with control group, the 16s rDNA sequencing of AGA rat faeces revealed an increase in the relative abundance of Lachnospiraceae, Muribaculaceae, and Bifidobacteriaceae species. While the relative abundance of Lactobacillaceae, Erysipelotrichaceae, Ruminococcaceae, Prevotellaceae and Enterobacteriaceae showed a relative decrease in species abundance. Of these, the reduction in species abundance of Enterobacteriaceae was associated with a reduction in amino acid metabolism and environmental perception. After MBD therapeutic intervention, the disturbance of the intestinal flora caused by AGA was restored. Conclusion: In summary, MBD is an effective agent for the treatment of AGA, with the potential mechanism being the regulation of intestinal flora to control inflammation. This would help to promote the therapeutic effect of MBD on AGA.
Collapse
Affiliation(s)
- Xianyang Wang
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haishan Long
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming Chen
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China
| | - Zongbo Zhou
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China
| | - Qinlin Wu
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China
| | - Shijie Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Shijie Xu, ; Geng Li, ; Zhifu Lu,
| | - Geng Li
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Shijie Xu, ; Geng Li, ; Zhifu Lu,
| | - Zhifu Lu
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China,*Correspondence: Shijie Xu, ; Geng Li, ; Zhifu Lu,
| |
Collapse
|
25
|
Tian J, Zhou D, Xiang L, Xie B, Wang B, Li Y, Liu X. Calycosin represses AIM2 inflammasome-mediated inflammation and pyroptosis to attenuate monosodium urate-induced gouty arthritis through NF-κB and p62-Keap1 pathways. Drug Dev Res 2022; 83:1654-1672. [PMID: 36069386 DOI: 10.1002/ddr.21985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 07/05/2022] [Accepted: 08/10/2022] [Indexed: 11/06/2022]
Abstract
Gouty arthritis is an inflammatory disease induced by monosodium urate (MSU), and is closely related to the activation of inflammasomes. Calycosin plays an anti-inflammatory role in arthritis. This study explored the mechanism of Calycosin in MSU-induced gouty arthritis. MSU-induced gouty arthritis mouse models with or without treatment of Calycosin were established, and physiological and pathological indicators were determined. Similarly, peripheral blood mononuclear cells (PBMCs) and THP-1 macrophages were used in vitro. Lactate dehydrogenase (LDH) was tested. The degree of centrifugal infiltration was detected by immunofluorescence. ELISA and quantitative reverse-transcription polymerase chain reaction were conducted to determine the levels of inflammatory factors. Immunohistochemistry, immunofluorescence, and flow cytometry were utilized to detect the content of caspase-1. Protein expressions of NF-κB-, p62-Keap1 pathway-, and pyroptosis-related factors were examined by western blot. In MSU-induced mouse models, calycosin increased mechanical hyperalgesia but decreased the swelling index of the mouse knee joint in a time-dependent manner. MSU treatment increased inflammatory cells and LysM-eGFP+ neutrophils recruitment in vivo, and promoted the LDH content in vitro, and meanwhile, calycosin reversed the aforementioned effects of MSU. In addition, calycosin repressed the release of inflammatory factors, promoted p62 level and diminished the levels of AIM2, caspase-1, ASC, IL-1β, Keap1, Cleaved GSDMD, and Cleaved caspase-1 and phosphorylation of p65 and IκBα in MSU-induced mouse or cell models. Furthermore, AIM2 silencing also inhibited MSU-induced inflammation and pyroptosis. Collectively, calycosin may inhibit AIM2 inflammasomes-mediated inflammation and pyroptosis through NF-κB and p62-Keap1 pathways, ultimately playing a protective role in gouty arthritis.
Collapse
Affiliation(s)
- Jing Tian
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Dapeng Zhou
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Liangbi Xiang
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Bing Xie
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Baichuan Wang
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Yang Li
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| | - Xinwei Liu
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
26
|
Gout and Diet: A Comprehensive Review of Mechanisms and Management. Nutrients 2022; 14:nu14173525. [PMID: 36079783 PMCID: PMC9459802 DOI: 10.3390/nu14173525] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Gout is well known as an inflammatory rheumatic disease presenting with arthritis and abnormal metabolism of uric acid. The recognition of diet-induced systemic metabolic pathways have provided new mechanistic insights and potential interventions on gout progression. However, the dietary recommendations for gouty patients generally focus on food categories, with few simultaneous considerations of nutritional factors and systemic metabolism. It is worthwhile to comprehensively review the mechanistic findings and potential interventions of diet-related nutrients against the development of gout, including purine metabolism, urate deposition, and gouty inflammation. Although piecemeal modifications of various nutrients often provide incomplete dietary recommendations, understanding the role of nutritional factors in gouty development can help patients choose their healthy diet based on personal preference and disease course. The combination of dietary management and medication may potentially achieve enhanced treatment effects, especially for severe patients. Therefore, the role of dietary and nutritional factors in the development of gout is systematically reviewed to propose dietary modification strategies for gout management by: (1) reducing nutritional risk factors against metabolic syndrome; (2) supplementing with beneficial nutrients to affect uric acid metabolism and gouty inflammation; and (3) considering nutritional modification combined with medication supplementation to decrease the frequency of gout flares.
Collapse
|
27
|
Leung N, Yip K, Pillinger MH, Toprover M. Lowering and Raising Serum Urate Levels: Off-Label Effects of Commonly Used Medications. Mayo Clin Proc 2022; 97:1345-1362. [PMID: 35787862 PMCID: PMC9575594 DOI: 10.1016/j.mayocp.2022.02.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 11/22/2022]
Abstract
Drug-induced hyperuricemia and gout present an increasingly prevalent problem in clinical practice. Herein, we review the urate-lowering or urate-raising effects of commonly used agents. We performed a PubMed search using the terms gout, urate, and medication, along with the specific agents/classes described herein. Reports were reviewed until 2022, and original studies were considered if they primarily or secondarily reported the effects of 1 or more drugs on serum urate level. Previous reviews were assessed for references to additional studies that described urate-altering effects of medications. Urate-changing drugs are summarized regarding their magnitude of effect, mechanism of action, and clinical significance. Potentially urate-lowering drugs include angiotensin II receptor blockers, calcium channel blockers, high-dose aspirin and salicylates, some nonsalicylate nonsteroidal anti-inflammatory drugs, angiotensin-converting enzyme inhibitors, sodium-glucose cotransporter 2 inhibitors, statins, and fenofibrate. Potentially urate-increasing drugs discussed include diuretics, β-blockers, insulin, pyrazinamide, ethambutol, calcineurin inhibitors, low-dose aspirin, testosterone, and lactate. In patients who have or are at risk for hyperuricemia or gout, an increased awareness of drugs that affect serum urate level may allow for prescribing that effectively treats the indicated problem while minimizing adverse effects on hyperuricemia and gout.
