Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial hyperplasia, inflammation, and pannus formation. Although conventional therapies are effective, they are often associated with significant side effects. Traditional Chinese medicine (TCM), known for its high safety profile and multitarget therapeutic effects, has shown unique potential in the treatment of RA. In vitro models, including synovial organoids and cell culture systems, serve as crucial platforms for replicating the structure and function of human synovial tissue. Synovial organoids closely mimic the pathological microenvironment of RA, providing an ideal tool for drug screening and efficacy evaluation. In contrast, cell culture models, which offer greater experimental flexibility, allow precise control of conditions to facilitate an in-depth investigation of the roles and mechanisms of individual or interacting cell types in RA pathogenesis. This review first covers the fundamental structure and pathophysiology of the synovium, followed by a detailed discussion of the development and construction techniques of synovial organoids. Building on this foundation, it explores the therapeutic potential of TCM using synovial organoids and cell culture models, highlighting how TCM exerts therapeutic effects by inhibiting synovial hyperplasia and modulating immune responses through its multitarget and multipathway mechanisms. Finally, the review envisions the broad application prospects of integrating synovial organoids and cell culture models with TCM in RA treatment, underscoring the importance of interdisciplinary collaboration and innovation. This study aims to provide a novel perspective on the integration of TCM with modern biotechnological approaches and promote innovative advances in RA therapy.


1. Introduction
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent inflammation, synovial hyperplasia, pannus formation, and progressive destruction of articular cartilage and bone tissue. , The synovium, a critical component of the joint structure, not only provides lubrication and protection but also plays a central role in the pathogenesis of RA. , Key pathological features of RA include aberrant proliferation of synovial cells, excessive secretion of pro-inflammatory cytokines, and the formation of invasive pannus tissue. , Current treatment strategies for RA, such as nonsteroidal anti-inflammatory drugs, disease-modifying antirheumatic drugs, and glucocorticoids, are effective in controlling symptoms but are often associated with significant side effects, particularly with long-term use, which can adversely impact patients' quality of life and treatment compliance. − Although newly developed biological agents and targeted small-molecule drugs have shown impressive therapeutic outcomes, their high cost poses a considerable financial burden on patients and healthcare systems alike. , Therefore, there is an urgent need to develop safe, effective, and affordable therapeutic alternatives for RA.
Traditional Chinese medicine (TCM), known for its favorable safety profile and low incidence of adverse effects, is gaining increasing global attention as a complementary or alternative therapeutic approach. In many Asian countries, the use of herbal medicine for RA has a history spanning thousands of years. This time-honored medical practice, supported by extensive clinical experience and an increasing body of scientific evidence, has demonstrated significant therapeutic potential. , One of the distinguishing features of TCM is its multitarget, multipathway mode of action, allowing it to exert therapeutic effects through diverse biological mechanisms. , Specifically, many Chinese herbal formulations possess strong antirheumatic properties, including anti-inflammatory, analgesic, and immunomodulatory effects, as well as the ability to inhibit abnormal synovial cell proliferation and pathological angiogenesis. , Numerous herbs and their active constituents have shown promising efficacy in suppressing synovial hyperplasia, providing novel insights and potential breakthroughs in RA therapy.
Organoids are three-dimensional cellular structures formed through intrinsic genetic reprogramming mechanisms, primarily initiated by tightly regulated soluble biochemical niche signals derived from either suspension cultures or three-dimensional extracellular matrix (ECM)-like systems that closely mimic in vivo environments. These signals are essential for promoting cellular self-organization and the development of tissue-specific organ-like structures. − In recent years, organoids have attracted considerable attention in biomedical research due to their ability to replicate the complex architecture and function of human organs in vitro. Their high fidelity in modeling organ development, disease progression, and drug response makes them powerful platforms for investigating organogenesis, elucidating disease mechanisms, and conducting drug screening and evaluation studies. ,
Among these, synovial organoids have emerged as critical models for recapitulating the structural and functional features of human synovial tissue in vitro. In recent years, they have shown substantial progress in the fields of disease modeling, pharmacological research, and tissue engineering. − The establishment of physiologically functional synovial organoids not only enhances our understanding of RA pathogenesis but also offers invaluable experimental systems for drug development, therapeutic evaluation, and tissue repair. TCM has demonstrated notable therapeutic effects in such models. For example, Danggui Buxue Decoction (DBD) has been shown to target SFRP4 and modulate the Wnt signaling pathway, thereby effectively suppressing inflammation in RA fibroblast-like synoviocytes (RA-FLS) and slowing disease progression. Similarly, Baihu Guizhi Decoction (BHGZD) inhibits synovial cell hyperproliferation and inflammation by suppressing Toll-like receptor 4-mediated NLRP3 inflammasome activation, offering promising support for symptom relief in RA patients. In an effort to further elucidate the mechanisms of TCM, researchers have developed an innovative in vitro coculture model in which peripheral blood mononuclear cells (PBMCs) from RA patients are used to stimulate FLS, mimicking the pathological immune environment of RA. Using this model, the TCM formula Huangqin Qingre Chubi Capsule (HQC) was found to exert significant anti-inflammatory effects, likely through inhibition of the hsa_circ_0091685/EIF4A3/IL-17 regulatory axis, thereby contributing to disease amelioration. Moreover, recent studies have identified sesamol as a novel p53 stabilizer with therapeutic potential in RA. Notably, sesamol not only alleviates RA symptoms but also inhibits the growth of synovial organoids, providing a new direction for TCM applications in RA management. Collectively, these findings highlight the promising results and future potential of TCM in single-cell, coculture, and synovial organoid-based models. They underscore the growing importance of TCM in RA treatment and suggest a substantial opportunity for its further development.
This article aims to explore the potential of synovial organoids and cell culture models as innovative platforms for applying TCM in the treatment of RA. It provides a comprehensive review of the developmental trajectory and construction methodologies of organoids and discusses in depth the therapeutic applications and advantages of TCM in these systems. Through this investigation, we seek to offer new insights and perspectives on the integration of TCM with modern biotechnological approaches for advancing RA therapy.
2. Basic Structure and Pathophysiology of the Synovium
The human synovium, a membranous tissue lining the inner surface of articulating joints, plays a crucial role in maintaining joint stability and facilitating diverse physiological functions. The synovial membrane (SM) is composed of two distinct layers: the intimal layer and the subintimal layer. , The intimal layer, forming the innermost portion of the SM, typically consists of one or two layers of macrophage-like synoviocytes (MLS, also referred to as type A synovial cells) and FLS (type B synovial cells). These cells are primarily responsible for secreting synovial fluid (SF), which lubricates joint cartilage, supports chondrocyte function, and supplies essential nutrients. Notably, type B synoviocytes synthesize hyaluronic acid, a critical glycosaminoglycan that significantly contributes to the viscosity and lubricating properties of SF. , Beneath the intimal layer lies the subintimal region of the SM, consisting of two to three layers of synovial cells supported by a loose connective tissue matrix abundant in fibroblasts.
Synovial dysfunction is closely associated with the initiation and progression of joint disorders with synovitis being one of the most prominent manifestations. Synovitis is characterized by synovial hyperplasia, excessive cell proliferation, and massive infiltration of immune cells. These pathological alterations are accompanied by elevated levels of pro-inflammatory mediatorssuch as cytokines, nitric oxide (NO), and prostaglandin E2 (PGE2)secreted by resident synovial cells. In the context of inflammatory arthritis, particularly RA, the synovium is regarded as the primary site of pathology. Persistent synovitis plays a central role in driving secondary damage to adjacent cartilage and bone structures. , During RA flareups, the synovium undergoes two major pathological changes. First, activation of FLS and MLS leads to a marked expansion of the intimal layer. This expansion facilitates adhesion and proliferation of inflammatory mediators and matrix-degrading enzymes, including selectins, integrins, and members of the immunoglobulin superfamily. Second, the infiltration of activated immune cells into the SM further amplifies the local inflammatory milieu, contributing to persistent inflammation and disease progression (Figure ).
1.
Basic structure and pathophysiology of the synovium.
3. Evolution of Synovial Organoid Models
3.1. From Monolayer Culture to Coculture and 3D Organoid Development
The evolution of synovial research models has undergone a significant transitionfrom basic to advanced systems and from two-dimensional (2D) to three-dimensional (3D) configurations. While each model offers distinct advantages, no single approach can fully capture the complex pathological landscape of synovial diseases (Figure ). Monolayer cultures and coculture systems remain widely used due to their operational simplicity and cost-effectiveness. Monolayer cultures, in particular, are well-suited for large-scale experiments and circumvent the technical challenges associated with coculturing heterogeneous cell populations. However, their limited ability to replicate tissue complexity and dynamic intercellular interactions often restrict investigations to isolated components rather than the system as a whole. To overcome these limitations, 3D culture systems and tissue explant models have been developed, providing more physiologically relevant platforms for studying synovial diseases such as RA. These models enable the simulation of intercellular communication and stromal interactions more accurately than 2D systems, making them valuable tools for drug screening and mechanistic research. As a result, they have significantly improved the translational efficiency of preclinical studies. ,
2.
Comparison of synovial organoids to other preclinical models.
With growing insight, researchers have recognized that 2D models, such as cultures of synovial fibroblasts or commonly used cell lines such as RAW 264.7 and K4IM, are insufficient to fully elucidate disease mechanisms. These models often fail to incorporate critical components such as synovial macrophages and ECM elements. , Consequently, newer strategies have emerged to simulate 3D tissue structures. These include the direct use of tissue explants and the construction of cell-scaffold composites, both of which represent promising research directions. Notably, the synovial joint capsule (SJC), which serves as a conduit between intra- and extra-articular environments and contains functionally important synovial tissue, has become a valuable model for investigating glycoproteinase hydrolysis mechanisms. However, anatomical variability among SJC samples presents challenges to standardization and reproducibility in experimental studies. , Interestingly, coculturing synovial and capsular tissues with articular cartilage has been shown to enhance glycosidase activity, offering insights into the complex interplay within the joint microenvironment. A major breakthrough was achieved by Stefani and colleagues, who developed a highly biomimetic, tissue-engineered synovial model by embedding bovine synovial fibroblasts and macrophage-like cells in Matrigel. In this system, Matrigel functions as a regulatory ECM scaffold that guides cellular behavior and mimics inflammatory responses. This model provides a powerful experimental platform for investigating the role of synovial cells in joint homeostasis and inflammation.
