Effects and Mechanisms of Traditional Chinese Medicine and Related Active Constituents Treating Gout
Hao Wen1, Le Yang2, Ruicheng Liu1, Guangli Yan1, Hui Sun1*, Ling Kong1, Ye Sun2, Ying Han1, Qiqi Zhao1, Shuyu Kang1, Xijun Wang1,2*
1 National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
2 State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
*Corresponding authors: Xijun Wang and Hui Sun, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
Received Date: 10 February 2023
Accepted Date: 17 February 2023
Published Date: 22 February 2023
Citation: Wen H, Yang L, Liu R, Yan G, Sun H, et al. (2023) Effects and Mechanisms of Traditional Chinese Medicine and Related Active Constituents Treating Gout. Curr Res Cmpl Alt Med 7: 170. DOI: https://doi.org/10.29011/2577-2201.100070
Abstract
Gout is an inherited metabolic disorder resulting from abnormalities in purine metabolism manifesting as episodes of acute inflammatory arthritis. The incidence of gout shows an increasing tendency each year, due to the complicated pathogenesis, the currently available drugs have poor treatment effects on it. Therefore, the development of effective drugs for gout treatment is urgently needed. Because of its high biological activity and safety, Traditional Chinese medicine (TCM) has significant advantages in the prevention and treatment of gout. This paper systematically reviewed the related research on TCM’s treatment of gout in the previous five years, and summarized the actions and mechanisms of 25 Chinese herbal extracts, 38 Chinese medicinal monomers, and 19 Chinese medicinal compounds in the treatment of gout, including serum uric acid levels reduction, anti-inflammation, and anti-oxidative stress. These primarily involve the signaling pathways NLRP3/ASC/caspase, TLRs/MYD88/NF-κB, ATP/ P2X7R, and others. Furthermore, the limitations and shortcomings of the experimental study on the treatment of gout with TCM are discussed and appropriate measures are proposed. To summarize, TCM is likely to be a potential candidate for gout treatment, but more clinical research is needed to fully understand its role in the treatment of gout.
Keywords: Active ingredients; Anti-inflammation; Gout; Pathogenesis; Pharmacological effects; Traditional Chinese medicine
Abbreviations: TCM- Traditional Chinese medicine; GAG- Glycosaminogly; URAT1- Uric acid transporter 1; GLUT9- Glucose transporter 9; OAT- Organic anion transporter; ATP- Adenosine triphosphate; ABCG2- (ATP)–binding cassette transporter subfamily G member 2; MSU- Monosodium urate; NLRP3- NOD-like receptor pyrin containing 3; TLR- Toll-like receptor; IL-1R- Interleukin-1 receptor; I-RAK- IL-1R–related kinase; TNF- Tumor necrosis factor; LPS- Lipopolysaccharide; NLRs- NOD–like receptors; NF-κB- Nuclear factor kappa B; PGE2Prostaglandin E2; COX- Cyclooxygenase; ROS- Reactive oxygen species; MPO- Myeloperoxidase; IL-1β- Interleukin-1 beta; IL-18- Interleukin-18; IL-6- Interleukin-6; TGF-β- Transforming growth factor beta; MAPK- Mitogen-activated protein kinase; SDF-1: Stromal cell-derived factor 1; MCP-1: Monocyte chemoattractant protein-1; IL-10- Interleukin-10; OCT- Organic cation; PTGS2- Prostaglandin-endoperoxide synthasetransporter; UA- Uric acid.
Introduction
Gout is a common inherited metabolic disorder resulting from abnormalities in purine metabolism or reduced uric acid excretion and is characterized by hyperuricemia-associated inflammatory arthritis, belonging to the category of metabolic rheumatism [13]. Acute gouty arthritis, the main form of gout [4], occurs due to the deposition of sodium urate crystals in the joints and other tissues because of the high level of uric acid persisting in the body for a long time [5]. The common clinical manifestations of gout are redness and swelling of the affected joints accompanied by severe pain [6]. The first gouty attack episode involves a single joint, with the unilateral first metatarsophalangeal joint being the most common one [7]. Notably, when gout repeatedly recurs for a long period, tophi formation occurs in the joints and soft tissues, and with disease progression, joint deformity, urinary calculi, and renal impairment can occur, which confers a great effect on the health and quality of life of the affected patients [8,9]. Current epidemiology worldwide indicates an increasing trend in morbidity and prevalence in both developed and developing countries [10,11]. Therefore, identifying an effective treatment for gout has attracted increasing attention and has become a top priority.
Because of the complexity of the pathogenesis of gout, no drugs available so far can completely cure gout. In western medicine, nonsteroidal anti-inflammatory drugs, glucocorticoids, and colchicine are mainly used for treating acute gouty attacks owing to their excellent analgesic and anti-inflammatory effects in rapidly relieving acute gouty attacks [12]. Febuxostat and allopurinol are mainly used for the treatment of gout in the chronic phase to reduce uric acid levels [13]. These first-line anti-gout drugs exert not only a significant therapeutic effect but also significant side effects, and these drugs cannot fundamentally eliminate the disease [14]. Hence, there is an urgent need to develop new anti-gout drugs. TCM has a long history of therapeutic effect on gout and has demonstrated good clinical effects. Thus far, a large number of experimental studies have focused on the anti-gout effect of TCM. It is believed that TCM exerts the anti-gout effect mainly by reducing uric acid levels, inflammation, and oxidative stress. Currently, there is a lack of a comprehensive review on gout treatment with TCM. We, therefore, present a review on the pathophysiological mechanism of gout and mechanisms of effective of TCM and related effective constituents for treating gout to provide novel ideas and directions for the prevention and treatment of gout in the future.
Pathogenesis of Gout
Hyperuricemia, which is the most important factor for gout development, is characterized by elevated level of uric acid in the body, with a subsequent increase in the risk of gout [15]. Hyperuricemia is mainly caused by two factors: increased uric acid production and reduced uric acid excretion. Uric acid is the end product of purine nucleotide degradation in the body, and the dietary source is attributed to the absorption of purine from purine-rich foods by the small intestine. Xanthine oxidase (XOD) catalyzes the oxidation reactions of hypoxanthine to xanthine and finally to uric acid, which directly affects uric acid formation [16]. Additionally, uric acid excretion is regulated by the kidneys and intestines, particularly through glomerular filtration, as well as active and passive reabsorption at the beginning and straight segments of the proximal tubule. Many transporters are involved in the filtration and reabsorption processes, namely, URAT1; GLUT9; OAT1, OAT2, and OAT3; and secretion transporter ABCG2 [1722]. The expression and dysfunction of these transporters affect uric acid excretion.
Uric acid mostly exists as soluble urate in the body [23]. When the uric acid concentration in the body exceeds the solubility threshold of 7.0 mg/dL [24], MSU crystals are deposited in the joints, soft tissues, cartilage, and kidneys. The MSU crystal is an injury-related pathogen that activates not only the innate immune response but also the macrophages and monocytes, leading to the activation of NLRP3 inflammasomes. NLRP3 inflammasomes are closely associated with gout episodes [25]. The activation of NLRP3 inflammasomes depends on two steps, namely, initiation and activation [26]. The initiation step affects the assembly and activation of the NLRP3 inflammasomes and is related to the TLR signaling pathway. TLR2 and TLR4 play a vital role in the production of inflammatory factors [27]. The complex formed by the binding of MSU crystals to CD14 receptors located on the cell surface is recognized by the TLRs; IL-1R–related kinase (I-RAK) is activated by the recruitment of myeloid differentiation factor (MYD88) through IL-1R and binds to tumor necrosis factor (TNF) receptor–related factor 6 (TRAF6), which then activates IκB kinase to allow free NF-κB to enter the nucleus. Subsequently, the transcription of chemokines and proinflammatory mediators, including pro-interleukin (pro-IL)-1β, pro-IL-18, and TNF-α [2830]. It is worth noting that the MSU crystals activate TLRs only by binding with the TLR ligand (fatty acid/ LPS) [31]. Additionally, the activation step of the NLRP3 inflammasomes is influenced by the MSU crystals. The NLRP3 inflammasome comprises three parts: nucleotide oligomerization domain (NOD)–like receptors (NLRs), apoptosis-associated dot-like protein (apoptosisassociated speck-like protein containing a caspase recruitment domain [ASC]), and pro-cysteine–dependent aspartate-directed protease-1 ( pro-caspase-1). NLRs are cytoplasmic pattern recognition receptors that sense a danger signal from the MSU crystals, connect with the ASC and pro-caspase-1, and assemble into an inflammasome, thereby leading to Caspase-1 cleavage and activation, which cleaves pro-IL-1β and pro-IL-18 into mature IL-1β and IL-18, respectively [32-34]. The MSU crystal can also induce the monocytes to secrete COX-2, which first transforms arachidonic acid into PGE2 and then stimulates the inflammatory reaction [35,36].
Excessive extracellular ATP concentration can also promote the activation of NLRP3 inflammasomes and thus produce more inflammatory factors. Particularly, the abnormally high ATP level activates the P2X7R receptor on the cell surface and induces the formation of cation channels, which promote the influx of extracellular Na ions and Ca ions and the efflux of K ions simultaneously [37]. K ion outflow promotes the activation of the NLRP3 inflammasome [38]. Additionally, the engulfment of the MSU crystals by the macrophages punctures the mitochondria, leading to the production of ROS [39], further activation of the NLRP3 inflammasome, and increased production of the inflammatory factor IL-1β [40].
To summarize, MSU crystals can upregulate the expression of many inflammatory factors, including TNF-α, IL-1β, and IL-18, among others, and IL-1β is the main inflammatory factor causing gout. IL-1β stimulates an inflammatory response that generates the recruitment of monocytes and neutrophils at the site of MSU crystal deposition, and the resulting inflammatory response causes gout [41]. Notably, continuous accumulation of IL-1β activates matrix-degrading enzymes, causing cartilage damage and bone destruction [42] (Figure 1).
Figure 1: Schematic diagram of the pathology of gout. Gout is characterized by high serum uric acid levels, MSU crystal deposition, inflammation, and oxidative stress. Increased uric acid production and decreased uric acid excretion can result in increased serum uric acid levels, and persistently high serum uric acid levels can result in the deposition of MSU crystals, which trigger inflammatory reactions that lead to gout. Oxidative stress promotes the formation and activation of inflammasomes, as well as the production of more inflammatory factors, aggravating the inflammatory response.
Therapeutic Effects of Herbal Medicines
Herbal Medicines Originated from Asteraceae
Atractylodis rhizome, a dry rhizome from Atractylodes lancea (Thunb.) DC., is a common Chinese herbal medicine with antiinflammatory, anticancer, antibacterial, and other pharmacological effects [43]. Li et al. identified the effective components and the mechanism of atractylodis rhizome extract in the treatment of gouty arthritis through network pharmacology and verified their results through experiments. Atractylodes macrocephala could lower the levels of TGF-β1, PGE2, UA, IL-1β, IL-6, and TNF-α and XOD activity and can regulate apoptosis-related pathways, which may be the mechanism of atractylodes macrocephala for gout treatment [44]. The atractylodes macrocephala extract can inhibit XOD activity, reduce uric acid level, and decrease inflammation in gouty arthritis rats by hindering the formation of inflammatory factors such as IL-1β and TNF-α [45]. The chicory extract regulates the NF-κB and NLRP3 signaling pathways and exerts a therapeutic effect on gouty arthritis. In vivo studies have shown that the chicory extract significantly reduced the degree of joint swelling in mice and blocked the NF-κB/NLRP3 signaling pathways, thereby inhibiting the release of IL-1β. Similar results were obtained under in vitro conditions. Chicory and chicoric acid inhibited IL-1β expression [46]. Siegesbeckia orientalis L. reduces serum uric acid concentration by inhibiting XOD activity. It can also reduce carrageenan-induced foot swelling and synovitis in vivo [47]. The sunflower (Helianthus annuus) head extract (SHE) can significantly inhibit MSU crystal–induced ankle joint swelling in rats by reducing the serum IL-10 level and inhibiting MCP-1 protein expression. The SHE extract can also decrease the uric acid level in hyperuricemic mice by inhibiting XOD activity in vivo [48] (Table 1).
Herbal Medicines Originated from Dioscoreaceae
Dioscorea collettii extract (DCE) exerts a therapeutic effect on gouty arthritis. DCE reduces the expression of NALP3 and ASC proteins in vivo, decreases the expression of the NALP3 protein, and inhibits the activation of Caspase-1 protein in THP1 macrophages in vitro [49]. Lu et al. reported that the Rhizoma Dioscoreae Nipponicae(Dioscoreaceae) extract may exert a pharmacological effect in gouty arthritis by regulating the p38 MAPK and SDF-1/CXCR-4 signaling pathways. In addition, its action mechanism is also closely related to the metabolism of glycerophospholipids, inositol phosphate and galactose [50,51] (Table 1).
Herbal Medicines Originated from Liliaceae
The extract of Smilax glabra Roxb. can stimulate uric acid excretion and confer protection against uric acid nephropathy. The mechanism of Smilax glabra Roxb. in lowering uric acid levels is related to the upregulated expression of ABCG2, OAT1, organic cation transporter 2 (OCT2), and organic cation/carnitine transporter 2 (OCTN2). Smilax glabra Roxb. may thus exert a therapeutic effect on gout by reducing uric acid levels. Smilax glabra Roxb. can also improve renal damage caused by urate [52]. Interestingly, Smilax glabra Roxb. can also reduce serum uric acid levels by inhibiting XOD activity. In vivo experiments have confirmed that Smilax glabra Roxb. can treat gouty arthritis by reducing the expression of the inflammasome [53,54]. Wu et al. demonstrated that Smilax riparia can stimulate uric acid excretion by inhibiting mURAT1 expression, suggesting that it may exert a therapeutic impact on gout by reducing uric acid levels [55]. The extract of Smilax china L. can decrease uric acid levels, and its mechanism of action may be associated with the inhibition of XOD activity, suggesting its possible therapeutic impact on gout [56] (Table 1).
