1
|
Chenchula S, Ghanta MK, Alhammadi M, Mohammed A, Anitha K, Nuthalapati P, Raju GSR, Huh YS, Bhaskar L. Phytochemical compounds for treating hyperuricemia associated with gout: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03686-4. [PMID: 39636406 DOI: 10.1007/s00210-024-03686-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Gout is a prevalent metabolic disorder characterized by increased uric acid (UA) synthesis or decreased UA clearance from the bloodstream, leading to the formation of urate crystals in joints and surrounding tissues. Hyperuricemia (HUA), the underlying cause of gout, poses a growing challenge for healthcare systems in developed and developing countries. Currently, the most common therapeutic approaches for gouty HUA primarily involve the use of allopathic or modern medicine. However, these treatments are often accompanied by adverse effects and may not be universally effective for all patients. Therefore, this systematic review aims to provide a comprehensive outline of phytochemical compounds that have emerged as alternative treatments for HUA associated with gout and to examine their specific mechanisms of action. A systematic search was conducted to identify phytochemicals that have previously been evaluated for their effectiveness in reducing HUA. From a review of > 800 published articles, 100 studies reporting on 50 phytochemicals associated with the management of HUA and gout were selected for analysis. Experimental models were used to investigate the effects of these phytochemicals, many of which exhibited multiple mechanisms beneficial for managing HUA. This review offers valuable insights for identifying and developing novel compounds that are safer and more effective for treating HUA associated with gout.
Collapse
Affiliation(s)
- Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Mohan Krishna Ghanta
- Department of Pharmacology, MVJ Medical College and Research Hospital, Bangalore, 562114, Karnataka, India
| | - Munirah Alhammadi
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon, 22212, Republic of Korea
| | - Arifullah Mohammed
- Department of Agriculture Science, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, 17600, Jeli, Kelantan, Malaysia
- College of Agriculture, KL University, Vaddeswaram Campus, Guntur, Andhra Pradesh, 522302, India
| | - Kuttiappan Anitha
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, 474005, Madhya Pradesh, India
| | - Poojith Nuthalapati
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon, 22212, Republic of Korea.
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, 495009, India.
| |
Collapse
|
2
|
Wang Y, Liang Y, Jiang L, Luo X, Cheng T, Long X. A network pharmacology and transcriptome analysis of the therapeutic effects of tea tree oil on the lungs of chicks exposed to hydrogen sulfide. Poult Sci 2024; 103:104180. [PMID: 39305611 PMCID: PMC11458973 DOI: 10.1016/j.psj.2024.104180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 10/11/2024] Open
Abstract
This study investigated the use of tea tree oil (TTO) in the treatment of H2S-induced lung injury in chickens, focusing on the detoxification mechanism. H2S can damage the respiratory system and reduce growth performance. TTO can improve immune inflammation and growth performance. The mechanism by which TTO mitigates the harmful effects of H2S on chicken lungs remains unclear. Therefore, the experimental model was established by H2S exposure and TTO addition in drinking water. The 240 one-day-old Roman pink chicks were selected for the experiment. The trial was divided into control group (CON), treatment group (TTG, 0.02 mL/L TTO+H2S) and H2S exposure group (AVG, H2S). There were 4 replicates in each group and the trial lasted for 42 d. The therapeutic effect of TTO on lung injury in chickens were determined by growth performance evaluation, transcription sequencing and network pharmacology analysis. The results showed that in the test's third week, the body weights of the chickens in the CON were higher than those in the AVG and TTG (P < 0.05). Pathological sections showed that TTO alleviated the symptoms of lung inflammation and bleeding caused by ROS. As showed by transcriptional sequencing, the mRNA expression of apoptosis-related genes Caspase-9, BAK-1, BCL-2 and BAX were significantly altered (P < 0.05). Meanwhile, the mRNA expression of inflammation-related genes IL-2, IL-6, and IL-17 were downregulated (P < 0.05). Network pharmacological analysis showed that CA2, CA4, GABRA5 and ADH1C were the key targets of TTO. The TTO treatment significantly altered these targets (P < 0.05). Molecular docking confirmed the strong binding ability between the active component and the targets. This study showed that TTO inhibits H2S-induced oxidative damage to the lungs, thereby improving their health status. This provides a new solution for the prevention of harmful gas in the poultry industry.
Collapse
Affiliation(s)
- Yachao Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China.
| | - Yilei Liang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Li Jiang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Xuegang Luo
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Tingting Cheng
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Xiaoyan Long
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| |
Collapse
|
3
|
Li D, Dong J, Xiong T, Zhou X, Li Y, Chen C, Li S, Song Z, Xu N, Yang M, Yan X, Liu T, Liu S. Transdermal delivery of iguratimod and colchicine ethosome by dissolving microneedle patch for the treatment of recurrent gout. Colloids Surf B Biointerfaces 2024; 242:114087. [PMID: 39003846 DOI: 10.1016/j.colsurfb.2024.114087] [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: 04/16/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
This study introduces a novel approach of repetitive modeling to simulate the pathological process of recurrent gout attacks in humans. This methodology addresses the instability issues present in rat models of gout, providing a more accurate representation of the damage recurrent gout episodes inflict on human skeletal systems. A soluble nanoneedle system encapsulating colchicine and iguratimod ethosomal formulations was developed. This system aims to modulate inflammatory cytokines and inhibit osteoclast activity, thereby treating inflammatory pain and bone damage associated with recurrent gout. Additionally, a comprehensive evaluation of the microneedles' appearance, morphology, mechanical properties, and penetration capability confirmed their effectiveness in penetrating the stratum corneum. Dissolution tests and skin irritation assessments demonstrated that these microneedles dissolve rapidly without irritating the skin. In vitro permeation studies indicated that transdermal drug delivery via these microneedles is more efficient and incurs lower drug loss compared to traditional topical applications. In vivo pharmacodynamic assessments conducted in animal models revealed significant analgesic and anti-inflammatory effects when both types of microneedles were used together. Further analyses, including X-ray imaging, hematoxylin and eosin (H&E) staining, Safranin-O/fast green staining, tartrate-resistant acid phosphatase staining, and quantification of osteoclasts, confirmed the bone-protective effects of the microneedle combination. In conclusion, the findings of this research underscore the potential of this novel therapeutic approach for clinical application in the treatment of recurrent gout.
Collapse
Affiliation(s)
- Du Li
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Jindian Dong
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Tong Xiong
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Xingyu Zhou
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Yanhui Li
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Chuncheng Chen
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Shijie Li
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Zhuoyue Song
- Bioengineering Laboratory, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, Guangdong 510070, PR China
| | - Nenggui Xu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Mingjing Yang
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Xiaoxia Yan
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Tao Liu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Shihui Liu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| |
Collapse
|
4
|
Zhang Z, Wang P, Xiong Q, Xu S, Kang D, He Z, Yao C, Jian G. Advancements in the study of IL-6 and its receptors in the pathogenesis of gout. Cytokine 2024; 182:156705. [PMID: 39053079 DOI: 10.1016/j.cyto.2024.156705] [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: 05/02/2024] [Revised: 06/11/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Gout is an autoinflammatory disease characterized by the deposition of monosodium urate crystals in or around the joints, primarily manifesting as inflammatory arthritis that recurs and resolves spontaneously. Interleukin-6 (IL-6) is a versatile cytokine with both anti-inflammatory and pro-inflammatory capabilities, linked to a variety of inflammatory diseases such as gouty arthritis, rheumatoid arthritis, inflammatory bowel disease, vasculitis, and several types of cancer. The rapid production of IL-6 during infections and tissue damage aids in host defense. However, excessive synthesis of IL-6 and dysregulation of its receptor signaling (IL-6R) might contribute to the pathology of diseases. Recent advancements in clinical and basic research, along with developments in animal models, have established the significant role of IL-6 and its receptors in the pathogenesis of gout, although the precise mechanisms remain to be fully elucidated. This review discusses the role of IL-6 and its receptors in gout progression and examines contemporary research on modulating IL-6 and its signaling pathways for treatment. It aims to provide insights into the pathogenesis of gout and to advance the development of targeted therapies for gout-related inflammation.
Collapse
Affiliation(s)
- Zeng Zhang
- The Third People's Hospital of Suining City, Suining 629000, Sichuan, China
| | - Peng Wang
- Xichong County People's Hospital, Nanchong 637200, Sichuan, China
| | - Qin Xiong
- The Third People's Hospital of Suining City, Suining 629000, Sichuan, China
| | - Shanshan Xu
- The Third People's Hospital of Suining City, Suining 629000, Sichuan, China
| | - Dong Kang
- The Third People's Hospital of Suining City, Suining 629000, Sichuan, China
| | - Zhengguang He
- The Third People's Hospital of Suining City, Suining 629000, Sichuan, China
| | - Chengjiao Yao
- Affiliated Hospital of Sichuan Bei Medical College, Nanchong 637000, Sichuan, China
| | - Guilin Jian
- The Third People's Hospital of Suining City, Suining 629000, Sichuan, China.
| |
Collapse
|
5
|
Matos IDA, Dallazen JL, Reis LR, Souza LF, Bevevino RC, de Moura RD, Ronsein GE, Hoch NC, da Costa Júnior NB, Costa SKP, Meotti FC. Targeting Myeloperoxidase Ameliorates Gouty Arthritis: A Virtual Screening Success Story. J Med Chem 2024; 67:12012-12032. [PMID: 38991154 DOI: 10.1021/acs.jmedchem.4c00721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
This study presents a new approach for identifying myeloperoxidase (MPO) inhibitors with strong in vivo efficacy. By combining inhibitor-like rules and structure-based virtual screening, the pipeline achieved a 70% success rate in discovering diverse, nanomolar-potency reversible inhibitors and hypochlorous acid (HOCl) scavengers. Mechanistic analysis identified RL6 as a genuine MPO inhibitor and RL7 as a potent HOCl scavenger. Both compounds effectively suppressed HOCl production in cells and neutrophils, with RL6 showing a superior inhibition of neutrophil extracellular trap release (NETosis). In a gout arthritis mouse model, intraperitoneal RL6 administration reduced edema, peroxidase activity, and IL-1β levels. RL6 also exhibited oral bioavailability, significantly reducing paw edema when administered orally. This study highlights the efficacy of integrating diverse screening methods to enhance virtual screening success, validating the anti-inflammatory potential of potent inhibitors, and advancing the MPO inhibitor research.
Collapse
Affiliation(s)
- Isaac de A Matos
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jorge L Dallazen
- Department of Pharmacology, Institute of Biological Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Lorenna R Reis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Luiz Felipe Souza
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Regina C Bevevino
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Rafael D de Moura
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Graziella E Ronsein
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Nicolas Carlos Hoch
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Soraia Kátia P Costa
- Department of Pharmacology, Institute of Biological Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Flavia C Meotti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
6
|
Ea HK, Kischkel B, Chirayath TW, Klück V, Aparicio C, Loeung HU, Manivet P, Jansen T, Zarka M, Lioté F, Latourte A, Bardin T, Gauffenic A, Vicaut E, Crișan TO, Netea MG, Richette P, Joosten LA. Systemic inflammatory cytokine profiles in patients with gout during flare, intercritical and treat-to-target phases: TNFSF14 as new biomarker. Ann Rheum Dis 2024; 83:945-956. [PMID: 38373842 DOI: 10.1136/ard-2023-225305] [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: 11/20/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024]
Abstract
INTRODUCTION Untreated gout is characterised by monosodium urate (MSU) crystal accumulation responsible for recurrent flares that are commonly separated by asymptomatic phases. Both phases are inflammatory conditions of variable intensity. Gout flares are self-limited inflammatory reactions involving multiple mediators. This study aimed to characterise the inflammatory profiles of gout at different phases. METHODS Using the Olink targeted proteomics, levels of 92 inflammation-related proteins were measured in plasma samples of a prospective gout population (GOUTROS), collected at gout flare (T1), the intercritical phase (T2) and after reaching the target serum urate level under urate-lowering therapy (T3). Results were validated in an independent cohort (OLT1177-05) with plasmas collected at T1 and T2. Ex vivo and in vitro experiments were performed to assess the inflammatory properties of new biomarkers. RESULTS In total, 21 inflammatory new biomarkers were differentially expressed during the three time-points of gout disease. The levels of four of these proteins (interleukin 6 (IL-6), colony-stimulating factor 1, vascular endothelial growth factor A and tumour necrosis factor superfamily 14 (TNFSF14)) were increased during gout flare in an independent cohort. IL-6 and TNFSF14 had the highest fold change in expression during T1 versus T2 or T3. TNFSF14 was produced at the inflamed joint and enhanced the inflammatory response induced by lipopolysaccharide and MSU crystal stimulation. Conversely, TNFSF14 blockade reduced the inflammatory response. Additionally, single nucleotide polymorphisms of TNFSF14 affected the ability of myeloid cells to produce inflammatory cytokines. CONCLUSION Gout flare involves multiple inflammatory mediators that may be used as potential therapeutic targets.
