Skip to main content

SYSTEMATIC REVIEW article

Front. Pharmacol., 25 January 2021
Sec. Ethnopharmacology
This article is part of the Research Topic Integrative Pharmacology-based Research on Traditional Medicine: Methodologies, Medical and Pharmacological Applications View all 73 articles

The Efficacy and Mechanism of Chinese Herbal Medicines in Lowering Serum Uric Acid Levels: A Systematic Review

Liqian Chen,&#x;Liqian Chen1,2Zhengmao Luo&#x;Zhengmao Luo3Ming Wang&#x;Ming Wang1Jingru ChengJingru Cheng4Fei LiFei Li5Hanqi Lu,Hanqi Lu1,2Qiuxing HeQiuxing He2Yanting YouYanting You2Xinghong ZhouXinghong Zhou2Hiu Yee KwanHiu Yee Kwan6Xiaoshan Zhao
Xiaoshan Zhao2*Lin Zhou
Lin Zhou7*
  • 1Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
  • 2Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Chinese Medicine, Southern Medical University, Guangzhou, China
  • 3Department of Nephrology, General Hospital of Southern Theatre Command, PLA, Guangzhou, China
  • 4Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
  • 5Department of Traditional Chinese Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
  • 6School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
  • 7Endocrinology Department, Nanfang Hospital, Southern Medical University, Guangzhou, China

Background. Chinese herbal medicines are widely used to lower serum uric acid levels. However, no systemic review summarizes and evaluates their efficacies and the underlying mechanisms of action. Objectives. To evaluate the clinical and experimental evidences for the effectiveness and the potential mechanism of Chinese herbal medicines in lowering serum uric acid levels. Methods. Four electronic databases PubMed, Wed of Science, the Cochrane Library and Embase were used to search for Chinese herbal medicines for their effects in lowering serum uric acid levels, dated from 1 January 2009 to 19 August 2020. For clinical trials, randomized controlled trials (RCTs) were included; and for experimental studies, original articles were included. The methodological quality of RCTs was assessed according to the Cochrane criteria. For clinical trials, a meta-analysis of continuous variables was used to obtain pooled effects. For experimental studies, lists were used to summarize and integrate the mechanisms involved. Results. A total of 10 clinical trials and 184 experimental studies were included. Current data showed that Chinese herbal medicines have promising clinical efficacies in patients with elevated serum uric acid levels (SMD: −1.65, 95% CI: −3.09 to −0.22; p = 0.024). There was no significant difference in serum uric acid levels between Chinese herbal medicine treatments and Western medicine treatments (SMD: −0.13, 95% CI: −0.99 to 0.74; p = 0.772). Experimental studies revealed that the mechanistic signaling pathways involved in the serum uric acid lowering effects include uric acid synthesis, uric acid transport, inflammation, renal fibrosis and oxidative stress. Conclusions. The clinical studies indicate that Chinese herbal medicines lower serum uric acid levels. Further studies with sophisticated research design can further demonstrate the efficacy and safety of these Chinese herbal medicines in lowering serum uric acid levels and reveal a comprehensive picture of the underlying mechanisms of action.

Introduction

Hyperuricemia refers to an abnormally high concentration of serum uric acid (sUA), typically defined as >7 mg/dL in men and >6 mg/dL in women. The data from the National Health and Nutrition Examination Survey (NHANES) 2007–2016 showed that the prevalence rates of hyperuricemia were 20.2% among men and 20.0% among women between 2015 and 2016 in the United States (Chen-Xu et al., 2019). A cross-sectional survey in China showed that the prevalence of hyperuricemia was 8.4% among Chinese adults from 2009 to 2010 (Liu et al., 2014a). Hyperuricemia and gout remain as a considerable burden, which not only adversely affect patients’ health and quality of life (Burke et al., 2015; Gamala and Jacobs, 2019), but also cast an economic burden in the society (Rai et al., 2015). SUA is associated with cardiovascular diseases, such as hypertension (Kuwabara et al., 2018) and atrial fibrillation (Tang et al., 2014). Elevated sUA can lead to decreased renal function, which in turn reduces the excretion of UA in urine, resulting in an increased risk of hyperuricemia or gout (Sato et al., 2019). Elevated sUA may also contribute to the pathogenesis of metabolic syndrome (Battelli et al., 2018; Battelli et al., 2019), non-alcoholic fatty liver disease (Xu et al., 2015), diabetes (Kim et al., 2015). Urate-lowering therapy is a therapeutic strategy for controlling gout, chronic kidney disease, metabolic syndrome and many other diseases. Current interventions for elevated sUA include xanthine oxidase inhibitors, uricosuric agents and anti-inflammatory drugs. Although both febuxostat and allopurinol are effective in reducing sUA, allopurinol may produce a mild skin rash and severe cutaneous reactions (Strilchuk et al., 2019), while febuxostat has a higher risk of all-cause and cardiovascular mortality (White et al., 2018). Benzbromarone is a uricosuric drug which is widely used, but studies have reported possible complications such as hepatotoxicity (Strilchuk et al., 2019). Although these drugs are clinically used, their efficacies are unsatisfactory, and are usually coupled with adverse side effects in the long-term use (Dalbeth et al., 2016).

Hyperuricemia belongs to the arthromyodynia disease category in traditional Chinese medicine. Chinese herbal medicine has been used to treat hyperuricemia for a long time and has significant clinical efficacy (Lin et al., 2016). Chinese herbal medicines with high efficacy and low incidence of adverse reactions have drawn increasing attention from scholars. Studies have compared Chinese herbal medicine with Western medicine for their efficacies in lowering sUA levels. However, these studies differ in their treatment protocols and evaluation methodologies, which greatly limit their clinical applicability. In addition, the mechanism of Chinese herbal medicine in lowering sUA levels is still being explored. Some herbs such as Phellodendri Chinrnsis Cortex, Atractylodes Lancea (Thunb.)Dc. (Chen et al., 2015b), Smilacis Glabrae Rhixoma (Liu et al., 2015), reduce UA intake and/or increase UA excretion by regulating various physiological and cellular pathways. Some herbs like bergenin (Chen et al., 2020a), alpinia oxyphylla seed extract (Lee et al., 2019b) and rhizoma smilacis glabrae extracts (Liang et al., 2019), promote renal and gut uric acid excretion in hyperuricemia models and also decrease the serum levels of inflammatory cytokines. Most Chinese herbal medicines act on multiple targets to achieve their sUA lowering effects. Therefore, the aim of this systematic review is to evaluate the evidence for the efficacy of Chinese herbal medicines in lowering sUA levels in patients with hyperuricemia, compared to no intervention, placebo or urate-lowering agents, and to comprehensively summarize the mechanisms underlying the sUA lowering effects reported from experimental studies.

Materials and Methods

Data Source and Search Strategy

Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) (Moher et al., 2009) was used to construct the report of the current study and the completed checklist is provided in Supplementary material, Supplementary Table S1. The electronic databases PubMed, Wed of Science, the Cochrane Library and Embase were systematically searched by three researchers (Liqian Chen, Zhengmao Luo and Ming Wang). For any discrepancies between researchers, consensus was reached through discussion. We reviewed literatures published from 1 January 2009 to 19 August 2020 on elevated sUA that had been treated with Chinese herbal medicines. The following combination of terms were used as search keywords: (Traditional Chinese Medicine OR Herbal Medicine OR Chinese Herbal Drugs OR Chinese Plant Extracts OR (Plants, Medicinal) OR Phytochemicals OR herb* OR natural product) combined with (gout OR pain paralysis OR hyperuricemia OR uric acid). The full search strategy in PubMed was provided in Supplementary material, Supplementary Table S2. The search did not exclude articles based on language. To look for additional relevant studies, references of all potentially relevant articles were also retrieved, and authors of studies that met the inclusion criteria but lacked data would be contacted. Abstracts, meeting proceedings, and personal communications were not used for the purpose of this study.

Study Selection

The articles in this review included clinical trials and in vivo and in vitro studies. When screening clinical trials, the followings are the inclusion criteria:

A). Types of trials: All randomized controlled trials (RCTs) investigating the use of Chinese herbal medicine in lowering sUA will be eligible for inclusion.

B). Types of participants: In accordance with the NHANES-III laboratory definition, hyperuricemia is typically defined as >7 mg/dL in men and >6 mg/dL in women (Choi et al., 2020). All adult patients (18 years and older, no upper age limit) with a diagnosis of hyperuricemia will be considered for this review.

C). Types of interventions: According the General guidelines for methodologies on research and evaluation of traditional medicine from WHO, herbal medicines are defined as materials or products derived from plants that have medical or other beneficial effects on human health, including herbs, herbal materials, herbal products, finished herbal products that contain parts of plants or other plant materials or compositions as active ingredients, as well as materials of inorganic or animal sources. The interventions included in this study include Chinese herbal medicine in various prescriptions, such as herbal formulas, herbal extracts, active ingredients of herbs. Clinical trials comparing Chinese herbal medicine with no intervention, placebo or urate-lowering agents were included in our study. Chinese herbal medicine plus placebo or combined with urate-lowering agents compared to the same medications was also included. We do not limit the formulations or administration of herbal preparations for clinical use.

D). Types of outcome measures: Outcome measures should include at least one essential outcome, such as change in sUA levels after treatment and the overall efficacy.

When screening in vivo or vitro experimental studies using the herbs, the following conditions should be met before inclusion: A) Original article investigating the use of Chinese herbal medicine in lowering sUA will be eligible for inclusion. B) Studies using experimental models of mice, rats, rabbits or cell cultures will be considered. All models used should present corresponding pathological symptoms. C) The model should only be treated with Chinese herbal medicine before or after intervention. If medication other than herbs were being used, both the treatment group and the control group must be administered. D) Outcome measures should include change in sUA after treatment.

Data extraction

The data were extracted by three researchers (Jingru Cheng, Fei Li and Hanqi Lu) independently to obtain the following information: A) the design of study; B) characteristics of trial participants (including sample size, age, period); C) type of intervention (including dose regimen, duration); D) type of outcome measure (including the level of sUA). The reported mean (standard deviation [SD]) or risk estimates and 95% confidence intervals (CIs) of sUA were also extracted.

Quality Assessment

The quality assessment of the studies was performed independently by three researchers (Qiuxing He, Yanting You and Xinghong Zhou), and all discrepancies between researchers were resolved through discussion. The methodological quality of RCTs was assessed according to the Cochrane Handbook for Systematic reviews of Interventions. The scores for each bias domain and the final score of risk of systematic bias were graded as low, high or unclear risk. The overall level of evidence was considered “strong” if there were consistent findings among multiple high quality RCTs, and “moderate” if findings were consistent among multiple low-quality RCTs and/or one high-quality RCT. Level of evidence was “conflicting” if findings were inconsistent across the studies, and “no evidence from trials,” if there were no RCTs.

Statistical Analysis

Statistical analysis was performed with the software STATA version 16.0. The d index and the standard deviation (SDd) values of sUA for each RCT were calculated before using STATA. The d index and SDd values of sUA were continuous, standard mean difference (SMD) with 95% confidence interval (95% CI) was calculated. The overall effect was calculated by a Z-test, and p < 0.05 (2-tailed) was deemed statistically significant. Potential heterogeneity was assessed by I2 statistics. A fixed effects model was chosen if I2 ≤ 50%, otherwise, a random effects model was applied. Subgroup analysis was performed to illuminate the heterogeneity according to the study characteristics, such as interventions other than Chinese herbal medicine. A ‘leave-one-out’ sensitivity analysis was carried out to test the reliability of the results. Potential publication bias was evaluated by the Egger’s test and Begg’s test. When the geometric mean and CIs were reported, tools provided in the Cochrane Handbook were used to convert the geometric mean and CIs to arithmetic mean and SD of the raw data.

Results

Literature Flow

The initial electronic search of the literature yielded 4981 potentially relevant citations. After duplicate removal and title/abstract screening, 374 full-text articles were retrieved for detailed assessment. Of these studies, 180 articles did not meet the inclusion criteria. Finally, 194 articles were included in the review with 10 clinical trials, 169 in vivo experiments, 0 in vitro experiments, and 15 that were a combination of both in vitro and in vivo experiments (Figure 1).

FIGURE 1
www.frontiersin.org

Figure 1. Flowchart of study selection for the systematic review.

Results of clinical trials

Study Characteristics

The specified characteristics of the included clinical trials as well as their study populations were summarized in Table 1. All clinical trials included were RCTs which mainly explored the effects of Chinese herbal medicine on patients with elevated sUA levels such as hyperuricemia and gout. In order to objectively observe the therapeutic effects of herbs on lowering sUA levels, the changes of sUA levels before and after treatment in 10 clinical trials were listed in Supplementary material, Supplementary Table S3. Of the 10 included trials, nine were treated with herbal formula, and the remaining one was treated with a combination of herbal extracts and active ingredients. In the subgroup analyses of intervention, two studies compared the therapeutic effects of Chinese herbal medicine and placebo (Rozza et al., 2016; Xie et al., 2017), three studies compared Chinese herbal medicine and Western medicine (Zhang et al., 2009; Zhou et al., 2013; Wang et al., 2014), two studies compared Chinese herbal medicine, Western medicine and placebo or no intervention (Zhang et al., 2011; Wang et al., 2019b), one compared two kinds of Chinese herbal medicine and Western medicine (Yu et al., 2018), and two compared Chinese herbal medicine combined Western medicine and Western medicine (Chen et al., 2009; Xiang et al., 2009).

TABLE 1
www.frontiersin.org

TABLE 1. Clinical trials for herbs lowering serum uric acid.

Meta-Analysis

In order to investigate the efficacy of Chinese herbal medicine in lowering sUA, we performed a subgroup analysis based on interventions. A random effects model was used for the analysis because I2 = 96.4%.

Chinese Herbal Medicine vs Placebo or No Intervention

Four RCTs were analyzed. The subgroup meta-analysis showed that all of them were statistically significant differences between Chinese herbal medicine and placebo or no intervention. The combined SMD was −1.65 with a 95% CI of −3.09 to −0.22 (p = 0.024). Therefore, there was a significant difference between the Chinese herbal medicine and placebo or no intervention in the reduction of sUA (Figure 2).

FIGURE 2
www.frontiersin.org

Figure 2. Effects of Chinese herbal medicine on serum uric acid in patients with elevated serum uric acid.

Chinese Herbal Medicine vs Western Medicine

Seven RCTs were analyzed. On subgroup meta-analysis, the combined SMD was −0.13 with a 95% CI of −0.99 to 0.74 (p = 0.772), indicating no significant difference between the Chinese herbal medicine and Western medicine in the reduction of sUA (Figure 2).

Chinese Herbal Medicine Plus Western Medicine vs Western Medicine

Two RCTs were analyzed. Subgroup meta-analysis (Figure 2) showed no statistical significance between the Chinese herbal medicine plus Western medicine and Western medicine in the reduction of sUA (SMD −2.27, 95% CI: −5.84 to 1.31, p = 0.214).

Meta-regression Analyses

The meta-regression showed that samples size, duration of treatment, type of diseases and intervention other than Chinese herbal medicine did not influence these results (all p values >0.05) (Supplementary Figure S1).

Quality Assessment

RCT was assessed according to the Cochrane Handbook for Systematic reviews of Interventions (Table 2). Generally, the methodological quality was assessed to be moderate. Most of the studies (8/10, 80%) have details on the random grouping of patients, but only half of the studies (5/10, 50%) fully reported the scheme of concealment allocation. Only four trials (4/10, 40%) had their subjects and investigators blinded during the study, and four trials (4/10, 40%) had all the subjects, investigators and outcome evaluators blinded.

TABLE 2
www.frontiersin.org

TABLE 2. Methodological quality assessment of randomized controlled trials according to the Cochrane Handbook.

Sensitivity Analysis

Sensitivity analysis was conducted to confirm the efficacy Chinese herbal medicine in lowering sUA. The pooled SMDs were repeated by sequentially removing one of the included studies with a random-effects model (Supplementary Figure S2). None of the studies changed the overall effect.

Publication Bias

There was no evidence of publication bias according to the Begg’s test (p = 0.428) and Egger’s test (p = 0.344) for the meta-analysis of Chinese herbal medicine on lowering sUA (Supplementary Figure S3).

Results of experimental studies

We categorized 184 in vivo and in vitro experiments into three groups: 56 active ingredients (Table 3), 78 natural products (Table 4), and 52 herbal formulas (Table 5). Among them, one article described two active ingredients (Lin et al., 2018), and one article (Su et al., 2014) studied one active ingredient and one natural product concurrently. To assure the quality of the studies included, detailed information on herbs (including source, concentration, quality assessment, chemical analysis, and compound purity) was summarized in Supplementary material, Supplementary Tables S4–S6.

