International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disorder primarily characterized by symmetrical, multi-joint pain and swelling, often accompanied by local joint destruction. The underlying pathogenesis remains incompletely understood, but it involves a complex interplay of genetic, environmental, and immune factors, ultimately leading to the breakdown of immune tolerance and systemic immune dysregulation (1). RA markedly impairs the quality of life of patients. Over the past three decades, its global prevalence has steadily increased, with the 2020 RA Global Burden of Disease Study reporting an estimated prevalence of ~0.2%, placing a substantial burden on the global economy (2).
Despite significant advances in the treatment of RA, outcomes remain unsatisfactory. Due to its complex pathogenesis, no drug or therapy can fully halt RA progression, and most treatments are associated with notable side effects (3). Epidemiological and translational studies have increasingly emphasized the critical role of mucosal interactions with symbiotic bacteria in RA development (4–6). Research on high-risk RA populations has revealed lung mucosal inflammation, as well as the production of local anti-citrullinated protein antibodies (ACPAs) and rheumatoid factor, supporting the ‘mucosal origin hypothesis’ (7,8). Further investigations have identified distinct mucosal mechanisms, including those of the intestinal mucosa, that drive RA progression (9). The intestine, home to the largest concentration of innate and adaptive immune cells, forms a vital interface between the internal environment and external factors, linking environmental influences to systemic immunity (10,11). Alterations in the gut microbiota, which may respond to external environmental and changes or be driven by systemic inflammation can occur before the onset of arthritis. Emerging research increasingly highlights the critical role of the ‘gut-joint axis’ in the transition from preclinical to clinical RA.
The relationship between intestinal microecological imbalance and RA involves complex physiological and pathological mechanisms. Studies from RA animal models and preclinical RA (PreRA) populations indicate that gut alterations may precede the onset of the disease and potentially serve as latent triggers of systemic inflammation (12–16). Conversely, a reverse ‘gut-joint axis’ mechanism may also exist, although further research is required to substantiate this hypothesis. For example, increasing levels of inflammatory factors in the joints may spread peripherally, contributing to or exacerbating damage to the intestinal mucosal barrier and microbiota dysbiosis (17,18). Additionally, pain may alter the metabolic products of gut microbiota and intestinal hormones (19,20). The present review summarized the ‘gut-joint axis’ mechanism in RA, highlighting changes in gut microbiota, interactions between microbiota and genetic risks, alterations in intestinal permeability, dysregulated mucosal immune responses, molecular mimicry, disruption of microbial-derived metabolites, gut hormone regulation and sex bias. These factors, alone or in combination, contribute to the onset and progression of RA (Fig. 1).
Natural compounds, biologically active substances derived from natural sources, have garnered significant attention due to their diverse biological effects, including antioxidant, anti-inflammatory, anti-fibrotic, anti-tumor and anti-angiogenesis properties. Given the limitations of current RA treatments, the present review explored several natural compounds that exert anti-RA effects through the ‘gut-joint axis’ mechanism, offering new insights and potential strategies for innovative RA therapies (Fig. 2).
The present review conducted a systematic literature search across PubMed (https://pubmed.ncbi.nlm.nih.gov; version: Baseline 2025 released December 2024), Web of Science (https://www.webofscience.com; version: 2025–09 weekly update), SpringerLink (https://link.springer.com; version: Content release 2025.09), ScienceDirect (https://www.sciencedirect.com; version: 2025 issue 10), ACS Publications (https://pubs.acs.org; version: 2025 volume 147), Wiley Online Library (https://onlinelibrary.wiley.com; version: 2025 volume releases) and CNKI (https://www.cnki.net; version: CNKI 2025 Master Release) for studies published between January 1995 and September 2025. The following keywords were used: ‘Rheumatoid Arthritis’, ‘Gut’, ‘Natural compounds’, ‘Alkaloids’, ‘Polyphenols’, ‘Polysaccharides’, ‘Glycosides’, ‘Flavonoids’, ‘Terpenoids’, ‘CD4+T cells’ and ‘Hormone’. Pending clinical trials were retrieved from ClinicalTrials.gov and the Center for Drug Evaluation (http://www.cde.org.cn; version: 2025 Master Release), National Medical Products Administration (https://www.nmpa.gov.cn; version: 2025 Master Release). Studies included were clinical studies, laboratory studies (including animal studies or in vitro cellular studies, or combined animal and in vitro studies), meta-analyses and bioinformatics studies, irrespective of language. Exclusion criteria were: i) Duplicate publications; ii) clinical trials lacking relevant data; iii) newspapers, conference proceedings, commentaries, and non-experimental information; iv) low-quality studies or those making erroneous conclusions; and v) retention of multiple reports on the same topic for comparison and discussion.
The gut microbiota plays a pivotal role in regulating host immunity and influencing the progression of RA through various mechanisms (21,22). Several recurring patterns have been identified in the intestinal microbiota of patients with RA, including an increase in the abundance of Prevotella and Lactobacillus and a decrease in Faecalibacterium and Bacteroidetes (14,23–27). Among these, the pathogenic role of Prevotella in RA has been extensively studied. Prevotella is closely associated with the RA risk gene human leukocyte antigen (HLA)-DRB1 and is involved in the disease's adaptive immune response (12). Notably, antibodies against Prevotella copri (P. copri) are specific to patients with RA, with antibody levels to other symbiotic bacteria in patients with RA are similar or lower compared with those in other patients with arthritis or healthy controls (28,29). Moreover, when P. copri enters the systemic circulation, it can selectively accumulate in the joints, potentially inducing synovial hyperplasia and deformation by promoting synovial tissue growth (30). P. copri is one of several intestinal microbiota species with pathogenic effects on RA and their mechanisms have been comprehensively reviewed (31,32).
Genome-wide association studies have identified several RA susceptibility alleles, including HLA-DRB1, PTPN22, and PADI4 genes (33,34). Alleles within the class II loci of HLA, particularly those in DRB1, show a strong association with RA risk in individuals positive for autoantibodies, with >70% of ACPA-positive patients with RA carrying the HLA-DRB1 allele (35). Environmental factors may serve as significant triggers for individuals with a genetic predisposition to develop RA. Notable microbiome changes have been observed in the intestines of healthy individuals carrying the RA risk allele HLA-DRB1, including an increase in Lachnospiraceae, Clostridiaceae and Bifidobacterium longum species (36). Wells et al (12) classified Prevotella species based on predicted types and assigned numerical names according to the SILVA database. The authors cohort study found that, even in the absence of clinically detectable disease, the genetic risk of RA was associated with specific gut microbiota. The Prevotella_7 strain (whose predicted type remains uncertain) was particularly strongly linked to the RA genotype.
The intestine plays a critical role as a conduit between the body's internal environment and external factors. When the intestinal mucosal barrier is compromised, non-self antigens can penetrate and invade the body, triggering and exacerbating systemic inflammatory responses. This can lead to the breakdown of self-immune tolerance and the disruption of immune homeostasis (37). Additionally, increased intestinal permeability allows immune cells to migrate beyond the intestine, potentially causing extraintestinal inflammation (38,39). Clinical studies have shown that intestinal permeability is elevated in both preclinical and established patients with RA (24,39–42). Individuals with high zonulin levels (>10 ng/ml) during the early stages of RA are at greater risk of developing the disease within a year (39). Furthermore, persistent arthritis may further compromise intestinal permeability, facilitating the invasion of joint synovial fluid by intestinal microorganisms in the IV stage of RA (17). In vitro studies have also indicated that intestinal bacteria from PreRA subjects directly modulate the expression of zonula occludens-1 in Caco-2 cells at both the transcriptional and protein levels (5). Collinsella, a genus within the Actinobacteria phylum, is markedly enriched in the gut microbiota of patients with RA. Its overgrowth disrupts intestinal barrier function and is independently associated with disease-related inflammatory activity (14,43). In collagen-induced arthritis (CIA) mice, serum zonulin levels and intestinal permeability are elevated prior to arthritis onset, with this barrier dysfunction being microbiota-driven. Transfer of CIA-derived gut microbiota to germ-free (GF) recipients results in increased gut permeability. Moreover, pre-arthritic administration of a zonulin antagonist selectively restores epithelial integrity and reduces subsequent joint inflammation (39). Hypoxia-inducible factor (HIF) plays a pivotal role in regulating tight junction proteins such as claudin-1, thereby orchestrating adaptive barrier responses in the mucosa (44,45). HIF-2α, specifically expressed in the mucosal epithelium, is essential for maintaining gut barrier integrity and regulating intestinal iron absorption. Certain bacterial metabolites, such as 1,3-diaminopropane, reuterin, and short-chain fatty acids (SCFAs), suppress HIF-2α, influencing intestinal barrier function (46). Wen et al (42) demonstrated that inhibiting intestinal HIF-2α preserves epithelial barrier integrity by selectively downregulating claudin-15, thereby reducing arthritis severity. Consequently, restoring intestinal tight junctions may be a key intervention in the prevention and treatment of RA.
Studies suggest that the initiation of autoimmune responses in RA begins in gut-associated lymphoid tissue (GALT), distant from the joints, involving both innate and adaptive immune responses (5,39,47,48). Increased intestinal permeability facilitates the transport of intestinal immune cells to the joints. Takahashi et al (49) performed cell transport analysis using KikGR mice and observed that both follicular helper T (Tfh) cells and B cells migrated from the colonic patch to the draining lymph nodes following immunization. In the CIA model, it has been confirmed the distal GALT is a primary site for initiating the autoimmune response. The intestinal lamina propria is recognized as the main source of T helper 17 (Th17) cells, and during the immune initiation phase in CIA mice, an increase in Th17 cell content in intestinal lamina propria correlates strongly with arthritis severity (47,50,51). In transgenic K/BxN arthritis mice, a population of α4β7-expressing Th17 cells was found in the spleen, where they assist in the generation of autoantibodies against glucose-6-phosphate isomerase (52). Another potential source of α4β7+ Th17 cells in the spleen is the migration of intestinal CD103+ dendritic cells (DCs) to the spleen. Marietta et al (38) demonstrated that CD103+ DCs, located in the intestinal lamina propria, migrate to the spleen in CIA mice, suggesting that these DCs may have originated from the intestine.
Molecular mimicry, a process in which microbial antigens share structural similarities with autoantigens, can lead to the cross-activation of autoreactive T or B cells, thereby triggering autoimmunity. Certain gut microbiota have been implicated in causing autoimmune responses and systemic inflammation through molecular mimicry, resulting in joint tissue damage (53). For instance, Escherichia coli heat-shock protein (DnaJ) shares a molecular mimicry mechanism with the HLA-DRB1*0401 molecule, as both exhibit the amino acid sequence QKRAA (54). Preclinical RA studies have identified potential epitopes in Citrobacter, Clostridium, Bacteroides and Eggerthella that resemble collagen XI and HLA-DRB1*0401 epitopes (12,13,55,56). Additionally, Bacteroides fragilis 3_1_12, a strain from the Bacteroidaceae family, may mimic the collagen II peptide (57–59). Similarly, Prevotella epitopes show high sequence homology with T-cell epitopes of N-acetylglucosamine-6-sulfonamide and serpin A, presented by HLA-DR (60–62). These microorganisms may trigger autoimmune responses and antibody production via molecular mimicry, thereby accelerating RA progression. Thus, molecular mimicry may represent a key mechanism by which the gut microbiota influences the pathogenesis and development of RA.
The disruption of metabolites derived from the intestinal microbiota plays a pivotal role in the ‘gut-joint axis’ of RA. Preclinical high-risk populations have already shown intestinal microbiota dysbiosis and alterations in metabolomics (5). Among these metabolites, SCFAs have been studied most extensively studied and are considered key immunomodulatory compounds closely associated with RA (63). SCFAs primarily exert their effects on the immune system by modulating histone acetyltransferases and deacetylases (64). In patients with RA, fecal levels of acetate, propionate, butyrate, and valerate are markedly reduced (49,63,65), with a characteristic deficiency of butyrate producers and an enrichment of butyrate consumers in the intestinal microbiota of these individuals (66,67). The protective role of butyrate in RA has been widely investigated, particularly its effects on balancing Th17 and T regulatory (Treg) cells in the intestine system, as well as its regulation of B cells and autoantibody production (63,67,68). Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis has revealed alterations in amino acid and lipid metabolism in PreRA and established patients with RA (5,69). A retrospective clinical cohort study in Sweden found that patients with RA and positive ACPA exhibited elevated levels of lysophospholipid and tryptophan metabolism several years before diagnosis, compared with undiagnosed individuals with RA (70). Following this, Seymour et al (71) demonstrated in animal studies that indole, a tryptophan metabolite produced by bacteria such as P. copri, Collinsella and Ruminococcus, plays a pivotal role in the development of CIA by enhancing Th17 immunity both locally in the gut and systemically. Bile acids (BAs) are vital for immune regulation, particularly in balancing Th17 and Treg cell populations (72–75), and contribute to the maintenance of fat metabolism. However, high concentrations of BAs can have cytotoxic effects on colonic epithelial cells (76). While studies on BA levels in RA animal models remain inconclusive (77–79), accumulating evidence suggests that BAs play a protective and preventative role in RA (18,80).
