1
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, De Sanctis JB, Martí-Amarista CE, Hidalgo R, Alegría-Barrero E, Riera Lizardo RJ, Correa-Pérez A. Colchicine for the primary prevention of cardiovascular events. Cochrane Database Syst Rev 2025; 2:CD015003. [PMID: 39927511 DOI: 10.1002/14651858.cd015003.pub2] [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] [Indexed: 02/11/2025]
Abstract
BACKGROUND Atherosclerotic cardiovascular diseases (ACVDs), a condition characterised by lipid accumulation in arterial walls, which is often exacerbated by chronic inflammation disorders, is the major cause of mortality and morbidity worldwide. Colchicine, with its first medicinal use in ancient Egypt, is an inexpensive drug with anti-inflammatory properties. However, its role in primary prevention of ACVDs in the general population remains unknown. OBJECTIVES To assess the clinical benefits and harms of colchicine as primary prevention of cardiovascular outcomes in the general population. SEARCH METHODS We searched the Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, Web of Science, and LILACS. We searched ClinicalTrials.gov and WHO ICTRP for ongoing and unpublished studies. We also scanned the reference lists of relevant included studies, reviews, meta-analyses, and health technology reports to identify additional studies. There were no limitations on language, date of publication, or study setting. The search results were updated on 31 May 2023. SELECTION CRITERIA Randomised controlled trials (RCTs) in any setting, recruiting adults without pre-existing cardiovascular disease. We included trials that compared colchicine versus placebo, non-steroidal anti-inflammatory drugs, corticosteroids, immunomodulating drugs, or usual care. Our primary outcomes were all-cause mortality, non-fatal myocardial infarction, stroke, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors independently selected studies, extracted data, and performed risk of bias and GRADE assessments. MAIN RESULTS We identified 15 RCTs (1721 participants randomised; 1412 participants analysed) with follow-up periods ranging from 4 to 728 weeks. The intervention was oral colchicine compared with placebo, immunomodulating drugs, or usual care or no treatment. Due to biases and imprecision, the evidence was very uncertain for all outcomes. All trials but one had a high risk of bias. Five out of seven meta-analyses included fewer than six trials (71.4%). The objectives of the review were to assess cardiovascular outcomes in the general population, but many of the included trials focused on liver disease. Colchicine compared to placebo Colchicine may reduce all-cause mortality compared to placebo in primary prevention, but the evidence is very uncertain (risk ratio (RR) 0.68, 95% confidence interval (CI) 0.51 to 0.91; 6 studies, 463 participants; very low-certainty evidence; number needed to treat for an additional beneficial outcome (NNTB) 11, 95% CI 6 to 67). Colchicine may result in little to no difference in non-fatal myocardial infarction, but the evidence is very uncertain (RR 0.87, 95% CI 0.41 to 1.82; 1 study, 100 participants; very low-certainty evidence). Colchicine may not reduce the incidence of stroke, but the evidence is very uncertain (RR 2.43, 95% CI 0.67 to 8.86; 1 study, 100 participants; very low-certainty evidence). Regarding adverse events, colchicine may increase the incidence of diarrhoea (RR 3.99, 95% CI 1.44 to 11.06; 8 studies, 605 participants; very low-certainty evidence; number needed to treat for an additional harmful outcome (NNTH) 10, 95% CI 6 to 17), and may have little to no effect on neurological outcomes such as seizure or mental confusion (RR 0.72, 95% CI 0.31 to 1.66; 2 studies, 155 participants; very low-certainty evidence), but the evidence is very uncertain. The effect of colchicine on cardiovascular mortality is also very uncertain (RR 1.27, 95% CI 0.03 to 62.43; 2 studies, 160 participants; very low-certainty evidence). Colchicine may not reduce post-cardiac procedure atrial fibrillation, but the evidence is very uncertain (RR 0.74, 95% CI 0.25 to 2.19; 1 study, 100 participants). We found no trials reporting on pericardial effusion, peripheral artery disease, heart failure, or unstable angina. Colchicine compared to methotrexate (immunomodulating drug) Colchicine may result in little to no difference in all-cause mortality compared to methotrexate, but the evidence is very uncertain (RR 0.42, 95% CI 0.12 to 1.51; 1 study, 85 participants; very low-certainty evidence). We found no trials reporting other cardiovascular outcomes or adverse events for this comparison. Colchicine compared to usual care or no treatment The evidence is very uncertain about the effect of colchicine compared with usual care on all-cause mortality in primary prevention (RR 1.07, 95% CI 0.90 to 1.27; 2 studies, 729 participants; very low-certainty evidence). Regarding adverse events, colchicine may increase the incidence of diarrhoea compared to usual care, but the evidence is very uncertain (RR 3.32, 95% CI 1.56 to 7.03; 2 studies, 729 participants; very low-certainty evidence; NNTH 18, 95% CI 12 to 42). No trials reported other cardiovascular outcomes for this comparison. AUTHORS' CONCLUSIONS This Cochrane review evaluated the clinical benefits and harms of using colchicine for the primary prevention of cardiovascular events in the general population. Comparisons were made against placebo, immunomodulating medications, or usual care or no treatment. However, the certainty of the evidence for the predefined outcomes was very low, highlighting the pressing need for high-quality, rigorous studies to ascertain colchicine's clinical impact definitively. We identified numerous biases and inaccuracies in the included studies, limiting their generalisability and precluding a conclusive determination of colchicine's efficacy in preventing cardiovascular events. The existing evidence regarding colchicine's potential cardiovascular benefits or harms for primary prevention is inconclusive owing to the limitations inherent in the current studies. More robust clinical trials are needed to bridge this evidence gap effectively.
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador, Universidad UTE, Quito, Ecuador
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Mario A Gemmato-Valecillos
- Department of Medicine, NYC Health + Hospitals / Elmhurst, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | - Juan Bautista De Sanctis
- The Institute of Molecular and Translational Medicine, Palacky University Olomouc, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | | | - Ricardo Hidalgo
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador., Universidad UTE, Quito, Ecuador
| | | | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
- Medicine Department, Rheumatology Unit, Universidad de Carabobo, Valencia, Venezuela
| | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Department of Hospital Pharmacy and Medical Devices, Hospital Central de la Defensa Gómez Ulla, Madrid, Spain
| |
Collapse
|
2
|
Li G, Xu J, Li H, Yan W, Chen F, Yuan A, Zhang J. Network Pharmacology Suggests Mechanisms for Therapeutic Effects of Caulis Sinomenii on Avian Gout. J Poult Sci 2025; 62:2025002. [PMID: 39781185 PMCID: PMC11701077 DOI: 10.2141/jpsa.2025002] [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: 10/21/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
Avian gout (AG) is detrimental to the survival and production performance of poultry and effective drugs are lacking. Caulis sinomenii has shown clinical efficacy against arthritis and may have potential value in AG prevention and treatment. In the present study, the components and targets of C. sinomenii and AG-related targets were identified using relevant databases. The common targets, target interactions, and signaling pathways involved in the prevention and treatment of AG by C. sinomenii were determined using software to explore the potential mechanisms of action. Sixteen components of C. sinomenii, eight of which were active ingredients with 351 targets and 2993 AG-related targets, were identified using several databases. A total of 156 common targets were associated with 202 biological processes and 34 pathways. Toll-like receptor 4 (TLR4) and prostaglandin endoperoxide synthase 2 were core targets. These targets may exert therapeutic effects on AG through four pathways: the nucleotide-binding oligomerization domain (NOD)-like receptor, mammalian target of rapamycin, TLR, and mitogen-activated protein kinase signaling pathways. In summary, C. sinomenii has potential therapeutic efficacy against AG through multicomponent, multi-target, and multi-pathway mechanisms.
Collapse
Affiliation(s)
- Guanyang Li
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- College of Biological and Food Engineering/Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province/Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Huaihua University, Huaihua 418000, China
| | - Junfei Xu
- College of Biological and Food Engineering/Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province/Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Huaihua University, Huaihua 418000, China
| | - Huanhuan Li
- College of Biological and Food Engineering/Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province/Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Huaihua University, Huaihua 418000, China
| | - Wenxin Yan
- College of Biological and Food Engineering/Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province/Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Huaihua University, Huaihua 418000, China
| | - Fengting Chen
- College of Biological and Food Engineering/Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province/Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Huaihua University, Huaihua 418000, China
| | - Anwen Yuan
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Juzuo Zhang
- Department of Clinic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- College of Biological and Food Engineering/Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province/Hunan Provincial Higher Education Key Laboratory of Intensive Processing Research on Mountain Ecological Food, Huaihua University, Huaihua 418000, China
| |
Collapse
|
3
|
Chen F, Li Y, Zhao L, Lin C, Zhou Y, Ye W, Wan W, Zou H, Xue Y. Anti-inflammatory effects of MerTK by inducing M2 macrophage polarization via PI3K/Akt/GSK-3β pathway in gout. Int Immunopharmacol 2024; 142:112942. [PMID: 39217874 DOI: 10.1016/j.intimp.2024.112942] [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: 04/09/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Mer tyrosine kinase (MerTK) has been found to regulate the secretion of inflammatory factors and exert immunosuppressive effects, but its role in gout remains unclear. In this study, we aimed to clarify the immnue effects of MerTK in gout. MerTK in synovium or serum of gout patients was determined by immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), and real-time quantitative polymerase chain reaction (RT-qPCR). In monosodium urate (MSU)-induced gout mice, the effect of MerTK inhibitor (UNC2250) on inflammation and polarization was also assessed. After inhibition, knockdown or overexpression of MerTK, inflammatory response and polarization level in THP1-derived macrophages were evaluated by RT-qPCR and flow cytometry. Regulation of MerTK inhibitors on mitochondrial function and downstream pathway in THP1-derived macrophages were detected. MerTK in synovium and serum of gout patients were increased. MerTK inhibitor stimulated the inflammation and M1 polarization in MSU-induced gout mice. MerTK inhibition, knock-down, or overexpression affected inflammatory response, polarization and mitochondrial function in vitro in gout model. The PI3K/Akt/GSK-3β pathway was identified to reduce after MerTK inhibition and the relevant results were as expected, validated by knock-down or overexpressing MerTK. In conclusion, MerTK was detected to increase in both gout patients and model. MerTK influenced inflammatory response and polarization markers through PI3K/Akt/GSK-3β pathway. Interfering MerTK/PI3K/Akt/GSK-3β axis may provide a new therapeutic target for gout.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yixuan Li
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Li Zhao
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Cong Lin
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yingzi Zhou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Weiguo Wan
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Xiong Z, Ding Z, Sun J, Jiang X, Cong H, Sun H, Qiao F. In vivo assembly in tobacco cells to elucidate and engineer the biosynthesis of 4-hydroxydihydrocinnamaldehyde from Gloriosa superba. PLANT CELL REPORTS 2024; 43:235. [PMID: 39299972 DOI: 10.1007/s00299-024-03318-4] [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: 08/05/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
KEY MESSAGE This study described the biosynthesis of 4-hydroxydihydrocinnamaldehyde sharing with monolignol pathway and supplemented the biosynthesis of colchicine in G. superba, 4-hydroxydihydrocinnamaldehyde produced in tobacco BY2 cells provided an important stepstone. The precursor, 4-hydroxydihydrocinnamaldehyde (4-HDCA), participates in the biosynthesis of the carbon skeleton of colchicine, which is derived from L-phenylalanine. However, one hypothesis proposed that 4-HDCA is synthesized by sharing the early part of the monolignol pathway in G. superba. In this study, we validated this prediction and identified the enzymatic functions involved in this pathway. GsDBR1 is a crucial enzyme to illustrate 4-HDCA diverging from monolignol pathway, we first confirmed its reductase activity on 4-coumaraldehyde, an important intermediate compound in monolignol biosynthesis. Then, the biochemical function of recombinant enzymes belonging to the other four families were verified to elucidate the entire process of 4-HDCA biosynthesis from L-phenylalanine. After reconstruction, the 4-HDCA was 78.4 ng/g with fresh weight (FW) of transgenic tobacco cells, and the yield increased to 168.22 ng/g·FW after improved treatment with methyl jasmonate (MeJA). The elucidation of 4-HDCA biosynthesis sharing the monolignol pathway supplemented the biosynthesis of colchicine in G. superba, and the production of 4-HDCA in tobacco cells provides an important step in the development of plant cell cultures as heterologous bio-factories for secondary metabolite production.