TABLE 3
www.frontiersin.org

TABLE 3. Active ingredients on lowering serum uric acid based on in vivo and in vitro studies.

TABLE 4
www.frontiersin.org

TABLE 4. Natural products that lower serum uric acid based on in vivo and in vitro studies.

TABLE 5
www.frontiersin.org

TABLE 5. Herbal formulas that lower serum uric acid based on in vivo and in vitro studies.

Active Ingredients That Lower Serum Urate in vivo and in vitro Studies

Active ingredient is a single ingredient and studies have shown that it plays an important therapeutic role in reducing sUA. Among the active ingredients with potential of lowering sUA (Table 3), 25 of which act on uric acid synthesis, 28 target uric acid transporter, 19 resolve inflammation, nine possess kidney protective function, and four regulate the oxidative stress.

Natural Products That Lower Serum Urate in vivo and in vitro Studies

Natural products include herbs and relatively complex extracts derived from herbs. Among the natural products with potential of lowering sUA (Table 4), 40 of which act on uric acid synthesis, 29 target uric acid transporter, 17 resolve inflammation, eight possess kidney protective functions, and three regulate the oxidative stress.

Herbal Formulas That Lower Serum Urate in vivo and in vitro Studies

Herbal formula is a combination of a variety of herbs. The composition of the herbal formula included in the current study were listed in Supplementary material, Supplementary Table S7. Among the herbal formula with lowering sUA effects (Table 5), 18 of which act on uric acid synthesis, 15 target uric acid transporter, 15 resolve inflammation, seven possess kidney protective functions, and five regulate the oxidative stress.

Discussion

The efficacies of Chinese herbal medicines lowering Serum uric Acid levels

This systematic review compares the efficacies of the Chinese herbal medicines and the Western medicine in lowering sUA levels by analysing10 RCTs with a total of 1,060 patients. The meta-analysis has three important findings. First, there is a significant difference between the Chinese herbal medicine, placebo or no intervention in the reduction of sUA levels. Second, the efficacies of Chinese herbal medicines in lowering sUA levels are comparable to that of Western medicine. Third, the efficacies of Chinese herbal medicines plus Western medicine in lowering sUA levels are comparable to that of Western medicine. The heterogeneity of the meta-analysis was high. To investigate the source of heterogeneity in our analysis, we conducted a sensitivity analysis, removing one study at one time from the primary analysis did not change the main finding. In addition, our findings were confirmed by the lack of publication bias and effect modifiers according to the Begg’s test, Egger’s test and meta-regression analysis.

The results indicate that the efficacies of Chinese herbal medicines in lowering sUA levels are comparable to that of Western medicine, which is consistent with the analysis result of Lin et al. (Lin et al., 2016). Compared with the study of Lin et al., our study is more comprehensive, because we also compared the sUA lowering efficacy between Chinese herbal medicine and placebo or no intervention, and between Chinese herbal medicine plus western medicine and western medicine. In addition, we have summarized the underlying mechanisms of herbs in lowering sUA. It should be noted that among the 10 RCTs included, a total of six RCTs used one or several components of Simiao Pills, which is a famous formula in traditional Chinese medicine, including Cortex Phellodendri Chinensis, Atractylodes Lancea (Thunb.) DC., Coix lacryma-jobi L.var.ma-yuen (Roman.) Stapf (Yi Yi), Cyathula officinalis Kuan (Xiang et al., 2009; Zhang et al., 2009; Zhang et al., 2011; Zhou et al., 2013; Xie et al., 2017; Yu et al., 2018). The specific doses of herbs in each study have been listed in the Supplementary Tables S4–S6. According to the Pharmacopoeia of the People’s Republic of China revised by the Food and Drug Administration in 2015, except for the use of Cortex Phellodendri Chinensis, Atractylodes Lancea (Thunb.) DC., Cyathula officinalis Kuan in one RCT (Xiang et al., 2009) exceeded the recommended dosage, the dosages of these four herbs in the remaining five RCTs were within the recommended dosage range. Except for the RCT in which the dosage exceeded the recommended dosage (Xiang et al., 2009), the dosage of these four herbs in the remaining five RCTs were all at the high level or even reached the critical value within the recommended range. Despite the high dosage, the three RCTs describing the adverse reactions in the six RCTs showed that the incidence of adverse reactions was either lower than that of the placebo group or was not statistically significant compared with the control group.

In addition to clinical studies, experimental studies have also suggested the urate lowering effects of Simiao Pills, which act on xanthine oxidase (XO), uric acid transport-related proteins urate anion transporter 1 (URAT1) and glucose transporter 9 (GLUT9), and also modulate the inflammation, oxidative stress and other processes (Hu et al., 2010; Hua et al., 2012; Shi et al., 2013; Pan et al., 2014; Ma et al., 2015; Lin et al., 2020). The finding suggests that Chinese herbal medicines are mostly multi-targeted or have interplay with other signaling pathways to lower sUA levels. In contrast, conventional Western medicines used to lower sUA levels include allopurinol, probenecid and benzbromarone, which generally act on specific targets. Allopurinol competitively inhibits xanthine oxidase (Strilchuk et al., 2019). Probenecid and benzbromarone are typical urate-promoting drugs target at URAT1 (Azevedo et al., 2019). These may partly explain why the combination of Chinese and Western medicine can improve the index of renal function while lowering sUA levels in patients with hyperuricemia (Chen et al., 2009; Xiang et al., 2009).

Nevertheless, several limitations of the meta-analysis are worth considering. Due to limited reports, participants with elevated sUA levels in this meta-analysis included patients with hyperuricemia and gout, and most participants were from China. These facts indicate that the current meta-analysis could have potential bias. Further trials need to be carried out in a larger comprehensive population to demonstrate the efficacy of Chinese herbal medicines in lowering sUA. In addition, long-term tracking of sUA is necessary to determine whether Chinese herbal medicine can effectively control sUA. However, long-term follow-ups were not available in the current included studies. The incidence of adverse reactions is an important indicator to compare the efficacy and safety of Chinese herbal medicine and Western medicine in lowering sUA levels. However, of the six included studies comparing Chinese herbal medicine and Western medicine, only 2 has reported the adverse reaction events in details. Thus, we cannot evaluate the safety of Chinese herbal medicine vs. western medicine in lowering sUA levels. Moreover, the standardization of the methodologies and the small number of the included trials may lead to an overestimation of the overall efficacy of Chinese herbal medicine. Therefore, studies with high quality are needed to confirm the efficacy and safety of Chinese herbal medicine in lowering sUA levels.

The mechanisms of Chinese herbal medicines in lowering Serum uric Acid

In current review, 186 in vivo and in vitro experiments with 56 active ingredients (Table 3), 78 natural products (Table 4), and 52 herbal formulas (Table 5) are included to explore the common mechanism of Chinese herbal medicine in lowering sUA levels. According to the summary of the targets of Chinese herbal medicine in lowering sUA, it is clearly revealed that most Chinese herbal medicine lowers sUA by acting on multiple targets or multiple pathways. The therapeutic mechanism of Chinese herbal medicine included in this study mainly involved the UA synthesis, UA transport, inflammation, renal fibrosis and oxidative stress.

Uric acid, produced primarily in the liver, is the final product of diet and endogenous purine metabolism. Problems with key enzymes involved in UA production can cause abnormal UA levels, including phosphoribosyl pyrophosphate (PRPP) synthetase, purine nucleoside phosphorylase, xanthine oxidase, hypoxanthine-guanine phosphoribosyl transferase (HGPRT) (Cammalleri and Malaguarnera, 2007; Maiuolo et al., 2016). In the circulation, UA exists mainly in the form of urate anion under physiologic pH. The saturation level of monosodium urate in human plasma is limited. Hence, UA must be excreted continuously to prevent its accumulation and reduce the toxicity. The primary scavenger of urate is the kidney, which expels about 75% of urate every day (Chaudhary et al., 2013). In recent years, studies have revealed the complex interaction of transporters involved in urate metabolism (Wright et al., 2010). Several transporters involved in urate metabolism have been identified, including URAT1, GLUT9, organic anion transporters (OAT1/3), and adenosine triphosphate (ATP)-binding cassette transporter 2 (ABCG2) (Enomoto et al., 2002; Lipkowitz, 2012; Mandal and Mount, 2015). Previous studies have found that inflammation, oxidative stress, mitochondrial dysfunction and other factors cause abnormalities in these critical proteins, and thus lead to the disorders in UA metabolism (Lanaspa et al., 2012; Zhou et al., 2018b). On the contrary, abnormal UA levels can also lead to the release of some cytokines, such as tumor necrosis factor-α (TNF-α), nuclear factor-kappa B (NF-κΒ), the NOD-like receptor P3 (NLRP3) inflammasome, interleukin-6 (IL-6), interleukin-1β (IL-1β), and so on (Major et al., 2018; Singh et al., 2019).

Herbs Lower Serum Uric Acid by Targeting Uric Acid Synthesis

The substrate for UA synthesis is ribose-5-phosphate which can be converted to PRPP via PRPP synthase and then to inosine monophosphate. This intermediate compound produces adenosine monophosphate and guanosine monophosphate, which subsequently release adenosine and guanosine molecules, respectively. Adenosine deaminase converts adenosine to inosine, while guanosine to free guanine. Inosine is degraded to hypoxanthine via purine nucleoside phosphorylase. Xanthine oxidase, one of the key enzymes involved in UA synthesis, converts hypoxanthine to xanthine which then converts to UA. Guanine is directly converted to xanthine, which subsequently to UA by XO. On the other hand, hypoxanthine and guanine enter a salvage pathway through the activity of HGPRT, which converts these purine into their respective nucleotides (Lipkowitz, 2012; Lima et al., 2015; Maiuolo et al., 2016). Thus, PRPP synthase, XO and HGPRT are all key enzymes that can cause abnormal sUA levels. PRPP gene mutations have been implicated in a number of human diseases. Overexpression of PRPP results in the enhanced activity of phosphoribosyl pyrophosphate synthetase-I, which can lead to excessive production of purine. Patients with active phosphoribosyl pyrophosphate synthetase-I may result in UA over production (Mittal et al., 2015). Lesch-Nyhan disease is caused by a wide variety of mutations in the HGPRT gene and is one of the models of gout caused by the increased production of UA (Fu et al., 2014; Harris, 2018). Lack of HGPRT can lead to Kelley-Seegmiller syndrome, which is characterized by hyperuricemia, hyperuricosuria, gouty arthritis and urolithiasis (Chavarriaga et al., 2019). Among these key enzymes, XO is the most commonly studied enzyme. 3,5,2′,4′-tetrahydroxychalcone significantly inhibits the activities of XO in liver, and the decreased content of PRPP in liver will suppresse the UA production (Niu et al., 2018). In our review, nearly half of the studies (83/186, 45%) confirm that herbs regulate XO synthesis and activity. For example, pallidifloside D, a saponin glycoside constituent from the total saponins of Smilax riparia, enhances the hypouricemic effect of allopurinol by regulating XO activity. The combination of allopurinol and Pallidifloside D significantly up-regulates HGPRT expression and down-regulates the expressions of PRPP in PC12 cells (Li et al., 2016).

Herbs Lower Serum Uric Acid by Targeting Uric Acid Transporter

About two-thirds of UA is excreted by the kidneys, and the remaining is excreted via the gastrointestinal tract. Renal excretion of UA consists of four steps: 1) glomerular filtration, 2) presecretory reabsorption, 3) secretion and 4) post-secretory reabsorption (Maiuolo et al., 2016). Urate is filtered through the glomerulus, so the reabsorption and secretion of UA after glomerulus play an important role in regulating the excretion of UA. Most of the urate (99%) filtered through the glomerulus is reabsorbed in the early S1 segment of the proximal tubule (presecretory reabsorption). Uric acid is then secreted in the S2 segment of the proximal tubule to return approximately 50% of the filtered urate into the tubule lumen. Post-secretory reabsorption occurs primarily in the distal S3 segment of the proximal tubule, followed by about 10% of the secreted urate in the urine (Diamond and Paolino, 1973; Mandal and Mount, 2015). Urate transporters such as URAT1, OAT1, OAT3, GLUT9, ABCG2 play important roles in regulating sUA, and their dysfunction may cause abnormal urate transport. URAT1, the main urate-anion exchanger in the luminal membrane of the proximal tubules, can be inhibited by the uricosuric agents, likes probenecid, benzbromarone and losartan (Enomoto et al., 2002). URAT1 mutations have been found in patients with familial hypouricemia and UA levels below 1 mg/dL, and the UA transport is inactivated when this mutant transporter is expressed in xenopus oocytes (Dinour et al., 2011). Clinical studies have shown that inhibition of URAT1 effectively reduces sUA levels and resolves the gout symptoms (Lee et al., 2019a). URAT1 is a branch of the organic anion transporter (OAT). OAT1 and OAT3 exchange urate with bivalent anions, suggesting that they are suitable for basolateral entry of urate during urate secretion (Mandal and Mount, 2015). Among the 14 members of the GLUT family transport glucose or other monosaccharides, GLUT9 does transport essentially urate (Caulfield et al., 2008). GLUT9 mediates urate transport, which is independent of sodium, chlorine and anions, but voltage-dependent (Anzai et al., 2008). Compared with the mutation in URAT1, the complete loss of GLUT9 results in the net secretion of urate (Preitner et al., 2009). ABCG2 is a multidrug resistance transporter that is also implicated as an important urate transporter. Its gene variation has become the main cause of elevated sUA levels (Dehghan et al., 2008). ABCG2-mediated loss or reduction of renal urate secretion will lead to increased renal urate reabsorption (Woodward et al., 2009).

In our review, more than one-third of the studies (72/186, 39%) show that Chinese herbal medicines target uric acid transporters. Alpinia oxyphylla seed extract enhances UA excretion in the kidney by reducing URAT1 and up-regulating OTA1 (Lee et al., 2019b). Wuling San down-regulates mRNA and protein levels of URAT1 and GLUT9, as well as up-regulates OAT1 in the kidney of hyperuricemic mice. Moreover, Wuling San also up-regulates organic cation/carnitine transporters which are associated with impaired renal function, leading to kidney protection (Ding et al., 2013). Gypenosides, natural saponins extracted from Gynostemma pentaphyllum, significantly lower sUA levels by reducing XO and increasing in urate excretion through regulating URAT1, GLUT9, and OAT1 (Pang et al., 2017).

Herbs Lowering Serum Uric Acid Resolve Inflammation

Uric acid belongs to the damage-associated molecular patterns, altered metabolites of necrotic or stressed cells that the innate immune system sees as an alarm signal (Patel, 2018). Elevated UA levels alters the physiology, boosting the expressions of inflammatory proteins by triggering complex pro-inflammatory cascades that damage cells and tissues (Chen and Lan, 2017). Clinical trials have shown that serum levels of IL-6 and TNF-α are significantly higher in hyperuricemia patients than in healthy people, and that of IL-6 and TNF-α are significantly increased as sUA levels increase (Zhou et al., 2018b). SUA above 9 mg/dl is associated with a gouty arthritis incidence of 4.9%. The accumulation of monosodium urate (MSU) crystals induce a mass of inflammatory cells (such as neutrophils and monocytes) to infiltrate into the site of MSU crystal deposits in patients, resulting in an acute inflammatory response, manifested as acute gout flares (Dalbeth et al., 2016). IL-1 released from these immune cells further triggers the release of various pro-inflammatory cytokines and chemokines, such as IL-8, IL-6 and TNF-α, which can further enhance neutrophil recruitment (El Ridi and Tallima, 2017). Inflammatory cytokines, especially IL-1β, are the key mediators of gouty inflammation. A phase III, international safety study of patients with acute gout arthritis treated with rilonacept (an IL-1 blocker) for 16 weeks shows that rilonacept significantly reduces the risk of gout attacks by 70.3% (Sundy et al., 2014). In experimental studies included in this review, 37 articles (11 active ingredients, 13 natural products, and 13 herbal formulas) describe the effects of these herbs on IL-1. It is worth mentioning that these herbs not only act on IL-1, but also as XO, UA transporter, other inflammatory factors such as IL-6, IL-8, TNF-α, NF-κB, NLRP3, caspase 1, etc.