In conclusion, the progression from monolayer culture to coculture systems and 3D organoid models has enabled increasingly realistic simulations of the synovial microenvironment. These advancements have greatly enhanced our capacity to investigate disease mechanisms and develop novel therapeutic strategies.
3.2. Construction of 3D Synovial Organoids
3.2.1. Starting Cell Types
In the development of synovial organoids, stem cells play a central role as the foundational cell source owing to their exceptional self-renewal capacity and multipotent differentiation potential. Among these, adult stem cells (ASCs)especially when cultured as spheroidshave emerged as promising candidates for organoid generation, as they can faithfully recapitulate tissue morphogenesis and perform at least one function characteristic of the target tissue or organ. Within the context of musculoskeletal regenerative medicine, synovial-derived mesenchymal stem cells (MSCs) have attracted growing interest due to their distinctive biological characteristics. These cells reside within a specialized synovial niche, where local fibroblasts contribute to ECM production and joint homeostasis by secreting essential biomolecules. To enhance the precision of patient-specific therapeutic modeling, researchers primarily utilize two sources of fibroblasts in synovial organoid construction: fibroblasts directly derived from synovial MSCs and those generated via induced pluripotent stem cell (iPSC) technology. Each cell source presents unique advantages and application prospects.
Synovial-derived MSCs (SMSCs), a relatively recent addition to the MSC family, have garnered attention for their robust proliferative capacity and pronounced chondrogenic potential. When seeded at high densities, SMSCs rapidly expand, making them suitable for clinical-scale applications. Moreover, under 3D culture conditions, SMSCs demonstrate superior biocompatibility and enhanced chondrogenic differentiation, establishing a solid foundation for synovial organoid development. , Concurrently, the differentiation behavior of fibroblastscritical for maintaining tissue architecture and functionhas become a key focus in organoid research. , Recent studies have highlighted the pivotal role of Notch signaling, mediated by vascular endothelial cells, in regulating the differentiation of CD90+ fibroblast subpopulations, offering new mechanistic insights into fibroblast behavior within synovial organoid systems. iPSCs, another essential fibroblast source, not only provide a renewable and scalable cell population but also originate from patients with diverse clinical backgrounds. This makes them particularly suitable for disease modeling and the development of personalized therapies. , iPSC-derived constructs offer significant potential in high-throughput drug screening and in vitro disease modeling, thus accelerating progress in precision medicine and therapeutic innovation. In one notable study, researchers constructed a 3D synovial organoid model by coculturing iPSCs with MSC-derived fibroblasts and macrophages while introducing polyethylene particles to simulate mechanical and pathological stimuli. This system successfully recapitulated native tissue architecture and physiological responses under a variety of clinical conditions, providing a robust platform for diagnostic evaluation and the development of novel therapeutic strategies (Figure A).
3.
Current understanding and research progress on synovial organoids. (A) Inducing differentiation of pluripotent stem cells and mesenchymal stem cells into the synovium. (B) Using poor culture medium and heterogeneous culture medium components in organoid culture. (C) Various methods and scans have been used to construct synovial organoids.
3.2.2. Culture Environment and Signaling Factors
In the construction of synovial organoids, the physical and biochemical characteristics of the culture environment are of critical importance (Figure B). In recent years, significant progress in tissue engineeringparticularly in the development of 3D microsystems, organoid platforms, and microfluidic organ-on-a-chip technologieshas enabled increasingly sophisticated exploration of the complex structure and function of synovial tissue. − Utilizing 3D synovial organoid culture systems, researchers have been able to investigate cellular mechanisms involved in synovial tissue formation and inflammatory remodeling, highlighting the pivotal role of FLS in driving synovial inflammation under physiologically relevant conditions. , Innovative models of synovial angiogenesis based on a spheroid culture have also been developed. These models employ growth factor stimulation to recapitulate the complex process of angiogenesis and integrate monocyte-derived macrophages to simulate in vivo intercellular interactions, thereby deepening our understanding of immune and vascular contributions to synovial inflammation. − Since 2006, researchers such as Kiener's group have introduced 3D synovial micromass organoid models that allow for the spontaneous formation of synovial-like structures. These models have been successfully employed to investigate the regulatory roles of FLS in inflammatory cascades. , To further dissect the interplay between synovial and cartilage tissues, hydrogel-based 3D coculture models have been proposed. These systems aim to mimic intertissue communication and provide insight into joint pathophysiology by replicating the dynamic crosstalk between synovial and chondral organoids. Technological advances have also led to the emergence of microphysiological systems and organ-on-a-chip platforms, offering integrated, dynamic, and high-throughput environments for modeling complex aspects of arthritic disease. These platforms provide novel tools for elucidating disease mechanisms and facilitating therapeutic development. A notable example includes the work of Ertl et al., who established a coculture model involving human FLS (using the synovial sarcoma cell line SW982), murine osteoclast precursors (RAW264.7), and bone marrow-derived MSCs (BMSCs). This system was employed to investigate how FLS migration and invasion contribute to bone erosion in RA, offering new insights into the mechanisms underlying joint destruction. In synovial organoid cultures, the use of solid ECM materials is fundamental for supporting 3D morphogenesis and cellular organization. ECM components such as gelatin and alginate, which closely mimic native ECM properties, have demonstrated excellent biocompatibility and structural versatility for building 3D bioscaffolds. , A recent study effectively leveraged these materials in combination with 3D printing technology to construct an in vitro model that recapitulates the pathological features of RA-associated pannus formation, underscoring the immense potential of 3D biomanufacturing in biomedical applications.
The formation of synovial organoids depends on specific signaling factors that initiate and guide the self-assembly of cellular developmental pathways at defined temporal stages. Although multiple construction protocols have been reported, a unified standard for optimal practices has yet to be established. One study employing a 3D in vitro organ culture system of FLS identified cadherin-11 as a critical mediator of FLS adhesion. Cadherin-11 was shown to interact with the actin cytoskeleton, promoting structural remodeling and playing a decisive role in the organization and stabilization of synovial-like structures in vitro. In efforts to optimize scaffold design for synovial organoid construction, researchers have investigated how architectural features of biomaterials influence the cellular mechanical microenvironment. Hydrogels, as cellular carriers, were found to more evenly distribute and transmit mechanical forces compared to porous scaffolds. − Moreover, coculturing FLS with human umbilical vein endothelial cells (HUVECs) and monocyte-derived macrophages within a 3D collagen-based scaffoldsupplemented with vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (bFGF)significantly promoted spheroid growth and increased the density of both FLS and ECs in the growth region, in contrast to unstimulated controls. To further enhance the structural and functional fidelity of engineered synovial tissue, researchers have adopted advanced volumetric extrusion bioprinting techniques. By utilizing gelatin methacryloyl (GelMA) as a bioink, they successfully fabricated bioartificial synovial tissue constructs that recapitulate key features of native tissue and are well-suited for in vitro experimentation (Figure C).
In conclusion, the construction of synovial organoids is a multidisciplinary and complex endeavor involving the selection of appropriate starting cell types, precise control of the culture environment, regulation of key signaling pathways, and incorporation of cutting-edge technologies. These advances provide powerful platforms for investigating synovial inflammation and offer a robust foundation for developing targeted therapeutic strategies for diseases such as RA.
4. Involvement of Synovial Organoids and Cell Culture Models in TCM Treatment for RA
4.1. Single-Cell Cultivation in TCM Therapy for RA
The introduction of single-cell cultivation technology has significantly advanced our ability to mimic the microenvironment of synovial cells in RA, allowing for precise investigation of the mechanisms by which individual compounds and complex formulations from TCM influence cell proliferation, differentiation, and function. This approach not only deepens our understanding of RA pathogenesis but also provides valuable experimental evidence for identifying molecular targets of TCM therapies.
Through the application of single-cell cultivation systems, the active ingredients and pharmacological mechanisms of various TCM agents in RA treatment have been progressively elucidated. For instance, Forsythiaside A has shown potent antimigratory and anti-inflammatory effects in an in vitro RA model using IL-1β-stimulated MH7A cells. Its activity is mediated through modulation of the JAK/STAT signaling pathway, identifying novel molecular targets for cell-level RA therapy. Similarly, Qufeng Zhitong Capsule (QFZTC) directly targets RA cells, significantly reducing cell viability and alleviating inflammatory responses, thereby underscoring the unique regulatory capacity of TCM in modulating RA cellular behavior. Hedyotis diffusa Willd (HDW) has been shown to mitigate RA progression by targeting the MMP9/miR-204–5p/MIAT axis, providing further evidence of the multipathway, multitarget potential of TCM in disease intervention. Siegesbeckiae Herba (SBH) offers a novel strategy for inflammatory control by promoting apoptosis in RA-FLS and reducing immune-mediated inflammation. Xinfeng Capsules (XFC), a well-characterized TCM formulation, suppress inflammation and oxidative stress by upregulating LINC00638 and activating the Nrf2/HO-1 pathway. XFC also modulates the expression of lncRNA MAPKAPK5-AS1 to induce apoptosis in RA-FLS, expanding the application spectrum of TCM in RA therapy. , Triptolide further demonstrates efficacy by significantly inhibiting RA-FLS proliferation and inflammation via the hsa-circ-0003353/microRNA-31–5p/CDK1 axis and downregulating ENST00000619282, introducing novel molecular avenues for RA intervention. , Complex formulations such as Huangqin Qingre Chubi Capsule (HQC) have been shown to delay RA progression through regulation of the CUL4B/Wnt pathway, illustrating the systemic therapeutic effects of TCM. Astragaloside, another TCM-derived compound, inhibits RA-FLS proliferation in rat models by modulating the lncRNA LOC100912373/miR-17–5p/PDK1 axis, providing strong support for its application in precision medicine approaches. Notably, combination therapies such as mangiferin and cinnamic acid, key components of Baihu-Guizhi Decoction (BHGZD), have been found to inhibit TLR4/NF-κB/NLRP3-mediated pyroptosis. Berberine also exhibits significant antiproliferative and antiadhesive effects on RA-FLS through modulation of the RAS/MAPK/FOXO/HIF-1 signaling pathways, providing yet another promising strategy for RA management. For a detailed summary of research findings regarding the role of single-cell cultivation technology in TCM-based RA treatment, refer to Table .
