Education, Science, Technology, Innovation and Life
Open Access
Sign In

Research Progress in the Prevention and Treatment of Hyperuricemia with Gout by Traditional Chinese Medicine

Download as PDF

DOI: 10.23977/medcm.2023.050910 | Downloads: 18 | Views: 352

Author(s)

Haoran Li 1, Yun Tian 2

Affiliation(s)

1 Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
2 Shaanxi Provincial Hospital of Chinese Medicine, Xi'an, Shaanxi, 710003, China

Corresponding Author

Yun Tian

ABSTRACT

Hyperuricaemia is a metabolic disorder whose pathogenesis is directly related to the level of uric acid in the blood. In addition, hyperuricaemia is also related to gout, diabetes mellitus and kidney disease, therefore, hyperuricaemia should be taken seriously. Traditional Chinese medicine (TCM) has unique advantages in the treatment of hyperuricaemia. In this paper, the pathogenesis of hyperuricemia and the mechanism of action of TCM in the prevention and treatment of hyperuricemia are reviewed to provide a theoretical basis for the research of anti-hyperuricemia drugs.

KEYWORDS

Hyperuricaemia; pathogenesis; Chinese medicine; mechanism of action

CITE THIS PAPER

Haoran Li, Yun Tian, Research Progress in the Prevention and Treatment of Hyperuricemia with Gout by Traditional Chinese Medicine. MEDS Chinese Medicine (2023) Vol. 5: 79-86. DOI: http://dx.doi.org/10.23977/medcm.2023.050910.