There are 16 herbs included in this review that interfere with the NLRP3 inflammasome. Consistent with reported studies, elevated UA can be effectively reduced by regulating NLRP3 inflammasome - IL-1 pathway (Dhanasekar and Rasool, 2016; Szekanecz et al., 2019). Acute gout is an inflammatory response to MSU crystals. Innate immune pathways are essential in the pathogenesis of gout, particularly the activation of NLRP3 inflammasome, which leads to the release of IL-1β and other pro-inflammatory cytokines (So and Martinon, 2017). MSU crystals must first be coated with serum proteins and then interact with articular cell’s surface membrane directly or via receptors, to stimulate a cytosolic molecular platform involved in innate immunity and promote the assembly and activation of the NLRP3 inflammasome (El Ridi and Tallima, 2017). NLRP3 inflammasomes are formed by the recruitment of the apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and subsequent recruitment of caspase-1. Caspase-1 activates pro-inflammatory cytokines IL-1β and IL-18 by cleaving their respective precursor proteins, pro-IL-1β and pro-IL-18 (So and Martinon, 2017). Neutrophils are recruited and activated in response to the spillover of IL-1, producing ROS, proteolytic enzymes, pro-inflammatory chemokines, cytokines and so on, which recruit and activate macrophages (El Ridi and Tallima, 2017). Thus, the release of IL-1β mediated by inflammasome and the rapid recruitment of neutrophils lead to acute inflammatory episodes in gout patients. It is noteworthy that hyperuricemia may stimulate inflammatory leukocytes through epigenetic modification such as histone methylation, even without MSU crystals, thereby increasing production of IL-1β, IL-6, and TNF-α (Wasilewska et al., 2012; Zhou et al., 2018b).

The activation and maturation of IL-1β in response to endogenous and exogenous stimuli are also involved in the NF-κB pathway (Chen and Lan, 2017). In our review, there are 13 articles focused on NF-κB signaling pathway, showing the inhibitory effect of herbs on NF-κB activation under elevated sUA levels. It has been proved that NF-κB can be activated by UA (Spiga et al., 2017). Its activation transcribes a large number of pro-inflammatory genes. In a resting state, NF-κB combines with IκBα (inhibitor of NF-κB kinase subunit α) form a dimer in the cytoplasm, after IκBα kinase is activated, it phosphorylates IκBα. Subsequently, NF-κB is transferred from the cytoplasm to the nucleus, leading to the transcription and expression of genes related to inflammation. Studies have shown that inhibition of NF-κB restrains inflammation and improves hyperuricemia or gout conditions (Chen et al., 2019a; Wang et al., 2019c). Interestingly, studies focused on NF-κB activity is also related to NLRP3. NF-κB is essential for the initiation, assembly and activation of NLRP3 inflammasome, which is also a key step in the release of inflammatory cytokines in MSU crystal-induced inflammation (So and Martinon, 2017). P38 mitogen-activated protein kinase (MAPK) is also involved in the inflammatory cascade of NF-κB. Uric acid activates NF-κB through MAPK signal pathway, thus leading to the release of inflammatory factors such as TNF-α (Tang et al., 2017).

Herbs Lowering Serum Uric Acid Protect Against Renal Fibrosis

Evidence suggests that UA levels can be used to predict the prognosis of chronic kidney disease and end-stage kidney disease. Elevated sUA level is an independent risk factor for kidney disease and lead to renal fibrosis (Fan et al., 2019). Mice with systemic GLUT9 knockout showed moderate hyperuricemia, excessive hyperaciduria and obstructive nephropathy, along with progressive inflammatory fibrosis (Preitner et al., 2009). Once UA exceeds the maximum amount the kidneys can excrete, it gets deposit in the kidney and firstly causes direct pathological damage to the kidney. Secondly, the deposition of UA in the kidney results in the accumulation of neutrophils and monocytes as well as the release of inflammatory factors, which lead to glomerulosclerosis and interstitial fibrosis and aggravate renal injury (Ye et al., 2018). Prevention and treatment of renal fibrosis is the best treatment for kidney diseases caused by elevated sUA. However, at present, modern medical treatments are not effective in reducing renal fibrosis and preventing the progression of diseases.

A total of 24 studies included in this review are focused on hyperuricemic nephropathy, suggesting that herbal medicines improve renal injury induced by elevated sUA levels by regulating renal fibrosis-related signal pathways. Fibrosis is usually associated with strong inflammatory reactions and immunocyte infiltration. Therefore, inhibition of inflammatory cytokines might be a potential method to prevent fibrosis. Vaticaffinol, one of the herbs included in this review, markedly down-regulates NLRP3, ASC, caspase-1, IL-1β, IL-18, IL-6 and TNF-α in hyperuricemic mice, thus significantly decreases sUA levels and improves kidney function (Chen et al., 2017b). Transforming growth factor-β1/Mothers against decapentaplegic homolog 3 (TGF-β1/Smad3) signaling is the most potent fibrogenic factor in the regulation of renal interstitial fibrosis process (Loeffler and Wolf, 2015). Pterostilbene, also mentioned in this review, suppresses the activation of TGF-β1/Smad3 and proto-oncogene tyrosine-protein kinase Src/Signal transducer and activator of transcription 3 (Src/STAT3) signaling pathway as to decrease sUA level and alleviate renal fibrosis in hyperuricemic mice (Pan et al., 2019). These findings highlight the fact that herbal medicines may be the potential antifibrotic therapeutics for hyperuricemic nephropathy treatment.

Herbs Lowering Serum Uric Acid Modulate Oxidative Stress

In addition to the inflammatory process, oxidative stress is one of the early events related to the elevated sUA. Uric acid entering cells can rapidly induce oxidative stress (Ko et al., 2019; Yin et al., 2019). This state of oxidative stress is governed by the balance between ROS production and their elimination by antioxidants. Since the cell membrane is impermeable to urate anion, cellular concentration of urate depends on the specific transporter of urate and xanthine oxidoreductase (XOR). In mammals, this enzyme exists in two forms, xanthine dehydrogenase (XDH) and XO. XDH transfers electron to NAD+ and generates NADH, and XO transfers electron to O2 and generates oxidative stress (Isaka et al., 2016). In addition, XOR may lead to the production of the superoxide anion and nitric oxide, especially under low pH or hypoxia conditions (Godber et al., 2000; Battelli et al., 2019). It can be seen that the generation of UA mediated by XO is closely related to the production of ROS. On the other hand, antioxidant enzymes that scavenge ROS are ubiquitous, including superoxide dismutase, glutathione peroxidase and catalase, the changes of these enzymatic activities may lead to oxidative stress. However, in inflammatory diseases including hyperuricemia, the production of the superoxide anion is often faster than its removal by superoxide dismutase (Zamudio-Cuevas et al., 2015).

There is an increasing interest in using herbs to resolve the inflammatory conditions, including hyperuricemia, because they play an anti-inflammatory role by inhibiting the production of ROS, such as procyanidins, Shizhifang, Zisheng Shenqi decoction, quercetin, Modified Simiao decoction and other herbs included in this review. Antioxidants like quercetin inhibits inflammation in rat models of chronic MSU-induced arthritis by decreasing inflammatory mediators such as IL-1β (Huang et al., 2012). Other herbs mentioned above also show significant antioxidant effects. For example, Shizhifang effectively suppresses the NLRP3-ASC-caspase-1 axis through accommodating the ROS pathway, thereby alleviates potassium oxonate - induced hyperuricemia (Wu et al., 2017). In summary, the herbs that possess anti-oxidant effects may be developed as anti-inflammatory agents to treat inflammatory diseases such as gout and hyperuricemia.

Limitations and Perspectives From the Experimental Studies

As shown in Tables 35, there are fewer studies on the active ingredients of herbs than on the herbal extracts and herbal formulas. Aside from herbal formula, there can be hundreds of active ingredients in a single-flavored herb, making it difficult to identify the ones that actually mediate the therapeutic effects. This partly explains why multiple signaling pathways are involved in herbal treatments. Extraction of components that mediate the therapeutic effects is difficult. In general, safe and effective single-flavored herb can be screened out from literatures or experimental studies, and its effective active ingredients can be separated by pharmacological methods. Then, the mechanism of action of the drug can be explored based on the identified active ingredients from the herbs. In recent years, there are new approaches to identify the active ingredients of herbs and predict their targets, such as Systems Pharmacology. Researchers used Systems Pharmacology as a basis to screen out active ingredients from herbal formulas or herbs, and predict the targets of active ingredients, and finally verify them through in vivo and in vitro experimental studies (Zhou et al., 2016; Hong et al., 2017; Zhao et al., 2019). It really opens up new ideas for the studies of herbs and shortens the time to find out the mechanisms of action underlying the therapeutic effects of the herbs. However, with the increasing number of herbal studies using Systems Pharmacology, we can find that it also has disadvantages, such as limited Systems Pharmacology databases, data not updated, limited prediction results and so on. In view of this, it is still necessary to explore how to efficiently screen out the active ingredients of the herbs and identify their molecular targets.

Conclusion

In conclusion, the results of meta-analysis indicate that Chinese herbal medicines have potent therapeutic effects in lowering sUA levels. The signaling pathways involved in the sUA lowering effects include UA synthesis, UA transport, inflammation, renal fibrosis and oxidative stress. Further studies with sophisticated research design can further demonstrate the efficacy and safety of these Chinese herbal medicines in lowering sUA levels. Identification of the active ingredients and delineation of the underlying mechanisms of action can facilitate the clinical translation and application of these ingredients.

Author Contributions

All the authors contributed sufficiently for their participation in the study, as follows: XZ and LZ conceived, designed, and supervised the study; LC, ZL, MW, JC, FL, and HL conducted the literature search, data extraction, data analysis and interpretation; QH, YY, and XZ appraised the articles; LC, ZL, and MW wrote the paper, HK modified the paper. All authors have read and approved the final manuscript.

Funding

This work was supported by the Key Project of National Natural Science Foundation of China (No. 81830117), the National Science Foundation of China (Nos. 81774212, 81760821, 81703952), the Natural Science Foundation of Guangdong Province, China (Nos. 2017A030313722, 2018A030313375, 2019A1515010400), and the Science & Technical Plan of Guangzhou, Guangdong, China (No.201903010069).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

Abbreviations can be referred to Tables 3, 4. FABP1, fatty acid-binding protein; HPRT1, hypoxanthine-guanine phosphoribosyl transferase; APOB, apolipoprotein B; FOS, one subunit of activator protien-1; FN1, fibronectin1; MIP-1α, MIP-1 β, serum proinflammatory cytokines; TXNIP, thioredoxin interacting proteins; PTEN, phosphate and tension homology deleted on chromsome ten; NALP1/6, NACHT, LRR and PYD domains-containing protein one and six; ADAMTs, a family of metalloproteinases with thrombospondin motifs; TIMP, tissue inhibitor of metalloproteinase; CD2AP, CD2-associated protein.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fphar.2020.578318/full#supplementary-material.

References

Adachi, S. I., Nihei, K. I., Ishihara, Y., Yoshizawa, F., and Yagasaki, K. (2017). Anti-hyperuricemic effect of taxifolin in cultured hepatocytes and model mice. Cytotechnology. 69 (2), 329–336. doi:10.1007/s10616-016-0061-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Amat, N., Umar, A., Hoxur, P., Anaydulla, M., Imam, G., Aziz, R., et al. (2015). Traditional Uighur Medicine Karapxa decoction, inhibits liver xanthine oxidase and reduces serum uric acid concentrations in hyperuricemic mice and scavenges free radicals in vitro. BMC Compl. Alternative Med. 15, 131. doi:10.1186/s12906-015-0644-1

CrossRef Full Text | Google Scholar

Anzai, N., Ichida, K., Jutabha, P., Kimura, T., Babu, E., Jin, C. J., et al. (2008). Plasma urate level is directly regulated by a voltage-driven urate efflux transporter URATv1 (SLC2A9) in humans. J. Biol. Chem. 283 (40), 26834–26838. doi:10.1074/jbc.C800156200

PubMed Abstract | CrossRef Full Text | Google Scholar

Azevedo, V. F., Kos, I. A., Vargas-Santos, A. B., da Rocha Castelar Pinheiro, G., and Dos Santos Paiva, E. (2019). Benzbromarone in the treatment of gout. Adv Rheumatol. 59 (1), 37. doi:10.1186/s42358-019-0080-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Battelli, M. G., Bortolotti, M., Polito, L., and Bolognesi, A. (2018). The role of xanthine oxidoreductase and uric acid in metabolic syndrome. Biochim. Biophys. Acta (BBA) - Mol. Basis Dis. 1864 (8), 2557–2565. doi:10.1016/j.bbadis.2018.05.003

CrossRef Full Text | Google Scholar

Battelli, M. G., Bortolotti, M., Polito, L., and Bolognesi, A. (2019). Metabolic syndrome and cancer risk: the role of xanthine oxidoreductase. Redox Biol. 21, 101070. doi:10.1016/j.redox.2018.101070

PubMed Abstract | CrossRef Full Text | Google Scholar

Burke, B. T., Kottgen, A., Law, A., Windham, B. G., Segev, D., Baer, A. N., et al. (2015). Physical function, hyperuricemia, and gout in older adults. Arthritis Care Res. 67 (12), 1730–1738. doi:10.1002/acr.22648

PubMed Abstract | CrossRef Full Text | Google Scholar

Cammalleri, L., and Malaguarnera, M. (2007). Rasburicase represents a new tool for hyperuricemia in tumor lysis syndrome and in gout. Int. J. Med. Sci. 4 (2), 83–93. doi:10.7150/ijms.4.83

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, W., Xu, D., Wu, W., Xu, T., and Li, T. (2011). Experimental study on anti-inflammation and analgesic effects of extracts of compound Shuiniujiao. Pharmaceut. Care Res. 11 (2), 103–106. doi:10.5428/pcar20110209

CrossRef Full Text | Google Scholar

Caulfield, M. J., Munroe, P. B., O'Neill, D., Witkowska, K., Charchar, F. J., Doblado, M., et al. (2008). SLC2A9 is a high-capacity urate transporter in humans. PLoS Med. 5 (10), e197. doi:10.1371/journal.pmed.0050197

PubMed Abstract | CrossRef Full Text | Google Scholar

Chaudhary, K., Malhotra, K., Sowers, J., and Aroor, A. (2013). Uric Acid - key ingredient in the recipe for cardiorenal metabolic syndrome. Cardiorenal Med. 3 (3), 208–220. doi:10.1159/000355405

PubMed Abstract | CrossRef Full Text | Google Scholar

Chavarriaga, J., Ocampo, M., Fakih, N., and Silva Herrera, J. (2019). Kelley-seegmiller syndrome: urolithiasis, renal uric acid deposits, and gout: what is the role of the urologist?. Urol. Int. 102 (2), 233–237. doi:10.1159/000494360

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Q., Ma, L., and Akebaier, W. (2009). Clinical study on treatment of hyperuricaemia by retention enema of Chinese herbal medicine combined with allopurinol. Chin. J. Integr. Med. 15 (6), 431–434. doi:10.1007/s11655-009-0431-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J. W., Zhou, Y., Xue, Z. Y., Li, C., Guo, J., Zhou, L. Y., et al. (2013a). [Effect of jianpihuashi decoction on rats with hyperuricemia]. Zhong Yao Cai. 36 (9), 1486–1489

PubMed Abstract | Google Scholar

Chen, L., Mola, M., Deng, X., Mei, Z., Huang, X., Shu, G., et al. (2013b). Dolichos falcata Klein attenuated the inflammation induced by monosodium urate crystals in vivo and in vitro. J. Ethnopharmacol. 150 (2), 545–552. doi:10.1016/j.jep.2013.08.063

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, C. Y., Huang, C. C., Tsai, K. C., Huang, W. J., Huang, W. C., Hsu, Y. C., et al. (2014a). Evaluation of the antihyperuricemic activity of phytochemicals from Davallia formosana by enzyme assay and hyperuricemic mice model. Evid Based Complement Alternat Med. 2014, 873607. doi:10.1155/2014/873607

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, G. L., Wu, S., Na, S., and Li, L. (2014b). [Effect of total saponin of dioscorea on uric acid excretion indicators in chronic hyperuricemia rats]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 34 (1), 75–80

Google Scholar

Chen, Y., Lu, Y., Wang, Y. N., Lin, Z. C., Gu, W., Tan, L., et al. (2014c). [Effect of compound qingqin liquid on the expression of toll-like receptor in the renal tissue of rats with urate nephropathy]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 34 (6), 722–727

PubMed Abstract | Google Scholar

Chen, G., Tan, M. L., Li, K. K., Leung, P. C., and Ko, C. H. (2015a). Green tea polyphenols decreases uric acid level through xanthine oxidase and renal urate transporters in hyperuricemic mice. J. Ethnopharmacol. 175, 14–20. doi:10.1016/j.jep.2015.08.043