Collapse
Affiliation(s)
- Hang-Korng Ea
- Bioscar, INSERM UMR-1132, hôpital Lariboisière, centre Viggo Petersen, DMU Locomoteur, AP-HP, Universite Paris Cite, Paris, France
| | - Brenda Kischkel
- Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Viola Klück
- Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Philippe Manivet
- Assistance Publique-Hôpitaux de Paris, Centre de Ressources Biologiques, Paris, France
| | - Tim Jansen
- Rheumatology, VieCuri, Venlo, The Netherlands
| | - Mylène Zarka
- Bioscar, INSERM UMR-1132, Universite Paris Cite, Paris, France
| | - Frédéric Lioté
- Bioscar, INSERM UMR-1132, hôpital Lariboisière, centre Viggo Petersen, DMU Locomoteur, AP-HP, Universite Paris Cite, Paris, France
| | - Augustin Latourte
- Bioscar, INSERM UMR-1132, hôpital Lariboisière, centre Viggo Petersen, DMU Locomoteur, AP-HP, Universite Paris Cite, Paris, France
| | - Thomas Bardin
- Bioscar, INSERM UMR-1132, hôpital Lariboisière, centre Viggo Petersen, DMU Locomoteur, AP-HP, Universite Paris Cite, Paris, France
| | - Alan Gauffenic
- Bioscar, INSERM UMR-1132, hôpital Lariboisière, centre Viggo Petersen, DMU Locomoteur, AP-HP, Universite Paris Cite, Paris, France
| | - Eric Vicaut
- Unité de recherche clinique, Groupe hospitalier Lariboisiere Fernand-Widal, Paris, France
| | - Tania Octavia Crișan
- Department of Medical Genetics, Universitatea de Medicina si Farmacie Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Mihai G Netea
- Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pascal Richette
- Bioscar, INSERM UMR-1132, hôpital Lariboisière, centre Viggo Petersen, DMU Locomoteur, AP-HP, Universite Paris Cite, Paris, France
| | - Leo Ab Joosten
- Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Universitatea de Medicina si Farmacie Iuliu Hatieganu, Cluj-Napoca, Romania
| |
Collapse
|
7
|
Fan Y, Liu W, Jin Y, Lu H, Liu C, Wang A, Gu Q, Ka Y. To Investigate the Mechanism of Qinpi Tongfeng Formula in Treating Acute Gouty Arthritis by UHPLC-Q-Orbitrap-MS, Network Pharmacology and Experimental Validation. J Inflamm Res 2024; 17:3475-3498. [PMID: 38828049 PMCID: PMC11144412 DOI: 10.2147/jir.s454098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
Background Acute gouty arthritis (AGA) is characterized by the accumulation of monosodium urate crystals within the joints, leading to inflammation and severe pain. Western medicine treatments have limitations in addressing this condition. Previous studies have shown the efficacy of Qinpi Tongfeng formula (QPTFF) in treating AGA, but further investigation is needed to understand its mechanism of action. Methods We used ultra-high-performance liquid chromatography tandem Q-Exactive Orbitrap high-resolution mass spectrometry (UHPLC-Q-Orbitrap-MS) to identify compounds in QPTFF. Target proteins regulated by these compounds were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform, Chemistry Database, and Swiss Target Prediction Database. AGA-related targets were searched and screened from various databases, including Genecards, PharmGKB, Drugbank, etc. Intersection targets of QPTFF and AGA were analyzed for protein-protein interaction networks, GO function enrichment, and KEGG pathway enrichment. We then verified QPTFF's mechanism of action using an AGA rat model, assessing pathological changes via H&E staining and target expression via ELISA, RT-qPCR, and Western blot. Results UHPLC-Q-Orbitrap-MS identified 207 compounds in QPTFF, with 55 selected through network pharmacology. Of 589 compound-regulated targets and 1204 AGA-related targets, 183 potential targets were implicated in QPTFF's treatment of AGA. Main target proteins included IL-1β, NFKBIA, IL-6, TNF, CXCL8, and MMP9, with the IL-17 signaling pathway primarily regulated by QPTFF. Experimental results showed that medium and high doses of QPTFF significantly reduced serum inflammatory factors and MMP-9 expression, and inhibited IL-17A, IL-6, IKK-β, and NF-κB p65 mRNA and protein expression in AGA rats compared to the model group. Conclusion Key targets of QPTFF include IL-1β, NFKBIA, IL-6, TNF-α, CXCL8, and MMP9. QPTFF effectively alleviates joint inflammation in AGA rats, with high doses demonstrating no liver or kidney toxicity. Its anti-inflammatory mechanism in treating AGA involves the IL-17A/NF-κB p65 signaling pathway.
Collapse
Affiliation(s)
- Yihua Fan
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
- Department of Rheumatism and Immunity, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Wei Liu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Yue Jin
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Hang Lu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Chunliu Liu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Aihua Wang
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Qingxiang Gu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Yuxiu Ka
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| |
Collapse
|
8
|
Yang QB, Zhang MY, Yang L, Wang J, Mi QS, Zhou JG. Deficiency of histone deacetylases 3 in macrophage alleviates monosodium urate crystals-induced gouty inflammation in mice. Arthritis Res Ther 2024; 26:96. [PMID: 38711064 PMCID: PMC11071232 DOI: 10.1186/s13075-024-03335-4] [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: 10/31/2023] [Accepted: 05/01/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Gout is caused by monosodium urate (MSU) crystals deposition to trigger immune response. A recent study suggested that inhibition of Class I Histone deacetylases (HDACs) can significantly reduce MSU crystals-induced inflammation. However, which one of HDACs members in response to MSU crystals was still unknown. Here, we investigated the roles of HDAC3 in MSU crystals-induced gouty inflammation. METHODS Macrophage specific HDAC3 knockout (KO) mice were used to investigate inflammatory profiles of gout in mouse models in vivo, including ankle arthritis, foot pad arthritis and subcutaneous air pouch model. In the in vitro experiments, bone marrow-derived macrophages (BMDMs) from mice were treated with MSU crystals to assess cytokines, potential target gene and protein. RESULTS Deficiency of HDAC3 in macrophage not only reduced MSU-induced foot pad and ankle joint swelling but also decreased neutrophils trafficking and IL-1β release in air pouch models. In addition, the levels of inflammatory genes related to TLR2/4/NF-κB/IL-6/STAT3 signaling pathway were significantly decreased in BMDMs from HDAC3 KO mice after MSU treatment. Moreover, RGFP966, selective inhibitor of HDAC3, inhibited IL-6 and TNF-α production in BMDMs treated with MSU crystals. Besides, HDAC3 deficiency shifted gene expression from pro-inflammatory macrophage (M1) to anti-inflammatory macrophage (M2) in BMDMs after MSU challenge. CONCLUSIONS Deficiency of HDAC3 in macrophage alleviates MSU crystals-induced gouty inflammation through inhibition of TLR2/4 driven IL-6/STAT3 signaling pathway, suggesting that HDAC3 could contribute to a potential therapeutic target of gout.
Collapse
Affiliation(s)
- Qi-Bin Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People's Republic of China.
- Henry Ford Immunology Program, Departments of Dermatology and Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.
| | - Meng-Yun Zhang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People's Republic of China
- Department of Integrated TCM and Western Medicine, General Hospital of Central Theater, PLA, Wuhan, Hubei Province, 430070, China
- Henry Ford Immunology Program, Departments of Dermatology and Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA
| | - Liu Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, People's Republic of China
| | - Jie Wang
- Henry Ford Immunology Program, Departments of Dermatology and Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA
| | - Qing-Sheng Mi
- Henry Ford Immunology Program, Departments of Dermatology and Internal Medicine, Henry Ford Health System, 1 Ford Place, Detroit, MI, 48202, USA.
| | - Jing-Guo Zhou
- Department of Rheumatology and Immunology, Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan Province, 610500, People's Republic of China.
| |
Collapse
|
9
|
Zi X, Su R, Su R, Wang H, Li B, Gao C, Li X, Wang C. Elevated serum IL-2 and Th17/Treg imbalance are associated with gout. Clin Exp Med 2024; 24:9. [PMID: 38240927 PMCID: PMC10799120 DOI: 10.1007/s10238-023-01253-4] [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: 07/16/2023] [Accepted: 12/18/2023] [Indexed: 01/22/2024]
Abstract
Gout is considered an auto-inflammatory disorder, and the immunological drivers have not been fully unraveled. This study compared the peripheral lymphocyte and CD4+T cell subsets, and cytokines in gout and healthy controls (HCs) to explore the contributions of T helper 17 (Th17) cells, T regulatory (Treg) cells and cytokines to the pathogenesis of gout. We enrolled 126 gout patients (53 early-onset gout with age of first presentation < 40 years, and 73 late-onset gout with age of first presentation ≥ 40 years) and 77 HCs. Percentage and absolute numbers of peripheral lymphocyte and CD4+T cell subpopulations in each group were detected by flow cytometry. The serum cytokine levels were determined by flow cytometric bead array. For circulating CD4+T cell subsets, Th17/Treg ratio was significantly higher in early-onset gout, late-onset gout and gout without tophus than HCs; Th17 cells were significantly elevated in early-onset gout and gout without tophus, while the percentage of Treg cells was significantly decreased in early-onset and late-onset gout. Additionally, gout patients had significantly higher cytokines levels (including IL-2, IL-4, IL-6, IL-10, IL-17, IFN-γ, and TNF-α) than HCs; IL-2 levels were positively correlated with Treg cells and negatively correlated with ESR. ROC analysis showed that disease duration, CRP and fibrinogen, had moderate predictive performances for tophus in gout (the AUCs were 0.753, 0.703 and 0.701, respectively). Our study suggests that early-onset and late-onset gout differ in Th17/Treg imbalance, which in early-onset gout is due to elevated Th17 cells and in late-onset gout is due to decreased Treg cells. And increased serum cytokine levels, especially IL-2, may play an essential role in that. Restoring Th17/Treg balance may be a crucial way to improve the prognosis of gout patients.
Collapse
Affiliation(s)
- Xiaoyu Zi
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Ronghui Su
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Hui Wang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Baochen Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China.
| |
Collapse
|
10
|
Lu S, Qian T, Cao F, Kang T, Liu X, Wang C, Xia Y, Hou X. Prevalence and treatment rate of gout by depressive symptom severity: A cross-sectional analysis of NHANES 2007-2018. Int J Rheum Dis 2024; 27:e14959. [PMID: 37984452 DOI: 10.1111/1756-185x.14959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/02/2023] [Accepted: 10/15/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND The co-disease of depression and gout is becoming more common in the modern era. However, the relationship between the severity of depressive symptoms and gout prevalence and treatment rate was still unclear. OBJECTIVE This study aimed to determine the relationship between the prevalence, treatment rate of gout, and the severity of depression in the United States. METHOD The cross-sectional analysis of the 2007-2018 National Health and Nutrition Examination Survey (NHANES) for participants with depression was performed. According to their Patient Health Questionnaire-9 (PHQ-9) scores, participants were categorized as none, mild, moderate, moderately severe, and severe. To learn the correlation between the severity of depressive symptoms and the prevalence and treatment rate of gout using multivariate logistic regression to control for confounder interference. RESULTS A total of 25 022 patients were included in this study. As the severity of the depressive symptoms worsened (Mild, Moderate and Moderately severe), the risk of gout increased in non-adjusted model and model 1,2,3 (p-value for trend =.01 in non-adjusted model, <.0001 in model 1, <.01 in models 2 and 3; prevalence group in Model 1, aOR1.71, 95% CI (1.40, 2.08) in the mild group, aOR1.68, 95% CI (1.19, 2.39) in the moderate group, aOR1.31,95% CI (0.82, 2.11) in the moderately severe group, aOR1.21, 95% CI (0.62, 2.38) in the severe group). However, the lower gout prevalence trend has no statistical significance after adjusting all factors in Model 4(p-value for trend =.98). Compared with patients without depression, only a few patients received treatment, especially patients with severe depression (none, 80.1%; severe, 0.2%). The more severe the depression, the lower the treatment rate (p-value for trend: non-adjusted model, p < .001; model 1, p = .05; model 2, p = .02; model 3, p = .03). CONCLUSION Compared with patients without depression, the patients with depression had a higher risk of gout. With the aggravation of depression, the prevalence of gout and the rate of treatment both were decreased. Patients with gout and depression need to receive multidisciplinary care after diagnosis. However, currently, treatment cannot meet the needs of the current patients.
Collapse
Affiliation(s)
- Siyi Lu
- Department of Rheumatology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Tangliang Qian
- Department of Rheumatology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Fengjiao Cao
- Department of Rheumatology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Tianlun Kang
- Department of Rheumatology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoping Liu
- Department of Rheumatology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Chaoxin Wang
- Department of Functional Test, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yun Xia
- IRB (Institutional Review Board) Office, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xiujuan Hou
- Department of Rheumatology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
11
|
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: 0] [Impact Index Per Article: 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
|
12
|
Liu W, Peng J, Wu Y, Ye Z, Zong Z, Wu R, Li H. Immune and inflammatory mechanisms and therapeutic targets of gout: An update. Int Immunopharmacol 2023; 121:110466. [PMID: 37311355 DOI: 10.1016/j.intimp.2023.110466] [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: 03/20/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023]
Abstract
Gout is an autoimmune disease characterized by acute or chronic inflammation and damage to bone joints induced due to the precipitation of monosodium urate (MSU) crystals. In recent years, with the continuous development of animal models and ongoing clinical investigations, more immune cells and inflammatory factors have been found to play roles in gouty inflammation. The inflammatory network involved in gout has been discovered, providing a new perspective from which to develop targeted therapy for gouty inflammation. Studies have shown that neutrophil macrophages and T lymphocytes play important roles in the pathogenesis and resolution of gout, and some inflammatory cytokines, such as those in the interleukin-1 (IL-1) family, have been shown to play anti-inflammatory or proinflammatory roles in gouty inflammation, but the mechanisms underlying their roles are unclear. In this review, we explore the roles of inflammatory cytokines, inflammasomes and immune cells in the course of gout development and the research status of therapeutic drugs used for inflammation to provide insights into future targeted therapy for gouty inflammation and the direction of gout pathogenesis research.