Collapse
Affiliation(s)
- Nicole Leung
- Divison of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Orthopedic Hospital, New York, NY.
| | - Kevin Yip
- Department of Rheumatology, Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Michael H Pillinger
- Rheumatology Section, New York Harbor Healthcare System, New York Campus, U.S. Department of Veterans Affairs
| | - Michael Toprover
- Rheumatology Section, New York Harbor Healthcare System, New York Campus, U.S. Department of Veterans Affairs
| |
Collapse
|
28
|
Tsai PH, Kuo CF, See LC, Li PR, Chen JS, Tseng WY. Stroke Risk in Patients with Gout: A Nationwide Retrospective Cohort Study in Taiwan. J Clin Med 2022; 11:jcm11133779. [PMID: 35807064 PMCID: PMC9267343 DOI: 10.3390/jcm11133779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 01/25/2023] Open
Abstract
Objectives: To estimate stroke risk in Taiwanese patients with gout. Methods: We enrolled patients from the Taiwan National Health Insurance Database, with gout diagnosed from 2000 to 2008, and followed them up until December 2018. This cohort was propensity score-matched according to birth year, sex, the date of diagnosis of gout, comorbidities, and co-medications with individuals without gout (controls) (n = 310,820 in each group). Stroke was defined as the primary diagnosis at discharge after the index date. To evaluate ischemic and hemorrhagic stroke risks, we calculated their incidence, hazard ratio (HR), and two-year moving average incidence rate. Results: The incidence (95% CI) and HR of ischemic stroke were lower in the gout group than in the control group in the first 3 years (incidence: 4.74 [4.60–4.88] vs. 5.17 [5.03–5.32] per 1000 person-years; HR: 0.92 [0.88–0.96]), then became significantly higher than in the control group after 3 years (incidence: 4.10 [4.04–4.16] vs. 3.81 [3.75–3.87] per 1000 person-years; HR: 1.08 [1.05–1.10]). Similarly, the incidence (95% CI) and HR of hemorrhagic stroke was lower in the gout group than in the control group in the first 3 years (incidence: 1.51 [1.43–1.59] vs. 1.70 [1.62–1.79] per 1000 person-years; HR: 0.88 [0.82–0.92]), then became significantly higher than in controls after 3 years (incidence: 1.43 [1.39–1.46] vs. 1.26 [1.22–1.29] per 1000 person-years; HR: 1.14 [1.10–1.18]). Conclusions: In Taiwan, patients with gout had higher risks of ischemic and hemorrhagic stroke after 3 years.
Collapse
Affiliation(s)
- Ping-Han Tsai
- Division of Rheumatology, Allergy and Immunology, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei City 236, Taiwan;
| | - Chang-Fu Kuo
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan City 333, Taiwan; (C.-F.K.); (L.-C.S.)
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
| | - Lai-Chu See
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan City 333, Taiwan; (C.-F.K.); (L.-C.S.)
- Department of Public Health, Chang Gung University, Taoyuan City 333, Taiwan;
- Biostatistics Core Laboratory, Molecular Medicine Research Center, Chang Gung University, Taoyuan City 333, Taiwan
| | - Pei-Ru Li
- Department of Public Health, Chang Gung University, Taoyuan City 333, Taiwan;
| | - Jung-Sheng Chen
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan City 333, Taiwan;
| | - Wen-Yi Tseng
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital-Keelung, No. 222, Mijin Road, Keelung City 204, Taiwan
- Correspondence: ; Tel.: +886-2-24313131 (ext. 6204)
| |
Collapse
|
29
|
Zhu KJ, Deng GS, Zhang LY, Yang YC, Xu Q, Zhang MY. Association of neutrophil-to-lymphocyte ratio with renal impairment among patients with acute gouty arthritis. Int Urol Nephrol 2022; 54:2995-3000. [PMID: 35612782 DOI: 10.1007/s11255-022-03239-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 05/04/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Gouty arthritis (GA) is an inflammatory disease, and renal impairment may occur to varying degrees with the progress of disease. The neutrophil-to-lymphocyte ratio (NLR) is an inflammatory marker. In this study, we aimed to explore the association between NLR and renal impairment in GA. MATERIALS AND METHODS The subjects comprised 499 patients with gouty arthritis (GA) (473 men, 26 women; age range, 39-61 years old) from our hospital. They were divided into a chronic kidney disease (CKD) group (n = 206) and non-CKD group (n = 293) according to the glomerular filtration rate. Blood samples were collected during the gout flares. The differences in NLR, general data, and laboratory indexes of patients with GA between the two groups were compared, such as serum uric acid (SUA), serum creatinine (SCREA), erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), high-density lipoprotein (HDL), and low-density lipoprotein (LDL). RESULTS NLR (3.38 vs. 2.38 (p < 0.001)) was higher in the CKD group, compared to the non-CKD group. Similarly, both SUA (527 vs. 507 (p < 0.05)) and SCREA (122 vs. 87 (p < 0.001)) were higher in the CKD group than in the non-CKD group. Multivariate logistic regression analysis showed that NLR (OR = 1.122, p < 0.05), age, hypertension, and SUA were risk factors for CKD in patients with GA, although HDL and HGB were protective factors. The receiver operating characteristic (ROC) curve analysis indicated that the area under the curve of NLR for predicting CKD in patients with GA was 0.646 (95% CI 0.597-0.694). CONCLUSION Our data showed that NLR might be an important potential factor for evaluating renal impairment in GA during flares.
Collapse
Affiliation(s)
- Kai-Jun Zhu
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Zhengzhou Second Hospital, Zhengzhou, 450000, China
| | - Guo-Shu Deng
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Li-Yu Zhang
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ye-Chun Yang
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qiang Xu
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Ming-Ying Zhang
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
30
|
Wu C, Li F, Zhang X, Xu W, Wang Y, Yao Y, Han Z, Xia D. (-)-Epicatechin Ameliorates Monosodium Urate-Induced Acute Gouty Arthritis Through Inhibiting NLRP3 Inflammasome and the NF-κB Signaling Pathway. Front Pharmacol 2022; 13:799552. [PMID: 35462936 PMCID: PMC9019746 DOI: 10.3389/fphar.2022.799552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Gouty arthritis is a common and complex inflammatory disease that will reduce the life quality of human beings (-)-Epicatechin (EC) is famous for antioxidant and anti-inflammatory activities. Thus, the aim of this study was to investigate the therapeutic effect of EC on gouty arthritis and its mechanisms. Methods and results: EC was added into a monosodium urate (MSU)-stimulated THP-1 cell that was induced by phorbol 12-myristate 13-acetate and lipopolysaccharide (LPS) in advance to establish a gout model in vitro. The efficiency of EC on acute gouty arthritis mice induced by MSU was further investigated. The results showed that EC concentration-dependently improved the cell viability of LPS and MSU stimulated THP-1 cells, and significantly alleviated MSU-induced ankle edema in mice in a dose-dependent manner. In addition, EC inhibited the infiltration of inflammatory cells and local cascular congestion in ankle joint tissue. Furthermore, the secretion of inflammatory cytokines (IL-1β, IL-18, IL-6, and TNF-α) activation of NLRP3 inflammasome and NF-κB signaling pathway were markedly suppressed by EC in vitro and in vivo. Conclusion: These results indicated that EC could effectively improve MSU-induced acute gouty arthritis via inhibiting NLRP3 inflammasome and the NF-κB signaling pathway in vitro and in vivo, which suggested that EC might be a promising active ingredient for the prevention and treatment of gouty arthritis.