The intestine harbors a significant number of endocrine cells that secrete various intestinal hormones. Hormones such as vasoactive intestinal peptide (VIP), somatostatin (SOM), substance P (SP) and gastrin-releasing peptide are implicated in the progression of RA (81–83). VIP can promote the Th1/Th2 balance and enhance the differentiation of Treg cells and follicular regulatory T (Tfr) cells, thereby reducing joint inflammation in RA animal models (84–86). SP has been reported to stimulate the proliferation of synovial cells (87,88) and serum SP levels are considered indicative of disease activity and subclinical inflammation in patients with RA (89). However, other studies suggest that SP treatment can reduce inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-17 (IL-17), while increasing the expression of the anti-inflammatory cytokine IL-10, leading to significant improvements in RA symptoms (90,91). Consequently, the exact mechanism by which SP influences RA remains to be further elucidated.
Significant sex differences exist in RA, with a prevalence rate in females that is notably higher than in males, often at a ratio of 4:1 or more (92,93). The influence of sex hormones on RA has been extensively studied. A sudden drop in estrogen levels and a decrease in androgen can both increase RA risk (94). Sex hormone deficiency enhances intestinal permeability, expands the Th17 cell pool and upregulates osteoclastogenic factors, such as TNF-α, RANKL and IL-17, in the bone marrow, with the gut microbiota playing a central role in this process (95). However, butyrate supplementation has been shown to prevent estrogen-deficiency-induced bone loss (96) and alleviate arthritis by activating the estrogen-related receptor α on Th17 cells, modulating IL-17 and IL-10 production (68). Sex-specific differences in the gut microbiota have been well established (97). Compared to women, men have markedly lower levels of Prevotella (36) and P. copri antibodies, which are considered specific to RA, exhibit a sex disparity, with a male-to-female ratio of 1:9. This difference may markedly contribute to the female predominance observed in RA (28). Additionally, Eggerthella lenta (E. lenta), a potential pathogenic bacterium in RA, is found in higher levels in female patients with RA (98). Animal studies have shown that female mice treated with E. lenta display increased intestinal permeability, elevated IgG-RF levels, an expansion of IL-17- and IFN-γ-producing B cells, and higher concentrations of BAs and succinyl carnitine, indicating a sex-specific immune response to E. lenta (98). Despite some evidence, the role of sex-dependent differences in the human microbiota in RA remains poorly understood. Preclinical and clinical data are both still limited, and more comprehensive evidence is needed to support or challenge these findings.
Natural compounds exert a positive effect on human health by regulating various mechanisms, offering potential therapeutic benefits for a range of diseases. Several natural compounds, including sinomenine and Tripterygium wilfordii are widely used in the clinical treatment of RA in China. Clinical studies exploring the use of natural compounds for RA treatment have entered the initial exploration stage (Table I). However, these studies face challenges such as low quality, weak evidence and small sample size, necessitating high-quality clinical trials to improved support the therapeutic effects of these compounds on RA. Basic research has consistently confirmed the protective effects of natural compounds on RA, elucidating the underlying mechanisms. For most high-molecular-weight, low-bioavailability natural compounds, which accumulate in large quantities in the gastrointestinal tract after oral administration, the intestine appears to be a key target for their action in treating RA. This section focuses on natural compounds that treat or enhance RA therapy through the ‘gut-joint axis’ (Table II).
Alkaloids, primarily found in plants, are key components of numerous Chinese herbal medicines. Higher plants from the Liliaceae, Berberidaceae, Ranunculaceae, Leguminosae, Amaryllidaceae, Papaveraceae and Solanaceae families are rich in alkaloids. These compounds are characterized by nitrogen atoms in their chemical structure, which is a defining feature and their biological efficacy is attributed to the specific arrangement of atoms in their molecular structure (99). Alkaloids exhibit significant biological activities, including antibacterial, antiviral, anti-inflammatory, and anti-tumor effects, suggesting their potential medicinal value in the RA treatment (100–102).
Berberine, an isoquinoline alkaloid extracted from various medicinal plants such as Coptis, Phellodendron and Berberis, possesses a broad range of pharmacological effects, including anti-tumor, anti-oxidative, anti-inflammatory, hypoglycemic and lipid-lowering properties. Clinical studies have reported its therapeutic efficacy in treating conditions such as cancer, digestive system disorders, and metabolic diseases (103–107). Due to its ability to inhibit toxins and bacteria and protect the intestinal mucosal barrier, berberine has shown significant advantages in the treatment of digestive disorders (108). A large portion of orally administered berberine remains in the intestine, with an extremely low bioavailability of only 0.1% (109). Oral administration of 200 mg/kg berberine markedly improved CIA in rats, although the peak drug concentration in the blood was <0.06 µM (110), which is much lower than the in vitro minimum effective concentration of 1–50 µM (109,111). Additionally, intravenous berberine did not improve arthritis in rats, suggesting that its anti-arthritis effects are intestinally dependent (110). Further mechanistic studies revealed that oral berberine selectively increased cortistatin levels, a neuropeptide derived from the intestine, both in the intestines and serum of arthritic rats. It also promoted cortistatin expression in intestinal nerve and endocrine cells. The upregulated cortistatin then entered systemic circulation, where it suppressed the immune response of Th17 cells, alleviating arthritis symptoms (110). Moreover, berberine markedly increased the levels of intestinal SCFAs, particularly butyrate, enhanced the abundance of butyrate-producing bacteria and stimulated the expression and activity of butyryl-CoA: Acetate CoA transferase (BUT) (112). When BUT inhibitors or broad-spectrum antibiotics were applied, the regulatory effect of berberine on the intestinal internal environment and its anti-arthritis effects were markedly reduced, indicating that berberine promotes butyrate production through the intestinal microbiota, positioning it as a potential therapeutic agent for RA (112). This effect is highly dependent on the intestinal microbiota (112).
Sinomenine is an alkaloid extracted from the roots and stems of Sinomenium acutum and has demonstrated immunosuppressive and anti-arthritis effects (113). In China, sinomenine is widely used in clinical RA treatment and has been shown to improve arthritis symptoms and reduce inflammatory markers in patients (114–116). A study utilizing multicriteria decision analysis to evaluate the benefits and risks of sinomenine preparations in RA treatment found that combining sinomenine with conventional drugs yielded greater benefits than using it alone (117). However, animal studies have shown that oral administration of sinomenine does not achieve the minimum effective concentration in the synovium or serum (≥250 µM) required for in vitro efficacy, indicating that its precise anti-arthritis mechanism remains to be fully elucidated (118–120). Sinomenine may exert anti-inflammatory effects by inhibiting macrophage activation via the α 7-nicotinic acetylcholine receptor (α7nAChR) (121–123). Subsequent studies exploring the anti-arthritis mechanism of sinomenine through the neuroendocrine-inflammation axis revealed that oral sinomenine treatment improved systemic inflammation in CIA rats. It was found to interact with the residues Tyr184 and Tyr191, binding to α7nAChR and activating the α7nAChR-PI3K/Akt/mTOR pathway, which selectively promotes the generation of VIP in intestinal neuron-like cells of CIA rats (124). Research by Tong et al (120) further confirmed that the protective effect of sinomenine in CIA is intestine-dependent. Oral administration of sinomenine reduced the arthritis score in CIA rats, decreased serum IL-17A levels and increased IL-10 levels. Additionally, sinomenine increased the frequency of Treg cells in intestinal lymphoid tissue, decreased Th17 cell frequency and markedly elevated α4β7-positive cells in the joints. These findings suggest that sinomenine regulates immune cell migration within the gut-joint axis, facilitating lymphocyte migration from the intestine to the joint, thereby alleviating RA symptom. Notably, when administered intraperitoneally, sinomenine loses its therapeutic effect on arthritis.
Matrine, a natural bioactive compound extracted from the roots of the leguminous plant Sophora flavescens, is one of its main pharmacologically active components. It possesses a broad range of pharmacological and biological effects, including antiviral, anti-tumor, antioxidant and antibacterial properties, with the added advantage of low toxicity and minimal side effects (125). A number of studies have demonstrated that matrine can effectively alleviate arthritis symptoms in RA model animal by regulating immune responses, inhibiting synovial vessel formation, and suppressing fibroblast-such as synovial cell proliferation, among other mechanisms (126–128). Oxymatrine, a derivative of matrine with a distinct oxygen structure, exhibits similar pharmacological effects. Animal studies have shown that oxymatrine also exerts anti-RA effects (129–131). In CIA mice, oxymatrine treatment markedly improved arthritis symptoms, reduced the abundance of Firmicutes in the gut and increased the abundance of Bacteroidota, Patescibacteria and Campylobacterota, thereby reshaping the gut microbiota towards more normal levels (131). Moreover, oxymatrine treatment corrected the imbalance in CD4+ T cell subsets in CIA mice, restoring intestinal immune balance by regulating the Th1/Th2, Treg/Th17 and Tfr/Tfh cell ratios (131). Limited clinical studies also suggest that adding oxymatrine to routine RA treatment can more effectively reduce inflammatory markers and balance the immune response in patients (132,133).
Polyphenols, commonly found in plants such as vegetables, fruits and soybeans, consist primarily of aromatic rings and hydroxyl groups. These compounds are renowned for their potent anti-inflammatory and antioxidant properties, which offer significant benefits for human health and the prevention of chronic diseases. Due to these properties, polyphenols are emerging as promising candidate drugs for the management of RA.
Curcumin, the main active compound in turmeric, directly targets the intestines and exhibits potent anti-inflammatory, immune-regulatory and intestinal microbiota-modulating effects (134,135). Extensive basic and clinical studies have confirmed curcumin's significant benefits in improving RA (136). In clinical settings, curcumin not only alleviates symptoms and reduces serum inflammatory markers but also demonstrates a high safety profile (137–140). However, curcumin is characterized by poor gastrointestinal absorption and low oral bioavailability, with an absolute bioavailability of only 1%. In rats, oral administration of 100 mg/kg curcumin resulted in a plasma peak of only 0.02 µM, far below the minimum effective concentration of 10 µM needed to inhibit synovial cell and lymphocyte activation in vitro (141–144). Consequently, curcumin's therapeutic effects on RA may be primarily mediated through its extensive pharmacological effect on the intestine. Yang et al (145) indicated that the anti-arthritis effects of oral curcumin depend on the intestine. Curcumin activates the cAMP/PKA and Ca2+/CaMKII signaling pathways, increasing the number of SOM-positive cells in the small intestine and raising SOM levels in both the intestine and serum. This process alleviates inflammation and immune dysfunction, thereby inhibiting the progression of adjuvant-induced arthritis (AIA) in rats. However, when a SOM inhibitor is used or curcumin is administered intraperitoneally, this anti-arthritis effect is markedly diminished.
Quercetin, a flavonoid commonly found in fruits and vegetables, possesses multiple biological activities, including anti-inflammatory, antioxidant, anti-cancer and liver-protective effects. Clinical studies on quercetin for treating RA are limited. While quercetin appears to positively affect arthritis symptoms, its ability to enhance antioxidant capacity and modulate inflammation in patients with RA remains controversial (146,147). Animal studies, however, have shown that quercetin's potent antioxidant properties not only improve RA but also alleviate side effects caused by traditional RA treatments (148,149). Moreover, research has demonstrated that RA is associated with enteric neurodegenerative changes that impair intestinal functions such as absorption, secretion, and immunity (150,151). Oral administration of quercetin can increase the expression of glial cell-derived neurotrophic factor, glial fibrillary acidic protein and VIP in the intestine. This effect helps reverse the decreased density of intestinal neurons and glial cells in RA mice, restores the morphological changes in the myenteric and submucosal plexuses, and alleviates both intestinal and joint inflammation (152,153).
Natural polysaccharides are macromolecular substances primarily extracted from plants, algae, animals, fungi and bacteria. These polysaccharides exhibit a wide range of important biological activities, including anti-hyperlipidemic, antioxidant, anti-tumor, anti-hepatotoxicity, and immunomodulatory effects (154). Several Chinese herbal medicines, such as Angelica sinensis, wolfberry, Acanthopanax senticosus and Ephedra, contain natural polysaccharides that have been shown to alleviate RA by protecting the intestines and modulating the intestinal microbiota.