Collapse
Affiliation(s)
- Wenji Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China; The Second Clinical Medical College of Nanchang University, 330006 Nanchang, China
| | - Jie Peng
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China; The Second Clinical Medical College of Nanchang University, 330006 Nanchang, China
| | - Yixin Wu
- Queen Mary College of Nanchang University, 330006 Nanchang, China
| | - Zuxiang Ye
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China; The Second Clinical Medical College of Nanchang University, 330006 Nanchang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, 1 MinDe Road, 330006 Nanchang, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China.
| | - Hui Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, 330006 Nanchang, China.
| |
Collapse
|
13
|
Ma Y, Yin Z, Dai H, Wu D, Cong J, Huang X, Chen X, Zou L, Ye Z, Huang Z. Increased Metallothionein-1 Associated with Gout Activity and Tophi. Immunol Invest 2023; 52:319-331. [PMID: 36719801 DOI: 10.1080/08820139.2023.2173078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND AIMS Gout is a chronic self-limiting inflammatory arthritis. An increase in metallothionein-1 (MT-1) has been reported in rheumatoid arthritis and osteoarthritis, and it attenuates inflammation and the pathology of diseases. This study aims to detect MT-1 levels in patients with gout and to explore its correlation with disease activity, clinical indexes, and inflammatory cytokines. METHODS The expression of MT-1 messenger RNAs (mRNAs) and protein levels in patients with gout were measured using real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Correlations between MT-1 and clinical indexes or inflammatory mediators were analyzed using Spearman's correlation test. RESULTS Compared with healthy controls (HCs, n = 43), patients with active gout (n = 27) showed higher levels of MT-1 mRNA in peripheral blood mononuclear cells and protein in serum, particularly those with tophi. No significant difference in serum MT-1 levels was observed among patients with inactive gout, HCs, and patients with hyperuricemia without gout. Furthermore, no significant difference was observed between patients with gout with kidney damage and HCs. In addition, serum interleukin (IL)-1β, IL-6, and IL-8 levels were significantly increased in patients with active gout, particularly in those with tophi. The serum MT-1 level was positively correlated with C-reactive protein, as well as with IL-1β, IL-6, and IL-18. CONCLUSION The higher levels of MT-1 were found in patients with gout, which were correlated with disease activity and gout related pro-inflammatory cytokines. Indicating MT-1 may serve as a new marker for predicting disease activity.Abbreviations: IL-1β: Interleukin 1β; MT-1: Metallothionein-1; CRP: C-Reactive Protein; ROS: Reactive Oxygen Species; IL-10: Interleukin 10; TGF-β: Transforming Growth Factor Beta.
Collapse
Affiliation(s)
- Yanmei Ma
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Hanying Dai
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China
| | - Dandan Wu
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China
| | - Junxiao Cong
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Xinmin Huang
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Xinpeng Chen
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Linghua Zou
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhizhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhong Huang
- Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China.,Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| |
Collapse
|
14
|
Zhang J, Sun W, Gao F, Lu J, Li K, Xu Y, Li Y, Li C, Chen Y. Changes of serum uric acid level during acute gout flare and related factors. Front Endocrinol (Lausanne) 2023; 14:1077059. [PMID: 36896178 PMCID: PMC9989260 DOI: 10.3389/fendo.2023.1077059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
OBJECTIVE By studying the changes of serum uric acid (SUA) in acute stage and remission stage of gouty arthritis, we aimed to explore the relationship between the changes of SUA level and free glucocorticoids and inflammatory factors. METHODS A prospective, longitudinal study was conducted on 50 acute gout patients in the dedicated gout clinic of the Affiliated Hospital of Qingdao University. Blood and 24-hour urine samples were collected during the acute phase and two weeks after the initial visit. Patients with acute gouty arthritis were treated primarily with colchicine and nonsteroidal anti-inflammatory drugs. RESULTS A total of 32 patients completed the two-week follow-up trial. SUA levels were significantly downregulated during the acute flare than after the flare (464.14 ± 90.97 vs. 527.36 ± 86.90 μmol/L, p < 0.001). The 24-hour fractional excretion of uric acid (24 h FEur) (5.54 ± 2.82% vs. 4.68 ± 2.83%, p < 0.001) and 24-hour urinary uric acid excretion (24 h Uur) (663.08 ± 249.48 μmol/L vs. 540.87 ± 263.18 μmol/L, p = 0.001) increased significantly in patients during the acute phase. The percent change in SUA was associated with those in 24 h FEur and C-reactive protein. Meanwhile, the percent change in 24 h Uur was associated with those in 24-hour urinary free cortisol, percent change in interleukin 1β and interleukin 6. CONCLUSION Decreased SUA level during the acute gout flare was associated with increased excretion of urinary uric acid. Inflammatory factors and bioactive free glucocorticoids may play significant roles in this process.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Endocrinology and Metabolism, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenyan Sun
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fei Gao
- Department of Hand and Foot Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jie Lu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Kelei Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Yijun Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yushuang Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changgui Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, The Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Metabolic Diseases, Qingdao University, Qingdao, China
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
- *Correspondence: Ying Chen, ; Changgui Li,
| | - Ying Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
- *Correspondence: Ying Chen, ; Changgui Li,
| |
Collapse
|
15
|
Hao G, Xu X, Song J, Zhang J, Xu K. Lipidomics analysis facilitate insight into the molecular mechanisms of urate nephropathy in a gout model induced by combination of MSU crystals injection and high-fat diet feeding. Front Mol Biosci 2023; 10:1190683. [PMID: 37206890 PMCID: PMC10188924 DOI: 10.3389/fmolb.2023.1190683] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Renal injury is one of the most common clinical manifestations of patients with hyperuricaemia/gout. The precise pathophysiological mechanism(s) for the renal injury is still unknown. Furthermore, it is also unclear whether the clinical therapies (e.g., colchicine and febuxostat) could prevent its progression or not. Lipids are involved in almost all of important biological processes and play critical roles in maintaining the renal functions. Herein, shotgun lipidomics was performed for class-targeted lipid analysis of cellular lipidomes in renal tissue of a gouty model induced by combination of monosodium urate crystals injection and high-fat diet feeding with/without treatment with either colchicine or febuxostat. Serum uric acid (UA), proinflammatory cytokines (i.e., TNF-α and IL-6), xanthine oxidase activity, footpad swelling, and pain threshold were determined to evaluate the gouty severity. Renal histopathological changes, blood urea nitrogen, creatinine, and kidney index were used to reflect renal injury. Lipidomics analysis revealed that altered triacylglycerol (TAG) profile, impaired mitochondrial function resulted by decreased tetra 18:2 cardiolipin, reduced 4-hydroxyalkenal (HNE) species, and elevated lysophospholipids were already present in the kidneys at early stage of renal injury, probably contributing to its occurrence and development. In addition to significantly reduce the UA level and relief the gouty severity, treatment with either colchicine or febuxostat could restore HNE bioavailability, thereby delaying the progression of renal injury. However, both of them could not recover the altered TAG profile and the impaired mitochondrial function, indicating that treatment with either of them could not completely prevent the development of renal injury in the gouty model.
Collapse
Affiliation(s)
- Guifeng Hao
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Xiaofen Xu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jingyi Song
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jida Zhang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kejun Xu
- Emergency Medicine Department, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- *Correspondence: Kejun Xu,
| |
Collapse
|
16
|
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
|
17
|
Xu L, Cheng J, Lu J, Lin G, Yu Q, Li Y, Chen J, Xie J, Su Z, Zhou Q. Integrating network pharmacology and experimental validation to clarify the anti-hyperuricemia mechanism of cortex phellodendri in mice. Front Pharmacol 2022; 13:964593. [PMID: 36438835 PMCID: PMC9692208 DOI: 10.3389/fphar.2022.964593] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/24/2022] [Indexed: 08/12/2023] Open
Abstract
Hyperuricemia (HUA), a common metabolic disease, is treated as the second-largest metabolic disease after diabetes in China. Cortex Phellodendri (CP) is one of the most frequently used herbal medicines for treating gout or HUA. However, the mechanism underlying the anti-HUA effect of CP is still unrevealed. Hence, this study aimed to explore the pharmacological mechanism of CP against HUA using network pharmacology coupled with in vivo experimental validation. Active compounds and potential targets of CP, as well as the potential targets related to HUA, were retrieved from multiple open-source databases. The drug-disease overlapping targets were obtained by Venn diagram analysis and used to construct the herb-component-target (HCT), protein-protein-interaction (PPI), and component-target-pathway (CTP) networks. The functional enrichment analysis was also performed for further study. Furthermore, a HUA mouse model was induced by a combination of intraperitoneal injection of potassium oxonate (PO, 300 mg/kg) and intragastric administration of hypoxanthine (HX, 300 mg/kg) daily for 10 days. Different dosages of CP (200, 400, and 800 mg/kg) were orally given to mice 1 h after modeling. The results showed that 12 bioactive compounds and 122 drug-disease overlapping targets were obtained by matching 415 CP-related targets and 679 HUA-related targets, and berberine was one of the most important compounds with the highest degree value. The core targets of CP for treating HUA were TP53, MAPK8, MAPK3, IL-6, c-Jun, AKT1, xanthine oxidase (XOD), and ATP-binding cassette subfamily G member 2 (ABCG2). The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment results showed that the anti-HUA effect of CP mainly involved the pathways of inflammation and apoptosis, such as PI3K/Akt, TNF, MAPK, TLR, AMPK, NF-κB, and NLRP3 signaling pathways. In vivo animal experiment further confirmed the hypouricemic effect of CP in a HUA mouse model, as evidenced by significantly restored kidney histological deteriorations, and considerably decreased levels of serum uric acid (sUA), creatinine (Cre), blood urea nitrogen (BUN), and hepatic UA. Furthermore, the hypouricemic action of CP in vivo might be attributed to its suppression of XOD activity in the liver, rather than ABCG2 in the kidney. Real-time qPCR (RT-qPCR) and Western blot analysis also confirmed the key roles of the hub genes in CP against HUA. In conclusion, CP exhibited therapeutic effect against HUA via multi-compounds, multi-targets, and multi-pathways. It possessed anti-HUA and nephroprotective effects via suppressing XOD activity, and reversed the progression of renal injury by exerting anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Lieqiang Xu
- College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, China
| | - Juanjuan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jieyi Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiuxia Yu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| |
Collapse
|
18
|
Jiao C, Liang H, Liu L, Li S, Chen J, Xie Y. Transcriptomic analysis of the anti-inflammatory effect of Cordyceps militaris extract on acute gouty arthritis. Front Pharmacol 2022; 13:1035101. [PMID: 36313318 PMCID: PMC9614083 DOI: 10.3389/fphar.2022.1035101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Gouty arthritis (GA) is a common inflammatory disease that causes pain due to the deposition of monosodium urate (MSU) crystals into joints and surrounding tissues. Anti-inflammatory drugs have significant clinical anti-inflammatory and analgesic effects, but they have many side effects. Cordyceps militaris is an edible and medicinal fungus, and its extract (CME) has good anti-inflammatory and analgesic effects. This study aimed to investigate the anti-inflammatory effect of CME on GA and its underlying mechanism. Methods: The effect of CME on the expression of related inflammatory factors and histopathological changes in the MSU-induced acute inflammatory gout model in rats was studied by ELISA and HE, and its anti-inflammatory mechanism was analyzed by transcriptome combined with RT-qPCR. Results: CME significantly improved gait scores and joint swelling in GA rats, and reduced MSU-induced inflammatory cell infiltration. CME inhibited MSU-induced inflammatory responses by reducing the levels of pro-inflammatory factors TNF-α, IL-1β, IL-6, and Caspase-1 and increasing the anti-inflammatory factor IL-10. Transcriptome analysis showed that CME significantly altered inflammation-related cytokine pathways, and identified four major genes involved in regulation of inflammation, CCL7, CSF2RB, LIF, and IL-1β. In addition, RT-qPCR was performed to verify these differential genes. Conclusion: CME significantly alleviated the inflammatory progression of GA and ameliorated the onset of GA. The underlying mechanism may be related to triggering the cytokine-cytokine receptor interaction signaling pathway to inhibit the activation of the inflammasome and regulate the immune system. And it regulates the inflammatory response induced by MSU crystals through the genes CCL7, CSF2RB, and IL-1β.
Collapse
Affiliation(s)
- Chunwei Jiao
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, China
| | - Huijia Liang
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
| | - Li Liu
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
| | - Shunxian Li
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, China
| | - Jiaming Chen
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
| | - Yizhen Xie
- Guangdong Yuewei Edible Fungi Technology Co, Ltd., Guangzhou, China
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, China
- *Correspondence: Yizhen Xie,
| |
Collapse
|
19
|
Wu ZD, Yang XK, He YS, Ni J, Wang J, Yin KJ, Huang JX, Chen Y, Feng YT, Wang P, Pan HF. Environmental factors and risk of gout. ENVIRONMENTAL RESEARCH 2022; 212:113377. [PMID: 35500858 DOI: 10.1016/j.envres.2022.113377] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/30/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
Gout is a chronic disease with inflammatory arthritis caused by monosodium urate (MSU) crystals deposition, an elevated serum urate level (hyperuricaemia) is the critical factor leading to MSU crystals deposition and promoting the progression of gout. The onset and development of gout is generally the result of multiple factors, such as diet, heredity and environmental factors. Although genetics and diet are thought to play as major factors, a growing body of research evidence has highlighted that environmental factors also play a significant role in the onset and exacerbation of gout. Recent studies have shown that air pollutants such as particulate matter, sulfur dioxide (SO2) and carbon monoxide (CO) may increase the risk of hospitalizations for gout, and that the changes in temperature and humidity may affect uric acid (UA) levels. There is also seasonal trend in gout. It has been demonstrated that environmental factors may induce or accelerate the production and release of pro-inflammatory mediators, causing an unbalance oxidative stress and systemic inflammation, and then participating in the overall process or a certain link of gout. Moreover, several environmental factors have shown the ability to induce the production urate and regulate the innate immune pathways, involving in the pathogenesis of gout. Nevertheless, the role of environmental factors in the etiology of gout remains unclear. In this review, we summarized the recent literatures and aimed to discuss the relationship between environmental factors (such as microclimate, season, ambient/indoor air pollution and extreme weather) and gout. We further discussed the inflammatory mechanisms of environmental factors and gout and the comprehensive effects of environmental factors on gout. We also made a prospect of the management and treatment of gout, with special consideration to environmental factors associated with gout.