Collapse
Affiliation(s)
- Zhiqiang Xiong
- National Key Laboratory for Tropical Crops Breeding, Sanya, 572024, China
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Zhuoying Ding
- National Key Laboratory for Tropical Crops Breeding, Sanya, 572024, China
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Jingyi Sun
- National Key Laboratory for Tropical Crops Breeding, Sanya, 572024, China
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Xuefei Jiang
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Hanqing Cong
- National Key Laboratory for Tropical Crops Breeding, Sanya, 572024, China
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Huapeng Sun
- National Key Laboratory for Tropical Crops Breeding, Sanya, 572024, China.
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China.
| | - Fei Qiao
- National Key Laboratory for Tropical Crops Breeding, Sanya, 572024, China.
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China.
| |
Collapse
|
5
|
Li Y, Zhuang Y, Chen Y, Wang G, Tang Z, Zhong Y, Zhang Y, Wu L, Ji X, Zhang Q, Pan B, Luo Y. Euphorbia factor L2 alleviated gouty inflammation by specifically suppressing both the priming and activation of NLRP3 inflammasome. Int Immunopharmacol 2024; 138:112598. [PMID: 38981223 DOI: 10.1016/j.intimp.2024.112598] [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: 03/20/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
Euphorbia L. is a traditionally used herb and contains many newly identified compounds with novel chemical structures. Euphorbia factor L2 (EFL2), a diterpenoid derived from Euphorbia seeds, is reported to alleviate acute lung injury and arthritis by exerting anti-inflammatory effects. In this study, we aimed to test the therapeutic benefit and mechanisms of EFL2 in NLRP3 inflammasome-mediated gouty models and identified the potential molecular mechanism. A cell-based system was used to test the specific inhibitory effect of EFL2 on NLRP3-related inflammation. The gouty arthritis model and an air pouch inflammation model induced by monosodium urate monohydrate (MSU) crystals were used for in vivo experiments. Nlrp3-/- mice and in vitro studies were used for mechanistic exploration. Virtual molecular docking and biophysical assays were performed to identify the direct binding and regulatory target of EFL2. The inhibitory effect of EFL2 on inflammatory cell infiltration was determined by flow cytometry in vivo. The mechanism by which EFL2 activates the NLRP3 inflammasome signaling pathway was evaluated by immunological experiment and transmission electron microscopy. In vitro, EFL2 specifically reduced NLRP3 inflammasome-mediated IL-1β production and alleviated MSU crystal-induced arthritis, as well as inflammatory cell infiltration. EFL2 downregulated NF-κB phosphorylation and NLRP3 inflammasome expression by binding to glucocorticoid receptors. Moreover, EFL2 could specifically suppress the lysosome damage-mediated NLRP3 inflammasome activation process. It is expected that this work may be useful to accelerate the development of anti-inflammatory drugs originated from traditional herbs and improve therapeutics in gout and its complications.
Collapse
Affiliation(s)
- Yanhong Li
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuqing Zhuang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuehong Chen
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guan Wang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 6a004a, Sichuan, China
| | - Zhigang Tang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yutong Zhong
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuanyuan Zhang
- Sichuan Institute of Food Inspection, Chengdu, Sichuan, China
| | - Liang Wu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing Ji
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiuping Zhang
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Pan
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang 262700, Shandong, China
| | - Yubin Luo
- Department of Rheumatology & Immunology, Laboratory of Rheumatology & Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Liu F, Bai Y, Wan Y, He J, Li Q, Xie Y, Guo P. Mechanism of flavonoids in the treatment of gouty arthritis (Review). Mol Med Rep 2024; 30:132. [PMID: 38818832 PMCID: PMC11157185 DOI: 10.3892/mmr.2024.13256] [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: 12/20/2023] [Accepted: 04/17/2024] [Indexed: 06/01/2024] Open
Abstract
The present review expounds the advancements in the application and mechanisms of flavonoids in gouty arthritis, highlighting their significance in managing the disease. Gouty arthritis is among the most common and severe inflammatory diseases, caused by hyperuricemia and the deposition of sodium urate crystals in the joints and surrounding tissues, posing a serious threat to human life and health. Flavonoids, extracted from various herbs, have attracted significant attention due to their efficacy in improving gouty arthritis. The present study systematically reviews the in vivo studies and in vitro animal studies on flavonoids from herbal medicines for the treatment of gouty arthritis that have been previously published in the PubMed, ScienceDirect, Google Scholar and China National Knowledge Infrastructure databases between 2000 and 2023. The review of the literature indicated that flavonoids can improve gouty arthritis through multiple mechanisms. These include lowering xanthine oxidase activity, inhibiting uric acid (UA) synthesis, regulating UA transporters to promote UA excretion, reducing the inflammatory response and improving oxidative stress. These mechanisms predominantly involve regulating the NOD‑like receptor 3 inflammasome, the Toll‑like receptor 4/myeloid differentiation factor 88/nuclear factor‑κB signaling pathway, and the levels of UA transporter proteins, namely recombinant urate transporter 1, glucose transporter 9, organic anion transporter (OAT)1 and OAT3. Various flavonoids used in traditional Chinese medicine hold therapeutic promise for gouty arthritis and are anticipated to pave the way for novel pharmaceuticals and clinical applications.
Collapse
Affiliation(s)
- Feifan Liu
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuanmei Bai
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yan Wan
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Jinglin He
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Qiongchao Li
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuhuan Xie
- College of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Peixin Guo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
7
|
Oh TK, Park HY, Song IA. Insomnia disorder and cancer mortality in South Korea: a secondary analysis of musculoskeletal disease cohort. Sleep Breath 2024; 28:1311-1318. [PMID: 38418767 DOI: 10.1007/s11325-024-03009-0] [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: 10/31/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE The association between insomnia disorder and cancer-related mortality risk remains controversial. Therefore, this study aimed to investigate the correlation between insomnia disorder and cancer-related mortality. METHODS Patients who were diagnosed with musculoskeletal disease (MSD) between 2010 and 2015 were included in this study as a secondary analysis of a patient cohort with MSD in South Korea. Cancer mortality was evaluated between January 1, 2016, and December 31, 2020, using multivariable Cox regression modeling. Patients with and without insomnia disorder constituted the ID and non-ID groups, respectively. RESULTS The final analysis incorporated a total of 1,298,314 patients diagnosed with MSDs, of whom 11,714 (0.9%) died due to cancer. In the multivariable Cox regression model, the risk of total cancer-related mortality was 14% (hazard ratio [HR], 1.14; 95% confidence interval [CI], 1.10-1.19; P < 0.001) higher in the ID group than in the non-ID group. Moreover, the ID group had a higher risk of mortality due to esophageal (HR, 1.46; 95% CI, 1.08-1.96; P = 0.015), colorectal (HR, 1.20; 95% CI, 1.05-1.36; P = 0.007), head and neck (HR, 1.39; 95% CI, 1.01-1.94; P = 0.049), lung (HR, 1.17; 95% CI, 1.08-1.27; P < 0.001), and female genital organ (HR: 1.39, 95% CI: 1.09, 1.77; P = 0.008) cancers; leukemia; and lymphoma (HR, 1.30; 95% CI, 1.12-1.49; P < 0.001). CONCLUSION Insomnia disorder was associated with elevated overall cancer mortality in patients with MSDs, which was more evident for cancer mortality due to esophageal, colorectal, head and neck, lung, and female genital organ cancers; leukemia; and lymphoma.
Collapse
Affiliation(s)
- Tak Kyu Oh
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Gumi-ro, 173, Beon-gil, Bundang-gu, Seongnam, 13620, South Korea
- Department of Anesthesiology and Pain Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Hye Yoon Park
- Department of Psychiatry, Seoul National University Hospital, Seoul, South Korea
| | - In-Ae Song
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Gumi-ro, 173, Beon-gil, Bundang-gu, Seongnam, 13620, South Korea.
- Department of Anesthesiology and Pain Medicine, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
8
|
Javed C, Noreen R, Niazi SG, Kiyani MM, Ul Ain Q. Anti-gouty arthritis and anti-inflammatory effects of curcumin nanoparticles in monosodium urate crystals induced Balb/C mice. Inflammopharmacology 2024; 32:1929-1940. [PMID: 38556563 DOI: 10.1007/s10787-024-01450-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: 01/03/2024] [Accepted: 02/21/2024] [Indexed: 04/02/2024]
Abstract
Gout is a metabolic condition characterized by the accumulation of urate crystals in the synovial joints. These crystal depositions result in joint swelling and increased concentration of serum uric acid in blood. The commercially available drugs lower serum uric acid levels and reduce inflammation, but these standard therapies have many side effects. This study aimed to investigate anti-gout and anti-inflammatory properties of curcumin nanoparticles (CNPs). For this purpose, CNPs were prepared by dissolving curcumin into dichloromethane. Then, gout was induced by injecting monosodium urate crystals (MSU) in the ankle joint and in the intra-peritoneal cavity which caused ankle swelling and increased blood uric acid levels. CNPs in different concentrations (5, 10, and 20 ppm) and allopurinol were orally administered. The MSU crystals increased the xanthine oxidase levels both in serum and the liver. Moreover, MSU crystals increased the serum levels of interleukin 1β, interleukin-6, tumor necrosis factor-alpha, liver function tests markers, renal function tests markers, and lipid profiles. However, the administration of CNPs decreased the levels of all these variables. CNPs increased the serum high-density lipoprotein and interleukin-10 levels. Moreover, CNPs also reduced ankle swelling significantly. Hence, the levels of xanthine oxidase, uric acid and ankle swelling were reduced significantly by oral administration of CNPs. Our findings indicate that CNPs through their anti-inflammatory properties significantly alleviate gouty arthritis. Thus, the study concluded that CNPs can be developed as an efficient anti-gout agent with minimal side effects.
Collapse
Affiliation(s)
- Chanda Javed
- Department of Biochemistry, Government College University, Faisalabad, 38000, Pakistan
| | - Razia Noreen
- Department of Biochemistry, Government College University, Faisalabad, 38000, Pakistan.
| | - Samia Gul Niazi
- Faculty of Pharmacy, Hamdard University Islamabad Campus, Islamabad, Pakistan
| | - Mubin Mustafa Kiyani
- Shifa College of Medical Technology, Shifa Tameer e Milat University, Islamabad, Pakistan
| | - Qurat Ul Ain
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, Pakistan.
| |
Collapse
|
9
|
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
|
10
|
Elshiwy K, Amin GEED, Farres MN, Samir R, Allam MF. The role of colchicine in the management of COVID-19: a Meta-analysis. BMC Pulm Med 2024; 24:190. [PMID: 38641775 PMCID: PMC11031948 DOI: 10.1186/s12890-024-03001-0] [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: 07/04/2023] [Accepted: 04/08/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND The Coronavirus disease 2019 (COVID-19) pandemic has robustly affected the global healthcare and economic systems and it was caused by coronavirus-2 (SARS-CoV-2). The clinical presentation of the disease ranges from a flu-like illness to severe pneumonia and death. Till September 2022, the cumulative number of cases exceeded 600 million worldwide and deaths were more than 6 million. Colchicine is an alkaloid drug that is used in many autoinflammatory conditions e.g., gout, familial Mediterranean fever, and Behçet's syndrome. Colchicine inhibits the production of superoxide and the release of interleukins that stimulate the inflammatory cascade. Colchicine decreases the differentiation of myofibroblast and the release of fibrotic mediators including transforming growth factor (TGF-β1) that are related to the fibrosis. Moreover, colchicine has been used to traet viral myocarditis caused by CMV or EBV, interstitial pneumonia, and pericarditis resulting from influenza B infection. Additionally, colchicine is considered safe and affordable with wide availability. OBJECTIVE The aim of the current study was to assess the evidence of colchicine effectiveness in COVID-19 treatment. METHODS A comprehensive review of the literature was done till May 2022 and yielded 814 articles after ranking the articles according to authors and year of publication. Only 8 clinical trials and cohort studies fulfilling the inclusion criteria were included for further steps of data collection, analysis, and reporting. RESULTS This meta-analysis involved 16,488 patients; 8146 patients in the treatment group and 8342 patients in the control group. The results showed that colchicine resulted in a significant reduction in the mortality rate among patients received colchicine in comparison with placebo or standard care (RR 0.35, 95%CI: 0.15-0.79). Colchicine resulted in a significant decrease in the need for O2 therapy in patients with COVID-19 (RR 0.07, 95%CI 0.02-0.27, P = 0.000024). However, colchicine had no significant effect on the following outcomes among COVID-19 patients: the need for hospitalization, ICU admission, artificial ventilation, and hospital discharge rate. Among the PCR confirmed COVID-19 patients, colchicine decreased the hospitalization rate (RR 0.75, 95%CI 0.57-0.99, P = 0.042). However, colchicine had no effect on mortality and the need for mechanical ventilation among this subgroup. CONCLUSION Colchicine caused a significant clinical improvement among COVID-19 patients as compared with the standard care or placebo, in terms of the need for O2, and mortality. This beneficial effect could play a role in the management of COVID-19 especially severe cases to decrease need for oxygen and to decrease mortality among these patients.