CrossRef Full Text | Google Scholar

Chen, W. J., Wu, Y., Xu, C., Liu, S., Wang, W. Z., Song, F. R., et al. (2015b). [Study on therapeutic effects of ermiao pill and ermiao pill categorized formula in hyperuricemic rats using spectroscopic methods]. Guang Pu Xue Yu Guang Pu Fen Xi. 35 (4), 956–960

Google Scholar

Chen, J., Zhou, J., Wei, S., Xie, Z., Wen, C., and Xu, G. (2016a). Effect of a traditional Chinese medicine prescription Quzhuotongbi decoction on hyperuricemia model rats studied by using serum metabolomics based on gas chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 1026, 272–278. doi:10.1016/j.jchromb.2015.10.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Chen, X. L., Xiang, T., Sun, B. G., Luo, H. X., Liu, M. T., et al. (2016b). Total saponins from dioscorea septemloba thunb reduce serum uric acid levels in rats with hyperuricemia through OATP1A1 up-regulation. J Huazhong Univ Sci Technolog Med Sci. 36 (2), 237–242. doi:10.1007/s11596-016-1573-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, W. J., Wu, Y., Bi, R. B., Liu, S., Liu, Z. Y., Liu, Z. Q., et al. (2017a). Therapeutic effects of selaginella tamariscina on the model of acute gout with hyperuricemia in rats based on metabolomics analysis. Chin. J. Chem. 35 (7), 1117–1124. doi:10.1002/cjoc.201600810

CrossRef Full Text | Google Scholar

Chen, Y. S., Chen, C. J., Yan, W., Ge, H. M., and Kong, L. D. (2017b). Anti-hyperuricemic and anti-inflammatory actions of vaticaffinol isolated from Dipterocarpus alatus in hyperuricemic mice. Chin. J. Nat. Med. 15 (5), 330–340. doi:10.1016/s1875-5364(17)30053-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, B., Li, H., Ou, G., Ren, L., Yang, X., and Zeng, M. (2019a). Curcumin attenuates MSU crystal-induced inflammation by inhibiting the degradation of IkappaBalpha and blocking mitochondrial damage. Arthritis Res. Ther. 21 (1), 193. doi:10.1186/s13075-019-1974-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H.-f., Zhang, C., Yao, Y., Li, J.-m., Du, W.-d., Li, M.-l., et al. (2019b). Study on anti-hyperuricemia effects and active ingredients of traditional Tibetan medicine TongFengTangSan (TFTS) by ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. J. Pharmaceut. Biomed. Anal. 165, 213–223. doi:10.1016/j.jpba.2018.11.038

CrossRef Full Text | Google Scholar

Chen, M., Ye, C., Zhu, J., Zhang, P., Jiang, Y., Lu, X., et al. (2020a). Bergenin as a novel urate-lowering therapeutic strategy for hyperuricemia. Frontiers in Cell and Developmental Biology. 8. doi:10.3389/fcell.2020.00703

CrossRef Full Text | Google Scholar

Chen, W. D., Zhao, Y. L., Sun, W. J., He, Y. J., Liu, Y. P., Jin, Q., et al. (2020b). Kidney tea" and its bioactive secondary metabolites for treatment of gout. J. Agric. Food Chem. doi:10.1021/acs.jafc.0c03848

CrossRef Full Text | Google Scholar

Chen, L., and Lan, Z. (2017). Polydatin attenuates potassium oxonate-induced hyperuricemia and kidney inflammation by inhibiting NF-kappaB/NLRP3 inflammasome activation via the AMPK/SIRT1 pathway. Food Funct. 8 (5), 1785–1792. doi:10.1039/c6fo01561a

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen-Xu, M., Yokose, C., Rai, S. K., Pillinger, M. H., and Choi, H. K. (2019). Contemporary prevalence of gout and hyperuricemia in the United States and decadal trends: the national health and nutrition examination survey, 2007-2016. Arthritis Rheum 71 (6), 991–999. doi:10.1002/art.40807

CrossRef Full Text | Google Scholar

Cheng, L. C., Murugaiyah, V., and Chan, K. L. (2015). Flavonoids and phenylethanoid glycosides from Lippia nodiflora as promising antihyperuricemic agents and elucidation of their mechanism of action. J. Ethnopharmacol. 176, 485–493. doi:10.1016/j.jep.2015.11.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, S., Sun, H., Li, X., Yan, J., Peng, Z., You, Y., et al. (2019). Effects of alismatis rhizoma and rhizoma smilacis glabrae decoction on hyperuricemia in rats. Evid. base Compl. Alternative Med. 2019, 1–12. doi:10.1155/2019/4541609

CrossRef Full Text | Google Scholar

Cho, S. S., Song, S. H., Choi, C. Y., Park, K. M., Shim, J. H., and Park, D. H. (2018). Optimization of the extraction conditions and biological evaluation of dendropanax morbifera H. Lev as an anti-hyperuricemic source. Molecules. 23 (12), 3313. doi:10.3390/molecules23123313

CrossRef Full Text | Google Scholar

Choi, H. K., McCormick, N., Lu, N., Rai, S. K., Yokose, C., and Zhang, Y. (2020). Population impact attributable to modifiable risk factors for hyperuricemia. Arthritis Rheum 72 (1), 157–165. doi:10.1002/art.41067

CrossRef Full Text | Google Scholar

Cui, D., Liu, S., Tang, M., Lu, Y., Zhao, M., Mao, R., et al. (2020). Phloretin ameliorates hyperuricemia-induced chronic renal dysfunction through inhibiting NLRP3 inflammasome and uric acid reabsorption. Phytomedicine. 66, 153111. doi:10.1016/j.phymed.2019.153111

PubMed Abstract | CrossRef Full Text | Google Scholar

Dalbeth, N., Merriman, T. R., and Stamp, L. K. (2016). Gout. Lancet. 388 (10055), 2039–2052. doi:10.1016/s0140-6736(16)00346-9

CrossRef Full Text

Dang, Y. X., Liang, D. L., Zhou, X. X., Qin, Y., Gao, Y., and Li, W. M. (2019). Protective effect of Mori Cortex on kidney in rats with hyperlipidemia and hyperuricemia based on molecular docking technique. Chin. Tradit. Herb. Drugs. 50 (5), 1175–1181. doi:10.7501/j.issn.0253-2670.2019.05.022

CrossRef Full Text | Google Scholar

Dehghan, A., Kottgen, A., Yang, Q., Hwang, S. J., Kao, W. L., Rivadeneira, F., et al. (2008). Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study. Lancet. 372 (9654), 1953–1961. doi:10.1016/s0140-6736(08)61343-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Dhanasekar, C., and Rasool, M. (2016). Morin, a dietary bioflavonol suppresses monosodium urate crystal-induced inflammation in an animal model of acute gouty arthritis with reference to NLRP3 inflammasome, hypo-xanthine phospho-ribosyl transferase, and inflammatory mediators. Eur. J. Pharmacol. 786, 116–127. doi:10.1016/j.ejphar.2016.06.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Diamond, H. S., and Paolino, J. S. (1973). Evidence for a postsecretory reabsorptive site for uric acid in man. J. Clin. Invest. 52 (6), 1491–1499. doi:10.1172/jci107323

PubMed Abstract | CrossRef Full Text | Google Scholar

Ding, X. Q., Pan, Y., Wang, X., Ma, Y. X., and Kong, L. D. (2013). Wuling san ameliorates urate under-excretion and renal dysfunction in hyperuricemic mice. Chin. J. Nat. Med. 11 (3), 214–221. doi:10.1016/s1875-5364(13)60019-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Dinour, D., Bahn, A., Ganon, L., Ron, R., Geifman-Holtzman, O., Knecht, A., et al. (2011). URAT1 mutations cause renal hypouricemia type 1 in Iraqi Jews. Nephrol. Dial. Transplant. 26 (7), 2175–2181. doi:10.1093/ndt/gfq722

PubMed Abstract | CrossRef Full Text | Google Scholar

El Ridi, R., and Tallima, H. (2017). Physiological functions and pathogenic potential of uric acid: a review. J. Adv. Res. 8 (5), 487–493. doi:10.1016/j.jare.2017.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Enomoto, A., Kimura, H., Chairoungdua, A., Shigeta, Y., Jutabha, P., Cha, S. H., et al. (2002). Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature. 417 (6887), 447–452. doi:10.1038/nature742

PubMed Abstract | CrossRef Full Text | Google Scholar

Fan, H., Zhao, Y., Fu, J., Zhang, H., Wang, D., Gao, Y., et al. (2018). Anti-inflammatory effect of Tongfengning Capsule in rats with acute gouty arthritis and its mechanism. J. Jilin Univ. - Med. Ed. 44 (2), 270–274. doi:10.13481/j.1671-587x.20180212

CrossRef Full Text | Google Scholar

Fan, S., Zhang, P., Wang, A. Y., Wang, X., Wang, L., Li, G., et al. (2019). Hyperuricemia and its related histopathological features on renal biopsy. BMC Nephrol. 20 (1), 95. doi:10.1186/s12882-019-1275-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, C., Chen, L., He, M., Luo, Y., Zhou, M., Zhang, N., et al. (2019). Molecular mechanistic insight into the anti-hyperuricemic effect of Eucommia ulmoides in mice and rats. Pharm. Biol. 57 (1), 112–119. doi:10.1080/13880209.2019.1568510

PubMed Abstract | CrossRef Full Text | Google Scholar

Fei, Y., Ye, D., Fan, X. F., and Dong, F. Q. (2016). Effect of Dioscorea tokoro Makino extract on hyperuricemia in mice. Trop. J. Pharmaceut. Res. 15 (9), 1883–1887. doi:10.4314/tjpr.v15i9.10

CrossRef Full Text | Google Scholar

Fu, R., Ceballos-Picot, I., Torres, R. J., Larovere, L. E., Yamada, Y., Nguyen, K. V., et al. (2014). Genotype-phenotype correlations in neurogenetics: lesch-Nyhan disease as a model disorder. Brain. 137 (Pt 5), 1282–1303. doi:10.1093/brain/awt202

PubMed Abstract | CrossRef Full Text | Google Scholar

Gamala, M., and Jacobs, J. W. G. (2019). Gout and hyperuricaemia: a worldwide health issue of joints and beyond. Rheumatology.. doi:10.1093/rheumatology/kez272

CrossRef Full Text | Google Scholar

Godber, B. L., Doel, J. J., Sapkota, G. P., Blake, D. R., Stevens, C. R., Eisenthal, R., et al. (2000). Reduction of nitrite to nitric oxide catalyzed by xanthine oxidoreductase. J. Biol. Chem. 275 (11), 7757–7763. doi:10.1074/jbc.275.11.7757

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Y., Jiang, Q., Gui, D., and Wang, N. (2015). Chinese herbal formulas Si-Wu-tang and Er-Miao-San synergistically ameliorated hyperuricemia and renal impairment in rats induced by adenine and potassium oxonate. Cell. Physiol. Biochem. 37 (4), 1491–1502. doi:10.1159/000438517

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, L. F., Chen, X., Lei, S. S., Li, B., Zhang, N. Y., Ge, H. Z., et al. (2020). Effects and mechanisms of dendrobium officinalis six nostrum for treatment of hyperuricemia with hyperlipidemia. Evid Based Complement Alternat Med. 2020, 2914019. doi:10.1155/2020/2914019

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, J., Xie, Y., Sui, F., Liu, C., Du, X., Liu, C., et al. (2016). Zisheng Shenqi decoction ameliorates monosodium urate crystal-induced gouty arthritis in rats through anti-inflammatory and anti-oxidative effects. Mol. Med. Rep. 14 (3), 2589–2597. doi:10.3892/mmr.2016.5526

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, B., Zhu, C. X., Shi, W., Huang, H. Z., Hu, X. G., Zhou, X. M., et al. (2017). Effect of Rhizoma Polygoni Cuspidati and Ramulus Cinnamomi compatibility on uric acid metabolism and urinary neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 in rats with hyperuricemia. Chin. J. Integr. Med. 23 (7), 535–542. doi:10.1007/s11655-016-2649-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, B., Gong, M., Li, Z., Qiu, Y., and Zou, Z. (2020). NMR-based metabonomic study reveals intervention effects of polydatin on potassium oxonate-induced hyperuricemia in rats. Oxid Med Cell Longev. 2020, 6943860. doi:10.1155/2020/6943860

PubMed Abstract | CrossRef Full Text | Google Scholar

Harris, J. C. (2018). Lesch-Nyhan syndrome and its variants: examining the behavioral and neurocognitive phenotype. Curr. Opin. Psychiatr. 31 (2), 96–102. doi:10.1097/yco.0000000000000388

CrossRef Full Text | Google Scholar

Ho, S. T., Tung, Y. T., Huang, C. C., Kuo, C. L., Lin, C. C., Yang, S. C., et al. (2012). The hypouricemic effect of Balanophora laxiflora extracts and derived phytochemicals in hyperuricemic mice. Evid Based Complement Alternat Med. 2012, 910152. doi:10.1155/2012/910152

PubMed Abstract | CrossRef Full Text | Google Scholar

Honda, S., Kawamoto, S., Tanaka, H., Kishida, H., Kitagawa, M., Nakai, Y., et al. (2014). Administered chrysanthemum flower oil attenuates hyperuricemia: mechanism of action as revealed by DNA microarray analysis. Biosci. Biotechnol. Biochem. 78 (4), 655–661. doi:10.1080/09168451.2014.890028

PubMed Abstract | CrossRef Full Text | Google Scholar

Hong, Q., Yu, S., Mei, Y., Lv, Y., Chen, D., Wang, Y., et al. (2014). Smilacis Glabrae Rhizoma reduces oxidative stress caused by hyperuricemia via upregulation of catalase. Cell. Physiol. Biochem. 34 (5), 1675–1685. doi:10.1159/000366369

PubMed Abstract | CrossRef Full Text | Google Scholar

Hong, M., Li, S., Wang, N., Tan, H. Y., Cheung, F., and Feng, Y. (2017). A biomedical investigation of the hepatoprotective effect of radix salviae miltiorrhizae and network pharmacology-based prediction of the active compounds and molecular targets. Int. J. Mol. Sci. 18 (3). doi:10.3390/ijms18030620

CrossRef Full Text | Google Scholar

Hou, C. W., Lee, Y. C., Hung, H. F., Fu, H. W., and Jeng, K. C. (2012). Longan seed extract reduces hyperuricemia via modulating urate transporters and suppressing xanthine oxidase activity. Am. J. Chin. Med. 40 (5), 979–991. doi:10.1142/s0192415x12500723

PubMed Abstract | CrossRef Full Text | Google Scholar

Hou, P. Y., Mi, C., He, Y., Zhang, J., Wang, S. Q., Yu, F., et al. (2015). Pallidifloside D from Smilax riparia enhanced allopurinol effects in hyperuricemia mice. Fitoterapia. 105, 43–48. doi:10.1016/j.fitote.2015.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Q. H., Jiao, R. Q., Wang, X., Lv, Y. Z., and Kong, L. D. (2010). Simiao pill ameliorates urate underexcretion and renal dysfunction in hyperuricemic mice. J. Ethnopharmacol. 128 (3), 685–692. doi:10.1016/j.jep.2010.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Hua, J., Huang, P., Zhu, C. M., Yuan, X., and Yu, C. H. (2012). Anti-hyperuricemic and nephroprotective effects of Modified Simiao Decoction in hyperuricemic mice. J. Ethnopharmacol. 142 (1), 248–252. doi:10.1016/j.jep.2012.04.052

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Q. H., Miao, M. X., Lu, G., and Ji, H. (2013a). Effects of quercetin on expression of renal NLRP3 and TLRs in rats with uric acid nephtopathy. Chin. Tradit. Herb. Drugs. 44 (24), 3496–3502. doi:10.7501/j.issn.0253-2670.2013.24.013

CrossRef Full Text | Google Scholar

Hu, Q. H., Zhu, J. X., Ji, J., Wei, L. L., Miao, M. X., and Ji, H. (2013b). Fructus Gardenia Extract ameliorates oxonate-induced hyperuricemia with renal dysfunction in mice by regulating organic ion transporters and mOIT3. Molecules. 18 (8), 8976–8993. doi:10.3390/molecules18088976