Collapse
Affiliation(s)
- Zheng-Dong Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Xiao-Ke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Jie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Kang-Jia Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Yue Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Ya-Ting Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Peng Wang
- Teaching Center of Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.
| |
Collapse
|
20
|
Cao JF, Xingyu Yang, Li Xiong, Wu M, Chen S, Xu H, Gong Y, Zhang L, Zhang Q, Zhang X. Exploring the mechanism of action of dapansutrile in the treatment of gouty arthritis based on molecular docking and molecular dynamics. Front Physiol 2022; 13:990469. [PMID: 36105284 PMCID: PMC9465377 DOI: 10.3389/fphys.2022.990469] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/12/2022] [Indexed: 01/02/2023] Open
Abstract
Purpose: Dapansutrile is an orally active β-sulfonyl nitrile compound that selectively inhibits the NLRP3 inflammasome. Clinical studies have shown that dapansutrile is active in vivo and limits the severity of endotoxin-induced inflammation and joint arthritis. However, there is currently a lack of more in-depth research on the effect of dapansutrile on protein targets such as NLRP3 in gouty arthritis. Therefore, we used molecular docking and molecular dynamics to explore the mechanism of dapansutrile on NLRP3 and other related protein targets. Methods: We use bioinformatics to screen active pharmaceutical ingredients and potential disease targets. The disease-core gene target-drug network was established and molecular docking was used for verification. Molecular dynamics simulations were utilized to verify and analyze the binding stability of small molecule drugs to target proteins. The supercomputer platform was used to measure and analyze the binding free energy, the number of hydrogen bonds, the stability of the protein target at the residue level, the radius of gyration and the solvent accessible surface area. Results: The protein interaction network screened out the core protein targets (such as: NLRP3, TNF, IL1B) of gouty arthritis. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that gouty arthritis mainly played a vital role by the signaling pathways of inflammation and immune response. Molecular docking showed that dapansutrile play a role in treating gouty arthritis by acting on the related protein targets such as NLRP3, IL1B, IL6, etc. Molecular dynamics was used to prove and analyze the binding stability of active ingredients and protein targets, the simulation results found that dapansutrile forms a very stable complex with IL1B. Conclusion: We used bioinformatics analysis and computer simulation system to comprehensively explore the mechanism of dapansutrile acting on NLRP3 and other protein targets in gouty arthritis. This study found that dapansutrile may not only directly inhibit NLRP3 to reduce the inflammatory response and pyroptosis, but also hinder the chemotaxis and activation of inflammatory cells by regulating IL1B, IL6, IL17A, IL18, MMP3, CXCL8, and TNF. Therefore, dapansutrile treats gouty arthritis by attenuating inflammatory response, inflammatory cell chemotaxis and extracellular matrix degradation by acting on multiple targets.
Collapse
|
21
|
Cheng JJ, Ma XD, Ai GX, Yu QX, Chen XY, Yan F, Li YC, Xie JH, Su ZR, Xie QF. Palmatine Protects Against MSU-Induced Gouty Arthritis via Regulating the NF-κB/NLRP3 and Nrf2 Pathways. Drug Des Devel Ther 2022; 16:2119-2132. [PMID: 35812134 PMCID: PMC9259749 DOI: 10.2147/dddt.s356307] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose Gouty arthritis could be triggered by the deposition of monosodium uric acid (MSU) crystals. Palmatine (PAL), a protoberberine alkaloid, has been proven to possess compelling health-beneficial activities. In this study, we aimed to explore the effect of PAL on LPS plus MSU crystal-stimulated gouty arthritis in vitro and in vivo. Methods PMA-differentiated THP-1 macrophages were primed with LPS and then stimulated with MSU crystal in the presence or absence of PAL. The expression of pro-inflammatory cytokines and oxidative stress-related biomarkers and signal pathway key targets were determined by ELISA kit, Western blot, immunohistochemistry and qRT-PCR, respectively. In addition, the anti-inflammatory and antioxidant activities of PAL on MSU-induced arthritis mice were also evaluated. Results The results indicated that PAL (20, 40 and 80 μM) dose-dependently decreased the mRNA expression and levels of pro-inflammatory cytokines (interleukin-1beta (IL-1β), IL-6, IL-18 and tumor necrosis factor alpha (TNF-α)). The levels of superoxide dismutase (SOD) and glutathione (GSH) were remarkably enhanced, while the level of malondialdehyde (MDA) was reduced. Western blot analysis revealed that PAL appreciably inhibited NF-κB/NLRP3 signaling pathways through inhibiting the phosphorylation of p-65 and IκBα, blocking the expression of NLRP3, ASC, IL-1β and Caspase-1, as well as enhancing the antioxidant protein expression of Nrf2 and HO-1. In vivo, PAL attenuated MSU-induced inflammation in gouty arthritis, as evidenced by mitigating the joint swelling, and decreasing the productions of IL-1β, IL-6, IL-18, TNF-α and MDA, while enhancing the levels of SOD and GSH. Moreover, PAL further attenuated the infiltration of neutrophils into joint synovitis. Conclusion PAL protected against MSU-induced inflammation and oxidative stress via regulating the NF-κB/NLRP3 and Nrf2 pathways. PAL may represent a potential candidate for the treatment of gouty arthritis.
Collapse
Affiliation(s)
- Juan-Juan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Xing-Dong Ma
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Gao-Xiang Ai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Qiu-Xia Yu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China
| | - Xiao-Ying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Fang Yan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China.,Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Yu-Cui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Jian-Hui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, 510120, People's Republic of China
| | - Zi-Ren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Qing-Feng Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People's Republic of China.,Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| |
Collapse
|
22
|
The Role of Inflammasomes in Osteoarthritis and Secondary Joint Degeneration Diseases. Life (Basel) 2022; 12:life12050731. [PMID: 35629398 PMCID: PMC9146751 DOI: 10.3390/life12050731] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 12/23/2022] Open
Abstract
Osteoarthritis is age-related and the most common form of arthritis. The main characteristics of the disease are progressive loss of cartilage and secondary synovial inflammation, which finally result in pain, joint stiffness, and functional disability. Similarly, joint degeneration is characteristic of systemic inflammatory diseases such as rheumatoid arthritis and gout, with the associated secondary type of osteoarthritis. Studies suggest that inflammation importantly contributes to the progression of the disease. Particularly, cytokines TNFα and IL-1β drive catabolic signaling in affected joints. IL-1β is a product of inflammasome activation. Inflammasomes are inflammatory multiprotein complexes that propagate inflammation in various autoimmune and autoinflammatory conditions through cell death and the release of inflammatory cytokines and damage-associated molecule patterns. In this article, we review genetic, marker, and animal studies that establish inflammasomes as important drivers of secondary arthritis and discuss the current evidence for inflammasome involvement in primary osteoarthritis. The NLRP3 inflammasome has a significant role in the development of secondary osteoarthritis, and several studies have provided evidence of its role in the development of primary osteoarthritis, while other inflammasomes cannot be excluded. Inflammasome-targeted therapeutic options might thus provide a promising strategy to tackle these debilitating diseases.
Collapse
|
23
|
Verma AK, Hossain MS, Ahmed SF, Hussain N, Ashid M, Upadhyay SK, Vishvakarma NK, Bhojiya AA, Srivastava SK. " In silico identification of ethoxy phthalimide pyrazole derivatives as IL-17A and IL-18 targeted gouty arthritis agents". J Biomol Struct Dyn 2022:1-15. [PMID: 35532103 DOI: 10.1080/07391102.2022.2071338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Two proinflammatory cytokines, IL17A and IL18, are observed to be elevated in the serum of gout patients and they play a crucial role in the development and worsening of inflammation, which has severe effects. In present study, we have combined molecular docking, molecular dynamics studies and MM-PBSA analysis to study the effectiveness of ethoxy phthalimide pyrazole derivatives (series 3a to 3e) as potential inhibitors against cytokines IL17A and IL18 as a druggable targets. The binding energy of the docked series ranges from -13.5 to -10.0 kcal/mol and extensively interacts with the amino acids in the active pocket of IL17A and IL18. Compound 3e had the lowest binding energy with IL17A at -12.6 kcal/mol compared to control allopurinol (3.32 kcal/mol). With IL18, compound 3a seems to have the lowest binding energy of -9.6 kcal/mol compared to control allopurinol (3.18 kcal/mol). In MD simulation studies, compound 3a forms a stable and energetically stabilized complex with the target protein. Depending on properties of the bound IL17A-3a and IL18-3a complexes was compared by means of MM-PBSA analysis. These derivatives can be used as a scaffold to develop promising IL17A and IL18 inhibitors to assess their potential for gouty arthritis and other related diseases.
Collapse
Affiliation(s)
- Abhishek Kumar Verma
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Off Jaipur-Ajmer Expressway, Jaipur, Rajasthan, India
| | - Md Shahadat Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Sonapur, Noakhali, Bangladesh
| | - Sk Faisal Ahmed
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Sonapur, Noakhali, Bangladesh
| | - Nasir Hussain
- Department of Chemistry, Faculty of Science and Technology, Mewar University, Chittorgarh, Rajasthan, India
| | - Mohammad Ashid
- Department of Chemistry, Faculty of Science and Technology, Mewar University, Chittorgarh, Rajasthan, India
| | - Sudhir K Upadhyay
- Department of Environmental Science, V.B.S. Purvanchal University, Jaunpur, Uttar Pradesh, India
| | | | - Ali Asger Bhojiya
- Department of Science, U.S. Ostwal Science, Arts & Commerce College, Chittorgarh, India
| | - Sandeep Kumar Srivastava
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Off Jaipur-Ajmer Expressway, Jaipur, Rajasthan, India
| |
Collapse
|
24
|
Wu C, Li F, Zhang X, Xu W, Wang Y, Yao Y, Han Z, Xia D. (-)-Epicatechin Ameliorates Monosodium Urate-Induced Acute Gouty Arthritis Through Inhibiting NLRP3 Inflammasome and the NF-κB Signaling Pathway. Front Pharmacol 2022; 13:799552. [PMID: 35462936 PMCID: PMC9019746 DOI: 10.3389/fphar.2022.799552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Gouty arthritis is a common and complex inflammatory disease that will reduce the life quality of human beings (-)-Epicatechin (EC) is famous for antioxidant and anti-inflammatory activities. Thus, the aim of this study was to investigate the therapeutic effect of EC on gouty arthritis and its mechanisms. Methods and results: EC was added into a monosodium urate (MSU)-stimulated THP-1 cell that was induced by phorbol 12-myristate 13-acetate and lipopolysaccharide (LPS) in advance to establish a gout model in vitro. The efficiency of EC on acute gouty arthritis mice induced by MSU was further investigated. The results showed that EC concentration-dependently improved the cell viability of LPS and MSU stimulated THP-1 cells, and significantly alleviated MSU-induced ankle edema in mice in a dose-dependent manner. In addition, EC inhibited the infiltration of inflammatory cells and local cascular congestion in ankle joint tissue. Furthermore, the secretion of inflammatory cytokines (IL-1β, IL-18, IL-6, and TNF-α) activation of NLRP3 inflammasome and NF-κB signaling pathway were markedly suppressed by EC in vitro and in vivo. Conclusion: These results indicated that EC could effectively improve MSU-induced acute gouty arthritis via inhibiting NLRP3 inflammasome and the NF-κB signaling pathway in vitro and in vivo, which suggested that EC might be a promising active ingredient for the prevention and treatment of gouty arthritis.
Collapse
Affiliation(s)
- Chenxi Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fenfen Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoxi Zhang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenjing Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanjing Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ziwei Han
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
25
|
Sansone A, Reisman Y, Jannini EA. Relationship between hyperuricemia with deposition and sexual dysfunction in males and females. J Endocrinol Invest 2022; 45:691-703. [PMID: 34997558 PMCID: PMC8741558 DOI: 10.1007/s40618-021-01719-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022]
Abstract
PURPOSE The association between gout, the most common crystal arthropathy, and sexual dysfunctions has often been investigated by studies in the last decades. Despite the presence of shared risk factors and comorbidities and the possible effects on sexual health of long-term gout complications, awareness of this association is severely lacking and the pathogenetic mechanisms have only partially been identified. In the present review, we aimed to investigate the current evidence regarding the potential mechanisms linking sexual dysfunctions and gout. METHODS A comprehensive literature search within PubMed was performed to provide a summary of currently available evidence regarding the association between gout and sexual dysfunctions. RESULTS Gout and sexual dysfunctions share several risk factors, including diabesity, chronic kidney disease, hypertension, metabolic syndrome, and peripheral vascular disease. Gout flares triggered by intense inflammatory responses feature severe pain and disability, resulting in worse sexual function, and some, but not all, treatments can also impair sexual health. Long-term gout complications can result in persistent pain and disability due to joint deformity, fractures, or nerve compression, with negative bearing on sexual function. The presence of low-grade inflammation impairs both sex steroids synthesis and endothelial function, further advancing sexual dysfunctions. The psychological burden of gout is another issue negatively affecting sexual health. CONCLUSIONS According to currently available evidence, several biological and psychological mechanisms link sexual dysfunctions and gout. Addressing risk factors and providing adequate treatment could potentially have beneficial effects on both conditions. Appropriate clinical evaluation and multidisciplinary approach are recommended to improve patient care.
