Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Molecular Medicine Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1791-2997 Online ISSN: 1791-3004
Journal Cover
January-2026 Volume 33 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
January-2026 Volume 33 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review)

  • Authors:
    • Ruipu Zhang
    • Liang Zhang
    • Bin Tian
    • Yiqun Wang
    • Xin Kang
    • Jiang Zheng
  • View Affiliations / Copyright

    Affiliations: Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
    Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 23
    |
    Published online on: October 31, 2025
       https://doi.org/10.3892/mmr.2025.13733
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Osteoarthritis (OA) is a prevalent chronic joint disorder with a notable global health burden, characterized by articular cartilage degeneration, abnormal bone remodeling and synovial inflammation. Traditional treatments mainly focus on symptom management rather than addressing the underlying disease mechanisms. The gut microbiome serves a potential role in OA through the gut‑bone‑cartilage axis. Notably, the gut microbiome and its metabolites can influence bone and cartilage homeostasis, and the Wnt/β‑catenin signaling pathway has been implicated in OA pathogenesis. The present study comprehensively reviews the emerging evidence supporting the gut‑bone‑cartilage axis in OA and the role of microbial regulation of Wnt/β‑catenin signaling in joint remodeling. The current understanding of the influence of the gut microbiome on OA pathogenesis is summarized, discussing the mechanisms underlying the gut‑bone‑cartilage axis and exploring the therapeutic potential of targeting this axis. Future research should focus on developing targeted therapies that modulate the gut microbiome and the Wnt/β‑catenin pathway, as well as exploring the potential of gene editing and carrier technologies for OA treatment.

Introduction

Osteoarthritis (OA) is a prevalent chronic joint disorder that poses a notable global health burden. With the aging population, the prevalence of OA is expected to rise, making it a critical public health issue (1). According to recent statistics, OA affects >300 million people worldwide, with a higher prevalence in individuals aged ≥60 years (1). The economic cost of OA is substantial, with direct medical expenses and indirect costs due to lost productivity estimated at billions of dollars annually; this underscores the urgent need for effective therapeutic strategies to manage and potentially reverse OA progression (2).

The pathophysiological hallmarks of OA include articular cartilage degeneration, abnormal bone remodeling and synovial inflammation (3). Cartilage degeneration is characterized by the loss of chondrocytes and the breakdown of the extracellular matrix, leading to reduced joint function and increased pain (4). Abnormal bone remodeling manifests as subchondral bone sclerosis and the formation of osteophytes, which further contribute to joint dysfunction (5). Synovial inflammation is characterized by the infiltration of inflammatory cells and the production of proinflammatory cytokines such as IL-1β) and tumor necrosis factor-α (TNF-α), exacerbating cartilage and bone damage. These pathological changes are interconnected, creating a vicious cycle that accelerates OA progression (5).

Despite the notable burden of OA, traditional treatments have limitations (6). Current therapeutic approaches primarily focus on symptom management, such as pain relief and improved joint function, rather than addressing the underlying disease mechanisms (7). Most research has concentrated on local factors within the joint microenvironment, such as chondrocyte apoptosis and inflammatory mediators (8,9). However, these approaches have shown limited efficacy in halting or reversing OA progression; therefore, there is a growing recognition of the need to explore systemic factors that may influence OA development and progression.

One such systemic factor is the gut microbiome, which has emerged as a potential regulator of OA through the gut-bone-cartilage axis (10,11). The gut microbiome comprises trillions of microorganisms that serve crucial roles in host metabolism, immune function and inflammation (12). Previous studies have shown that the gut microbiota and its metabolites can influence bone and cartilage homeostasis through various mechanisms, including immune modulation and signaling pathways such as Wnt/β-catenin (10,13–15). For example, animal studies have demonstrated that alterations in the gut microbiota composition can lead to changes in bone density and cartilage integrity, suggesting a potential link between gut dysbiosis and OA development (11,16). Additionally, the Wnt/β-catenin signaling pathway has been implicated in OA pathogenesis, with a study showing that its dysregulation can contribute to cartilage degeneration and bone remodeling (17). These findings highlight the potential of targeting the gut microbiome and Wnt/β-catenin signaling as novel therapeutic strategies for OA.

The present study aims to provide a comprehensive review of the emerging evidence supporting the gut-bone-cartilage axis in OA and the role of microbial regulation of Wnt/β-catenin signaling in joint remodeling. The current understanding of the influence of the gut microbiome on OA pathogenesis is summarized, discussing the mechanisms underlying the gut-bone-cartilage axis and exploring the therapeutic potential of targeting this axis. By integrating insights from various studies, the present review aims to provide information on novel options for OA treatment and management.

Gut microbiota-derived metabolites: Regulators of Wnt/β-catenin signaling

The gut microbiome, comprising a diverse array of microorganisms, serves a crucial role in host metabolism and immune function (18). These microorganisms, which include bacteria, fungi and viruses, reside primarily in the gastrointestinal tract and contribute to the breakdown of dietary fibers, the production of vitamins and the regulation of immune responses (19). Among the various metabolites produced by the gut microbiome, short-chain fatty acids (SCFAs), bile acids and tryptophan derivatives have garnered marked attention due to their systemic effects on bone and cartilage metabolism (20–22) (Table I).

Table I.

Gut microbial metabolites and their regulation of the Wnt/β-catenin pathway in OA.

Table I.

Gut microbial metabolites and their regulation of the Wnt/β-catenin pathway in OA.