ASP, the main active components of Angelica sinensis, enhance immunity and are non-starch polysaccharides that are not digested by the small intestine but serve as a primary energy source for the intestinal flora in the large intestine, thus supporting intestinal health (155). ASP intervention has been shown to relieve joint swelling and inflammation in RA animal models. Pseudo-aseptic tests confirmed that this relief is dependent on specific intestinal bacteria, such as Lactobacillus, Bifidobacterium and Faecalibaculum. Moreover, the metabolites produced by these bacteria, resulting from the interaction between ASP and the microbiota, contribute to the improvement of RA (156). ASP also alters gene expression in the colon and the intestinal microbiota. It promotes the expression of Cldn5 (a gene related to tight junctions), thereby enhancing the intestinal epithelial cell tight junctions in CIA mice, increasing the abundance of anti-inflammatory bacteria such as Norank f Norank o Clostridia UCG-014, Lactobacillus, Norank f Oscillospiraceae and Norank f Desulfovibrionaceae and reducing the migration of inflammatory factors. Additionally, ASP boosts the expression of proteins that inhibit osteoclast differentiation, such as Slit3 and Rgs18, promoting bone formation, and alleviating RA symptoms (27).
In traditional Chinese medicine, Acanthopanax senticosus has long been used to treat arthritis (157,158). ASPS, one of its key active ingredients, possesses functions including immune regulation, antioxidation and anti-diabetic properties (159–161). Studies have explored the mechanism of ASPS in treating RA, indicating that its therapeutic effect in CIA mice is partly dependent on the gut microbiota. This is because when CIA mice were treated with an antibiotic cocktail, arthritis worsens and when both ASPS and an antibiotic cocktail were administered, the protective effect of ASPS on joints was markedly weakened (162). Non-targeted UPLCMS metabolomics analysis of serum samples and PICRUSt2 (a method for accurately predicting 16S rRNA functional genes) analysis of fecal samples revealed that, following ASPS treatment, γ-glutamylcysteine may act as a metabolite of the gut microbiota and contribute to the anti-CIA effect of ASPS. This effect is likely mediated through the inhibition of NLRP3 inflammasome activation by γ-glutamylcysteine, leading to a reduction in IL-1β and caspase-1 expression, thereby exerting an anti-inflammatory effect (162).
LBP, a key active component of wolfberry, is not directly digested or absorbed by the human body. Instead, it reaches the large intestine where it is metabolized by the gut microbiota. LBP is highly valued for its medicinal properties, including enhancing barrier function, anti-inflammatory, antioxidant, anti-tumor effects and its ability to regulate the intestinal microbiota (163–167). LBP exerts an anti-CIA effect regulates chondrocyte proliferation, reduces the expression of inflammatory cytokines such as IL-1α, IL-1β, IL-12 and IL-17 and restores anti-inflammatory cytokine IL-10 levels (168–170). Additionally, LBP may alter the gut microbiota composition, increasing the abundance of microbial taxa that produce S-adenosylmethionine, which induces DNA hypermethylation of RA-related genes (such as Dpep3, Gstm6, Slc27a2, Col11a2, Sycp2, SNORA22) in intestinal epithelial cells. This suppresses gene expression, reduces inflammatory cytokine levels, and mitigates inflammation (170). The elimination of LBP's anti-CIA effect by antibiotics further suggests that its therapeutic action relies on the intestinal microbiota (171).
Ephedra, a herbaceous plant, has a long history in Chinese medicine for treating colds, edema and other ailments. Studies have identified its anti-inflammatory properties, with its primary component, ESP, showing efficacy in improving RA symptoms (172,173). ESP modulates the intestinal microbiota and its metabolites. After ESP treatment, the gut microbiota dysbiosis in CIA mice was corrected, leading to increased abundances of Dubosiella, Bifidobacterium, Clostridium and Pseudoramibactere, elevated colonic butyrate levels, reduced ileal and colonic permeability, and alleviated intestinal inflammation (174). Additionally, the upregulation of HDAC1 and HDAC2 in response to intestinal inflammation induces NF-κB phosphorylation and activates TLR4 and MyD88 in the synovial tissue, key factors in RA-associated immune imbalance. ESP counteracts this by reducing the nuclear translocation of the NF-κB p65 subunit, inhibiting the TLR4 signaling pathway and modulating the HDAC/TLR4/NF-κB pathway. This reduces the release of inflammatory cytokines, limiting their entry from the intestine into the joints, thereby minimizing joint damage and alleviating RA symptoms (173,174).
Paeony, a traditional Chinese herbal medicine, is highly regarded for its medicinal properties. TGP, extracted from the roots of paeony, serves as an immunomodulator and is widely used in China for RA treatment. Although clinical studies have not yet conclusively demonstrated that combining TGP with conventional drugs offers superior efficacy in treating RA, they have shown that TGP can mitigate liver function damage caused by drug treatments (175–178). The primary chemical components of TGP include paeoniflorin, albiflorin, hydroxy-paeoniflorin, paeonin and benzoylpaeoniflorin, all of which are monoterpene glycosides with low bioavailability and poor absorption (179,180). These compounds tend to accumulate in the intestine and can be metabolized by the intestinal microbiota (181–183). TGP treatment reduces arthritis severity in CIA rats, restructures the gut microbiota by correcting 78% of differential taxonomic profiles and enriches beneficial symbionts, thereby restoring intestinal ecological balance. Additionally, TGP markedly downregulates intestinal IFN-γ and secretory IgA production, modulates mucosal immunity, induces autoimmune tolerance and suppressed inflammatory responses (180). TGP also inhibits the expression of VEGF in CIA rats, regulating synovial neovascularization and abnormal synovial cell proliferation (184). These findings suggest that the therapeutic effects of TGP on RA may be mediated through intestinal regulatory mechanisms.
Polysaccharides and glycosides of Aralia echinocaulis. Aralia echinocaulis (A. echinocaulis), an Aralia species (Araliaceae), is a medicinal herb primarily found in southwestern China and in Zhejiang and Anhui provinces. Rich in polysaccharides and glycosides, it is commonly used in medicinal wine and has shown therapeutic benefits for RA (185). The total polysaccharide and glycoside (TPG) from A. echinocaulis can reduce serum IL-1β and TNF-α levels in CIA rats, alleviate synovial hyperplasia and modulate the gut microbiota composition. KEGG pathway and COG functional analyses reveal that TPG-induced changes in the gut microbiota are linked to the transport and metabolism of coenzymes and nucleotides, which may generate anti-RA metabolites, thereby suppressing CIA. However, TPG from A. echinocaulis showed no effect in antibiotic-treated CIA rats, indicating that its anti-arthritic action depends on the regulation of the gut microbiota (186).
THH is a commonly used drug in China, known for its immunosuppressive, anti-tumor and anti-inflammatory properties (187). Numerous studies have demonstrated that THH extract improves joint indices, reduces swelling and mitigates joint damage in RA animal models (188,189). Further investigations have revealed that THH extract markedly elevates the colonic mRNA levels of tight-junction proteins and mucins in RA mice. It also reduces the abundance of pathogenic bacteria, such as Marvinbryantia, Desulfovibrio, Parabobacterides, Bacteroides and Butyricimonas, while enriching beneficial taxa such as ifidobacterium, Akkermansia, Lactobacillus and Roseburia. These alterations help restore microbial balance, reprogram metabolic pathways, enhance SCFA production, and modulate BA metabolism (190,191). Additionally, THH treatment reduces the expression of pro-inflammatory cytokines (IL-1β, IL-8, IL-17) and increases anti-inflammatory cytokine IL-10 in the colon of RA model mice. It also inhibits NLRP3 inflammasome activation and blocks the TLR4/MyD88/MAPK signaling pathways in muscles and plasma, reducing joint inflammation and protecting the joints (190,191). However, these therapeutic effects were not observed in RA mice with gut microbiota deficiency, confirming that a healthy gut microbiota is essential for the therapeutic efficacy of THH extract (190,191).
RTA, a liana primarily found in China and Southeast Asia, has anti-inflammatory and analgesic properties, making it widely used for treating chronic pain and gastrointestinal disorders (192–194). RTA extract reduces serum inflammatory markers in RA model animals, increases chondrocyte survival in inflammatory environments, and improves arthritis symptoms (194–196). Research by Qin et al (197) suggests that the therapeutic the mechanism of RTA extract in RA may involve modulating intestinal immune responses and restoring microbial balance. Following RTA treatment, Th17 (IL-17A, RORC, IL-1β and IL-6) and Treg (IL-10 and FOXP3) cell-related protein and mRNA levels in the colon tissues of AIA rats were downregulated and upregulated, respectively, promoting the restoration of the Th17//Treg balance. Additionally, RTA extract reshaped the intestinal microbiota of AIA rats, increasing beneficial bacteria and reducing RA-associated strains, such as Liilactobacillus and Streptococcus.
Paederia scandens, a herb native to countries such as China, India and Vietnam, is commonly used to treat rheumatism and digestive tract diseases and is regarded as a safe plant. In vitro cell experiments have demonstrated that Paederia scandens extract (PSE) reduces the mRNA levels of pro-inflammatory cytokines (IL-6, IL-1β and IL-17) in fibroblast-like synovial cells from patients with RA and inhibits cell proliferation by suppressing the activation of the JAK-STAT pathway (198). In vivo studies further confirmed PSE's therapeutic effect on CIA mice, restoring the intestinal microbiota balance disturbed by collagen induction. PSE reduced relative abundance of inflammation-related microbiota, such as Desulfovibrio, Mucispirillum, Helicobacter and Lachnospiraceae. However, whether the intestinal regulatory mechanism plays a pivotal role in PSE's treatment of RA remains to be further explored (199).
ATO, a compound with a long history of use in treating various diseases, was approved by the U.S. Food and Drug Administration in 2000 for treating acute promyelocytic leukemia, eventually emerging as a cutting-edge treatment for lymphoma, solid tumors, and other conditions (200). Studies suggest that ATO holds potential in RA treatment by balancing immune cells and inhibiting angiogenesis (201–203). Additionally, regulating intestinal microecology and abnormal metabolites may be part of ATO's therapeutic action in RA (204). Specifically, ATO treatment increased the richness and diversity of the intestinal microbiota in CIA mice, restoring the imbalance between the two major phyla, Firmicutes and Bacteroidetes (204). On a metabolic level, ATO regulates several metabolites, including (−)-β-pinene, a substrate involved in lipid metabolism related to RA. ATO may alleviate RA symptoms by regulating lipid metabolism through the modulation of (−)-β-pinene levels (204).
Columbianadin. Angelicae pubescentis radix (APR), the dried root of Angelica pubescens Maxim. f. biserrata Shan et Yuan, has a long history of use in treating rheumatic joint pain. A natural component extracted from APR, columbianadin, exhibits anti-inflammatory, analgesic, and anti-tumor properties (205–208). Columbianadin effectively inhibits the JAK1/STAT3 and NF-κB inflammatory pathways, improves the Keap1/Nrf2 oxidative stress pathway and modulates the expression of inflammatory factors (downregulating IL-1β, TNF-α and IL-6) and antioxidant markers (upregulating glutathione and superoxide dismutase). This results in reduced swelling and joint protection (209). Furthermore, columbianadin restores microbial diversity in the small intestine of CIA mice, modulates energy- and amino-acid-metabolic pathways, and helps maintain micro-ecological homeostasis with minimal adverse effects (209).
The ‘gut-joint axis’ is not exclusive to RA but also occurs in other immune-mediated arthritis diseases, with distinct mechanisms at play. Studies have shown that ~50% of patients with spondyloarthritis (SpA) develop subclinical intestinal inflammation, which can progress to inflammatory bowel disease (IBD), most commonly Crohn's disease (210,211). By contrast, subclinical intestinal inflammation in RA predominantly involves the small intestine and is characterized by lymphocyte and macrophage infiltration (39,210,211). Furthermore, the composition of the intestinal microbiota differs between RA and SpA: Prevotella is markedly and specifically enriched in RA, whereas no distinct microbial signature has yet been identified for SpA. However, dysbiosis-driven activation of the IL-23/IL-17 axis plays a pivotal role in the pathogenesis of both SpA and IBD (212). Additionally, whereas CD4+ T cells that dominate the ‘gut-joint axis’ in RA, CD8+ T cells bearing identical T-cell receptor clonotypes are the predominant immune population in both the gut and joints of SpA patients (210,213).