Collapse
Affiliation(s)
- A Sansone
- Chair of Endocrinology and Medical Sexology (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy
| | - Y Reisman
- Flare-Health, Amstelveen, The Netherlands
| | - E A Jannini
- Chair of Endocrinology and Medical Sexology (ENDOSEX), Department of Systems Medicine, University of Rome Tor Vergata, via Montpellier 1, 00133, Rome, Italy.
| |
Collapse
|
26
|
Feng Y, Yu Y, Chen Z, Wang L, Ma J, Bai X, Sun Y, Wang D. Effects of β-Carotin and Green Tea Powder Diets on Alleviating the Symptoms of Gouty Arthritis and Improving Gut Microbiota in C57BL/6 Mice. Front Microbiol 2022; 13:837182. [PMID: 35145506 PMCID: PMC8821968 DOI: 10.3389/fmicb.2022.837182] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
As a chronic metabolic disease caused by disorders of purine metabolism, gout has shown increasing incidence rate worldwide. Considering that gout is not easily treated and cured, further studies are explored to prevent gout development through diet modification. Both β-carotin and green tea powder are rich in dietary fiber, which helps maintain the balance of gut microbiota in humans. The aim of this study was to investigate the effects of β-carotin and green tea powder diet on the prevention of gouty arthritis in relation to the bacterial structure of gut microbiota in mice. We successfully induced gouty arthritis in C57BL/6 mice by injecting monosodium urate (MSU) crystals and feeding high-fat diet (HFD), and further investigated the effects of additional β-carotin and green tea powder in the diets of mice on the prevention of gouty arthritis in mice. Our results showed that diet of β-carotin and green tea powder reduced the joint swelling and pain in mice with gout, reduced the levels of serum uric acid (UA) and three types of pro-inflammatory cytokines, i.e., interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), improved the gut microbiota profile, and reduced the metabolic levels of purines and pyrimidines. In conclusion, our study provided evidence to support the application of β-carotin and green tea powder diet as a dietary adjustment method to prevent and treat gouty arthritis.
Collapse
Affiliation(s)
- Yu Feng
- Department of Orthopedic, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yanbo Yu
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| | - Zheng Chen
- Department of Orthopedic, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lili Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingyu Ma
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaohui Bai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yundong Sun
- Department of Microbiology, Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunology of Shandong Province, School of Basic Medicine, Shandong University, Jinan, China
| | - Dawei Wang
- Department of Orthopedic, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Orthopedic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
27
|
Biochemical and molecular effects of a commercial diuretic with herbal extract on experimentally induced urolithiasis in chickens. Vet Res Commun 2021; 46:419-430. [PMID: 34846629 DOI: 10.1007/s11259-021-09866-4] [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: 05/19/2021] [Accepted: 11/19/2021] [Indexed: 01/10/2023]
Abstract
This study evaluated the diuretic, antioxidant, anti-inflammatory, and immunological effects of a commercial diuretic (CD) (composed of ammonium chloride, potassium citrate, sodium chloride, ascorbic acid, biotin, halfa bar extract, and hexamine) on chickens with induced urolithiasis. A total of 100 one-day-old white Hy-Line chicks were fed a basal diet containing 20% crude protein (CP) and 1% Ca until they reached 48 days of age. Then, the birds were divided into five groups (G1-G5). G1 was fed a basal diet and kept as a negative control, G2 was fed a high protein (HP) diet containing 25% crude protein, G3 was fed high calcium (HC) diet containing 5% Ca, G4 was fed HP diet supplemented with CD, and G5 was fed HC diet supplemented with CD. The CD was supplemented with drinking water (at a dose of 0.5 ml/ liter) for 1 week. The experiment was held for 78 days. Clinical signs, postmortem lesions, and mortality rates were observed. Biochemical analytes, redox status biomarkers, and expression of interleukin-6 (IL-6) and interferon-gamma (IFN-γ) were measured. Tissue samples were taken for histopathological examination. No signs of CD toxicity were observed during the toxicity test prior to the experiment. Compared to all groups, birds in G2 and G3 showed impaired renal function and alterations in biochemical, redox status, lipid peroxidation, post-mortem, and histopathological lesions along with upregulation of IL-6 and IFN-γ in the kidney and spleen. In conclusion, commercial diuretic supplementation for one week improves renal function, redox status, immune and anti-inflammatory responses in chickens with induced urolithiasis.
Collapse
|
28
|
Kimura Y, Tsukui D, Kono H. Uric Acid in Inflammation and the Pathogenesis of Atherosclerosis. Int J Mol Sci 2021; 22:ijms222212394. [PMID: 34830282 PMCID: PMC8624633 DOI: 10.3390/ijms222212394] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/06/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Hyperuricemia is a common metabolic syndrome. Elevated uric acid levels are risk factors for gout, hypertension, and chronic kidney diseases. Furthermore, various epidemiological studies have also demonstrated an association between cardiovascular risks and hyperuricemia. In hyperuricemia, reactive oxygen species (ROS) are produced simultaneously with the formation of uric acid by xanthine oxidases. Intracellular uric acid has also been reported to promote the production of ROS. The ROS and the intracellular uric acid itself regulate several intracellular signaling pathways, and alterations in these pathways may result in the development of atherosclerotic lesions. In this review, we describe the effect of uric acid on various molecular signals and the potential mechanisms of atherosclerosis development in hyperuricemia. Furthermore, we discuss the efficacy of treatments for hyperuricemia to protect against the development of atherosclerosis.
Collapse
Affiliation(s)
- Yoshitaka Kimura
- Department of Internal Medicine, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan; (Y.K.); (D.T.)
- Department of Microbiology and Immunology, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan
| | - Daisuke Tsukui
- Department of Internal Medicine, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan; (Y.K.); (D.T.)
| | - Hajime Kono
- Department of Internal Medicine, Faculty of Medicine, Teikyo University of Medicine, Tokyo 173-8605, Japan; (Y.K.); (D.T.)
- Correspondence: ; Tel.: +81-3-3964-1211
| |
Collapse
|
29
|
Mansouri N, Marjani M, Tabarsi P, von Garnier C, Mansouri D. Successful Treatment of Covid-19 Associated Cytokine Release Syndrome with Colchicine. A Case Report and Review of Literature. Immunol Invest 2021; 50:884-890. [PMID: 32633162 PMCID: PMC7441796 DOI: 10.1080/08820139.2020.1789655] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We describe the case of a 42 year old, healthy patient with Covid-19 who despite improvement in his respiratory symptoms developed a mild to moderate cytokine release syndrome (CRS) and an associated monoarticular gout flare. Since the patient refused admission to the hospital and had stable vital signs, we chose to treat him with a safe anti-inflammatory and non-immunosuppressive therapy. To hit two birds with one stone, we considered colchicine, as it has systemic anti-inflammatory effects and is also effective in gout flare. Unexpectedly, 48 hours after treatment, not only did his ongoing fever and toe pain disappear, he also had significant improvements in his general state of health and all his inflammatory markers including fibrinogen, ferritin, D-dimer, and IL-6 levels normalized. To our knowledge, the use of colchicine in Covid-19 and CRS has not been reported. This observation merits the consideration of colchicine as a safe, inexpensive and oral medication for the treatment of mild to moderate CRS in Covid-19 patients. More importantly, in Covid-19 patients with early lung involvement colchicine may be an appropriate candidate to prevent CRS in adjunction with routine antiviral agents. Indeed, multicenter, randomized controlled studies are required to evaluate the benefits of this therapy.
Collapse
Affiliation(s)
- Nahal Mansouri
- Division of Pulmonary Medicine, Department of Medicine, Lausanne University Hospital (CHUV), University of Lausanne
(UNIL), Lausanne, Switzerland
- Department of Clinical Immunology and Infectious Diseases,
National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Marjani
- The Clinical Tuberculosis and Epidemiology Research
Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payam Tabarsi
- The Clinical Tuberculosis and Epidemiology Research
Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Christophe von Garnier
- Division of Pulmonary Medicine, Department of Medicine, Lausanne University Hospital (CHUV), University of Lausanne
(UNIL), Lausanne, Switzerland
| | - Davood Mansouri
- Department of Clinical Immunology and Infectious Diseases,
National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- The Clinical Tuberculosis and Epidemiology Research
Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Pediatric Respiratory Diseases Research Center, National
Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
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
|
31
|
Gualberto Cardoso PR, Diniz Lopes Marques C, de Melo Vilar K, Dantas AT, Branco Pinto Duarte AL, Pitta IDR, Galdino da Rocha Pitta M, Barreto de Melo Rêgo MJ. Interleukin-18 in Brazilian Rheumatoid Arthritis Patients: Can Leflunomide Reduce It? Autoimmune Dis 2021; 2021:6672987. [PMID: 34055402 PMCID: PMC8131162 DOI: 10.1155/2021/6672987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/30/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Rheumatoid arthritis affects about 1% of the world's population. This is a chronic autoimmune disease. It is predominant in females with progressive joint damage. Immune cells are involved, especially Th1/Th17 lymphocytes and their inflammatory cytokines. These proteins have different functions in the immune system, such as IL-16 is a chemotactic factor; IL-18 can activate NFκB transcription producing inflammatory proteins; IL-31 can activate the JAK/STAT pathway which leads to the production of inflammatory factors in chronic diseases; IL-33 promotes IL-16 secretion which causes lymphocyte recruitment, and IL-32 and IL-34 appear to increase TNF secretion by macrophages activation in AR. The aim of this study was to evaluate serum levels of IL-16, IL-18, IL-31, IL-32, IL-33, and IL-34 and compare them with the severity and treatment of RA patients if there are any correlations. METHODS A total of 140 RA patients and 40 healthy donors were recruited from the Department of Rheumatology at Hospital das Clínicas from the Federal University of Pernambuco. 60 AR patients were naïve for any treatment. Serum cytokine levels were determined using an ELISA kit. RESULTS Serum IL-16 (p = 0.0491), IL-18 (p < 0.0001), IL-31 (p = 0.0004), and IL-32 (p = 0.0040) levels were significantly increased in RA patients compared with healthy donors. It was observed that patients using leflunomide had the lowest IL-18 levels, close to controls levels (p = 0.0064). CONCLUSION IL-16, IL-18, IL-31, and IL-32 are increased in the serum of RA patients. IL-18 is at lower levels in those AR who are taking leflunomide as treatment.
Collapse
Affiliation(s)
- Pablo Ramon Gualberto Cardoso
- Laboratory of Immunomodulation and New Therapeutic Approaches (LINAT), Research Group on Immunomodulation and New Therapeutic Approaches Suely Galdino (Nupit SG), Federal University of Pernambuco, Recife, Brazil
| | | | - Kamila de Melo Vilar
- Laboratory of Immunomodulation and New Therapeutic Approaches (LINAT), Research Group on Immunomodulation and New Therapeutic Approaches Suely Galdino (Nupit SG), Federal University of Pernambuco, Recife, Brazil
| | | | | | - Ivan da Rocha Pitta
- Laboratory of Immunomodulation and New Therapeutic Approaches (LINAT), Research Group on Immunomodulation and New Therapeutic Approaches Suely Galdino (Nupit SG), Federal University of Pernambuco, Recife, Brazil
| | - Maira Galdino da Rocha Pitta
- Laboratory of Immunomodulation and New Therapeutic Approaches (LINAT), Research Group on Immunomodulation and New Therapeutic Approaches Suely Galdino (Nupit SG), Federal University of Pernambuco, Recife, Brazil
| | - Moacyr Jesus Barreto de Melo Rêgo
- Laboratory of Immunomodulation and New Therapeutic Approaches (LINAT), Research Group on Immunomodulation and New Therapeutic Approaches Suely Galdino (Nupit SG), Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
32
|
Kurniasari MD, Karwur FF, Rayanti RE, Dharmana E, Rias YA, Chou KR, Tsai HT. Second-Hand Smoke and Its Synergistic Effect with a Body-Mass Index of >24.9 kg/m 2 Increase the Risk of Gout Arthritis in Indonesia. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:4324. [PMID: 33921811 PMCID: PMC8073587 DOI: 10.3390/ijerph18084324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/16/2022]
Abstract
To analyze the association between smoking status (active smoking and exposure to Second-Hand Smoking (SHS)) and the synergistic effect of smoking status and BMI with gout risk, a community-based case-control design was undertaken among 385 participants, including 304 healthy controls and 81 gout patients from seven community health services. Adjusted Odd Ratios (AORs) and 95% Confidence Interval (CIs) of gout for active smoking and SHS were 3.26 (95% CI = 1.07~9.90) and 4.67 (95% CI = 2.18~10.00) compared to non-smokers. Time-dependent manner of active smoking and SHS significantly increased gout risk with AORs and 95% CIs of 5.95 (1.41~25.03) and 10.12 (3.51~29.14). Dose-dependency of active smokers and SHS showed AORs and 95% CIs of 5.15 (1.28~20.63) and 4.37 (1.33~14.28). Smoking 20 cigarettes (one pack) per day for one year is equivalent to one pack-year. Active smoking >20 pack-year and SHS > 26.5 pack-year increased gout risk with AORs and 95% CIs of 7.18 (1.53~33.67) and 9.95 (3.64~27.22). Participants who smoked (active smoking and SHS) and with Body Mass Index (BMI) of > 24.9 kg/m2 synergistically increased gout risk, with an AOR of 9.65 and 95% CI of 3.25~28.65, compared to BMI ≤ 24.9 kg/m2 and non-smoker. Smoking status (active smoking and SHS) and the synergistic effect of smoking status and BMI increased gout risk in Indonesia.