First author, yearMetabolite categorySpecific metaboliteSource of metaboliteMechanism of action on Wnt/β-catenin pathwayModel/study typeKey findings(Refs.)
Montalvany-Antonucci et al, 2019SCFAsButyrateProduced by gut microbiota through fermentation of dietary fibersInhibits HDACs, enhancing β-catenin stability; this leads to increased expression of Wnt target genes in osteoblasts, promoting bone formationAnimal modelButyrate supplementation reduces cartilage degradation and inflammation in animal models of OA by modulating the Wnt/β-catenin pathway(20)
Hou et al, 2023Bile acidsSecondary bile acidsMetabolized by gut microbiota from primary bile acids Wnt-induced inflammationFXR activation inhibits the Wnt/β-catenin pathway in osteoblasts, reducing bone resorption; in chondrocytes, it suppressesMechanistic studyAltered bile acid metabolism is associated with changes in bone density and cartilage integrity in patients with OA(21)
Lucas et al, 2018Tryptophan derivativesIndole-3-propionic acidProduced by gut microbiota from tryptophanActs as an antioxidant and modulates Wnt/β-catenin signaling by interacting with aryl hydrocarbon receptors, reducing inflammatory responses in chondrocytesCell cultureIndole-3-propionic acid protects chondrocytes from oxidative stress and inhibits Wnt/β-catenin pathway overactivation(22)
Lee et al, 2019SCFAsPropionateProduced by gut microbiota through fermentation of dietary fibersIt upregulates Wnt inhibitors like DKK1, reducing Wnt activation in osteoblasts and chondrocytesAnimal modelPropionate supplementation has a protective effect on cartilage in animal models of OA(25)
Rios et al, 2019Bile acidsDeoxycholic acidMetabolized by gut microbiota from primary bile acidsActivates TLR4 and NF-κB pathways, leading to increased production of pro-inflammatory cytokinesAnimal modelDeoxycholic acid administration is associated with exacerbated joint inflammation and cartilage damage in animal models of OA(26)
Guan et al, 2020Vitamin metabolitesVitamin K2Influenced by gut microbiota metabolismVitamin K2 deficiency decreases osteocalcin activation, impairing Wnt-mediated bone formationAnimal modelDysbiosis affects vitamin K2 availability, impacting bone metabolism and potentially contributing to OA(27)
Collins et al, 2021LPSGram-negative bacterial cell wall componentReleased by gut microbiota, particularly in dysbiosisActivates TLR4 pathway, leading to pro-inflammatory cytokine productionAnimal modelElevated LPS levels are associated with increased joint inflammation and cartilage damage in animal models of OA(28)
Pedersini et al, 2020Vitamin metabolitesVitamin DInfluenced by gut microbiota, metabolismRegulates Wnt/β-catenin signaling by enhancing the expression of vitamin D receptor and influencing the activity of related proteinsAnimal modelVitamin D deficiency is associated with exacerbated OA progression while supplementation improves joint health(29)
Xie et al, 2020LPSLipid AReleased by gut microbiota, particularly in dysbiosisActivates TLR4 and NF-κB pathways, leading to increased production of pro-inflammatory cytokinesAnimal modelLipid A administration is associated with increased joint inflammation and cartilage damage in animal models of OA(33)

[i] DKK1, Dickkopf-1; FXR, farnesoid X receptor; HDACs, histone deacetylases; LPS, lipopolysaccharide; OA, osteoarthritis; SCFAs, short-chain fatty acids; TLR4, Toll-like receptor 4.

Key metabolites linking the microbiota to the Wnt pathway [SCFAs, bile acids and lipopolysaccharide (LPS)]

The gut microbiome is characterized by its high diversity, with thousands of different species coexisting in a complex ecosystem (19). Bacteria from the Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria phyla are the most abundant. These microorganisms perform a variety of functions, including the fermentation of indigestible carbohydrates to produce SCFAs, such as acetate, propionate and butyrate (20). These SCFAs not only serve as an energy source for colonic epithelial cells but also have systemic effects on metabolism and inflammation (18,22) (Fig. 1).

The gut-bone-cartilage triad.
Schematic representation of the gut-bone-cartilage triad,
illustrating the interactions between gut microbiota, bone and
cartilage in OA. This figure was generated by Figdraw (www.figdraw.com; ID no. TIAWO27ea2). OA,
osteoarthritis.

Figure 1.

The gut-bone-cartilage triad. Schematic representation of the gut-bone-cartilage triad, illustrating the interactions between gut microbiota, bone and cartilage in OA. This figure was generated by Figdraw (www.figdraw.com; ID no. TIAWO27ea2). OA, osteoarthritis.

The metabolites produced by the gut microbiome notably impact bone metabolism, influencing both bone formation and resorption (23,24). SCFAs, particularly butyrate, have been shown to regulate bone density through multiple mechanisms. Butyrate inhibits osteoclast differentiation, thereby reducing bone resorption and protecting bone density (25). This effect is mediated by the inhibition of histone deacetylases (HDACs), which serve a crucial role in osteoclastogenesis. On the other hand, LPS, a component of the outer membrane of gram-negative bacteria, can promote bone resorption by activating the Toll-like receptor 4 (TLR4) pathway. This leads to the production of pro-inflammatory cytokines such as IL-1β and TNF-α, which stimulate osteoclast activity and bone resorption (26) (Fig. 1).

Vitamin metabolism is another area where the gut microbiome exerts its influence on bone health. Vitamin K2, essential for the activation of bone matrix proteins such as osteocalcin, is influenced by the gut microbiome. Guan et al (27) showed that alterations in the gut microbiome can affect the availability of vitamin K2, thereby impacting bone metabolism (Fig. 1).

Metabolite-driven Wnt activation/inhibition mechanisms

The gut microbiome also indirectly influences cartilage metabolism through the induction of systemic inflammation. Dysbiosis, or an imbalance in the gut microbiome, has been associated with increased levels of pro-inflammatory cytokines such as IL-1β and TNF-α (28). These cytokines can promote the degradation of cartilage matrix components, such as collagen and proteoglycans, by stimulating the production of matrix metalloproteinases (MMPs) and aggrecanases in chondrocytes (29). This inflammatory cascade accelerates cartilage degeneration and contributes to the progression of OA.

Moreover, the gut microbiome is implicated in the development of metabolic inflammation and obesity-related OA (30). Obesity is an important risk factor for OA, and studies have shown that the gut microbiome can influence energy metabolism and fat storage (31,32). Dysbiosis can lead to increased energy harvest from the diet and promote adipose tissue expansion, contributing to obesity (33). The resulting mechanical stress on joints, combined with systemic inflammation, exacerbates cartilage degeneration and joint damage in OA.

Butyrate (from Firmicutes) inhibits HDACs, enhancing β-catenin nuclear translocation and activating Wnt signaling in osteoblasts (20). Conversely, LPS (from gram-negative bacteria) binds TLR4, suppressing Wnt via NF-κB-induced sclerostin expression (34), promoting osteoclastogenesis and cartilage breakdown.

Notably, the influence of the gut microbiota on Wnt/β-catenin signaling is not limited to direct metabolic products. The gut microbiota also modulates the immune response of the host, which in turn affects Wnt/β-catenin signaling. For example, probiotics such as Lactobacillus have been shown to modulate the immune system and reduce inflammation by regulating the balance between T helper 17 (Th17) and regulatory T (Treg) cells, thereby inhibiting the overactivation of the Wnt/β-catenin pathway (35). This immune-mediated regulation provides another layer of complexity in the role of the gut microbiota in OA pathogenesis.