1. Synovial Organoid Involvement in TCM Treatment of RA.
| evolution | reference | cell (model) | pathophysiological mechanism | index | axis | effects | TCM |
|---|---|---|---|---|---|---|---|
| monocell culture | [] | MH7A | inflammation | TNF-α, IL-6, IL-8 | JAK/STAT | Forsythiaside A exhibits antimigratory and anti-inflammatory effects in an RA in vitro model (IL-1β-stimulated MH7A cells) by modulating the JAK/STAT pathway | forsythiaside A |
| [] | RA-FLS | inflammation | IL-6, IL-1β, TNF-α, VEGF | MAPK | QFZTC inhibits cell viability and inflammatory response in RA | Qufeng Zhitong capsule (QFZTC) | |
| [] | RA-FLS | identification of specific mRNA- miRNA- lncRNA networks | MMP9, miR-204–5p, MIAT | MMP9/miR-204–5p/MIAT | HDW may affect the progression of RA by regulating the MMP9/miR-204–5p/MIAT axis | Hedyotis diffusa Willd (HDW) | |
| [] | RA-FLS | immune inflammation | Bax, caspase3, caspase8, caspase9, COX2, TNF, and IL-17 | - | Siegesbeckiae Herba can promote apoptosis of RA-FLS and alleviate the immune inflammatory response of RA | Siegesbeckiae Herba (SBH) | |
| [] | RA-FLS | inflammation, oxidative stress | IL-6, IL-17, ROS, SOD2, RNS | LINC00638/Nrf2/HO-1 | XFC inhibits inflammation and oxidative stress in RA by up-regulating LINC00638 and activating the Nrf2/HO-1 pathway | Xinfeng capsules (XFC) | |
| [] | HUVEC, RA-FLS | synovial hyperplasia, angiogenesis | VEGF, CD31 | VEGFR/PI3K/AKT | EAF effectively inhibits synovial proliferation and angiogenesis in arthritic joints, a mechanism attributed to the modulation of the VEGFR/PI3K/AKT signaling pathway | ethanol extract of Anemone flaccida Fr. Schmidt (EAF) | |
| [] | RA-FLS | bone destruction | FGFR1, p-FGFR1, ERK1/2, p-ERK1/2, RANKL, MMP2, MMP3 | FGFR1 | JBQGF may play a therapeutic role in preventing and treating RA joint bone destruction by inhibiting the phosphorylation of FGFR1 | Juanbi Qianggu Formula (JBQGF) | |
| [] | RA-FLS | migration, oxidative stress | ROS, MDA, SOD, MMP2, MMP9 | Nrf2-Keap1 | PCA reduces H2O2-induced migration and oxidative stress of RA-FLS through activation of the Nrf2-Keap1 signaling pathway | protocatechuic acid (PCA) | |
| [] | RA-FLS | proliferation, adhesion | MMP-1, MMP-3, RANKL, TNF-α | RAS/MAPK/FOXO/HIF-1 | Berberine suppresses RA-FLS cell proliferation and adhesion via the RAS/MAPK/FOXO/HIF-1 signaling pathway, offering a potential treatment for rheumatoid arthritis | berberine | |
| [] | RA-FLS | inflammation | TNF-α, IL-1β, IL-6, IL-10 | Wnt/β-catenin | DBD inhibits RA progression and may control the disease by suppressing inflammatory cytokine responses through Wnt signaling, targeting SFRP4, to delay RA development | Danggui Buxue Decoction (DBD) | |
| [] | RA-FLS | cell growth, inflammatory response | IL-1β, IL-6, IL-4, IL-10 | hsa-circ-0003353/microRNA-31–5p/CDK1 axis | Triptolide inhibits fibroblast-like synoviocyte growth and inflammation by modulating the hsa-circ-0003353/microRNA-31–5p/CDK1 axis | triptolide | |
| [] | MH7A | inflammation, apoptosis | MMP-1, MMP-3, IL-1β, IL-2, IL-6, IL-17, IFN-γ, MCP-1, IL-10 | PI3K/AKT, JAK/STAT | IBC promotes cell apoptosis through the PI3K/AKT and JAK/STAT signaling pathways, ultimately inhibiting the progression of synovitis | isobavachalcone (IBC) | |
| [] | RA synovial cells | inflammation and chemotaxis | TNF-α, IL-1β, CCL5, CXCL10, CXCR2, IL2 | Toll, TNF | Dioscin significantly inhibits the proliferation and induces apoptosis of synovial cells in RA rats by reducing the secretion of TNF-α and IL-1β, suppressing the abnormal expression of CCL5, CXCL10, CXCR2, and IL-2 | dioscin | |
| [] | MH7A | inflammation | Bax, Bcl-2, IL-6, IL-8, COX-2, iNOS, MMP-1, MMP-2, MMP-3, MMP-9 | NF-κB, MAPK | Corilagin exerts antiproliferative and anti-inflammatory effects in rheumatoid arthritis by downregulating NF-κB and MAPK signaling pathways | corilagin | |
| [] | MH7A, RAW264.7 | inflammation, pyroptosis | TLR4, NLRP3, ASC, caspase-1, GSDMD-NT, IL-1β, IL-18 | TLR4/NFκB/NLRP3 | The novel drug combination of mangiferin and cinnamic acid in BHGZD alleviates rheumatoid arthritis by inhibiting TLR4/NF-κB/NLRP3 activation-induced pyroptosis | Baihu-Guizhi decoction (BHGZD) | |
| [] | HFLS-RA | inflammation and apoptosis | TNF-α, IL-6, MMP-1, MMP-3, COX-2, PGE2 | NF-κB, MAPKs | FLA inhibits inflammatory mediator expression and synthesis in HFLS-RA by suppressing NF-κB and MAPK signaling pathways while inducing apoptosis via the mitochondrial pathway | Fuzi lipid-soluble alkaloids (FLA) | |
| [] | MH7A | inflammation | IL-1β, IL-6, IL-8, IL-17A | DA extracts exert anti-inflammatory and anti-arthritic effects on human rheumatoid arthritis fibroblast-like synoviocytes and distinct mouse arthritis | Deer velvet antler (DA) | ||
| [] | RA-SF | monocyte adhesion | VCAM-1, CD11b | MEK1/2-ERK, p38, AP-1 | Antcin K inhibits VCAM-1-dependent monocyte adhesion in human rheumatoid arthritis synovial fibroblasts | antcin K | |
| [] | RA-FLS | apoptosis, inflammation | IL-4, IL-17, Bax, Bcl-2 | lncRNA MAPKAPK5-AS1 | XFC promotes RA-FLS apoptosis and attenuates inflammation by regulating lncRNA MAPKAPK5-AS1 | Xinfeng Capsules (XFC) | |
| [] | RA-FLS | apoptosis, inflammation | TNF-α, IL-1β, IL-6, IL-8, IL-4, IL-10 | lncRNA ENST00000619282 | Triptolide decreases ENST00000619282 to stimulate the apoptosis and relieves the inflammation of RA-FLS | triptolide | |
| [] | RA-FLS | inflammation | IL-1, IL-6, IL-8, MMP3, Fibronectin | circ_0015756/CUL4B/Wnt | RA pathogenesis is delayed by the traditional Chinese medicine compound Huangqin Qingre Chubi Capsule via the CUL4B/Wnt pathway | Huangqin Qingre Chubi Capsule (HQC) | |
| [] | RA-FLS | proliferation | PDK1, AKT | LOC100912373/miR-17–5p/PDK1 | Astragaloside regulates lncRNA LOC100912373 and the miR-17–5p/PDK1 axis to inhibit the proliferation of fibroblast-like synoviocytes in rats with rheumatoid arthritis | astragaloside | |
| [] | HFLS-RA | inflammation, proliferation | EGFR, MMP9, IL2, MAPK14, KDR | Rap1, PI3K-Akt, Ras | ARF can partially weaken RA by regulating the expression of multiple targets in the inflammatory immune system | antirheumatic arthritis fraction (ARF) of G. yunnanensis | |
| [] | RA-FLS | autophagy | Atg1, Atg5, Atg14, LC3-II, PI3K, Akt, mTOR | PI3K/Akt/mTOR | JWJGC regulates autophagy by inhibiting the PI3K/Akt/mTOR pathway in RA | Jinwu Jiangu Capsule (JWJGC) | |
| [] | RA-FLS | pyroptosis | caspase-1/3/4/5, NLRP3, GSDMD, ASC, IL-1β, IL-18, LDH | NLRP3/CAPSES/GSDMD | JWJGC inhibits pyroptosis through the NLRP3/CAPSES/GDMD pathway and treats fibroblast like synovial cells in rheumatoid arthritis | Jinwu Jiangu Capsule (JWJGC) | |
| [] | FLS | inflammation | IL-1β, IL-6, IL-10, IL-4 | JAK2/STAT3 | NBTL ameliorates rheumatoid arthritis in rats through inhibiting the JAK2/STAT3 signaling pathway | New Bi Tong Ling (NBTL) | |
| [] | RA-FLS | inflammation | NLRP3, caspase-1 p20, IL-1β, TNF-α | NLRP3 | The myrtenal and β-carboxymethyllene oxides selected from PEL can inhibit the activation of the NLRP3 inflammasome, thereby alleviating RA symptoms | Liquidabaris Fructus (LF) petroleum ether extract (PEL) | |
| [] | MH7A, RAW264.7 cells | immune inflammation, pyroptosis | caspase-1, IL-1β, LDH, NLRP3, ASC, TLR4, GSDMD | Toll-like signaling | BHGZD alleviates rheumatoid arthritis by inhibiting NLRP3 inflammasome activation mediated by Toll-like receptor 4 | Baihu-Guizhi decoction (BHGZD) | |
| [] | RA-FLS | energy metabolism, apoptosis, autophagy | ROS, ATP, PKM2, GLUT1, HK2, Pklr, Aldob, Pgam2, Pfkm, Acss2, caspase 3, Bax, LC3-II/LC3-I | PI3K/AKT/mTOR | Shikonin induces programmed death of fibroblast synovial cells in rheumatoid arthritis by inhibiting energy pathways | shikonin | |
| [] | RA-FLS | inflammation, bone destruction | TNF-α, NO | Inhibition of rheumatoid arthritis using bark, leaf, and male flower extracts of Eucommia ulmoides | Eucommia ulmoides Oliv. | ||
| [] | RA-SF | immune inflammation | IL-17A, TNF-α, IFN-γ, IL-6, TLR2, TLR4 | The inhibitory effect of asarinin drug serum on RASFs may be achieved by inhibition of T helper cell (Th)1/Th17 cytokines through suppression of TLR2 and TLR4 | asarinin | ||
| [] | RA-FLS | inflammation, proliferation | IL-6, IL-1β, FOX3 | miR-155/FOX3 | Paeonol prevents TNF-α-induced FLS proliferation and cytokine release by reducing the expression of miR-155 and upregulating its target FOXO3 | Paeonol (Pae) | |
| [] | MH7A | inflammation, proliferation | IL-1β, IL-6 | MAPKs | XAN selectively regulates MAPK signaling and has subsequent antiproliferative and anti-inflammatory activities on MH7A cells | 1,7-dihydroxy-3,4-dimethoxyxanthone (XAN) | |
| [] | MH7A | apoptosis, lipid accumulation, oxidative stress | ROS, CHOP | HO treatment induces lipid accumulation, ROS production, CHOP expression, and apoptosis in MH7A cells. CHOP plays a downstream antirheumatoid factor role in MH7A cells | hempseed oil (HO) | ||
| cell coculture | [] | Coculture of RA-PBMC and RA-FLS | inflammation | IL-17, IL-6, IL-23, TNF-α | hsa_circ_0091,685/EIF4A3/IL-17 | HQC may exert anti-inflammatory effects by blocking the hsa_circ_0091685/EIF4A3/IL-17 axis in RA | Huangqin Qingre Chubi Capsule (HQC) |
| [] | Coculture of rat peripheral blood lymphocytes and FLS | chemotaxis, inflammation | IL-6, TNF-α, IFN-γ, IL-4, CXCL10, CXCR3 | CXCL10/CXCR3 | Di-Long active ingredients exhibit good therapeutic effects on rheumatoid arthritis by inhibiting CXCL10/CXCR3 chemotaxis | Pheretima vulgaris | |