Collapse
Affiliation(s)
- Maria Dyah Kurniasari
- School of Nursing, College of Nursing, Taipei Medical University, Wu-Xing Street, No 250, Taipei City 11031, Taiwan; (M.D.K.); (K.R.C.)
- Department of Nursing, Faculty of Medicine and Health Sciences, Universitas Kristen Satya Wacana, Diponegoro Street, No 52-60, Salatiga City 50711, Indonesia;
| | - Ferry Fredy Karwur
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universitas Kristen Satya Wacana, Diponegoro Street, No 52-60, Salatiga City 50711, Indonesia;
| | - Rosiana Eva Rayanti
- Department of Nursing, Faculty of Medicine and Health Sciences, Universitas Kristen Satya Wacana, Diponegoro Street, No 52-60, Salatiga City 50711, Indonesia;
| | - Edi Dharmana
- Faculty of Medicine, Universitas Diponegoro, Prof. Sudarto Street, No.13, Semarang City 50275, Indonesia;
| | - Yohanes Andy Rias
- Faculty of Health and Medicine, Institut Ilmu Kesehatan Bhakti Wiyata Kediri, College of Nursing, KH Wachid Hasyim Street, No.65, Kediri City 64114, Indonesia;
| | - Kuei Ru Chou
- School of Nursing, College of Nursing, Taipei Medical University, Wu-Xing Street, No 250, Taipei City 11031, Taiwan; (M.D.K.); (K.R.C.)
- Center for Nursing and Healthcare Research in Clinical Practice Application, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Department of Nursing, Taipei Medical University-Shuang Ho Hospital, Taipei 23561, Taiwan
- Psychiatric Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Hsiu-Ting Tsai
- School of Nursing, College of Nursing, Taipei Medical University, Wu-Xing Street, No 250, Taipei City 11031, Taiwan; (M.D.K.); (K.R.C.)
- Post-Baccalaureate Program in Nursing, College of Nursing, Taipei Medical University, Wu-Xing Street, No 250, Taipei 11031, Taiwan
| |
Collapse
|
33
|
Ding L, Li H, Sun B, Wang T, Meng S, Huang Q, Hong X, Liu D. Elevated interleukin-37 associated with tophus and pro-inflammatory mediators in Chinese gout patients. Cytokine 2021; 141:155468. [PMID: 33647713 DOI: 10.1016/j.cyto.2021.155468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Interleukin-37(IL-37), a natural inhibitor of innate immunity, has been identified to protect against various inflammatory diseases, including monosodium urate (MSU)-induced inflammation. However, the association of IL-37 with clinical indexes and pro-inflammatory mediators in gout patients remains unclear. The aim of this study was to determine IL-37 level in hyperuricemia and gout patients with or without tophus, and to investigate the correlations of IL-37 with clinical indexs such as Uric Acid (UA), CRP(C-reactive protein), Creatinine Clearance Rate (Ccr), Erythrocyte Sedimentation Rate (ESR) and so on, as well as with the pro-inflammatory mediators in serum including Interleukin-1β(IL-1β), Interleukin-6(IL-6) and Interleukin-18(IL-18) from gout patients. METHODOLOGY The serum levels of IL-37, IL-1β, IL-6 and IL-18 levels in serum of gout patients were determined by ELISA; the correlations between IL-37 and clinical values or pro-inflammatory mediators in serum of gout were analyzed by Spearman correlation test. RESULTS The serum levels of IL-37 were higher in active gout patients than inactive gout patients and HCs, especially in active gout patients with tophus. No significant difference was observed in serum IL-37 levels between hyperuricemia and normal controls. IL-1β, IL-6 and IL-18 levels were significant elevated in gout patients with tophus than those without tophus; Serum IL-37 were positively correlated with CRP and ESR, as well as with IL-1β, IL-6 and IL-18, negatively correlated with Ccr, and not correlated with UA, creatinine (Cr) and triglyceride (TG) in gout patients. CONCLUSIONS IL-37 increased in gout patients positively associated CRP and ESR, as well as with proinflammatory mediators IL-1β, IL-6, IL-18, the presence of tophus and chronic kidney disease in gout. It may be a novel marker for predicting this pathology.
Collapse
Affiliation(s)
- Liping Ding
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Heng Li
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Baodong Sun
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Tingting Wang
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Shuhui Meng
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Qin Huang
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Xiaoping Hong
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China.
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China.
| |
Collapse
|
34
|
Cho YN, Jeong HS, Park KJ, Kim HS, Kim EH, Jin HM, Jung HJ, Ju JK, Choi SE, Kang JH, Park DJ, Kim TJ, Lee SS, Kee SJ, Park YW. Altered distribution and enhanced osteoclastogenesis of mucosal-associated invariant T cells in gouty arthritis. Rheumatology (Oxford) 2021; 59:2124-2134. [PMID: 32087015 DOI: 10.1093/rheumatology/keaa020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 01/08/2020] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE This study was designed to investigate the role of mucosal-associated invariant T (MAIT) cells in gouty arthritis (GA) and their effects on osteoclastogenesis. METHODS Patients with GA (n = 61), subjects with hyperuricaemia (n = 11) and healthy controls (n = 30) were enrolled in this study. MAIT cells, cytokines, CD69, programmed death-1 (PD-1) and lymphocyte-activation gene 3 (LAG-3) levels were measured by flow cytometry. In vitro osteoclastogenesis experiments were performed using peripheral blood mononuclear cells in the presence of M-CSF and RANK ligand. RESULTS Circulating MAIT cell levels were significantly reduced in GA patients. However, their capacities for IFN-γ, IL-17 and TNF-α production were preserved. Expression levels of CD69, PD-1 and LAG-3 in MAIT cells were found to be elevated in GA patients. In particular, CD69 expression in circulating MAIT cells was increased by stimulation with MSU crystals, suggesting that deposition of MSU crystals might contribute to MAIT cell activation. Interestingly, MAIT cells were found to be accumulated in synovial fluid and infiltrated into gouty tophus tissues within joints. Furthermore, activated MAIT cells secreted pro-resorptive cytokines (i.e. IL-6, IL-17 and TNF-α) and facilitated osteoclastogenesis. CONCLUSION This study demonstrates that circulating MAIT cells are activated and numerically deficient in GA patients. In addition, MAIT cells have the potential to migrate to inflamed tissues and induce osteoclastogenesis. These findings provide an important role of MAIT cells in the pathogenesis of inflammation and bone destruction in GA patients.
Collapse
Affiliation(s)
- Young-Nan Cho
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hae-Seong Jeong
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Ki-Jeong Park
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic MedicineChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Eun-Hee Kim
- Department of Forensic MedicineChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hye-Mi Jin
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hyun-Ju Jung
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Jae Kyun Ju
- Department of Surgery, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Sung-Eun Choi
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Ji-Hyoun Kang
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Dong-Jin Park
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Tae-Jong Kim
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Shin-Seok Lee
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Yong-Wook Park
- Department of RheumatologyChonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| |
Collapse
|
35
|
Kondratiuk VE, Tarasenko OM, Karmazina OM, Taranchuk VV. Impact of the Synbiotics and Urate-Lowering Therapy on Gut Microbiota and Cytokine Profile in Patients with Chronic Gouty Arthritis. J Med Life 2021; 13:490-498. [PMID: 33456597 PMCID: PMC7803318 DOI: 10.25122/jml-2020-0065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The main goal of our study is the impact evaluation of complex urate-lowering therapy with the synbiotic addition on fecal microbiota and cytokine profile in patients with primary gout. During our study, 130 men (mean age 55.5 ± 9.4 years) with gout (duration 7.7 ± 7.1 years) were examined. All patients were divided into two treatment groups. The main group (n = 68) was taking allopurinol at 300 mg per day dose and additionally a synbiotic. The comparison group (n = 62) received allopurinol monotherapy without synbiotic intake. The therapy duration was 3 months. Evaluation of therapy efficiency was marked by blood uric acid changes, cytokine levels, CRP and fecal microbiota condition. After treatment, stabilization of the gut microbiota parameters was observed, which was leading to normalization uricemia levels (40.3% vs. 21%, p <0.01) in the main group patients. Addition of synbiotic to allopurinol leads to a blood uric acid lowering (18.7% vs. 13.3%, p <0.01), CRP reduction (75% vs. 26.3%, p <0.01) as well as decrease of cytokines level: IL-1β, IL-6, IL-8, IL-10 and TNFα (all p <0.001). After a 3-month gout treatment, a group of patients who received complex therapy with synbiotic inclusion showed signs of disease remission characterized by inflammation activity reducing, fecal microbiota condition normalization and a more pronounced decrease in laboratory markers comparing to control group.
Collapse
|
36
|
Chen S, Huang X, Huang Y, Zhao W, Zheng S, Huang Q, Li T. Role of plasma fibrinogen in assessing disease activity of patients with gout. Clin Chim Acta 2020; 510:483-487. [PMID: 32795545 DOI: 10.1016/j.cca.2020.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Gout is an inflammatory disease characterized by the deposition of monosodium urate (MSU) in synovial fluid and other tissues. Many studies have shown that the activation of coagulation system had been proposed correlated with systemic inflammation. The concentrations of plasma fibrinogen and D-dimer are increased in abnormal coagulation, emerging as available indicators to predict systemic inflammation. The aim of this study is to reveal the predictive value of plasma fibrinogen, D-dimer in the disease activity of gout patients. METHODS This retrospective study included 334 gout patients and 101 age- and gender- matched healthy controls. The gout patients were divided into two groups according to the gout activity score (GAS = 0.09 × last 12 month attacks + 1.01 × sUA + 0.34 × VAS patient + 0.53 × ln(1 + tophi number). The remission group included 46 patients with GAS of lower than 2.5 and the active group included 288 patients with GAS of 2.5 or higher. Clinical and laboratory data were recorded. The correlations between plasma fibrinogen, D-dimer and GAS were analyzed by Spearman's correlation analysis and Partial correlation analysis. Receiver operating characteristic (ROC) was used to evaluate the diagnostic value for the active group compared with remission group. The predictive value of fibrinogen, D-dimer to the disease activity of gout patients was tested by Binary logistic regression analysis. RESULTS Plasma fibrinogen and D-dimer in gout patients (3.66 (2.88, 5.20), 0.29 (0.22, 0.80)) were increased as compared with the control group (2.88 (2.51, 3.24), 0.22 (0.22, 0.32), both P < 0.001). Fibrinogen and D-dimer in active group (3.91 (3.00, 5.53), 0.34 (0.22, 0.86)) were higher than those in remission group (2.88 (2.34, 3.22), 0.22 (0.22, 0.26), both P < 0.001)). Plasma fibrinogen, D-dimer, ESR and CRP were positively correlated with GAS (r = 0.606, r = 0.419, r = 0.570, r = 0.440, all P < 0.001). ROC curve showed fibrinogen yielded a highest AUC than D-dimer, ESR, CRP. In addition, the optimal cutoff value of fibrinogen for active group was 3.60, with a specificity of 89.1% and sensitivity of 58.3%. Binary logistic regression analysis showed fibrinogen (odds ratio = 2.71, 95% confidence interval: 1.28-5.74, p = 0.011) was a predictor for gout disease activity. CONCLUSION Fibrinogen was increased in active gout group. Fibrinogen can serve as a reliable inflammatory marker for monitoring inflammatory response and disease activity in gout patients.
Collapse
Affiliation(s)
- Shuyang Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guangdong Province, China
| | - Xuechan Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guangdong Province, China
| | - Yukai Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guangdong Province, China
| | - Wenkai Zhao
- University of South China, Hengyang 421000, Hunan Province, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guangdong Province, China
| | - Shaoling Zheng
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guangdong Province, China
| | - Qidang Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guangdong Province, China
| | - Tianwang Li
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou 510317, Guangdong Province, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China.
| |
Collapse
|
37
|
Klück V, Liu R, Joosten LAB. The role of interleukin-1 family members in hyperuricemia and gout. Joint Bone Spine 2020; 88:105092. [PMID: 33129923 DOI: 10.1016/j.jbspin.2020.105092] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Interleukin (IL)-1 family cytokines and their receptors have important roles in innate and partly in adaptive immunity. The family consists of 11 members of which IL-1α, IL-1β, IL-18, IL-33, IL-36α, IL-36β and IL-36γ are considered pro-inflammatory and IL-1Ra, IL-36Ra, IL-37 and IL-38 anti-inflammatory. Whereas IL-1β has a known pivotal role in gout, increasing evidence suggests other IL-1 family members are also involved in the pathogenesis of hyperuricemia and gout flares. FINDINGS Studies indicate IL-1α, like IL-1β, plays an essential role in the pathogenesis of gout flares. IL-18, although elevated in patients with gout, does not contribute to MSU crystal-induced inflammation, but may be involved in the subsequent development of cardiovascular disease in individuals with gout. The role of the pro-inflammatory cytokine IL-36 in gout remains elusive. In contrast, IL-1Ra, IL-33, IL-37 and IL-38 inhibit MSU crystal-induced inflammation and therefore have therapeutic potential for treatment of gout flares. In addition to existing IL-1β blockers, several new therapeutics to treat gout are being developed either inhibiting the transcription or maturation of IL-1β. CONCLUSION In this review, IL-1 family cytokines are discussed in the context of hyperuricemia and gout. Finally, current and novel therapeutic options for targeting IL-1 are reviewed.
Collapse
Affiliation(s)
- Viola Klück
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands.
| | - Ruiqi Liu
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands.