Collapse
Affiliation(s)
- Kholoud Elshiwy
- Department of Family Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Ghada Essam El-Din Amin
- Department of Family Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Community, Environmental and Occupational Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Nazmy Farres
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Rasha Samir
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Farouk Allam
- Department of Family Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Preventive Medicine and Public Health, Faculty of Medicine, University of Cordoba, 14004, Cordoba, Spain
| |
Collapse
|
11
|
Alarabei AA, Abd Aziz NAL, AB Razak NI, Abas R, Bahari H, Abdullah MA, Hussain MK, Abdul Majid AMS, Basir R. Immunomodulating Phytochemicals: An Insight Into Their Potential Use in Cytokine Storm Situations. Adv Pharm Bull 2024; 14:105-119. [PMID: 38585461 PMCID: PMC10997936 DOI: 10.34172/apb.2024.001] [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: 02/18/2023] [Revised: 05/17/2023] [Accepted: 07/14/2023] [Indexed: 04/09/2024] Open
Abstract
Phytochemicals are compounds found in plants that possess a variety of bioactive properties, including antioxidant and immunomodulatory properties. Recent studies have highlighted the potential of phytochemicals in targeting specific signalling pathways involved in cytokine storm, a life-threatening clinical condition resulting from excessive immune cell activation and oversupply of proinflammatory cytokines. Several studies have documented the immunomodulatory effects of phytochemicals on immune function, including their ability to regulate essential cellular and molecular interactions of immune system cells. This makes them a promising alternative for cytokine storm management, especially when combined with existing chemotherapies. Furthermore, phytochemicals have been found to target multiple signalling pathways, including the TNF-α/NF-κB, IL-1/NF-κB, IFN-γ/JAK/STAT, and IL-6/JAK-STAT. These pathways play critical roles in the development and progression of cytokine storm, and targeting them with phytochemicals represents a promising strategy for controlling cytokine release and the subsequent inflammation. Studies have also investigated certain families of plant-related constituents and their potential immunomodulatory actions. In vivo and in vitro studies have reported the immunomodulatory effects of phytochemicals, which provide viable alternatives in the management of cytokine storm syndrome. The collective data from previous studies suggest that phytochemicals represent a potentially functional source of cytokine storm treatment and promote further exploration of these compounds as immunomodulatory agents for suppressing specific signalling cascade responses. Overall, the previous research findings support the use of phytochemicals as a complementary approach in managing cytokine storm and improving patient outcomes.
Collapse
Affiliation(s)
- Abdusalam Abdullah Alarabei
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nur Aimi Liyana Abd Aziz
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nur Izah AB Razak
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Razif Abas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Maizaton Atmadini Abdullah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Mohd Khairi Hussain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Amin Malik Shah Abdul Majid
- Natureceuticals Sdn Bhd, Kedah Halal Park, Kawasan Perindustrian Sg. Petani, 08000 Sg. Petani, Kedah, Malaysia
| | - Rusliza Basir
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
12
|
Jafari-Nozad AM, Jafari A, Yousefi S, Bakhshi H, Farkhondeh T, Samarghandian S. Anti-gout and Urate-lowering Potentials of Curcumin: A Review from Bench to Beside. Curr Med Chem 2024; 31:3715-3732. [PMID: 37488765 DOI: 10.2174/0929867331666230721154653] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/23/2023] [Accepted: 06/01/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Gouty arthritis is a complex form of inflammatory arthritis, triggered by the sedimentation of monosodium urate crystals in periarticular tissues, synovial joints, and other sites in the body. Curcumin is a natural polyphenol compound, isolated from the rhizome of the plant Curcuma longa, possessing countless physiological features, including antioxidant, anti-inflammatory, and anti-rheumatic qualities. OBJECTIVE This study aimed to discuss the beneficial impacts of curcumin and its mechanism in treating gout disease. METHODS Ten English and Persian databases were used to conduct a thorough literature search. Studies examining the anti-gouty arthritis effects of curcumin and meeting the inclusion criteria were included. RESULTS According to the studies, curcumin has shown xanthine oxidase and urate transporter- 1 inhibitory properties, uric acid inhibitory characteristics, and antioxidant and anti- inflammatory effects. However, some articles found no prominent reduction in uric acid levels. CONCLUSION In this review, we emphasized the potency of curcumin and its compounds against gouty arthritis. Despite the potency, we suggest an additional well-designed evaluation of curcumin, before its therapeutic effectiveness is completely approved as an antigouty arthritis agent.
Collapse
Affiliation(s)
| | - Amirsajad Jafari
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Saman Yousefi
- Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Hasan Bakhshi
- Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
| |
Collapse
|
13
|
Ling M, Gan J, Hu M, Pan F, Liu M. IL1A regulates the inflammation in gout through the Toll-like receptors pathway. Int J Med Sci 2024; 21:188-199. [PMID: 38164346 PMCID: PMC10750337 DOI: 10.7150/ijms.88447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/21/2023] [Indexed: 01/03/2024] Open
Abstract
Objective: Gout is a dangerous metabolic condition related to monosodium urate (MSU). Our aim is to study the molecular mechanisms underlying gout and to identify potential clinical biomarkers by bioinformatics analysis and experimental validation. Methods: In this study, we retrieved the overlapping genes between GSE199950-Differential Expressed Genes (DEGs) dataset and key module in Weighted Gene Co-Expression Network Analysis (WGCNA) on GSE199950. These genes were then analyzed by protein-protein interaction (PPI) network, expression and Gene Set Enrichment Analysis to identify the hub gene related to gout. Then, the gene was investigated by peripheral blood mononuclear cells (PBMCs), immunoassay and cell experiments like western blotting to uncover its underlying mechanism in gout cells. Results: From the turquoise module and 83 DEGs, we identified 62 overlapping genes, only 11 genes had mutual interactions in PPI network and these genes were highly expressed in MSU-treated samples. Then, it was found that the IL1A (interleukin 1 alpha) was the only one gene related to Toll-like receptor signaling pathway that was associated with the occurrence of gout. Thus, IL1A was determined as the hub gene in this study. In immunoassay, IL1A was significantly positively correlated with B cells and negatively correlated with macrophages. Moreover, IL1A is highly expressed in gout patients,it has a good clinical diagnostic value. Finally, the results of in vitro experiments showed that after knocking down IL1A, the expressions of pro-inflammatory cytokines and Toll-like receptor signaling pathway-related proteins (TLR2, TLR4, MyD88) were all reduced. Conclusion: It is confirmed that IL1A is a promoting gene in gout with a good diagnostic value, and specifically it affects the inflammation in gout through Toll-like receptor pathway. Our research offers fresh perspectives on the pathophysiology of gout and valuable directions for future diagnosis and treatment.
Collapse
Affiliation(s)
- Meirong Ling
- Emergency Medical Department, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Jiaqi Gan
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Mengting Hu
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Fei Pan
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| | - Mei Liu
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, 201199, Shanghai, China
| |
Collapse
|
14
|
Mohapatra A, Mohanty A, Sathiyamoorthy P, Chahal S, Vijayan V, Rajendrakumar SK, Park IK. Targeted treatment of gouty arthritis by biomineralized metallic nanozyme-mediated oxidative stress-mitigating nanotherapy. J Mater Chem B 2023; 11:7684-7695. [PMID: 37464890 DOI: 10.1039/d3tb00669g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Gouty arthritis is characterized by chronic deposition of monosodium urate (MSU) crystals in the joints and other tissues, resulting in the production of excess reactive oxygen species (ROS) and proinflammatory cytokines that intensify synovial inflammation. This condition is mainly associated with inflammatory M1 macrophage activation and oxidative stress production. Hence, gout symptoms can often be resolved by eliminating M1 macrophage activation and scavenging oxidative stress in the inflamed areas. Herein, we developed M1-macrophage-targeting biomineralized metallic nanozymes (FALNZs) that deplete oxidative stress and reduce the M1 macrophage levels to mitigate gouty arthritis. Intra-articular injection of the FALNZs targets inflammatory macrophages and suppresses ROS levels in joints with MSU-crystal-induced arthritis. In addition, the FALNZs alleviate joint swelling, inflammatory cytokine production, and pathological features of the joints. Overall, the proposed therapeutic approach is biocompatible and is an effective ROS scavenger for the treatment of gouty pathogenesis.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Ayeskanta Mohanty
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Padmanaban Sathiyamoorthy
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Sahil Chahal
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | - Veena Vijayan
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| | | | - In-Kyu Park
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju 61469, South Korea.
| |
Collapse
|
15
|
Yu H, Xue W, Yu H, Gu H, Qin L, Peng A. Joint Application of Multiple Inflammatory Cytokines in Diagnosis of Gout Flare. J Inflamm Res 2023; 16:1771-1782. [PMID: 37113627 PMCID: PMC10128086 DOI: 10.2147/jir.s408929] [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: 02/16/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Purpose This study aimed to explore the accuracy for joint application of inflammatory cytokines in diagnosis of gout flare by comparison with peripheral blood cells. Methods We collected the clinical data of 96 acute gout patients and 144 remission gout patients, and compared the levels of peripheral blood cells, inflammatory cytokines and blood biochemistry indexes between acute and remission gout. We respectively assessed the area under curves (AUCs) for single and multiple inflammatory cytokines including C-reactive protein (CRP), interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), and single and multiple peripheral blood cells including platelet (PLT), white blood cell (WBC), percentages of neutrophils (N%), lymphocytes (L%), eosinophils (E%), basophils (B%) in diagnosis of acute gout by receiver operating characteristic (ROC) curve analysis. Results By contrast with remission gout, the levels of PLT, WBC, N%, CRP, IL-1β, IL-6 and TNF-α increased, and the levels of L%, E% and B% decreased in acute gout. The AUCs of PLT, WBC, N%, L%, E% and B% in diagnosis of acute gout were respectively 0.591, 0.601, 0.581, 0.567, 0.608 and 0.635, while the AUC for joint application of these peripheral blood cells was 0.674. Moreover, the AUCs of CRP, IL-1β, IL-6 and TNF-α in diagnosis of acute gout were respectively 0.814, 0.683, 0.622 and 0.746, while the AUC for joint application of these inflammatory cytokines was 0.883, reflecting significantly higher levels than peripheral blood cells. Conclusion The joint application of multiple inflammatory cytokines can better distinguish acute gout from remission gout compared with peripheral blood cells.
Collapse
Affiliation(s)
- Hanqing Yu
- Center for Nephrology and Clinical Metabolomics, Division of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Wen Xue
- Center for Nephrology and Clinical Metabolomics, Division of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Hanjie Yu
- Center for Nephrology and Clinical Metabolomics, Division of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Hongchen Gu
- Center for Nephrology and Clinical Metabolomics, Division of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Ling Qin
- Center for Nephrology and Clinical Metabolomics, Division of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Ai Peng
- Center for Nephrology and Clinical Metabolomics, Division of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Ai Peng; Ling Qin, Center for Nephrology and Clinical Metabolomics, Division of Nephrology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072, People’s Republic of China, Email ;
| |
Collapse
|
16
|
Xu H, Wu J, Wang S, Xu L, Liu P, Shi Y, Wu S, Deng L, Chen X. Network pharmacology and in vivo experiments reveal the pharmacological effects and molecular mechanisms of Simiao Powder in prevention and treatment for gout. BMC Complement Med Ther 2022; 22:152. [PMID: 35672755 PMCID: PMC9175488 DOI: 10.1186/s12906-022-03622-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background Gout is a common disease with high incidence due to unhealthy diet and living habits. Simiao Powder, as a classic formula consisted of four common herbs, has been widely used in clinical practice since ancient times to prevent and treat gout. However, the pharmacological mechanism of Simiao Powder is still unclear. Methods Based on network pharmacology, Simiao Powder active compounds were identified in TCMSP, ETCM and BATMAN database, used to establish a network of interaction between potential targets of Simiao Powder and known therapeutic targets of gout. Subsequently, the key potential targets are being used for protein–protein interaction, GO enrichment analysis and KEGG pathway enrichment analysis through several authoritative open databases. Molecular docking through AutoDockTools software can verify interaction between molecules. Finally, to validate the predicted results, in vivo experiments based on hyperuricemic-gout mice model were designed and treated with Simiao powder and allopurinol. Serum levels of uric acid (UA), creatinine (Cr), blood urea nitrogen (BUN) and xanthine oxidase (XOD) were determined using a customized assay kit while the expression of PPAR-γ, PTGS1, IL-6 and Bcl2 mRNA were analyzed through qRT-PCR. Results Disease-target-compound network was visualized basing on the 20 bioactive compounds and the 19 potential targets using Cytoscape software. The results of PPI analysis, GO enrichment and KEGG pathway enrichment analysis indicate that the potential mechanism of Simiao Powder in treating gout may be achieved by regulating immune and inflammatory reactions, improving metabolism and endocrine. The results of molecular docking show that most of the targets and components have good binding activity. In vivo experiments revealed that Simiao powder can decreased serum UA and XOD levels in hyperuricemic-gout mice, and improved renal function. Furthermore, Simiao powder certainly regulates the expression of PPAR-γ, PTGS1, IL-6 and Bcl2 mRNA in ankle tissue in hyperuricemic-gout mice. Conclusion Collectively, this research predicted a multiple compounds, targets, and pathways model mechanism of Simiao Powder in the prevention and treatment of gout, providing new ideas and methods for in-depth research, via vivo experiments.