| [] | Coculture of T cells and synovial cells | inflammatory response, imbalance of Th17/Treg cells | IL-17, IL-1β, TNF-α, IL-10, Foxp3, RORγt | - | DTYMT significantly reduced the proliferation ability of RA fibroblast-like synovial cells under normal conditions, as well as the proliferation ability of T cells cocultured with RA fibroblast-like synovial cells after IL-6 activation | Duanteng-Yimu Tang (DTYMT) | |
| 3D organoids | [] | Synovial organoids | ubiquitin | p53, p21, PUMA, CCNB1, CCNA2, CDK1, CDK2, BIRC5, vimentin | p53 | Sesamol serves as a p53 stabilizer to relieve rheumatoid arthritis progression and inhibits the growth of synovial organoids | sesamol |
In summary, single-cell cultivation has become a pivotal tool in the exploration of TCM therapies for RA. It has not only facilitated the dissection of active compounds and their molecular mechanisms but also laid a solid foundation for the development of more precise and effective therapeutic strategies. However, despite its widespread application, owing to operational simplicity and cost-effectiveness, the single-cell model has intrinsic limitations. Specifically, it primarily reflects localized pathological processes, making it insufficient to fully capture the systemic physiological and pathological complexity of the RA.
4.2. Coculture Models in TCM Treatment of RA
Building upon single-cell models, the development of coculture systems enables the simulation of complex interactions among synovial cells, immune cells, fibroblasts, and other cell types, offering new opportunities to dissect the intricate immune networks involved in RA and the therapeutic mechanisms of TCM. The integration of both individual active ingredients and compound TCM formulations within coculture platforms has demonstrated broader therapeutic efficacy, thereby reinforcing the holistic treatment strategies characteristic of TCM for RA.
In exploring how coculture technology facilitates TCM research in RA therapy, a number of TCM formulations have revealed distinct mechanisms of action. For instance, Huangqin Qingre Chubi Capsule has demonstrated notable anti-inflammatory effects in coculture systems, such as models incorporating PBMCs from RA patients and RA-FLS. Specifically, this formulation acts by blocking the hsa_circ_0091685/EIF4A3/IL-17 regulatory axis, thereby modulating the inflammatory microenvironment of RA. These findings reveal a complex regulatory network and offer a novel mechanistic basis for the application of TCM in coculture-based models. Additionally, active compounds extracted from Di-Long (earthworm) have shown significant efficacy in suppressing inflammation by inhibiting CXCL10/CXCR3-mediated chemotaxis. This effect, validated in a coculture model of rat peripheral blood lymphocytes and RA-FLS, effectively disrupts the migration and accumulation of inflammatory cells, highlighting the crucial role of TCM in modulating intercellular communication and immune cell behavior in RA. Notably, the traditional formula of Duanteng-Yimu Tang significantly reduced the proliferation of RA-FLS under normal conditions in coculture models. Furthermore, it inhibited the proliferation of RA-FLS in the presence of T cells following IL-6 activation. These results demonstrate not only a direct suppressive effect on synovial fibroblasts but also the ability to modulate immune cell-synoviocyte interactions, offering promising insights into the immunomodulatory potential of TCM in RA treatment. For a detailed overview, refer to Table .
In conclusion, coculture models serve as a powerful experimental platform for elucidating the mechanisms by which TCM exerts its therapeutic effects in RA. By replicating the complex intercellular interactions within the synovial microenvironment, these models provide deeper insight into how TCM modulates pathological processes, laying a robust scientific foundation for the development of more precise and effective treatment strategies. However, despite their value in reflecting multicellular dynamics, coculture systems still face limitations, particularly in their inability to fully recapitulate the highly complex and dynamic in vivo environment. These challenges must be addressed to further advance their application in TCM-based RA research.
4.3. 3D Organoids in TCM Treatment of RA
The emergence of 3D organoid technology has significantly transformed in vitro modeling approaches for RA. By constructing 3D synovial organoids, researchers can more faithfully replicate the histopathological changes and drug response patterns observed in RA. This approach not only enhances translational research but also lays a foundation for the development of personalized treatment strategies. A recent study led by Lin and colleagues successfully established a synovial organoid model derived from RA-FLS to investigate the therapeutic potential of sesamol in RA. Their results revealed that sesamol exerted a dose-dependent inhibitory effect on synovial organoid growth. Intriguingly, organoids derived from different individuals exhibited varied sensitivities to sesamol, underscoring the feasibility of using organoid systems to guide personalized treatment decisions. To further elucidate the molecular mechanisms underlying sesamol’s action, the authors employed RNA sequencing (RNA-seq), which demonstrated that sesamol significantly upregulated the p53 signaling pathway and apoptosis-related genes but downregulated the key regulators of cell cycle progression and DNA replication. This transcriptional profile strongly supports sesamol's capacity to reverse pathological phenotypes in RA-FLS. Furthermore, the study revealed that sesamol enhances p53 pathway activity by inhibiting its ubiquitination and subsequent degradation, thereby functioning as a novel p53 stabilizer. The novelty of this research lies in its multidimensional validation of sesamol's therapeutic efficacy, utilizing not only traditional in vivo models such as collagen-induced arthritis mice and predictive modeling but also the RA-FLS-derived synovial organoid platform. This comprehensive approach offers valuable new insights and strategies for advancing TCM-based therapies for RA. Despite its advantages, the widespread application of 3D organoid technology remains constrained by several factors, including complex operational protocols, technical challenges, and relatively high costs. These limitations continue to pose barriers to their large-scale adoption and deeper exploration in basic and translational research (Table ).
In conclusion, the progression from single-cell cultures to coculture systems and ultimately to 3D organoids reflects the continuous advancement of biotechnological approaches in RA research. This evolution has opened new frontiers for exploring the therapeutic potential of TCM, particularly through the lens of synovial organoid models. With ongoing technological innovation and expanded research efforts, TCM strategies, especially those grounded in organoid-based platforms, are poised to play an increasingly critical role in the prevention, management, and treatment of RA.
5. Conclusions and Outlook
Synovial organoids and cell culture models represent promising platforms for studying the therapeutic potential of TCM in the treatment of RA. However, their development and application still face numerous notable challenges. First, while single-cell culture models offer operational simplicity, they fail to capture the full complexity and systemic nature of RA pathophysiology. Coculture models, though more representative of in vivo conditions, remain limited in their ability to fully simulate the dynamic and multifactorial environment of the human joint. Second, the current technology for constructing synovial organoids is still in its infancy. The efficient and stable generation of physiologically functional synovial organoids remains a significant technical hurdle. Additionally, the integration of TCM with synovial organoid research presents additional limitations that warrant careful attention: (1) Variability in Herbal Formulations: The multicomponent and multitarget nature of TCM prescriptions leads to considerable batch-to-batch variability in active compounds, making it difficult to achieve reproducible dose–response relationships. (2) Standardization Challenges: The lack of universally accepted quality control protocols for TCM extracts hinders the cross-laboratory validation of experimental findings across different laboratories and organoid platforms. (3) Technical Reproducibility: Differences in organoid generation methods (e.g., cell sources, scaffold materials, and cytokine cocktails) may result in inconsistent responses to TCM interventions. Overcoming these challenges will require coordinated interdisciplinary efforts. Key steps include the establishment of standardized TCM identification and optimization of organoid culture protocols among other solutions. Looking ahead, future research should focus on breaking through current technical barriers in organoid fabrication, developing more advanced and physiologically accurate in vitro models that better reflect the pathogenesis of RA, and further integrating TCM theories with modern biotechnological tools. Such advancements will significantly accelerate the translational application of organoid-based platforms in RA research, particularly in disease modeling, drug screening and evaluation, and design of personalized treatment regimens.