Herbal Medicines Originated from Other Families
Some Chinese herbal extracts can reduce serum uric acid levels by inhibiting the formation of uric acid, including corni fructus, which is the dry and mature pulp of Cornus officinalis Sieb. et Zucc (Cornaceae), Salvia plebeia R. Br. (Lamiaceae), Alocasia longiloba Miq. (Araceae), Olea europaea L. (Oleaceae) and Camellia japonica L.(Theaceae). The extracts of Chinese herbal medicines can reduce uric acid production by inhibiting XOD activity, which reduces serum uric acid levels, thus contributing to the prevention and treatment of gout [57-61].
Some Chinese herbal extracts reduce uric acid levels by promoting uric acid excretion. The ethanolic extract of the barks of Liriodendron chinense (Hemsl.) Sarg (Magnoliaceae) (EELC) can promote uric acid excretion, reduce uric acid accumulation in the kidneys, and decrease blood uric acid levels. The uric acid– lowering effect occurs through the upregulated expression of ABCG2, OAT1, and OTA3 proteins in the kidney. EELC can also inhibit the JAK2/STAT3 and NF-κB pathways. The enhancement of the ASK1/c-jun signaling pathway reduces the release of inflammatory factors and alleviates the symptoms of hyperuricemic nephropathy [62]. The extract of Polygonum capitatum Buch. Ham. ex D. Don (Polygonaceae)can downregulate the expression of GLUT9 and URAT1 to promote uric acid excretion. It is worth noting that the extract can also reduce uric acid production by hindering XOD activity [63]. Another study reported a similar result. The Mori Ramulus (Moraceae) extract can promote uric acid excretion by inhibiting the mRNA and protein expression of URAT1, GLUT9, and OAT1 and upregulating the mRNA and protein expression of OCT1/2 and OCTN1/2, respectively [64]. Han et al. reported that the extract of Urtica hyperborea Jacq. ex (Urticaceae) can significantly reduce uric acid levels in a hyperuricemia mouse model. In vitro studies have shown that the extract acts by upregulating OAT1 expression in HK2 cells and downregulating URAT1 expression [65]. The aforementioned herbals have therapeutic effects on gout, mainly by lowering uric acid levels (Table 1).
However, some herbals can treat gouty arthritis through their anti-inflammatory effect. For example, relative to the control group, the model group, and the treatment group, the Phyllanthus emblica (Phyllanthaceae) extract significantly reduced the inflammatory response in the ankle joint of rats with gout; it significantly inhibited the expression of NLRP3 and Caspase-1 in the synovial cells of the ankle joints of rats and decreased the levels of inflammatory factors such as IL-10, and TNF-α [66]. The seed extract of Apium graveolens L. (Apiaceae) can reduce the levels of IL-1β and TNF-α. [67]. The Aconitum carmichaelii Debeaux (Ranunculaceae) extract can evidently downregulate the mRNA expression of Caspase-1, PTGS2, spleen tyrosine kinase, MAPK14, matrix metalloproteinase (MMP) 9, and COX-2 in MSU-treated THP-1 cells in vitro. In vivo studies have shown that the extract can significantly inhibit the expression of serum IL-1β and IL-18 in gouty arthritis rats induced by MSU crystals [68]. Similarly, the extract of Cinnamomum cassia (Lauraceae) can reduce the secretion of the inflammatory factor IL-1β by activating AIM2, NLRP3 inflammasome [69]. Interestingly, the extract of Ginkgo biloba L. (Ginkgoaceae) leaves can also inhibit the production of serum IL-1β [70]. Liu et al. confirmed that the Eurycoma longifolia (Simaroubaceae) extract can effectively inhibit joint swelling in rats with gouty arthritis induced by MSU crystals. Network pharmacology, molecular docking, and the enrichment method of KEGG demonstrated that the Eurycoma longifolia extract might have a therapeutic effect on gouty arthritis by synergistically regulating TLRs, NLRs, and MAPK signaling pathway [71]. Similarly, Hua et al. speculated that the extract of Stauntonia brachyanthera Hand-Mazz (Lardizabalaceae) plays an anti-gout role by regulating MAPK, TLRs, NLRs, and PI3K-Akt signaling pathway [72] (Table 1).
herbals |
Model |
Functions |
Mechanisms |
Ref. |
|
Asteraceae |
Atractylodes
Lancea
(Thunb.) DC. |
SPF
SD rats RAW264.7
macrophages |
Anti-inflammatory |
IL-1β↓;
IL-6↓; TNF-α↓; PTGS2↓
|
[44] |
Atractylodes
macrocephala
Koidz. |
Male
SD rats |
Anti-inflammatory Anti-hyperuricemia Improve
renal injury |
IL-1β↓;
TNF-a↓; XOD↓ |
[45] |
|
Cichorium
intybus L. |
Male
SD rats macrophages |
Anti-inflammatory |
NLRP3↓;IL-1β↓; NF-κB
p65↓ |
[46] |
|
Siegesbeckia
orientalis L. |
Male
Wistar rats |
Anti-inflammatory Anti-hyperuricemia Inhibit
uric acid production |
XOD↓ |
[47] |
|
Helianthus
annuus Head |
Male
BALB/c mice |
Anti-inflammatory Anti-hyperuricemia |
IL-10↓;
MCP-1↓;XOD↓ |
[48] |
|
Dioscoreaceae |
Dioscorea
collettii |
THP‑1 cell |
Anti-inflammatory
|
NLRP3↓;IL-1β↓;
IL-18↓; TNF-α↓; |
[49] |
Discorea
nipponica Makino |
Male
SD rats
|
Anti-inflammatory
|
glutathione
metabolism, glycerol phospholipid metabolism, inositol phosphate metabolism, galactose
metabolism p38
MAPK, SDF-1/CXCR-4 |
||
Liliaceae |
Smilax
glabra Roxb |
Male
SD rats Male
BALB/c mice |
Anti-inflammatory Anti-hyperuricemia Promote
uric acid excretion Improve
kidney injury Inhibit
uric acid production |
IL-1β↓;
IL-6↓; IL-12↓; TNF-α↓;
TGF-β1↓; ABCG2↑;
OAT1↑; OCT2↑;
OCTN2↑ mURAT1↓;
XOD↓ |
[52-55] |
Smilax
china L. |
Male
ICR mice |
Anti-hyperuricemia |
XOD↓ |
[56] |
|
Cornaceae |
Cornus
officinalis Sieb. |
Male
ICR mice |
Anti-hyperuricemia |
XOD↓ |
[57] |
Lamiaceae |
Saluia
plebeia R.
Br. |
Male
ICR mice |
Anti-hyperuricemia |
XOD↓ |
[58] |
Araceae |
Alocasia
longiloba Miq. |
Male
SD rats |
Anti-hyperuricemia |
XOD↓ |
[59] |
Oleaceae |
Olea
europaea L. leaf |
Male
SD rats |
Anti-hyperuricemia |
XOD↓ |
[60] |
Theaceae |
Camellia
japonica L. |
Male
SD rats |
Anti-hyperuricemia |
XOD↓ |
[61] |
Magnoliaceae |
Liriodendron
chinense (Hemsl.) Sarg. |
Male
C57BL/6 mice |
Anti-inflammatory Anti-hyperuricemia Improve
kidney injury Promote
uric acid excretion |
TNF-α↓;
NF-κB
p65↓ OAT1↓; OAT3↓; ABCG2↓ |
[62] |
Polygonaceae |
Polygonum
capitatum
Buch. Ham. ex D. Don |
Male
Kunming mice |
Anti-inflammatory Anti-hyperuricemia Promote
uric acid excretion Improve
kidney injury Inhibit
uric acid production |
IL-1β↓;
IL-6↓; TNF-α↓; GLUT9↓;
URAT1↓; OAT1↓; XOD↓
|
[63] |
Moraceae |
Mori
Ramulus |
Male
Kunming mice |
Anti-hyperuricemia Promote
uric acid excretion
|
GLUT9↓;
URAT1↓; OAT1↓; mOCT1↑; mOCT2↑; mOCTN1↑; mOCTN2↑ |
[64] |
Urticaceae |
Urtica
hyperborea
Jacq. |
Human
renal tubular epithelial HK2 cells Male
Kunming mice |
Anti-hyperuricemia Promote
uric acid excretion Inhibit
uric acid production |
URAT1↓;
OAT1↓; XOD↓ |
[65] |
Phyllanthaeae |
Phyllanthus
emblica L. |
SPF
Male SD rats |
Anti-inflammatory |
NLRP3↓; caspase-1↓; MMP13↓; IL-1β↓;
IL-10↓; TNF-α↓; |
[66] |
Apiaceae |
Apium
graveolens L.
seed |
Male
BalB/c mice |
Anti-inflammatory Antioxidant
stress |
IL-1β↓;
IL-10↓; ROS↓ |
[67] |
Ranunculaceae |
Aconitum
carmichaelii
Debeaux |
Male
SD rats THP-1
cells |
Anti-inflammatory |
IL-1β↓;
IL-18↓; TNF-α↓; MAPK14↓; MMP9↓; PTGS2↓; |
[68] |
Lauraceae |
Cinnamomum
cassia Presl |
Bone
marrow-derived macrophages Male
C57BL/6 mice |
Anti-inflammatory |
NLRP3↓;
IL-1β↓ |
[69] |
Ginkgoaceae |
Ginkgo
biloba L.
leaves |
Male
SD rats |
Anti-inflammatory |
L-1β↓ |
[70] |
Simaroubaceae |
Eurycoma
longifolia Jack. |
Adult
Wistar albino rats |
Anti-inflammatory |
Toll-like
receptor, MAPK, chemokine, NOD-like receptor, T cell receptor signaling pathway |
[71] |
Lardizabalaceae |
Stauntonia
brachyantherand-Mazz |
|
Anti-inflammatory |
MAPK,
PI3K-Akt, Toll-like
Receptor, NOD-like
receptor signaling pathway |
[72] |
Table 1: Summary of Single herbs for gout
Therapeutic Effects of Natural Compounds from TCM for Gout
Polysaccharides
Polysaccharides isolated from herbal medicines have important pharmacological properties, including anti-inflammatory, anticancer, antioxidant, antidiabetic, antiviral, hypolipidemic, and immune regulation properties [73,74]. Isatidis Radix is a biennial herb of the Brassicaceae family. In TCM, Isatidis Radix plays an important role in heat clearance and detoxification [75]. As reported in modern pharmacological research, Isatidis Radix contains many important components, including polysaccharides, alkaloids, and organic acids [76].
Isatidis Radix polysaccharide (IRP) extracted from Isatidis Radix could inhibit the activation of multiple inflammasomes and alleviate gouty arthritis, as seen both in vivo and in vitro experiments. Under in vitro conditions, IRP can inhibit caspase-1 cleavage and IL-1β secretion by blocking NLRP3 inflammasome activation. Under in vivo conditions, IRP can alleviate joint swelling caused by MSU crystals in mice and reduce the secretion of MPO, IL-6, IL-18, and IL-1β in the swollen joints [77].
Plantaginis semen polysaccharide (PSP) can alleviate gout and renal damage. Oral administration of PSP (0.27 g/kg body weight, daily for 20 days) can significantly reduce serum uric acid and urine protein levels. PSP can also inhibit the expression of TNF-α, TGF-β, and IL-6 and downregulate the expression of caspase-1, ASC, and NLRP3 proteins. To sum up, PSP can alleviate gout by reducing uric acid levels, and PSP can alleviate gout nephropathy by reducing urinary protein levels and inhibiting the release of inflammatory factors by reducing the expression of caspase-1, ASC, and NLRP3 proteins [78].
Lonicera japonica polysaccharide (i.p., 0.1, 0.2, and 0.3 g/kg body weight) exerted anti-hyperuricemic effects in rats by inhibiting XOD activity. It also decreased the flares of gouty arthritis and alleviated ankle swelling in mice by downregulating the expression of IL-1β, IL-6, TNF-α, and COX-2–related inflammatory factors in murine serum [79]. (Table 2).
Sources |
Model |
Functions |
Mechanisms |
Ref. |
|
Isatidis
Radix
Polysaccharide |
Isatis
tinctoria
Linnaeus (Brassicaceae) |
male
C57BL/6 mice THP-1
cells BMDMs
cells |
Anti-inflammatory |
NLRP3↓;
IL-1β↓; IL-6↓; IL18↓; TNF-α↓ |
[77] |
|
Plantago
asiatica L. (Plantaginaceae) |
Male
SD rats |
Anti-inflammatory Improve
renal function |
NLRP3↓;
IL-6↓; TNF-α↓; TGF-β↓ |
[78] |
Lonicera
japonica Polysaccharide |
Lonicera
japonica
Thunb. (Caprifoliaceae) |
Male
SD rats |
Anti-inflammatory Anti-hyperuricemia
|
IL-1β↓;
IL-6↓; TNF-α↓; COX-2↓XOD↓ |
[79] |
Table 2: Summary of Polysaccharides for gout
Alkaloids
Dihydroberberine (DHB) is a derivative of berberine. Xu et al. investigated the effects of DHB on the reduction of uric acid levels and renal protection in hyperuricemic mice. Oral DHB (25 and 50 mg/kg body weight) decreased the uric acid level triggered by potassium oxazinate and hypoxanthine in a dosedependent manner. DHB significantly inhibits XOD activity, thereby decreasing uric acid production. DHB can also promote uric acid excretion by downregulating the expression of URAT1 and GLUT9. Additionally, DHB inhibits the mRNA expression of NLRP3, ASC, caspase-1, and IL-1β in the kidney to protect renal function [83].