Additionally, a recent study has highlighted the role of bile acids in regulating Wnt/β-catenin signaling (21). Bile acids, upon metabolism by gut microbiota, can activate farnesoid X receptor (FXR) and other signaling pathways that intersect with Wnt/β-catenin (21). This crosstalk is particularly relevant in the context of bone and cartilage homeostasis, as evidenced by altered bile acid profiles in patients with OA, characterized by a significant decrease in the levels of secondary bile acids (such as deoxycholic acid and lithocholic acid) and a relative increase in primary bile acids (such as cholic acid and chenodeoxycholic acid) compared to healthy individuals (21).

This crosstalk is particularly relevant in the context of bone and cartilage homeostasis, as evidenced by studies showing altered bile acid profiles in patients with OA, characterized by a significant decrease in the levels of secondary bile acids (such as deoxycholic acid and lithocholic acid) and a relative increase in primary bile acids (such as cholic acid and chenodeoxycholic acid) compared to healthy individuals (21).

In summary, gut microbiota-derived metabolites exert notable effects on Wnt/β-catenin signaling through multiple mechanisms, involving direct metabolic actions, immune modulation and interactions with other signaling pathways. These findings underscore the multifaceted role of the gut microbiome in OA pathogenesis and highlight potential therapeutic targets for modulating joint remodeling in OA.

Wnt/β-catenin dysregulation: Direct driver of OA structural pathology

The Wnt/β-catenin signaling pathway serves a crucial role in bone formation, cartilage homeostasis and joint development (36). This pathway is involved in the regulation of cell proliferation, differentiation and survival, and it is thus essential in maintaining the structural integrity of the musculoskeletal system. In OA, the Wnt/β-catenin pathway exhibits a dual role, with both protective and pathological effects (36). Understanding this dual role is essential for developing targeted therapeutic strategies to manage OA progression (Fig. 2).

Wnt/β-catenin signaling pathway in
OA. The dual role of the Wnt/β-catenin signaling pathway in OA
exerts both protective and pathological effects. This figure was
generated by Figdraw (www.figdraw.com; ID no. YTOSUdf022). HDAC, histone
deacetylases; OA, osteoarthritis; SCFAs, short chain fatty
acids.

Figure 2.

Wnt/β-catenin signaling pathway in OA. The dual role of the Wnt/β-catenin signaling pathway in OA exerts both protective and pathological effects. This figure was generated by Figdraw (www.figdraw.com; ID no. YTOSUdf022). HDAC, histone deacetylases; OA, osteoarthritis; SCFAs, short chain fatty acids.

Wnt hyperactivation

The Wnt/β-catenin signaling pathway is a highly conserved pathway that regulates various cellular processes, including bone formation and cartilage homeostasis (36). In the canonical Wnt pathway, Wnt ligands bind to Frizzled (FZD) receptors and LRP5/6 co-receptors, leading to the accumulation of β-catenin in the cytoplasm (37,38). This accumulated β-catenin translocates to the nucleus, where it interacts with TCF/LEF transcription factors to regulate the expression of target genes involved in OA pathogenesis, such as MMP13, ADAMTS5 (a disintegrin and metalloproteinase with thrombospondin motifs 5), and cyclin D1 (37). These genes are involved in cell proliferation, differentiation and survival, making the Wnt/β-catenin pathway a critical regulator of bone and cartilage metabolism (38). β-catenin accumulation in chondrocytes, driven by canonical Wnt signaling activation, functions as a transcriptional co-activator that directly binds to T-cell factor/lymphoid enhancer factor (TCF/LEF) sites in the promoters of genes encoding catabolic enzymes. This leads to the upregulation of MMP13 and ADAMTS5 (39–41). MMP13 specifically cleaves type II collagen, the primary structural component of articular cartilage, while ADAMTS5 is the major aggrecanase responsible for aggrecan core protein degradation (39,40). This direct degradation of collagen II and aggrecan is a central mechanism driving cartilage matrix loss in OA. Elevated fecal LPS levels, indicative of gut dysbiosis and increased gram-negative bacterial load/intestinal permeability, promote systemic inflammation and can activate TLR4 signaling within joint tissues. TLR4 signaling has been shown to potentiate Wnt/β-catenin signaling and/or induce the expression of inflammatory cytokines (such as IL-1β and TNF-α) that further stimulate chondrocyte catabolism, including MMP13 and ADAMTS5 production (28,34). Thus, the clinical association between elevated fecal LPS levels and accelerated cartilage loss can be mechanistically explained, at least in part, by LPS-mediated enhancement of the Wnt/β-catenin pathway and downstream inflammatory cascades, culminating in the upregulation of these key matrix-degrading enzymes and subsequent cartilage destruction (28,34).

Wnt dysregulation

In OA, the Wnt/β-catenin pathway is often dysregulated, leading to both protective and pathological effects (39). At the chondrocyte level, excessive activation of the Wnt/β-catenin pathway can result in the hypertrophy of chondrocytes and the upregulation of MMPs, which contribute to cartilage degradation (40,41). This process is characterized by the loss of cartilage matrix components, such as collagen and proteoglycans, leading to reduced joint function and increased pain.

On the other hand, the Wnt/β-catenin pathway also serves a role in bone remodeling (42). β-catenin promotes the differentiation of osteoblasts, which are responsible for bone formation (43,44). However, abnormal activation of this pathway can lead to the formation of osteophytes, bony outgrowths that contribute to joint deformity and reduced mobility (45,46). This dual role of the Wnt/β-catenin pathway in OA highlights the complexity of its regulation and the need for targeted therapeutic interventions.

Regulation of the Wnt pathway by the gut microbiome

The gut microbiome has emerged as a key regulator of the Wnt/β-catenin pathway, influencing both bone and cartilage metabolism (47). Furthermore, the gut-brain axis may be involved in the indirect regulation of the Wnt/β-catenin pathway through neural communication, although further research is needed to elucidate its role in bone and cartilage metabolism (48). Overall, these diverse mechanisms highlight the complexity of how gut microbial metabolites modulate the Wnt/β-catenin signaling pathway in distant bones and cartilages, and underscore the potential of targeting this axis for OA treatment.

At the cellular level, the Wnt/β-catenin pathway exhibits distinct effects on different cell types. In osteoblasts, β-catenin promotes differentiation and bone formation by upregulating osteogenic genes such as Runx2 and Osterix (49). Conversely, in chondrocytes, excessive activation of the Wnt/β-catenin pathway can lead to hypertrophy and the upregulation of MMPs, which contribute to cartilage degradation (50). This cell-specific regulation underscores the complexity of the Wnt/β-catenin pathway in OA pathogenesis.