Acknowledgments
We acknowledge Biorender medical art (www.biorender.com/) for providing some elements included in the figures and the TOC graphic.
Glossary
Abbreviations
- ASCs
Adult stem cells
- BHGZD
Baihu Guizhi Decoction
- DBD
Danggui Buxue Decoction
- ECM
Extracellular matrix
- FLS
Fibroblast-like synovial cells
- HQC
Huangqin Qingre Chubi Capsule
- iPSC
Induced pluripotent stem cell
- NO
Nitric oxide
- PBMCs
Peripheral blood mononuclear cells
- PGE2
Prostaglandin E2
- RA
Rheumatoid arthritis
- SF
Synovial fluid
- SM
Synovium
- TCM
Traditional Chinese medicine
- 2D
two-dimensional
- 3D
three-dimensional
- XFC
Xinfeng Capsules
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
All authors read and approved the final manuscript. F.W. and J.L. conceptualized and designed the study. F.W. and J.W. prepared the original draft. Y.S. and M.H. analyzed and interpreted the results. J.L. reviewed and edited the final draft.
This study was supported by the following projects: National Nature Fund Program (82274490, 82205090); National Traditional Chinese Medicine Inheritance and Innovation Project Fund (Development and Reform Commission[2022] No. 366); National High Level Key Discipline of Traditional Chinese Medicine, Traditional Chinese Medicine Arthralgia (number Chinese Medicine Ren Jiao Han [2023] No. 85).
No potential conflict of interest was reported by the author(s).
The authors declare no competing financial interest.
References
- Niu Q., Gao J., Wang L., Liu J., Zhang L.. Regulation of differentiation and generation of osteoclasts in rheumatoid arthritis. Front. Immunol. 2022;13:1034050. doi: 10.3389/fimmu.2022.1034050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang J., Xiong J., Sun Y., Gu L., Chen Y., Guo Y., Liu C., Sun J.. B7-H3 promotes angiogenesis in rheumatoid arthritis. Mol. immunol. 2024;165:19–27. doi: 10.1016/j.molimm.2023.12.002. [DOI] [PubMed] [Google Scholar]
- Smith M. D., Barg E., Weedon H., Papengelis V., Smeets T., Tak P. P., Kraan M., Coleman M., Ahern M. J.. Microarchitecture and protective mechanisms in synovial tissue from clinically and arthroscopically normal knee joints. Ann. rheum dis. 2003;62(4):303–307. doi: 10.1136/ard.62.4.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alivernini S., Firestein G. S., McInnes I. B.. The pathogenesis of rheumatoid arthritis. Immunity. 2022;55(12):2255–2270. doi: 10.1016/j.immuni.2022.11.009. [DOI] [PubMed] [Google Scholar]
- Tsaltskan V., Firestein G. S.. Targeting fibroblast-like synoviocytes in rheumatoid arthritis. Curr. opin pharmacol. 2022;67:102304. doi: 10.1016/j.coph.2022.102304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Y., Chen Y., Wang J., Zhu X., Wang K., Li Y., Zhou F.. Ginkgolide J protects human synovial cells SW982 via suppression of p38-dependent production of pro-inflammatory mediators. Mol. med rep. 2021;24(2):555. doi: 10.3892/mmr.2021.12194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conigliaro P., Triggianese P., De Martino E., Fonti G. L., Chimenti M. S., Sunzini F., Viola A., Canofari C., Perricone R.. Challenges in the treatment of Rheumatoid Arthritis. Autoimmun rev. 2019;18(7):706–713. doi: 10.1016/j.autrev.2019.05.007. [DOI] [PubMed] [Google Scholar]
- Mueller A. L., Payandeh Z., Mohammadkhani N., Mubarak S. M. H., Zakeri A., Alagheband Bahrami A., Brockmueller A., Shakibaei M.. Recent Advances in Understanding the Pathogenesis of Rheumatoid Arthritis: New Treatment Strategies. Cells. 2021;10(11):3017. doi: 10.3390/cells10113017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Andrea E., Desai R. J., He M., Glynn R. J., Lee H., Weinblatt M. E., Kim S. C.. Cardiovascular Risks of Hydroxychloroquine vs Methotrexate in Patients With Rheumatoid Arthritis. J. am coll cardiol. 2022;80(1):36–46. doi: 10.1016/j.jacc.2022.04.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jalal H., O’Dell J. R., Bridges S. L., Cofield S., Curtis J. R., Mikuls T. R., Moreland L. W., Michaud K.. Cost-Effectiveness of Triple Therapy Versus Etanercept Plus Methotrexate in Early Aggressive Rheumatoid Arthritis. Arthrit care res. 2016;68(12):1751–1757. doi: 10.1002/acr.22895. [DOI] [PubMed] [Google Scholar]
- Kour G., Choudhary R., Anjum S., Bhagat A., Bajaj B. K., Ahmed Z.. Phytochemicals targeting JAK/STAT pathway in the treatment of rheumatoid arthritis: Is there a future? Biochem pharmacol. 2022;197:114929. doi: 10.1016/j.bcp.2022.114929. [DOI] [PubMed] [Google Scholar]
- Williamson E. M., Lorenc A., Booker A., Robinson N.. The rise of traditional Chinese medicine and its materia medica: a comparison of the frequency and safety of materials and species used in Europe and China. J. ethnopharmacol. 2013;149(2):453–462. doi: 10.1016/j.jep.2013.06.050. [DOI] [PubMed] [Google Scholar]
- Liu X., Wang Z., Qian H., Tao W., Zhang Y., Hu C., Mao W., Guo Q.. Natural medicines of targeted rheumatoid arthritis and its action mechanism. Front Immunol. 2022;13:945129. doi: 10.3389/fimmu.2022.945129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li T. P., Zhang A. H., Miao J. H., Sun H., Yan G. L., Wu F. F., Wang X. J.. Applications and potential mechanisms of herbal medicines for rheumatoid arthritis treatment: a systematic review. RSC Adv. 2019;9(45):26381–26392. doi: 10.1039/C9RA04737A. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma Q., Jiang J. G.. Functional Components from Nature-Derived Drugs for the Treatment of Rheumatoid Arthritis. Curr. drug targets. 2016;17(14):1673–1686. doi: 10.2174/1389450117666160527122233. [DOI] [PubMed] [Google Scholar]
- Keisuke I., Bian B. L., Li X. D., Takashi S., Akira I.. Action mechanisms of complementary and alternative medicine therapies for rheumatoid arthritis. Chin j integr med. 2011;17(10):723–730. doi: 10.1007/s11655-011-0871-3. [DOI] [PubMed] [Google Scholar]
- Lü S., Wang Q., Li G., Sun S., Guo Y., Kuang H.. The treatment of rheumatoid arthritis using Chinese medicinal plants: From pharmacology to potential molecular mechanisms. J. ethnopharmacol. 2015;176:177–206. doi: 10.1016/j.jep.2015.10.010. [DOI] [PubMed] [Google Scholar]
- Yuan H. Y., Zhang X. L., Zhang X. H., Meng L., Wei J. F.. Analysis of patents on anti-rheumatoid arthritis therapies issued in China. Expert opin ther pat. 2015;25(8):909–930. doi: 10.1517/13543776.2015.1044972. [DOI] [PubMed] [Google Scholar]
- Rao Q., Zhao X., Wu F., Guo X., Xu Y., Yu H., Cai D., Zhao G.. [Corrigendum] Alcohol extracts from Anemone flaccida Fr. Schmidt treat rheumatoid arthritis via inhibition of synovial hyperplasia and angiogenesis. Mol. med rep. 2024;30(4):179. doi: 10.3892/mmr.2024.13302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boonekamp K. E., Dayton T. L., Clevers H.. Intestinal organoids as tools for enriching and studying specific and rare cell types: advances and future directions. J. mol cell biol. 2020;12(8):562–568. doi: 10.1093/jmcb/mjaa034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin X., Farin H. F., van Es J. H., Clevers H., Langer R., Karp J. M.. Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nat. methods. 2014;11(1):106–12. doi: 10.1038/nmeth.2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shyer A. E., Huycke T. R., Lee C., Mahadevan L., Tabin C. J.. Bending gradients: how the intestinal stem cell gets its home. Cell. 2015;161(3):569–580. doi: 10.1016/j.cell.2015.03.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clevers H.. Modeling Development and Disease with Organoids. Cell. 2016;165(7):1586–1597. doi: 10.1016/j.cell.2016.05.082. [DOI] [PubMed] [Google Scholar]
- Zhao Z., Chen X., Dowbaj A. M., Sljukic A., Bratlie K., Lin L., Fong E. L. S., Balachander G. M., Chen Z., Soragni A., Huch M., Zeng Y. A., Wang Q., Yu H.. Organoids. Nat. Rev. Methods Primers. 2022;2:94. doi: 10.1038/s43586-022-00174-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin X., Lin T., Wang X., He J., Gao X., Lyu S., Wang Q., Chen J.. Sesamol serves as a p53 stabilizer to relieve rheumatoid arthritis progression and inhibits the growth of synovial organoids. Phytomedicine. 2023;121:155109. doi: 10.1016/j.phymed.2023.155109. [DOI] [PubMed] [Google Scholar]