Palmatine (PAL) is the main active ingredient in Corydalis yanhusuo. Juanjuan et al. demonstrated that PAL has the potential as an effective therapeutic candidate for gouty arthritis because of its anti-inflammatory activities. PAL treatment at doses of 20, 40, and 80 µm alleviated MSU-induced gouty arthritis in a rat model. PAL treatment dose-dependently inhibits the mRNA expression of IL-1β, IL-6, IL-18, and TNF-α. Additionally, it can promote the production of superoxide dismutase and glutathione (GSH) and reduce the production of malondialdehyde (MDA). Western blotting has shown that PAL can effectively inhibit the NF-κB/ NLRP3 signaling pathway by inhibiting the expression of NLRP3, ASC, IL-1β, and Caspase-1, while enhancing the expression of the antioxidant proteins Nrf2 and HO-1. Moreover, PAL further reduces the infiltration of neutrophils into synovitis [84]. Tetrahydropalmatine (THP), another effective compound from Corydalis yanhusuo, can attenuate MSU-induced gouty arthritis. Oral THP (10, 20, and 40 mg/kg body weight) dose-dependently reduced MSU-induced plantar swelling, and when compared with the model group, the levels of inflammatory factors in the treatment group decreased and the infiltration of inflammatory cells weakened, both of which are attributed to the role of THP in inhibiting NLRP3 inflammasome activation and caspase-1 formation. THP also reduces ROS production induced by MSU crystal deposition by increasing the activity of antioxidant enzymes under both in vivo and in vitro conditions. It is worth noting that the impact of a high dose is equivalent to that of a positive drug [85].
Coptisine, the main effective component of Coptis chinensis, can significantly inhibit the secretion of mature IL-1β from RAW264.7 macrophages stimulated by LPS together with MSU, ATP, and nigericin, by inhibiting caspase-1 activation. Moreover, it blocks the NF-κB signaling pathway to inhibit NLRP3 expression, thereby preventing the priming of inflammasome. Under in vivo conditions, oral administration of coptisine reduced the degree of foot swelling in mice of the treatment group. The aforementioned findings suggest that coptisine can be used to treat gouty arthritis caused by NLRP3 inflammasomes [86]. (Table 3).
Monomers |
Sources |
Model |
Functions |
mechanisms |
Ref. |
Berberrubine |
phellodendri chinensis cortex (Rutaceae) |
male KM mice |
Anti-inflammatory Anti-hyperuricemia Improve kidney injury Inhibit uric acid production Promote uric acid excretion |
IL-1β↓; IL-6↓; TNF-α↓; URAT1↓; GLUT9↓; OAT1/3↑; ABCG2↑; XOD↓ |
[82] |
Dihydroberberine |
phellodendri chinensis cortex (Rutaceae) |
male KM mice |
Anti-inflammatory Anti-hyperuricemia Improve kidney injury Inhibit uric acid production Promote uric acid excretion |
NLRP3↓; IL-1β↓; IL-6↓; IL18↓; TNF-α↓; URAT1↓; GLUT9↓; XOD↓ |
[83] |
Palmatine |
Corydalis yanhusuo (Papaveraceae) |
male KM mice THP-1 cells |
Anti-inflammatory
|
IL-1β↓; IL-6↓; IL18↓; TNF-α↓; MDA↓; Nrf2↓; HO-1↓ |
[84] |
Tetrahydropalmatine |
Corydalis yanhusuo (Papaveraceae) |
Male C57BL/6 mice RAW264.7 macrophage |
Anti-inflammatory Antioxidant stress reaction |
NLRP3↓;IL-1β↓; IL-6↓; IL18↓; TNF-α↓;
ROS↓ |
[85] |
Coptisine |
Male Kun Ming mice RAW 264.7 cell macrophage |
Anti-inflammatory
|
NLRP3↓; IL-1β↓; TNF-α↓ |
[87] |
Table 3: Summary of Alkaloids for gout
Phenolic Compounds
Phenolic compounds exert a wide range of pharmacological activities, including antioxidation, anti-gout, mprovement of vascular endothelial function, anti-atherosclerosis, and inhibition of platelet aggregation [87].
Caffeic acid can effectively reduce serum uric acid levels in hyperuricemic mice triggered by potassium oxazinate, and it acts by inhibiting XOD activity in the liver [88,89].
Chlorogenic acid alleviates mouse foot swelling induced by MSU crystals by blocking the expression of IL-6, IL-1β, and TNF-α. Moreover, chlorogenic acid alleviates gouty arthritis [90]. With regard to the mechanism of chlorogenic acid in relieving gouty arthritis, its action may show an association with the regulation of mitogen-activated protein kinase (MAPK), PI3KAkt, and NF-κB signaling pathways and the intervention of TLRs, affecting the release of cellular inflammatory factors [91].
Vanillin, a phenolic compound, is an effective component in Amomum villosum Lour. Under in vitro conditions, vanillic acid inhibits XOD activity [92], but its effect on uric acid has not been investigated. Based on previous experimental results, we speculate that vanillic acid may have a beneficial effect on gouty arthritis by reducing serum uric acid levels.
Paeonol is an effective component of Moutan Cortex. Chen et al. reported that paeonol can attenuate gouty arthritis induced by MSU crystals. Specifically, oral paeonol reduced foot swelling induced by MSU crystals in rats; reduced the expression of TNF-α, IL-1β, and IL-6; upregulated the protein expression of P65 in the nucleus; and significantly inhibited NF-κB DNA binding activity. The aforementioned results indicate that paeonol may play an antiinflammatory role and hinder the expression of proinflammatory cytokines and NF-κB [93].
6-Shogaol is an active ingredient extracted from Zingiber officinale Roscoe. that can effectively reduce serum uric acid levels by inhibiting XOD activity. Moreover, 6-shogaol can also reduce the production of inflammatory cytokines such as IL-1β and TNF-α [94] (Table 4).
Monomers |
Sources |
Model |
Functions |
Mechanisms |
Ref. |
Caffeic acid |
Tabebuia roseoalba (Bignoniaceae) |
male albino Swiss mice |
Anti-inflammatory Anti-hyperuricemia |
XOD↓ |
|
Chlorogenic acid |
Tabebuia roseoalba (Bignoniaceae) Porana sinensis Hemsl. (Convolvulaceae) Smilax china L. (Liliaceae) |
male albino Swiss mice male Kun-Ming strain of mice |
Anti-inflammatory Anti-hyperuricemia Inhibit uric acid production
|
IL-1β↓; IL-4↓; IL-6↓; TNF-α↓; TGF-β↓; XOD↓ |
|
Vanillic acid |
Amomum villosum Lour. |
|
Anti-hyperuricemia Inhibit uric acid production
|
XOD↓ |
[92] |
Paeonol |
Paeonia suffruticosa Andr. |
Male SD rats |
Anti-inflammatory
|
IL-1β↓; IL-6↓; TNF-α↓ |
[93] |
6-shogaol |
Zingiber
officinale Roscoe |
Male SD rats |
Anti-inflammatory Anti-hyperuricemia |
IL-1β↓; TNF-α↓; XOD↓ |
[94] |
Table 4: Summary of Phenolic compounds for gout
Flavonoids
Most Chinese herbal medicines contain flavonoids. They have a wide range of pharmacological activities, including antiinflammatory, antioxidant, immune enhancement, anticancer, and other effects [95].
Icariin is the main active ingredient in Epimedium. Cao et al. confirmed the therapeutic effect of icariin against gouty arthritis and reported that icariin (20, 40, and 80 mg/kg body weight) inhibits the expression of IL-1β, IL-6, PGE2, and TNF-α in the synovial tissue, alleviates the infiltration of inflammatory factors, and weakens ankle joint swelling in rats. Moreover, icariin can reduce the expression of the NALP3 inflammasome by inhibiting the nuclear transcription of proteins participating in the NF-κB pathway. In vitro studies showed that icariin reduced the activity of chondrocytes and inhibited the levels of the inflammasome and glycosaminogly [97].
Luteolin and luteolin-4′-O-glucoside, effective components extracted from Gnaphalium affine D. Don., can inhibit the protein expression of URAT1 and the activity of XOD to reduce uric acid production and increase uric acid excretion, thereby alleviating hyperuricemia symptoms. It can also reduce foot swelling induced by MSU crystals and the production of inflammatory cytokines such as IL-1, IL-β, and TNF-α [98]. Other studies have obtained consistent results, that is, luteolin-4′-O-glucoside and luteolin-4′O-glucoside can alleviate hyperuricemia and gouty arthritis [99].
Chrysin, a flavonoid compound extracted from Oroxylum indicum (L.) (Bignoniaceae) has the effect of anti-inflammation, anti-oxidation and anti-hyperuricemia. Chang et al. reported that oral administration of chrysin (50, 100, and 150 mg/kg body weight, daily for 28 days) lowered serum uric acid levels in hyperuricemia induced by high-fructose syrup in a dose-dependent manner. Moreover, chrysin can inhibit XOD activity; western blotting analysis demonstrated that the protein expression of URAT1 and GLUT9 showed an evident decrease, whereas that of OAT1 and human ABCG2 protein was increased. Chrysin can significantly reduce IL-1β levels in the kidney and serum of rats and MDA levels, suggesting that chrysin has significant antioxidant and antiinflammatory activities. It is worth noting whether such a high dose (50mg/kg body weight) will produce toxic and side effects in experimental animals [100].
Quercetin, a flavonoid compound widely dispersed throughout the plant kingdom has the effect of anticancer, antioxidant and anti-gouty arthritis. Feng et al. predicted 72 potential targets of quercetin for gout using network pharmacology analysis and constructed an interaction network between quercetin and gout. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the IL-17, TNF-α, MAPK, and PI3K signaling pathways were the main involved pathways. Quercetin administration to mice with MSU-induced gouty arthritis, relative to the model group and the control group, significantly reduced ankle joint swelling, hindered the expression of IL-6/17A/F in the IL-17 signaling pathway, and reduced the expression of retinoic acid–related orphan receptor, TNF-α, and TGF-β, all of which are in line with those obtained previously [101].
(-)-Epicatechin (EC) is the main active component in some medicinal plants and food, with anti-inflammatory and anti-oxidant effects. Chen et al. conducted in-depth research on its role and mechanism in gouty arthritis treatment. First, in vitro experiments showed that EC could increase the activity of THP-1 cells treated with MSU; reduce the levels of IL-6, IL-18, IL-1β, and TNF-α in the supernatant of THP-1 cells; and significantly increase the expression of ASC, NLRP3, and caspace-1 induced by MSU administration. Nuclear P65 expression was found to be reduced, suggesting that EC restricted P65 from entering the nucleus. The same results were obtained during the in vivo experiments either [102].
Baeckein E (BF-2) is an active compound isolated from Baeckea frutescens L. BF-2 evidently blocks IL-1β secretion, inhibits the thermophilicity of macrophages, and alleviates ankle swelling. BF-2 also alleviates oxidative stress by inhibiting the combination of pre-Caspase 1 and ASC, thereby inhibiting the assembly of the NLRP3 inflammasome. More interestingly, BF-2 can block the activation of the NLRP3 inflammasome by inhibiting the MAPK/NF-κB signaling pathway [103] (Table 5).
Monomers |
Sources |
Model |
Functions |
Mechanisms |
Ref. |
Kaempferol |
Cudrania tricuspidata Leaf (Moraceae) |
male ICR mice |
Anti-hyperuricemia |
XOD↓ |
[96] |
Icariin |
Epimedium brevicornu Maxim (Berberidaceae) |
Male SD rats chondrocytes |
Anti-inflammatory |
NLRP3↓;IL-1β↓; IL- 6↓; TNF-α↓; PGE2↓; |
[97] |
Luteolin |
Gnaphalium affine D. Don. (Asteraceae) |
male Kun-Ming mice male ICR mice |
Anti-inflammatory Anti-hyperuricemia Improve kidney injury Inhibit uric acid production Promote uric acid excretion |
IL-1β↓; TNF-α↓; URAT1↓; GLUT9↓; XOD↓ |
[98,99] |
Luteolin-4′-O-glucoside |
|||||
Chrysin |
Many plants |
Male SD rats |
Anti-inflammatory Anti-hyperuricemia Antioxidant stress reaction Inhibit uric acid production Promote uric acid excretion |
IL-1β↓; ROS↓; URAT1↓; GLUT9↓; OAT1↑; ABCG2↑ |
[100] |
Quercetin |
Many plants |
Male SD rats |
Anti-inflammatory |
IL-1β↓; IL-6↓; IL-17A↓; IL-17F↓; TNF-α↓ |
[101] |
(-)-Epicatechin (EC) |
Many plants |
Male C57BL/6 mice THP-1 cells |
Anti-inflammatory |
NLRP3↓;IL-1β↓; IL6↓; IL-18↓; TNF-α↓; |
[102] |
Baeckein E |
Baeckea frutescens L. (Myrtaceae) |
J774A.1 macrophages male C57BL/6 mice |
Anti-inflammatory Antioxidant stress reaction |
NLRP3↓;IL-1β↓; ROS↓ |
[103] |
Table 5: Summary of Flavonoids for gout
Quinones
Rhein, a active components of Rheum tanguticum Maxim. ex Regel, can significantly inhibit the production of IL-1β and TNF-α by macrophages and caspase-1 protein. The rate of inhibition of IL-1β by rhein at a concentration of 2.5 µg/mL is 47%, and rhein (5 µg/mL) can significantly reduce ASC content and inhibit NLRP3 expression. To sum up, rhein alleviates gouty arthritis by inhibiting the activation of the NLRP3 inflammasome and macrophages [104].
Polydatin , a quinone compound isolated from Reynoutria japonica Houtt., has a wide range of pharmacological effects, including its efficacy in preventing and treating diabetes, atherosclerosis, hyperlipidemia, and gout [105]. Oral administration of polydatin (20 and 50 mg/kg body weight) to hyperuricemic rats induced by potassium oxazinate reduced the levels of uric acid and creatinine in blood and urine respectively and achieved the same effect as that of the positive drug allopurinol. It can also restore TNF-α, IL-1, IL-β, and IL-6 levels in the serum and kidney of the rats in the model group to the original level. Polydatin blocked NF-κB protein expression and significantly lowered IκBα phosphorylation. Additionally, western blotting showed that the expression of ASC, Caspase-1, and NLRP3 was inhibited. Furthermore, polydatin upregulated the expression of p-AMPK and SIRT1 proteins in the kidney. It is speculated that polydatin may inhibit the production of NF-κB and NLRP3 inflammasome in the kidney by regulating the AMPK/SIRT1 signaling pathway in gout treatment [106] (Table 6).