Collapse
Affiliation(s)
- Chenxi Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fenfen Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoxi Zhang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenjing Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanjing Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ziwei Han
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
31
|
Duan L, Zhong J, Yang Y, Zhu X. Editorial: Advances in Pathogenesis and Therapies of Gout. Front Immunol 2022; 13:890204. [PMID: 35432327 PMCID: PMC9010866 DOI: 10.3389/fimmu.2022.890204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Lihua Duan
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- *Correspondence: Lihua Duan,
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Yang
- Department of Medicine, University of Florida, FL, Gainesville, United States
| | - Xiaoxia Zhu
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Zhou M, Hua L, Wang YF, Chen ST, Yang CM, Zhang M, Li X, Li B. Oral Huzhang granules for the treatment of acute gouty arthritis: protocol for a double-blind, randomized, controlled trial. Trials 2022; 23:248. [PMID: 35365187 PMCID: PMC8973546 DOI: 10.1186/s13063-022-06188-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/20/2022] [Indexed: 11/10/2022] Open
Abstract
Background Acute gouty arthritis (GA) is the main clinical manifestation and the most common initial symptom of gout. The treatment of acute GA involves the use of colchicine, non-steroidal anti-inflammatory drugs, and corticosteroids. Because of the side effects of these drugs, their clinical applications are limited. The use of traditional Chinese medicine for the treatment of acute GA has unique advantages. The aim of this trial is to clarify the treatment efficacy, safety, and recurrence control efficacy of Huzhang granules (HZG) in patients with GA showing dampness-heat syndrome. Methods/design This double-blind, randomized, controlled trial was planned to be conducted between July 1, 2020, and December 31, 2022. A sample size of 267 participants (89 per group) with GA will be randomly assigned to three treatment groups in the ratio of 1:1:1: HZG, etoricoxib, and placebo groups. The study duration is 13 days, including a 1-day screening period, 5-day intervention period, and 1-week follow-up period. The primary outcome is analgesic effectiveness, assessed as pain in the worst-affected joint, which will be measured using the visual analog scale. Secondary outcomes include the patient’s assessment of pain in the primary study joint, patient’s global assessment of response to therapy, investigator’s global assessment of response to therapy, investigator’s assessment of tenderness and swelling of the study joint, and TCM syndromes. Furthermore, the number, nature, and severity of adverse events will be recorded. Discussion This study will provide evidence regarding the clinical efficacy and safety of Chinese medicine treatment for acute gouty arthritis. This study will provide noteworthy findings. Trial registration ClinicalTrials.gov NCT04462666. Registered on July 05, 2020 (first version).
Collapse
Affiliation(s)
- Mi Zhou
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Liang Hua
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yi-Fei Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Si-Ting Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chun-Mei Yang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ming Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China. .,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China. .,Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW This review gives an overview of recently published articles on COVID-19 and gout. RECENT FINDINGS People with gout are likely to be at an increased risk of poor outcomes after COVID-19 infection due to comorbid cardiometabolic conditions. The effects of chronic hyperuricemia on trained immunity, and the hyperinflammatory state induced by gout itself may also play a role. Frequent courses of glucocorticoids for gout flares may be associated with adverse outcomes after COVID-19 infection and reduced immunogenicity to the COVID-19 vaccination. Similarities between the pathophysiology of gout flares and the dysregulated inflammatory response of severe COVID-19 have been identified. Medications used in the treatment of gout, including colchicine and interleukin-1 inhibitors, have shown promise in the treatment of COVID-19 in clinical trials. Overall, the COVID-19 pandemic has had a negative impact on gout care, with patients reporting more difficulty with disease control, accessing medications and healthcare, and poorer quality of life. SUMMARY The COVID-19 pandemic has created many challenges for people with gout. At present, there is a lack of guidance on the management of gout during the pandemic and paucity of research assessing outcomes of COVID-19 infection in people with gout.
Collapse
Affiliation(s)
- Vicky Tai
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Philip C Robinson
- University of Queensland School of Clinical Medicine, Faculty of Medicine
- Royal Brisbane & Women's Hospital, Metro North Hospital & Health Service, Herston Road, Herston, Queensland, Australia
| | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
McCormick N, Yokose C, Lu N, Joshi AD, Curhan GC, Choi HK. Impact of adiposity on risk of female gout among those genetically predisposed: sex-specific prospective cohort study findings over >32 years. Ann Rheum Dis 2021; 81:556-563. [PMID: 34857519 DOI: 10.1136/annrheumdis-2021-221635] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/09/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVES To evaluate the joint (combined) association of excess adiposity and genetic predisposition with the risk of incident female gout, and compare to their male counterparts; and determine the proportion attributable to body mass index (BMI) only, genetic risk score (GRS) only, and to their interaction. METHODS We prospectively investigated potential gene-BMI interactions in 18 244 women from the Nurses' Health Study and compared with 10 888 men from the Health Professionals Follow-Up Study. GRS for hyperuricaemia was derived from 114 common urate-associated single nucleotide polymorphisms. RESULTS Multivariable relative risk (RR) for female gout was 1.49 (95% CI 1.42 to 1.56) per 5 kg/m2 increment of BMI and 1.43 (1.35 to 1.52) per SD increment in the GRS. For their joint association of BMI and GRS, RR was 2.18 (2.03 to 2.36), more than the sum of each individual factor, indicating significant interaction on an additive scale (p for interaction <0.001). The attributable proportions of joint effect for female gout were 42% (37% to 46%) to adiposity, 37% (32% to 42%) to genetic predisposition and 22% (16% to 28%) to their interaction. Additive interaction among men was smaller although still significant (p interaction 0.002, p for heterogeneity 0.04 between women and men), and attributable proportion of joint effect was 14% (6% to 22%). CONCLUSIONS While excess adiposity and genetic predisposition both are strongly associated with a higher risk of gout, the excess risk of both combined was higher than the sum of each, particularly among women.