- Wei K., Korsunsky I., Marshall J. L., Gao A., Watts G. F. M., Major T., Croft A. P., Watts J., Blazar P. E., Lange J. K., Thornhill T. S., Filer A., Raza K., Donlin L. T., Albrecht J., Anolik J. H., Apruzzese W., Boyce B. F., Boyle D. L., Bridges S. L., Buckner J. H., Bykerk V. P., DiCarlo E., Dolan J., Eisenhaure T. M., Firestein G. S., Fonseka C. Y., Goodman S. M., Gravallese E. M., Gregersen P. K., Guthridge J. M., Gutierrez-Arcelus M., Hacohen N., Holers V. M., Hughes L. B., Ivashkiv L. B., James E. A., James J. A., Jonsson A. H., Keegan J., Kelly S., Lee Y. C., Lederer J. A., Lieb D. J., Mandelin A. M., McGeachy M. J., McNamara M. A., Mears J. R., Meednu N., Mizoguchi F., Moreland L., Nguyen J. P., Nusbaum C., Noma A., Orange D. E., Perlman H., Pitzalis C., Rangel-Moreno J., Rao D. A., Rohani-Pichavant M., Ritchlin C., Robinson W. H., Salomon-Escoto K., Seshadri A., Seifert J., Slowikowski K., Sutherby D., Tabechian D., Turner J. D., Utz P. J., Zhang F., Siebel C. W., Buckley C. D., Raychaudhuri S., Brenner M. B.. Notch signalling drives synovial fibroblast identity and arthritis pathology. Nature. 2020;582(7811):259–264. doi: 10.1038/s41586-020-2222-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan J. F., Li S., Guo C. A., Xu D. L., Zhang F., Yan Z. Q., Mo X. M.. Evaluation of synovium-derived mesenchymal stem cells and 3D printed nanocomposite scaffolds for tissue engineering. Sci. technol adv mat. 2015;16(4):045001. doi: 10.1088/1468-6996/16/4/045001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang X., He Y., Zhao Y., Pan Z., Wang Y.. Danggui Buxue Decoction exerts its therapeutic effect on rheumatoid arthritis through the inhibition of Wnt/β-catenin signaling pathway. J. Orthop Surg Res. 2023;18(1):944. doi: 10.1186/s13018-023-04439-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W., Mao X., Wang X., Liu Y., Wang K., Li C., Li T., Zhang Y., Lin N.. Disease-Modifying Anti-rheumatic Drug Prescription Baihu-Guizhi Decoction Attenuates Rheumatoid Arthritis via Suppressing Toll-Like Receptor 4-mediated NLRP3 Inflammasome Activation. Front Pharmacol. 2021;12:743086. doi: 10.3389/fphar.2021.743086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Y., Liu J., Qi Y., Zhou Q., Li Y., Cong C., Chen Y.. Integrating clinical data mining, network analysis and experimental validation reveal the anti-inflammatory mechanism of Huangqin Qingre Chubi Capsule in rheumatoid arthritis treatment. J. ethnopharmacol. 2024;329:118077. doi: 10.1016/j.jep.2024.118077. [DOI] [PubMed] [Google Scholar]
- Smith M. D.. The normal synovium. Open Rheumatol. J. 2011;5(null):100–106. doi: 10.2174/1874312901105010100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwanaga T., Shikichi M., Kitamura H., Yanase H., Nozawa-Inoue K.. Morphology and functional roles of synoviocytes in the joint. Arch histol cytol. 2000;63(1):17–31. doi: 10.1679/aohc.63.17. [DOI] [PubMed] [Google Scholar]
- Bottini N., Firestein G. S.. Duality of fibroblast-like synoviocytes in RA: passive responders and imprinted aggressors. Nat. rev rheumatol. 2013;9(1):24–33. doi: 10.1038/nrrheum.2012.190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Sousa E. B., Aguiar D. P., Barcelos J. F., Duarte M. E., Olej B.. Approaches to preserve human osteochondral allografts. Cell tissue bank. 2015;16(3):425–31. doi: 10.1007/s10561-014-9486-1. [DOI] [PubMed] [Google Scholar]
- Stefani R. M., Halder S. S., Estell E. G., Lee A. J., Silverstein A. M., Sobczak E., Chahine N. O., Ateshian G. A., Shah R. P., Hung C. T.. A Functional Tissue-Engineered Synovium Model to Study Osteoarthritis Progression and Treatment. Tissue eng pt a. 2019;25(7–8):538–553. doi: 10.1089/ten.tea.2018.0142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma H. P., Deng X., Chen D. Y., Zhu D., Tong J. L., Zhao T., Ma J. H., Liu Y. Q.. A microfluidic chip-based co-culture of fibroblast-like synoviocytes with osteoblasts and osteoclasts to test bone erosion and drug evaluation. R Soc. Open Sci. 2018;5(9):180528. doi: 10.1098/rsos.180528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Makkar R., Behl T., Kumar A., Uddin M. S., Bungau S.. Untying the correlation between apolipoproteins and rheumatoid arthritis. Inflamm res. 2021;70(1):19–28. doi: 10.1007/s00011-020-01410-5. [DOI] [PubMed] [Google Scholar]
- Cooles F. A., Isaacs J. D.. Pathophysiology of rheumatoid arthritis. Curr. opin rheumatol. 2011;23(3):233–240. doi: 10.1097/BOR.0b013e32834518a3. [DOI] [PubMed] [Google Scholar]
- Badillo-Mata J. A., Camacho-Villegas T. A., Lugo-Fabres P. H.. 3D Cell Culture as Tools to Characterize Rheumatoid Arthritis Signaling and Development of New Treatments. Cells. 2022;11(21):3410. doi: 10.3390/cells11213410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Damerau A., Gaber T.. Modeling Rheumatoid Arthritis In Vitro: From Experimental Feasibility to Physiological Proximity. Int. J. Mol. Sci. 2020;21(21):7916. doi: 10.3390/ijms21217916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mosser D. M., Edwards J. P.. Exploring the full spectrum of macrophage activation. Nat. rev immunol. 2008;8(12):958–969. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ilic M. Z., Vankemmelbeke M. N., Holen I., Buttle D. J., Clem Robinson H., Handley C. J.. Bovine joint capsule and fibroblasts derived from joint capsule express aggrecanase activity. Matrix biol. 2000;19(3):257–265. doi: 10.1016/S0945-053X(00)00069-X. [DOI] [PubMed] [Google Scholar]
- Broeren M. G. A., Waterborg C. E. J., Wiegertjes R., Thurlings R. M., Koenders M. I., Van Lent P., Van der Kraan P. M., Van de Loo F. A. J.. A three-dimensional model to study human synovial pathology. Altex-altern anim ex. 2019;36(1):18–28. doi: 10.14573/altex.1804161. [DOI] [PubMed] [Google Scholar]
- Vankemmelbeke M. N., Ilic M. Z., Handley C. J., Knight C. G., Buttle D. J.. Coincubation of bovine synovial or capsular tissue with cartilage generates a soluble ″Aggrecanase″ activity. Biochem bioph res co. 1999;255(3):686–691. doi: 10.1006/bbrc.1999.0266. [DOI] [PubMed] [Google Scholar]
- Baptista L. S., Kronemberger G. S., Côrtes I., Charelli L. E., Matsui R. A. M., Palhares T. N., Sohier J., Rossi A. M., Granjeiro J. M.. Adult Stem Cells Spheroids to Optimize Cell Colonization in Scaffolds for Cartilage and Bone Tissue Engineering. Int. J. Mol. Sci. 2018;19(5):1285. doi: 10.3390/ijms19051285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sekiya I., Katano H., Ozeki N.. Characteristics of MSCs in Synovial Fluid and Mode of Action of Intra-Articular Injections of Synovial MSCs in Knee Osteoarthritis. Int. J. Mol. Sci. 2021;22(6):2838. doi: 10.3390/ijms22062838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Y., Koga H., Nakagawa Y., Nakamura T., Katagiri H., Takada R., Katakura M., Tsuji K., Sekiya I., Miyatake K.. Characteristics of the synovial microenvironment and synovial mesenchymal stem cells with hip osteoarthritis of different bone morphologies. Arthritis res ther. 2024;26(1):17. doi: 10.1186/s13075-023-03252-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okamura G., Ebina K., Hirao M., Chijimatsu R., Yonetani Y., Etani Y., Miyama A., Takami K., Goshima A., Yoshikawa H., Ishimoto T., Nakano T., Hamada M., Kanamoto T., Nakata K.. Promoting Effect of Basic Fibroblast Growth Factor in Synovial Mesenchymal Stem Cell-Based Cartilage Regeneration. Int. J. Mol. Sci. 2020;22(1):300. doi: 10.3390/ijms22010300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J. C., Lee S. Y., Min H. J., Han S. A., Jang J., Lee S., Seong S. C., Lee M. C.. Synovium-derived mesenchymal stem cells encapsulated in a novel injectable gel can repair osteochondral defects in a rabbit model. Tissue eng pt a. 2012;18(19–20):2173–2186. doi: 10.1089/ten.tea.2011.0643. [DOI] [PubMed] [Google Scholar]
- Fülber J., Agreste F. R., Seidel S. R. T., Sotelo E. D. P., Barbosa Â., Michelacci Y. M., Baccarin R. Y. A.. Chondrogenic potential of mesenchymal stem cells from horses using a magnetic 3D cell culture system. World J. Stem Cells. 2021;13(6):645–658. doi: 10.4252/wjsc.v13.i6.645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koliaraki V., Prados A., Armaka M., Kollias G.. The mesenchymal context in inflammation, immunity and cancer. Nat. immunol. 2020;21(9):974–982. doi: 10.1038/s41590-020-0741-2. [DOI] [PubMed] [Google Scholar]
- Davidson S., Coles M., Thomas T., Kollias G., Ludewig B., Turley S., Brenner M., Buckley C. D.. Fibroblasts as immune regulators in infection, inflammation and cancer. Nat. rev immunol. 2021;21(11):704–717. doi: 10.1038/s41577-021-00540-z. [DOI] [PubMed] [Google Scholar]