Monomers |
Sources |
Model |
Functions |
Mechanisms |
Ref. |
Rhein |
Rheum tanguticum Maxim. ex Regel (Polygonaceae) |
THP-1 cells |
Anti-inflammatory |
IL-1β↓; TNF-α↓; caspase-1↓ |
[104] |
Polydatin |
Reynoutria japonica Houtt. (Polygonaceae) |
Male SD rats |
Anti-inflammatory Anti-hyperuricemia Improve kidney injury |
IL-1β↓; IL-6↓; TNF-α↓; NF-κB p65↓ |
[106] |
Table 6: Summary of Quinones for gout
Saponins
Saponin is an effective active component of many Chinese herbal medicines. Modern pharmacology has demonstrated that saponin has diverse biological activities, including anti-inflammatory, anticancer, hypoglycemic, and other effects [107].
Smilaxchinoside A and Smilaxchinoside C are steroidal saponins isolated from the ethanolic extract of Smilax riparia. Wu et al. reported that these two compounds inhibit uric acid production and stimulate uric acid excretion by inhibiting mURAT1 expression in the kidney and XOD activity. Smilaxchinoside A and Smilaxchinoside C are, therefore, potential clinical drugs for gout treatment [108]. Riparoside B and timosaponin J, which are also extracted from Smilax riparia, have the same effect as that of Smilaxchinoside A and Smilaxchinoside C and promote uric acid excretion by inhibiting URAT1 expression [109] (Table 7).
Monomers |
Sources |
Model |
Functions |
Mechanisms |
Ref. |
Smilaxchinoside A |
Smilax riparia . (Liliaceae) |
Male Kunming mice |
Anti-hyperuricemia Inhibit uric acid production Promote uric acid excretion |
mURAT1↓; mGLUT9↓; XOD↓; OAT1↑ |
[108] |
Smilaxchinoside C |
|||||
Riparoside B |
Smilax riparia (Liliaceae) |
Male Kunming mice |
Anti-hyperuricemia Inhibit uric acid production Promote uric acid excretion |
mURAT1↓; XOD↓ |
[109] |
Timosaponin J |
Table 7: Summary of Saponins for gout
Terpenoids
Terpenoids are widely present in many plants and have extensive pharmacological and biological activities [110].
Andrographolide (AND), the main component of Andrographis paniculata (Burm. f.) Nees, can inhibit IL-1β production by bone marrow macrophages in mice with gout induced by MSU crystals and infiltration of monocytes. AND interacts with the IKK/NF-κB signaling pathway and inhibits the activation of the NLRP3 inflammasome (endotoxin induced), leading to the downregulation of NLRP3 and Pro-IL-1β protein expression. AND also inhibits HO-1 protein expression and reduces ROS production. It is worth noting that AND reduces the combination of ASC and NLRP3. AND alleviates gouty arthritis by inhibiting ROS-induced NLRP3 inflammasome assembly and IL-1β release by increasing HO-1 expression [111].
Cucurbitacin B (CUB) is a terpenoid compound isolated from plants belonging to Cucurbitaceae. Xue et al. confirmed that CUB could significantly reduce IL-1β secretion by blocking the formation of NLRP3 inflammasome and inhibiting the key enzymes of macrophage glycolysis. Through histopathological analysis, the authors also showed that CUB improved foot swelling and inflammatory cell infiltration in gouty arthritis mice [112].
Tanshinones is a class of effective biological components in Salvia miltiorrhiza Bge. Yue et al. identified 15 tanshinones that can inhibit NLRP3 inflammasome activation, among which tanshinone IIA, isocryptotanshinone, and dihydrotanshinone I can reduce the production of oxygen free radicals in macrophages. Tanshinone protects mitochondria by promoting autophagic protein kinase and AMP-activated protein kinase, suggesting that tanshinone is effective for treating gouty arthritis [113].
Budlein A is a metabolite in the biosynthesis of sesquiterpene lactones in Asteraceae plants. Budlein A can alleviate knee joint swelling, reduce neutrophil aggregation, inhibit neutrophil phagocytosis due to the deposition of MSU crystals, and block the mRNA expression of IL-1β and TNF-α in the knee joint tissue homogenate. It is worth noting that inhibition of NF-κB activation may reduce TNF-α production. The aforementioned results suggest that Budlein A alleviates gouty arthritis by activating NF-κB and promoting the assembly of inflammatory factors [114].
Tripterine pentacyclic triterpene compound isolated from Tripterygium wilfordii Hook., can alleviate gouty arthritis. Specifically, it can reduce the swelling degree in MSU-induced joint swelling in mice. A high dose of tripterine (1 mg/kg body weight) can reduce the swelling degree by 83.9%, and a low dose can reduce the swelling degree by 72.4%. Hematoxylineosin staining analysis has shown that tripterine can reduce the infiltration of inflammatory cells. Additionally, enzyme-linked immunosorbent assay has demonstrated that tripterine inhibited the secretion of IL-1β. The protein expression of Caspase-1 was also downregulated [115] (Table 8).
Monomers |
Sources |
Model |
Functions |
Mechanisms |
Ref. |
Andrographolide |
Andrographis paniculata (Burm. f.)
Nees (Acanthaceae) |
male C57BL/6J mice Bone marrow-derived macrophages |
Anti-inflammatory Antioxidant stress |
NLRP3↓;IL-1β↓; caspase-1↓; HO-1↓; ROS↓ |
[111] |
Cucurbitacin B |
Cucurbitaceae plants |
Male C57BL/6 mice Bone marrow-derived
macrophages |
Anti-inflammatory
|
IL-1β↓; IL-18↓; HK1↓; HK2↓; PKM2↓; LDHA↓; PDH↓; |
[112] |
Tanshinone IIA |
Salvia miltiorrhiza Bge. (Lamiaceae) |
Male C57BL/6 mice J774A.1 macrophages |
Anti-inflammatory Antioxidant stress |
IL-1β↓; ROS↓ |
[113] |
Isocryptotanshinone |
|||||
Dihydrotanshinone I |
|||||
Budlein A |
Asteraceae plants |
Male C57BL/6 mice Bone marrow-derived macrophages |
Anti-inflammatory
|
IL-1β↓; TNF-α↓; NF-κB p65↓ |
[114] |
Tripterine |
Tripterygium wilfordii Hook.
f. (Celastraceae) |
male WT C57BL/6J mice HEK293T cells THP-1 cells Bone marrow-derived macrophages |
Anti-inflammatory Improve liver injury |
NLRP3↓; IL-1β↓; |
[115] |
Table 8: Summary of Terpenoids for gout
Glycosides
Gentiopicroside (GPS) is an iridoid glycoside isolated from Gentiana Macrophylla Pall. GPS can reduce foot swelling caused by MSU crystals; block the secretion of inflammatory factors, namely, IL-1β, IL-6, IL-18, and TNF-α; and significantly inhibit neutrophil infiltration. Immunosuppression analysis has demonstrated that GPS significantly restored the expression of NLRP3, ASC, and Caspase-1 proteins in the model group. GPS can inhibit the activation of NLRP3 inflammasomes in macrophages under in vitro conditions. GPS can be used to treat gouty arthritis by hindering the activation of the NLRP3 inflammasome [116].
Kinsenoside is the main effective ingredient in Anoectochilus roxburghii (Wall.) Lindl. in the treatment of gouty joints. Kinsenoside can negatively regulate the NF-κB/MAPK signaling pathway, thereby reducing the expression of inflammatory cytokines present downstream of NF-κB and inhibiting the biological activity of vascular endothelial cells. Kinsenoside can also inhibit the expression of NALP3 and TLR2 and the uptake of CD14 mediated by MSU crystals in macrophages. Under in vivo conditions, kinsenoside can significantly reduce inflammatory cell infiltration and endothelial damage and alleviate ankle swelling [117].
Total glucosides of paeony,the main components of Paeoniae Radix Alba, regulates the interaction among MALAT1, MIMR8765p, and NLRP3 proteins, blocks the TLR4/MYD88/NF-κB signaling pathway, and inhibits the THP-1 macrophage inflammation induced by MSU crystals, thus producing a therapeutic effect on gouty arthritis [118] (Table 9).
Monomer s |
Sources |
Model |
Functions |
Mechanisms |
Ref. |
Gentiopicroside |
Gentiana Macrophylla Pall. (Gentianaceae) |
Male C57BL/6 mice RAW264.7 macrophage cell |
Antiinflammatory |
NLRP3↓; IL-1β↓; IL-6↓; IL18↓; |
[116] |
Kinsenoside |
Anoectochilus roxburghii (Wall.) Lindl. (Orchidaceae) |
human umbilical vein endothelial cell Male SD rats |
Antiinflammatory |
IL-1β↓; IL-6↓; TNF-α↓; COX-2↓; NF-κB p65↓; PGE2↓; |
[117] |
Total glucosides of paeony |
Paeonia lactiflora Pall. (Paeoniaceae) |
THP-1 cells |
Antiinflammatory |
NLRP3↓; Caspase-1↓; IL-1β↓; MALAT1/miR-876-5p/NLRP3 axis TLR4/MyD88/NF-κB |
[118] |
Table 9: Summary of Glycosides for gout
TCM Prescription for Gout
Ermiao Pill is made up of phellodendron chinense Schneid. (Rutaceae) and Atractylodes lancea (Thunb.) DC. (Asteraceae), and it was introduced in “Dan Xi Xin Fa” by Zhu Danxi during the Yuan Dynasty in China. Used in the treatment of gouty arthritis, rheumatoid arthritis, and arthralgia [119,120]. The mechanism of the uric acid lowering effect of Ermiao Pill was linked to the regulation of purine metabolism, taurine metabolism, hypotaurine metabolism, glyceride metabolism, amino acid metabolism, and primary bile acid metabolism [121]. By inhibiting the expression of the proinflammatory factor IL-6, it can also reduce ankle swelling in gouty arthritis rats. Ermiao Wan can be used to treat gouty arthritis by lowering uric acid levels and inhibiting inflammatory reactions; however, its effective components and mechanism need to be investigated further [122].
Sanmiao Pill is a Chinese medicinal compound derived from the Ming Dynasty medical book “Yi Xue Zheng Zhuan”, which consists of three components, Phellodendron chinense Schneid. (Rutaceae ), Atractylodes lancea (Thunb.) DC. (Asteraceae), and Achyranthes bidentata Blume ( Amaranthaceae). Due to dampheat in the lower energizer, it is primarily used in the clinical treatment of gout [123]. Zhu et al. demonstrated that Sanmiao Pill could treat gout by inhibiting cartilage matrix degradation. The cartilage matrix is primarily made up of proteoglycan and collagen. In particular, when compared to the model group, the treatment group’s oral administration of Sanmiao Pill significantly reduced glycosaminoglycan expression while increasing proteoglycan levels. Notably, Sanmiao Pill significantly inhibited MMP-3 and aggrecanase-4 expression while increasing TIMP-1 and TIMP-3 expression [124]. Furthermore, Sanmiao pill reduced joint swelling in a gouty arthritis mouse model induced by MSU crystals and inhibited the production of IL-1β, TNF-α, and MPO. Surprisingly, the alcoholic extract of Sanmiao pill has a stronger anti-inflammatory effect than the aqueous extract [125].
Simiao Pill was derived from China’s “Cheng Fang Bian Du” in the Qing Dynasty, which comprises four individual herbs, namelyPhellodendron chinense Schneid. (Rutaceae),Atractylodes lancea (Thunb.) DC. (Asteraceae), Achyranthes bidentata Blume (Amaranthaceae), Coix lacryma-jobi L.var.mayuen( Roman.) Stapf (Poaceae). It is clinically used to treat damp-heat pouring downward gout [126]. Cao et al. found that by promoting PI3K/Akt activation and M2 polarization in macrophages, Simiao Pill (1 and 10 mg/ kg body weight) could significantly inhibit IL-6 and IL-1β levels and alleviate the inflammatory response caused by MSU crystals [127]. Simiao Pill can also reduce serum inflammatory factors like MIP-1α, MIP-1β, interferon-γ, and IL-9, downregulate NLRP3 expression, reduce intestinal cell apoptosis, and improve intestinal microflora to treat gouty arthritis [128]. Network pharmacology has discovered that Simiao Pill can also inhibit IL-17 and TNF-α [129]. Simiao Pill has been shown to lower serum uric acid levels in other studies. Simiao Pill, in particular, inhibited XOD activity and promoted uric acid excretion by inhibiting the expression of OAT1, URAT1, and GLUT9 transporters in the kidney [130,131]. Furthermore, modified Simiao pill can reduce joint swelling in rats with gouty arthritis caused by MSU crystals and can inhibit the expression of inflammatory factors like IL-6, IL-1β, and TNF-α. The p-STAT3 and MMP3 proteins were upregulated in C28/I2 cells in cartilage tissue, but the tissue inhibitor of metalloproteinase (TIMP)-3 protein was downregulated [132-134].
The Quzhuo-Tongbi decoction (QTD) is a TCM empirical prescription used clinically to treat acute gouty arthritis that consists of nine different Chinese herbal medicines (Table 10). Oral administration of QTD (0.5 g/kg body weight) inhibited the expression of NLRP3, reduced the production of IL-1β and TNF-α, and alleviated ankle swelling in rats with gouty arthritis caused by MSU crystals [135]. Other research has found that QTD can treat gout by regulating intestinal flora. QTD specifically improves intestinal barrier function by increasing the abundance of butyrate-producing bacteria and encouraging the production of SCFAs. Notably, a lack of SCFAs was linked to increased glycolysis. Furthermore, QTD can lower serum uric acid levels, and its mechanism may be linked to increased ABCG2 expression [136,137].
Zisheng-Shenqi decoction (ZSD), derived from “the Golden mirror of medicine” in China’s Qing dynasty, is made up of ten Chinese herbal medicines (Table 10) and has the function of tonifying the kidneys and removing dampness. It is used to treat gout in clinical settings. According to network pharmacology, ZSD can inhibit inflammatory reactions, resist oxidative stress, and relieve pain by regulating the ATP-P2X7R and NLR signaling pathways, thereby treating gout [138]. Another study found comparable results. ZSD can effectively treat gouty arthritis in vivo due to its anti-inflammatory and antioxidant properties. Oral ZSD (400 mg/10 g body weight) administration could inhibit the expression of inflammatory factors TNF-α and IL-1β, as well as the activation of NF-κB. Furthermore, glutathione peroxidase and superoxide dismutase activated the antioxidant stress effects. This study’s findings provided sufficient evidence for its use in gout treatment [139].