Collapse
Affiliation(s)
- Natalie McCormick
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA .,Arthritis Research Canada, Vancouver, British Columbia, Canada.,Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Chio Yokose
- Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Rheumatology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Na Lu
- Arthritis Research Canada, Vancouver, British Columbia, Canada.,Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Amit D Joshi
- Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Clinical Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gary C Curhan
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Hyon K Choi
- Arthritis Research Canada, Vancouver, British Columbia, Canada .,Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Nada D, Gaber R, Mahmoud AS, Elkhouly R, Alashkar D. Hyperuricemia Among Egyptian Rheumatoid Arthritis Patients. Is It an Association or an Inflammatory Marker? A Cross-Sectional Observational Study. Open Access Rheumatol 2021; 13:305-314. [PMID: 34675698 PMCID: PMC8502065 DOI: 10.2147/oarrr.s331488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To detect the prevalence of hyperuricemia in Egyptian rheumatoid arthritis (RA) patients as well as to assess its association with the severity of joint inflammation and disease-modifying antirheumatic drugs (DMARDs) in those patients. METHODS A total of 150 RA patients were recruited; all patients were subjected to (1) clinical and functional assessment by disease activity score in 28 joints (DAS28) and modified health assessment questionnaire (MHAQ). (2) Laboratory investigations: serum uric acid (SUA) level, complete blood count (CBC), erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), rheumatoid factor (RF), anti-cyclic citrullinated peptide (anti-CCP), tumor necrosis factor α (TNF-α), interleukin 1 (IL1), and interleukin 6 (IL6) levels. (3) Radiological evaluation: (A) plain X-ray of both hands and feet; (B) musculoskeletal ultrasound (MSUS) of both wrists, hands, shoulder, ankle, and knee joints. RESULTS SUA was significantly correlated with disease activity by DAS28. Acute-phase reactants and inflammatory markers (IL1β, IL6, and TNF-α) were also significantly elevated in RA patients with low and high hyperuricemia compared to those with normal SUA. A total of 90% of RA patients with low hyperuricemia had synovial proliferation with power Doppler (1+ and 2+), and 30 patients had mild effusion (1+), while nearly all patients with high hyperuricemia had hypoechoic synovial proliferation (2+ and 3+), and 20 patients had moderate effusion. However, 70% RA patients with normal serum uric acid showed mild synovitis and effusion (1+). No significant association was found between the administered DMARDs and levels of SUA as well as inflammatory markers; however, high-dose steroid treatment was associated with high SUA level. CONCLUSION Elevation of serum uric acid levels in Egyptian RA patients was prevalent and might be an inflammatory marker for severity of joint inflammation. Moreover, higher doses of steroids could be considered a cause of hyperuricemia.
Collapse
Affiliation(s)
- Doaa Nada
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Tanta University, Tanta, Al Gharbeia Governorate, Egypt
| | - Rasha Gaber
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Al Gharbeia Governorate, Egypt
| | - Al Shymaa Mahmoud
- Radiology Departments, Faculty of Medicine, Tanta University, Tanta, Al Gharbeia Governorate, Egypt
| | - Radwa Elkhouly
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Tanta University, Tanta, Al Gharbeia Governorate, Egypt
| | - Doaa Alashkar
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Tanta University, Tanta, Al Gharbeia Governorate, Egypt
| |
Collapse
|
36
|
Georgel PT, Georgel P. Where Epigenetics Meets Food Intake: Their Interaction in the Development/Severity of Gout and Therapeutic Perspectives. Front Immunol 2021; 12:752359. [PMID: 34603340 PMCID: PMC8484966 DOI: 10.3389/fimmu.2021.752359] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/31/2021] [Indexed: 01/02/2023] Open
Abstract
Gout is the most frequent form of inflammatory arthritis in the world. Its prevalence is particularly elevated in specific geographical areas such as in the Oceania/Pacific region and is rising in the US, Europe, and Asia. Gout is a severe and painful disease, in which co-morbidities are responsible for a significant reduction in life expectancy. However, gout patients remain ostracized because the disease is still considered "self-inflicted", as a result of unhealthy lifestyle and excessive food and alcohol intake. While the etiology of gout flares is clearly associated with the presence of monosodium urate (MSU) crystal deposits, several major questions remain unanswered, such as the relationships between diet, hyperuricemia and gout flares or the mechanisms by which urate induces inflammation. Recent advances have identified gene variants associated with gout incidence. Nevertheless, genetic origins of gout combined to diet-related possible uric acid overproduction account for the symptoms in only a minor portion of patients. Hence, additional factors must be at play. Here, we review the impact of epigenetic mechanisms in which nutrients (such as ω-3 polyunsaturated fatty acids) and/or dietary-derived metabolites (like urate) trigger anti/pro-inflammatory responses that may participate in gout pathogenesis and severity. We propose that simple dietary regimens may be beneficial to complement therapeutic management or contribute to the prevention of flares in gout patients.
Collapse
Affiliation(s)
- Philippe T Georgel
- Department of Biological Sciences, Cell Differentiation and Development Center, Joan C. Edwards School of Medicine, Byrd Biotechnology Science Center, Marshall University, Huntington, WV, United States
| | - Philippe Georgel
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Unité de Recherche et d'Expertise Immunity and Inflammation, Institut Pasteur in New Caledonia, Pasteur Network, Nouméa, New Caledonia
| |
Collapse
|
37
|
Deng SH, Dang WT, Liu J, Bai Y, You LL, Hu J, Luo H. Differential Diagnosis of Acute and Chronic Gouty Arthritis by Multijoint Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:2853-2859. [PMID: 34325959 DOI: 10.1016/j.ultrasmedbio.2021.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 04/26/2021] [Accepted: 05/06/2021] [Indexed: 06/13/2023]
Abstract
To investigate whether multi-joint ultrasound (US) findings in patients with gouty arthritis could be used to distinguish between acute and chronic stages, we performed a retrospective study with 129 enrolled patients from the Rheumatology Department of the First Affiliated Hospital of Chengdu Medical College from September 1, 2018 to June 12, 2019. Patients with acute or non-acute gout were categorized using clinical data, and US imaging findings of the knees, ankles and first metatarsophalangeal joints were analyzed and compared between groups. Notably, we found that the most prevalent sign detected by US was the hyperechoic spot in the synovium, followed by arthrosynovitis, aggregates, double contour signs and tophi; meanwhile, bone erosions were the least common. Additionally, synovitis was more frequently detected in the acute joints of gouty arthritis (49%) compared with the non-acute joints (35%), whereas grade 1 or 2 blood flow classifications (97%), tophi and bone lesions were more often seen in the latter. Overall, our data suggest that multi-joint US scanning might be used to evaluate disease severity and discriminate between stages of gouty arthritis.
Collapse
Affiliation(s)
- Si-Hui Deng
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wan-Tai Dang
- Department of Rheumatism and Immunity, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jian Liu
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Yang Bai
- Department of Rheumatism and Immunity, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Lan-Lan You
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jin Hu
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Hui Luo
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
38
|
Gromova MA, Tsurko VV, Melekhina AS. GOUT AND THE CENTRAL ASIAN DIET. CENTRAL ASIAN JOURNAL OF MEDICAL HYPOTHESES AND ETHICS 2021. [DOI: 10.47316/cajmhe.2021.2.2.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Patients with gout are advised to follow a certain diet. The search for healthy eating habits continues. This overview aims to explore the impact of the Central Asian diet on the development and course of hyperuricemia and gout and to propose related preventive measures. We processed English articles reflecting on the impact of the Central Asian diet on the development of gout and the frequency of gout attacks. Central Asian dishes such as lagman, pilaf, and manty are distinguished for high nutritional value and abundance of saturated fats, red meat, and trans fats. Moderate consumption of Central Asian traditional dishes is advisable to prevent weight gain and gout attacks. Patients with gout are additionally encouraged to consume low-calorie and non-fat products and regularly intake fruits and vegetables during meals. Patients with gout should be provided with information about a healthy diet and lifestyle. More evidence-based studies are warranted to provide better quality guidance on gout prevention and treatment.