Overall, alterations in the microbiota are a key factor in the ‘gut-joint axis’ of RA. These changes not only influence mucosal and systemic autoimmune responses via metabolites, molecular mimicry and other mechanisms but are also closely linked to the efficacy and side effects of RA treatments, particularly methotrexate (24,214,215). Research focusing on specific microbiota and metabolomics may offer a more targeted approach to understanding the ‘gut-joint axis’ in RA. Since the intestinal microbiota is influenced by numerous factors, broadly studying all microbiota changes to pinpoint the pathogenesis of diseases may lead to incorrect conclusions.
Natural compounds possess diverse biological activities and can improve RA by regulating the intestinal microecology. Importantly, the anti-RA effects of certain compounds are dependent on the intestine. The intestinal microbiota represents a potential target for the treatment of RA, particularly with high-molecular-weight and low-bioavailability natural compounds. Although existing reviews have catalogued the potential mechanisms by which natural compounds ameliorate RA and occasionally mention the gut-joint axis, the present review presented a more comprehensive and systematic summary of the anti-RA effects of natural compounds based on this mechanism (216–219). The present review highlighted the protective effects of natural compounds such as alkaloids, polyphenols, polysaccharides and plant extracts on the intestinal mucosa and the regulation of intestinal flora homeostasis in RA model animals, ultimately improving RA symptoms. It explored the anti-RA potential and mechanisms of these compounds, providing a scientific foundation and new insights for the development of novel therapeutic drugs. Several natural compounds have already been widely used in clinical practice, while others, with solid scientific support for drug development, should be prioritized. For example, berberine, a well-studied natural compound, has shown significant therapeutic effects across various diseases, suggesting its broad potential in RA drug development. Lycium barbarum polysaccharides, one of the key active components of L. barbarum, also exhibit notable efficacy in treating RA. Moreover, L. barbarum, being a readily available and low-cost source, could be a valuable candidate for drug development.
Despite their therapeutic potential, most natural compounds suffer from low oral bioavailability and tend to accumulate in the gastrointestinal tract after oral administration. While this accumulation may be advantageous for RA treatment through the ‘gut-joint axis’ mechanism, improving the bioavailability of these compounds could further enhance their efficacy in disease recovery. Additionally, challenges in translating natural compounds into clinical drugs include structural instability, poor solubility and unclear toxic or side effects. Hence, novel drug discovery remains a protracted, multi-parameter endeavor that demands rigorous, head-to-toe optimization and integrated assessment of all pharmacological, pharmacokinetic and safety attributes. However, as pharmacological mechanisms of natural compounds are being dissected at ever-greater depth, our knowledge of these compounds has broadened and their multifaceted profiles are now improved understood.
At present, new drug delivery methods such as nanoparticles are developing rapidly. It is expected that in the future, there will be more basic research and clinical trials on the treatment of RA using natural compounds through the ‘gut-joint axis’ pathway. This is precisely what is needed at present.
Not applicable.
Funding: No funding was received.
Not applicable.
WH was responsible for conceptualization, writing the original draft, writing, reviewing and editing. RL was responsible for writing, reviewing and editing and providing illustrations and tables. ZZ was responsible for writing, reviewing and editing. Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
|
Di Matteo A, Bathon JM and Emery P: Rheumatoid arthritis. Lancet. 402:2019–2033. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
GBD 2021 Rheumatoid Arthritis Collaborators, . Global, regional, and national burden of rheumatoid arthritis, 1990–2020, and projections to 2050: A systematic analysis of the Global Burden of Disease Study 2021. Lancet Rheumatol. 5:e594–e610. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Aletaha D and Smolen JS: Diagnosis and management of rheumatoid arthritis: A Review. JAMA. 320:1360–1372. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Brewer RC, Lanz TV, Hale CR, Sepich-Poore GD, Martino C, Swafford AD, Carroll TS, Kongpachith S, Blum LK, Elliott SE, et al: Oral mucosal breaks trigger anti-citrullinated bacterial and human protein antibody responses in rheumatoid arthritis. Sci Transl Med. 15:eabq84762023. View Article : Google Scholar : PubMed/NCBI | |
|
Luo Y, Tong Y, Wu L, Niu H, Li Y, Su LC, Wu Y, Bozec A, Zaiss MM, Qing P, et al: Alteration of gut microbiota in individuals at High-Risk for rheumatoid arthritis associated with disturbed metabolome and the initiation of arthritis through the triggering of mucosal immunity imbalance. Arthritis Rheumatol. 75:1736–1748. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
de Jesus VC, Singh M, Schroth RJ, Chelikani P and Hitchon CA: Association of bitter taste receptor T2R38 polymorphisms, oral microbiota, and rheumatoid arthritis. Curr Issues Mol Biol. 43:1460–1472. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
van Delft MAM and Huizinga TWJ: An overview of autoantibodies in rheumatoid arthritis. J Autoimmun. 110:1023922020. View Article : Google Scholar : PubMed/NCBI | |
|
Kronzer VL and Sparks JA: Occupational inhalants, genetics and the respiratory mucosal paradigm for ACPA-positive rheumatoid arthritis. Ann Rheum Dis. 82:303–305. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Holers VM, Demoruelle KM, Buckner JH, James EA, Firestein GS, Robinson WH, Steere AC, Zhang F, Norris JM, Kuhn KA and Deane KD: Distinct mucosal endotypes as initiators and drivers of rheumatoid arthritis. Nat Rev Rheumatol. 20:601–613. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao T, Wei Y, Zhu Y, Xie Z, Hai Q, Li Z and Qin D: Gut microbiota and rheumatoid arthritis: From pathogenesis to novel therapeutic opportunities. Front Immunol. 13:10071652022. View Article : Google Scholar : PubMed/NCBI | |
|
Jiao Y, Wu L, Huntington ND and Zhang X: Crosstalk between gut microbiota and innate immunity and its implication in autoimmune diseases. Front Immunol. 11:2822020. View Article : Google Scholar : PubMed/NCBI | |
|
Wells PM, Adebayo AS, Bowyer RCE, Freidin MB, Finckh A, Strowig T, Lesker TR, Alpizar-Rodriguez D, Gilbert B, Kirkham B, et al: Associations between gut microbiota and genetic risk for rheumatoid arthritis in the absence of disease: A cross-sectional study. Lancet Rheumatol. 2:e418–e427. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Zhang D, Jia H, Feng Q, Wang D, Liang D, Wu X, Li J, Tang L, Li Y, et al: The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med. 21:895–905. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Wright K, Davis JM, Jeraldo P, Marietta EV, Murray J, Nelson H, Matteson EL and Taneja V: An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 8:432016. View Article : Google Scholar : PubMed/NCBI | |
|
Inamo J: Non-causal association of gut microbiome on the risk of rheumatoid arthritis: A Mendelian randomisation study. Ann Rheum Dis. 80:e1032021. View Article : Google Scholar : PubMed/NCBI | |
|
Jeong Y, Kim JW, You HJ, Park SJ, Lee J, Ju JH, Park MS, Jin H, Cho ML, Kwon B, et al: Gut microbial composition and function are altered in patients with early rheumatoid arthritis. J Clin Med. 8:6932019. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng M, Zhao Y, Cui Y, Zhong C, Zha Y, Li S, Cao G, Li M, Zhang L, Ning K and Han J: Stage-specific roles of microbial dysbiosis and metabolic disorders in rheumatoid arthritis. Ann Rheum Dis. 81:1669–1677. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun H, Guo Y, Wang H, Yin A, Hu J, Yuan T, Zhou S, Xu W, Wei P, Yin S, et al: Gut commensal Parabacteroides distasonis alleviates inflammatory arthritis. Gut. 72:1664–1677. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu X, Zhan G, Chen R, Wang D, Guan S and Xu H: Gut microbiota and its role in stress-induced hyperalgesia: Gender-specific responses linked to different changes in serum metabolites. Pharmacol Res. 177:1061292022. View Article : Google Scholar : PubMed/NCBI | |
|
Barton JR, Londregan AK, Alexander TD, Entezari AA, Covarrubias M and Waldman SA: Enteroendocrine cell regulation of the gut-brain axis. Front Neurosci. 17:12729552023. View Article : Google Scholar : PubMed/NCBI | |
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, et al: Gut microbiota bridges dietary nutrients and host immunity. Sci China Life Sci. 66:2466–2514. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen S, Dan L, Xiang L, He Q, Hu D and Gao Y: The role of gut flora-driven Th cell responses in preclinical rheumatoid arthritis. J Autoimmun. 154:1034262025. View Article : Google Scholar : PubMed/NCBI | |
|
Alpizar-Rodriguez D, Lesker TR, Gronow A, Gilbert B, Raemy E, Lamacchia C, Gabay C, Finckh A and Strowig T: Prevotella copri in individuals at risk for rheumatoid arthritis. Ann Rheum Dis. 78:590–593. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, et al: Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2:e012022013. View Article : Google Scholar : PubMed/NCBI | |
|
Picchianti-Diamanti A, Panebianco C, Salemi S, Sorgi ML, Di Rosa R, Tropea A, Sgrulletti M, Salerno G, Terracciano F, D'Amelio R, et al: Analysis of gut microbiota in rheumatoid arthritis patients: Disease-related dysbiosis and modifications induced by etanercept. Int J Mol Sci. 19:29382018. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Zeng B, Zhang J, Li W, Mou F, Wang H, Zou Q, Zhong B, Wu L, Wei H and Fang Y: Role of the gut microbiome in modulating arthritis progression in mice. Sci Rep. 6:305942016. View Article : Google Scholar : PubMed/NCBI | |
|
Hu Q, Wu C, Yu J, Luo J and Peng X: Angelica sinensis polysaccharide improves rheumatoid arthritis by modifying the expression of intestinal Cldn5, Slit3 and Rgs18 through gut microbiota. Int J Biol Macromol. 209:153–161. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Pianta A, Arvikar S, Strle K, Drouin EE, Wang Q, Costello CE and Steere AC: Evidence of the immune relevance of Prevotella copri, a gut microbe, in patients with rheumatoid arthritis. Arthritis Rheumatol. 69:964–975. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Pianta A, Chiumento G, Ramsden K, Wang Q, Strle K, Arvikar S, Costello CE and Steere AC: Identification of novel, immunogenic HLA-DR-Presented Prevotella copri peptides in patients with rheumatoid arthritis. Arthritis Rheumatol. 73:2200–2205. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Chen B, Li S, Yang L, Zhu D, Wang Y, Wang H, Wang T, Shi B, Gai Z, et al: Detection and characterization of bacterial nucleic acids in culture-negative synovial tissue and fluid samples from rheumatoid arthritis or osteoarthritis patients. Sci Rep. 8:143052018. View Article : Google Scholar : PubMed/NCBI | |
|
Peng Y, Huang Y, Li H, Li C, Wu Y, Wang X, Wang Q, He J and Miao C: Associations between rheumatoid arthritis and intestinal flora, with special emphasis on RA pathologic mechanisms to treatment strategies. Microb Pathog. 188:1065632024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Wu Y, Luo Y, Wei S, Lu C, Zhou Y, Wang J, Miao T, Lin H, Zhao Y, et al: Self-Balance of intestinal flora in spouses of patients with rheumatoid arthritis. Front Med (Lausanne). 7:5382020. View Article : Google Scholar : PubMed/NCBI | |
|