Collapse
|
17
|
Xiong Z, Wang L, Sun J, Jiang X, Cong H, Sun H, Qiao F. Functional characterization of a Colchicum autumnale L. double-bond reductase (CaDBR1) in colchicine biosynthesis. PLANTA 2022; 256:95. [PMID: 36214872 DOI: 10.1007/s00425-022-04003-0] [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: 07/27/2022] [Accepted: 09/21/2022] [Indexed: 06/16/2023]
Abstract
An alkenal double-bond reductase enzyme (CaDBR1) was cloned from Colchicum autumnale L. The encoded enzyme catalysed 4-coumaraldehyde to 4-hydroxydihydrocinnamaldehyde (4-HDCA). Its functional characterization increased the understanding of colchicine biosynthesis. As a traditional medical plant, Colchicum autumnale L. is famous for producing colchicine, a widely used drug for alleviating gout attacks. The biosynthetic pathway of colchicine was revealed most recently, and 4-hydroxydihydrocinnamaldehyde (4-HDCA) has been verified as a crucial intermediate derived from L-phenylalanine. However, the functional gene that catalyses the formation of 4-HDCA remains controversial. In this study, the alkenal double-bond reductase (DBR) gene member CaDBR1 was cloned and characterized from C. autumnale. Bioinformatics analysis predicted and characterized the basic physicochemical properties of CaDBR1. Recombinant CaDBR1 protein was heterologously expressed in Escherichia coli and purified by a Ni-NTA column. In vitro enzyme assays indicated that CaDBR1 could catalyse 4-coumaraldehyde to form 4-HDCA but could not generate 4-HDCA by taking cinnamaldehyde as a substrate. Stable transformation into tobacco BY-2 cells revealed that CaDBR1 localized in the cytoplasm, and tissue-specific expression results showed that CaDBR1 had the highest expression in bulbs. All these results verify and confirm the participation and contribution of CaDBR1 in the biosynthesis pathway of 4-HDCA and colchicine alkaloids in C. autumnale.
Collapse
Affiliation(s)
- Zhiqiang Xiong
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Liang Wang
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Jingyi Sun
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Xuefei Jiang
- Hainan Key Laboratory of Sustainable Utilization of Tropical Bioresources, Key Laboratory for Quality Regulation of Tropical Horticultural Plants of Hainan Province, Sanya Nanfan Research Institute, College of Horticulture, Hainan University, Haikou, 570228, China
| | - Hanqing Cong
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Huapeng Sun
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China.
| | - Fei Qiao
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement in Southern China, Ministry of Agriculture, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| |
Collapse
|
18
|
Lu L, Xiong Y, Lin Z, Chu X, Panayi AC, Hu Y, Zhou J, Mi B, Liu G. Advances in the therapeutic application and pharmacological properties of kinsenoside against inflammation and oxidative stress-induced disorders. Front Pharmacol 2022; 13:1009550. [PMID: 36267286 PMCID: PMC9576948 DOI: 10.3389/fphar.2022.1009550] [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: 08/02/2022] [Accepted: 09/13/2022] [Indexed: 11/19/2022] Open
Abstract
Extensive research has implicated inflammation and oxidative stress in the development of multiple diseases, such as diabetes, hepatitis, and arthritis. Kinsenoside (KD), a bioactive glycoside component extracted from the medicinal plant Anoectochilus roxburghii, has been shown to exhibit potent anti-inflammatory and anti-oxidative abilities. In this review, we summarize multiple effects of KD, including hepatoprotection, pro-osteogenesis, anti-hyperglycemia, vascular protection, immune regulation, vision protection, and infection inhibition, which are partly responsible for suppressing inflammation signaling and oxidative stress. The protective action of KD against dysfunctional lipid metabolism is also associated with limiting inflammatory signals, due to the crosstalk between inflammation and lipid metabolism. Ferroptosis, a process involved in both inflammation and oxidative damage, is potentially regulated by KD. In addition, we discuss the physicochemical properties and pharmacokinetic profiles of KD. Advances in cultivation and artificial synthesis techniques are promising evidence that the shortage in raw materials required for KD production can be overcome. In addition, novel drug delivery systems can improve the in vivo rapid clearance and poor bioavailability of KD. In this integrated review, we aim to offer novel insights into the molecular mechanisms underlying the therapeutic role of KD and lay solid foundations for the utilization of KD in clinical practice.
Collapse
Affiliation(s)
- Li Lu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xiangyu Chu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Adriana C. Panayi
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Hand-, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Yiqiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Juan Zhou
- Department of Cardiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- *Correspondence: Bobin Mi, ; Guohui Liu,
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- *Correspondence: Bobin Mi, ; Guohui Liu,
| |
Collapse
|
19
|
Identification of Interleukin-1-Beta Inhibitors in Gouty Arthritis Using an Integrated Approach Based on Network Pharmacology, Molecular Docking, and Cell Experiments. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2322417. [PMID: 36193152 PMCID: PMC9526673 DOI: 10.1155/2022/2322417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022]
Abstract
Background This study aimed to investigate the molecular mechanism of Tongfengding capsule (TFDC) in treating immune-inflammatory diseases of gouty arthritis (GA) and interleukin-1-beta (IL-1β) inhibitors by using network pharmacology, molecular docking, and cell experiments. Methods In this study, the compounds of TFDC and the potential inflammatory targets of GA were obtained from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), Online Mendelian Inheritance in Man (OMIM), and GeneCards databases. The TFDC-GA-potential targets interaction network was accomplished by the STRING database. The TFDC-active compound-potential target-GA network was constructed using Cytoscape software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were used to further explore the GA mechanism and therapeutic effects of TFDC. Quantitative real-time PCR (qPCR) was used to verify whether the TFDC inhibited IL-1β in GA. Molecular docking technology was used to analyze the optimal effective compounds from the TFDC for docking with IL-1β. Result 133 active compounds and 242 targets were screened from the TFDC, and 25 of the targets intersected with GA inflammatory targets, which were considered as potential therapeutic targets. Network pharmacological analysis showed that the TFDC active compounds such as quercetin, stigmasterol, betavulgarin, rutaecarpine, naringenin, dihydrochelerythrine, and dihydrosanguinarine had better correlation with GA inflammatory targets such as PTGS2, PTGS1, NOS2, SLC6A3, HTR3A, PPARG, MAPK14, RELA, MMP9, and MMP2. The immune-inflammatory signaling pathways of the active compounds for treating GA are IL-17 signaling pathway, TNF signaling pathway, NOD-like receptor signaling pathway, NF-kappa B signaling pathway, Toll-like receptor signaling pathway, HIF-1 signaling pathway, etc. The TFDC reduced IL-1β mRNA expression in GA by qPCR. Molecular docking results suggested that rutaecarpine was the most appropriate natural IL-1β inhibitor. Conclusion Our findings provide an essential role and bases for further immune-inflammatory studies on the molecular mechanisms of TFDC and IL-1β inhibitors development in GA.
Collapse
|
20
|
Wang J, Sun YC. Revealing the pharmacological effect and mechanism of darutoside on gouty arthritis by liquid chromatography/mass spectrometry and metabolomics. Front Mol Biosci 2022; 9:942303. [PMID: 36090056 PMCID: PMC9448993 DOI: 10.3389/fmolb.2022.942303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Darutoside is a diterpenoids compound with significant anti-inflammatory activity, however the pharmacological action and mechanism are still unclear. Metabolomics strategy was used to uncovering the pharmacological action and effective mechanism of darutoside against acute gouty arthritis rats. Liquid chromatography coupled with mass spectrometry technique was performed to explore the serum metabolites and potential pathways. We found that darutoside can up-regulate the level of glutamate, alanine, chenodeoxycholic acid, 1-methyladenosine, aspartic acid, citric acid, and down-regulate the level of valine, isoleucine, glutamine, alanyl-threonine, pyruvic acid, gamma-aminobutyric acid, uric acid. Metabolic pathway analysis showed that the therapeutic effect of darutoside was involved in amino acid metabolism, sugar metabolism, fatty acid metabolism, energy metabolism, purine metabolism and butanoate metabolism. It indicated that darutoside protect against acute gouty arthritis by regulating the expression of the key protein targets. It revealed that the mechanism of darutoside on acute gouty arthritis, which may be leading to the changes of serum metabolites, metabolic pathways and key protein targets to improve immune system response, inhibit oxidative stress and inflammatory response. It provides a novel method for molecular mechanisms of natural product in the disease treatment.
Collapse
Affiliation(s)
- Jing Wang
- School Hospital, Harbin University of Science and Technology, Harbin, China
| | - Yan-Chun Sun
- School Hospital, Harbin University of Science and Technology, Harbin, China
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences /Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin, China
- *Correspondence: Yan-Chun Sun,
| |
Collapse
|
21
|
Martí-Carvajal AJ, De Sanctis JB, Hidalgo R, Martí-Amarista CE, Alegría E, Correa-Pérez A, Monge Martín D, Riera Lizardo RJ. Colchicine for the primary prevention of cardiovascular events. Hippokratia 2022. [DOI: 10.1002/14651858.cd015003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Arturo J Martí-Carvajal
- Cochrane Ecuador. Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC). Facultad de Ciencias de la Salud Eugenio Espejo ; Universidad UTE; Quito Ecuador
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
- Cátedra Rectoral de Médicina Basada en la Evidencia; Universidad de Carabobo; Valencia Venezuela
| | - Juan Bautista De Sanctis
- The Institute of Molecular and Translational Medicine; Palacky University Olomouc, Faculty of Medicine and Dentistry; Czech Republic Czech Republic
| | - Ricardo Hidalgo
- Cochrane Ecuador. Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC). Facultad de Ciencias de la Salud Eugenio Espejo; Universidad Tecnológica Equinoccial; Quito Ecuador
| | | | - Eduardo Alegría
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
| | - Andrea Correa-Pérez
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
- Clinical Biostatistics Unit; Hospital Universitario Ramón y Cajal (IRYCIS); Madrid Spain
| | | | | |
Collapse
|
22
|
Xu X, Yuan Z, Zhang S, Li G, Zhang G. Regulation of TRPV1 channel in monosodium urate-induced gouty arthritis in mice. Inflamm Res 2022; 71:485-495. [PMID: 35298670 DOI: 10.1007/s00011-022-01561-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE The transient receptor potential vanilloid subtype 1 (TRPV1) channel is considered to play an important regulatory role in the process of pain. The purpose of this study is to observe the change characteristics of TRPV1 channel in MSU-induced gouty arthritis and to find a new target for clinical treatment of gout pain. METHODS Acute gouty arthritis was induced by injection of monosodium urate (MSU) crystals into the ankle joint of mice. The swelling degree was evaluated by measuring the circumference of the ankle joint. Mechanical hyperalgesia was conducted using the electronic von Frey. Calcium fluorescence and TRPV1 current were recorded by applying laser scanning confocal microscope and patch clamp in dorsal root ganglion (DRG) neurons, respectively. RESULTS MSU treatment resulted in significant inflammatory response and mechanical hyperalgesia. The peak swelling degree appeared at 12 h, and the minimum pain threshold appeared at 8 h after MSU treatment. The fluorescence intensity of capsaicin-induced calcium response and TRPV1 current were increased in DRG cells from MSU-treated mice. The number of cells that increased calcium response after MSU treatment was mainly distributed in small-diameter DRG cells. However, the action potential was not significantly changed in small-diameter DRG cells after MSU treatment. CONCLUSIONS These findings identified an important role of TRPV1 in mediating mechanical hyperalgesia in MSU-induced gouty arthritis and further suggest that TRPV1 can be regarded as a potential new target for the clinical treatment of gouty arthritis.