Collapse
|
39
|
Henson MA. Interrogation of the perturbed gut microbiota in gouty arthritis patients through in silico metabolic modeling. Eng Life Sci 2021; 21:489-501. [PMID: 34257630 PMCID: PMC8257998 DOI: 10.1002/elsc.202100003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/26/2021] [Accepted: 05/01/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies have shown perturbed gut microbiota associated with gouty arthritis, a metabolic disease characterized by an imbalance between uric acid production and excretion. To mechanistically investigate altered microbiota metabolism associated with gout disease, 16S rRNA gene amplicon sequence data from stool samples of gout patients and healthy controls were computationally analyzed through bacterial community metabolic models. Patient-specific community models constructed with the metagenomics modeling pipeline, mgPipe, were used to perform k-means clustering of samples according to their metabolic capabilities. The clustering analysis generated statistically significant partitioning of samples into a Bacteroides-dominated, high gout cluster and a Faecalibacterium-elevated, low gout cluster. The high gout cluster was predicted to allow elevated synthesis of the amino acids D-alanine and L-alanine and byproducts of branched-chain amino acid catabolism, while the low gout cluster allowed higher production of butyrate, the sulfur-containing amino acids L-cysteine and L-methionine, and the L-cysteine catabolic product H2S. By expanding the capabilities of mgPipe to provide taxa-level resolution of metabolite exchange rates, acetate, D-lactate and succinate exchanged from Bacteroides to Faecalibacterium were predicted to enhance butyrate production in the low gout cluster. Model predictions suggested that sulfur-containing amino acid metabolism generally and H2S more specifically could be novel gout disease markers.
Collapse
Affiliation(s)
- Michael A. Henson
- Department of Chemical Engineering and the Institute for Applied Life SciencesUniversity of MassachusettsAmherstMAUSA
| |
Collapse
|
40
|
Abstract
Urate is the end-product of the purine metabolism in humans. The dominant source of urate is endogenous purines and the remainder comes through diet. Approximately two thirds of urate is eliminated via the kidney with the rest excreted in the feces. While the transporter BCRP, encoded by ABCG2, has been found to play a role in both the gut and kidney, SLC22A12 and SLC2A9 encoding URAT1 and GLUT9, respectively, are the two transporters best characterized. Only 8-12% of the filtered urate is excreted by the kidney. Renal elimination of urate depends substantially on specific transporters, including URAT1, GLUT9 and BCRP. Studies that have assessed the biologic effects of urate have produced highly variable results. Although there is a suggestion that urate may have anti-oxidant properties in some circumstances, the majority of evidence indicates that urate is pro-inflammatory. Hyperuricemia can result in the formation of monosodium urate (MSU) crystals that may be recognized as danger signals by the immune system. This immune response results in the activation of the NLRP3 inflammasome and ultimately in the production and release of interleukin-1β, and IL-18, that mediate both inflammation, pyroptotic cell death, and necroinflammation. It has also been demonstrated that soluble urate mediates effects on the kidney to induce hypertension and can induce long term epigenetic reprogramming in myeloid cells to induce "trained immunity." Together, these sequelae of urate are thought to mediate most of the physiological effects of hyperuricemia and gout, illustrating this biologically active molecule is more than just an "end-product" of purine metabolism.
Collapse
Affiliation(s)
- Robert T Keenan
- Division of Rheumatology, Duke University School of Medicine, Durham 27710, NC, USA.
| |
Collapse
|
41
|
Pisaniello HL, Fisher MC, Farquhar H, Vargas-Santos AB, Hill CL, Stamp LK, Gaffo AL. Efficacy and safety of gout flare prophylaxis and therapy use in people with chronic kidney disease: a Gout, Hyperuricemia and Crystal-Associated Disease Network (G-CAN)-initiated literature review. Arthritis Res Ther 2021; 23:130. [PMID: 33910619 PMCID: PMC8080370 DOI: 10.1186/s13075-021-02416-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/04/2021] [Indexed: 12/27/2022] Open
Abstract
Gout flare prophylaxis and therapy use in people with underlying chronic kidney disease (CKD) is challenging, given limited treatment options and risk of worsening renal function with inappropriate treatment dosing. This literature review aimed to describe the current literature on the efficacy and safety of gout flare prophylaxis and therapy use in people with CKD stages 3-5. A literature search via PubMed, the Cochrane Library, and EMBASE was performed from 1 January 1959 to 31 January 2018. Inclusion criteria were studies with people with gout and renal impairment (i.e. estimated glomerular filtration rate (eGFR) or creatinine clearance (CrCl) < 60 ml/min/1.73 m2), and with exposure to colchicine, interleukin-1 inhibitors, non-steroidal anti-inflammatory drugs (NSAIDs), and glucocorticoids. All study designs were included. A total of 33 studies with efficacy and/or safety analysis stratified by renal function were reviewed-colchicine (n = 20), anakinra (n = 7), canakinumab (n = 1), NSAIDs (n = 3), and glucocorticoids (n = 2). A total of 58 studies reported these primary outcomes without renal function stratification-colchicine (n = 29), anakinra (n = 10), canakinumab (n = 6), rilonacept (n = 2), NSAIDs (n = 1), and glucocorticoids (n = 10). Most clinical trials excluded study participants with severe CKD (i.e. eGFR or CrCl of < 30 mL/min/1.73 m2). Information on the efficacy and safety outcomes of gout flare prophylaxis and therapy use stratified by renal function is lacking. Clinical trial results cannot be extrapolated for those with advanced CKD. Where possible, current and future gout flare studies should include patients with CKD and with study outcomes reported based on renal function and using standardised gout flare definition.