The gut microbiome can indirectly regulate the Wnt/β-catenin pathway through the immune-metabolic network. Dysbiosis, or an imbalance in the gut microbiome, can lead to increased levels of pro-inflammatory cytokines, such as IL-1β and TNF-α. These cytokines can influence the expression of Wnt pathway-related genes, such as sclerostin, an inhibitor of Wnt signaling (51). Furthermore, the Wnt/β-catenin pathway involves intricate feedback mechanisms. For example, the upregulation of Dickkopf-1 and secreted FZD-related protein acts as a negative feedback loop to modulate Wnt signaling intensity (52,53). These feedback mechanisms are essential for maintaining the balance of Wnt/β-catenin signaling in bone and cartilage homeostasis.

Gut-joint connection: Evidence, mechanisms and debates

The gut-bone-cartilage triad has emerged as a promising area of research in understanding the pathogenesis of OA (10). However, despite advancements, there are still controversies and gaps in the knowledge that need to be addressed. The present review aims to integrate the existing evidence and highlight the controversies in current research (Table II).

Table II.

Gut microbiota and OA-related studies.

Table II.

Gut microbiota and OA-related studies.

First author, yearGut microbiota category/interventionModel/study typeKey findings(Refs.)
Ulici et al, 2018Germ-free miceAnimal modelOA induced by destabilization of the medial meniscus is reduced in germ-free mice(54)
Chen et al, 2021FMTAnimal modelFMT from patients with OA to germ-free mice results in joint inflammation and cartilage damage(55)
Fortuna et al, 2021Comparative analysis of fecal microbiotaClinical studyPatients with OA show decreased gut microbiota diversity and specific bacterial species associated with disease severity(56)
Hahn et al, 2021Probiotic interventionClinical trialProbiotic supplementation improves OA symptoms in some patients but not in others(57)
Ilesanmi-Oyelere et al, 2021Gut microbiota and Wnt/β-catenin pathwayMechanistic studyMicrobial metabolites can directly modulate Wnt/β-catenin signaling in chondrocytes and osteoblasts(58)
Lan et al, 2021Time-dependent effect of gut microbiotaLongitudinal studyEarly-life exposure to specific gut microbiota may influence joint health later in life(59)
Luna et al, 2021Probiotic interventionAnimal model Lactobacillus can regulate the balance between Th17 and Treg cells, inhibiting the overactivation of the Wnt/β-catenin pathway(35)
Pedersini et al, 2021Prebiotic interventionAnimal modelA high-fiber diet increases the production of butyrate, reducing inflammation and cartilage degradation(60)
Mi et al, 2023High-fiber diet and gut microbiotaIntervention studyA high-fiber diet promotes the growth of beneficial gut bacteria and increases butyrate production, reducing inflammation and cartilage degradation(61)
Piva et al, 2023FMTAnimal modelFMT alleviates joint inflammation and cartilage degeneration by restoring a healthy gut microbiome(62)
Wang et al, 2023Small molecule inhibitorsAnimal modelICG-001 inhibits the Wnt/β-catenin pathway and decreases cartilage degeneration(63)
Zhao et al, 2024Gene editing (CRISPR/Cas9)Animal modelCRISPR/Cas9 editing of genes involved in the Wnt/β-catenin pathway, such as sclerostin, enhances bone formation and reduces cartilage degeneration(65)

[i] FMT, fecal microbiota transplantation; HFD, high-fat diet; OA, osteoarthritis; SCFAs, short-chain fatty acids; Th17, T helper 17; TLR4, toll-like receptor 4; Treg, regulatory T.

Evidence for the microbiota-metabolite-Wnt-structural axis

Rodent models have provided robust pre-clinical evidence linking the gut microbiome to OA pathogenesis. In germ-free C57BL/6 mice subjected to destabilization of the medial meniscus (DMM), Ulici et al (54) observed more severe cartilage degeneration and subchondral bone sclerosis than in specific-pathogen-free (SPF) controls, indicating that the absence of commensal microbes exacerbates OA-related joint damage. Complementing these findings, Chen et al (55) performed fecal microbiota transplantation (FMT) from human knee-OA patients into 8-week-old germ-free Swiss Webster mice and documented increased synovial inflammation and accelerated cartilage loss within eight weeks. Collins et al (28) employed high-fat diet (HFD)-fed C57BL/6J mice to model metabolic OA. The authors demonstrated that HFD-induced dysbiosis, characterized by reduced Lactobacillus and increased Desulfovibrionaceae, was associated with elevated systemic lipopolysaccharide (LPS) levels and greater cartilage erosion independent of body weight. In a rat model, Rios et al (26) demonstrated that prebiotic supplementation (oligofructose) in male Wistar rats mitigated HFD-driven joint inflammation by restoring gut-barrier integrity and reducing TLR4/NF-κB activation. Collectively, these murine and rat studies establish a causal role for the microbiome-joint axis in OA development and provide multiple translational platforms for mechanistic dissection.

Resolving controversies through a unified framework

While animal studies have provided evidence for the role of the gut microbiome in OA, clinical research has yielded contradictory results. Fortuna et al (56) reported a decrease in gut microbiota diversity in patients with OA, with specific bacterial species, such as Prevotella, showing a positive association with disease severity. However, other studies have failed to replicate these findings, suggesting that the relationship between gut microbiota and OA may be more complex (55,57). For instance, Chen et al reported no significant difference in α-diversity between older women with OA and healthy controls, and found no consistent association between specific bacterial taxa and OA status (55). Similarly, Hahn et al demonstrated that neither global microbiome diversity nor the relative abundance of Prevotella correlated with OA severity or pain scores in a mouse model (57), suggesting that the relationship between gut microbiota and OA may be more complex than initially thought.

Probiotic interventions have also shown inconsistent results in clinical trials. Hahn et al (57) reported improvements in OA symptoms following probiotic supplementation, whereas Fortuna et al (56) observed no significant changes in knee joint function or systemic inflammation after 12-week prebiotic/probiotic intervention in obese adults with knee OA, and Ilesanmi-Oyelere et al (58) found no notable effects of synbiotic supplementation on OA-related biomarkers in a randomized controlled trial. These discrepancies may be attributed to differences in study design, patient populations and the specific probiotic strains used.

Despite the growing body of evidence linking the gut microbiome to OA, gaps remain in the understanding of the underlying mechanisms. One key area of uncertainty is the cell-specific interaction between gut microbiota and the Wnt/β-catenin pathway. Ilesanmi-Oyelere et al (58) suggested that microbial metabolites can directly modulate Wnt/β-catenin signaling in chondrocytes and osteoblasts, but the exact cell types and molecular mechanisms involved remain unclear.