Padyukov L: Genetics of rheumatoid arthritis. Semin Immunopathol. 44:47–62. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Saevarsdottir S, Stefansdottir L, Sulem P, Thorleifsson G, Ferkingstad E, Rutsdottir G, Glintborg B, Westerlind H, Grondal G, Loft IC, et al: Multiomics analysis of rheumatoid arthritis yields sequence variants that have large effects on risk of the seropositive subset. Ann Rheum Dis. 81:1085–1095. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Larid G, Pancarte M, Offer G, Clavel C, Martin M, Pradel V, Auger I, Lafforgue P, Roudier J, Serre G and Balandraud N: In rheumatoid arthritis patients, HLA-DRB1*04:01 and rheumatoid nodules are associated with ACPA to a particular fibrin epitope. Front Immunol. 12:6920412021. View Article : Google Scholar : PubMed/NCBI | |
|
Asquith M, Sternes PR, Costello ME, Karstens L, Diamond S, Martin TM, Li Z, Marshall MS, Spector TD, le Cao KA, et al: HLA alleles associated with risk of ankylosing spondylitis and rheumatoid arthritis influence the gut microbiome. Arthritis Rheumatol. 71:1642–1650. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Paray BA, Albeshr MF, Jan AT and Rather IA: Leaky gut and autoimmunity: An Intricate balance in individuals health and the diseased State. Int J Mol Sci. 21:97702020. View Article : Google Scholar : PubMed/NCBI | |
|
Marietta EV, Murray JA, Luckey DH, Jeraldo PR, Lamba A, Patel R, Luthra HS, Mangalam A and Taneja V: Suppression of inflammatory arthritis by human Gut-derived prevotella histicola in humanized mice. Arthritis Rheumatol. 68:2878–2888. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Tajik N, Frech M, Schulz O, Schälter F, Lucas S, Azizov V, Dürholz K, Steffen F, Omata Y, Rings A, et al: Targeting zonulin and intestinal epithelial barrier function to prevent onset of arthritis. Nat Commun. 11:19952020. View Article : Google Scholar : PubMed/NCBI | |
|
Audo R, Sanchez P, Riviere B, Mielle J, Tan J, Lukas C, Macia L, Morel J and Immediato Daien C: Rheumatoid arthritis is associated with increased gut permeability and bacterial translocation which are reversed by inflammation control. Rheumatology (Oxford). Aug 10–2022.(Epub ahead of print). doi: 10.1093/rheumatology/keac454. | |
|
Heidt C, Kammerer U, Fobker M, Ruffer A, Marquardt T and Reuss-Borst M: Assessment of intestinal permeability and inflammation Bio-markers in patients with rheumatoid arthritis. Nutrients. 15:23862023. View Article : Google Scholar : PubMed/NCBI | |
|
Wen J, Lyu P, Stolzer I, Xu J, Gießl A, Lin Z, Andreev D, Kachler K, Song R, Meng X, et al: Epithelial HIF2α expression induces intestinal barrier dysfunction and exacerbation of arthritis. Ann Rheum Dis. 81:1119–1130. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ruiz-Limón P, Mena-Vázquez N, Moreno-Indias I, Manrique-Arija S, Lisbona-Montañez JM, Cano-García L, Tinahones FJ and Fernández-Nebro A: Collinsella is associated with cumulative inflammatory burden in an established rheumatoid arthritis cohort. Biomed Pharmacother. 153:1135182022. View Article : Google Scholar : PubMed/NCBI | |
|
Schwartz AJ, Das NK, Ramakrishnan SK, Jain C, Jurkovic MT, Wu J, Nemeth E, Lakhal-Littleton S, Colacino JA and Shah YM: Hepatic hepcidin/intestinal HIF-2α axis maintains iron absorption during iron deficiency and overload. J Clin Invest. 129:336–348. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Saeedi BJ, Kao DJ, Kitzenberg DA, Dobrinskikh E, Schwisow KD, Masterson JC, Kendrick AA, Kelly CJ, Bayless AJ, Kominsky DJ, et al: HIF-dependent regulation of claudin-1 is central to intestinal epithelial tight junction integrity. Mol Biol Cell. 26:2252–2262. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Das NK, Schwartz AJ, Barthel G, Inohara N, Liu Q, Sankar A, Hill DR, Ma X, Lamberg O, Schnizlein MK, et al: Microbial metabolite signaling is required for systemic Iron homeostasis. Cell Metab. 31:115–130.e6. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rogier R, Evans-Marin H, Manasson J, van der Kraan PM, Walgreen B, Helsen MM, van den Bersselaar LA, van de Loo FA, van Lent PL, Abramson SB, et al: Alteration of the intestinal microbiome characterizes preclinical inflammatory arthritis in mice and its modulation attenuates established arthritis. Sci Rep. 7:156132017. View Article : Google Scholar : PubMed/NCBI | |
|
Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, et al: Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med. 14:eabn51662022. View Article : Google Scholar : PubMed/NCBI | |
|
Takahashi D, Hoshina N, Kabumoto Y, Maeda Y, Suzuki A, Tanabe H, Isobe J, Yamada T, Muroi K, Yanagisawa Y, et al: Microbiota-derived butyrate limits the autoimmune response by promoting the differentiation of follicular regulatory T cells. EBioMedicine. 58:1029132020. View Article : Google Scholar : PubMed/NCBI | |
|
Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al: Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 139:485–498. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Tan TG, Sefik E, Geva-Zatorsky N, Kua L, Naskar D, Teng F, Pasman L, Ortiz-Lopez A, Jupp R, Wu HJ, et al: Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proc Natl Acad Sci USA. 113:E8141–E8150. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C and Mathis D: Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 32:815–827. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Rojas M, Restrepo-Jimenez P, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, Leung PSC, Ansari AA, Gershwin ME and Anaya JM: Molecular mimicry and autoimmunity. J Autoimmun. 95:100–123. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Albani S, Keystone EC, Nelson JL, Ollier WE, La Cava A, Montemayor AC, Weber DA, Montecucco C, Martini A and Carson DA: Positive selection in autoimmunity: Abnormal immune responses to a bacterial dnaJ antigenic determinant in patients with early rheumatoid arthritis. Nat Med. 1:448–452. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Alcaide-Ruggiero L, Molina-Hernandez V, Granados MM and Dominguez JM: Main and minor types of collagens in the articular cartilage: The role of collagens in repair tissue evaluation in chondral defects. Int J Mol Sci. 22:133292021. View Article : Google Scholar : PubMed/NCBI | |
|
Gomez A, Luckey D, Yeoman CJ, Marietta EV, Berg Miller ME, Murray JA, White BA and Taneja V: Loss of sex and age driven differences in the gut microbiome characterize arthritis-susceptible 0401 mice but not arthritis-resistant 0402 mice. PLoS One. 7:e360952012. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou C, Zhao H, Xiao XY, Chen BD, Guo RJ, Wang Q, Chen H, Zhao LD, Zhang CC, Jiao YH, et al: Metagenomic profiling of the pro-inflammatory gut microbiota in ankylosing spondylitis. J Autoimmun. 107:1023602020. View Article : Google Scholar : PubMed/NCBI | |
|
Miyoshi M and Liu S: Collagen-induced arthritis models. Methods Mol Biol. 2766:3–7. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liang B, Ge C, Lönnblom E, Lin X, Feng H, Xiao L, Bai J, Ayoglu B, Nilsson P, Nandakumar KS, et al: The autoantibody response to cyclic citrullinated collagen type II peptides in rheumatoid arthritis. Rheumatology (Oxford). 58:1623–1633. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Pianta A, Arvikar SL, Strle K, Drouin EE, Wang Q, Costello CE and Steere AC: Two rheumatoid arthritis-specific autoantigens correlate microbial immunity with autoimmune responses in joints. J Clin Invest. 127:2946–2956. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang JY and Roehrl MH: Glycosaminoglycans are a potential cause of rheumatoid arthritis. Proc Natl Acad Sci USA. 99:14362–14367. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Hua R, Ni Q, Eliason TD, Han Y, Gu S, Nicolella DP, Wang X and Jiang JX: Biglycan and chondroitin sulfate play pivotal roles in bone toughness via retaining bound water in bone mineral matrix. Matrix Biol. 94:95–109. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rosser EC, Piper CJM, Matei DE, Blair PA, Rendeiro AF, Orford M, Alber DG, Krausgruber T, Catalan D, Klein N, et al: Microbiota-derived metabolites suppress arthritis by amplifying Aryl-hydrocarbon receptor activation in regulatory B cells. Cell Metab. 31:837–851.e10. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Correa-Oliveira R, Fachi JL, Vieira A, Sato FT and Vinolo MA: Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunol. 5:e732016. View Article : Google Scholar : PubMed/NCBI | |
|
Yao Y, Cai X, Zheng Y, Zhang M, Fei W, Sun D, Zhao M, Ye Y and Zheng C: Short-chain fatty acids regulate B cells differentiation via the FFA2 receptor to alleviate rheumatoid arthritis. Br J Pharmacol. 179:4315–4329. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Wei J, Zhang W, Doherty M, Zhang Y, Xie H, Li W, Wang N, Lei G and Zeng C: Gut dysbiosis in rheumatic diseases: A systematic review and meta-analysis of 92 observational studies. EBioMedicine. 80:1040552022. View Article : Google Scholar : PubMed/NCBI | |
|
He J, Chu Y, Li J, Meng Q, Liu Y, Jin J, Wang Y, Wang J, Huang B, Shi L, et al: Intestinal butyrate-metabolizing species contribute to autoantibody production and bone erosion in rheumatoid arthritis. Sci Adv. 8:eabm15112022. View Article : Google Scholar : PubMed/NCBI | |
|
Kim DS, Kwon JE, Lee SH, Kim EK, Ryu JG, Jung KA, Choi JW, Park MJ, Moon YM, Park SH, et al: Attenuation of rheumatoid inflammation by sodium butyrate through reciprocal targeting of HDAC2 in osteoclasts and HDAC8 in T cells. Front Immunol. 9:15252018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang M, Huang J, Fan H, He D, Zhao S, Shu Y, Li H, Liu L, Lu S, Xiao C and Liu Y: Treatment of rheumatoid arthritis using combination of methotrexate and tripterygium glycosides Tablets-A quantitative plasma pharmacochemical and pseudotargeted metabolomic approach. Front Pharmacol. 9:10512018. View Article : Google Scholar : PubMed/NCBI | |
|
Surowiec I, Arlestig L, Rantapaa-Dahlqvist S and Trygg J: Metabolite and lipid profiling of biobank plasma samples collected prior to onset of rheumatoid arthritis. PLoS One. 11:e01641962016. View Article : Google Scholar : PubMed/NCBI | |
|
Seymour BJ, Trent B, Allen BE, Berlinberg AJ, Tangchittsumran J, Jubair WK, Chriswell ME, Liu S, Ornelas A, Stahly A, et al: Microbiota-dependent indole production stimulates the development of collagen-induced arthritis in mice. J Clin Invest. 134:e1676712023. View Article : Google Scholar : PubMed/NCBI | |
|
Cao S, Meng X, Li Y, Sun L, Jiang L, Xuan H and Chen X: Bile acids elevated in chronic periaortitis could activate Farnesoid-X-receptor to suppress IL-6 production by macrophages. Front Immunol. 12:6328642021. View Article : Google Scholar : PubMed/NCBI | |
|
Fiorucci S, Biagioli M, Zampella A and Distrutti E: Bile acids activated receptors regulate innate immunity. Front Immunol. 9:18532018. View Article : Google Scholar : PubMed/NCBI | |
|
Hu J, Wang C, Huang X, Yi S, Pan S, Zhang Y, Yuan G, Cao Q, Ye X and Li H: Gut microbiota-mediated secondary bile acids regulate dendritic cells to attenuate autoimmune uveitis through TGR5 signaling. Cell Rep. 36:1097262021. View Article : Google Scholar : PubMed/NCBI | |
|
Zou F, Qiu Y, Huang Y, Zou H, Cheng X, Niu Q, Luo A and Sun J: Effects of short-chain fatty acids in inhibiting HDAC and activating p38 MAPK are critical for promoting B10 cell generation and function. Cell Death Dis. 12:5822021. View Article : Google Scholar : PubMed/NCBI | |
|
Ignacio Barrasa J, Olmo N, Perez-Ramos P, Santiago-Gómez A, Lecona E, Turnay J and Antonia Lizarbe M: Deoxycholic and chenodeoxycholic bile acids induce apoptosis via oxidative stress in human colon adenocarcinoma cells. Apoptosis. 16:1054–1067. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng X, Pi Z, Zheng Z, Liu S, Song F and Liu Z: Combined 16S rRNA gene sequencing and metabolomics to investigate the protective effects of Wu-tou decoction on rheumatoid arthritis in rats. J Chromatogr B Analyt Technol Biomed Life Sci. 1199:1232492022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu M, Li S, Cao N, Wang Q, Liu Y, Xu Q, Zhang L, Sun C, Xiao X and Yao J: Intestinal flora, intestinal metabolism, and intestinal immunity changes in complete Freud's adjuvant-rheumatoid arthritis C57BL/6 mice. Int Immunopharmacol. 125:1110902023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Yu Y, Liao J, Hu W, Bian X, Wu J and Zhu YZ: S-Propargyl-Cysteine remodels the gut microbiota to alleviate rheumatoid arthritis by regulating bile acid metabolism. Front Cell Infect Microbiol. 11:6705932021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, Peng F, Cheng H, Zhang D, Zhang Y, Wang L, Tang F, Wang J, Wan Y, Wu J, et al: Chronic cold environment regulates rheumatoid arthritis through modulation of gut microbiota-derived bile acids. Sci Total Environ. 903:1668372023. View Article : Google Scholar : PubMed/NCBI | |