Collapse
Affiliation(s)
- Huai Leng Pisaniello
- Discipline of Medicine, Faculty of Health and Medical Sciences, the University of Adelaide, Adelaide, South Australia, Australia
| | - Mark C Fisher
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA.,Prima CARE, Fall River, MA, USA
| | - Hamish Farquhar
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | | | - Catherine L Hill
- Discipline of Medicine, Faculty of Health and Medical Sciences, the University of Adelaide, Adelaide, South Australia, Australia.,Rheumatology Unit, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Angelo L Gaffo
- Division of Rheumatology and Clinical Immunology, University of Alabama, 1720 2nd Avenue South, Birmingham, AL, 35294, USA. .,Birmingham VA Medical Center, Birmingham, USA.
| |
Collapse
|
42
|
Integrated Molecular Docking with Network Pharmacology to Reveal the Molecular Mechanism of Simiao Powder in the Treatment of Acute Gouty Arthritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5570968. [PMID: 34007291 PMCID: PMC8100412 DOI: 10.1155/2021/5570968] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/12/2021] [Accepted: 04/16/2021] [Indexed: 12/26/2022]
Abstract
Background The incidence of gout has been rapidly increasing in recent years with the changing of diet. At present, modern medications used in the clinical treatment of gout showed several side effects, such as gastrointestinal damage and the increased risk of cardiovascular disease. The traditional Chinese prescription Simiao Powder (SMP) has a long history in the treatment of acute gouty arthritis (AGA) and has a good curative effect. However, the mechanism and target of its therapeutic effects are still not completely understood. Methods Potential active compounds (PACs) and targets of SMP were found in the TCMSP database, and the disease target genes related to AGA were obtained by searching CTD, DisGeNET, DrugBank, GeneCards, TTD, OMIM, and PharmGKB disease databases with “acute gouty arthritis” and “Arthritis, Gouty” as keywords, respectively. The network of “Traditional Chinese medicine (TCM)-PACs-potential targets of acute gouty arthritis” was constructed with the Cytoscape 3.7.2 software, and the target genes of acute gouty arthritis were intersected with genes regulated by active compounds of SMP. The resultant common gene targets were input into Cytoscape 3.7.2 software, and the BisoGenet plug-in was used to construct a PPI network. The GO functional enrichment analysis and KEGG pathway enrichment analysis of the intersecting target proteins were performed using R software and corresponding program packages. The molecular docking verification was carried out between the potentially active compounds of SMP and the core target at the same time. Results 40 active components and 203 targets were identified, of which 95 targets were common targets for the drugs and diseases. GO function enrichment analysis revealed that SMP regulated several biological processes, such as response to lipopolysaccharide and oxidative stress, RNA polymerase II transcription regulator complex, protein kinase complex, and other cellular and molecular processes, including DNA-binding transcription factor binding. Results of KEGG pathway analysis showed that SMP was associated with AGA-related pathways such as interleukin-17 (IL-17), tumor necrosis factor (TNF), p53, and hypoxia-inducible factor 1 (HIF-1) signaling pathways. The results of molecular docking showed that active compounds in SMP exhibited strong binding to five core protein receptors (TP53, FN1, ESR1, CDK2, and HSPA5). Conclusions Active components of SMP, such as quercetin, kaempferol, wogonin, baicalein, beta-sitosterol, and rutaecarpine, showed therapeutic effects on AGA. These compounds were strongly associated with core target proteins (such as TP53, FN1, ESR1, CDK2, and HSPA5). This study reveals that IL-17, TNF, p53, and HIF-1 signaling pathways mediate the therapeutic effects of SMP on AGA. These findings expand our understanding of the mechanism of SMP in the treatment of AGA.
Collapse
|
43
|
Autoinflammatory Features in Gouty Arthritis. J Clin Med 2021; 10:jcm10091880. [PMID: 33926105 PMCID: PMC8123608 DOI: 10.3390/jcm10091880] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022] Open
Abstract
In the panorama of inflammatory arthritis, gout is the most common and studied disease. It is known that hyperuricemia and monosodium urate (MSU) crystal-induced inflammation provoke crystal deposits in joints. However, since hyperuricemia alone is not sufficient to develop gout, molecular-genetic contributions are necessary to better clinically frame the disease. Herein, we review the autoinflammatory features of gout, from clinical challenges and differential diagnosis, to the autoinflammatory mechanisms, providing also emerging therapeutic options available for targeting the main inflammatory pathways involved in gout pathogenesis. This has important implication as treating the autoinflammatory aspects and not only the dysmetabolic side of gout may provide an effective and safer alternative for patients even in the prevention of possible gouty attacks.
Collapse
|
44
|
Topless RK, Phipps-Green A, Leask M, Dalbeth N, Stamp LK, Robinson PC, Merriman TR. Gout, Rheumatoid Arthritis, and the Risk of Death Related to Coronavirus Disease 2019: An Analysis of the UK Biobank. ACR Open Rheumatol 2021; 3:333-340. [PMID: 33856739 PMCID: PMC8126763 DOI: 10.1002/acr2.11252] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/26/2021] [Indexed: 12/25/2022] Open
Abstract
Objectives The objectives for this study were to assess whether gout and/or rheumatoid arthritis (RA) are risk factors for coronavirus disease 2019 (COVID‐19) diagnosis and to assess whether gout and/or RA are risk factors for death from COVID‐19. Methods We used data from the UK Biobank. Multivariable‐adjusted logistic regression was employed in the following analyses: analysis A, to test for association between gout and/or RA and COVID‐19 diagnosis (n = 473,139); analysis B, to test for association between gout and/or RA and death from COVID‐19 in a case‐control cohort of people who died of or survived COVID‐19 (n = 2059); analysis C, to test for association between gout and/or RA and death from COVID‐19 in the entire UK Biobank cohort (n = 473,139). Results RA, but not gout, was associated with COVID‐19 diagnosis in analysis A. Neither RA nor gout was associated with risk of death in the group diagnosed with COVID‐19 in analysis B. However, RA was associated with risk of death related to COVID‐19 by using the UK Biobank cohort in analysis C, independent of comorbidities and other measured risk factors (odds ratio [OR] 1.9; 95% confidence interval CI 1.2–3.0). Gout was not associated with death related to COVID‐19 in the same UK Biobank analysis (OR 1.2; 95% CI 0.8–1.7). Conclusion RA is a risk factor for death from COVID‐19 by using the UK Biobank cohort. These findings require replication in larger data sets that also allow for inclusion of a wider range of factors.
Collapse
Affiliation(s)
| | | | - Megan Leask
- University of Otago, Dunedin, New Zealand, and University of Alabama at Birmingham
| | | | - Lisa K Stamp
- University of Otago, Christchurch, Christchurch, New Zealand
| | | | - Tony R Merriman
- University of Otago, Dunedin, New Zealand, and University of Alabama at Birmingham
| |
Collapse
|
45
|
Zisheng Shenqi Decoction Ameliorates Monosodium Urate-Mediated Gouty Arthritis in Rats via Promotion of Autophagy through the AMPK/mTOR Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6918026. [PMID: 33505502 PMCID: PMC7806400 DOI: 10.1155/2021/6918026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/11/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022]
Abstract
Gouty arthritis (GA) is an inflammatory disease owing to the accumulation of monosodium urate (MSU) in joints, leading to redness and burning pain. In this study, the effect of Zisheng Shenqi Decoction (ZSD) on a rat model of MSU-induced GA was investigated. ZSD obviously diminished the right paw thickness, the degree of the swelling of the paw, and the infiltration of the inflammatory cell, as well as cartilage erosion, and widened the joint space in MSU-treated rats. Besides, MSU remarkably elevated the release of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, and IL-18; however, ZSD treatment dose dependently lowered these levels and resulted in a significant decrease in articular elastase activity. Also, ZSD administration increased the activities of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) but declined malondialdehyde (MDA) and nitrogen monoxide (NO) contents. Importantly, western blotting analysis revealed that NOD-like receptor protein 3 (NLRP3), cleaved caspase-1, IL-1β, nuclear factor-E2-related factor 2 (Nrf2) in the cytoplasm, phosphorylated mammalian target of rapamyclin (p-mTOR), and p62 expressions were downregulated, whereas the levels of nuclear Nrf2, phosphorylated AMP-activated protein kinase (p-AMPK), Beclin-1, and LC3II/I were upregulated by ZSD. Immunofluorescence assay indicated that ZSD evidently promoted nuclear translocation of LC3. Taken together, ZSD inhibited inflammation and oxidative stress and facilitated autophagy through the activation of the AMPK pathway and suppression of the mTOR signaling pathway, demonstrating its potential for preventing and curing GA.