Collapse
Affiliation(s)
- Xiuqi Xu
- Department of Clinical Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Ziqi Yuan
- Department of Clinical Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Shijia Zhang
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221000, China
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research of Southwest Medical University, Luzhou, 646000, China
| | - Guangqin Zhang
- Department of Clinical Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, China.
| |
Collapse
|
23
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:919-929. [DOI: 10.1093/jpp/rgac024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 04/03/2022] [Indexed: 11/14/2022]
|
24
|
Luo Z, Yang F, Hong S, Wang J, Chen B, Li L, Yang J, Yao Y, Yang C, Hu Y, Wang S, Xu T, Wu J. Role of microRNA alternation in the pathogenesis of gouty arthritis. Front Endocrinol (Lausanne) 2022; 13:967769. [PMID: 36034424 PMCID: PMC9402903 DOI: 10.3389/fendo.2022.967769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022] Open
Abstract
Gouty arthritis is a common inflammatory disease. The condition is triggered by a disorder of uric acid metabolism, which causes urate deposition and gout flares. MicroRNAs are a class of conserved small non-coding RNAs that bind to the 3' untranslated region (UTR) of mRNA and regulate the expression of a variety of proteins at the post-transcriptional level. In recent years, attention has been focused on the role of miRNAs in various inflammatory diseases, including gouty arthritis. It is thought that miRNAs may regulate immune function and inflammatory responses, thereby influencing the onset and progression of the disease. This article mainly reviewed the roles of miRNAs in the pathogenesis of gouty arthritis and prospected their potential as diagnostic and prognostic relevant biomarkers and as possible therapeutic targets.
Collapse
Affiliation(s)
- Zhipan Luo
- The First Affifiliated Hospital, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- Anhui Institute of Innovative Drugs, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Fan Yang
- The First Affifiliated Hospital, Anhui Medical University, Hefei, China
| | - Shaocheng Hong
- The First Affifiliated Hospital, Anhui Medical University, Hefei, China
| | - Jianpeng Wang
- The First Affifiliated Hospital, Anhui Medical University, Hefei, China
| | - Bangjie Chen
- The First Affifiliated Hospital, Anhui Medical University, Hefei, China
| | - Liangyun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- Anhui Institute of Innovative Drugs, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Junfa Yang
- Institute of clinical pharmacology, Anhui Medical University, Hefei, China
| | - Yan Yao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- Anhui Institute of Innovative Drugs, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Chenchen Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- Anhui Institute of Innovative Drugs, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ying Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- Anhui Institute of Innovative Drugs, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Shuxian Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- Anhui Institute of Innovative Drugs, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- Anhui Institute of Innovative Drugs, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
- *Correspondence: Tao Xu, ; Jun Wu,
| | - Jun Wu
- Geriatric Department, The First Affifiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Tao Xu, ; Jun Wu,
| |
Collapse
|
25
|
Fu XL, Zhou J, Tang WW, Liu Y, Li ZL, Li P, Chen J. Study on the compatibility effect and active constituents of Atractylodis Rhizoma in Ermiao Wan against Acute Gouty Arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114353. [PMID: 34161798 DOI: 10.1016/j.jep.2021.114353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ermiao Wan (EMW), composed of Atractylodis Rhizoma (AR) and Phellodendri Chinensis Cortex (PC), is a classical traditional Chinese medicine prescription having been used to treat the disease named "Tong Feng", which is described as "ache in bones and joints" with the same symptom of modern disease named acute gouty arthritis for many years in TCM clinical practice. Besides, both PC and AR were considered to be effective in anti-inflammatory according to modern pharmacological research. AIM OF THE STUDY Present study was undertaken to probe the compatibility rationality between the two herbs PC and AR in EMW and the active constituents of AR against acute gouty arthritis (AGA). MATERIALS AND METHODS Rat model of AGA was induced by intra-articular injection of monosodium urate (MSU) crystal suspension, and PC combined with or without different AR extracts were used for AGA treatment. Ankle joint swelling, proinflammatory cytokines in serum and pathological changes of synovium were investigated. Using the developed UHPLC-QQQ-MS method, the plasma concentrations of the primary alkaloids in PC, such as berberine, phellodendrine, magnoflorine, jatrorrhizine, berberrubine, palmatine, and tetrahydropalmatine, in AGA rat were determined, and pharmacokinetics properties were compared following oral administration of PC, PC combined with or without different AR extracts. RESULTS PC, PC combined with AR volatile oil (VO) extract or PC combined with whole AR extract significantly attenuated the ankle joint swelling of AGA rats. Besides, the combination of PC and VO extract of AR showed superior efficacy than other groups in ameliorating ankle joint swelling, reducing the IL-6 expression in serum and improving tissue lesions of ankle joints. Furthermore, it turned out that the VO extract of AR increased the blood exposure level of PC related alkaloids than non-volatile oil (NVO) extract of AR, by comparing the pharmacokinetic results of each group. CONCLUSIONS The VO components of AR were the key compatible materials to combine with PC in EMW for AGA treatment. Moreover, the enhanced anti-AGA activity of PC after combining with VO extract of AR may attribute to the influence of VO on the pharmacokinetics of PC. This study may provide useful information for elucidating the compatibility effects of AR in EMW against AGA.
Collapse
MESH Headings
- Administration, Oral
- Alkaloids/chemistry
- Alkaloids/isolation & purification
- Alkaloids/pharmacokinetics
- Animals
- Anti-Inflammatory Agents/administration & dosage
- Anti-Inflammatory Agents/chemistry
- Anti-Inflammatory Agents/pharmacology
- Arthritis, Gouty/drug therapy
- Arthritis, Gouty/physiopathology
- Atractylodes/chemistry
- Chromatography, High Pressure Liquid/methods
- Disease Models, Animal
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Male
- Mass Spectrometry/methods
- Oils, Volatile/chemistry
- Oils, Volatile/isolation & purification
- Oils, Volatile/pharmacology
- Phellodendron/chemistry
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Xiao-Ling Fu
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jie Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wei-Wei Tang
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zi-Lu Li
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Jun Chen
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
26
|
Anti-inflammatory activity of curcumin-loaded tetrahedral framework nucleic acids on acute gouty arthritis. Bioact Mater 2021; 8:368-380. [PMID: 34541407 PMCID: PMC8429917 DOI: 10.1016/j.bioactmat.2021.06.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 02/05/2023] Open
Abstract
Gouty arthritis is a very familiar inflammatory arthritis. Controlling inflammation is the key to preventing gouty arthritis. However, colchicine, the most highly represented drug used in clinical practice, has strict contraindications owing to some severe side effects. Curcumin (Cur), a natural anti-inflammatory drug, has demonstrated good safety and efficacy. However, the rapid degradation, poor aqueous solubility, and low bioavailability of Cur limit its therapeutic effect. To strengthen the effectiveness and bioavailability of Cur. Cur loaded tetrahedral framework nucleic acids (Cur-TFNAs) were synthesized to deliver Cur. Compared with free Cur, Cur-TFNAs exhibit a preferable drug stability, good biocompatibility (CCK-8 assay), ease of uptake (immunofluorescence), and higher tissue utilization (in vivo biodistribution). Most importantly, Cur-TFNAs present better anti-inflammatory effect than free Cur both in vivo and in vitro experiments through the determination of inflammation-related cytokines expression. Therefore, we believe that Cur-TFNAs have great prospects for the prevention of gout and similar inflammatory diseases. The drug curcumin system based on DNA nanostructures (Cur-TFNAs) were developed to obtain a novel nanomaterial with high water solubility, large encapsulation efficiency, sustained drug release and excellent drug stability. Based on cellular uptake and in vivo drug imaging, Cur-TFNAs improve the retention of Cur in cells and tissues. Cur-TFNAs prevent acute gouty arthritis through mediating anti-inflammatory and antioxidant responses of macrophage.
Collapse
|
27
|
Wang CC, Lu JW, Peng YJ, Lee CH, Lee HS, Chu YH, Huang CJ, Ho YJ, Liu FC, Wu CC. Ameliorative Effects of Cardamonin on Monosodium Urate-Induced Gouty Arthritis through Inhibiting NLRP3 Inflammasome Mediation. ACTA ACUST UNITED AC 2021; 57:medicina57090898. [PMID: 34577821 PMCID: PMC8471485 DOI: 10.3390/medicina57090898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 11/16/2022]
Abstract
Background and Objectives: Gouty arthritis is an acute inflammatory response caused by the precipitation of monosodium urate (MSU) crystals in joints. The triggering of MSU leads to increased production of inflammatory cytokines, such as interleukin-1β, which in turn lead to the formation of macromolecular complexes, referred to as inflammasomes. Thorough characterization of the NLRP3 inflammasome can be used as an indicator of an immune response against harmful stimuli. Cardamonin is a chalcone, mainly found in the seeds of Alpinia katsumadai, and exhibits anti-inflammatory activity by inhibiting the release of pro-inflammatory cytokines in vitro. However, the mechanism by which cardamonin treatment alleviates gouty arthritis has yet to be fully elucidated. Materials and Methods: In vitro or in vivo models were used to study whether cardamonimn inhibited NLRP3 inflammasome activation or suppressed gouty inflammation. Results: In the current study, we determined that most NLRP3 was released passively after MSU stimulation, and this release of NLRP3 promoted caspase-1 activation and IL-1β secretion. Cardamonin was shown to decrease both the activity of caspase-1 and secretion of IL-1β in J774A.1 macrophage cells subjected to MSU stimulation. Cardamonin was also shown to attenuate the production of COX-2 in MSU-stimulated J774A.1 macrophage cells. Finally, cardamonin reduced the thickness of the synovial lining and the infiltration of gouty arthritis in a rat model. Conclusions: Overall, cardamonin significantly attenuated IL-1β secretion, caspase-1 activity, and COX-2 production stimulated by MSU. These findings provide new insights into the molecular mechanisms underlying the effects of cardamonin treatment for gouty arthritis.
Collapse
Affiliation(s)
- Chih-Chien Wang
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
| | - Jeng-Wei Lu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT-Alliance for Research and Technology, Singapore 138602, Singapore;
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-J.P.); (C.-J.H.)
| | - Chian-Her Lee
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
| | - Herng-Sheng Lee
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan;
| | - You-Hsiang Chu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; (Y.-H.C.); (Y.-J.H.)
| | - Chun-Jung Huang
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-J.P.); (C.-J.H.)
| | - Yi-Jung Ho
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; (Y.-H.C.); (Y.-J.H.)
- School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | - Feng-Cheng Liu
- Rheumatologym, Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
| | - Chia-Chun Wu
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
- Correspondence:
| |
Collapse
|
28
|
A brief review on in vivo models for Gouty Arthritis. Metabol Open 2021; 11:100100. [PMID: 34189452 PMCID: PMC8219997 DOI: 10.1016/j.metop.2021.100100] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/29/2021] [Accepted: 06/11/2021] [Indexed: 01/21/2023] Open
Abstract
Gout is more common in men than in women, by a factor of 3.1–10.1. Gout prevalence and incidence have increased in recent decades, with prevalence reaching 11–13% and incidence reaching 0.4% in people over the age of 80. Age-related renal impairment, altered drug distribution, and increased prevalence of comorbidities have significant consequences for safe and effective gout pharmacotherapy. The Discovery of Fruitful in-vivo animal models needs the effective screening of drugs or formulations used in the treatment of gout. In vivo animal models of Gouty arthritis are extensively used to investigate pathogenic mechanisms governing inflammation-driven bone and cartilage damage. Four commonly utilized models include the Potassium oxonate induced hyperuricemic model, MSU crystals induced gouty arthritis animal model, Animal Model of Acute Gouty Arthritis with Hyperuricemia, and Diet-induced hyperuricemia. These offer unique advantages for correlating different aspects of gouty arthritis with human disease. In-vivo animal models served as testing beds for novel biological therapies, including cytokine blockers and small molecule inhibitors of intracellular signaling that have revolutionized gouty arthritis treatment. This review highlights a brief overview of in vivo experimental models for assessment of hypouricemic, anti-inflammatory, as well as renal protective effects of test compounds with some evaluation parameters in detail.