|
Gonzalez-Rey E, Delgado-Maroto V, Souza Moreira L and Delgado M: Neuropeptides as therapeutic approach to autoimmune diseases. Curr Pharm Des. 16:3158–3172. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Delgado M, Pozo D and Ganea D: The significance of vasoactive intestinal peptide in immunomodulation. Pharmacol Rev. 56:249–290. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Imhof AK, Gluck L, Gajda M, Lupp A, Bräuer R, Schaible HG and Schulz S: Differential antiinflammatory and antinociceptive effects of the somatostatin analogs octreotide and pasireotide in a mouse model of immune-mediated arthritis. Arthritis Rheum. 63:2352–2362. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Jimeno R, Leceta J, Martínez C, Gutiérrez-Cañas I, Pérez-García S, Carrión M, Gomariz RP and Juarranz Y: Effect of VIP on the balance between cytokines and master regulators of activated helper T cells. Immunol Cell Biol. 90:178–186. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Jimeno R, Gomariz RP, Gutierrez-Canas I, Martinez C, Juarranz Y and Leceta J: New insights into the role of VIP on the ratio of T-cell subsets during the development of autoimmune diabetes. Immunol Cell Biol. 88:734–745. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Leceta J, Garin MI and Conde C: Mechanism of immunoregulatory properties of vasoactive intestinal peptide in the K/BxN mice model of autoimmune arthritis. Front Immunol. 12:7018622021. View Article : Google Scholar : PubMed/NCBI | |
|
O'Connor TM, O'Connell J, O'Brien DI, Goode T, Bredin CP and Shanahan F: The role of substance P in inflammatory disease. J Cell Physiol. 201:167–180. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Raap T, Justen HP, Miller LE, Cutolo M, Scholmerich J and Straub RH: Neurotransmitter modulation of interleukin 6 (IL-6) and IL-8 secretion of synovial fibroblasts in patients with rheumatoid arthritis compared to osteoarthritis. J Rheumatol. 27:2558–2565. 2000.PubMed/NCBI | |
|
Barbosa-Cobos RE, Lugo-Zamudio G, Flores-Estrada J, Becerril-Mendoza LT, Rodríguez-Henríquez P, Torres-González R, Moreno-Eutimio MA, Ramirez-Bello J and Moreno J: Serum substance P: An indicator of disease activity and subclinical inflammation in rheumatoid arthritis. Clin Rheumatol. 37:901–908. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang MH, Chung E, Chi GF, Ahn W, Lim JE, Hong HS, Kim DW, Choi H, Kim J and Son Y: Substance P induces M2-type macrophages after spinal cord injury. Neuroreport. 23:786–792. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Hong HS and Son Y: Substance P ameliorates collagen II-induced arthritis in mice via suppression of the inflammatory response. Biochem Biophys Res Commun. 453:179–184. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Galarza-Delgado DA, Azpiri-Lopez JR, Colunga-Pedraza IJ, Cárdenas-de la Garza JA, Vera-Pineda R, Wah-Suárez M, Arvizu-Rivera RI, Martínez-Moreno A, Ramos-Cázares RE, Torres-Quintanilla FJ, et al: Prevalence of comorbidities in Mexican mestizo patients with rheumatoid arthritis. Rheumatol Int. 37:1507–1511. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Castillo-Canon JC, Trujillo-Caceres SJ, Bautista-Molano W, Valbuena-Garcia AM, Fernandez-Avila DG and Acuna-Merchan L: Rheumatoid arthritis in Colombia: A clinical profile and prevalence from a national registry. Clin Rheumatol. 40:3565–3573. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Niu Q, Hao J, Li Z and Zhang H: Helper T cells: A potential target for sex hormones to ameliorate rheumatoid arthritis? (Review). Mol Med Rep. 30:2152024. View Article : Google Scholar : PubMed/NCBI | |
|
Li JY, Chassaing B, Tyagi AM, Vaccaro C, Luo T, Adams J, Darby TM, Weitzmann MN, Mulle JG, Gewirtz AT, et al: Sex steroid deficiency-associated bone loss is microbiota dependent and prevented by probiotics. J Clin Invest. 126:2049–2063. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Porwal K, Pal S, Kulkarni C, Singh P, Sharma S, Singh P, Prajapati G, Gayen JR, Ampapathi RS, Mullick A and Chattopadhyay N: A prebiotic, short-chain fructo-oligosaccharides promotes peak bone mass and maintains bone mass in ovariectomized rats by an osteogenic mechanism. Biomed Pharmacother. 129:1104482020. View Article : Google Scholar : PubMed/NCBI | |
|
Dominianni C, Sinha R, Goedert JJ, Pei Z, Yang L, Hayes RB and Ahn J: Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS One. 10:e01245992015. View Article : Google Scholar : PubMed/NCBI | |
|
Balakrishnan B, Luckey D, Wright K, Davis JM, Chen J and Taneja V: Eggerthella lenta augments preclinical autoantibody production and metabolic shift mimicking senescence in arthritis. Sci Adv. 9:eadg11292023. View Article : Google Scholar : PubMed/NCBI | |
|
Bhambhani S, Kondhare KR and Giri AP: Diversity in chemical structures and biological properties of plant alkaloids. Molecules. 26:33742021. View Article : Google Scholar : PubMed/NCBI | |
|
Ettefagh KA, Burns JT, Junio HA, Kaatz GW and Cech NB: Goldenseal (Hydrastis canadensis L.) extracts synergistically enhance the antibacterial activity of berberine via efflux pump inhibition. Planta Med. 77:835–840. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu M, Wang H, Chen J and Zhu H: Sinomenine improve diabetic nephropathy by inhibiting fibrosis and regulating the JAK2/STAT3/SOCS1 pathway in streptozotocin-induced diabetic rats. Life Sci. 265:1188552021. View Article : Google Scholar : PubMed/NCBI | |
|
Samra YA, Said HS, Elsherbiny NM, Liou GI, El-Shishtawy MM and Eissa LA: Cepharanthine and Piperine ameliorate diabetic nephropathy in rats: Role of NF-kappaB and NLRP3 inflammasome. Life Sci. 157:187–199. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Naz I, Masoud MS, Chauhdary Z, Shah MA and Panichayupakaranant P: Anti-inflammatory potential of berberine-rich extract via modulation of inflammation biomarkers. J Food Biochem. 46:e143892022. View Article : Google Scholar : PubMed/NCBI | |
|
Alorabi M, Cavalu S, Al-Kuraishy HM, Al-Gareeb AI, Mostafa-Hedeab G, Negm WA, Youssef A, El-Kadem AH, Saad HM and Batiha GE: Pentoxifylline and berberine mitigate diclofenac-induced acute nephrotoxicity in male rats via modulation of inflammation and oxidative stress. Biomed Pharmacother. 152:1132252022. View Article : Google Scholar : PubMed/NCBI | |
|
Luo R, Liao Z, Song Y, Yin H, Zhan S, Li G, Ma L, Lu S, Wang K, Li S, et al: Berberine ameliorates oxidative stress-induced apoptosis by modulating ER stress and autophagy in human nucleus pulposus cells. Life Sci. 228:85–97. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Samadi P, Sarvarian P, Gholipour E, Asenjan KS, Aghebati-Maleki L, Motavalli R, Hojjat-Farsangi M and Yousefi M: Berberine: A novel therapeutic strategy for cancer. IUBMB Life. 72:2065–2079. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pirillo A and Catapano AL: Berberine, a plant alkaloid with lipid- and glucose-lowering properties: From in vitro evidence to clinical studies. Atherosclerosis. 243:449–461. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Song D, Hao J and Fan D: Biological properties and clinical applications of berberine. Front Med. 14:564–582. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu YT, Hao HP, Xie HG, Lai L, Wang Q, Liu CX and Wang GJ: Extensive intestinal first-pass elimination and predominant hepatic distribution of berberine explain its low plasma levels in rats. Drug Metab Dispos. 38:1779–1784. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Yue M, Xia Y, Shi C, Guan C, Li Y, Liu R, Wei Z and Dai Y: Berberine ameliorates collagen-induced arthritis in rats by suppressing Th17 cell responses via inducing cortistatin in the gut. FEBS J. 284:2786–2801. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tan XS, Ma JY, Feng R, Ma C, Chen WJ, Sun YP, Fu J, Huang M, He CY, Shou JW, et al: Tissue distribution of berberine and its metabolites after oral administration in rats. PLoS One. 8:e779692013. View Article : Google Scholar : PubMed/NCBI | |
|
Yue M, Tao Y, Fang Y, Lian X, Zhang Q, Xia Y, Wei Z and Dai Y: The gut microbiota modulator berberine ameliorates collagen-induced arthritis in rats by facilitating the generation of butyrate and adjusting the intestinal hypoxia and nitrate supply. FASEB J. 33:12311–12323. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Yao Y and Ding CZ: A combination of sinomenine and methotrexate reduces joint damage of collagen induced arthritis in rats by modulating osteoclast-related cytokines. Int Immunopharmacol. 18:135–141. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Liu W, Zhang Y, Zhu W, Ma C, Ruan J, Long H and Wang Y: Sinomenine inhibits the progression of rheumatoid arthritis by regulating the secretion of inflammatory cytokines and Monocyte/macrophage subsets. Front Immunol. 9:22282018. View Article : Google Scholar : PubMed/NCBI | |
|
Huang RY, Pan HD, Wu JQ, Zhou H, Li ZG, Qiu P, Zhou YY, Chen XM, Xie ZX, Xiao Y, et al: Comparison of combination therapy with methotrexate and sinomenine or leflunomide for active rheumatoid arthritis: A randomized controlled clinical trial. Phytomedicine. 57:403–410. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chen JJ, Sun Y and Huang CB: Clinical observation of sinomenine hydrochloride in the treat-to-target therapy of rheumatoid arthritis and patient-reported outcomes. J Chin Med Materials. 228–231. 2025. | |
|
Xiang G, Gao M, Qin H, Shen X, Huang H, Hou X and Feng Z: Benefit-risk assessment of traditional Chinese medicine preparations of sinomenine using multicriteria decision analysis (MCDA) for patients with rheumatoid arthritis. BMC Complement Med Ther. 23:372023. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, He L, Hu Y, Duan H, Li X, Tan S, Zou M, Gu C, Zeng X, Yu L, et al: Sinomenine suppresses osteoclast formation and Mycobacterium tuberculosis H37Ra-induced bone loss by modulating RANKL signaling pathways. PLoS One. 8:e742742013. View Article : Google Scholar : PubMed/NCBI | |
|
Chen DP, Wong CK, Leung PC, Fung KP, Lau CB, Lau CP, Li EK, Tam LS and Lam CW: Anti-inflammatory activities of Chinese herbal medicine sinomenine and Liang Miao San on tumor necrosis factor-α-activated human fibroblast-like synoviocytes in rheumatoid arthritis. J Ethnopharmacol. 137:457–468. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Tong B, Yu J, Wang T, Dou Y, Wu X, Kong L, Dai Y and Xia Y: Sinomenine suppresses collagen-induced arthritis by reciprocal modulation of regulatory T cells and Th17 cells in gut-associated lymphoid tissues. Mol Immunol. 65:94–103. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Liao H, Ochani M, Justiniani M, Lin X, Yang L, Al-Abed Y, Wang H, Metz C, Miller EJ, et al: Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nat Med. 10:1216–1221. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Yi L, Luo JF, Xie BB, Liu JX, Wang JY, Liu L, Wang PX, Zhou H and Dong Y: α7 nicotinic acetylcholine receptor is a novel mediator of sinomenine Anti-Inflammation effect in macrophages stimulated by lipopolysaccharide. Shock. 44:188–195. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Hayashi S, Hamada T, Zaidi SF, Oshiro M, Lee J, Yamamoto T, Ishii Y, Sasahara M and Kadowaki M: Nicotine suppresses acute colitis and colonic tumorigenesis associated with chronic colitis in mice. Am J Physiol Gastrointest Liver Physiol. 307:G968–G978. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yue M, Zhang X, Dou Y, Wei Z, Tao Y, Xia Y and Dai Y: Gut-sourced vasoactive intestinal polypeptide induced by the activation of α7 nicotinic acetylcholine receptor substantially contributes to the anti-inflammatory effect of sinomenine in collagen-induced arthritis. Front Pharmacol. 9:6752018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Chen L, Sun X, Yang Q, Wan L and Guo C: Matrine: A promising natural product with various pharmacological activities. Front Pharmacol. 11:5882020. View Article : Google Scholar : PubMed/NCBI | |