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, J., Zhu, M., Tao, Y., Wang, S., Chen, J., Sun, W., et al. (2012). Therapeutic properties of quercetin on monosodium urate crystal-induced inflammation in rat. J. Pharm. Pharmacol. 64 (8), 1119–1127. doi:10.1111/j.2042-7158.2012.01504.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, B., Hu, X., Wang, J., Li, P., and Chen, J. (2019a). Study on chemical constituents of herbal formula Er Miao Wan and GC-MS based metabolomics approach to evaluate its therapeutic effects on hyperuricemic rats. J Chromatogr B Analyt Technol Biomed Life Sci. 1118–1119, 101–108. doi:10.1016/j.jchromb.2019.04.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, L., Deng, J., Chen, G., Zhou, M., Liang, J., Yan, B., et al. (2019b). The anti-hyperuricemic effect of four astilbin stereoisomers in Smilax glabra on hyperuricemic mice. J. Ethnopharmacol. 238, 111777. doi:10.1016/j.jep.2019.03.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Hui, W., Yongliang, Y., Yongde, C., Guo, L., Li, L., Zhonglin, Y., et al. (2016). Hypouricemic and nephroprotective effects of emodinol in oxonate-induced hyperuricemic mice are mediated by organic ion transporters and OIT3. Planta Med. 82 (4), 289–297. doi:10.1055/s-0035-1558212

PubMed Abstract | CrossRef Full Text | Google Scholar

Huijuan, W., Xiaoxu, C., Rui, S., Xinghui, L., Beibei, T., and Jianchun, M. (2017). Qi-Zhu-Xie-Zhuo-Fang reduces serum uric acid levels and ameliorates renal fibrosis in hyperuricemic nephropathy rats. Biomed. Pharmacother. 91, 358–365. doi:10.1016/j.biopha.2017.04.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Huo, L. N., Wang, W., Zhang, C. Y., Shi, H. B., Liu, Y., Liu, X. H., et al. (2015). Bioassay-guided isolation and identification of xanthine oxidase inhibitory constituents from the leaves of perilla frutescens. Molecules. 20 (10), 17848–17859. doi:10.3390/molecules201017848

PubMed Abstract | CrossRef Full Text | Google Scholar

Isaka, Y., Takabatake, Y., Takahashi, A., Saitoh, T., and Yoshimori, T. (2016). Hyperuricemia-induced inflammasome and kidney diseases. Nephrol. Dial. Transplant. 31 (6), 890–896. doi:10.1093/ndt/gfv024

PubMed Abstract | CrossRef Full Text | Google Scholar

Jhang, J. J., Ong, J. W., Lu, C. C., Hsu, C. L., Lin, J. H., Liao, J. W., et al. (2016). Hypouricemic effects of Mesona procumbens Hemsl. through modulating xanthine oxidase activity in vitro and in vivo. Food Funct. 7 (10), 4239–4246. doi:10.1039/c6fo00822d

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, Y., You, X. Y., Fu, K. L., and Yin, W. L. (2012). Effects of extract from mangifera indica leaf on monosodium urate crystal-induced gouty arthritis in rats. Evid Based Complement Alternat Med. 2012, 967573. doi:10.1155/2012/967573

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, Y., Lin, Y., Hu, Y. J., Song, X. J., Pan, H. H., and Zhang, H. J. (2017). Caffeoylquinic acid derivatives rich extract from Gnaphalium pensylvanicum willd. Ameliorates hyperuricemia and acute gouty arthritis in animal model. BMC Compl. Alternative Med. 17 (1), 320. doi:10.1186/s12906-017-1834-9

CrossRef Full Text | Google Scholar

Kang, L., Miao, J. X., Cao, L. H., Miao, Y. Y., Miao, M. S., Liu, H. J., et al. (2020). Total glucosides of herbaceous peony (Paeonia lactiflora Pall.) flower attenuate adenine- and ethambutol-induced hyperuricaemia in rats. J. Ethnopharmacol. 261, 113054. doi:10.1016/j.jep.2020.113054

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, S. C., Liu, J., and Solomon, D. H. (2015). Risk of incident diabetes in patients with gout: a cohort study. Arthritis Rheum 67 (1), 273–280. doi:10.1002/art.38918

CrossRef Full Text | Google Scholar

Kim, J. K., Kim, W. J., Hyun, J. M., Lee, J. S., Kwon, J. G., Seo, C., et al. (2017). Salvia plebeia extract inhibits xanthine oxidase activity in vitro and reduces serum uric acid in an animal model of hyperuricemia. Planta Med. 83 (17), 1335–1341. doi:10.1055/s-0043-111012

PubMed Abstract | CrossRef Full Text | Google Scholar

Ko, J., Kang, H. J., Kim, D. A., Kim, M. J., Ryu, E. S., Lee, S., et al. (2019). Uric acid induced the phenotype transition of vascular endothelial cells via induction of oxidative stress and glycocalyx shedding. Faseb. J., fj201901148R. doi:10.1096/fj.201901148R

CrossRef Full Text | Google Scholar

Kodithuwakku, N. D., Pan, M., Zhu, Y. L., Zhang, Y. Y., Feng, Y. D., Fang, W. R., et al. (2013). Anti-inflammatory and antinociceptive effects of Chinese medicine SQ gout capsules and its modulation of pro-inflammatory cytokines focusing on gout arthritis. J. Ethnopharmacol. 150 (3), 1071–1079. doi:10.1016/j.jep.2013.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Kodithuwakku, N. D., Feng, Y. D., Zhang, Y. Y., Pan, M., Fang, W. R., and Li, Y. M. (2015). The molecular insight into the antihyperuricemic and renoprotective effect of Shuang Qi gout capsule in mice. J. Ethnopharmacol. 163, 278–289. doi:10.1016/j.jep.2015.01.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Kondo, M., Hirano, Y., Nishio, M., Furuya, Y., Nakamura, H., and Watanabe, T. (2013). Xanthine oxidase inhibitory activity and hypouricemic effect of aspalathin from unfermented rooibos. J. Food Sci. 78 (12), H1935–H1939. doi:10.1111/1750-3841.12304

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuo, C.-Y., Kao, E.-S., Chan, K.-C., Lee, H.-J., Huang, T.-F., and Wang, C.-J. (2012). Hibiscus sabdariffa L. extracts reduce serum uric acid levels in oxonate-induced rats. Journal of Functional Foods. 4 (1), 375–381. doi:10.1016/j.jff.2012.01.007

CrossRef Full Text | Google Scholar

Kou, Y. Y., Li, Y. F., Xu, M., Li, W. Y., Yang, M., and Li, R. L. (2015). Effects of RuPeng15 powder (RPP15) on monosodium urate crystal-induced gouty arthritis in rats. Evid Based Complement Alternat Med. 2015, 527019. doi:10.1155/2015/527019

PubMed Abstract | CrossRef Full Text | Google Scholar

Kou, Y., Li, Y., Ma, H., Li, W., Li, R., and Dang, Z. (2016). Uric acid lowering effect of Tibetan Medicine RuPeng15 powder in animal models of hyperuricemia. J. Tradit. Chin. Med. 36 (2), 205–210

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuwabara, M., Hisatome, I., Niwa, K., Hara, S., Roncal-Jimenez, C. A., Bjornstad, P., et al. (2018). Uric acid is a strong risk marker for developing hypertension from prehypertension: a 5-year Japanese cohort study. Hypertension. 71 (1), 78–86. doi:10.1161/hypertensionaha.117.10370

PubMed Abstract | CrossRef Full Text | Google Scholar

Lanaspa, M. A., Sanchez-Lozada, L. G., Choi, Y. J., Cicerchi, C., Kanbay, M., Roncal-Jimenez, C. A., et al. (2012). Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: potential role in fructose-dependent and -independent fatty liver. J. Biol. Chem. 287 (48), 40732–40744. doi:10.1074/jbc.M112.399899

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. P., Lin, Y. Y., Duh, C. Y., Huang, S. Y., Wang, H. M., Wu, S. F., et al. (2015). Lemnalol attenuates mast cell activation and osteoclast activity in a gouty arthritis model. J. Pharm. Pharmacol. 67 (2), 274–285. doi:10.1111/jphp.12331

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, Y. M., Shon, E. J., Kim, O. S., and Kim, D. S. (2017). Effects of Mollugo pentaphylla extract on monosodium urate crystal-induced gouty arthritis in mice. BMC Compl. Alternative Med. 17 (1), 447. doi:10.1186/s12906-017-1955-1

CrossRef Full Text | Google Scholar

Lee, H. A., Yu, K. S., Park, S. I., Yoon, S., Onohara, M., Ahn, Y., et al. (2019a). URC102, a potent and selective inhibitor of hURAT1, reduced serum uric acid in healthy volunteers. Rheumatology.. doi:10.1093/rheumatology/kez140

CrossRef Full Text | Google Scholar

Lee, Y. S., Sung, Y. Y., Yuk, H. J., Son, E., Lee, S., Kim, J. S., et al. (2019b). Anti-hyperuricemic effect of Alpinia oxyphylla seed extract by enhancing uric acid excretion in the kidney. Phytomedicine. 62, 152975. doi:10.1016/j.phymed.2019.152975

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y. Q., Wei, Y. H., and Han, X. (2010). Effects of Danggui Buxue Tang on heart muscle function in hyperuricemic rats. Chin. Pharmaceut. J. 45 (7), 524–526

Google Scholar

Li, H. G., Hou, P. Y., Zhang, X., He, Y., Zhang, J., Wang, S. Q., et al. (2016). Hypouricemic effect of allopurinol are improved by Pallidifloside D based on the uric acid metabolism enzymes PRPS, HGPRT and PRPPAT. Fitoterapia. 113, 1–5. doi:10.1016/j.fitote.2016.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, L., Teng, M., Liu, Y., Qu, Y., Zhang, Y., Lin, F., et al. (2017a). Anti-Gouty arthritis and antihyperuricemia effects of sunflower (helianthus annuus) head extract in gouty and hyperuricemia animal models. BioMed Res. Int. 2017, 5852076. doi:10.1155/2017/5852076

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y. P., Wu, S., Ran, A., Xu, D. Y., Wei, J. M., and Zhao, Z. L. (2017b). Aristolochia bracteolate retz. Attenuates hyperuricemia IN a metabolic arthritis rat model. Afr. J. Tradit., Complementary Altern. Med. 14 (4), 180–187. doi:10.21010/ajtcam.v14i4.21

CrossRef Full Text | Google Scholar

Li, Z. J., Li, Z., Dong, X. Y., Lu, L. F., and Wang, C. L. (2017c). Hypouricemic and nephroprotective effects of total flavonoids from the residue of supercritical CO2 extraction of Humulus lupulus in potassium oxonate-induced mice. Pak. J. Pharm. Sci. 30 (2), 493–497

PubMed Abstract | Google Scholar

Liang, G., Nie, Y., Chang, Y., Zeng, S., Liang, C., Zheng, X., et al. (2019). Protective effects of Rhizoma smilacis glabrae extracts on potassium oxonate- and monosodium urate-induced hyperuricemia and gout in mice. Phytomedicine. 59, 152772. doi:10.1016/j.phymed.2018.11.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Lima, W. G., Martins-Santos, M. E., and Chaves, V. E. (2015). Uric acid as a modulator of glucose and lipid metabolism. Biochimie. 116, 17–23. doi:10.1016/j.biochi.2015.06.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, J., Chen, S., Li, S., Lu, M., Li, Y., and Su, Y. (2016). Efficacy and safety of Chinese medicinal herbs for the treatment of hyperuricemia: a systematic review and meta-analysis. Evid Based Complement Alternat Med. 2016, 2146204. doi:10.1155/2016/2146204

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, Y., Liu, P. G., Liang, W. Q., Hu, Y. J., Xu, P., Zhou, J., et al. (2018). Luteolin-4'-O-glucoside and its aglycone, two major flavones of Gnaphalium affine D. Don, resist hyperuricemia and acute gouty arthritis activity in animal models. Phytomedicine. 41, 54–61. doi:10.1016/j.phymed.2018.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, X., Shao, T., Huang, L., Wen, X., Wang, M., Wen, C., et al. (2020). Simiao decoction alleviates gouty arthritis by modulating proinflammatory cytokines and the gut ecosystem. Front. Pharmacol. 11, 955. doi:10.3389/fphar.2020.00955

PubMed Abstract | CrossRef Full Text | Google Scholar

Lipkowitz, M. S. (2012). Regulation of uric acid excretion by the kidney. Curr. Rheumatol. Rep. 14 (2), 179–188. doi:10.1007/s11926-012-0240-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Xu, L. L., and Xu, Y. (2013). Antigout active fractions in tongfeng granule. Chin. Tradit. Herb. Drugs. 44 (5), 590–594. doi:10.7501/j.issn.0253-2670.2013.05.018

CrossRef Full Text | Google Scholar

Liu, H., Zhang, X. M., Wang, Y. L., and Liu, B. C. (2014a). Prevalence of hyperuricemia among Chinese adults: a national cross-sectional survey using multistage, stratified sampling. J. Nephrol. 27 (6), 653–658. doi:10.1007/s40620-014-0082-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X. H., Huang, M. Q., Lin, Z. W., Zhen, H. S., Liu, A. T., and Zhang, K. Y. (2014b). Anti-gout effect of Poecilobdella manillensis. Chin. Tradit. Herb. Drugs. 45 (12), 1747–1750. doi:10.7501/j.issn.0253-2670.2014.12.018

CrossRef Full Text | Google Scholar

Liu, Y. W., Sun, W. F., Zhang, X. X., Li, J., and Zhang, H. H. (2015). Compound Tufuling Granules ([characters: see text]) regulate glucose transporter 9 expression in kidney to influence serum uric acid level in hyperuricemia mice. Chin. J. Integr. Med. 21 (11), 823–829. doi:10.1007/s11655-015-2052-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, H. J., Pan, X. X., Liu, B. Q., Gui, X., Hu, L., Jiang, C. Y., et al. (2017). Grape seed-derived procyanidins alleviate gout pain via NLRP3 inflammasome suppression. J. Neuroinflammation. 14 (1), 74. doi:10.1186/s12974-017-0849-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X. H., Zhao, Y. X., Zhou, Y. M., Huang, M. Q., Huang, S. S., Zhen, H. S., et al. (2018). Anti-gout effect of hirudin and its mechanism. Chin. Tradit. Herb. Drugs. 49 (6), 1365–1370. doi:10.7501/j.issn.0253-2670.2018.06.020

CrossRef Full Text | Google Scholar

Liu, N., Wang, Y., Yang, M., Bian, W., Zeng, L., Yin, S., et al. (2019). New rice-derived short peptide potently alleviated hyperuricemia induced by potassium oxonate in rats. J. Agric. Food Chem. 67 (1), 220–228. doi:10.1021/acs.jafc.8b05879

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, G., Chen, X., Lu, X., Zhao, J., and Li, X. (2020). Sunflower head enzymatic hydrolysate relives hyperuricemia by inhibiting crucial proteins (xanthine oxidase, adenosine deaminase, uric acid transporter1) and restoring gut microbiota in mice. Journal of Functional Foods. 72. doi:10.1016/j.jff.2020.104055

CrossRef Full Text | Google Scholar

Lo, H. C., Wang, Y. H., Chiou, H. Y., Lai, S. H., and Yang, Y. (2010). Relative efficacy of casein or soya protein combined with palm or safflower-seed oil on hyperuricaemia in rats. Br. J. Nutr. 104 (1), 67–75. doi:10.1017/s0007114510000310

PubMed Abstract | CrossRef Full Text | Google Scholar

Loeffler, I., and Wolf, G. (2015). Epithelial-to-Mesenchymal transition in diabetic nephropathy: fact or fiction?. Cells. 4 (4), 631–652. doi:10.3390/cells4040631

PubMed Abstract | CrossRef Full Text | Google Scholar

Lou, X. J., Wang, Y. Z., Lei, S. S., He, X., Lu, T. T., Zhan, L. H., et al. (2020). Beneficial effects of macroporous resin extract of dendrobium candidum leaves in rats with hyperuricemia induced by a high-purine diet. Evid Based Complement Alternat Med. 2020, 3086106. doi:10.1155/2020/3086106

PubMed Abstract | CrossRef Full Text | Google Scholar

Louxin, B., Miaoyanyan, F., and Miaomingsan, (2018). Effect of arctiin on rat hyperuricemia model. Acta Med. Mediterr. 34 (6), 1739–1742. doi:10.19193/0393-6384_2018_6_266

CrossRef Full Text | Google Scholar

Lü, Y. Z., Hu, Q. H., Wang, X., Ouyang, Z., and Kong, L. D. (2010). Effects of Ermiao Pill water extracts on imbalance of urate levels and its related genes and protein levels in hyperuricemic mice. Chin. Tradit. Herb. Drugs. 41 (3), 418–423