Collapse
|
29
|
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
|
30
|
Tavares ALDF, Reginato A, Neves M, Pradal LDA, Leal TSDS, Ribeiro LDFC, Menolli RA, Bertolini GRF. Analysis of Wistar Rats Submitted to a Gout Model, Treated with Double Cryotherapy Protocol. Ther Hypothermia Temp Manag 2021; 12:30-37. [PMID: 33904783 DOI: 10.1089/ther.2021.0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Gout arthritis commonly affects joint regions by deposition of crystals, promoting functional damage mainly during periods of exacerbation. Cryotherapy is a commonly used resource to contain inflammatory processes, however, its use during a gout crisis is not yet well understood. Therefore, the objective was to evaluate the parameters of Wistar rats submitted to an experimental gout model and treated with dual cryotherapy protocol. Twenty-one male Wistar rats were used, separated into three groups: control group (CG), lesion group (LG), and lesion + cryotherapy group (LCG). Gout model induction was through intra-articular injection, with urate crystal solution, in the right knee and cryoimmersion treatment was performed for 20 minutes at a temperature of 5° ± 2°C. Seven evaluations and two treatment moments were performed, and the following parameters were analyzed: joint edema, grip strength, joint disability, motor function, and leukocyte migration through synovial lavage. In the statistical analysis we used SPSS 20.0 with Generalized Linear Models, with least significant difference posttest, always with 5% significance level. The treatment reduced edema, promoted strength recovery, and was effective in reducing total leukocytes in the synovial fluid. No difference was observed between the injured groups for joint disability and motor function. Cryotherapy promoted edema reduction and increased pelvic limb grip strength in Wistar rats during the acute period.
Collapse
Affiliation(s)
| | - Aline Reginato
- Universidade Estadual do Oeste do Paraná-Unioeste, Cascavel, Brazil
| | - Morgana Neves
- Universidade Estadual do Oeste do Paraná-Unioeste, Cascavel, Brazil
| | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
The Role of Oxidative Stress in Hyperuricemia and Xanthine Oxidoreductase (XOR) Inhibitors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1470380. [PMID: 33854690 PMCID: PMC8019370 DOI: 10.1155/2021/1470380] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022]
Abstract
Uric acid is the end product of purine metabolism in humans. Hyperuricemia is a metabolic disease caused by the increased formation or reduced excretion of serum uric acid (SUA). Alterations in SUA homeostasis have been linked to a number of diseases, and hyperuricemia is the major etiologic factor of gout and has been correlated with metabolic syndrome, cardiovascular disease, diabetes, hypertension, and renal disease. Oxidative stress is usually defined as an imbalance between free radicals and antioxidants in our body and is considered to be one of the main causes of cell damage and the development of disease. Studies have demonstrated that hyperuricemia is closely related to the generation of reactive oxygen species (ROS). In the human body, xanthine oxidoreductase (XOR) catalyzes the oxidative hydroxylation of hypoxanthine to xanthine to uric acid, with the accompanying production of ROS. Therefore, XOR is considered a drug target for the treatment of hyperuricemia and gout. In this review, we discuss the mechanisms of uric acid transport and the development of hyperuricemia, emphasizing the role of oxidative stress in the occurrence and development of hyperuricemia. We also summarize recent advances and new discoveries in XOR inhibitors.
Collapse
|
33
|
Cichoric Acid Ameliorates Monosodium Urate-Induced Inflammatory Response by Reducing NLRP3 Inflammasome Activation via Inhibition of NF- kB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8868527. [PMID: 33505510 PMCID: PMC7808822 DOI: 10.1155/2021/8868527] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/24/2020] [Accepted: 12/26/2020] [Indexed: 12/30/2022]
Abstract
Gouty arthritis is characterized by the deposition of monosodium urate (MSU) within synovial joints and tissues due to increased urate concentrations. Here, we elucidated the role of the natural compound cichoric acid (CA) on the MSU crystal-stimulated inflammatory response. The THP-1-derived macrophages (THP-Ms) were pretreated with CA and then stimulated with MSU suspensions. The protein levels of p65 and IκBα, the activation of the NF-κB signaling pathway by measuring the expression of its downstream inflammatory cytokines, and the activity of NLRP3 inflammasome were measured by western blotting and ELISA. CA treatment markedly inhibited the degradation of IκBα and the activation of NF-κB signaling pathway and reduced the levels of its downstream inflammatory genes such as IL-1β, TNF-α, COX-2, and PGE2 in the MSU-stimulated THP-M cells. Therefore, we infer that CA effectively alleviated MSU-induced inflammation by suppressing the degradation of IκBα, thereby reducing the activation of the NF-κB signaling pathway and the NLRP3 inflammasome. These results suggest that CA could be a novel therapeutic strategy in averting acute episodes of gout.
Collapse
|
34
|
Zhang JZ, Chen XY, Wu YJ, Li LM, Huang L, Yin QZ, Luo P, Liu Y. Identification of Active Compounds From Yi Nationality Herbal Formula Wosi Influencing COX-2 and VCAM-1 Signaling. Front Pharmacol 2021; 11:568585. [PMID: 33442381 PMCID: PMC7797783 DOI: 10.3389/fphar.2020.568585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
The Yi nationality herbal formula Wosi is used in China as a folk medicine to treat arthritis and related diseases. Despite its widespread use, the active ingredients, and pharmacological mechanisms are not performed. This is the first time to identify the active compounds from Wosi with the aim at providing the potential effect of Wosi and exploring its underlying anti-inflammatory mechanism in monosodium urate crystals (MSU)-induced arthritis rats. In this study, anti-hyperuricemia effect was assessed by reducing the serum uric acid levels and increasing uric acid excretion in the urine for the hyperuricemia rat model. Wosi significantly suppressed the degree of joint swelling and improved the symptoms of inflammation induced by MSU crystals. The inhibition of IL-2, IL-1β, IFN-γ, and IL-6 secretion and IL-10 increase in the serum were also observed. This study also focuses on the screening of the main compounds from Wosi against cyclooxygenase for anti-inflammatory properties using molecular docking. The result showed 3-O-[α-L-pyran rhamnose(1-3)-β-D-pyran glucuronic acid]- oleanolic acid, 3-O-(β-D-pyran glucuronic acid)-oleanolic acid-28-O-β-D-pyran glucoside, and 3-O-[α-L-pyran rhamnose(1-3)-β-D-pyran glucuronic acid]-oleanolic acid-28-O-β-D-pyran glucoside with a higher binding affinity for COX-2 than COX-1 which indicated relatively higher interaction than COX-1. The preferential selectivity toward inhibiting COX-2 enzyme over COX-1 of three compounds from Wosi were evaluated using in-vitro cyclooxygenases 1 and 2 (COX-1/2) inhibition assays. Meanwhile, the down-regulated protein expression of COX-2 and VCAM-1 in synovial tissue sections from ankle joints of experiments rats were confirmed by immunohistochemistry analysis after the Wosi treatment. In conclusion, three oleanolic acid glycosides were implied as mainly efficient compounds in Yi nationality herbal formula Wosi for arthritis therapy via selectively influencing COX-2 and VCAM-1 signaling.
Collapse
Affiliation(s)
- Ji-Zhong Zhang
- Institute of Ethnic Medicine, Southwest Minzu University, Chengdu, China.,Sichuan Provincial Qiang-Yi Medicinal Resources Protection and Utilization Technology and Engineering Laboratory, Chengdu, China
| | - Xiao-Yi Chen
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - You-Jiao Wu
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Li-Min Li
- Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Li Huang
- Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | | | - Pei Luo
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Yuan Liu
- Institute of Ethnic Medicine, Southwest Minzu University, Chengdu, China.,Sichuan Provincial Qiang-Yi Medicinal Resources Protection and Utilization Technology and Engineering Laboratory, Chengdu, China
| |
Collapse
|
35
|
Andrographolide inhibits IL-1β release in bone marrow-derived macrophages and monocyte infiltration in mouse knee joints induced by monosodium urate. Toxicol Appl Pharmacol 2020; 410:115341. [PMID: 33242555 DOI: 10.1016/j.taap.2020.115341] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 01/05/2023]
Abstract
Andrographolide (AND) is the major diterpenoid in A. paniculata with wide clinical application and has been shown to be a potent anti-inflammatory agent. Gout is the leading inflammatory disease of the joints, and the deposition of urate in the articular cavity attracts immune cells that release inflammatory cytokines. Monosodium urate (MSU) is known to be one of the activators of the NLRP3 (NLR family pyrin domain containing 3) inflammasome. After activation, the NLRP3 inflammasome releases interleukin-1β (IL-1β), which causes the development of many inflammatory diseases. The aim of the present study was to investigate whether AND attenuates the release of IL-1β mediated by the NLRP3 inflammasome. The effects of AND were studied in bone marrow-derived macrophages (BMDMs) treated with lipopolysaccharide (LPS) and MSU and in mice with MSU-induced joint inflammation. AND suppressed MSU phagocytosis dose-dependently and markedly inhibited LPS- and MSU-induced IL-1β release in BMDMs. Moreover, AND pretreatment inhibited the LPS-induced NLRP3 inflammasome priming stage by inhibiting the IKK/NFκB signaling pathway, which resulted in decreased protein expression of NLRP3 and proIL-1β. AND induced HO-1 protein expression in a dose-dependent manner and attenuated MSU-induced ROS generation. Silencing HO-1 mitigated AND inhibition of LPS/MSU-induced IL-1β release in J774A.1 cells. In addition, AND decreased MSU-mediated ASC binding to NLRP3. Oral administration of AND attenuated MSU-induced monocyte infiltration in mouse knee joints. These results suggest that the working mechanisms by which AND down-regulates MSU-induced joint inflammation might be via HO-1 induction and attenuation of ROS-mediated NLRP3 inflammasome assembly and subsequent IL-1β release.
Collapse
|
36
|
Ribeiro SA, Lopes C. The therapeutic potential of colchicine in the complications of COVID19. Could the immunometabolic properties of an old and cheap drug help? Metabol Open 2020; 7:100045. [PMID: 32808940 PMCID: PMC7373059 DOI: 10.1016/j.metop.2020.100045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/30/2022] Open
Abstract
The present study analyzes the importance of the inflammasome that involves the NLRP3 complex in the state of hypercytokinemia observed in patients with COVID-19, significantly increasing IL-1β, IL18, IL-6, and TNF. Unfortunately, improving the immune response can sometimes worsen the outcome of the disease. Studies show that colchicine, among other actions, inhibits the assembly of NLRP3 complex that is responsible for generating the active form of Caspase-1 that will convert Pro-IL-1β and Pro-IL-18 into their active forms. We suggest using colchicine, a class of drugs with low-cost, extensively tested, well-tolerated medicine as a complementary treatment for patients with COVID-19, in early stages of the disease based on knowledge of its immunomodulatory properties.
Collapse
Affiliation(s)
| | - Cassio Lopes
- Hospital John Paul II, Intensive Medical Assistance, AMI, Porto Velho, RO, Brazil
| |
Collapse
|
37
|
Zhou M, Ze K, Wang Y, Li X, Hua L, Lu Y, Chen X, Ding X, Chen S, Ru Y, Zhang M, Li B. Huzhang Tongfeng Granule Improves Monosodium Urate-Induced Inflammation of Gouty Arthritis Rat Model by Downregulation of Cyr61 and Related Cytokines. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:9238797. [PMID: 32419834 PMCID: PMC7206887 DOI: 10.1155/2020/9238797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/18/2020] [Accepted: 04/06/2020] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Gouty arthritis (GA) is a noninfectious inflammatory disease characterized by self-limited and severe pain. Huzhang Tongfeng granule is one of the most effective traditional Chinese medicines in the treatment of acute GA. However, its effects on the inflammatory factors in the process of acute gout inflammation remain unknown. In the present study, we aimed to evaluate the effect of Huzhang Tongfeng granule on the expressions of Cyr61 and related inflammatory factors in both experimental gout models in vivo and in vitro. METHODS Huzhang Tongfeng granule was provided by the pharmaceutical preparation room of Yueyang Hospital of Integrated Traditional Chinese and Western Medicine. The expressions of Cyr61, IL-1β, TNF-α, and IL-6 in monosodium urate- (MSU-) induced rat models and fibroblast-like synoviocytes (FLSs) were determined by RT-PCR, Western blotting analysis, ELISA, immunohistochemistry, and hematoxylin and eosin staining. RESULTS Huzhang Tongfeng granule could downregulate the expressions of IL-1β, TNF-α, and IL-6 to some extent by inhibiting the expression of Cyr61. CONCLUSIONS Collectively, our findings indicated that Cyr61 was highly expressed in rat models of gout. By inhibiting the expression of Cyr61, Huzhang Tongfeng granule could partially attenuate the inflammation induced by MSU crystal.