Another area of controversy is the time-dependent effect of gut microbiota on OA development. Lan et al (59) suggested that early-life exposure to specific gut microbiota may influence joint health later in lif. However, subsequent germ-free and antibiotic perturbation studies by Ulici et al (54) and Chen et al (55) failed to detect any significant association between neonatal microbial composition and OA susceptibility in adulthood, indicating that the role of host developmental stages in modulating the impact of gut microbiota on OA progression remains to be elucidated.

Therapeutic strategies and translational medicine prospects

The gut-bone-cartilage triad and the role of microbial regulation of Wnt/β-catenin signaling in OA joint remodeling offer promising options for therapeutic interventions. The present review aims to explore various strategies targeting the gut microbiome and the Wnt/β-catenin pathway, as well as the potential of gene editing and carrier technologies in OA treatment (Table II). Therapies targeting the microbiota-metabolite-Wnt axis (such as probiotics increasing butyrate/Wnt inhibition and FMT restoring anti-inflammatory taxa) show promise in normalizing joint remodeling (35,60,61).

Targeting the gut microbiome
Probiotics and prebiotics

Probiotics, such as Lactobacillus, have shown potential in modulating the immune system and reducing inflammation. Luna et al (35) demonstrated that Lactobacillus can regulate the balance between Th17 and Treg cells, thereby inhibiting the overactivation of the Wnt/β-catenin pathway. Prebiotics, which are non-digestible food components that promote the growth of beneficial gut bacteria, can also serve a role in OA management. For example, a high-fiber diet has been shown to increase the production of butyrate, a SCFA that inhibits β-catenin signaling, thereby reducing inflammation and cartilage degradation (60).

Diet control

Dietary interventions are a promising approach to modulating the gut microbiome and its effects on OA. A high-fiber diet has been shown to promote the growth of beneficial gut bacteria, leading to increased production of butyrate, which can inhibit β-catenin signaling and reduce inflammation (61). Additionally, a balanced diet rich in antioxidants and anti-inflammatory nutrients can mitigate the effects of OA by reducing systemic inflammation and promoting joint health (61).

FMT

FMT, which involves transferring fecal microbiota from a healthy donor to a recipient, has shown promising results in OA animal models. Piva et al (62) demonstrated that FMT can alleviate joint inflammation and cartilage degeneration by restoring a healthy gut microbiome. This approach holds potential for clinical applications, although further research is needed to optimize the procedure and ensure its safety and efficacy.

Targeting the Wnt pathway with small molecule drugs

The Wnt/β-catenin pathway has emerged as a promising target for OA therapy. Small molecule inhibitors, such as ICG-001, have been shown to inhibit the Wnt/β-catenin pathway and reduce cartilage degeneration (63). However, the use of inhibitors must be balanced against the potential for adverse effects, as the Wnt/β-catenin pathway serves a crucial role in bone formation and homeostasis (36). On the other hand, agonists, such as lithium salts, can promote bone formation by activating the Wnt/β-catenin pathway (64). The therapeutic potential of these agents must be carefully evaluated in clinical trials to determine their safety and efficacy.

Gene editing technologies, such as CRISPR/Cas9, offer a powerful tool for modulating the Wnt/β-catenin pathway in OA (65). Zhao et al (65) demonstrated that CRISPR/Cas9 can be used to edit genes involved in the Wnt/β-catenin pathway, such as SCLEROSTIN, which is an inhibitor of Wnt signaling. Lentiviral vectors have been used to deliver CRISPR/Cas9 systems to target cells, enabling precise gene editing and modulation of the Wnt/β-catenin pathway. This approach holds potential for developing novel therapies for OA, although further research is needed to address technical challenges, and ensure the safety and efficacy of these technologies.

Critical analysis of intervention strategies

When evaluating the proposed intervention strategies for OA targeting the gut-bone-cartilage triad and Wnt/β-catenin signaling, a comprehensive analysis of feasibility, specificity, safety and ethical considerations is essential.

Gene editing technologies

Gene editing tools such as CRISPR/Cas9 offer precise modulation of the Wnt/β-catenin pathway. While preclinical studies show promise in enhancing bone formation and reducing cartilage degradation, several concerns must be addressed. The specificity of CRISPR/Cas9 remains a challenge due to potential off-target effects, which could lead to unintended genetic modifications in various cell types (66). Safety concerns include the risk of immune responses to the Cas9 protein and the long-term consequences of altering gene expression. Ethical considerations revolve around the permanent nature of germline editing, even though somatic cell editing is primarily used for OA. The irreversible changes to the genome necessitate the careful balancing of risks and benefits (67).

Targeting the gut microbiome

Interventions targeting the gut microbiome, such as probiotics, prebiotics, FMT and dietary modifications, show potential for regulating the Wnt/β-catenin pathway. These approaches are relatively feasible and have fewer ethical concerns compared with gene editing. However, challenges remain in achieving specificity, as the complex composition of the gut microbiome makes it difficult to target specific bacterial strains without affecting the overall microbial balance (68). Safety concerns include the risk of introducing pathogenic bacteria through FMT or altering the microbiome in ways that could lead to other health issues (69). Long-term studies are needed to assess the safety and effectiveness of these interventions.

Small molecule drugs

Small molecule inhibitors and agonists of the Wnt/β-catenin pathway provide another option for OA treatment. These drugs are relatively easy to administer and have well-established pharmacokinetic profiles, enhancing their feasibility (37). However, ensuring their specificity to the Wnt/β-catenin pathway is challenging, as they may interact with other signaling pathways. Safety concerns involve potential toxicities and side effects, particularly with long-term use (37,70). Rigorous clinical trials are necessary to evaluate their safety and efficacy in patients with OA.

Future research directions and challenges

The gut-bone-cartilage triad and the microbial regulation of Wnt/β-catenin signaling in OA joint remodeling present opportunities for future research and therapeutic development (71,72). However, several challenges need to be addressed to fully realize the potential of these approaches. The present review explores future research directions and challenges in this field.

One of the most promising future directions in OA research is the integration of multiomics approaches, including metagenomics, metabolomics and single-cell sequencing (73,74). These techniques can provide a comprehensive understanding of the complex interactions between the gut microbiome and the host, particularly in the context of OA (74). Metagenomics can identify specific microbial species and their functional capabilities, whereas metabolomics can reveal the metabolic products of these microorganisms and their effects on host metabolism (75). Single-cell sequencing can provide insights into the heterogeneity of cell populations within the joint, including chondrocytes, osteoblasts and immune cells, and how they respond to microbial signals (76). For example, SCFAs produced by the gut microbiota can influence bone metabolism by enhancing the bone morphogenetic protein, Wnt and osteoprotegerin signaling pathways, and inhibiting the RANKL signaling pathway (20,77,78). By integrating these multiomics approaches, researchers can identify key microbial species and their metabolites that serve a role in OA progression, and develop targeted therapies to modulate these interactions.