Google Scholar

Lu, F., Liu, L., Yu, D. H., Li, X. Z., Zhou, Q., and Liu, S. M. (2014). Therapeutic effect of Rhizoma Dioscoreae Nipponicae on gouty arthritis based on the SDF-1/CXCR 4 and p38 MAPK pathway: an in vivo and in vitro study. Phytother Res. 28 (2), 280–288. doi:10.1002/ptr.4997

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, X. Y., Zhou, F. H., Dong, Y. Q., Gong, L. N., Li, Q. Y., Tang, L., et al. (2018). Codonopsis tangshen oliv. Amelioration effect on diabetic kidney disease rats induced by high fat diet feeding combined with streptozotocin. Nat Prod Bioprospect. 8 (6), 441–451. doi:10.1007/s13659-018-0187-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lyu, S., Ding, R., Liu, P., OuYang, H., Feng, Y., Rao, Y., et al. (2019). LC-MS analysis of serum for the metabolomic investigation of the effects of pulchinenoside b4 administration in monosodium urate crystal-induced gouty arthritis rat model. Molecules. 24 (17). doi:10.3390/molecules24173161

CrossRef Full Text | Google Scholar

Ma, Y., Zhou, L. L., Yan, H. Y., and Liu, M. (2009). Effects of extracts from Paederia scandens (LOUR.) MERRILL (Rubiaceae) on MSU crystal-induced rats gouty arthritis. Am. J. Chin. Med. 37 (4), 669–683. doi:10.1142/s0192415x09007156

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, L., Zhang, S., Yuan, Y., and Gao, J. (2014). Hypouricemic actions of exopolysaccharide produced by Cordyceps militaris in potassium oxonate-induced hyperuricemic mice. Curr. Microbiol. 69 (6), 852–857. doi:10.1007/s00284-014-0666-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, C. H., Kang, L. L., Ren, H. M., Zhang, D. M., and Kong, L. D. (2015). Simiao pill ameliorates renal glomerular injury via increasing Sirt1 expression and suppressing NF-κB/NLRP3 inflammasome activation in high fructose-fed rats. J. Ethnopharmacol. 172, 108–117. doi:10.1016/j.jep.2015.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, T. H., Sheng, T., Tian, C. M., Xing, M. Y., Yan, L. J., and Xia, D. Z. (2019a). [Effect of ethanolic extract of Polygonum cuspidatum on acute gouty arthritis in mice through NLRP3/ASC/caspase-1 axis]. Zhongguo Zhongyao Zazhi. 44 (3), 546–552. doi:10.19540/j.cnki.cjcmm.20180925.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, W. G., Wang, J., Bu, X. W., Zhang, H. H., Zhang, J. P., Zhang, X. X., et al. (2019b). Effects of polygonum cuspidatum on AMPK-FOXO3alpha signaling pathway in rat model of uric acid-induced renal damage. Chin. J. Integr. Med. 25 (3), 182–189. doi:10.1007/s11655-017-2979-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, W. G., Wang, J., Bu, X. W., Zhang, H. H., Zhang, J. P., Zhang, X. X., et al. (2019c). Effects of polygonum cuspidatum on AMPK-FOXO3α signaling pathway in rat model of uric acid-induced renal damage. Chin. J. Integr. Med. 25 (3), 182–189. doi:10.1007/s11655-017-2979-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Maiuolo, J., Oppedisano, F., Gratteri, S., Muscoli, C., and Mollace, V. (2016). Regulation of uric acid metabolism and excretion. Int. J. Cardiol. 213, 8–14. doi:10.1016/j.ijcard.2015.08.109

PubMed Abstract | CrossRef Full Text | Google Scholar

Major, T. J., Dalbeth, N., Stahl, E. A., and Merriman, T. R. (2018). An update on the genetics of hyperuricaemia and gout. Nat. Rev. Rheumatol. 14 (6), 341–353. doi:10.1038/s41584-018-0004-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Mandal, A. K., and Mount, D. B. (2015). The molecular physiology of uric acid homeostasis. Annu. Rev. Physiol. 77, 323–345. doi:10.1146/annurev-physiol-021113-170343

PubMed Abstract | CrossRef Full Text | Google Scholar

Meng, Z., Yan, Y., Tang, Z., Guo, C., Li, N., Huang, W., et al. (2015). Anti-hyperuricemic and nephroprotective effects of rhein in hyperuricemic mice. Planta Med. 81 (4), 279–285. doi:10.1055/s-0034-1396241

PubMed Abstract | CrossRef Full Text | Google Scholar

Mittal, R., Patel, K., Mittal, J., Chan, B., Yan, D., Grati, M., et al. (2015). Association of PRPS1 mutations with disease phenotypes. Dis. Markers. 2015, 127013. doi:10.1155/2015/127013

PubMed Abstract | CrossRef Full Text | Google Scholar

Moher, D., Liberati, A., Tetzlaff, J., and Altman, D. G. (2009). Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann. Intern. Med. 151 (4), 264–269. doi:10.7326/0003-4819-151-4-200908180-00135

PubMed Abstract | CrossRef Full Text | Google Scholar

Niu, Y., Lu, W., Gao, L., Lin, H., Liu, X., and Li, L. (2012). Reducing effect of mangiferin on serum uric acid levels in mice. Pharm. Biol. 50 (9), 1177–1182. doi:10.3109/13880209.2012.663763

PubMed Abstract | CrossRef Full Text | Google Scholar

Niu, Y., Liu, J., Liu, H. Y., Gao, L. H., Feng, G. H., Liu, X., et al. (2016). Hypouricaemic action of mangiferin results from metabolite norathyriol via inhibiting xanthine oxidase activity. Pharm. Biol. 54 (9), 1680–1686. doi:10.3109/13880209.2015.1120322

PubMed Abstract | CrossRef Full Text | Google Scholar

Niu, Y., Zhou, Y., Lin, H., Gao, L. H., Xiong, W., Zhu, H., et al. (2018). Inhibition of 3,5,2',4'-tetrahydroxychalcone on production of uric acid in hypoxanthine-induced hyperuricemic mice. Biol. Pharm. Bull. 41 (1), 99–105. doi:10.1248/bpb.b17-00655

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, Y., Chu, Z., Wang, W., and Yang, B. (2013). Pretreatment with Jieduxiezhuo decoction impedes elevations in serum uric acid levels in mice. J. Tradit. Chin. Med. 33 (6), 794–797. doi:10.1016/s0254-6272(14)60014-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, H. Y., Shi, L., Xu, L., Yin, L., Zeng, W. P., Zhang, G. J., et al. (2014). Effect of active fractions from modified Simiao Wan on hyperuricemia and its mechanism. Chin. J. Pharmacol. Toxicol. 28 (3), 380–385. doi:10.3867/j.issn.1000-3002.2014.03.012

CrossRef Full Text | Google Scholar

Pan, J., Shi, M., Li, L., Liu, J., Guo, F., Feng, Y., et al. (2019). Pterostilbene, a bioactive component of blueberries, alleviates renal fibrosis in a severe mouse model of hyperuricemic nephropathy. Biomed. Pharmacother. 109, 1802–1808. doi:10.1016/j.biopha.2018.11.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, J., Zhang, C., Shi, M., Guo, F., Liu, J., Li, L., et al. (2020). Ethanol extract of Liriodendron chinense (Hemsl.) Sarg barks attenuates hyperuricemic nephropathy by inhibiting renal fibrosis and inflammation in mice. J. Ethnopharmacol. 113278. doi:10.1016/j.jep.2020.113278

CrossRef Full Text | Google Scholar

Pang, M., Fang, Y., Chen, S., Zhu, X., Shan, C., Su, J., et al. (2017). Gypenosides inhibits xanthine oxidoreductase and ameliorates urate excretion in hyperuricemic rats induced by high cholesterol and high fat food (lipid emulsion). Med. Sci. Mon. Int. Med. J. Exp. Clin. Res. 23, 1129–1140. doi:10.12659/msm.903217

CrossRef Full Text | Google Scholar

Patel, S. (2018). Danger-Associated molecular patterns (DAMPs): the derivatives and triggers of inflammation. Curr. Allergy Asthma Rep. 18 (11), 63. doi:10.1007/s11882-018-0817-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, A., Lin, L., Zhao, M., and Sun, B. (2019). Identifying mechanisms underlying the amelioration effect of Chrysanthemum morifolium Ramat. 'Boju' extract on hyperuricemia using biochemical characterization and UPLC-ESI-QTOF/MS-based metabolomics. Food Funct. 10 (12), 8042–8055. doi:10.1039/c9fo01821b

PubMed Abstract | CrossRef Full Text | Google Scholar

Preitner, F., Bonny, O., Laverriere, A., Rotman, S., Firsov, D., Da Costa, A., et al. (2009). Glut9 is a major regulator of urate homeostasis and its genetic inactivation induces hyperuricosuria and urate nephropathy. Proc. Natl. Acad. Sci. U. S. A. 106 (36), 15501–15506. doi:10.1073/pnas.0904411106

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, Z., Wang, S., Lin, Y., Zhao, Y., Yang, S., Song, J., et al. (2018). Antihyperuricemic effect of mangiferin aglycon derivative J99745 by inhibiting xanthine oxidase activity and urate transporter 1 expression in mice. Acta Pharm. Sin. B. 8 (2), 306–315. doi:10.1016/j.apsb.2017.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Rai, S. K., Burns, L. C., De Vera, M. A., Haji, A., Giustini, D., and Choi, H. K. (2015). The economic burden of gout: a systematic review. Semin. Arthritis Rheum 45 (1), 75–80. doi:10.1016/j.semarthrit.2015.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Rozza, F., Trimarco, V., Izzo, R., Grassi, D., and Ferri, C. (2016). Effects of a novel fixed combination of nutraceuticals on serum uric acid concentrations and the lipid profile in asymptomatic hyperuricemic patients : results from the PICONZ-UA study. High Blood Pres. Cardiovasc. Prev. 23 (4), 381–386. doi:10.1007/s40292-016-0168-x

CrossRef Full Text | Google Scholar

Sato, Y., Feig, D. I., Stack, A. G., Kang, D. H., Lanaspa, M. A., Ejaz, A. A., et al. (2019). The case for uric acid-lowering treatment in patients with hyperuricaemia and CKD. Nat. Rev. Nephrol. doi:10.1038/s41581-019-0174-z

CrossRef Full Text | Google Scholar

Shan, H. L., Shan, R. P., and Fu, X. C. (2015). [Hypouricemic effect of ethanol extracts from dioscoreae nipponicae rhizoma]. Zhejiang Da Xue Xue Bao Yi Xue Ban. 44 (1), 49–53

PubMed Abstract | Google Scholar

Shang, X. Z., Ma, W. G., Chen, Y., Lu, Y., Wang, Y. N., Xu, Y. M., et al. (2014). [Effect of compound qingqin liquid on the expression levels of ang II and COX-2 mRNA transcription and protein expression in the renal tissue of uric acid nephropathy rats: an experimental study]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 34 (7), 819–825

PubMed Abstract | Google Scholar

Sheu, S. Y., Fu, Y. T., Huang, W. D., Chen, Y. A., Lei, Y. C., Yao, C. H., et al. (2016). Evaluation of xanthine oxidase inhibitory potential and in vivo hypouricemic activity of dimocarpus longan lour. Extracts. Phcog. Mag. 12 (Suppl. 2), S206–S212. doi:10.4103/0973-1296.182176

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, Y. W., Wang, C. P., Wang, X., Zhang, Y. L., Liu, L., Wang, R. W., et al. (2012). Uricosuric and nephroprotective properties of Ramulus Mori ethanol extract in hyperuricemic mice. J. Ethnopharmacol. 143 (3), 896–904. doi:10.1016/j.jep.2012.08.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, L., Xu, L., Yang, Y., Song, H., Pan, H., and Yin, L. (2013). Suppressive effect of modified Simiaowan on experimental gouty arthritis: an in vivo and in vitro study. J. Ethnopharmacol. 150 (3), 1038–1044. doi:10.1016/j.jep.2013.10.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, L., Zhao, F., Zhu, F., Liang, Y., Yang, F., Zhang, G., et al. (2016a). Traditional Chinese Medicine Formula "Xiaofeng granules" suppressed gouty arthritis animal models and inhibited the proteoglycan degradation on chondrocytes induced by monosodium urate. J. Ethnopharmacol. 191, 254–263. doi:10.1016/j.jep.2016.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, Y. C., Lin, K. S., Jhai, Y. F., Lee, B. H., Han, Y., Cui, Z., et al. (2016b). Miracle fruit (synsepalum dulcificum) exhibits as a novel anti-hyperuricaemia agent. Molecules. 21 (2), 140. doi:10.3390/molecules21020140

CrossRef Full Text | Google Scholar

Silva, C. R., Frohlich, J. K., Oliveira, S. M., Cabreira, T. N., Rossato, M. F., Trevisan, G., et al. (2013). The antinociceptive and anti-inflammatory effects of the crude extract of Jatropha isabellei in a rat gout model. J. Ethnopharmacol. 145 (1), 205–213. doi:10.1016/j.jep.2012.10.054

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, A. K., Haque, M., O'Sullivan, K., Chourasia, M., Ouseph, M. M., and Ahmed, S. (2019). Suppression of monosodium urate crystal-induced inflammation by inhibiting TGF-beta-activated kinase 1-dependent signaling: role of the ubiquitin proteasome system. Cell. Mol. Immunol. doi:10.1038/s41423-019-0284-3

CrossRef Full Text | Google Scholar

So, A. K., and Martinon, F. (2017). Inflammation in gout: mechanisms and therapeutic targets. Nat. Rev. Rheumatol. 13 (11), 639–647. doi:10.1038/nrrheum.2017.155

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, S. H., Park, D. H., Bae, M. S., Choi, C. Y., Shim, J. H., Yoon, G., et al. (2018). Ethanol extract of cudrania tricuspidata leaf ameliorates hyperuricemia in mice via inhibition of hepatic and serum xanthine oxidase activity. Evid Based Complement Alternat Med. 2018, 8037925. doi:10.1155/2018/8037925

PubMed Abstract | CrossRef Full Text | Google Scholar

Spiga, R., Marini, M. A., Mancuso, E., Di Fatta, C., Fuoco, A., Perticone, F., et al. (2017). Uric acid is associated with inflammatory biomarkers and induces inflammation via activating the NF-kappaB signaling pathway in HepG2 cells. Arterioscler. Thromb. Vasc. Biol. 37 (6), 1241–1249. doi:10.1161/atvbaha.117.309128

PubMed Abstract | CrossRef Full Text | Google Scholar

Strilchuk, L., Fogacci, F., and Cicero, A. F. (2019). Safety and tolerability of available urate-lowering drugs: a critical review. Expet Opin. Drug Saf. 18 (4), 261–271. doi:10.1080/14740338.2019.1594771

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, J., Wei, Y., Liu, M., Liu, T., Li, J., Ji, Y., et al. (2014). Anti-hyperuricemic and nephroprotective effects of Rhizoma Dioscoreae septemlobae extracts and its main component dioscin via regulation of mOAT1, mURAT1 and mOCT2 in hypertensive mice. Arch Pharm. Res. (Seoul). 37 (10), 1336–1344. doi:10.1007/s12272-014-0413-6

CrossRef Full Text | Google Scholar

Su, Q., Su, H., Nong, Z., Li, D., Wang, L., Chu, S., et al. (2018). Hypouricemic and nephroprotective effects of an active fraction from polyrhachis vicina roger on potassium oxonate-induced hyperuricemia in rats. Kidney Blood Press. Res. 43 (1), 220–233. doi:10.1159/000487675

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, W. F., Zhu, M. M., Li, J., Zhang, X. X., Liu, Y. W., Wu, X. R., et al. (2015). Effects of Xie-Zhuo-Chu-Bi-Fang on miR-34a and URAT1 and their relationship in hyperuricemic mice. J. Ethnopharmacol. 161, 163–169. doi:10.1016/j.jep.2014.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Sundy, J. S., Schumacher, H. R., Kivitz, A., Weinstein, S. P., Wu, R., King-Davis, S., et al. (2014). Rilonacept for gout flare prevention in patients receiving uric acid-lowering therapy: results of RESURGE, a phase III, international safety study. J. Rheumatol. 41 (8), 1703–1711. doi:10.3899/jrheum.131226