- Ortiz-Vitali J. L., Darabi R.. iPSCs as a Platform for Disease Modeling, Drug Screening, and Personalized Therapy in Muscular Dystrophies. Cells. 2019;8(1):20. doi: 10.3390/cells8010020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma A., Sances S., Workman M. J., Svendsen C. N.. Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell stem cell. 2020;26(3):309–329. doi: 10.1016/j.stem.2020.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao Q., Li Z., Rhee C., Xiang S., Maruyama M., Huang E. E., Yao Z., Bunnell B. A., Tuan R. S., Lin H., Gold M. S., Goodman S. B.. Macrophages Modulate the Function of MSC- and iPSC-Derived Fibroblasts in the Presence of Polyethylene Particles. Int. J. Mol. Sci. 2021;22(23):12837. doi: 10.3390/ijms222312837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z. A., Sant S., Cho S. K., Goodman S. B., Bunnell B. A., Tuan R. S., Gold M. S., Lin H.. Synovial joint-on-a-chip for modeling arthritis: progress, pitfalls, and potential. Trends biotechnol. 2023;41(4):511–527. doi: 10.1016/j.tibtech.2022.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piluso S., Li Y., Abinzano F., Levato R., Moreira Teixeira L., Karperien M., Leijten J., van Weeren R., Malda J.. Mimicking the Articular Joint with In Vitro Models. Trends biotechnol. 2019;37(10):1063–1077. doi: 10.1016/j.tibtech.2019.03.003. [DOI] [PubMed] [Google Scholar]
- Mondadori C., Palombella S., Salehi S., Talò G., Visone R., Rasponi M., Redaelli A., Sansone V., Moretti M., Lopa S.. Recapitulating monocyte extravasation to the synovium in an organotypic microfluidic model of the articular joint. Biofabrication. 2021;13(4):045001. doi: 10.1088/1758-5090/ac0c5e. [DOI] [PubMed] [Google Scholar]
- Rothbauer M., Höll G., Eilenberger C., Kratz S. R. A., Farooq B., Schuller P., Olmos Calvo I., Byrne R. A., Meyer B., Niederreiter B., Küpcü S., Sevelda F., Holinka J., Hayden O., Tedde S. F., Kiener H. P., Ertl P.. Monitoring tissue-level remodelling during inflammatory arthritis using a three-dimensional synovium-on-a-chip with non-invasive light scattering biosensing. Lab chip. 2020;20(8):1461–1471. doi: 10.1039/C9LC01097A. [DOI] [PubMed] [Google Scholar]
- Maracle C. X., Kucharzewska P., Helder B., van der Horst C., Correa de Sampaio P., Noort A. R., van Zoest K., Griffioen A. W., Olsson H., Tas S. W.. Targeting non-canonical nuclear factor-κB signalling attenuates neovascularization in a novel 3D model of rheumatoid arthritis synovial angiogenesis. Rheumatology. 2017;56(2):294–302. doi: 10.1093/rheumatology/kew393. [DOI] [PubMed] [Google Scholar]
- Bhattacharjee M., Balakrishnan L., Renuse S., Advani J., Goel R., Sathe G., Keshava Prasad T. S., Nair B., Jois R., Shankar S., Pandey A.. Synovial fluid proteome in rheumatoid arthritis. Clin. Proteom. 2016;13:12. doi: 10.1186/s12014-016-9113-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philippon E. M. L., van Rooijen L. J. E., Khodadust F., van Hamburg J. P., van der Laken C. J., Tas S. W.. A novel 3D spheroid model of rheumatoid arthritis synovial tissue incorporating fibroblasts, endothelial cells, and macrophages. Front. Immunol. 2023;14:1188835. doi: 10.3389/fimmu.2023.1188835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiener H. P., Lee D. M., Agarwal S. K., Brenner M. B.. Cadherin-11 induces rheumatoid arthritis fibroblast-like synoviocytes to form lining layers in vitro. Am. j pathol. 2006;168(5):1486–1499. doi: 10.2353/ajpath.2006.050999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonelli M., Dalwigk K., Platzer A., Olmos Calvo I., Hayer S., Niederreiter B., Holinka J., Sevelda F., Pap T., Steiner G., Superti-Furga G., Smolen J. S., Kiener H. P., Karonitsch T.. IRF1 is critical for the TNF-driven interferon response in rheumatoid fibroblast-like synoviocytes: JAKinibs suppress the interferon response in RA-FLSs. Exp mol med. 2019;51(7):1–11. doi: 10.1038/s12276-019-0267-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rothbauer M., Byrne R. A., Schobesberger S., Olmos Calvo I., Fischer A., Reihs E. I., Spitz S., Bachmann B., Sevelda F., Holinka J., Holnthoner W., Redl H., Toegel S., Windhager R., Kiener H. P., Ertl P.. Establishment of a human three-dimensional chip-based chondro-synovial coculture joint model for reciprocal cross talk studies in arthritis research. Lab chip. 2021;21(21):4128–4143. doi: 10.1039/D1LC00130B. [DOI] [PubMed] [Google Scholar]
- Sun L., Yang H., Wang Y., Zhang X., Jin B., Xie F., Jin Y., Pang Y., Zhao H., Lu X., Sang X., Zhang H., Lin F., Sun W., Huang P., Mao Y.. Application of a 3D Bioprinted Hepatocellular Carcinoma Cell Model in Antitumor Drug Research. Front Oncol. 2020;10:878. doi: 10.3389/fonc.2020.00878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lv K., Zhu J., Zheng S., Jiao Z., Nie Y., Song F., Liu T., Song K.. Evaluation of inhibitory effects of geniposide on a tumor model of human breast cancer based on 3D printed Cs/Gel hybrid scaffold. Mat sci eng c-mater. 2021;119:111509. doi: 10.1016/j.msec.2020.111509. [DOI] [PubMed] [Google Scholar]
- Lin J., Sun A. R., Li J., Yuan T., Cheng W., Ke L., Chen J., Sun W., Mi S., Zhang P.. A Three-Dimensional Co-Culture Model for Rheumatoid Arthritis Pannus Tissue. Front Bioeng Biotechnol. 2021;9:764212. doi: 10.3389/fbioe.2021.764212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rossi G., Manfrin A., Lutolf M. P.. Progress and potential in organoid research. Nat. rev genet. 2018;19(11):671–687. doi: 10.1038/s41576-018-0051-9. [DOI] [PubMed] [Google Scholar]
- Delaine-Smith R. M., Reilly G. C.. Mesenchymal stem cell responses to mechanical stimuli. Muscles Ligaments Tendons J. 2012;2(3):169–180. [PMC free article] [PubMed] [Google Scholar]
- Ravalli S., Szychlinska M. A., Lauretta G., Musumeci G.. New Insights on Mechanical Stimulation of Mesenchymal Stem Cells for Cartilage Regeneration. Appl. Sci. 2020;10(76):292. doi: 10.3390/app10082927. [DOI] [Google Scholar]
- Grad S., Eglin D., Alini M., Stoddart M. J.. Physical stimulation of chondrogenic cells in vitro: a review. Clin orthop relat r. 2011;469(10):2764–2772. doi: 10.1007/s11999-011-1819-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petretta M., Villata S., Scozzaro M. P., Roseti L., Favero M., Napione L., Frascella F., Pirri C. F., Grigolo B., Olivotto E.. In Vitro Synovial Membrane 3D Model Developed by Volumetric Extrusion Bioprinting. Appl. Sci. 2023;13(3):1889. doi: 10.3390/app13031889. [DOI] [Google Scholar]
- Su M., Zhou D., Huang J., Yang T., Zhou Q., Tan Y.. Forsythiaside A exhibits anti-migration and anti-inflammation effects in rheumatoid arthritis in vitro model. Int. J. Rheum. Dis. 2024;27(1):e14976. doi: 10.1111/1756-185X.14976. [DOI] [PubMed] [Google Scholar]
- Lin H., Du X., Wang Y., Cai C., Gao J., Xiang H., Pan F.. The Potential Mechanisms of Qufeng Zhitong Capsule against Rheumatoid Arthritis Based on Network Pharmacology and In Vitro Experiments. Crit rev immunol. 2024;44(1):1–16. doi: 10.1615/CritRevImmunol.2023050214. [DOI] [PubMed] [Google Scholar]
- Jiang J., Huang M., Zhang S. S., Wu Y. G., Li X. L., Deng H., Qili X. Y., Chen J. L., Meng Y., Sun W. K.. Identification of Hedyotis diffusa Willd-specific mRNA-miRNA-lncRNA network in rheumatoid arthritis based on network pharmacology, bioinformatics analysis, and experimental verification. Sci. Rep. 2024;14(1):6291. doi: 10.1038/s41598-024-56880-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang F., Liu J., Fang Y., Wen J., He M., Li X., Han Q.. Effect of Siegesbeckiae Herba on immune-inflammation of rheumatoid arthritis: Data mining and network pharmacology. Eur. J. Integrative Med. 2023;59:102242. doi: 10.1016/j.eujim.2023.102242. [DOI] [Google Scholar]
- Sun Y., Liu J., Xin L., Wen J., Zhou Q., Chen X., Ding X., Zhang X.. Xinfeng capsule inhibits inflammation and oxidative stress in rheumatoid arthritis by up-regulating LINC00638 and activating Nrf2/HO-1 pathway. J. ethnopharmacol. 2023;301:115839. doi: 10.1016/j.jep.2022.115839. [DOI] [PubMed] [Google Scholar]
- Wen J. T., Liu J., Wang X., Wang J.. Xinfeng Capsules promotes apoptosis of synovial fibroblasts and attenuates inflammation in rheumatoid arthritis by regulating lncRNA MAPKAPK5-AS1. Zhongguo Zhong Yao Za Zhi. 2021;46(24):6542–6548. doi: 10.19540/j.cnki.cjcmm.20210914.501. [DOI] [PubMed] [Google Scholar]
- Wen J. T., Liu J., Wan L., Xin L., Guo J. C., Sun Y. Q., Wang X., Wang J.. Triptolide inhibits cell growth and inflammatory response of fibroblast-like synoviocytes by modulating hsa-circ-0003353/microRNA-31–5p/CDK1 axis in rheumatoid arthritis. Int. immunopharmacol. 2022;106:108616. doi: 10.1016/j.intimp.2022.108616. [DOI] [PubMed] [Google Scholar]
- Wen J., Liu J., Wang X., Wang J.. Triptolide promotes the apoptosis and attenuates the inflammation of fibroblast-like synoviocytes in rheumatoid arthritis by down-regulating lncRNA ENST00000619282. Phytother res. 2021;35(8):4334–4346. doi: 10.1002/ptr.7129. [DOI] [PubMed] [Google Scholar]