|
Ao L, Gao H, Jia L, Liu S, Guo J, Liu B and Dong Q: Matrine inhibits synovial angiogenesis in collagen-induced arthritis rats by regulating HIF-VEGF-Ang and inhibiting the PI3K/Akt signaling pathway. Mol Immunol. 141:13–20. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Niu Y, Dong Q and Li R: Matrine regulates Th1/Th2 cytokine responses in rheumatoid arthritis by attenuating the NF-κB signaling. Cell Biol Int. 41:611–621. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zou Y, Li Q, Liu D, Li J, Cai Q, Li C, Zhao Q and Xu W: Therapeutic effects of matrine derivate MASM in mice with collagen-induced arthritis and on fibroblast-like synoviocytes. Sci Rep. 7:24542017. View Article : Google Scholar : PubMed/NCBI | |
|
Cao G, Mao Z, Niu T, Wang P, Yue X, Wang X, Ma A and Zhang Y: Oxymatrine ameliorates rheumatoid arthritis by regulation of Tfr/Tfh cell balance via the TLR9-MyD88-STAT3 signaling pathway. J Sci Food Agric. 103:6017–6024. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang G, Liu B, Zeng Z, Chen Q, Feng Y and Ning X: Oxymatrine hydrazone (OMTH) synthesis and its protective effect for rheumatoid arthritis through downregulation of MEK/NF-κB pathway. Environ Toxicol. 36:2448–2453. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Cao G, Li J, Mao Z and Zhang Y: Oxymatrine alleviates Collagen-induced arthritis in mice by regulating the immune balance of T cells. Molecules. 28:58792023. View Article : Google Scholar : PubMed/NCBI | |
|
Shi F, Li S and Shi H: Oxymatrine modulates immune balance in rheumatoid arthritis and its effect on patients' inflammatory profile. Hainan Med J. 29:2997–3000. 2018. | |
|
Li W and Zhang SN: Effect of oxymatrine on immune balance and its influence on the inflammatory status in patients with rheumatoid arthritis. China Modern Med. 26:18–21. 2019. | |
|
Kacena C: Effects of the curcuminoid and Non-curcuminoid compounds of turmeric on the gut microbiome and inflammation: Potential use in the treatment and prevention of disease. Nutr Rev. 83:1771–1783. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Hasanzadeh S, Read MI, Bland AR, Majeed M, Jamialahmadi T and Sahebkar A: Curcumin: An inflammasome silencer. Pharmacol Res. 159:1049212020. View Article : Google Scholar : PubMed/NCBI | |
|
Kou H, Huang L, Jin M, He Q, Zhang R and Ma J: Effect of curcumin on rheumatoid arthritis: A systematic review and meta-analysis. Front Immunol. 14:11216552023. View Article : Google Scholar : PubMed/NCBI | |
|
Chandran B and Goel A: A randomized, pilot study to assess the efficacy and safety of curcumin in patients with active rheumatoid arthritis. Phytother Res. 26:1719–1725. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Pourhabibi-Zarandi F, Rafraf M, Zayeni H, Asghari-Jafarabadi M and Ebrahimi AA: The efficacy of curcumin supplementation on serum total antioxidant capacity, malondialdehyde, and disease activity in women with rheumatoid arthritis: A randomized, double-blind, placebo-controlled clinical trial. Phytother Res. 38:3552–3563. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Pourhabibi-Zarandi F, Rafraf M, Zayeni H, Asghari-Jafarabadi M and Ebrahimi AA: Effects of curcumin supplementation on metabolic parameters, inflammatory factors and obesity values in women with rheumatoid arthritis: A randomized, double-blind, placebo-controlled clinical trial. Phytother Res. 36:1797–1806. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Amalraj A, Varma K, Jacob J, Divya C, Kunnumakkara AB, Stohs SJ and Gopi S: A novel highly bioavailable curcumin formulation improves symptoms and diagnostic indicators in rheumatoid arthritis patients: A randomized, double-blind, placebo-controlled, two-dose, three-arm, and parallel-group study. J Med Food. 20:1022–1030. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yang KY, Lin LC, Tseng TY, Wang SC and Tsai TH: Oral bioavailability of curcumin in rat and the herbal analysis from Curcuma longa by LC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci. 853:183–189. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Jang DJ, Kim ST, Oh E and Lee K: Enhanced oral bioavailability and antiasthmatic efficacy of curcumin using redispersible dry emulsion. Biomed Mater Eng. 24:917–930. 2014.PubMed/NCBI | |
|
Kloesch B, Becker T, Dietersdorfer E, Kiener H and Steiner G: Anti-inflammatory and apoptotic effects of the polyphenol curcumin on human fibroblast-like synoviocytes. Int Immunopharmacol. 15:400–405. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Huang G, Xu Z, Huang Y, Duan X, Gong W, Zhang Y, Fan J and He F: Curcumin protects against collagen-induced arthritis via suppression of BAFF production. J Clin Immunol. 33:550–557. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Wu X, Wei Z, Dou Y, Zhao D, Wang T, Bian D, Tong B, Xia Y, Xia Y and Dai Y: Oral curcumin has anti-arthritic efficacy through somatostatin generation via cAMP/PKA and Ca(2+)/CaMKII signaling pathways in the small intestine. Pharmacol Res. 95-96:71–81. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Javadi F, Ahmadzadeh A, Eghtesadi S, Aryaeian N, Zabihiyeganeh M, Rahimi Foroushani A and Jazayeri S: The effect of quercetin on inflammatory factors and clinical symptoms in women with rheumatoid arthritis: A Double-blind, randomized controlled trial. J Am Coll Nutr. 36:9–15. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Javadi F, Eghtesadi S, Ahmadzadeh A, Aryaeian N, Zabihiyeganeh M, Foroushani AR and Jazayeri S: The effect of quercetin on plasma oxidative status, C-reactive protein and blood pressure in women with rheumatoid arthritis. Int J Prev Med. 5:293–301. 2014.PubMed/NCBI | |
|
Tang M, Zeng Y, Peng W, Xie X, Yang Y, Ji B and Li F: Pharmacological aspects of natural quercetin in rheumatoid arthritis. Drug Des Devel Ther. 16:2043–2053. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Tao T, Yao H, Zheng H, Wang F and Gao Y: Mechanism of action of quercetin in rheumatoid arthritis models: Meta-analysis and systematic review of animal studies. Inflammopharmacology. 31:1629–1645. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cojocaru M, Cojocaru IM, Silosi I and Vrabie CD: Gastrointestinal manifestations in systemic autoimmune diseases. Maedica (Bucur). 6:45–51. 2011.PubMed/NCBI | |
|
Souza ID, Ribeiro JS, Bersani-Amado CA and Zanoni JN: Analysis of myosin-V immunoreactive myenteric neurons from arthritic rats. Arq Gastroenterol. 48:205–210. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Piovezana Bossolani GD, Silva BT, Colombo Martins Perles JV, Lima MM, Vieira Frez FC, Garcia de Souza SR, Sehaber-Sierakowski CC, Bersani-Amado CA and Zanoni JN: Rheumatoid arthritis induces enteric neurodegeneration and jejunal inflammation, and quercetin promotes neuroprotective and anti-inflammatory actions. Life Sci. 238:1169562019. View Article : Google Scholar : PubMed/NCBI | |
|
Silva BTD, Martins-Perles JVC, Bossolani GDP, Lima MM, Sehaber-Sierakowski CC, Gremaschi LB, Cunha JPSE, Bersani-Amado CA and Zanoni JN: Quercetin and ibuprofen combination displayed anti-inflammatory effects and also extenuates the enteric neurons damage of arthritic rats. An Acad Bras Cienc. 96 (Suppl 1):e202302442024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Yan B, Wang Z, Li M and Zhao W: Natural polysaccharides with immunomodulatory activities. Mini Rev Med Chem. 20:96–106. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ho Do M, Seo YS and Park HY: Polysaccharides: Bowel health and gut microbiota. Crit Rev Food Sci Nutr. 61:1212–1224. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Luo J, Yang Q, Jiang W, Liu Y, Hu Q and Peng X: The interaction between Angelica sinensis polysaccharide ASP-2pb and specific gut bacteria alleviates rheumatoid arthritis in rats. Int J Biol Macromol. 301:1404732025. View Article : Google Scholar : PubMed/NCBI | |
|
Lim H, Min DS, Yun HE, Kim KT, Sun YN, Dat LD, Kim YH and Kim HP: Impressic acid from Acanthopanax koreanum, possesses matrix metalloproteinase-13 down-regulating capacity and protects cartilage destruction. J Ethnopharmacol. 209:73–81. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Takahashi Y, Tanaka M, Murai R, Kuribayashi K, Kobayashi D, Yanagihara N and Watanabe N: Prophylactic and therapeutic effects of Acanthopanax senticosus Harms extract on murine collagen-induced arthritis. Phytother Res. 28:1513–1519. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Kang MC, Kim SY, Kim EA, Lee JH, Kim YS, Yu SK, Chae JB, Choe IH, Cho JH and Jeon YJ: Antioxidant activity of polysaccharide purified from Acanthopanax koreanum Nakai stems in vitro and in vivo zebrafish model. Carbohydr Polym. 127:38–46. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Fu J, Fu J, Yuan J, Zhang N, Gao B, Fu G, Tu Y and Zhang Y: Anti-diabetic activities of Acanthopanax senticosus polysaccharide (ASP) in combination with metformin. Int J Biol Macromol. 50:619–623. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Lee JB, Tanikawa T, Hayashi K, Asagi M, Kasahara Y and Hayashi T: Characterization and biological effects of two polysaccharides isolated from Acanthopanax sciadophylloides. Carbohydr Polym. 116:159–166. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Liu A, Zhang M, Wu Y, Zhang C, Zhang Q, Su X, Zhu X, Shi W, Liu J, Zhang Y, et al: ASPS exhibits Anti-rheumatic effects by reprogramming gut microbiota and increasing serum γ-Glutamylcysteine Level. Adv Sci (Weinh). 10:e22056452023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu L, Sha XY, Wu YN, Chen MT and Zhong JX: Lycium barbarum polysaccharides protects retinal ganglion cells against oxidative stress injury. Neural Regen Res. 15:1526–1531. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ran L, Chen F, Zhang J, Mi J, Lu L, Yan Y and Cao Y: Antitumor effects of pollen polysaccharides from Chinese wolfberry on DU145 cells via the PI3K/AKT pathway in vitro and in vivo. Int J Biol Macromol. 152:1164–1173. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Gao M and Han T: Lycium barbarum polysaccharides ameliorate intestinal barrier dysfunction and inflammation through the MLCK-MLC signaling pathway in Caco-2 cells. Food Funct. 11:3741–3748. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Fu YW, Peng YF, Huang XD, Yang Y, Huang L, Xi Y, Hu ZF, Lin S, So KF and Ren CR: Lycium barbarum polysaccharide-glycoprotein preventative treatment ameliorates aversive. Neural Regen Res. 16:543–549. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kalinkovich A and Livshits G: A cross talk between dysbiosis and Gut-associated immune system governs the development of inflammatory arthropathies. Semin Arthritis Rheum. 49:474–484. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Lv J, Yang B, Liu F, Tian Z, Cai Y, Yang D, Ouyang J, Sun F, Shi Y and Xia P: Lycium barbarum polysaccharide attenuates type II collagen-induced arthritis in mice. Int J Biol Macromol. 78:318–323. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Cai S, Sun J and Wei X: Lycium barbarum polysaccharide inhibits NF-κB pathway to reduce the level of inflammatory cytokines in osteoarthritis chondrocytes. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 34:989–993. 2018.(In Chinese). PubMed/NCBI | |
|
Lai W, Wang C, Lai R, Peng X and Luo J: Lycium barbarum polysaccharide modulates gut microbiota to alleviate rheumatoid arthritis in a rat model. NPJ Sci Food. 6:342022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Liu L, Luo J and Peng X: Metabolites from specific intestinal bacteria in vivo fermenting Lycium barbarum polysaccharide improve collagenous arthritis in rats. Int J Biol Macromol. 226:1455–1467. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cao W, Liu J, Dai Y, Zhou Y, Li R and Yu P: Bibliometric analysis of marine traditional chinese medicine in pharmacopoeia of the People's Republic of China: Development, differences, and trends directions. Evid Based Complement Alternat Med. 2022:39719672022. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng Q, Mu X, Pan S, Luan R and Zhao P: Ephedrae herba: A comprehensive review of its traditional uses, phytochemistry, pharmacology, and toxicology. J Ethnopharmacol. 307:1161532023. View Article : Google Scholar : PubMed/NCBI | |