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
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kan Ze
- 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
| | - Yifei 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
| | - 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
| | - Liang Hua
- 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
| | - Yi Lu
- 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
| | - Xi 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
| | - Xiaojie Ding
- 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
| | - Siting 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
| | - Yi Ru
- 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
- 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
|
38
|
Activation Status of NLRP3 Inflammasome in Peripheral Blood Mononuclear Cells From Patients With Gout Flare. J Clin Rheumatol 2020; 26:S208-S212. [PMID: 32332275 DOI: 10.1097/rhu.0000000000001394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND/OBJECTIVE Although gout flares are featured by systemic signs of inflammation, cellular sources of inflammatory mediators are not yet properly characterized. Our objective was to evaluate serum levels and gene expression in peripheral blood mononuclear cells (PBMCs) of several molecules associated with the activation of NLRP3 inflammasome. METHODS Fifteen patients with gout flare and 15 individuals with asymptomatic hyperuricemia were cross-sectionally studied. Serum levels of interleukin 1β (IL-1β), IL-18, monocyte chemoattractant protein 1/chemokine (C-C motif) ligand 2 (CCL2), and vascular cell adhesion molecule 1 were measured as a reflection of systemic inflammation, whereas the expression of NLRP3, CASP1, IL18, and CCL2 genes was measured to assess the inflammatory characteristics of PBMCs. RESULTS Serum levels of IL-1β (1.27 [0.07-1.99] pg/mL vs. 0 [0-0.82] pg/mL, p = 0.032) and vascular cell adhesion molecule 1 (606 [435-748] pg/mL vs. 349 [305-422] pg/mL, p = 0.014) were significantly higher in patients with gout flare than in individuals with asymptomatic hyperuricemia, whereas differences in IL-18 and monocyte chemoattractant protein 1/CCL2 were not found. Notably, no differences were observed in the expression of NLRP3, CASP1, IL18, or CCL2 in PBMCs from individuals of one or another group. CONCLUSIONS Systemic inflammation during gout flares does not appear to be associated with NLRP3 inflammasome activation in PBMCs, suggesting that it may represent the systemic spread of local (synovial) inflammation to monosodium urate crystals, which provides a rationale for redirecting anti-inflammatory therapy from a systemic approach to one centered on the inflamed joint.
Collapse
|
39
|
Huang WS, Lin CL, Tsai CH, Chang KH. Association of gout with CAD and effect of antigout therapy on CVD risk among gout patients. J Investig Med 2020; 68:972-979. [PMID: 32098832 PMCID: PMC7306869 DOI: 10.1136/jim-2019-001140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2020] [Indexed: 12/20/2022]
Abstract
Hyperuricemia has been identified as an independent risk factor for coronary artery disease (CAD), with a dose-response association. In this study, we explored the causal association between gout and antigout medication and the risk of incidental CAD. We sampled data from the National Health Insurance Research Database and recruited 37,091 patients as the gout cohort, and 37,091 controls. Our primary endpoint was the diagnosis of CAD during follow-up. The overall study population was followed up until CAD diagnosis, withdrawal from the National Health Insurance program, or the end of the study. Cox proportional hazards regression models were used to examine the effect of gout on the risk of CAD, represented by the HR with the 95% CI. Patients with gout were at greater risk of CAD, compared with those without gout: HR=1.49 after adjusting for potential confounders. Non-steroidal anti-inflammatory drugs and prednisolone use was associated with a reduced risk of CAD: HR=0.63 and 0.50, respectively. Patients with gout, treated with antigout medication, exhibited a reduced risk of CAD compared with non-gout patients. Among patients with gout, those on antigout therapy had 32% lower risk compared with those not on antigout therapy: adjusted HR=0.68, 95% CI 0.63 to 0.73. Gout increases the risk of CAD, and the use of antigout medication reduces CAD risk. These results indicate that gout or hyperuricemia is a modifiable risk factor for CAD.
Collapse
Affiliation(s)
- Wei-Shih Huang
- Department of Neurology, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Chon-Haw Tsai
- Department of Neurology, China Medical University Hospital and College of Medicine, China Medical University, Taichung, Taiwan
| | - Kuang-Hsi Chang
- Department of Medical Research, Tungs' Taichung Metroharbor Hospital, Taichung City, Taiwan .,Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,General Education Center, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| |
Collapse
|
40
|
Luo Y, Wang L, Peng A, Liu JY. Metabolic profiling of human plasma reveals the activation of 5-lipoxygenase in the acute attack of gouty arthritis. Rheumatology (Oxford) 2019; 58:345-351. [PMID: 30247644 DOI: 10.1093/rheumatology/key284] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Indexed: 12/27/2022] Open
Abstract
Objective Monosodium urate-induced inflammation plays a vital role in acute gout (AG). Inflammation is a multi-stage process involved in the acute release of arachidonic acid and its metabolites. However, the function of the metabolism of arachidonic acid and other polyunsaturated fatty acids in AG is not well understood. This study aimed to investigate the modification of polyunsaturated fatty acid metabolism by AG. Methods Plasma samples from patients with an AG attack (n = 26) and gender-matched healthy controls (n = 26) were analysed by metabolic profiling of polyunsaturated fatty acids. The findings were further validated with a second cohort (n = 20 each group). The associated mechanisms were investigated in whole blood cells from the second cohort and neutrophils in vitro. Results Plasma metabolic profiling revealed a significant increase in leukotriene B4 (LTB4) for AG patients in both cohorts. The increase in plasma LTB4 was accounted for by the dynamic balance between the activation of 5-lipoxygenase and CYP4F3, the former mediating the biosynthesis of LTB4 and the latter mediating its metabolism. This was supported by significantly increased transcriptional levels of 5-lipoxygenase and CYP4F3 in whole blood cells from AG patients compared with those of controls, and the uric acid-caused dose-relevant and time-dependent activation of 5-lipoxygenase and CYP4F3 at the transcriptional and molecular levels in vitro. Conclusion Increased LTB4 in AG patients is mainly due to activation of 5-lipoxygenase. 5-Lipoxygenase inhibition may be of therapeutic value clinically.
Collapse
Affiliation(s)
- Ying Luo
- Center for Nephrology & Metabolomics, Division of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ling Wang
- Center for Nephrology & Metabolomics, Division of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ai Peng
- Center for Nephrology & Metabolomics, Division of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun-Yan Liu
- Center for Nephrology & Metabolomics, Division of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Chhana A, Pool B, Wei Y, Choi A, Gao R, Munro J, Cornish J, Dalbeth N. Human Cartilage Homogenates Influence the Crystallization of Monosodium Urate and Inflammatory Response to Monosodium Urate Crystals: A Potential Link Between Osteoarthritis and Gout. Arthritis Rheumatol 2019; 71:2090-2099. [DOI: 10.1002/art.41038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/05/2019] [Indexed: 01/11/2023]
Affiliation(s)
| | | | | | - Ally Choi
- University of Auckland Auckland New Zealand
| | - Ryan Gao
- University of Auckland Auckland New Zealand
| | | | | | | |
Collapse
|
42
|
Löffler C, Sattler H, Löffler U, Krämer BK, Bergner R. Size matters: observations regarding the sonographic double contour sign in different joint sizes in acute gouty arthritis. Z Rheumatol 2019. [PMID: 29536155 DOI: 10.1007/s00393-018-0425-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE In distinguishing urate arthritis (UA) from non-crystal-related arthritides, joint sonography including the detection of the double contour sign (DCS) and hypervascularization using power Doppler ultrasound (PDUS) is an important step in the diagnostic process. But are these sonographic features equally reliable in every accessible joint under real-life conditions? METHODS We retrospectively analyzed 362 patients with acute arthritis and evaluated the DCS and the degree of PDUS hypervascularization in patients with gout and in those with arthritis other than urate arthritis (non-UA). We classified all joints into the groups small, medium, and large. Sensitivities, specificities, positive and negative predictive values (PPV/NPV), and a binary regression model were calculated. We also evaluated the influence of serum uric acid levels (SUA) on the presence of a DCS in each joint category. RESULTS Sensitivity of the DCS in gout was 72.5% in the entire cohort, 66.0% in large, 78.8% in medium, and 72.3% in small joints. In wrist joints the DCS sensitivity maxed at 83.3%, with a specificity of 81.8%. The lowest rates of DCS sensitivity were found in gout patients with elbow joint involvement (42.9%). In all joints except metatarsophalangeal joint 1 (MTP-1), the incidence of a DCS increased by the increment of SUA levels above 7.5 mg/dl (p < 0.001). PDUS signals were most commonly found in medium and small joints and were only scarce in large joints, independent of the underlying diagnosis. CONCLUSIONS In our study we detected different rates of accuracy regarding DCS and PDUS in patients with acute arthritis. The best results were seen in medium-size joints, especially wrists.
Collapse
Affiliation(s)
- C Löffler
- Department of Nephrology, Endocrinology, Rheumatology, University Hospital Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - H Sattler
- Department of Oncology, Rheumatology, Nephrology, Klinikum Ludwigshafen, Bremserstr. 79, 67063, Ludwigshafen, Germany
| | - U Löffler
- Psychotherapy Clinic, Institute of Psychology, University of Heidelberg, Hauptstr. 49-51, 69117, Heidelberg, Germany
| | - B K Krämer
- Department of Nephrology, Endocrinology, Rheumatology, University Hospital Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - R Bergner
- Department of Oncology, Rheumatology, Nephrology, Klinikum Ludwigshafen, Bremserstr. 79, 67063, Ludwigshafen, Germany
| |
Collapse
|
43
|
Azevedo VF, Kos IA, Vargas-Santos AB, da Rocha Castelar Pinheiro G, Dos Santos Paiva E. Benzbromarone in the treatment of gout. Adv Rheumatol 2019; 59:37. [PMID: 31391099 DOI: 10.1186/s42358-019-0080-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 07/19/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Benzbromarone is a uricosuric drug that has been used in the treatment of gout over the last 30 years. Due to its potent inhibition of the dominant apical (luminal) urate exchanger in the human proximal tubule URAT1, it reduces the urate reabsorption, diminishing serum urate levels and therefore preventing gout flares. Through several clinical trials, Benzbromarone has been proved effective and safe, inclusive in patients with chronic kidney disease and as combination therapy with allopurinol. Due to hepatotoxicity reports, it was withdrawn from the European market by the manufacturer, however many authors have questioned the product's withdrawal due to a lack of clinical evidence in order to support its hepatotoxicity. Benzbromarone is still available in several European countries, New Zealand, Brazil and several other countries. Despite the product's marketing over more than 20 years after the first hepatotoxicity reports, we have found only five reports in our literature search, and no prospective or retrospective study correlating hepatotoxicity with benzbromarone use. SHORT CONCLUSION Benzbromarone is a safe and effective molecule for the treatment of gout. However, due to in vitro and in vivo data related to hepatotoxicity, it is prudent to prescribe it with some caution, especially for patients with an already known liver condition.
Collapse
Affiliation(s)
- Valderilio Feijó Azevedo
- Universidade Federal do Paraná, Rua General Carneiro 181, Centro, Curitiba, Paraná, Brazil. .,Edumed Educação em Saúde, Rua Bispo Dom José, 2495, Curitiba, Paraná, Brazil.
| | - Igor Age Kos
- Edumed Educação em Saúde, Rua Bispo Dom José, 2495, Curitiba, Paraná, Brazil
| | | | | | | |
Collapse
|
44
|
Zhou Z, Dong Y, Zhou H, Liu J, Zhao W. MiR-143-3p directly targets GLUT9 to reduce uric acid reabsorption and inflammatory response of renal tubular epithelial cells. Biochem Biophys Res Commun 2019; 517:413-420. [PMID: 31378369 DOI: 10.1016/j.bbrc.2019.07.114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/29/2019] [Indexed: 11/28/2022]
Abstract
GLUT9 is generally considered to be associated with the uric acid transport, which plays an important role in the regulation of serum uric acid level. In this study, the expression level of miR-143-3p was significantly decreased in hyperuricemia mice model group compared with the normal control by miRNA microarray, the same results were confirmed in the hyperuricemia patients and the healthy control group. It is predicted that GLUT9 may be the target gene of miR-143-3p by target scan and other net-software. GLUT9 as the downstream target gene of miR-143-3p was determinated by fluorescence enzyme activity assay. Western blotting and qRT-PCR indicated that the expression of GLUT9 in human renal tubular epithelial cells transfected with miR-143-3p mimics was significantly reduced. Meanwhile inflammatory factors IL-1β and MCP-1 significantly decreased. In conclusion, miR-143-3p can reduce uric acid reabsorption by inhibiting its downstream target gene GLUT9.