Collapse
|
46
|
Sun J, Fan Y, Yang Z, Jin R, Xin P, Cai X, Li X, Wang S. Efficacy and safety of fire-needle in the treatment of gouty arthritis: A protocol for systematic review and meta analysis. Medicine (Baltimore) 2020; 99:e21259. [PMID: 32791703 PMCID: PMC7386971 DOI: 10.1097/md.0000000000021259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Fire needle therapy is an ancient external treatment method of traditional Chinese medicine. This therapy is simple to operate and has fewer side effects. Gouty arthritis (GA) is common disease that is often characterized by high excruciating pain on joint. Evidence from clinical studies show that fire needle exerts therapeutic effects on gout arthritis, but no evidence-based medicine is available. This study aimed to evaluate the efficacy and safety of fire acupuncture in the treatment of gout arthritis. METHODS Randomized controlled trials of fire needle in the treatment of GA published until May 2020 will be searched in the English databases (PubMed, EMBASE, Web of Science, the Cochrane Library) and Chinese databases (China National Knowledge Infrastructure, the Chongqing VIP Chinese Science and Technology Periodical Database, Wanfang database, and China Biomedical Literature Database). Additional search will be performed on Google academy and Baidu Academy. Data will be extracted from the studies by 2 reviewers working independently. Subsequently, quality assessment and a meta-analysis will be carried out for the studies using RevMan 5.3. RESULTS The efficacy and safety of fire needle in the treatment of GA will be evaluated based on overall effective rate, visual analog scale, blood uric acid, C-reactive protein, joint swelling and pain score, adverse reaction rate, and other clinical outcomes. CONCLUSIONS The proposed systematic review and meta-analysis are expected to provide reliable evidence for the clinical benefits of fire-needle therapy in GA.
Collapse
Affiliation(s)
- Jiya Sun
- Tianjin University of Traditional Chinese Medicine
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion
| | - Yihua Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Rui Jin
- Tianjin University of Traditional Chinese Medicine
| | - Ping Xin
- Tianjin University of Traditional Chinese Medicine
| | - Xuemeng Cai
- Tianjin University of Traditional Chinese Medicine
| | - Xinju Li
- Tianjin University of Traditional Chinese Medicine
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion
| | - Shenjun Wang
- Tianjin University of Traditional Chinese Medicine
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion
| |
Collapse
|
47
|
Ye SM, Zhou MZ, Jiang WJ, Liu CX, Zhou ZW, Sun MJ, Hu QH. Silencing of Gasdermin D by siRNA-Loaded PEI-Chol Lipopolymers Potently Relieves Acute Gouty Arthritis through Inhibiting Pyroptosis. Mol Pharm 2020; 18:667-678. [PMID: 32579365 DOI: 10.1021/acs.molpharmaceut.0c00229] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gasdermin D (GSDMD) plays a causal role in NOD-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis eruption, which has been regarded as a potential therapeutic target for pyroptosis-related diseases including acute gouty arthritis. In the present study, the synthesized PEI-Chol (cholesterol grafted polyethylenimine) was assembled with GSDMD small interfering RNA (siRNA) to form PEI-Chol/siGSDMD polyplexes, which provided high transfection efficiency for siRNA-mediated GSDMD knockdown. Then we evaluated the effect of GSDMD siRNA-loaded PEI-Chol on inflammatory cascades in bone-marrow-derived macrophages (BMDMs) and acute gouty arthritis animal models under MSU exposure. When accompanied by pyroptosis blockade and decreased release of interleukin-1 beta (IL-1β), NLRP3 inflammasome activation was also suppressed by GSDMD knockdown in vivo and in vitro. Moreover, in MSU-induced acute gouty arthritis mice, blocking GSDMD with siRNA significantly improved ankle swelling and inflammatory infiltration observed in histopathological analysis. Furthermore, investigation using a mouse air pouch model verified the effect of siGSDMD-loaded PEI-Chol on pyroptosis of recruited macrophages and related signaling pathways in response to MSU. These novel findings exhibited that GSDMD knockdown relieved acute gouty arthritis through inhibiting pyroptosis, providing a possible therapeutic approach for MSU-induced acute gouty arthritis molecular therapy using PEI-Chol as a nucleic acid delivery carrier.
Collapse
Affiliation(s)
- Shu-Min Ye
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China.,School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Meng-Ze Zhou
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wen-Jiao Jiang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Chun-Xiao Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China.,School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Zhan-Wei Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Min-Jie Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Qing-Hua Hu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China.,School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| |
Collapse
|
48
|
Chiou A, England BR, Sayles H, Thiele GM, Duryee MJ, Baker JF, Singh N, Cannon GW, Kerr GS, Reimold A, Gaffo A, Mikuls TR. Coexistent Hyperuricemia and Gout in Rheumatoid Arthritis: Associations With Comorbidities, Disease Activity, and Mortality. Arthritis Care Res (Hoboken) 2020; 72:950-958. [PMID: 31074584 DOI: 10.1002/acr.23926] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/07/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Although hyperuricemia and gout can complicate the course of rheumatoid arthritis (RA), the impact of these factors on outcomes in RA is unclear. We undertook this study to examine associations of coexistent hyperuricemia and gout with RA disease measures, RA treatments, and survival. METHODS Participants from a longitudinal RA study were categorized by the presence of gout and serum urate (UA) status. Groups were compared by baseline patient characteristics, RA disease activity, treatments, and comorbidities. Associations of baseline serum UA levels with all-cause and cardiovascular disease (CVD)-related mortality were examined in multivariable survival analyses. RESULTS Of 1,999 participants with RA, 341 (17%) had serum UA concentrations of >6.8 mg/dl, and 121 (6.1%) were diagnosed with gout. There were no significant associations of serum UA concentration or gout with RA disease activity or treatment at enrollment, with the exception that those with gout were more likely to be receiving sulfasalazine and less likely to be receiving nonsteroidal antiinflammatory drugs. After adjustments for age and sex, moderate hyperuricemia (serum UA >6.8 to ≤8 mg/dl) was associated with an increased risk of CVD-related mortality (hazard ratio 1.56 [95% confidence interval 1.11-2.21]). This association was attenuated and not significant following additional adjustment for comorbidities that more commonly accompany hyperuricemia. Results corresponding with serum UA concentrations of >8.0 mg/dl were similar, although not reaching statistical significance in any model. There were no associations of baseline serum UA concentration with all-cause mortality. CONCLUSION Our study reports the frequency of hyperuricemia and gout in patients with RA. These results demonstrate strong associations of hyperuricemia with CVD mortality in this population, a risk that appears to be driven by excess comorbidity.