REFERENCES

[1] Su Hongyong, Yang Chen, Liang Dong, et al. Research Advances in the Mechanisms of Hyperuricemia-Induced Renal Injury [J]. BioMed research international, 2020, 2020:5817348.
[2] Tony R Merriman, Nicola Dalbeth. The genetic basis of hyperuricaemia and gout [J]. Joint Bone Spine, 2010, 78(1):35-40.
[3] Rashika El Ridi, Hatem Tallima. Physiological functions and pathogenic potential of uric acid: A review [J]. Journal of Advanced Research, 2017, 8(5):487-493.
[4] JK Maesaka, S Fishbane. Regulation of renal urate excretion: A critical review [J]. American Journal of Kidney Diseases, 1998, 32(6):917-933. [5] Sorensen L B. Role of the intestinal tract in the elimination of uric acid [J]. Arthritis and rheumatism, 1965, 8(5):694-706.
[6] Ichiro Hisatome, Kimiyoshi Ichida, Ikuo Mineo, et al. Japanese Society of Gout and Uric & Nucleic Acids 2019 Guidelines for Management of Hyperuricemia and Gout 3rd edition[J]. Gout Uric Nucleic Acids, 2020, 44:1–40.
[7] Dalbeth N, Merriman T. Crystal ball gazing: new therapeutic targets for hyperuricaemia and gout [J]. Rheumatology (Oxford, England), 2009, 48(3):222-226.
[8] Caulfield Mark J, Munroe Patricia B, O'Neill Deb, et al. SLC2A9 is a high-capacity urate transporter in humans [J]. PLoS medicine, 2008, 5(10):e197.
[9] Enomoto Atsushi, Kimura Hiroaki, Chairoungdua Arthit, et al. Molecular identification of a renal urate anion exchanger that regulates blood urate levels [J]. Nature, 2002, 417(6887):447-452.
[10] Woodward Owen M, Köttgen Anna, Coresh Josef, et al. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout [J]. Proceedings of the National Academy of Sciences of the United States of America, 2009, 106(25):10338-10342.
[11] Kimura Toru, Takahashi Michi, Yan Kunimasa, et al. Expression of SLC2A9 isoforms in the kidney and their localization in polarized epithelial cells [J]. PloS one, 2014, 9(1):e84996.
[12] Caulfield Mark J, Munroe Patricia B, O'Neill Deb, et al. SLC2A9 is a high-capacity urate transporter in humans [J]. PLoS medicine, 2008, 5(10):e197.
[13] Burckhardt Gerhard. Drug transport by Organic Anion Transporters (OATs) [J]. Pharmacology & therapeutics, 2012, 136(1):106-130.
[14] Xu Liuqing, Shi Yingfeng, Zhuang Shougang, et al. Recent advances on uric acid transporters [J]. Oncotarget, 2017, 8(59):100852-100862.
[15] Major Tanya J, Dalbeth Nicola, Stahl Eli A, et al. An update on the genetics of hyperuricaemia and gout. [J]. Nature reviews. Rheumatology, 2018, 14(6):341-353.
[16] Yisireyili Maimaiti, Hayashi Motoharu, Wu Hongxian, et al. Xanthine oxidase inhibition by febuxostat attenuates stress-induced hyperuricemia, glucose dysmetabolism, and prothrombotic state in mice [J]. Scientific reports, 2017, 7(1):1266.
[17] Lin Hui, Li Zhimin, Lu Zhifu. Effect of modified Baihu decoction combined with colchicine on acute gouty arthritis and the effect of inflammatory factors in patients [J]. Chinese Medicine, 2020, 15(8):1292-1296.
[18] Latourte Augustin, Bardin Thomas, Richette Pascal, et al. Prophylaxis for acute gout flares after initiation of urate-lowering therapy [J]. Rheumatology (Oxford, England), 2014, 53(11):1920-1926.
[19] Zhang MaryAnn, Solomon Daniel H, Desai Rishi J, et al. Assessment of Cardiovascular Risk in Older Patients With Gout Initiating Febuxostat Versus Allopurinol: Population-Based Cohort Study [J]. Circulation, 2018, 138(11): 1116-1126.
[20] Hamburger Max, Baraf Herbert S B, Adamson Thomas C, et al. 2011 recommendations for the diagnosis and management of gout and hyperuricemia [J]. The Physician and sportsmedicine, 2011, 39(4):98-123.
[21] Feng Dan, Ling Wenhua, Duan Ruidong. Lycopene suppresses LPS-induced NO and IL-6 production by inhibiting the activation of ERK, p38MAPK, and NF-κB in macrophages [J]. Inflammation Research, 2010, 59(2):115-121.
[22] Jin Du, Liang Yimin, Mou Xiaoyue. Comparison of the relieving effects of different doses of colchicine on pain and inflammation in patients with acute attack of gout [J]. Chinese Journal of General Practice, 2020, 18(7):1143-1146. 
[23] Liu Lijun, Zhong Lijun, Chen Yan. Analysis of the application of commonly used antigout drugs in a hospital [J]. Chinese Journal of Clinical Rational Drug Use, 2022, (26):160-162.