Personalized medicine, which is based on the host genotype and gut microbiome characteristics of the host, holds potential for OA treatment (79). The gut microbiome varies among individuals and this variability can influence the efficacy of therapeutic interventions. By understanding the specific microbial composition and functional capabilities of the gut microbiome of individuals, clinicians can tailor treatments to target the specific pathways involved in OA progression (14,16,17). For example, studies have shown that certain bacterial species, such as Lactobacillus rhamnosus GG, can increase the production of the osteogenic Wnt ligand Wnt10b, promoting bone formation (80). By identifying individuals with specific microbial profiles, clinicians can recommend personalized interventions, such as probiotics or prebiotics, to modulate the gut microbiome and improve joint health.

The development of new therapeutic strategies for OA requires the integration of cross-disciplinary technologies, including gene editing and carrier technologies (81,82). Carrier technologies, such as lentiviral vectors, can be used to deliver CRISPR/Cas9 systems to target cells, enabling precise gene editing and modulation of the Wnt/β-catenin pathway (83). These technologies can be further optimized to enhance their safety and efficacy, making them viable options for clinical application.

Conclusion

The gut-bone-cartilage triad and the microbial regulation of Wnt/β-catenin signaling in OA joint remodeling offer promising opportunities for future research and therapeutic development. By integrating multiomics approaches, personalized medicine and cross-disciplinary technologies, researchers may gain a deeper understanding of the complex interactions between the gut microbiome and the host, and develop targeted therapies to modulate these interactions. Future research should focus on addressing the challenges in these areas to fully realize the potential of these approaches in OA treatment.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

RZ, LZ, YW, BT, XK and JZ conceived the study. RZ and JZ performed the literature review. RZ, LZ, YW, BT and XK wrote the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wang Z, Xiao Z, Sun C, Xu G and He J: Global, regional and national burden of osteoarthritis in 1990–2021: A systematic analysis of the global burden of disease study 2021. BMC Musculoskelet Disord. 25:10212024. View Article : Google Scholar : PubMed/NCBI

2 

Laires PA, Canhão H, Rodrigues AM, Eusébio M, Gouveia M and Branco JC: The impact of osteoarthritis on early exit from work: Results from a population-based study. BMC Public Health. 18:4722018. View Article : Google Scholar : PubMed/NCBI

3 

Peng R, Shang J, Jiang N, Chi-Jen H, Gu Y, Xing B, Hu R, Wu B, Wang D, Xu X and Lu H: Klf10 is involved in extracellular matrix calcification of chondrocytes alleviating chondrocyte senescence. J Transl Med. 22:522024. View Article : Google Scholar : PubMed/NCBI

4 

Akkiraju H and Nohe A: Role of chondrocytes in cartilage formation, progression of osteoarthritis and cartilage regeneration. J Dev Biol. 3:177–192. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Cho Y, Jeong S, Kim H, Kang D, Lee J, Kang SB and Kim JH: Disease-modifying therapeutic strategies in osteoarthritis: Current status and future directions. Exp Mol Med. 53:1689–1696. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Fleischmann R: Have we found a true disease-modifying osteoarthritis drug (DMOAD) or is there still much work to be done? Rheumatology (Oxford). 64:2345–2346. 2025. View Article : Google Scholar : PubMed/NCBI

7 

Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S and Xiao G: Osteoarthritis: Pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther. 8:562023. View Article : Google Scholar : PubMed/NCBI

8 

Guan M, Yu Q, Zhou G, Wang Y, Yu J, Yang W and Li Z: Mechanisms of chondrocyte cell death in osteoarthritis: Implications for disease progression and treatment. J Orthop Surg Res. 19:5502024. View Article : Google Scholar : PubMed/NCBI

9 

Deng H, Xue P, Zhou X, Wang Y and Liu W: CCL4/CCR5 regulates chondrocyte biology and OA progression. Cytokine. 183:1567462024. View Article : Google Scholar : PubMed/NCBI

10 

Hao X, Shang X, Liu J, Chi R, Zhang J and Xu T: The gut microbiota in osteoarthritis: Where do we stand and what can we do? Arthritis Res Ther. 23:422021. View Article : Google Scholar : PubMed/NCBI

11 

Chisari E, Wouthuyzen-Bakker M, Friedrich AW and Parvizi J: The relation between the gut microbiome and osteoarthritis: A systematic review of literature. PLoS One. 16:e02613532021. View Article : Google Scholar : PubMed/NCBI

12 

Nigam M, Devi K, Coutinho HDM and Mishra AP: Exploration of gut microbiome and inflammation: A review on key signalling pathways. Cell Signal. 118:1111402024. View Article : Google Scholar : PubMed/NCBI

13 

Li J, Ho WTP, Liu C, Chow SK, Ip M, Yu J, Wong HS, Cheung WH, Sung JJY and Wong RMY: The role of gut microbiota in bone homeostasis. Bone Joint Res. 10:51–59. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Lyu Z, Hu Y, Guo Y and Liu D: Modulation of bone remodeling by the gut microbiota: A new therapy for osteoporosis. Bone Res. 11:312023. View Article : Google Scholar : PubMed/NCBI

15 

Marchese L, Contartese D, Giavaresi G, Di Sarno L and Salamanna F: The complex interplay between the gut microbiome and osteoarthritis: A systematic review on potential correlations and therapeutic approaches. Int J Mol Sci. 25:1432023. View Article : Google Scholar : PubMed/NCBI

16 

Yu F, Zhu C and Wu W: Senile osteoarthritis regulated by the gut microbiota: From mechanisms to treatments. Int J Mol Sci. 26:15052025. View Article : Google Scholar : PubMed/NCBI

17 

Wang Y, Fan X, Xing L and Tian F: Wnt signaling: A promising target for osteoarthritis therapy. Cell Commun Signal. 17:972019. View Article : Google Scholar : PubMed/NCBI

18 

Yang W and Cong Y: Gut microbiota-derived metabolites in the regulation of host immune responses and immune-related inflammatory diseases. Cell Mol Immunol. 18:866–877. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Grant ET, Parrish A, Boudaud M, Hunewald O, Hirayama A, Ollert M, Fukuda S and Desai MS: Dietary fibers boost gut microbiota-produced B vitamin pool and alter host immune landscape. Microbiome. 12:1792024. View Article : Google Scholar : PubMed/NCBI