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Street Pasteur nr. 6, 400349 Cluj-Napoca, Romania.
| |
Collapse
|
38
|
Shiels RG, Hewage W, Pennell EN, Vidimce J, Grant G, Pearson AG, Wagner KH, Morgan M, Bulmer AC. Biliverdin and bilirubin sulfonate inhibit monosodium urate induced sterile inflammation in the rat. Eur J Pharm Sci 2020; 155:105546. [PMID: 32927072 DOI: 10.1016/j.ejps.2020.105546] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 08/07/2020] [Accepted: 08/31/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Biliverdin, a by-product of haem catabolism, possesses potent endogenous antioxidant and anti-inflammatory properties. Bilirubin-C10-sulfonate (BRS), an active metabolite formed after enteral administration of BV in the rat, also possess antioxidant properties. Therefore, we investigated the anti-inflammatory and antioxidant activity of BV and BRS in an in vivo model of monosodium urate induced sterile inflammation. METHODS Subcutaneous air pouches were created on the dorsal flanks of Wistar rats (10-12 weeks of age). Prior to stimulation of the 6-day old pouch with monosodium urate (25 mg), groups were pre-treated with intraperitoneal BRS (27 mg/kg) and BV (27 mg/kg). Total and differential leukocyte counts were determined in pouch fluid aspirate at 1, 6, 12, 24 and 48 h after monosodium urate stimulation. Biliverdin (BV), BRS and unconjugated bilirubin (UCB) concentrations in the serum and pouch fluid were quantified using liquid chromatography-mass spectrometry. Pouch fluid cytokine concentrations (IL-1β, IL-1α, TNF-α, IL-17A, IL-12, GM-CSF, IL-33, IFN-γ, IL-18, IL-10, MCP-1, CXCL-1 and IL-6) were assessed after 6 h. In addition, 24 h protein carbonyl and chloramine concentrations were assessed in pouch fluid using ELISA and spectrophotometry, respectively. RESULTS BRS and BV significantly (p < 0.05) inhibited leukocyte (total, neutrophil and macrophage) infiltration into the pouch fluid from 6 to 48 h. For example, after 6 h neutrophil counts decreased following BRS (0.32 ± 0.11 × 106 cells mL-1) and BV (0.17 ± 0.03 × 106 cells mL-1) compared to MSU only (3.51 ± 1.07 × 106 cells mL-1). Both BV and BRS significantly (p < 0.05) reduced pouch GM-CSF (BV: 5.8 ± 1.2 pg mL-1, BRS: 6.9 ± 1.5 pg mL-1 vs MSU only: 13.0 ± 1.9 pg mL-1) and MCP-1 concentrations at 6 h (BV: 1804 ± 269 pg mL-1, BRS: 7927 ± 2668 pg mL-1 vs MSU only: 17,290 ± 4503 pg ml-1), whilst BV additionally inhibited IL-6 (4354 ± 977 pg mL-1 vs MSU only: 25,070 ± 5178 pg mL-1) and IL-18 (17.6 ± 2.0 pg mL-1 vs MSU only: 81.5 ± 19.9 pg mL-1) concentrations at 6 h (p < 0.05). Despite these differences, no change in pouch chloramine or protein carbonyl concentrations occurred at 24 h (p > 0.05). Serum BV concentrations rapidly diminished over 6 h, however, BRS was readily detected in the serum over 48 h, and in pouch fluid over 12 h. CONCLUSIONS This study is the first to elucidate anti-inflammatory activity of BRS and the efficacy of BV administration in a model of gouty inflammation. Reduced leukocyte infiltration and cytokine production in response to sterile inflammation further support the importance of these molecules in physiology and their therapeutic potential in sterile inflammation.
Collapse
Affiliation(s)
- Ryan G Shiels
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Wenu Hewage
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Evan N Pennell
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Josif Vidimce
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Gary Grant
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Andrew G Pearson
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Karl-Heinz Wagner
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Michael Morgan
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew C Bulmer
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia; Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
39
|
Singh M, Jadeja SD, Vaishnav J, Mansuri MS, Shah C, Mayatra JM, Shah A, Begum R. Investigation of the Role of Interleukin 6 in Vitiligo Pathogenesis. Immunol Invest 2020; 51:120-137. [PMID: 32865069 DOI: 10.1080/08820139.2020.1813756] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Interleukin-6 (IL6) is involved in pathogenesis of several autoimmune disorders including vitiligo. Hence, we aimed to investigate the association of IL6 -174 G/C and -572 G/C polymorphisms and its transcript levels with vitiligo; to evaluate the effect of IL-6 on normal human melanocyte (NHM) viability and expression of IL6R, MITF and TYR. IL6 -174 G/C and -572 G/C polymorphisms were genotyped by ARMS-PCR and PCR-RFLP respectively in 343 controls and 322 vitiligo patients. IL6 transcript levels were estimated from PBMCs (96 controls and 77 patients) and skin samples (15 controls and 15 patients) by qPCR. NHM viability was assessed by MTT; IL6R, MITF and TYR transcript and protein levels were monitored by qPCR and ICC respectively. Genetic analyses revealed no association of IL6 -174 G/C polymorphism (p> .05) with vitiligo. Analysis of IL6 -572 G/C revealed reduced risk of vitiligo in individuals with GC/CC genotypes compared to GG genotype (p = .010). IL6 expression was significantly increased (p = .0197) in PBMCs of patients. Further, IL6 expression was significantly higher in non-lesional skin compared to controls (p = .009). In-vitro NHM viability was decreased upon IL-6 exposure (10-50 ng/ml; p< .05), with significantly increased IL6R transcript (p = .042) and protein levels (p = .003) however, MITF transcript (p = .0003) and protein levels (p = .016), and TYR transcript levels (p = .001) were significantly decreased. The results suggest that IL6 -572 G/C polymorphism might be associated with vitiligo susceptibility in Gujarat population. Moreover, increased IL6 expression in vitiligo patients and its effect on NHM suggest a potential role in melanocyte biology. CONCLUSION The results suggest that IL6 - 572 G/C polymorphism might be associated with vitiligo susceptibility in Gujarat population. Moreover, increased IL6 expression in vitiligo patients and its effect on NHM suggest a potential role in melanocyte biology.
Collapse
Affiliation(s)
- Mala Singh
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, Vadodara, India
| | - Shahnawaz D Jadeja
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, Vadodara, India
| | - Jayvadan Vaishnav
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, Vadodara, India
| | - Mohmmad Shoab Mansuri
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, Vadodara, India
| | - Chandni Shah
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, Vadodara, India
| | - Jay M Mayatra
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, Vadodara, India
| | - Atul Shah
- Department of Reconstructive Surgery, Solace Hospital, Vadodara, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, Vadodara, India
| |
Collapse
|
40
|
El Sebaei MG, Arafat N, El-Shafei RA, El-Adl MA, Farag A, Aziza AE, Eladl AH. Biochemical and molecular investigation of oxidative stress associated with urolithiasis induced by increased dietary calcium or protein in chickens. J Anim Physiol Anim Nutr (Berl) 2020; 105:129-139. [PMID: 32790029 DOI: 10.1111/jpn.13436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 07/02/2020] [Accepted: 07/11/2020] [Indexed: 01/10/2023]
Abstract
This study was carried out to evaluate the effects of induced urolithiasis by high dietary calcium (Ca) or protein levels on biochemical analyte levels, redox status, selected inflammatory cytokines and histopathology in chickens. A total of 90 one-day-old white Hy-Line chicks were fed basal control diets containing 20% crude protein (CP) and 1% Ca until they reached 44 days of age. After that, the birds were divided into three groups (30 birds per group). All management factors (light, temperature, ventilation, stock density and diet) were identical among the three groups throughout the study except for the dietary Ca and protein percentages. Group I was fed a control diet containing 20% CP and 1% Ca, group II was fed a high-Ca diet containing 5% Ca, and group III was fed a high-protein diet containing 25% CP. Our findings clearly demonstrated that dietary imbalance (caused by high-Ca or high-CP levels) per se in chickens was physiologically harmful, as it was accompanied by post-mortem lesions; biochemical, redox status and histopathological alterations; and upregulation of inflammatory cytokines (interleukin (IL)-1β and IL-6). In particular, the birds fed the high-Ca diet clearly exhibited the most obvious alterations in most of the endpoints. In conclusion, this study constitutes the first extensive investigation of the effects of high-Ca or high-protein diets induced urolithiasis on growth performance, redox status, inflammatory cytokine levels and pathological characterization in chickens.
Collapse
Affiliation(s)
- Mahmoud G El Sebaei
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia.,Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Nagah Arafat
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Reham A El-Shafei
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed A El-Adl
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Amany Farag
- Department of Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Abeer E Aziza
- Department of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Abdelfattah H Eladl
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
41
|
Cyr61 Promotes Inflammation of a Gouty Arthritis Model in Rats. Mediators Inflamm 2020; 2020:8298615. [PMID: 32774151 PMCID: PMC7396108 DOI: 10.1155/2020/8298615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/06/2020] [Indexed: 11/17/2022] Open
Abstract
Background Cyr61 is considered a novel proinflammatory factor. Gouty arthritis (GA) is a self-limited inflammatory reaction caused by monosodium urate (MSU) crystals. In this study, we assessed the role of Cyr61 in the inflammatory process of GA. Methods We investigated the expression of Cyr61 in MSU-induced rat gout models and MSU-stimulated rat fibroblast-like synovial (FLS) cells. After inhibiting the expression of Cyr61, levels of IL-1β, TNF-α, and IL-6 were detected by ELISA, qPCR, western blot, and immunohistochemical methods. We probed the downstream NF-κB signaling pathway using the NF-κB inhibitor PDTC, and levels of NF-κB and p-NF-κB were detected by western blot and qPCR. Results Our results demonstrate that Cyr61 plays a potent role in the formation of local inflammation in vitro and in vivo. Cyr61 was highly expressed in synovial tissues of gout models, and the expression of Cyr61 protein was also significantly increased in MSU-stimulated FLS cells. Cyr61 promoted MSU-induced acute inflammation via the NF-κB signaling pathway. Conclusions Our study has revealed that Cyr61 is an important regulatory factor for the initiation of inflammation in GA. The high expression of Cyr61 protein can induce synovial cells to produce many inflammatory cytokines, such as IL-1β, TNF-α, and IL-6, partly in an NF-κB-dependent manner. Thus, inhibition of Cyr61 could be a new target and strategy for the prevention and treatment of GA.
Collapse
|
42
|
Chuang JP, Kao CY, Lee JC, Ling P, Maa MC, Leu TH. EPS8 regulates an NLRP3 inflammasome-independent caspase-1 activation pathway in monosodium urate crystal-treated RAW264.7 macrophages. Biochem Biophys Res Commun 2020; 530:487-493. [PMID: 32595041 DOI: 10.1016/j.bbrc.2020.05.084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/12/2020] [Indexed: 11/19/2022]
Abstract
Gout is an inflammatory arthritis caused by the phagocytosis of monosodium urate (MSU) crystal deposition in joints. NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome-dependent caspase-1 activation is implicated in the processing of interleukin-1β (IL-1β), which is the major effector cytokine in the acute inflammatory response of gout. Mechanisms underlying caspase-1 activation remain unclear. Epidermal growth factor receptor pathway substrate no. 8 (Eps8) is a signal transducer and actin filament organizer that plays a key role in lipopolysaccharide-stimulated phagocytosis in macrophages. Here, RAW264.7 macrophages that have no intact NLRP3 inflammasomes were used to investigate the role of Eps8 in MSU crystal-mediated caspase-1 activation. A kinetic study revealed that the induction of Eps8 expression by MSU crystals occurred before NLRP3, p46/p33 caspase-1, and mature IL-1β in RAW 264.7 cells. In addition, actin cytoskeleton dynamics was required for Eps8 induction and caspase-1 activation in MSU crystal stimulation. Silencing Eps8 had no effect on the basal expression of p46/p33 caspase-1 and NLRP3, but nearly abolished MSU crystal-induced NLRP3 expression and caspase-1 activation. Furthermore, MSU crystals induced Eps8-pro-caspase-1 complex formation and Eps8 formed a stable complex with p33 caspase-1, but not with NLRP3. In summary, our results demonstrated for the first time the importance of Eps8 in MSU crystal-mediated caspase-1 activation without the involvement of NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Jen-Pin Chuang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Department of Surgery, Puzi Hospital, Ministry of Health and Welfare, Chiayi, Taiwan, ROC
| | - Chuan-Yu Kao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Jenq-Chang Lee
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Pin Ling
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Ming-Chei Maa
- Department of Biochemistry, College of Medicine, China Medical University, Taichung, 40402, Taiwan, ROC.
| | - Tzeng-Horng Leu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
| |
Collapse
|
43
|
Klück V, Jansen TLTA, Janssen M, Comarniceanu A, Efdé M, Tengesdal IW, Schraa K, Cleophas MCP, Scribner CL, Skouras DB, Marchetti C, Dinarello CA, Joosten LAB. Dapansutrile, an oral selective NLRP3 inflammasome inhibitor, for treatment of gout flares: an open-label, dose-adaptive, proof-of-concept, phase 2a trial. THE LANCET. RHEUMATOLOGY 2020; 2:e270-e280. [PMID: 33005902 PMCID: PMC7523621 DOI: 10.1016/s2665-9913(20)30065-5] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
SUMMARY BACKGROUND Gout flares are driven by interleukin (IL)-1β. Dapansutrile inhibits the NLRP3 inflammasome and subsequent activation of IL-1β. In this study we aimed to investigate the safety and efficacy of orally administered dapansutrile in patients with a gout flare. METHODS In this open-label, proof-of-concept, phase 2a trial, adult patients (aged 18-80 years) with a monoarticular monosodium urate crystal-proven gout flare were enrolled at an outpatient clinic in the Netherlands and sequentially assigned using a dose-adaptive design to receive 100 mg/day, 300 mg/day, 1000 mg/day, or 2000 mg/day oral dapansutrile for 8 days. The coprimary outcomes were change in patient-reported target joint pain from baseline to day 3 and from baseline to day 7, assessed in the per-protocol population (all patients who received at least 80% of the study drug and had no major protocol deviations). Safety was assessed in the intention-to-treat population. This trial is registered with the EU Clinical Trials Register, EudraCT 2016-000943-14, and is completed. FINDINGS Between May 18, 2017, and Jan 21, 2019, 144 patients were assessed for eligibility, of whom 34 were enrolled and 29 were included in the per-protocol population (three patients were excluded due to receiving <80% of study drug and two had major protocol deviations): eight patients received 100 mg/day, seven received 300 mg/day, six received 1000 mg/day, and eight received 2000 mg/day. Between baseline and day 3, there was a mean reduction in patient-reported target joint pain of 52·4% (SD 32·94; p=0∙016) for the 100 mg/day group, 68·4% (34·29; p=0∙016) for the 300 mg/day group, 55·8% (44·90; p=0∙063) for the 1000 mg/day group, and 57·6% (38·72; p=0∙016) for the 2000 mg/day group. At day 7, there was a mean reduction of 82·1% (22·68; p=0∙031) for the 100 mg/day group, 84·2% (16·33; p=0∙016) for the 300 mg/day group, 68·9% (34·89; p=0∙031) for the 1000 mg/day group, and 83·9% (15·44; p=0∙008) for the 2000 mg/day group, compared to baseline. 25 (73·5%) of 34 patients reported a total of 45 treatment-emergent adverse events, most of which were metabolism and nutrition disorders (17 [37·8%]) and gastrointestinal disorders (ten [22·2%]). Two serious adverse events occurred during the study, admission to hospital because of worsening of gout flare at day 3, and admission to hospital because of coronary stenosis 18 days after the patient received their last dose; these were considered moderate in severity and unrelated to the study drug. INTERPRETATION Dapansutrile is a specific NLRP3 inflammasome inhibitor with a satisfactory safety profile and efficacy in the reduction of target joint pain in this study. Future studies are needed to confirm the clinical potential of dapansutrile.