PubMed Abstract | CrossRef Full Text | Google Scholar

Szekanecz, Z., Szamosi, S., Kovacs, G. E., Kocsis, E., and Benko, S. (2019). The NLRP3 inflammasome - interleukin 1 pathway as a therapeutic target in gout. Arch. Biochem. Biophys. 670, 82–93. doi:10.1016/j.abb.2019.01.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, R. B., Dong, J. Z., Yan, X. L., Du, X., Kang, J. P., Wu, J. H., et al. (2014). Serum uric acid and risk of left atrial thrombus in patients with nonvalvular atrial fibrillation. Can. J. Cardiol. 30 (11), 1415–1421. doi:10.1016/j.cjca.2014.06.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, H., Yang, L., Li, W., Li, J., and Chen, J. (2016). Exploring the interaction between Salvia miltiorrhiza and xanthine oxidase: insights from computational analysis and experimental studies combined with enzyme channel blocking. RSC Adv. 6 (114), 113527–113537. doi:10.1039/c6ra24396g

CrossRef Full Text | Google Scholar

Tang, L., Xu, Y., Wei, Y., and He, X. (2017). Uric acid induces the expression of TNFalpha via the ROSMAPKNFkappaB signaling pathway in rat vascular smooth muscle cells. Mol. Med. Rep. 16 (5), 6928–6933. doi:10.3892/mmr.2017.7405

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, C., Wang, Y., Chang, H., Li, J., and La, X. (2018). Spleen-kidney supplementing formula alleviates renal fibrosis in diabetic rats via TGF-β 1-miR-21-PTEN signaling pathway. Evid. base Compl. Alternative Med. 2018, 1–9. doi:10.1155/2018/3824357

CrossRef Full Text | Google Scholar

Tung, Y. T., Lin, L. C., Liu, Y. L., Ho, S. T., Lin, C. Y., Chuang, H. L., et al. (2015). Antioxidative phytochemicals from Rhododendron oldhamii Maxim. leaf extracts reduce serum uric acid levels in potassium oxonate-induced hyperuricemic mice. BMC Compl. Alternative Med. 15, 423. doi:10.1186/s12906-015-0950-7

CrossRef Full Text | Google Scholar

Wang, C. P., Wang, X., Zhang, X., Shi, Y. W., Liu, L., and Kong, L. D. (2010a). Morin improves urate excretion and kidney function through regulation of renal organic ion transporters in hyperuricemic mice. J. Pharm. Pharmaceut. Sci. 13 (3), 411–427. doi:10.18433/j3q30h

CrossRef Full Text | Google Scholar

Wang, X., Wang, C. P., Hu, Q. H., Lv, Y. Z., Zhang, X., Ouyang, Z., et al. (2010b). The dual actions of Sanmiao wan as a hypouricemic agent: down-regulation of hepatic XOD and renal mURAT1 in hyperuricemic mice. J. Ethnopharmacol. 128 (1), 107–115. doi:10.1016/j.jep.2009.12.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C. P., Wang, Y., Wang, X., Zhang, X., Ye, J. F., Hu, L. S., et al. (2011). Mulberroside a possesses potent uricosuric and nephroprotective effects in hyperuricemic mice. Planta Med. 77 (8), 786–794. doi:10.1055/s-0030-1250599

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Wang, L., Li, E., Li, Y., Wang, Z., Sun, X., et al. (2014). Chuanhu anti-gout mixture versus colchicine for acute gouty arthritis: a randomized, double-blind, double-dummy, non-inferiority trial. Int. J. Med. Sci. 11 (9), 880–885. doi:10.7150/ijms.9165

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, M. X., Liu, Y. L., Yang, Y., Zhang, D. M., and Kong, L. D. (2015). Nuciferine restores potassium oxonate-induced hyperuricemia and kidney inflammation in mice. Eur. J. Pharmacol. 747, 59–70. doi:10.1016/j.ejphar.2014.11.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Xu, B. B., Zeng, J. X., Zhu, J. X., Wang, X. Y., Ren, G., et al. (2016a). Effect of Dendropanaxchevalieri extracts on uric acid level in hyperuricemic mice and the possible mechanism. Chinese Journal of New Drugs. 25 (3), 334–338

Google Scholar

Wang, R., Ma, C. H., Zhou, F., and Kong, L. D. (2016b). Siwu decoction attenuates oxonate-induced hyperuricemia and kidney inflammation in mice. Chin. J. Nat. Med. 14 (7), 499–507. doi:10.1016/s1875-5364(16)30059-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, W. L., Sheu, S. Y., Huang, W. D., Chuang, Y. L., Tseng, H. C., Hwang, T. S., et al. (2016c). Phytochemicals from tradescantia albiflora kunth extracts reduce serum uric acid levels in oxonate-induced rats. Phcog. Mag. 12 (Suppl. 2), S223–S227. doi:10.4103/0973-1296.182171

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X. Y., Zhou, J. B., Shi, B., Guo, X. L., and Yan, Q. C. (2016d). Hypouricemic and nephroprotective effects of Jianpi Huashi decoction in a rat model of hyperuricemia. Int. J. Clin. Exp. Med. 9 (1), 455–465

Google Scholar

Wang, Y., Zhao, M., Xin, Y., Liu, J., Wang, M., and Zhao, C. (2016e). (1)H NMR and MS based metabolomics study of the therapeutic effect of Cortex Fraxini on hyperuricemic rats. J. Ethnopharmacol. 185, 272–281. doi:10.1016/j.jep.2016.03.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Lin, Z., Zhang, B., Nie, A., and Bian, M. (2017a). Cichorium intybus L. promotes intestinal uric acid excretion by modulating ABCG2 in experimental hyperuricemia. Nutr. Metab. 14, 38. doi:10.1186/s12986-017-0190-6

CrossRef Full Text | Google Scholar

Wang, Y., Lin, Z. J., Nie, A. Z., Li, L. Y., and Zhang, B. (2017b). [Effect of Chinese herb chicory extract on expression of renal transporter Glut9 in rats with hyperuricemia]. Zhongguo Zhongyao Zazhi. 42 (5), 958–963. doi:10.19540/j.cnki.cjcmm.2017.0029

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Zhao, M., Ye, H., Shao, Y., Yu, Y., Wang, M., et al. (2017c). Comparative pharmacokinetic study of the main components of cortex fraxini after oral administration in normal and hyperuricemic rats. Biomed. Chromatogr. 31 (8), e3934. doi:10.1002/bmc.3934

CrossRef Full Text | Google Scholar

Wang, Q., Yang, Q., Cao, X., Wei, Q., Firempong, C. K., Guo, M., et al. (2018). Enhanced oral bioavailability and anti-gout activity of [6]-shogaol-loaded solid lipid nanoparticles. Int. J. Pharm. 550 (1-2), 24–34. doi:10.1016/j.ijpharm.2018.08.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., Fang, Y., Yu, X., Guo, L., Zhang, X., and Xia, D. (2019a). The flavonoid-rich fraction from rhizomes of Smilax glabra Roxb. ameliorates renal oxidative stress and inflammation in uric acid nephropathy rats through promoting uric acid excretion. Biomed. Pharmacother. 111, 162–168. doi:10.1016/j.biopha.2018.12.050

CrossRef Full Text | Google Scholar

Wang, Y., Dong, L., Liu, P., Chen, Y., Jia, S., and Wang, Y. (2019b). A randomized controlled trial of chuanhutongfeng mixture for the treatment of chronic gouty arthritis by regulating miRNAs. Evid Based Complement Alternat Med. 2019, 5917269. doi:10.1155/2019/5917269

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Lin, Z., Zhang, B., Jiang, Z., Guo, F., and Yang, T. (2019c). Cichorium intybus L. Extract suppresses experimental gout by inhibiting the NF-kappaB and NLRP3 signaling pathways. Int. J. Mol. Sci. 20 (19). doi:10.3390/ijms20194921

CrossRef Full Text | Google Scholar

Wang, Y., Lin, Z., Zhang, B., Wang, X., and Chu, M. (2019d). Chicory (Cichorium intybus L.) inhibits renal reabsorption by regulating expression of urate transporters in fructose-induced hyperuricemia. Journal of Traditional Chinese Medical Sciences. 6 (1), 84–94. doi:10.1016/j.jtcms.2019.01.001

CrossRef Full Text | Google Scholar

Wang, Z., Ci, X. Y., Cui, T., Wei, Z. H., Zhang, H. B., Liu, R., et al. (2019e). Effects of Chinese herb ingredients with different properties on OAT4, URAT1 and serum uric acid level in acute hyperuricemia mice. Chin. Tradit. Herb. Drugs. 50 (5), 1157–1163. doi:10.7501/j.issn.0253-2670.2019.05.020

CrossRef Full Text | Google Scholar

Wasilewska, A., Tenderenda, E., Taranta-Janusz, K., Tobolczyk, J., and Stypulkowska, J. (2012). Markers of systemic inflammation in children with hyperuricemia. Acta Paediatr. 101 (5), 497–500. doi:10.1111/j.1651-2227.2011.02582.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, G. N., Su, Q. B., He, F., Zeng, X. B., Ya, Q. K., Lü, J. H., et al. (2013). Screening and chemical component analysis of anti-hyperuricemic active fraction of ethanol extract from Polyrhachis vicina Roger in hyperuricemia model mice. Chin. J. Pharmacol. Toxicol. 27 (4), 673–677. doi:10.3867/j.issn.1000-3002.2013.04.012

CrossRef Full Text | Google Scholar

Wei, Z., Xu, C., Liu, S., Song, F., Liu, Z., and Qu, X. (2018). Metabonomics study of the effects of traditional Chinese medicine formula Ermiaowan on hyperuricemic rats. J. Separ. Sci. 41 (2), 560–570. doi:10.1002/jssc.201700985

CrossRef Full Text | Google Scholar

White, W. B., Saag, K. G., Becker, M. A., Borer, J. S., Gorelick, P. B., Whelton, A., et al. (2018). Cardiovascular safety of febuxostat or allopurinol in patients with gout. N. Engl. J. Med. 378 (13), 1200–1210. doi:10.1056/NEJMoa1710895

PubMed Abstract | CrossRef Full Text | Google Scholar

Woodward, O. M., Kottgen, A., Coresh, J., Boerwinkle, E., Guggino, W. B., and Kottgen, M. (2009). Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc. Natl. Acad. Sci. U. S. A. 106 (25), 10338–10342. doi:10.1073/pnas.0901249106

PubMed Abstract | CrossRef Full Text | Google Scholar

Wright, A. F., Rudan, I., Hastie, N. D., and Campbell, H. (2010). A 'complexity' of urate transporters. Kidney Int. 78 (5), 446–452. doi:10.1038/ki.2010.206

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., Liu, L., Xie, H., Liao, J., Zhou, X., Wan, J., et al. (2012a). Tanshinone IIA prevents uric acid nephropathy in rats through NF-kappaB inhibition. Planta Med. 78 (9), 866–873. doi:10.1055/s-0031-1298487

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., Liu, L., Xie, H., Liao, J., Zhou, X., Wan, J., et al. (2012b). Tanshinone IIA prevents uric acid nephropathy in rats through NF-κB inhibition. Planta Med. 78 (9), 866–873. doi:10.1055/s-0031-1298487

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. H., Ruan, J. L., Zhang, J., Wang, S. Q., and Zhang, Y. W. (2014a). Pallidifloside D, a saponin glycoside constituent from Smilax riparia, resist to hyperuricemia based on URAT1 and GLUT9 in hyperuricemic mice. J. Ethnopharmacol. 157, 201–205. doi:10.1016/j.jep.2014.09.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. H., Wang, C. Z., Zhang, J., Wang, S. Q., Han, L., Zhang, Y. W., et al. (2014b). Effects of Smilaxchinoside A and Smilaxchinoside C, two steroidal glycosides from Smilax riparia, on hyperuricemia in a mouse model. Phytother Res. 28 (12), 1822–1828. doi:10.1002/ptr.5207

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. H., Yu, C. H., Zhang, C. F., Anderson, S., and Zhang, Y. W. (2014c). Smilax riparia reduces hyperuricemia in mice as a potential treatment of gout. Am. J. Chin. Med. 42 (1), 257–259. doi:10.1142/s0192415x14200018

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. H., Zhang, J., Wang, S. Q., Yang, V. C., Anderson, S., and Zhang, Y. W. (2014d). Riparoside B and timosaponin J, two steroidal glycosides from Smilax riparia, resist to hyperuricemia based on URAT1 in hyperuricemic mice. Phytomedicine. 21 (10), 1196–1201. doi:10.1016/j.phymed.2014.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X. H., Wang, C. Z., Wang, S. Q., Mi, C., He, Y., Zhang, J., et al. (2015). Anti-hyperuricemia effects of allopurinol are improved by Smilax riparia, a traditional Chinese herbal medicine. J. Ethnopharmacol. 162, 362–368. doi:10.1016/j.jep.2015.01.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., He, F., Li, Y., Wang, H., Shi, L., Wan, Q., et al. (2017). Effects of shizhifang on NLRP3 inflammasome activation and renal tubular injury in hyperuricemic rats. Evid Based Complement Alternat Med. 2017, 7674240. doi:10.1155/2017/7674240

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, P., Li, J., Zhang, X., Zeng, F., Liu, Y., and Sun, W. (2018a). Study of the treatment effects of compound tufuling granules in hyperuricemic rats using serum metabolomics. Evid Based Complement Alternat Med. 2018, 3458185. doi:10.1155/2018/3458185

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Wang, Y., Ou, J., Wan, Q., Shi, L., Li, Y., et al. (2018b). Effect and mechanism of ShiZhiFang on uric acid metabolism in hyperuricemic rats. Evid Based Complement Alternat Med. 2018, 6821387. doi:10.1155/2018/6821387

PubMed Abstract | CrossRef Full Text | Google Scholar

Xia, N., Li, B. A., Liu, H. J., Fan, J. B., Shen, W., and He, X. G. (2017). Anti-hyperuricemic effect of Plantago depressa Willd extract in rats. Trop. J. Pharmaceut. Res. 16 (6), 1365–1368. doi:10.4314/tjpr.v16i6.21

CrossRef Full Text | Google Scholar

Xiang, S. W., Lai, S. C., and Meng, Y. H. (2009). [Clinical study on modified sanmiao powder in treating chronic uric acid nephropathy]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 29 (11), 979–981

PubMed Abstract | Google Scholar

Xie, Z., Wu, H., Jing, X., Li, X., Li, Y., Han, Y., et al. (2017). Hypouricemic and arthritis relapse-reducing effects of compound tufuling oral-liquid in intercritical and chronic gout: a double-blind, placebo-controlled, multicenter randomized trial. Medicine (Baltim.). 96 (11), e6315. doi:10.1097/MD.0000000000006315

CrossRef Full Text | Google Scholar

Xiong, W. W., Zhang, H. Y., Wen, L., Wang, X. Y., Zhong, G. Y., Shi, Y. F., et al. (2018). Effect of Lagotis brachystachys Maxim extract on xanthine oxidase and renal urate transporters in hyperuricemia mice. Chinese Journal of New Drugs. 27 (13), 1538–1543

Google Scholar

Xu, W. A., Yin, L., Pan, H. Y., Shi, L., Xu, L., Zhang, X., et al. (2013). Study on the correlation between constituents detected in serum from Rhizoma Smilacis Glabrae and the reduction of uric acid levels in hyperuricemia. J. Ethnopharmacol. 150 (2), 747–754. doi:10.1016/j.jep.2013.09.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, C., Wan, X., Xu, L., Weng, H., Yan, M., Miao, M., et al. (2015). Xanthine oxidase in non-alcoholic fatty liver disease and hyperuricemia: one stone hits two birds. J. Hepatol. 62 (6), 1412–1419. doi:10.1016/j.jhep.2015.01.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, K., Liu, S., Zhao, X., Zhang, X., Fu, X., Zhou, Y., et al. (2019). Treating hyperuricemia related non-alcoholic fatty liver disease in rats with resveratrol. Biomed. Pharmacother. 110, 844–849. doi:10.1016/j.biopha.2018.12.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, M., An, Y. T., Li, J., Wu, Z. Z., and Wang, T. (2014). [Regulatory effect of leonurus extracts on hyperuricemia in rats]. Zhongguo Zhongyao Zazhi. 39 (24), 4856–4859

PubMed Abstract | Google Scholar

Yang, H. X., Su, J. X., Jiang, G. Q., and Chen, X. P. (2013). Effect of Tongxi Powder on blood uric acid of mice with acute hyperuricemia. Chin. Tradit. Herb. Drugs. 44 (12), 1635–1637. doi:10.7501/j.issn.0253-2670.2013.12.021