20 

Montalvany-Antonucci CC, Duffles LF, de Arruda JAA, Zicker MC, de Oliveira S, Macari S, Garlet GP, Madeira MFM, Fukada SY, Andrade I Jr, et al: Short-chain fatty acids and FFAR2 as suppressors of bone resorption. Bone. 125:112–121. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Hou Y, Li J and Ying S: Tryptophan metabolism and gut microbiota: A novel regulatory axis integrating the microbiome, immunity, and cancer. Metabolites. 13:11662023. View Article : Google Scholar : PubMed/NCBI

22 

Lucas S, Omata Y, Hofmann J, Böttcher M, Iljazovic A, Sarter K, Albrecht O, Schulz O, Krishnacoumar B, Krönke G, et al: Short-chain fatty acids regulate systemic bone mass and protect from pathological bone loss. Nat Commun. 9:552018. View Article : Google Scholar : PubMed/NCBI

23 

Yan J, Herzog JW, Tsang K, Brennan CA, Bower MA, Garrett WS, Sartor BR, Aliprantis AO and Charles JF: Gut microbiota induce IGF-1 and promote bone formation and growth. Proc Natl Acad Sci USA. 113:E7554–E7563. 2016. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Lee JY, Mannaa M, Kim Y, Kim J, Kim GT and Seo YS: Comparative analysis of fecal microbiota composition between rheumatoid arthritis and osteoarthritis patients. Genes (Basel). 10:7482019. View Article : Google Scholar : PubMed/NCBI

26 

Rios JL, Bomhof MR, Reimer RA, Hart DA, Collins KH and Herzog W: Protective effect of prebiotic and exercise intervention on knee health in a rat model of diet-induced obesity. Sci Rep. 9:38932019. View Article : Google Scholar : PubMed/NCBI

27 

Guan Z, Jia J, Zhang C, Sun T, Zhang W, Yuan W, Leng H and Song C: Gut microbiome dysbiosis alleviates the progression of osteoarthritis in mice. Clin Sci (Lond). 134:3159–3174. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Collins KH, Schwartz DJ, Lenz KL, Harris CA and Guilak F: Taxonomic changes in the gut microbiota are associated with cartilage damage independent of adiposity, high fat diet, and joint injury. Sci Rep. 11:145602021. View Article : Google Scholar : PubMed/NCBI

29 

Pedersini P, Turroni S and Villafañe JH: Gut microbiota and physical activity: Is there an evidence-based link? Sci Total Environ. 727:1386482020. View Article : Google Scholar : PubMed/NCBI

30 

Cani PD and Van Hul M: Gut microbiota in overweight and obesity: Crosstalk with adipose tissue. Nat Rev Gastroenterol Hepatol. 21:164–183. 2024. View Article : Google Scholar : PubMed/NCBI

31 

Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, Geurts L, Naslain D, Neyrinck A, Lambert DM, et al: Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 58:1091–1103. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Collins KH, Paul HA, Reimer RA, Seerattan RA, Hart DA and Herzog W: Relationship between inflammation, the gut microbiota, and metabolic osteoarthritis development: Studies in a rat model. Osteoarthritis Cartilage. 23:1989–1998. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Xie LL, Zhao YL, Yang J, Cheng H, Zhong ZD, Liu YR and Pang XL: Electroacupuncture prevents osteoarthritis of high-fat diet-induced obese rats. Biomed Res Int. 2020:93809652020. View Article : Google Scholar : PubMed/NCBI

34 

Zeng XZ, Zhang YY, Yang Q, Wang S, Zou BH, Tan YH, Zou M, Liu SW and Li XJ: Artesunate attenuates LPS-induced osteoclastogenesis by suppressing TLR4/TRAF6 and PLCγ1-Ca2+-NFATc1 signaling pathway. Acta Pharmacol Sin. 41:229–236. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Luna M, Guss JD, Vasquez-Bolanos LS, Alepuz AJ, Dornevil S, Strong J, Alabi D, Shi Q, Pannellini T, Otero M, et al: Obesity and load-induced posttraumatic osteoarthritis in the absence of fracture or surgical trauma. J Orthop Res. 39:1007–1016. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Hu L, Chen W, Qian A and Li YP: Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res. 12:392024. View Article : Google Scholar : PubMed/NCBI

37 

Maurice MM and Angers S: Mechanistic insights into Wnt-β-catenin pathway activation and signal transduction. Nat Rev Mol Cell Biol. 26:371–388. 2025. View Article : Google Scholar : PubMed/NCBI

38 

Ren DN, Chen J, Li Z, Yan H, Yin Y, Wo D, Zhang J, Ao L, Chen B, Ito TK, et al: LRP5/6 directly bind to Frizzled and prevent Frizzled-regulated tumour metastasis. Nat Commun. 6:69062015. View Article : Google Scholar : PubMed/NCBI

39 

Ma B, van Blitterswijk CA and Karperien M: A Wnt/β-catenin negative feedback loop inhibits interleukin-1-induced matrix metalloproteinase expression in human articular chondrocytes. Arthritis Rheum. 64:2589–2600. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Landman EB, Miclea RL, van Blitterswijk CA and Karperien M: Small molecule inhibitors of WNT/β-catenin signaling block IL-1β- and TNFα-induced cartilage degradation. Arthritis Res Ther. 15:R932013. View Article : Google Scholar : PubMed/NCBI

41 

Takamatsu A, Ohkawara B, Ito M, Masuda A, Sakai T, Ishiguro N and Ohno K: Verapamil protects against cartilage degradation in osteoarthritis by inhibiting Wnt/β-catenin signaling. PLoS One. 9:e926992014. View Article : Google Scholar : PubMed/NCBI

42 

Bao Q, Chen S, Qin H, Feng J, Liu H, Liu D, Li A, Shen Y, Zhao Y, Li J and Zong Z: An appropriate Wnt/β-catenin expression level during the remodeling phase is required for improved bone fracture healing in mice. Sci Rep. 7:26952017. View Article : Google Scholar : PubMed/NCBI

43 

Wang X, Qu Z, Zhao S, Luo L and Yan L: Wnt/β-catenin signaling pathway: Proteins' roles in osteoporosis and cancer diseases and the regulatory effects of natural compounds on osteoporosis. Mol Med. 30:1932024. View Article : Google Scholar : PubMed/NCBI

44 

Bao Q, Chen S, Qin H, Feng J, Liu H, Liu D, Li A, Shen Y, Zhong X, Li J and Zong Z: Constitutive β-catenin activation in osteoblasts impairs terminal osteoblast differentiation and bone quality. Exp Cell Res. 350:123–131. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Salazar VS, Mbalaviele G and Civitelli R: The pro-osteogenic action of beta-catenin requires interaction with BMP signaling, but not Tcf/Lef transcriptional activity. J Cell Biochem. 104:942–952. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Feng SY, Cao MN, Gao CC, Li YX, Lei J and Fu KY: Akt2 inhibition alleviates temporomandibular joint osteoarthritis by preventing subchondral bone loss. Arthritis Res Ther. 27:432025. View Article : Google Scholar : PubMed/NCBI