Collapse
Affiliation(s)
| | | | - Matthijs Janssen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Antoaneta Comarniceanu
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Monique Efdé
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Isak W Tengesdal
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Kiki Schraa
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Maartje C P Cleophas
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Curtis L Scribner
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Damaris B Skouras
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Carlo Marchetti
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Charles A Dinarello
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| |
Collapse
|
44
|
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
|
45
|
Temmoku J, Fujita Y, Matsuoka N, Urano T, Furuya MY, Asano T, Sato S, Matsumoto H, Watanabe H, Kozuru H, Yatsuhashi H, Kawakami A, Migita K. Uric acid-mediated inflammasome activation in IL-6 primed innate immune cells is regulated by baricitinib. Mod Rheumatol 2020; 31:270-275. [PMID: 32148148 DOI: 10.1080/14397595.2020.1740410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Gout is an inflammatory arthropathy caused by the deposition of monosodium urate (MSU). The synthesis and release of IL-1β is crucial for MSU-induced synovial inflammation. The aim of the present study was to investigate the mechanism of MSU crystal-induced autoinflammatory processes. METHODS In vitro studies were used to evaluate the role of IL-6 in inflammasome activation in human neutrophils cultured with MSU crystals. Human neutrophils were stimulated with MSU in the presence or absence of IL-6 priming to determine NLRP3 inflammasome activation and subsequent cleaved caspase-1 induction or IL-1β production. RESULTS IL-6 or MSU stimulation alone did not result in the efficient IL-1β production from human neutrophils. However, MSU stimulation induced marked IL-1β production from IL-6-primed neutrophils. Pretreatment with baricitinib, which blocks IL-6 receptor signaling, prevented MSU-induced cleaved caspase-1 or IL-1β induction in IL-6-primed neutrophils. Tocilizumab pretreatment also inhibited MSU-mediated IL-1β production from IL-6-primed neutrophils. CONCLUSION Priming of human neutrophils with IL-6 promotes uric acid-mediated IL-1β secretion in the absence of microbial stimulation. These results suggest that an endogenous cytokine, IL-6, is involved in MSU-mediated NLRP3 inflammasome activation and subsequent IL-1β production from innate immune cells and has a crucial role in MSU crystal-induced synovial inflammation. These findings provide insights into uric acid-mediated autoinflammation in the innate immune system.
Collapse
Affiliation(s)
- Jumpei Temmoku
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Naoki Matsuoka
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takeshi Urano
- Department of Biochemistry, Shimane University School of Medicine, Izumo, Japan
| | - Makiko Yashiro Furuya
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Haruki Matsumoto
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroshi Watanabe
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hideko Kozuru
- Clinical Research Center, NHO Nagasaki Medical Center, Nagasaki, Japan
| | | | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan.,Clinical Research Center, NHO Nagasaki Medical Center, Nagasaki, Japan
| |
Collapse
|
46
|
Spel L, Martinon F. Inflammasomes contributing to inflammation in arthritis. Immunol Rev 2020; 294:48-62. [DOI: 10.1111/imr.12839] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Lotte Spel
- Departement of Biochemistry University of Lausanne Epalinges Switzerland
| | - Fabio Martinon
- Departement of Biochemistry University of Lausanne Epalinges Switzerland
| |
Collapse
|
47
|
Salandari S, Shomali T, Mosleh N, Nazifi S. A comparative study on anti-inflammatory drug combinations in domestic pigeons with experimentally induced acute arthritis. Acta Vet Hung 2019; 67:588-601. [PMID: 31842592 DOI: 10.1556/004.2019.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The study compares the effect of one-time administration of nonsteroidal and/or steroidal anti-inflammatory combinations by topical or intramuscular (IM) routes to pigeons with monosodium urate (MSU)-induced arthritis. Forty-five adult domestic pigeons were assigned into nine equal groups: NC, negative control; PC, positive control with arthritis; sham, sham control; T1, meloxicam + hydrocortisone; T2, dexamethasone + piroxicam; T3, meloxicam + dexamethasone; T4, hydrocortisone + piroxicam; T5, dexamethasone + hydrocortisone; T6, meloxicam + piroxicam. Arthritis was also induced in T1 to T6 birds. Meloxicam and dexamethasone were administered by IM injection and the other drugs topically right after the induction of arthritis. Different drug combinations significantly decreased one-leg standing time. Induction of arthritis significantly increased TNF-α and IL-6 levels in synovial fluid and serum corticosterone and epinephrine in the PC group. Administration of drugs to birds of Groups T1 and T5 did not significantly change corticosterone concentration, while all different drug combinations decreased epinephrine level. Drug combinations that demonstrated better analgesic effect more strongly reduced serum epinephrine concentration. Meloxicam + hydrocortisone was the most effective combination in reducing inflammatory cytokines. In conclusion, one-time combination therapy with anti-inflammatory agents was effective in the acute management of inflammatory pain due to MSU-induced arthritis in pigeons, even by the topical route.
Collapse
Affiliation(s)
- Sajedeh Salandari
- 1Division of Pharmacology and Toxicology, Department of Basic Sciences and School of Veterinary Medicine, Shiraz University, P.O. Box 71441-69155, Shiraz, Iran
| | - Tahoora Shomali
- 1Division of Pharmacology and Toxicology, Department of Basic Sciences and School of Veterinary Medicine, Shiraz University, P.O. Box 71441-69155, Shiraz, Iran
| | - Najmeh Mosleh
- 2Department of Clinical Studies, School of Veterinary Medicine, Shiraz University, P.O. Box 71441-69155, Shiraz, Iran
| | - Saeed Nazifi
- 2Department of Clinical Studies, School of Veterinary Medicine, Shiraz University, P.O. Box 71441-69155, Shiraz, Iran
| |
Collapse
|
48
|
Interaction of the p.Q141K Variant of the ABCG2 Gene with Clinical Data and Cytokine Levels in Primary Hyperuricemia and Gout. J Clin Med 2019; 8:jcm8111965. [PMID: 31739430 PMCID: PMC6912417 DOI: 10.3390/jcm8111965] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/09/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Gout is an inflammatory arthritis influenced by environmental risk factors and genetic variants. The common dysfunctional p.Q141K allele of the ABCG2 gene affects gout development. We sought after the possible association between the p.Q141K variant and gout risk factors, biochemical, and clinical determinants in hyperuricemic, gouty, and acute gouty arthritis cohorts. Further, we studied the correlation of p.Q141K allele and levels of pro-/anti-inflammatory cytokines. Coding regions of the ABCG2 gene were analyzed in 70 primary hyperuricemic, 182 gout patients, and 132 normouricemic individuals. Their genotypes were compared with demographic and clinical parameters. Plasma levels of 27 cytokines were determined using a human multiplex cytokine assay. The p.Q141K variant was observed in younger hyperuricemic/gout individuals (p = 0.0003), which was associated with earlier disease onset (p = 0.004), trend toward lower BMI (p = 0.056), and C-reactive protein (CRP, p = 0.007) but a higher glomerular filtration rate (GFR, p = 0.035). Levels of 19 cytokines were higher, mainly in patients with acute gouty arthritis (p < 0.001), irrespective of the presence of the p.Q141K variant. The p.Q141K variant influences the age of onset of primary hyperuricemia or gout and other disease-linked risk factors and symptoms. There was no association with cytokine levels in the circulation.
Collapse
|
49
|
Phytochemicals as Novel Therapeutic Strategies for NLRP3 Inflammasome-Related Neurological, Metabolic, and Inflammatory Diseases. Int J Mol Sci 2019; 20:ijms20122876. [PMID: 31200447 PMCID: PMC6627634 DOI: 10.3390/ijms20122876] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
Several lines of evidence point out the relevance of nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome as a pivotal player in the pathophysiology of several neurological and psychiatric diseases (i.e., Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), amyotrophic lateral sclerosis, and major depressive disorder), metabolic disorders (i.e., obesity and type 2 diabetes) and chronic inflammatory diseases (i.e., intestinal inflammation, arthritis, and gout). Intensive research efforts are being made to achieve an integrated view about the pathophysiological role of NLRP3 inflammasome pathways in such disorders. Evidence is also emerging that the pharmacological modulation of NLRP3 inflammasome by phytochemicals could represent a promising molecular target for the therapeutic management of neurological, psychiatric, metabolic, and inflammatory diseases. The present review article has been intended to provide an integrated and critical overview of the available clinical and experimental evidence about the role of NLRP3 inflammasome in the pathophysiology of neurological, psychiatric, metabolic, and inflammatory diseases, including PD, AD, MS, depression, obesity, type 2 diabetes, arthritis, and intestinal inflammation. Special attention has been paid to highlight and critically discuss current scientific evidence on the effects of phytochemicals on NLRP3 inflammasome pathways and their potential in counteracting central neuroinflammation, metabolic alterations, and immune/inflammatory responses in such diseases.
Collapse
|
50
|
Lu X, Zeng R, Lin J, Hu J, Rong Z, Xu W, Liu Z, Zeng W. Pharmacological basis for use of madecassoside in gouty arthritis: anti-inflammatory, anti-hyperuricemic, and NLRP3 inhibition. Immunopharmacol Immunotoxicol 2019; 41:277-284. [PMID: 31084401 DOI: 10.1080/08923973.2019.1590721] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objectives: Gouty arthritis is caused by the deposition of monosodium urate (MSU) crystals in joints, which is associated with the rise of serum urate content. This study aims to investigate the therapeutic effect of Madecassoside on gouty arthritis and hyperuricemia. Methods: DBA/1 mice were intradermally injected with MSU to stimulate joint inflammation or intraperitoneally injected with MSU to trigger peritonitis. Moreover, ICR mice were exposed to potassium oxonate to stimulate hyperuricemia. Results: Madecassoside repressed MSU-triggered pad swelling, joint 99mTc uptake, and joint inflammation in DBA/1 mice with gouty arthritis. Neutrophil infiltration and IL-1β & IL-6 & MCP-1 secretion was also alleviated in lavage fluids from DBA/1 mice with peritonitis due to Madecassoside treatment. Furthermore, Madecassoside decreased MSU-induced neutrophil cytosolic factor 1, caspase-1 and NLRP3 expression in mice with peritoneal inflammation. In hyperuricemic mice, Madecassoside improved renal dysfunction. Serum uric acid, BUN, and creatinine were down-regulated by Madecassoside. Conclusion: These findings indicate that Madecassoside has potential to ameliorate inflammation in both acute gouty arthritis model and peritonitis model, probably via regulating IL-1β and NLRP3 expression. Practical point: Madecassoside also exhibited a urate-lowering effect and a renal protective effect in hyperuricemic mice.
Collapse
Affiliation(s)
- Xiaohui Lu
- a Department of Orthopedic Surgery , The First Affiliate Hospital of Shantou University Medical College , Shantou , China
| | - Runming Zeng
- a Department of Orthopedic Surgery , The First Affiliate Hospital of Shantou University Medical College , Shantou , China
| | - Jing Lin
- b Department of Oncology , The First Affiliate Hospital of Shantou University Medical College , Shantou , China
| | - Jun Hu
- a Department of Orthopedic Surgery , The First Affiliate Hospital of Shantou University Medical College , Shantou , China
| | - Zhijie Rong
- a Department of Orthopedic Surgery , The First Affiliate Hospital of Shantou University Medical College , Shantou , China
| | - Weicai Xu
- a Department of Orthopedic Surgery , The First Affiliate Hospital of Shantou University Medical College , Shantou , China
| | - Zewa Liu
- b Department of Oncology , The First Affiliate Hospital of Shantou University Medical College , Shantou , China
| | - Wanting Zeng
- c Division of Medicine , University College London , London , United Kingdom
| |
Collapse
|