Collapse
Affiliation(s)
| | - Bryant R England
- University of Nebraska Medical Center and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | | | - Geoffrey M Thiele
- University of Nebraska Medical Center and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Michael J Duryee
- University of Nebraska Medical Center and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Joshua F Baker
- Corporal Michael J. Crescenz VAMC and University of Pennsylvania, Philadelphia
| | | | - Grant W Cannon
- Salt Lake City VAMC and University of Utah, Salt Lake City
| | - Gail S Kerr
- Washington, DC VAMC, Georgetown University, and Howard University, Washington, DC
| | | | - Angelo Gaffo
- Birmingham VAMC and University of Alabama at Birmingham
| | - Ted R Mikuls
- University of Nebraska Medical Center and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| |
Collapse
|
49
|
Bousoik E, Qadri M, Elsaid KA. CD44 Receptor Mediates Urate Crystal Phagocytosis by Macrophages and Regulates Inflammation in A Murine Peritoneal Model of Acute Gout. Sci Rep 2020; 10:5748. [PMID: 32238827 PMCID: PMC7113258 DOI: 10.1038/s41598-020-62727-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/16/2020] [Indexed: 02/04/2023] Open
Abstract
Gout is a chronic arthritis caused by the deposition of poorly soluble monosodium urate monohydrate (MSU) crystals in peripheral joints. Resident macrophages initiate inflammation in response to MSU mediated by NF-κB nuclear translocation and NLRP3 inflammasome activation. We investigated the role of CD44, a transmembrane receptor, in mediating MSU phagocytosis by macrophages. We used an antibody that sheds the extracellular domain (ECD) of CD44 to study the role of the receptor and its associated protein phosphatase 2A (PP2A) in macrophage activation. We also studied the significance of CD44 in mediating MSU inflammation in-vivo. Cd44−/− BMDMs showed reduced MSU phagocytosis, LDH release, IL-1β expression and production compared to Cd44+/+ BMDMs. Elevated CD44 staining was detected intracellularly and CD44 colocalized with α-tubulin as a result of MSU exposure and ECD-shedding reduced MSU phagocytosis in murine and human macrophages. Anti-CD44 antibody treatment reduced NF-κB p65 subunit nuclear levels, IL-1β expression, pro-IL-1β and IL-8 production in MSU stimulated THP-1 macrophages (p < 0.01). The effect of the antibody was mediated by an enhancement in PP2A activity. CD44 ECD-shedding reduced the conversion of procaspase-1 to active caspase-1, caspase-1 activity and resultant generation of mature IL-1β in macrophages. Neutrophil and monocyte influx and upregulated production of IL-1β was evident in wildtype mice. MSU failed to trigger neutrophil and monocyte recruitment in Cd44−/− mice and lower IL-1β levels were detected in peritoneal lavages from Cd44−/− mice (p < 0.01). Anti-CD44 antibody treatment reduced neutrophil and monocyte recruitment and resulted in reduced lavage IL-1β levels in the same model. CD44 plays a biologically significant role in mediating phagocytosis of MSU and downstream inflammation and is a novel target in gout treatment.
Collapse
Affiliation(s)
- Emira Bousoik
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.,School of Pharmacy, Omar-Al-Mukhtar University, Derna, Libya
| | - Marwa Qadri
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.,Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, 82826, Saudi Arabia
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.
| |
Collapse
|
50
|
Wang Z, Zhao Y, Phipps-Green A, Liu-Bryan R, Ceponis A, Boyle DL, Wang J, Merriman TR, Wang W, Terkeltaub R. Differential DNA Methylation of Networked Signaling, Transcriptional, Innate and Adaptive Immunity, and Osteoclastogenesis Genes and Pathways in Gout. Arthritis Rheumatol 2020; 72:802-814. [PMID: 31738005 DOI: 10.1002/art.41173] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In gout, autoinflammatory responses to urate crystals promote acute arthritis flares, but the pathogeneses of tophi, chronic synovitis, and erosion are less well understood. Defining the pathways of epigenomic immunity training can reveal novel pathogenetic factors and biomarkers. The present study was undertaken to seminally probe differential DNA methylation patterns utilizing epigenome-wide analyses in patients with gout. METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from a San Diego cohort of patients with gout (n = 16) and individually matched healthy controls (n = 14). PBMC methylome data were processed with ChAMP package in R. ENCODE data and Taiji data analysis software were used to analyze transcription factor (TF)-gene networks. As an independent validation cohort, whole blood DNA samples from New Zealand Māori subjects (n = 13 patients with gout, n = 16 control subjects without gout) were analyzed. RESULTS Differentially methylated loci clearly separated gout patients from controls, as determined by hierarchical clustering and principal components analyses. IL23R, which mediates granuloma formation and cell invasion, was identified as one of the multiple differentially methylated gout risk genes. Epigenome-wide analyses revealed differential methylome pathway enrichment for B and T cell receptor signaling, Th17 cell differentiation and interleukin-17 signaling, convergent longevity regulation, circadian entrainment, and AMP-activated protein kinase signaling, which are pathways that impact inflammation via insulin-like growth factor 1 receptor, phosphatidylinositol 3-kinase/Akt, NF-κB, mechanistic target of rapamycin signaling, and autophagy. The gout cohorts overlapped for 37 (52.9%) of the 70 TFs with hypomethylated sequence enrichment and for 30 (78.9%) of the 38 enriched KEGG pathways identified via TFs. Evidence of shared differentially methylated gout TF-gene networks, including the NF-κB activation-limiting TFs MEF2C and NFATC2, pointed to osteoclast differentiation as the most strongly weighted differentially methylated pathway that overlapped in both gout cohorts. CONCLUSION These findings of differential DNA methylation of networked signaling, transcriptional, innate and adaptive immunity, and osteoclastogenesis genes and pathways suggest that they could serve as novel therapeutic targets in the management of flares, tophi, chronic synovitis, and bone erosion in patients with gout.
Collapse
Affiliation(s)
| | | | | | - Ru Liu-Bryan
- University of California, San Diego and San Diego VAMC
| | | | | | - Jun Wang
- University of California, San Diego
| | | | - Wei Wang
- University of California, San Diego
| | | |
Collapse
|