Guizhi-Shaoyao-Zhimu decoction (GSZD) is a classical prescription in China, which is derived from “Outline of the Golden Chamber”. It is a combination of nine Chinese herbal medicines
(Table 10) that is clinically used to treat gout and osteoarthritis [140]. By blocking the NLRP3/NF-κB signaling pathway, Zhou et al. demonstrated that GSZD could inhibit the expression of IL-1β, IL-6 inflammatory factor, and MCP-1 levels. Therefore, an antiinflammatory effect is produced. Furthermore, GSZD can lower serum uric acid levels [141,142].
The Shuang-Qi gout capsule (SQGC) is made up of six different herbs:Phragmites australis (Cav.) Trin. ex Steud. (Poaceae), Berchemia floribunda (Wall.) Brongn. (Rhamnaceae), Mallotus apelta (Lour.) Muell. Arg. (Euphorbiaceae), Schefflera arboricola Hay. (Araliaceae), Cinnamomum camphora (L.) presl (Lauraceae), Panax notoginseng (Burkill) F. H. Chen ( Araliaceae). It is used in the treatment of gouty arthritis. SQGC (57, 113, and 225 mg/kg body weight) reduced ankle joint swelling in a dose-dependent manner while also significantly inhibiting the expression of IL-1β and TNF-α. It is worth noting that the high-dose Shuang-qi gout capsule followed the same pattern as the positive drug [143]. Furthermore, SQGC can increase uric acid excretion. The mechanism is to inhibit URAT1 and GLUT9 expression while increasing the expression of OCT1, OCT2, and OAT1 [144].
Huzhen-Tongfeng formula (HTF) is an empirical prescription developed by Jinan University based on clinical experience with gouty arthritis.polygonum cuspidatum Sieb. et Zucc. (Polygonaceae),ligustri Lucidi Fructus (Oleaceae),plantago asiatica L. (Plantaginaceae), and polistes olivaceus (DeGeer) (Vespidae) are included in the prescription. Deng et al. revealed the mechanism of HTF for the first time in the treatment of gouty arthritis. Network pharmacology analysis predicted that the arachidonic acid metabolic pathway was most closely related to its anti-inflammatory action. In vivo studies revealed that oral administration of HTF reduced ankle swelling and inflammatory cell infiltration in rabbits with gouty arthritis. In vitro, HTF inhibited the expression of COX-1, COX-2, and 5-lipoxygenase [145]. Furthermore, HTF can be used to treat gouty arthritis by inhibiting the release of proinflammatory factors such as IL-1, IL6, TNF-α, and ASC oligomerization [146].
Jiangsuan-Chubi formula (JCF) was a traditional Chinese medicine prescription obtained through clustering analysis and frequency of use, consisting of 12 Chinese herbal medicines (Table 10) and used clinically to treat gouty arthritis. JCF has been shown in studies to inhibit the secretion of the inflammatory factor IL-1β by inhibiting the expression of the NRLP3 protein, resulting in an anti-gouty arthritis effect [147]. Qingre-Lishi decoction (QLD) is made up of 12 Chinese herbal medicines (Table 10) and is used to promote blood circulation, remove blood stasis, and relieve pain. It is used in the treatment of gout. Yu et al. confirmed that QLD could treat gouty arthritis by inhibiting NRLP3 protein expression and inflammatory factor IL-1β secretion [148]. Huzhang-Guizhi formula (HGF) is a traditional Chinese medicine derived from “Taiping Shenghui Fang,” which contains polygonum cuspidatum Sieb.et Zucc. (Polygonaceae) and cassia Twig ( Lauraceae). HGF, according to research, can inhibit the production of TNF-α and IL-1β in synovial fluid and the expression of the NF-KB p65 protein in synovial tissue, resulting in anti-gouty arthritis effects [149]. The ingredients in Shaoyao-Gancao decoction (SGD) are
Paeonia lactiflora Pall. (Paeoniaceae) and Glycyrrhiza uralensis Fisch. (Fabaceae). SGD significantly reduced PGE2 production in vitro by inhibiting the release of inflammatory factors COX2, IL-1β, and IL-6, thereby exerting an anti-gouty arthritis effect [150]. Tongfengding capsule is a traditional prescription that has been approved by the China Pharmacopoeia Committee (2020). It contains eight Chinese herbal medicines (Table 10). The medication is clinically used to treat gout and rheumatoid arthritis. Tongfengding capsule has been shown in studies to have an anti-gouty arthritis effect by inhibiting the secretion of the inflammatory factors TNF-α and IL-6. Furthermore, its mechanism may be linked to the inhibition of the arachidonic acid signaling pathway [151]. Xiaofeng granule is made from a new formula that has been improved from modified simiaowan and has heatclearing and dampness-drying properties. Shi et al. discovered that Xiaofeng granules can help with gout by inhibiting cartilage matrix degradation [152].
Shenling-Baizhu San (SBS) is a well-known prescription that is derived from “Prescriptions of the Bureau of Taiping People’s Welfare” and contains ten Chinese herbal medicines. Used in the treatment of gout and hyperuricemia. According to studies, SBS lowers serum uric acid levels by increasing uric acid excretion. Furthermore, it has been shown to inhibit the expression of p-PPARγSer273 in renal tubular epithelial cells [153]. Erding Formula (EF) is a traditional Chinese medicine prescription included in the China Pharmacopoeia, and it consists of viola philippica (Violaceae),taraxacum mongolicum Hand.-Mazz. (Asteraceae),lobelia chinensis Lour. (Campanulaceae) and isatis indigotica Fortune (Brassicaceae). According to pharmacological studies, EF can promote uric acid excretion by inhibiting URAT1 expression and increasing OAT3 expression [154]. Tongbixiao Pills (TBX) were a Chinese medicinal prescription developed by the Gout TCM Research Cooperation Group of Jingzhou Hospital of Traditional Chinese Medicine for the clinical treatment of gout. TBX has been shown in studies to have anti-gouty arthritis properties. Furthermore, it can lower serum uric acid levels by increasing uric acid excretion [155]. Tongfengtai granules (TFT) are a combination of seven Chinese herbal medicines that are clinically used to treat gouty nephropathy and rheumatism. Studies have shown that TFT can reduce serum uric acid levels by inhibiting XOD activity [156]. (Table 10).
Prescription |
Composition of prescription |
Model |
Effect |
Action mechanism |
Ref |
Er Miao pill |
Phellodendron chinense Schneid. (Rutaceae)Atractylodes lancea (Thunb.) DC. (Asteraceae) |
Male SD rats |
Antiinflammation Antihyperuricemia Promote uric acid excretion |
IL-6↓ |
[121,122] |
Sanmiao pill |
Phellodendron chinense Schneid. (Rutaceae) Atractylodes lancea (Thunb.) DC. (Asteraceae) Achyranthes bidentata Blume (Amaranthaceae) |
Male SD rats chondrocyte |
Antiinflammation Lowering uric acid level Inhibit cartilage matrix degradation |
IL-1β↓; TNF-α↓; IL-6↓ MMPs-3↓; ADAMTS-4↓ TIMPs-1↑; TIMPs -3↑ |
[124,125] |
Simiao pill |
Phellodendron chinense Schneid. (Rutaceae) Atractylodes lancea (Thunb.) DC. (Asteraceae) Achyranthes bidentata Blume (Amaranthaceae) Coix lacryma-jobi L.var.mayuen (Roman.)Stapf (Poaceae) |
THP-1 cells; Male C57BL/6 mice Male Kun- Ming strain of mice |
Antiinflammation Antihyperuricemia Regulating intestinal flora Lowering uric acid level Promote uric acid excretion |
NLRP3↓; IL6↓; IL-1β↓; TNF-α↓; p-Akt↑; MPO↓; XOD↓; ADA↓; mURAT1↓; mGLUT9↓; mOAT1↑; mOCT2↑ |
[127-131] |
Modified Simiao pill |
Phellodendron chinense Schneid. (Rutaceae) Atractylodes lancea (Thunb.) DC. (Asteraceae) Achyranthes bidentata Blume (Amaranthaceae) Coix lacryma-jobi L.var.mayuen (Roman.)Stapf )Poaceae) Smilax glabra Roxb. (Liliaceae) Lonicerajaponica Thunb (Caprifoliaceae) |
Male SD rats C28/I2 cell THP-1 cell |
Antiinflammation Antihyperuricemia |
IL-1β↓; TNF-α↓; IL-6↓; MMP-3↓; TIMP-3↑ |
[132-134] |
Quzhuo-tongbi decoction |
Smilax glabra Roxb. (Liliaceae) Dioscorea septemloba Thunb (Dioscoreaceae) Zea mays L. ( Poaceae) Coix lacryma-jobi L.var.mayuen (Roman.)Stapf )Poaceae) Siegesbeckia orientalis L. (Asteraceae) Curcuma longa L . (Zingiberaceae) Taxillus sutchuenensis (Lecomte) Danser (Loranthaceae) Corydalisyanhusuo W.T.Wang (Papaveraceae) CitrusmedicaLvar.sarcodac-tylis Swingle (Rutaceae) |
Male SD rats male C57BL/6 mice |
Antiinflammation Antihyperuricemia Regulating intestinal flora |
NLRP3↓; IL-1β↓; TNF-α↓; ABCG2↑; SCFAs↑ |
[135-137] |
Zisheng-shenqi decoction |
Rehmannia glutinosa (Gaert.) Libosch. (Scrophulariaceae) Cornus officinalis Sieb. (Cornaceae) Poria cocos(Schw.)Wolf (Polyporaceae) Cortex moutan ( Paeoniaceae) Dioscorea polystachya Turczaninow (Dioscoreaceae) Alisma orientalis (Sam.) Juzep. (Alismataceae) Amomum villosum Lour. (Zingiberaceae) |
male Wistar rats |
Antiinflammation Antioxidant stress |
IL-1β↓; TNF-α↓; NF-κB↓ SOD↓; GSH-Px↓ |
[138,139] |
Guizhi-Shaoyao- Zhimu decoction |
Cinnamomum cassia Presl (Lauraceae) Atractylodes macrocephala Koidz. (Asteraceae) Glycyrrhiza uralensis Fisch. (Fabaceae) Ephedra sinica Stapf (Ephedraceae) Zingiber officinale Roscoe (Zingiberaceae) Atractylodes macrocephala Koidz. (Asteraceae) Anemarrhena asphodeloides Bunge (Liliaceae) Saposhnikovia divaricata ( Trucz.) Schischk. (Apiaceae) Aconitum carmichaelii Debx (Ranunculaceae) |
Male C57BL/6J mice |
Antiinflammation Antihyperuricemia |
NLRP3↓; IL-1β↓; IL-6↓; MCP-1↓; IKKβ↓; p65↓ |
[140-142] |
Shuang-Qi gout capsule |
Phragmites australis (Cav.) Trin. ex Steud. (Poaceae) Berchemia floribunda (Wall.) Brongn. (Rhamnaceae) Mallotus apelta (Lour.) Muell. Arg. ( Euphorbiaceae) Schefflera arboricola Hay. (Araliaceae) Cinnamomum camphora (L.) presl (Lauraceae) Panax notoginseng (Burkill) F. H. Chen ( Araliaceae) |
Male ICR mice Male SD rats HUVE cells |
Antiinflammation Antihyperuricemia Promote uric acid excretion |
IL-1β↓; TNF-α↓; mURAT1↓; mABCG2↑ mOAT1↑; mOCT1↑; mOCT2↑ mGLUT9↓ |
[143,144] |
Huzhen-Tongfeng formula |
Polygonum cuspidatum Sieb.et Zucc. (Polygonaceae) Ligustri Lucidi Fructus (Oleaceae) Plantago asiatica L. (Plantaginaceae) Polistes olivaceus (DeGeer) (Vespidae) |
Male SD rats RAW264.7 cells THP-1 cell Male Japanese big ear white rabbits |
Antiinflammation Antioxidant stress |
IL-1β↓; IL-6↓; TNF-α↓; PGES2↓; COX-1↓; COX-2↓; |
[145,146] |
Jiang-Suan-Chu-Bi formula |
Smilax glabra Roxb. (Liliaceae) Paeonia lactiflora Pall. (Paeoniaceae) Polygonum cuspidatum Sieb.et Zucc. (Polygonaceae) Iphigenia indica Kunth (Liliaceae) Rheum officinale Baill. (Polygonaceae) Clematis chinensis Osbeck (Ranunculaceae)Heracleum hemsleyanum Diels ( Apiaceae) Phellodendron chinense Schneid. (Rutaceae) Atractylodes lancea( Thunb.) DC. (Asteraceae) Glycyrrhiza uralensis Fisch. (Fabaceae) Salvia miltiorrhiza Bge. (Labiatae) Angelica sinensis (Oliv.) Diels (Apiaceae) |
THP-1 cells |
Antiinflammation |
NLRP3↓; IL- 1β↓; ACS↓; caspase-1↓ |
[147] |
heat-clearing and diuresis-promoting prescription |
Tulipa gesneriana L. (Liliaceae) Lysimachia christiniae Hance (Primulaceae) Atractylodes Lancea (Thunb.) DC. (Asteraceae) Clematis chinensis Osbeck (Ranunculaceae) Smilax glabra Roxb. ( Liliaceae) Achyranthes bidentata Bl. (Amaranthaceae) Coix lacryma-jobi L.var.mayuen(Roman.)Stapf (Poaceae) Phellodendron chinense Schneid. (Rutaceae) Lycopus lucidus Turcz.vat.hirtus Regel (Labiatae) Anemarrhena asphodeloides Bunge (Liliaceae) Dioscorea polystachya Turczaninow (Dioscoreaceae) Polygonum cuspidatum Sieb.et Zucc. (Polygonaceae) |
Clinical patient |
Antiinflammation Antihyperuricemia |
NLRP3↓; IL1β↓ |
[148] |
Huzhang-Guizhi herb pair |
Polygonum cuspidatum Sieb.et Zucc. (Polygonaceae) CassiaTwig (Lauraceae) |
male SD rats |
Antiinflammation |
IL-1β↓; TNF-α↓ p65↓ |
[149] |
Shaoyao-gancao decoction |
Paeonia lactiflora Pall. (Paeoniaceae)Glycyrrhiza uralensis Fisch. ( Fabaceae) |
Male ICR mice RAW264.7 cells |
Antiinflammation |
IL-1β↓; IL-6↓; TNF-α↓; COX-2↓; PGES2↓ |
[150] |
Tongfengding capsule |
Gentiana macrophμlla Pall. (Gentianaceae) Phellodendron chinense Schneid. (Rutaceae) Corydalis yanhusuo W. T. Wang (Papaveraceae) Paeonia lactiflora Pall. (Paeoniaceae) Cyathula officinalis Kuan (Amaranthaceae) Alisma plantago-aquatica Linn. (Alismataceae) Plantago asiatica L. (Plantaginaceae) Smilax glabra Roxb. (Liliaceae) |