Collapse
Affiliation(s)
- Zhichao Zhou
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, 300134, China
| | - Ya Dong
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, 300134, China
| | - Hanchi Zhou
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, 300134, China
| | - Jia Liu
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, 300134, China
| | - Wei Zhao
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, 300134, China.
| |
Collapse
|
45
|
Decreased Expression of CD14 in MSU-Mediated Inflammation May Be Associated with Spontaneous Remission of Acute Gout. J Immunol Res 2019; 2019:7143241. [PMID: 31312662 PMCID: PMC6595337 DOI: 10.1155/2019/7143241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/07/2019] [Indexed: 12/27/2022] Open
Abstract
Gout is a common metabolic disease in humans, and it is due to persistently elevated levels of uric acid in the blood. At high levels, uric acid crystallizes and the crystals deposit in joints and surrounding tissues, resulting in an attack of gout. Interestingly, the gout attack can spontaneously resolve within a few days. However, the self-limited mechanism of gout remains elusive. It has been demonstrated that CD14 plays an important role in self-remission of gout. In this study, we found that the proportion of CD14-positive PBMCs was decreased in gout patients when compared with healthy controls and the serum sCD14 level was also considerably decreased in gout patients in comparison to healthy controls. In addition, sCD14 levels were positively correlated with CRP levels. Furthermore, the effect of MSU on the levels of CD14 in healthy volunteer's PBMC was explored in in vitro experiment. The results showed that CD14 expression on macrophage and sCD14 levels in the culture supernatants were significantly decreased after MSU treatment. However, there was no significance in the levels of membrane CD14 and sCD14 in healthy volunteer's PBMC stimulated by LPS. Taken together, these results suggest that CD14 might play an important role in self-remission of gout.
Collapse
|
46
|
Janssen CA, Oude Voshaar MAH, Vonkeman HE, Jansen TLTA, Janssen M, Kok MR, Radovits B, van Durme C, Baan H, van de Laar MAFJ. Anakinra for the treatment of acute gout flares: a randomized, double-blind, placebo-controlled, active-comparator, non-inferiority trial. Rheumatology (Oxford) 2019; 58:5270863. [PMID: 30602035 DOI: 10.1093/rheumatology/key402] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES To evaluate the efficacy and safety of anakinra in treating acute gout flares in a randomized, double-blind, placebo-controlled, active comparator, non-inferiority (NI) trial. METHODS Patients with a crystal-proven acute gout flare were randomized (1: 1) to treatment with anakinra or treatment as usual (free choice: either colchicine, naproxen or prednisone). The primary end point was the change in pain between baseline and the averaged pain score on days 2-4 measured on a five-point rating scale. NI of anakinra would be established if the upper bound of the 95% CI of the numeric difference in changed pain scores between treatment groups did not exceed the NI limit of 0.4 in favour of treatment as usual, in the per-protocol (PP) and intention-to-treat (ITT) populations, assessed in an analysis of covariance model. Secondary outcomes included safety assessments, improvement in pain, swelling, tenderness and treatment response after 5 days, assessed using linear mixed models and binary logistic regression models. RESULTS Forty-three patients received anakinra and 45 treatment as usual. Anakinra was non-inferior (mean difference; 95% CI) to treatment as usual in both the PP (-0.13; -0.44, 0.18) and ITT (-0.18; -0.44, 0.08) populations. No unexpected or uncommon (serious) adverse events were observed in either treatment arm. Analyses of secondary outcomes showed that patients in both groups reported similar significant reductions in their gout symptoms. CONCLUSION Efficacy of anakinra was shown to be non-inferior to treatment as usual for the treatment of acute gout flares, suggesting that anakinra is an effective treatment alternative for acute gout flares. TRIAL REGISTRATION Het Nederlands Trial Register, www.trialregister.nl, NTR5234.
Collapse
Affiliation(s)
- Carly A Janssen
- Arthritis Center Twente, Department of Psychology, Health and Technology, University of Twente, The Netherlands
| | - Martijn A H Oude Voshaar
- Arthritis Center Twente, Department of Psychology, Health and Technology, University of Twente, The Netherlands
| | - Harald E Vonkeman
- Arthritis Center Twente, Department of Psychology, Health and Technology, University of Twente, The Netherlands
- Department of Rheumatology and Clinical Immunology, Medisch Spectrum Twente, Ensche, The Netherlands
| | - Tim L Th A Jansen
- Department of Rheumatology, VieCuri Medical Center, Venlo, The Netherlands
| | - Matthijs Janssen
- Department of Rheumatology, VieCuri Medical Center, Venlo, The Netherlands
- Department of Rheumatology, Rijnstate Hospital, Arnhem, The Netherlands
| | - Marc R Kok
- Department of Rheumatology and Clinical Immunology, Maasstad Hospital, Rotterdam, The Netherlands
| | - Bea Radovits
- Department of Rheumatology, Bernhoven Hospital, Uden, The Netherlands
| | - Caroline van Durme
- Department of Rheumatology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Hetty Baan
- Department of Rheumatology and Clinical Immunology, ZiekenhuisGroep Twente, Almelo, The Netherlands
| | - Mart A F J van de Laar
- Arthritis Center Twente, Department of Psychology, Health and Technology, University of Twente, The Netherlands
- Department of Rheumatology and Clinical Immunology, Medisch Spectrum Twente, Ensche, The Netherlands
| |
Collapse
|
47
|
Kadiyoran C, Zengin O, Cizmecioglu HA, Tufan A, Kucuksahin O, Cure MC, Cure E, Kucuk A, Ozturk MA. Monocyte to Lymphocyte Ratio, Neutrophil to Lymphocyte Ratio, and Red Cell Distribution Width are the Associates with Gouty Arthritis. ACTA MEDICA (HRADEC KRALOVE) 2019; 62:99-104. [PMID: 31663502 DOI: 10.14712/18059694.2019.132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neutrophils, monocytes, and macrophages activations are associated with a gout attack. Monocyte to lymphocyte ratio (MLR), neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), red cell distribution width (RDW), and mean platelet volume (MPV) are well-known inflammation markers. In this study, we aimed to investigate whether they could be a predictive marker to the gout attack. MATERIAL AND METHODS A hundred and ten gout patients (male/female, 86/24) and 90 (male/female, 64/26) age-, gender-, and body mass index-matched volunteer controls were included in the study. Blood samples were obtained in the intercritical and attack period of the patients. Hemogram, serum uric acid (SUA), C-reactive protein (CRP), and erythrocyte sedimentation rate (ESR) values were studied. RESULTS In the attack period NLR (p < 0.001), PLR (p < 0.05), MLR (p < 0.001), RDW (p < 0.05), MPV (p < 0.05), ESR (p < 0.001), CRP (p < 0.001) and SUA (p < 0.001) values were significantly higher than intercritical period values. According to the results of regression analysis; There was an independent strong relationship between the gout attack and SUA, (Beta [β] = 0.352, p < 0.001), ESR (β = 0.329, p < 0.001), CRP (β = 0.286, p < 0.001), MLR (β = 0.126, p < 0.001), RDW (β = 0.100, p = 0.003) and NLR (β = 0.082, p = 0.014). CONCLUSIONS MLR, RDW, and NLR may be a strong predictive marker for a gout attack. MPV and PLR values in the gout attack may be associated with systemic inflammation.
Collapse
Affiliation(s)
- Cengiz Kadiyoran
- Department of Radiology, Necmettin Erbakan University, Konya, Turkey
| | - Orhan Zengin
- Department of Rheumatology, Adiyaman Training and Research Hospital, Adiyaman, Turkey
| | | | - Abdurrahman Tufan
- Division of Rheumatology, Department of Internal Medicine, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Orhan Kucuksahin
- Division of Rheumatology, Department of Internal Medicine, Ankara Liv Hospital, Kavaklidere, Ankara, Turkey
| | | | - Erkan Cure
- Department of Internal Medicine, Ota&Jinemed Hospital, Istanbul, Turkey.
| | - Adem Kucuk
- Division of Rheumatology, Department of Internal Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Mehmet Akif Ozturk
- Division of Rheumatology, Department of Internal Medicine, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
48
|
Lin TM, Chi JE, Chang CC, Kang YN. Do etoricoxib and indometacin have similar effects and safety for gouty arthritis? A meta-analysis of randomized controlled trials. J Pain Res 2018; 12:83-91. [PMID: 30588082 PMCID: PMC6305166 DOI: 10.2147/jpr.s186004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Gout, a common medical condition that causes pain, can be treated by painkillers and anti-inflammatories. Indometacin and etoricoxib are two such drugs. However, no synthesized evidence exists comparing etoricoxib with indometacin in treating patients with gout. Methods We searched PubMed, Embase, Ovid MEDLINE, Web of Science, ScienceDirect, and the Cochrane Library without restrictions on language or publication date for potential randomized clinical trials comparing etoricoxib with indometacin for gout. The meta-analysis was conducted using a random-effects model. Results Search results yielded 313 references from six electronic databases, four of which met the eligibility criteria. These four were randomized clinical trials, and they involved a total of 609 patients with gouty arthritis. No significant differences were observed in pain score change, tenderness, or swelling between etoricoxib and indometacin; the mean differences were −0.05 (95% CI, −0.21 to 0.10), −0.06 (95% CI, −0.18 to 0.05), and −0.04 (95% CI, −0.17 to 0.09). However, the pooled data revealed that significantly fewer overall adverse events occurred in the etoricoxib group (n=105, 33.5%) than in the indometacin group (n=130, 44.1%) and the risk ratio was 0.77 (95% CI, 0.62–0.94). Conclusion Our meta-analysis revealed that etoricoxib and indometacin have similar effects on pain relief. However, etoricoxib has a significantly lower risk of adverse events than does indometacin, especially digestive system-related adverse events.
Collapse
Affiliation(s)
- Tzu-Min Lin
- Center for Evidence-Based Medicine, Department of Education, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China, .,Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China
| | - Jia-En Chi
- Center for Evidence-Based Medicine, Department of Education, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China, .,School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Chi-Ching Chang
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China.,Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Yi-No Kang
- Center for Evidence-Based Medicine, Department of Education, Taipei Medical University Hospital, Taipei, Taiwan, Republic of China,
| |
Collapse
|
49
|
Estevez‐Garcia IO, Gallegos‐Nava S, Vera‐Pérez E, Silveira LH, Ventura‐Ríos L, Vancini G, Hernández‐Díaz C, Sánchez‐Muñoz F, Ballinas‐Verdugo MA, Gutierrez M, Pineda C, Rodriguez‐Henriquez P, Castillo‐Martínez D, Amezcua‐Guerra LM. Levels of Cytokines and Micro
RNA
s in Individuals With Asymptomatic Hyperuricemia and Ultrasonographic Findings of Gout: A Bench‐to‐Bedside Approach. Arthritis Care Res (Hoboken) 2018; 70:1814-1821. [DOI: 10.1002/acr.23549] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 02/13/2018] [Indexed: 01/25/2023]
Affiliation(s)
| | - Selma Gallegos‐Nava
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Hospital General Dr. Darío Fernández Fierro, and Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado Mexico CityMexico
| | - Erika Vera‐Pérez
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Mexico CityMexico
| | - Luis H. Silveira
- Instituto Nacional de Cardiología Ignacio Chávez Mexico CityMexico
| | - Lucio Ventura‐Ríos
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Mexico CityMexico
| | - Gonzalo Vancini
- Instituto Nacional de Cardiología Ignacio Chávez Mexico CityMexico
| | | | | | | | - Marwin Gutierrez
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Mexico CityMexico
| | - Carlos Pineda
- Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Mexico CityMexico
| | | | - Diana Castillo‐Martínez
- Hospital General de Zona 32 and Instituto Mexicano del Seguro Social Coyoacán, Mexico CityMexico
| | - Luis M. Amezcua‐Guerra
- Instituto Nacional de Cardiología Ignacio Chávez Universidad Autónoma Metropolitana−Xochimilco, and Unidad de Investigación Traslacional UNAM/INC Mexico City Mexico
| |
Collapse
|
50
|
Galindo T, Reyna J, Weyer A. Evidence for Transient Receptor Potential (TRP) Channel Contribution to Arthritis Pain and Pathogenesis. Pharmaceuticals (Basel) 2018; 11:E105. [PMID: 30326593 PMCID: PMC6315622 DOI: 10.3390/ph11040105] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023] Open
Abstract
Based on clinical and preclinical evidence, Transient Receptor Potential (TRP) channels have emerged as potential drug targets for the treatment of osteoarthritis, rheumatoid arthritis, and gout. This review summarizes the relevant data supporting a role for various TRP channels in arthritis pain and pathogenesis, as well as the current state of pharmacological efforts to ameliorate arthritis symptoms in patient populations.
Collapse
Affiliation(s)
- Tabitha Galindo
- School of Physical Therapy and Athletic Training, Pacific University, Hillsboro, OR 97116, USA.
| | - Jose Reyna
- School of Physical Therapy and Athletic Training, Pacific University, Hillsboro, OR 97116, USA.
| | - Andy Weyer
- Biological Sciences Department, City College of San Francisco, San Francisco, CA 94112, USA.
| |
Collapse
|