[24] Wang Yi, Ren Jingtian. Evaluation and significance of benefit-risk analysis of benzbromarone-induced liver injury [J]. Chinese Journal of Pharmacovigilance, 2022, 19(02):189-192.
[25] Wu Lili, Chen Yulian, Liu Han, et al. Emodin-induced hepatotoxicity was exacerbated by probenecid through inhibiting UGTs and MRP2 [J]. Toxicology and Applied Pharmacology, 2018, 359:91-101.
[26] Zhang Mingyuan, Niu Junqi, Wen Xiaoyu, et al. Liver failure associated with benzbromarone: A case report and review of the literature [J]. World journal of clinical cases, 2019, 7(13):1717-1725.
[27] Wang Fanhong, Shi Zhengfang. Treatment of primary gout by traditional Chinese medicine in 28 cases [J]. Zhejiang Journal of Chinese Medicine, 1994, 29(5):208-209.
[28] Kang Le, Miao Jinxin, Cao Lihua, et al. Total glucosides of herbaceous peony (Paeonia lactiflora Pall.) flower attenuate adenine-and ethambutol-induced hyperuricaemia in rats [J]. Journal of Ethnopharmacology, 2020, 261: 113054.
[29] Wang Yajie, Zhang Guowen. Inhibition Effect of Morin on Xanthine Oxidase Activity [J]. Food Science, 2014, 35(13): 143-146.
[30] Federico Dajas. Life or death: Neuroprotective and anticancer effects of quercetin [J]. Journal of Ethnopharmacology, 2012, 143(2):383-396.
[31] Spagnuolo Carmela, Russo Maria, Bilotto Stefania, et al. Dietary polyphenols in cancer prevention: the example of the flavonoid quercetin in leukemia [J]. Annals of the New York Academy of Sciences, 2012, 1259(1):95-103.
[32] Xie Kaili, Li Zhaohua, Dong Xianzhi, et al. Research Progress of Quercetin on Inhibiting the Activity of Xanthine Oxidase [J]. Lishizhen Medicine and Materia Medica Research, 2019, 30(9):2223-2225. 
[33] Chen Haiqing, Zhou Xuan, Wang Xiuxiu. Mechanism study of quercetin in the treatment of hyperuricaemia [J]. Guangming Journal of Chinese Medicine, 2019, 34(9):1340-1344.
[34] Zhu Lingling, Chen Baojun. Aescin Total Saponins Reducing Uric Acid in Vitro and in Vivo and Its Effect on Rric Acid Transporters of Mice with Hyperuricemia [J]. Journal of New Chinese Medicine, 2018, 50(5):41-44. 
[35] Lin Fengping, Ren Kaiming, Song Enfeng, et al. Effect of Clematis on uric acid nephropathy in rats [J]. Chinese Traditional Patent Medicine, 2006, (06):842-845.
[36] Wang Jinpiao, Liu Yongmao, He Zhichao, et al. Effects of ethanol extract of Zea mays on potassium oxybate-induced hyperuricaemia model in rats [J]. Chinese Traditional Patent Medicine, 2017, 39(3):605-608. 
[37] Chen Yan, Chen Xiao Lin, Liu Mengting, et al. Exploration of uric acid-lowering mechanism of Dioscorea villosa based on oatp1a1 expression in hyperuricemic rats [J]. Lishizhen Medicine and Materia Medica Research, 2015, 26(10):2330-2332. 
[38] Chen Guangliang, Zhu Liran, Na Sha, et al. Effect of total saponin of Dioscorea on chronic hyperuricemia and expression of URAT1 in rats [J]. China Journal of Chinese Materia Medica, 2013, 38(14):2348-2353.
[39] Ma Shiwei. Efficacy of the Danggui Niantong decoction on hyperuricemia and its effect on inflammatory factor levels [J]. Clinical Journal of Chinese Medicine, 2022, (23):64-66.
[40] Zhong Xiaofeng. Observation on the efficacy of Tiger Cane Gout Granules in the treatment of acute gouty arthritis [J]. Chinese Journal of Clinical Rational Drug Use, 2013, 6(11):61-62.
[41] Zhou Mi, Ze Kan, Wang Yifei, et al. Huzhang Tongfeng Granule Improves Monosodium Urate-Induced Inflammation of Gouty Arthritis Rat Model by Downregulation of Cyr61 and Related Cytokines [J]. Evidence-based complementary and alternative medicine: eCAM, 2020, 2020:9238797.
[42] Chen Hanyu. Effect of Liuwei Tongfeng Decoction combined with benzbromarone on TRF1 and TRF2 in peripheral blood of patients with gouty arthritis [J]. Modern Journal of Integrated Traditional Chinese and Western Medicine, 2018, 27(34):3782-3785+3802.

Downloads: 4235
Visits: 167802

Sponsors, Associates, and Links


All published work is licensed under a Creative Commons Attribution 4.0 International License.

Copyright © 2016 - 2031 Clausius Scientific Press Inc. All Rights Reserved.