Male SD rats THP-1 cells |
Antiinflammation |
TNF-α↓; IL-6↓; PGES2↓ |
[151] |
Xiaofeng Granules |
Atractylodes lancea( Thunb.)DC. (Asteraceae) Phellodendron chinense Schneid. (Rutaceae) Coix lacryma-jobi L.var.mayuen(Roman.)Stapf (Poaceae) Achyranthes bidentata Bl. (Amaranthaceae) Smilax glabra Roxb. (Liliaceae) Lonicera japonica Thunb. (Caprifoliaceae) |
Male SD rats chondrocyte |
Antiinflammation Inhibit cartilage matrix degradation |
L-1β↓; IL-6↓; TNF-α↓ MMPs/TIMP- 1↓; ADAMTS-4/ TIMP-3↓ |
[152] |
Shenling Baizhu San |
Panax ginseng C.A.Mey. (Araliaceae) Poria cocos (Schw.) Wolf (Polyporaceae) Atractylodes macrocephala Koidz. (Compositae) Dioscorea opposita Thunb. (Dioscoreaceae) Dolichos lablab L. (Leguminosae) Nelumbo nucifera Gaertn. (Nelumbonaceae) Coix lacryma-jobi. var. ma-yuen. (Rom.Caill.) Stapf (Poaceae) Amomum villosum Lour. (Zingiberaceae) Platycodon grandiflorus (Jacq.) A. DC. (Campanulaceae) Glycyrrhiza uralensis Fisch. (Leguminosae) |
male quails |
Antihyperuricemia |
p-PPARγ↓ |
[153] |
Erding Formula |
Viola philippica (Violaceae) Taraxacum mongolicum Hand.-Mazz. (Asteraceae) Lobelia chinensis Lour. (Campanulaceae) Isatis indigotica Fortune (Brassicaceae) |
Male Kunming mice |
Antihyperuricemia Antiinflammation relieve pain |
mOAT3↑; mURAT1↓ |
[154] |
Tongbixiao Pills |
Rehmannia glutinosa (Gaetn.) Libosch. (Scrophulariaceae) Cynanchum otophyllum Schneid. (Asclepiadaceae) Gentiana macrophylla (Gentianaceae) Achyranthes bidentata Blume (Amaranthaceae) Homalomena occulta (Araceae) Cervus nippon Temminck (Cervidae) Carica papaya L. (Caricaceae) Coix lacryma-jobi L.var.mayuen(Roman.)Stapf (Poaceae) Poria cocos(Schw.)Wolf (Polyporaceae) Citrus reticulata Blanco (Rutaceae) Cynanchum atratum Bunge. (Asclepiadaceae) Phellodendron chinense Schneid. (Rutaceae) Atractylodes lancea (Thunb.) DC. (Asteraceae) Lonicera japonica Thunb. (Caprifoliaceae) Tetrapanax papyrifer (Hook.) K. Koch (Araliaceae)Plantago depressa Willd. ( Plantaginaceae) Taraxacum mongolicum Hand.-Mazz. (Asteraceae) Clematis chinensis Osbeck (Ranunculaceae) Aristolochia mollissima Hance (Aristolochiaceae) Caulis Sinomenii (Menispermaceae) Dioscorea septemloba Thunbt (Dioscoreaceae) Areca catechu L. (Arecaceae) |
Male SD rats |
Antihyperuricemia Antiinflammation |
serum creatinine levels↓; serum urea nitrogen levels↓ |
[155] |
TongFengTangSan |
Tinospora sagittata (Oliv.) Gagnep. (Menispermaceae) Terminalia chebula Retz. (Combretaceae) Faeces Trogopterpri |
Male SD rats |
Antihyperuricemia |
XOD↓ |
[156] |
Table 10: Summary of antigout TCM prescriptions
Conclusions and Future Perspectives
The prevalence of gout is increasing year by year worldwide, which severely influences the work and life of patients. Because of the large number of affected people, complicated pathogenesis, and few effective clinical treatment strategies, gout has become a research hotspot. Herein, we present a literature review of gout treatment with TCM in recent years and conclude that TCM exerts an anti-gout effect through various pathways, mainly including reduction in uric acid levels (inhibition of uric acid production and promotion of uric acid excretion), anti-inflammation, and prevention of oxidative stress, among others. Additionally, a few studies have also reported the regulation of apoptosis, bone metabolism, and intestinal flora. The findings reflect the fact that TCM has multiple effects, multiple targets, multiple signaling pathways, and other advantages in gout treatment.
However, at present, TCM exerts the therapeutic effect for gout mainly by regulating the NLRP3/ASC/Caspase, TLRs/ MyD88/NF-κB, and ATP/P2X7R signaling pathways, while reports of regulating the JAK/STAT, MAPK, and Nrf2/HO-1 signaling pathways are rare. Research on the mechanism of the signaling pathways involved in gout treatment is of great significance, and discovering more signaling pathways is conducive to finding more therapeutic targets. Most research methods currently remain at the gene and protein expression levels as well as metabolomics and protein omics; hence, newer technologies need to be explored, to gain an in-depth understanding of the pharmacodynamic material basis and mechanism of action of TCM against gout. Additionally, the syndrome types of animal models used in some studies are single, and the stability of the model is poor.
On the basis of the long-term development of TCM in treating gout, we have put forward the following suggestions. First, we should explore the ancient books of TCM to gain an in-depth understanding of more Chinese herbal medicines or Chinese herbal compounds with the potential to treat gout. Second, because gout treatment requires long-term medication, it is necessary to conduct more toxicology studies to ensure the safety of the medication. Indepth research on the components of Chinese herbal medicine and Chinese herbal compounds is needed to establish quality control standards for Chinese medicine to ensure the effectiveness of the drugs. Finally, combined with the treatment strategies in different stages of gout, the compatibility and dose of Chinese medicine compounds should be appropriately adjusted.
Funding
This work was supported by grants from the Key Program of Natural Science Foundation of State (No. 81830110), Key Research and Development Program of Heilongjiang Province (No. 2022ZX02004), Key research and development project of states key laboratory of dampness syndrome of Chinese medicine (SZ2021ZZ49).
Acknowledgments
All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication. The authors declare no conflict of interest.
References
- Choi HK, Willett W, Curhan G (2010) Fructose-rich beverages and risk of gout in women. JAMA 304:2270-2278.
- Qin N, Qin M, Shi W, Kong L,Wang L, et al. (2022) Investigation of pathogenesis of hyperuricemia based on untargeted and targeted Sci Rep 12:13980.
- Chen JH, Yeh WT, Chuang SY, Wu YY, Pan WH, et al. (2012) Genderspecific risk factors for incident gout: a prospective cohort study. Clin. Rheumatol 31:239-245.
- Kuo MC, Chang SJ, Hsieh MC (2015) Colchicine significantly reduces incident cancer in gout male patients: a 12-year cohort study. Medicine 94:e1570.
- Dalbeth N, Gosling AL, Gaffo A, Abhishek A (2021) Gout. Lancet 397:1843-1855.
- Yang QB, He YL, Zhang QB, Mi QS, Zhou JG (2019) Downregulation of transcription factor T-bet as a protective strategy in monosodium urate-induced gouty inflammation. Front Immunol 10:1199.
- Taylor WJ, Fransen J, Jansen TL, Dalbeth N, Schumacher HR, et al. (2015) Study for updated gout classification criteria: identification of features to classify gout. Arthritis Care Res 67:1304-1315.
- Doherty M, Jenkins W, Richardson H, Sarmanova A, Abhishek A, et (2018) Efficacy and cost-effectiveness of nurse-led care involving education and engagement of patients and a treat-to-target uratelowering strategy versus usual care for gout: a randomised controlled trial. Lancet 392:1403-1412.
- Kuo CF, Grainge MJ, See LC, Yu KH, Luo SF, et al. (2015) Epidemiology and management of gout in Taiwan: a nationwide population study. Arthritis Res Ther 17:13.
- Dehlin M, Jacobsson L, Roddy E (2020) Global epidemiology of gout: prevalence, incidence, treatment patterns and risk factors. Nat Rev Rheumatol 16:380-390.
- Choi HK, Curhan G (2005) Gout: epidemiology and lifestyle choices. Curr Opin Rheumatol 17:341-345.
- Yin C, Liu B, Li Y, Li X, Wang J, et al. (2020) IL-33/ST2 induces neutrophil-dependent reactive oxygen species production and mediates gout pain. Theranostics 10:12189-12203.
- Kojima S, Matsui K, Hiramitsu S, Hisatome I, Waki M, et al. (2019) Febuxostat for cerebral and cardiorenovascular events prevention Eur Heart J 40:1778-1786.
- Wu CH, Gan CH, Li LH, Chang JC, Chen ST, et al. (2020) A synthetic small molecule F240B decreases NLRP3 inflammasome activation by autophagy induction. Front Immunol 11:607564.
- Campion EW, Glynn RJ, Delabry LO (1987) Asymptomatic Risks and consequences in the Normative Aging Study. Am J Chin Med 82:421-426.
- Gaubert M, Bardin T, Cohen-Solal A, Diévart F, Fauvel JP, et al. (2020) Hyperuricemia and hypertension, coronary artery disease, kidney disease: from concept to practice. Int J Mol Sci 21:4066.
- Zhang Y, Tan X, Lin Z, Li F, Yang C, et al. (2021) Fucoidan from laminaria japonica inhibits expression of GLUT9 and URAT1 via PI3K/ Akt, JNK and NF-κB pathways in uric acid-exposed HK-2 cells. Mar Drugs 19:238.
- Ahmad A, Ogungbenro K, Kunze A, Jacobs F, Snoeys J, et al. (2021) Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters. CPT Pharmacometrics Syst Pharmacol 10:467
- Benn CL, Dua P, Gurrell R, Loudon P, Pike A, et al. (2018) Physiology of hyperuricemia and urate-lowering treatments. Front Med 5:160.
- Hsueh CH, Yoshida K, Zhao P, Meyer TW, Zhang L, et al. (2016) Identification and quantitative assessment of uremic solutes as inhibitors of renal organic anion transporters, OAT1 and OAT3. Mol Pharm 13:3130-3140.
- Woodward OM, Köttgen A, Coresh J, Boerwinkle E, Guggino WB, et (2009) Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci 106:1033810342.
- Haider AJ, Cox MH, Jones N, Goode AJ, Bridge KS, et al. (2015) Identification of residues in ABCG2 affecting protein trafficking and drug transport, using co-evolutionary analysis of ABCG sequences. Rep. 35: e00241.
- Oh YJ, Moon KW (2021) Presence of tophi is associated with a rapid decline in the renal function in patients with gout. Sci Rep 11:5684.
- Guma M, Dadpey B, Coras R, Mikuls TR, Hamilton B, et al. (2022) Xanthine oxidase inhibitor urate-lowering therapy titration to target decreases serum free fatty acids in gout and suppresses lipolysis by Arthritis Res Ther 24:175.
- Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J (2006) Goutassociated uric acid crystals activate the NALP3 inflammasome. Nature 440:237-241.
- Hsieh CY, Li LH, Lam Y, Fang Z, Gan CH, et al. (2020) Synthetic 4-hydroxy auxarconjugatin B, a novel Autophagy inducer, attenuates gouty inflammation by inhibiting the NLRP3 inflammasome. Cells 9:279.
- Hwangbo H, Ji SY, Kim MY, Kim SY, Lee H, et al. (2021) Antiinflammatory effect of auranofin on palmitic acid and LPS-Induced inflammatory response by modulating TLR4 and NOX4-mediated NF-κB signaling pathway in RAW264. 7 macrophages. Int J Mol Sci 22:5920.
- Vidya MK, Kumar VG, Sejian V, Bagath M, Krishnan G, et al. (2018) Toll-like receptors: significance, ligands, signaling pathways, and functions in mammals. Int Rev Immunol 37:20-36.
- Cabău G, Crișan TO, Klück V, Popp RA, Joosten LAB (2020) Urateinduced immune programming: Consequences for gouty arthritis and Immunol. Rev. 294:92-105.
- Joosten LAB, Abdollahi-Roodsaz S, Dinarello CA, O’Neill L, Netea MG (2016) Toll-like receptors and chronic inflammation in rheumatic diseases: new developments. Nat Rev Rheumatol 12:344-357.
- Joosten LAB, Netea MG, Mylona E, Koenders MI, Malireddi RKS, et al. (2010) Engagement of fatty acids with toll‐like receptor 2 drives interleukin‐1β production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal–induced gouty arthritis. Arthritis Rheum 62:3237-3248.
- Franchi L, Eigenbrod T, Muñoz-Planillo R, Nuñez G (2009) The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol 10:241-247.
- Dong Z, Zhuang Q, Ning M, Wu S, Lu L, et al. (2020) Palmitic acid stimulates NLRP3 inflammasome activation through TLR4-NF-κB signal pathway in hepatic stellate cells. Ann Transl Med 8:168.
- Hughes MM, O’Neill LAJ (2018) Metabolic regulation of nlrp 3. Immunol Rev 281:88-98.
- Leiguez E, Motta P, Maia Marques R, Lomonte B, Sampaio SV, et al. (2020) A Representative GIIA phospholipase A2 activates preadipocytes to produce inflammatory mediators implicated in obesity Biomolecules 10:1593.