|
Ma Y, Wei X, Peng J, Wei F, Wen Y, Liu M, Song B, Wang Y, Zhang Y and Peng T: Ephedra sinica polysaccharide regulate the anti-inflammatory immunity of intestinal microecology and bacterial metabolites in rheumatoid arthritis. Front Pharmacol. 15:14146752024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Z, Li XP, Li ZJ, Xu L and Li XM: Reduced hepatotoxicity by total glucosides of paeony in combination treatment with leflunomide and methotrexate for patients with active rheumatoid arthritis. Int Immunopharmacol. 15:474–477. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang N, Li XM, Zhang MJ, Zhao DB, Zhu P, Zuo XX, Yang M, Su Y, Li ZG, Chen Z and Li XP: Total glucosides of paeony can reduce the hepatotoxicity caused by Methotrexate and Leflunomide combination treatment of active rheumatoid arthritis. Int Immunopharmacol. 28:802–807. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y and Xing HY: Clinical observation on effect of total glucosides of paeony combined with methotrexate on rheumatoid arthritis. Zhongguo Zhong Xi Yi Jie He Za Zhi. 27:839–840. 2007.(In Chinese). PubMed/NCBI | |
|
Luo J, Jin DE, Yang GY, Zhang YZ, Wang JM, Kong WP and Tao QW: Total glucosides of paeony for rheumatoid arthritis: A systematic review of randomized controlled trials. Complement Ther Med. 34:46–56. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Fei F, Yang H, Peng Y, Wang P, Wang S, Zhao Y, Huang J, Yu X, Feng S, Sun R, et al: Sensitive analysis and pharmacokinetic study of the isomers paeoniflorin and albiflorin after oral administration of total glucosides of white paeony capsule in rats. J Chromatogr B Analyt Technol Biomed Life Sci. 1022:30–37. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Xia SM, Shen R, Sun XY, Shen LL, Yang YM, Ke Y, Wang Y, Chen DY and Han XM: Development and validation of a sensitive liquid chromatography-tandem mass spectrometry method for the determination of paeoniflorin in rat brain and its application to pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci. 857:32–39. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W and Dai SM: Mechanisms involved in the therapeutic effects of Paeonia lactiflora Pallas in rheumatoid arthritis. Int Immunopharmacol. 14:27–31. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Sun B, Dai G, Bai Y, Zhang W, Zhu L, Chu J, Pan R and Ju W: Determination of paeoniflorin in rat plasma by Ultra-high performance liquid Chromatography-tandem mass spectrometry and its application to a pharmacokinetic study. J Chromatogr Sci. 55:1006–1012. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tong L, Wan M, Zhou D, Gao J, Zhu Y and Bi K: LC-MS/MS determination and pharmacokinetic study of albiflorin and paeoniflorin in rat plasma after oral administration of Radix Paeoniae Alba extract and Tang-Min-Ling-Wan. Biomed Chromatogr. 24:1324–1331. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Peng J, Lu X, Xie K, Xu Y, He R, Guo L, Han Y, Wu S, Dong X, Lu Y, et al: Dynamic alterations in the gut microbiota of Collagen-induced arthritis rats following the prolonged administration of total glucosides of paeony. Front Cell Infect Microbiol. 9:2042019. View Article : Google Scholar : PubMed/NCBI | |
|
Bao BL and Wang ZZ: Resource and pharmacognostical study on Aralia echinocaulis. Zhongguo Zhong Yao Za Zhi. 29:508–510. 2004.(In Chinese). PubMed/NCBI | |
|
Li Y, Dai M, Wang L and Wang G: Polysaccharides and glycosides from Aralia echinocaulis protect rats from arthritis by modulating the gut microbiota composition. J Ethnopharmacol. 269:1137492021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Zhang F, Xiao X, Wu Z, Hu Q, Jiang Y, Zhang W, Wei S, Ma X and Zhang X: Tripterygium hypoglaucum (Levl.) hutch and its main bioactive components: recent advances in pharmacological activity, pharmacokinetics and potential toxicity. Front Pharmacol. 12:7153592021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Liu Q, Zhou X, Zhang J, Liu W, Zhao X and Luo N: THH Relieves CIA inflammation by reducing inflammatory-related cytokines. Cell Biochem Biophys. 78:367–374. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Long C, Yang Y, Wang Y, Zhang X, Zhang L, Huang S, Yang D, Qiao X, Yang Y and Guo Y: Role of Glutamine-glutamate/GABA cycle and potential target GLUD2 in alleviation of rheumatoid arthritis by Tripterygium hypoglaucum (levl.) Hutch based on metabolomics and molecular pharmacology. J Ethnopharmacol. 281:1145612021. View Article : Google Scholar : PubMed/NCBI | |
|
Hu J, Ni J, Zheng J, Guo Y, Yang Y, Ye C, Sun X, Xia H, Liu Y and Liu H: Tripterygium hypoglaucum extract ameliorates adjuvant-induced arthritis in mice through the gut microbiota. Chin J Nat Med. 21:730–744. 2023.PubMed/NCBI | |
|
Zheng J, Hu J, Yang Y, Xiong L, Yang H, Zhang Z, Jiang N and Liu H: Suppressive effect of Tripterygium hypoglaucum (Levl.) Hutch extract on rheumatoid arthritis in mice by modulating inflammasome and bile acid metabolism. Biomed Pharmacother. 167:1154942023. View Article : Google Scholar : PubMed/NCBI | |
|
Orwa JA, Jondiko IJ, Minja RJ and Bekunda M: The use of Toddalia asiatica (L) Lam. (Rutaceae) in traditional medicine practice in East Africa. J Ethnopharmacol. 115:257–262. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Sukieum S, Sang-Aroon W and Yenjai C: Coumarins and alkaloids from the roots of Toddalia asiatica. Nat Prod Res. 32:944–952. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yang K, Tong L, Chen C, Zhang P, Pi H, Ruan H and Wu J: Therapeutic effects of extracts from Radix Toddaliae Asiaticae on collagen-induced arthritis in Balb/c mice. J Ethnopharmacol. 146:355–362. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Tong L, Chen T, Chen Z, Zhang P, Pi H, Ruan H and Wu J: Anti-inflammatory activity of omphalocarpin isolated from radix toddaliae asiaticae. J Ethnopharmacol. 155:1553–1560. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yan C and Wu J: Effect of Toddalia asiatica extract combined with miR-483 on proliferation, apoptosis and inflammatory factors expression of osteoarthritis chondrocyte. Pak J Pharm Sci. 33:1333–1340. 2020.PubMed/NCBI | |
|
Qin H, Fu Y, Zhou K, Song H, Fang G, Chen Q and Pang Y: Toddalia asiatica extract attenuates adjuvant-induced arthritis by modulating colon Th17/Treg balance and colony homeostasis. J Ethnopharmacol. 313:1165422023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen BC, He HY, Niu K, Rui K, Huang JG, Xie YQ and Xiao M: Network pharmacology-based approach uncovers the JAK/STAT signaling mechanism underlying Paederia scandens extract treatment of rheumatoid arthritis. Am J Transl Res. 14:5295–5307. 2022.PubMed/NCBI | |
|
Xiao M, Fu X, Ni Y, Chen J, Jian S, Wang L, Li L and Du G: Protective effects of Paederia scandens extract on rheumatoid arthritis mouse model by modulating gut microbiota. J Ethnopharmacol. 226:97–104. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Swindell EP, Hankins PL, Chen H, Miodragovic DU and O'Halloran TV: Anticancer activity of small-molecule and nanoparticulate arsenic(III) complexes. Inorg Chem. 52:12292–12304. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Li C, Zheng Y, Lin Z, Zhang Y and Zhang Z: Inhibition of angiogenesis by arsenic trioxide via TSP-1-TGF-β1-CTGF-VEGF functional module in rheumatoid arthritis. Oncotarget. 8:73529–73546. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Ma Y, Zhang Y, Niu S, Chu M and Zhang Z: Angiogenesis is inhibited by arsenic trioxide through downregulation of the CircHIPK3/miR-149-5p/FOXO1/VEGF functional module in rheumatoid arthritis. Front Pharmacol. 12:7516672021. View Article : Google Scholar : PubMed/NCBI | |
|
Li C, Zhang J, Wang W, Wang H, Zhang Y and Zhang Z: Arsenic trioxide improves Treg and Th17 balance by modulating STAT3 in treatment-naive rheumatoid arthritis patients. Int Immunopharmacol. 73:539–551. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Niu S, Zhu X, Zhang J, Ma Y, Lang X, Luo L, Li W, Zhao Y and Zhang Z: Arsenic trioxide modulates the composition and metabolic function of the gut microbiota in a mouse model of rheumatoid arthritis. Int Immunopharmacol. 111:1091592022. View Article : Google Scholar : PubMed/NCBI | |
|
Kang JI, Hong JY, Choi JS and Lee SK: Columbianadin inhibits cell proliferation by inducing apoptosis and necroptosis in HCT116 colon cancer cells. Biomol Ther (Seoul). 24:320–327. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lim HJ, Lee JH, Choi JS, Lee SK, Kim YS and Kim HP: Inhibition of airway inflammation by the roots of Angelica decursiva and its constituent, columbianadin. J Ethnopharmacol. 155:1353–1361. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Li R, Zhao C, Yao M, Song Y, Wu Y and Wen A: Analgesic effect of coumarins from Radix angelicae pubescentis is mediated by inflammatory factors and TRPV1 in a spared nerve injury model of neuropathic pain. J Ethnopharmacol. 195:81–88. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Su X, Wu B, Zhang W, Ji YH, Wang Q and Tan ZY: Inhibitory effects of columbianadin on nociceptive behaviors in a neuropathic pain model, and on Voltage-gated calcium currents in dorsal root ganglion neurons in mice. Front Pharmacol. 10:15222019. View Article : Google Scholar : PubMed/NCBI | |
|
Chen S, Wang Y, Zhang L, Han Y, Liang C, Wang S, Qi L, Pang X, Li J and Chang Y: Therapeutic effects of columbianadin from Angelicae Pubescentis radix on the progression of collagen-induced rheumatoid arthritis by regulating inflammation and oxidative stress. J Ethnopharmacol. 316:1167272023. View Article : Google Scholar : PubMed/NCBI | |
|
Gracey E, Vereecke L, McGovern D, Fröhling M, Schett G, Danese S, De Vos M, Van den Bosch F and Elewaut D: Revisiting the Gut-joint axis: Links between gut inflammation and spondyloarthritis. Nat Rev Rheumatol. 16:415–433. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
de Winter JJ, van Mens LJ, van der Heijde D, Landewe R and Baeten DL: Prevalence of peripheral and extra-articular disease in ankylosing spondylitis versus non-radiographic axial spondyloarthritis: A meta-analysis. Arthritis Res Ther. 18:1962016. View Article : Google Scholar : PubMed/NCBI | |
|
Guggino G, Mauro D, Rizzo A, Alessandro R, Raimondo S, Bergot AS, Rahman MA, Ellis JJ, Milling S, Lories R, et al: Inflammasome activation in ankylosing spondylitis is associated with gut dysbiosis. Arthritis Rheumatol. 73:1189–1199. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mauro D, Nakamura A, Haroon N and Ciccia F: The gut-enthesis axis and the pathogenesis of Spondyloarthritis. Semin Immunol. 58:1016072021. View Article : Google Scholar : PubMed/NCBI | |
|
Fan J, Jiang T and He D: Advances in the implications of the gut microbiota on the treatment efficacy of disease-modifying anti-rheumatic drugs in rheumatoid arthritis. Front Immunol. 14:11890362023. View Article : Google Scholar : PubMed/NCBI | |
|
Qiao J, Zhang SX, Chang MJ, Cheng T, Zhang JQ, Zhao R, Song S, Liu GY, Chang JS and Li XF: Specific enterotype of gut microbiota predicted clinical effect of methotrexate in patients with rheumatoid arthritis. Rheumatology (Oxford). 62:1087–1096. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Wang Z, Qian H, Tao W, Zhang Y, Hu C, Mao W and Guo Q: Natural medicines of targeted rheumatoid arthritis and its action mechanism. Front Immunol. 13:9451292022. View Article : Google Scholar : PubMed/NCBI | |
|
Han Y, Chen S, Liu C, Sun H, Jia Z, Shi J, Li J and Chang Y: A comprehensive review of natural products in rheumatoid arthritis: Therapeutic potential and mechanisms. Front Immunol. 16:15010192025. View Article : Google Scholar : PubMed/NCBI | |
|
Guo W, Lv G, Yang D, Zhang W, Li N, Hu J, Wu Y, Pi Z and Lin Z: Advances in the effect of gut-joint axis dysfunction on rheumatoid arthritis and the intervention of natural products. Chin J Analytical Chemistry. 52:1003542024. View Article : Google Scholar | |
|
Liang Y, Liu M, Cheng Y, Wang X and Wang W: Prevention and treatment of rheumatoid arthritis through traditional Chinese medicine: Role of the gut microbiota. Front Immunol. 14:12339942023. View Article : Google Scholar : PubMed/NCBI |