CrossRef Full Text | Google Scholar

Yang, Y., Zhang, D. M., Liu, J. H., Hu, L. S., Xue, Q. C., Ding, X. Q., et al. (2015). Wuling San protects kidney dysfunction by inhibiting renal TLR4/MyD88 signaling and NLRP3 inflammasome activation in high fructose-induced hyperuricemic mice. J. Ethnopharmacol. 169, 49–59. doi:10.1016/j.jep.2015.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, A., Guo, H., Fu, M., and Liu, M. (2019a). Inhibitive effects of huashi pill on formation of renal stones by modulating urine biochemical indexes and osteopontin in renal stone rat models. Med. Sci. Mon. Int. Med. J. Exp. Clin. Res. 25, 8335–8344. doi:10.12659/msm.916247

CrossRef Full Text | Google Scholar

Yang, L. X., Xue, J. J., Meng, X. Y., Wang, Y. S., Wu, L. L., Lv, C. Y., et al. (2019b). Effects of total flavonoids from Oxytropis falcata Bunge on the SOCS/JAK/STAT inflammatory signaling pathway in the kidneys of diabetic nephropathy model mice. Eur. J. Inflamm. 17. doi:10.1177/2058739219861877

CrossRef Full Text | Google Scholar

Yang, T. H., Yan, D. X., Huang, X. Y., Hou, B., Ma, Y. B., Peng, H., et al. (2019c). Termipaniculatones A-F, chalcone-flavonone heterodimers from Terminthia paniculata, and their protective effects on hyperuricemia and acute gouty arthritis. Phytochemistry. 164, 228–235. doi:10.1016/j.phytochem.2019.05.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Y., Zhang, L., Jiang, G., Lei, A., Yu, Q., Xie, J., et al. (2019d). Evaluation of the protective effects of Ganoderma atrum polysaccharide on acrylamide-induced injury in small intestine tissue of rats. Food Funct. 10 (9), 5863–5872. doi:10.1039/c9fo01452g

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao, L., Dong, W., Lu, F., and Liu, S. (2012). An improved acute gouty arthritis rat model and therapeutic effect of rhizoma Dioscoreae nipponicae on acute gouty arthritis based on the protein-chip methods. Am. J. Chin. Med. 40 (1), 121–134. doi:10.1142/s0192415x12500103

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao, R., Geng, Z., Mao, X., Bao, Y., Guo, S., Bao, L., et al. (2020). Tu-teng-cao extract alleviates monosodium urate-induced acute gouty arthritis in rats by inhibiting uric acid and inflammation. Evid Based Complement Alternat Med. 2020, 3095624. doi:10.1155/2020/3095624

PubMed Abstract | CrossRef Full Text | Google Scholar

Ye, Y., Zhang, Y., Wang, B., Walana, W., Wei, J., Gordon, J. R., et al. (2018). CXCR1/CXCR2 antagonist G31P inhibits nephritis in a mouse model of uric acid nephropathy. Biomed. Pharmacother. 107, 1142–1150. doi:10.1016/j.biopha.2018.07.077

PubMed Abstract | CrossRef Full Text | Google Scholar

Yi, L. T., Li, J., Su, D. X., Dong, J. F., and Li, C. F. (2012). Hypouricemic effect of the methanol extract from Prunus mume fruit in mice. Pharm. Biol. 50 (11), 1423–1427. doi:10.3109/13880209.2012.683115

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, W., Zhou, Q. L., OuYang, S. X., Chen, Y., Gong, Y. T., and Liang, Y. M. (2019). Uric acid regulates NLRP3/IL-1beta signaling pathway and further induces vascular endothelial cells injury in early CKD through ROS activation and K(+) efflux. BMC Nephrol. 20 (1), 319. doi:10.1186/s12882-019-1506-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Yong, T., Zhang, M., Chen, D., Shuai, O., Chen, S., Su, J., et al. (2016). Actions of water extract from Cordyceps militaris in hyperuricemic mice induced by potassium oxonate combined with hypoxanthine. J. Ethnopharmacol. 194, 403–411. doi:10.1016/j.jep.2016.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Yong, T., Chen, S., Xie, Y., Chen, D., Su, J., Shuai, O., et al. (2017). Hypouricemic effects of ganoderma applanatum in hyperuricemia mice through OAT1 and GLUT9. Front. Pharmacol. 8, 996. doi:10.3389/fphar.2017.00996

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoon, I. S., Park, D. H., Ki, S. H., and Cho, S. S. (2016). Effects of extracts from Corylopsis coreana Uyeki (Hamamelidaceae) flos on xanthine oxidase activity and hyperuricemia. J. Pharm. Pharmacol. 68 (12), 1597–1603. doi:10.1111/jphp.12626

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoon, I. S., Park, D. H., Bae, M. S., Oh, D. S., Kwon, N. H., Kim, J. E., et al. (2017a). In Vitro and in vivo studies on quercus acuta thunb. (Fagaceae) extract: active constituents, serum uric acid suppression, and xanthine oxidase inhibitory activity, Evid. Based Complement. Alternat. Med. 2017, 4097195. doi:10.1155/2017/4097195

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoon, I. S., Park, D. H., Kim, J. E., Yoo, J. C., Bae, M. S., Oh, D. S., et al. (2017b). Identification of the biologically active constituents of Camellia japonica leaf and anti-hyperuricemic effect in vitro and in vivo. Int. J. Mol. Med. 39 (6), 1613–1620. doi:10.3892/ijmm.2017.2973

PubMed Abstract | CrossRef Full Text | Google Scholar

You, W., Wang, J., Zou, Y., Che, K., Hou, X., Fei, H., et al. (2019). Modified Chuanhu anti-gout mixture, a traditional Chinese medicine, protects against potassium oxonate-induced hyperuricemia and renal dysfunction in mice. J. Int. Med. Res. 47 (5), 1927–1935. doi:10.1177/0300060519831182

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, X. N., Wu, H. Y., Deng, Y. P., Zhuang, G. T., Tan, B. H., Huang, Y. Z., et al. (2018). Yellow-dragon Wonderful-seed Formula" for hyperuricemia in gout patients with dampness-heat pouring downward pattern: a pilot randomized controlled trial. Trials. 19 (1), 551. doi:10.1186/s13063-018-2917-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, X., Fan, Y. S., Xu, L., Xie, G. Q., Feng, X. H., and Qian, K. (2019). Jia-Wei-Si-Miao-Wan alleviates acute gouty arthritis by targeting NLRP3 inflammasome. J. Biol. Regul. Homeost. Agents. 33 (1), 63–71

Google Scholar

Yuk, H. J., Lee, Y. S., Ryu, H. W., Kim, S. H., and Kim, D. S. (2018). Effects of toona sinensis leaf extract and its chemical constituents on xanthine oxidase activity and serum uric acid levels in potassium oxonate-induced hyperuricemic rats. Molecules. 23 (12). doi:10.3390/molecules23123254

CrossRef Full Text | Google Scholar

Zamudio-Cuevas, Y., Hernandez-Diaz, C., Pineda, C., Reginato, A. M., Cerna-Cortes, J. F., Ventura-Rios, L., et al. (2015). Molecular basis of oxidative stress in gouty arthropathy. Clin. Rheumatol. 34 (10), 1667–1672. doi:10.1007/s10067-015-2933-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, J. X., Xu, B. B., Li, M., Li, X. W., Zhu, J. X., Wang, X. Y., et al. (2015). Effect of Lagotis brevituba Maxim. extract in reducing uric acid level in hyperuricemia mice and it's mechanism. Chinese Journal of New Drugs. 24 (21), 2489–2493

Google Scholar

Zhang, M., Wang, J. F., and Wang, Y. F. (2009). [Effects of serial gout granules on insulin-resistance in primary gout patients]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 29 (12), 1068–1072

PubMed Abstract | Google Scholar

Zhang, X. X., Sun, W. F., and Xu, W. (2011). [Assessment on the clinical efficacy and safety of xiezhuo chubi recipe in treating hyperuricemia]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 31 (9), 1216–1219

PubMed Abstract | Google Scholar

Zhang, D. M., Li, Y. C., Xu, D., Ding, X. Q., and Kong, L. D. (2012). Protection of curcumin against fructose-induced hyperuricaemia and renal endothelial dysfunction involves NO-mediated JAK-STAT signalling in rats. Food Chem. 134 (4), 2184–2193. doi:10.1016/j.foodchem.2012.04.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Z. C., Su, G. H., Luo, C. L., Pang, Y. L., Wang, L., Li, X., et al. (2015). Effects of anthocyanins from purple sweet potato (Ipomoea batatas L. cultivar Eshu No. 8) on the serum uric acid level and xanthine oxidase activity in hyperuricemic mice. Food Funct. 6 (9), 3045–3055. doi:10.1039/c5fo00499c

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H. J., Li, L. N., Zhou, J., Yang, Q. Q., Liu, P. G., Xu, P., et al. (2017a). Effects of Gnaphalium affine D. Don on hyperuricemia and acute gouty arthritis. J. Ethnopharmacol. 203, 304–311. doi:10.1016/j.jep.2017.03.057

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S., Zhuang, J., Yue, G., Wang, Y., Liu, M., Zhang, B., et al. (2017b). Lipidomics to investigate the pharmacologic mechanisms of ginkgo folium in the hyperuricemic rat model. J Chromatogr B Analyt Technol Biomed Life Sci. 1060, 407–415. doi:10.1016/j.jchromb.2017.06.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, R., Zhan, S., Li, S., Zhu, Z., He, J., Lorenzo, J. M., et al. (2018a). Anti-hyperuricemic and nephroprotective effects of extracts from Chaenomeles sinensis (Thouin) Koehne in hyperuricemic mice. Food Funct. 9 (11), 5778–5790. doi:10.1039/c8fo01480a

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Jin, L., Liu, J., Wang, W., Yu, H., Li, J., et al. (2018b). Effect and mechanism of dioscin from Dioscorea spongiosa on uric acid excretion in animal model of hyperuricemia. J. Ethnopharmacol. 214, 29–36. doi:10.1016/j.jep.2017.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, W., Du, W., Li, G., Zhang, C., Yang, W., Yang, S., et al. (2019a). Constituents and anti-hyperuricemia mechanism of traditional Chinese herbal formulae erding granule. Molecules. 24 (18). doi:10.3390/molecules24183248

CrossRef Full Text | Google Scholar

Zhang, X. Y., Cheng, J., Zhao, P., Chen, K. L., and Li, J. (2019b). Screening the best compatibility of selaginella moellendorffii prescription on hyperuricemia and gouty arthritis and its mechanism. Evid Based Complement Alternat Med. 2019, 7263034. doi:10.1155/2019/7263034

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Z. C., Zhou, Q., Yang, Y., Wang, Y., and Zhang, J. L. (2019c). Highly acylated anthocyanins from purple sweet potato ( Ipomoea batatas L.) alleviate hyperuricemia and kidney inflammation in hyperuricemic mice: possible attenuation effects on allopurinol. J. Agric. Food Chem. 67 (22), 6202–6211. doi:10.1021/acs.jafc.9b01810

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, P., Chen, K. L., Zhang, G. L., Deng, G. R., and Li, J. (2017). Pharmacological basis for use of selaginella moellendorffii in gouty arthritis: antihyperuricemic, anti-inflammatory, and xanthine oxidase inhibition. Evid Based Complement Alternat Med. 2017, 2103254. doi:10.1155/2017/2103254

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, J., Lv, C., Wu, Q., Zeng, H., Guo, X., Yang, J., et al. (2019). Computational systems pharmacology reveals an antiplatelet and neuroprotective mechanism of Deng-Zhan-Xi-Xin injection in the treatment of ischemic stroke. Pharmacol. Res. 147, 104365. doi:10.1016/j.phrs.2019.104365

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, M., Wang, Y. F., Zhou, R., Zhang, M., and Li, B. (2013). [Treatment of gouty arthritis in different phases by a series of tongfeng granule: an efficacy observation]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 33 (12), 1603–1607

PubMed Abstract | Google Scholar

Zhou, Q., Yu, D. H., Zhang, C., Liu, S. M., and Lu, F. (2014). Total saponins from Discorea nipponica ameliorate urate excretion in hyperuricemic mice. Planta Med. 80 (15), 1259–1268. doi:10.1055/s-0034-1383048

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q., Yu, D. H., Liu, S. M., and Liu, Y. (2015). Total saponins from Discorea nipponica makino ameliorate urate excretion in hyperuricemic rats. Phcog. Mag. 11 (43), 567–573. doi:10.4103/0973-1296.160442

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, W., Wang, J., Wu, Z., Huang, C., Lu, A., and Wang, Y. (2016). Systems pharmacology exploration of botanic drug pairs reveals the mechanism for treating different diseases. Sci. Rep. 6, 36985. doi:10.1038/srep36985

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q., Lin, F. F., Liu, S. M., and Sui, X. F. (2017). Influence of the total saponin fraction from Dioscorea nipponica Makino on TLR2/4-IL1R receptor singnal pathway in rats of gouty arthritis. J. Ethnopharmacol. 206, 274–282. doi:10.1016/j.jep.2017.04.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Y., Zhang, X., Li, C., Yuan, X., Han, L., Li, Z., et al. (2018a). Research on the pharmacodynamics and mechanism of Fraxini Cortex on hyperuricemia based on the regulation of URAT1 and GLUT9. Biomed. Pharmacother. 106, 434–442. doi:10.1016/j.biopha.2018.06.163

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Y., Zhao, M., Pu, Z., Xu, G., and Li, X. (2018b). Relationship between oxidative stress and inflammation in hyperuricemia: analysis based on asymptomatic young patients with primary hyperuricemia. Medicine (Baltim.). 97 (49), e13108. doi:10.1097/md.0000000000013108

CrossRef Full Text | Google Scholar

Zhu, W., Pang, M., Dong, L., Huang, X., Wang, S., and Zhou, L. (2012). Anti-inflammatory and immunomodulatory effects of iridoid glycosides from Paederia scandens (LOUR.) MERRILL (Rubiaceae) on uric acid nephropathy rats. Life Sci. 91 (11-12), 369–376. doi:10.1016/j.lfs.2012.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, F., Yin, L., Ji, L., Yang, F., Zhang, G., Shi, L., et al. (2016). Suppressive effect of Sanmiao formula on experimental gouty arthritis by inhibiting cartilage matrix degradation: an in vivo and in vitro study. Int. Immunopharm. 30, 36–42. doi:10.1016/j.intimp.2015.11.010

CrossRef Full Text | Google Scholar

Zhu, L., Dong, Y., Na, S., Han, R., Wei, C., and Chen, G. (2017). Saponins extracted from Dioscorea collettii rhizomes regulate the expression of urate transporters in chronic hyperuricemia rats. Biomed. Pharmacother. 93, 88–94. doi:10.1016/j.biopha.2017.06.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, C., Xu, Y., Liu, Z. H., Wan, X. C., Li, D. X., and Tai, L. L. (2018). The anti-hyperuricemic effect of epigallocatechin-3-gallate (EGCG) on hyperuricemic mice. Biomed. Pharmacother. 97, 168–173. doi:10.1016/j.biopha.2017.10.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Zuo, J., He, H., Zuo, Z., Bou-Chacra, N., and Lobenberg, R. (2018). Erding Formula in hyperuricaemia treatment: unfolding traditional Chinese herbal compatibility using modern pharmaceutical approaches. J. Pharm. Pharmacol. 70 (1), 124–132. doi:10.1111/jphp.12840

PubMed Abstract | CrossRef Full Text | Google Scholar

Zuo, J., Zhang, W., Jian, H., Bou-Chacra, N., and Löbenberg, R. (2020). Esculetin as bioactive marker: towards a rational scientific approach for the treatment of hyperuricemia using traditional Chinese medicine. Brazilian Journal of Pharmaceutical Sciences. 56. doi:10.1590/s2175-97902019000417827

CrossRef Full Text | Google Scholar

Keywords: Chinese herbal medicine, serum uric acid, serum urate, efficacy, mechanism

Citation: Chen L, Luo Z, Wang M, Cheng J, Li F, Lu H, He Q, You Y, Zhou X, Kwan HY, Zhao X and Zhou L (2021) The Efficacy and Mechanism of Chinese Herbal Medicines in Lowering Serum Uric Acid Levels: A Systematic Review. Front. Pharmacol. 11:578318. doi: 10.3389/fphar.2020.578318

Received: 30 June 2020; Accepted: 21 December 2020;
Published: 25 January 2021.

Edited by:

Hai Yu Xu, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, China

Reviewed by:

Souad Skalli, Mohammed V University, Morocco
Bagher Larijani, Tehran University of Medical Sciences, Iran

Copyright © 2021 Chen, Luo, Wang, Cheng, Li, Lu, He, You, Zhou, Kwan, Zhao and Zhou. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xiaoshan Zhao, zhaoxs0609@163.com; Lin Zhou, zlecho@163.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.