- Chen Z, Cai X, Li M, Yan L, Wu L, et al. (2019) CRISPR/Cas9-based liver-derived reporter cells for screening of mPGES-1 inhibitors. J Enzyme Inhib Med Chem 34:799-807.
- Paik S, Kim JK, Silwal P, Sasakawa C, Jo EK (2021) An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cell Mol Immunol 18:1141-1160.
- Manna S, Howitz WJ, Oldenhuis NJ, Eldredge AC, Shen J, et al. (2018) Immunomodulation of the NLRP3 inflammasome through structurebased activator design and functional regulation via lysosomal rupture. ACS Cent Sci 4:982-995.
- Zhou R, Yazdi AS, Menu P, Tschopp J (2011) A role for mitochondria in NLRP3 inflammasome activation. Nature 469:221-225.
- Harijith A, Ebenezer DL, Natarajan V (2014) Reactive oxygen species at the crossroads of inflammasome and inflammation. Front Physiol 5:352.
- Chentouf M, Guzman C, Hamze M, Gross R, Lajoix AD, et al. (2014) Possible protective effect of membrane lipid rafts against interleukin1β-mediated anti-proliferative effect in INS-1 cells. Plos One 9: e102889.
- Schlesinger N, Thiele RG (2010) The pathogenesis of bone erosions in gouty arthritis. Ann Rheum Dis 69:1907-1912.
- Koonrungsesomboon N, Na-Bangchang K, Karbwang J (2014) Therapeutic potential and pharmacological activities of Atractylodes lancea (Thunb.) DC. Asian Pac J Trop Med 7:421-428.
- Li C, Wang C, Guo YJ, Wen R, Yan LP, et al. (2022) Research on the effect and underlying molecular mechanism of Cangzhu in the treatment of gouty arthritis. Eur. J. Pharmacol. 927:175044.
- Qian X, Jiang Y, Luo Y, Jiang Y (2022) The Anti-hyperuricemia and Antiinflammatory Effects of Atractylodes Macrocephala in Hyperuricemia and Gouty Arthritis Rat Models. Comb Chem High Throughput Screen.
- Wang Y, Lin Z, Zhang B, Jiang Z, Guo F, et al. (2019) Cichorium intybus extract suppresses experimental gout by inhibiting the NF-κB and NLRP3 signaling pathways. Int. J. Mol. Sci. 20:4921.
- Nguyen TD, Thuong PT, Hwang IH, Hoang TKH, Nguyen MK, et al. (2017) Anti-hyperuricemic, anti-inflammatory and analgesic effects of Siegesbeckia orientalis L. Resulting from the fraction with high phenolic content. BMC Complement Altern Med 17:191.
- Li L, Teng M, Liu Y, Qu Y, Zhang Y (2017) Anti-gouty arthritis and antihyperuricemia effects of sunflower (Helianthus annuus) head extract in gouty and hyperuricemia animal models. Biomed Res Int 2017:5852076.
- Wang L, Zhu L, Duan C, Li L, Chen G (2020) Total saponin of Dioscorea collettii attenuates MSU crystal-induced inflammation via inhibiting the activation of the NALP3 inflammasome and caspase-1 in THP-1 macrophages. Mol Med Rep 21:2466-2474.
- Yu D, Wang Y, Yu C, Song M, Zhou Q, et al. (2020) High‐throughput serum metabolomics analysis of gouty arthritis rat treated by total saponins of Rhizoma Dioscoreae Makino by UPLC–Q/TOF–MS. Biomed Chromatogr 34: e4867.
- Lu F, Liu L, Yu D, Li X, Zhou Q, et al. (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 28:280-288.
- Wang S, Fang Y, Yu X, Guo L, Zhang X, et al. (2019) The flavonoidrich 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.
- Liang G, Nie Y, Chang Y, Zeng S, Liang C, et al. (2019) Protective effects of Rhizoma smilacis glabrae extracts on potassium oxonateand monosodium urate-induced hyperuricemia and gout in mice. Phytomedicine 59:152772.
- Xu WA, Yin L, Pan HY, Shi L, Xu L, 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 150:747-754.
- Wu XH, Yu CH, Zhang CF, Anderson S, Zhang YW (2014) Smilax riparia reduces hyperuricemia in mice as a potential treatment of gout. Am J Chin Med 42:257-259.
- Chen L, Yin H, Lan Z, Ma S, Zhang C, et al. (2011) Anti-hyperuricemic and nephroprotective effects of Smilax china L. J Ethnopharmacol 135:399-405.
- Wang CC, Li YL, Chiu PY, Chen C, Chen HC, et al. (2022) Protective effects of corni fructus extract in mice with potassium oxonate–induced J Vet Med Sci 84:1134-1141.
- Kim JK, Kim WJ, Hyun JM, Lee JS, Kwon JG, 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. 83:1335-1341.
- Abdulhafiz F, Mohammed A, Kayat F, Bhaskar M, Hamzah Z, et al. (2020) Xanthine oxidase inhibitory activity, chemical composition, antioxidant properties and GC-MS Analysis of Keladi Candik (Alocasia longiloba Miq). Molecules 25:2658.
- Flemmig J, Kuchta K, Arnhold J, Rauwald HW (2011) Olea europaea leaf (Ph. Eur.) extract as well as several of its isolated phenolics inhibit the gout-related enzyme xanthine oxidase. Phytomedicine 18:561
- Yoon IS, Park DH, Kim JE, Yoo JC, Bae MS, et al. (2017) Identification of the biologically active constituents of Camellia japonica leaf and anti-hyperuricemic effect in vitro and in vivo. Int J Mol Med 39:1613
- Pan J, Zhang C, Shi M, Guo F, Liu J, et al. (2021) Ethanol extract of Liriodendron chinense (Hemsl.) Sarg barks attenuates hyperuricemic nephropathy by inhibiting renal fibrosis and inflammation in mice. J Ethnopharmacol 264:113278.
- Zhang CL, Zhang JJ, Zhu QF, Guan HY, Yang YX, et al. (2021) Antihyperuricemia and antigouty arthritis effects of Persicaria capitata herba in mice. Phytomedicine 93:153765.
- Shi YW, Wang CP, Wang X, Zhang YL, Liu L, et al. (2012) Uricosuric and nephroprotective properties of Ramulus Mori ethanol extract in hyperuricemic mice. J Ethnopharmacol 143:896-904.
- Han S, Wei R, Han D, Zhu J, Luo W, et al. (2020) Hypouricemic effects of extracts from Urtica hyperborea Jacq. ex Wedd. in hyperuricemia mice through XOD, URAT1, and OAT1. Biomed Res Int 2020:2968135.
- Tao H, Zhong J, Mo Y, Liu W, Wang H (2022) Exploring the Mechanism through which Phyllanthus emblica L. Extract Exerts Protective Effects against Acute Gouty Arthritis: A Network Pharmacology Study and Experimental Validation. Evid Based Complement Alternat Med 2022:9748338.
- Li S, Li L, Yan H, Jiang X, Hu W, et al. (2019) Anti-gouty arthritis and anti-hyperuricemia properties of celery seed extracts in rodent models. Mol Med Rep 20:4623-4633.
- Ye X, Wu J, Zhang D, Lan Z, Yang S, et al. (2021) How Aconiti Radix cocta can treat gouty arthritis based on systematic pharmacology and UPLC-QTOF-MS/MS. Front Pharmacol 12:618844.
- Shin WY, Shim DW, Kim MK, Sun X, Koppula S, et al. (2017) Protective effects of Cinnamomum cassia (Lamaceae) against gout and septic responses via attenuation of inflammasome activation in experimental J Ethnopharmacol 205:173-177.
- Chen W, Liu Y, Wei M, Shi L, Wu Y, et al. (2017) Studies on effect of Ginkgo biloba L. leaves in acute gout with hyperuricemia model rats by using UPLC-ESI-Q-TOF/MS metabolomic approach. RSC Adv 7:42964-42972.
- Liu Y, Lai L, Ju Y, Liu C, Meng D (2019) Chemical constituents and synergistic anti-gout studies on Eurycoma longifolia and potential mechanisms evaluation based on systemic analysis approach. Bioorg Chem 92:103302.
- Hua Q, Liu, Y, Liu, C, Liu, L, Meng, D (2019) Revealing synergistic mechanism of multiple components in Stauntonia brachyanthera -Mazz. for gout by virtual screening and system pharmacological approach. Bioorg. Chem. 91:103118.
- Zeng P, Li J, Chen Y, Zhang L (2019) The structures and biological functions of polysaccharides from traditional Chinese herbs. Prog Mol Biol Transl Sci 163:423-444.
- Hou C, Chen L, Yang L, Ji X (2020) An insight into anti-inflammatory effects of natural polysaccharides. Int J Biol Macromol 153:248-255.
- Wong LW, Goh CBS, Tan JBL (2022) A Systemic Review forEthnopharmacological Studies on Isatis indigotica Fortune: Bioactive Compounds and their Therapeutic Insights. Am J Chin Med 50:161207.
- Chen Q, Lan HY, Peng W, Rahman K, Liu QC, et al. (2021) Isatis indigotica: a review of phytochemistry, pharmacological activities and clinical applications. J Pharm Pharmacol 73:1137-1150.
- Ren L, Li Q, Li H, Zhan X, Yang R, et al. (2022) Polysaccharide extract from Isatidis Radix inhibits multiple inflammasomes activation and alleviate gouty arthritis. Phytother Res 36:3295-3312.
- Zhao H, Xu J, Wang R, Tang W, Kong L, et al. (2021) Plantaginis Semen polysaccharides ameliorate renal damage through regulating NLRP3 inflammasome in gouty nephropathy rats. Food Funct. 12:2543-2553.
- Yang Q, Wang Q, Deng W, Sun C, Wei Q, et al. (2019) Antihyperuricemic and anti-gouty arthritis activities of polysaccharide purified from Lonicera japonica in model rats. Int J Biol Macromol 123:801-809.
- Sun Y, Lenon GB, Yang AWH (2019) Phellodendri cortex: a phytochemical, pharmacological, and pharmacokinetic review. Evid Based Complement Alternat Med 2019:7621929.
- Jin Y, Khadka D.B, Cho WJ (2016) Pharmacological effects of berberine and its derivatives: a patent update. Expert Opin Ther Pat 26: 229-243.
- Lin G, Yu Q, Xu L, Huang Z, Mai L, et al. (2021) Berberrubine attenuates potassium oxonate-and hypoxanthine-induced hyperuricemia by regulating urate transporters and JAK2/STAT3 signaling pathway. Eur J Pharmacol 912:174592.
- Xu L, Lin G, Yu Q, Li Q, Mai L, et al. (2021) Anti-hyperuricemic and nephroprotective effects of dihydroberberine in potassium oxonateand hypoxanthine-induced hyperuricemic mice. Front Pharmacol 12:645879.
- Cheng JJ, Ma XD, Ai GX, Yu QX, Chen XY, et al. (2022) Palmatine protects against MSU-induced gouty arthritis via regulating the NF-κB/ NLRP3 and Nrf2 pathways. Drug Des Devel Ther 16:2119-2132.
- Wang Y, Zhu W, Lu D, Zhang C, Wang Y (2021) Tetrahydropalmatine attenuates MSU crystal-induced gouty arthritis by inhibiting ROSmediated NLRP3 inflammasome activation. Int Immunopharmacol 100:108107.
- Wu J, Luo Y, Jiang Q, Li S, Huang W, et al. (2019) Coptisine from Coptis chinensis blocks NLRP3 inflammasome activation by inhibiting caspase-1. Pharmacol Res 147:104348.
- Yin Q, Wang L, Yu H, Chen D, Zhu W, et al. (2021) Pharmacological effects of polyphenol phytochemicals on the JAK-STAT signaling Front Pharmacol 12:716672.
- Ferraz-Filha ZS, Ferrari FC, Araújo MCdPM, Bernardes ACFPF (2017) Effects of the aqueous extract from Tabebuia roseoalba and phenolic acids on hyperuricemia and inflammation. Evid Based Complement Alternat Med 2017:2712108.
- Li X, Jin W, Zhang W, Zheng G (2022) The inhibitory kinetics and mechanism of quercetin-3-O-rhamnoside and chlorogenic acid derived from Smilax china L. EtOAc fraction on xanthine oxidase. Int J Biol Macromol 213:447-455.
- Meng ZQ, Tang ZH, Yan YX, Guo CR, Cao L, et al. (2014) Study on the anti-gout activity of chlorogenic acid: Improvement on hyperuricemia and gouty inflammation. Am J Chin Med 42:1471-1483.
- Du X, Zhao L, Yang Y, Zhang Z, Hu J, et al. (2020) Investigation of the mechanism of action of Porana sinensis Hemsl. against gout arthritis using network pharmacology and experimental validation. J Ethnopharmacol 252:112606.
- Zhou Q, Li X, Wang X, Shi D, Zhang S, et al. (2022) Vanillic acid as a promising xanthine oxidase inhibitor: Extraction from amomum villosum lour and biocompatibility improvement via extract nanoemulsion. Foods 11:968.
- Chen G, Jia P, Yin Z, Kong S, Xiang Z, et al. (2019) Paeonol ameliorates monosodium urate‐induced arthritis in rats through inhibiting nuclear factor‐κB‐mediated proinflammatory cytokine production. Phytother Res 33:2971-2978.
- Wang Q, Yang Q, Cao X, Wei Q, Firempong CK, et al. (2018) Enhanced oral bioavailability and anti-gout activity of [6]-shogaol-loaded solid lipid nanoparticles. Int J Pharm 550:24-34.
- Chiyomaru T, Yamamura S, Zaman M.S, Majid S, Deng G, et al. (2012) Genistein suppresses prostate cancer growth through inhibition of oncogenic microRNA-151. PLoS One 7: e43812.
- Song SH, Park DH, Bae MS, Choi CY, Shim JH, 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.
- Cao Y (2021) Icariin alleviates MSU‐induced rat GA models through NF‐κB/NALP3 pathway. Cell Biochem Funct 39:357-366.
- Lin Y, Liu PG, Liang WQ, Hu YJ, Xu P, et al. (2018) Luteolin-4′-Oglucoside and its aglycone, two major flavones of Gnaphalium affine Don, resist hyperuricemia and acute gouty arthritis activity in animal models. Phytomedicine 41:54-61.