- Wang X., Chang J., Zhou G., Cheng C., Xiong Y., Dou J., Cheng G., Miao C.. The Traditional Chinese Medicine Compound Huangqin Qingre Chubi Capsule Inhibits the Pathogenesis of Rheumatoid Arthritis Through the CUL4B/Wnt Pathway. Front Pharmacol. 2021;12:750233. doi: 10.3389/fphar.2021.750233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang H., Fan C., Lu Y., Cui X., Liu J.. Astragaloside regulates lncRNA LOC100912373 and the miR-17–5p/PDK1 axis to inhibit the proliferation of fibroblast-like synoviocytes in rats with rheumatoid arthritis. Int. J. Mol. Med. 2021;48(1):130. doi: 10.3892/ijmm.2021.4963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W., Wang K., Liu Y., Wu H., He Y., Li C., Wang Q., Su X., Yan S., Su W., Zhang Y., Lin N.. A Novel Drug Combination of Mangiferin and Cinnamic Acid Alleviates Rheumatoid Arthritis by Inhibiting TLR4/NFκB/NLRP3 Activation-Induced Pyroptosis. Front Immunol. 2022;13:912933. doi: 10.3389/fimmu.2022.912933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z., Chen M., Wang Z., Fan Q., Lin Z., Tao X., Wu J., Liu Z., Lin R., Zhao C.. Berberine inhibits RA-FLS cell proliferation and adhesion by regulating RAS/MAPK/FOXO/HIF-1 signal pathway in the treatment of rheumatoid arthritis. Bone Joint Res. 2023;12(2):91–102. doi: 10.1302/2046-3758.122.BJR-2022-0269.R1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bao Y., Hu S. N., Song Z. J., Shen H. J., Zhong W. L., Du S. Y.. Chinese medicine Di-long (Pheretima vulgaris) and its active fraction exhibit anti-rheumatoid arthritis effects by inhibiting CXCL10/CXCR3 chemotaxis in synovium. J. ethnopharmacol. 2024;332:118286. doi: 10.1016/j.jep.2024.118286. [DOI] [PubMed] [Google Scholar]
- Feng W., Wan X., Fan S., Liu C. Z., Zheng X. X., Liu Q. P., Liu M. Y., Liu X. B., Lin C. S., Zhang L. J., Li D. T., Xu Q.. Mechanism underlying the action of Duanteng-Yimu Tang in regulating Treg/Th17 imbalance and anti-rheumatoid arthritis. Heliyon. 2023;9(5):e15867. doi: 10.1016/j.heliyon.2023.e15867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng X. H., Zhong S., Han H. H., Shi Q., Sun S. T., Xiao L. B.. Effect of Juanbi Qianggu Formula on biological behaviors of fibroblast-like synoviocytes in rheumatoid arthritis by regulating FGFR1 signaling pathway based on network pharmacology and cell function experiments. Zhongguo Zhong Yao Za Zhi. 2023;48(18):4864–4873. doi: 10.19540/j.cnki.cjcmm.20230320.406. [DOI] [PubMed] [Google Scholar]
- Liu Y., Zhang Y., Zhang K., Wang Y.. Protocatechuic acid reduces H2O2-induced migration and oxidative stress of fibroblast-like synoviocytes in rheumatoid arthritis by activating Nrf2-Keap1 signaling pathway. Chinese j physiol. 2023;66(1):28–35. doi: 10.4103/cjop.CJOP-D-22-00087. [DOI] [PubMed] [Google Scholar]
- Wang S., Du Q., Sun J., Geng S., Zhang Y.. Investigation of the mechanism of Isobavachalcone in treating rheumatoid arthritis through a combination strategy of network pharmacology and experimental verification. J. ethnopharmacol. 2022;294:115342. doi: 10.1016/j.jep.2022.115342. [DOI] [PubMed] [Google Scholar]
- Sun J., Liu B., Wang R., Yuan Y., Wang J., Zhang L., Sainaghi P. P.. Computation-Based Discovery of Potential Targets for Rheumatoid Arthritis and Related Molecular Screening and Mechanism Analysis of Traditional Chinese Medicine. Dis. Markers. 2022;2022:1. doi: 10.1155/2022/1905077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen Y., Teng L., Qu Y., Liu J., Zhu X., Chen S., Yang L., Huang Y., Song Q., Fu Q.. Anti-proliferation and anti-inflammation effects of corilagin in rheumatoid arthritis by downregulating NF-κB and MAPK signaling pathways. J. ethnopharmacol. 2022;284:114791. doi: 10.1016/j.jep.2021.114791. [DOI] [PubMed] [Google Scholar]
- Guo C., He L., Hu N., Zhao X., Gong L., Wang C., Peng C., Li Y.. Aconiti Lateralis Radix Praeparata lipid-soluble alkaloids alleviates IL-1β-induced inflammation of human fibroblast-like synoviocytes in rheumatoid arthritis by inhibiting NF-κB and MAPKs signaling pathways and inducing apoptosis. Cytokine. 2022;151:155809. doi: 10.1016/j.cyto.2022.155809. [DOI] [PubMed] [Google Scholar]
- Cheng W. J., Yang H. T., Chiang C. C., Lai K. H., Chen Y. L., Shih H. L., Kuo J. J., Hwang T. L., Lin C. C.. Deer Velvet Antler Extracts Exert Anti-Inflammatory and Anti-Arthritic Effects on Human Rheumatoid Arthritis Fibroblast-Like Synoviocytes and Distinct Mouse Arthritis. Am. j chinese med. 2022;50(6):1617–1643. doi: 10.1142/S0192415X22500689. [DOI] [PubMed] [Google Scholar]
- Achudhan D., Li-Yun Chang S., Liu S. C., Lin Y. Y., Huang W. C., Wu Y. C., Huang C. C., Tsai C. H., Ko C. Y., Kuo Y. H., Tang C. H.. Antcin K inhibits VCAM-1-dependent monocyte adhesion in human rheumatoid arthritis synovial fibroblasts. Food Nutr. Res. 2022;66:10–29219. doi: 10.29219/fnr.v66.8645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- X., Wang ; Y., Sun ; L., Ling ; X., Ren ; X., Liu ; Y., Wang ; Y., Dong ; J., Ma ; R., Song ; A., Yu ; J., Wei ; Q., Fan ; M., Guo ; T., Zhao ; R., Dao ; G., She , Gaultheria leucocarpa var. yunnanensis for Treating Rheumatoid Arthritis-An Assessment Combining Machine Learning-Guided ADME Properties Prediction, Network Pharmacology, and Pharmacological Assessment; Front Pharmacol; 12 (2021) 704040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q., Yao X., Xu H., Lu D., Huang Y., Tang F., Xiao L., Ma W.. Jinwu Jiangu Capsule affects synovial cells in rheumatoid arthritis through PI3K/Akt/mTOR signaling pathway. Acta biochim pol. 2021;68(4):641–646. doi: 10.18388/abp.2020_5514. [DOI] [PubMed] [Google Scholar]
- Ling Y., Xiao M., Huang Z. W., Xu H., Huang F. Q., Ren N. N., Chen C. M., Lu D. M., Yao X. M., Xiao L. N., Ma W. K., Song W.. Jinwujiangu Capsule Treats Fibroblast-Like Synoviocytes of Rheumatoid Arthritis by Inhibiting Pyroptosis via the NLRP3/CAPSES/GSDMD Pathway. Evid-based compl alt. 2021;2021:1. doi: 10.1155/2021/4836992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X., Xie Y., Kang A., Wang Y.. New bitongling (NBTL) ameliorates rheumatoid arthritis in rats through inhibiting JAK2/STAT3 signaling pathway. Eur. J. Histochem. 2021;65(1):3202. doi: 10.4081/ejh.2021.3202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W. x., Qian P., Guo Y. t., Gu L., Jurat J., Bai Y., Zhang D. f.. Myrtenal and β-caryophyllene oxide screened from Liquidambaris Fructus suppress NLRP3 inflammasome components in rheumatoid arthritis. BMC Complement Med. Ther. 2021;21(1):242. doi: 10.1186/s12906-021-03410-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J., Pang J., Liu Z., Ge X., Zhen Y., Jiang C. C., Liu Y., Huo Q., Sun Y., Liu H.. Shikonin induces programmed death of fibroblast synovial cells in rheumatoid arthritis by inhibiting energy pathways. Sci. Rep. 2021;11(1):18263. doi: 10.1038/s41598-021-97713-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing Y. Y., Wang J. Y., Wang K., Zhang Y., Liu K., Chen X. Y., Yuan Y.. Inhibition of rheumatoid arthritis using bark, leaf, and male flower extracts of Eucommia ulmoides. Evid-based Compl. Alt. 2020;2020:3260278. doi: 10.1155/2020/3260278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai Q., Li Y., Wang M., Li Y., Li J.. TlR2 and TlR4 are involved in the treatment of rheumatoid arthritis synovial fibroblasts with a medicated serum of asarinin through inhibition of Th1/Th17 cytokines. Exp ther med. 2020;19(4):3009–3016. doi: 10.3892/etm.2020.8557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu N., Feng X., Wang W., Zhao X., Li X.. Paeonol protects against TNF-α-induced proliferation and cytokine release of rheumatoid arthritis fibroblast-like synoviocytes by upregulating FOXO3 through inhibition of miR-155 expression. Inflamm res. 2017;66(7):603–610. doi: 10.1007/s00011-017-1041-7. [DOI] [PubMed] [Google Scholar]
- Zuo J., Xia Y., Li X., Ou-Yang Z., Chen J. W.. Selective modulation of MAPKs contribute to the anti-proliferative and anti-inflammatory activities of 1,7-dihydroxy-3,4-dimethoxyxanthone in rheumatoid arthritis-derived fibroblast-like synoviocyte MH7A cells. J. ethnopharmacol. 2015;168:248–254. doi: 10.1016/j.jep.2015.03.069. [DOI] [PubMed] [Google Scholar]
- Jeong M., Cho J., Shin J. I., Jeon Y. J., Kim J. H., Lee S. J., Kim E. S., Lee K.. Hempseed oil induces reactive oxygen species- and C/EBP homologous protein-mediated apoptosis in MH7A human rheumatoid arthritis fibroblast-like synovial cells. J. ethnopharmacol. 2014;154(3):745–752. doi: 10.1016/j.jep.2014.04.052. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.