- Jiang Y, Lin Y, Hu YJ, Song XJ, Pan HH, et al. (2017) Caffeoylquinic acid derivatives rich extract from Gnaphalium pensylvanicum willd. Ameliorates hyperuricemia and acute gouty arthritis in animal model. BMC Complement Altern Med 17:320.
- Chang YH, Chiang YF, Chen HY, Huang YJ, Wang KL, et al. (2021) Anti-inflammatory and anti-hyperuricemic effects of chrysin on a high fructose corn syrup-induced hyperuricemia rat model via the amelioration of urate transporters and inhibition of nlrp3 inflammasome signaling pathway. Antioxidants 10:564.
- Feng W, Zhong XQ, Zheng XX, Liu QP, Liu, MY, et al. (2022) Study on the effect and mechanism of quercetin in treating gout arthritis. Int Immunopharmacol 111:109112.
- Wu C, Li F, Zhang X, Xu W, Wang Y, et al. (2022) (−)-Epicatechin Ameliorates Monosodium Urate-Induced Acute Gouty Arthritis Through Inhibiting NLRP3 Inflammasome and the NF-κB Signaling Front Pharmacol 13:799552.
- Lin X, Wang H, An X, Zhang J, Kuang J, et al. (2021) Baeckein E suppressed NLRP3 inflammasome activation through inhibiting both the priming and assembly procedure: Implications for gout therapy. Phytomedicine 84:153521.
- Chang WC, Chu MT, Hsu CY, Wu YJJ, Lee JY, et al. (2019) Rhein, an anthraquinone drug, suppresses the NLRP3 inflammasome and macrophage activation in urate crystal-induced gouty inflammation. Am J Chin Med 47:135-151.
- Luo J, Chen S, Wang L, Zhao X, Piao C (2022) Pharmacological effects of polydatin in the treatment of metabolic diseases: a review. Phytomedicine 102:154161.
- Chen L, Lan Z (2017) Polydatin attenuates potassium oxonateinduced hyperuricemia and kidney inflammation by inhibiting NF-κB/ NLRP3 inflammasome activation via the AMPK/SIRT1 pathway. Food 8:1785-1792.
- Villanueva DS, Poirier P, Standley PR, Broderick TL (2003) Prevention of ischemic heart failure by exercise in spontaneously diabetic BB Wor rats subjected to insulin withdrawal. Metabolism 52:791-797.
- Wu XH, Wang CZ, Zhang J, Wang SQ, Han L, et al. (2014) Effects of smilaxchinoside A and smilaxchinoside C, two steroidal glycosides from Smilax riparia, on hyperuricemia in a mouse model. Phytother Res 28:1822-1828.
- Wu XH, Zhang J, Wang SQ, Yang VC, Anderson S, et al. (2014) Riparoside B and timosaponin J, two steroidal glycosides from Smilax riparia, resist to hyperuricemia based on URAT1 in hyperuricemic Phytomedicine 21:1196-1201.
- Couillaud J, Leydet L, Duquesne K, Iacazio G (2021) The terpene mini-path, a new promising alternative for terpenoids bio-production. Genes 12:1974.
- Lo CW, Lii CK, Hong JJ, Chuang WT, Yang YC, et al. (2021) Andrographolide inhibits IL-1β release in bone marrow-derived macrophages and monocyte infiltration in mouse knee joints induced by monosodium urate. Toxicol Appl Pharmacol 410:115341.
- Xue Y, Li R, Fang P, Ye Z, Zhao Y, et al. (2021) NLRP3 inflammasome inhibitor cucurbitacin B suppresses gout arthritis in mice. J Mol Endocrinol 67:27-40.
- Yue H, Yang Z, Ou Y, Liang S, Deng W, et al. (2021) Tanshinones inhibit NLRP3 inflammasome activation by alleviating mitochondrial damage to protect against septic and gouty inflammation. Int Immunopharmacol 97:107819.
- Fattori V, Zarpelon AC, Staurengo-Ferrari L, Borghi SM, Zaninelli TH, et al. (2018) Budlein A, a sesquiterpene lactone from viguiera robusta, alleviates pain and inflammation in a model of acute gout arthritis in Front Pharmacol 9:1076.
- Yan CY, Ouyang SH, Wang X, Wu YP, Sun WY, et al. (2021) Celastrol ameliorates Propionibacterium acnes/LPS-induced liver damage and MSU-induced gouty arthritis via inhibiting K63 deubiquitination of Phytomedicine 80:153398.
- He M, Hu C, Chen M, Gao Q, Li L, et al. (2022) Effects of Gentiopicroside on activation of NLRP3 inflammasome in acute gouty arthritis mice induced by MSU. J Nat Med 76:178-187.
- Han Q, Bing W, Di Y, Hua L, Shi-He L, et al. (2016) Kinsenoside screening with a microfluidic chip attenuates gouty arthritis through inactivating NF-κB signaling in macrophages and protecting endothelial Cell Death Dis. 7: e2350.
- Meng Q, Meng W, Bian H, Zheng F, Gu H, et al. (2021) Total glucosides of paeony protects THP-1 macrophages against monosodium urateinduced inflammation via MALAT1/miR-876-5p/NLRP3 signaling cascade in gouty arthritis. Biomed Pharmacother 138:111413.
- Chen G, Li KK, Fung CH, Liu CL, Wong HL, et al. (2014) Er-MiaoSan, a traditional herbal formula containing Rhizoma Atractylodis and Cortex Phellodendri inhibits inflammatory mediators in LPS-stimulated 7 macrophages through inhibition of NF-κB pathway and MAPKs activation. J Ethnopharmacol 154:711-718.
- Liu Z, Wang B (2018) Predict “multi-efficacy of one formula”molecular mechanism of ermiaowan based on network pharmacology. Chin J Exp Tradit Med Formulae 24:198-206.
- Huang B, Hu X, Wang J, Li P, Chen J (2019) 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 1118-1119:101-108.
- Fu XL, Zhou J, Tang WW, Liu Y, Li ZL, et al. (2021) Study on the compatibility effect and active constituents of atractylodis rhizoma in ermiao wan against acute gouty arthritis. J. Ethnopharmacol. 279:114353.
- Liu CS, Hu YN, Luo ZY, Xia T, Tan XM (2021) Comparative analysis of pharmacokinetics and intestinal absorption of six alkaloids between Sanmiao Pills and Simiao Pills in rats. China J Chin Materia Medica 46:6271-6277.
- Zhu F, Yin L, Ji L, Yang F, Zhang G, 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 Immunopharmacol 30:36-42.
- Lee YM, Son E, Kim DS (2021) Comparative study of anti-gouty arthritis effects of sam-myo-whan according to extraction solvents. Plants 10:278.
- Zhang Y, Chen J, Ye B, Hao Z, Dai H (2022) Simiaowan up-regulates intestinal ABCG2 expression to promote intestinal uric acid excretion in hyperuricemia rats. Chin. J. Exp. Tradit. Med. Formulae 28:33-39.
- Cao L, Zhao T, Xue Y, Xue L, Chen Y, et al. (2022) The anti-inflammatory and uric acid lowering effects of si-miao-san on gout. Front Immunol 12:777522.
- Lin X, Shao T, Huang L, Wen X, Wang M, et al. (2020) Simiao decoction alleviates gouty arthritis by modulating proinflammatory cytokines and the gut ecosystem. Front Pharmacol 11:955.
- Fan Y, Liu W, Jin Y, Hou X, Zhang X, et al. (2021) Integrated molecular docking with network pharmacology to reveal the molecular mechanism of simiao powder in the treatment of acute gouty arthritis. Evid Based Complement Alternat Med 2021:5570968.
- Xu H, Wu J, Wang S, Xu L, Liu P, et al. (2022) Network pharmacology and in vivo experiments reveal the pharmacological effects and molecular mechanisms of Simiao Powder in prevention and treatment for gout. BMC Complementary Med Ther 22:152.
- Hu QH, Jiao RQ, Wang X, Lv YZ, Kong LD (2010) Simiao pill ameliorates urate underexcretion and renal dysfunction in hyperuricemic mice. J Ethnopharmacol 128:685-692.
- Shi L, Yuan Z, Liu J, Cai R, Hasnat M, et al. (2021) Modified Simiaowan prevents articular cartilage injury in experimental gouty arthritis by negative regulation of STAT3 pathway. J Ethnopharmacol 270:113825.
- Liu YF, Tu SH, Chen Z, Wang Y, Hu YH, et al. (2014) Effects of modified Simiao decoction on IL-1β and TNFα secretion in monocytic THP-1 cells with monosodium urate crystals-induced inflammation. Evid Based Complement Alternat Med 2014:406816.
- Zhao F, Guochun L, Yang Y, Shi L, Xu L, et al. (2015) A network pharmacology approach to determine active ingredients and rationality of herb combinations of Modified-Simiaowan for treatment of gout. J. 168:1-16.
- Lv H, Chen J, Liu F, Jin Y, Xu Z, et al. (2019) A traditional clinic Chinese medicine prescription qu-zhuo-tong-Bi (QZTB) alleviates gouty arthritis in model rats. Evid Based Complement Alternat Med 2019:9456318.
- Chen J, Zhou J, Wei S, Xie Z, Wen C, et al. (2016) 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.
- Wen X, Lou Y, Song S, He Z, Chen J, et al. (2021) Qu-Zhuo-Tong-Bi Decoction alleviates gouty arthritis by regulating butyrate-producing bacteria in mice. Front Pharmacol 11:610556.
- Li WH, Han JR, Ren PP, Xie Y, Jiang DY (2021) Exploration of the mechanism of Zisheng Shenqi decoction against gout arthritis using network pharmacology. Comput Biol Chem 90:107358.
- Han J, Xie Y, Sui F, Liu C, Du X, 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 14:2589-2597.
- Zhang Q, Li R, Liu J, Peng W, Fan W, et al. (2020) Efficacy and tolerability of Guizhi-Shaoyao-Zhimu decoction in gout patients: a systematic review and Meta-analysis. Pharm. Biol. 58: 1023-1034.
- Zhou GQ, Chen G, Yang J, Qin W, Ping J (2022) Guizhi-ShaoyaoZhimu decoction attenuates monosodium urate crystal-induced inflammation through inactivation of NF-κB and NLRP3 inflammasome. J Ethnopharmacol 283:114707.
- Yang Q, Zhang J, Li J (2022) Clinical Effect of the Guizhi Shaoyao Zhimu Decoction in the Treatment of Hyperuricemia. Biomed Res Int 2022:5186210.
- Kodithuwakku ND, Pan M, Zhu Y, Zhang Y, Feng Y, et al. (2013) Antiinflammatory and antinociceptive effects of Chinese medicine SQ gout capsules and its modulation of pro-inflammatory cytokines focusing on gout arthritis. J Ethnopharmacol 150:1071-1079.
- Kodithuwakku ND, Feng Y, Zhang Y, Pan M, Fang W, et al. (2015) The molecular insight into the antihyperuricemic and renoprotective effect of Shuang Qi gout capsule in mice. J. Ethnopharmacol. 163:278-289.
- Deng J, Wu Z, Chen C, Zhao Z, Li Y, et al. (2020) Chinese Medicine Huzhen tongfeng formula effectively attenuates gouty arthritis by inhibiting arachidonic acid metabolism and inflammatory mediators. Mediators Inflamm 2020:6950206.
- Wu Z, Xue Q, Zhao Z, Zhou P, Zhou Q, et al. (2019) Suppressive effect of huzhentongfeng on experimental gouty arthritis: an in vivo and in vitro study. Evid Based Complement Alternat Med 2019:2969364.
- Xiao N, Qu J, He S, Huang P, Qiao Y, et al. (2020) Exploring the Therapeutic Composition and Mechanism of Jiang-Suan-Chu-Bi Recipe on Gouty Arthritis Using an Integrated Approach Based on Chemical Profile, Network Pharmacology and Experimental Support Using Molecular Cell Biology. Front. Pharmacol. 10:1626.
- Yu J, Li L, Liu J, Chen Z (2022) Influence of intervention treatment by “heat-clearing and diuresis-promoting” prescription on NALP3, an inflammatory factor in acute gouty arthritis. J Orthop Surg Res 17:162.
- Han B, Huang H, Li Z, Gong M, Shi W, et al. (2016) Therapeutic effects of chinese medicine herb pair, huzhang and guizhi, on monosodium urate crystal-induced gouty arthritis in rats revealed by anti-inflammatory assessments and NMR-based metabonomics. Evid Based Complement Alternat Med 2016:9398435.
- Lee YM, Kim DS (2020) The extraction solvent influences the antiinflammatory effects of jakyakgamcho-tang in lipopolysaccharidestimulated macrophages and mice with gouty arthritis. Int J Mol Sci 21:9748.
- Yang W, Jiang X, Liu J, Qi D, Luo Z, et al. (2021) Integrated strategy from in vitro, in situ, in vivo to in silico for predicting active constituents and exploring molecular mechanisms of tongfengding capsule for treating gout by inhibiting inflammatory responses. Front Pharmacol 12:759157.
- Shi L, Zhao F, Zhu F, Liang Y, Yang F, et al. (2016) 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.
- Wang Y, Lin Z, Huang J, Chu M, Ding X, et al. (2022) An integrated study of Shenling Baizhu San against hyperuricemia: Efficacy evaluation, core target identification and active component discovery. J Ethnopharmacol 295:115450.
- Zuo J, He H, Zuo Z, Bou-Chacra N, Löbenberg R (2018) Erding Formula in hyperuricaemia treatment: unfolding traditional Chinese herbal compatibility using modern pharmaceutical approaches. J Pharm Pharmacol 70:124-132.
- Xi S, ZhiguoShao, Li L, Gui Z, Liu P, et al. (2022) Tongbixiao pills improve gout by reducing uric acid levels and inhibiting inflammation. Dose-Response 20:15593258221090340.
- Chen H, Zhang C, Yao Y, Li J, Du W, et al. (2019) Study on antihyperuricemia effects and active ingredients of traditional Tibetan medicine TongFengTangSan (TFTS) by ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass J Pharm Biomed Anal 165:213-223.
© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. Read More About Open Access Policy.