47 

Hao J, Liu C, Gu Z, Yang X, Lan X and Guo X: Dysregulation of Wnt/β-catenin signaling contributes to intestinal inflammation through regulation of group 3 innate lymphoid cells. Nat Commun. 15:28202024. View Article : Google Scholar : PubMed/NCBI

48 

Ohara TE and Hsiao EY: Microbiota-neuroepithelial signalling across the gut-brain axis. Nat Rev Microbiol. 23:371–384. 2025. View Article : Google Scholar : PubMed/NCBI

49 

Zhou X, Zhang Z, Feng JQ, Dusevich VM, Sinha K, Zhang H, Darnay BG and de Crombrugghe B: Multiple functions of Osterix are required for bone growth and homeostasis in postnatal mice. Proc Natl Acad Sci USA. 107:12919–12924. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Usami Y, Gunawardena AT, Iwamoto M and Enomoto-Iwamoto M: Wnt signaling in cartilage development and diseases: Lessons from animal studies. Lab Invest. 96:186–196. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Sharvari S, Meganathan P and Vedagiri H: Gut microbial dysbiosis induced exacerbations influence the progression of colorectal cancer. Biol Bull Rev. 14:724–739. 2024. View Article : Google Scholar

52 

Mehta A, Motavaf M, Raza D, McLure AJ, Osei-Opare KD, Bordone LA and Gru AA: Revolutionary approaches to hair regrowth: Follicle neogenesis, Wnt/ß-catenin signaling, and emerging therapies. Cells. 14:7792025. View Article : Google Scholar : PubMed/NCBI

53 

Kwack MH, Sung YK, Chung EJ, Im SU, Ahn JS, Kim MK and Kim JC: Dihydrotestosterone-inducible dickkopf 1 from balding dermal papilla cells causes apoptosis in follicular keratinocytes. J Invest Dermatol. 128:262–269. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Ulici V, Kelley KL, Azcarate-Peril MA, Cleveland RJ, Sartor RB, Schwartz TA and Loeser RF: Osteoarthritis induced by destabilization of the medial meniscus is reduced in germ-free mice. Osteoarthritis Cartilage. 26:1098–1109. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Chen J, Wang A and Wang Q: Dysbiosis of the gut microbiome is a risk factor for osteoarthritis in older female adults: A case control study. BMC Bioinformatics. 22:2992021. View Article : Google Scholar : PubMed/NCBI

56 

Fortuna R, Hart DA, Sharkey KA, Schachar RA, Johnston K and Reimer RA: Effect of a prebiotic supplement on knee joint function, gut microbiota, and inflammation in adults with co-morbid obesity and knee osteoarthritis: Study protocol for a randomized controlled trial. Trials. 22:2552021. View Article : Google Scholar : PubMed/NCBI

57 

Hahn AK, Wallace CW, Welhaven HD, Brooks E, McAlpine M, Christiansen BA, Walk ST and June RK: The microbiome mediates epiphyseal bone loss and metabolomic changes after acute joint trauma in mice. Osteoarthritis Cartilage. 29:882–893. 2021. View Article : Google Scholar : PubMed/NCBI

58 

Ilesanmi-Oyelere BL, Roy NC and Kruger MC: Modulation of bone and joint biomarkers, gut microbiota, and inflammation status by synbiotic supplementation and weight-bearing exercise: Human study protocol for a randomized controlled trial. JMIR Res Protoc. 10:e301312021. View Article : Google Scholar : PubMed/NCBI

59 

Lan H, Hong W, Qian D, Peng F, Li H, Liang C, Du M, Gu J, Mai J, Bai B and Peng G: Quercetin modulates the gut microbiota as well as the metabolome in a rat model of osteoarthritis. Bioengineered. 12:6240–6250. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Pedersini P, Savoldi M, Berjano P and Villafañe JH: A probiotic intervention on pain hypersensitivity and microbiota composition in patients with osteoarthritis pain: Study protocol for a randomized controlled trial. Arch Rheumatol. 36:296–301. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Mi Y, Yi N, Xu X, Zeng F, Li N, Tan X, Gong Z, Yan K, Kuang G and Lu M: Prebiotics alleviate cartilage degradation and inflammation in post-traumatic osteoarthritic mice by modulating the gut barrier and fecal metabolomics. Food Funct. 14:4065–4077. 2023. View Article : Google Scholar : PubMed/NCBI

62 

Piva F, Gervois P, Karrout Y, Sané F and Romond MB: Gut-joint axis: Impact of bifidobacterial cell wall lipoproteins on arthritis development. Nutrients. 15:48612023. View Article : Google Scholar : PubMed/NCBI

63 

Wang X, Wu Y, Liu Y, Chen F, Chen S, Zhang F, Li S, Wang C, Gong Y, Huang R, et al: Altered gut microbiome profile in patients with knee osteoarthritis. Front Microbiol. 14:11534242023. View Article : Google Scholar : PubMed/NCBI

64 

Meffre D, Grenier J, Bernard S, Courtin F, Dudev T, Shackleford G, Jafarian-Tehrani M and Massaad C: Wnt and lithium: A common destiny in the therapy of nervous system pathologies? Cell Mol Life Sci. 71:1123–1148. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Zhao L, Lai Y, Jiao H, Li J, Lu K and Huang J: CRISPR-mediated Sox9 activation and RelA inhibition enhance cell therapy for osteoarthritis. Mol Ther. 32:2549–2562. 2024. View Article : Google Scholar : PubMed/NCBI

66 

Naeem M, Majeed S, Hoque MZ and Ahmad I: Latest developed strategies to minimize the off-target effects in CRISPR-cas-mediated genome editing. Cells. 9:16082020. View Article : Google Scholar : PubMed/NCBI

67 

Klermund J, Rhiel M, Kocher T, Chmielewski KO, Bischof J, Andrieux G, El Gaz M, Hainzl S, Boerries M, Cornu TI, et al: On- and off-target effects of paired CRISPR-Cas nickase in primary human cells. Mol Ther. 32:1298–1310. 2024. View Article : Google Scholar : PubMed/NCBI

68 

Hitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J and Clavel T: Microbiome-based interventions to modulate gut ecology and the immune system. Mucosal Immunol. 15:1095–1113. 2022. View Article : Google Scholar : PubMed/NCBI

69 

Porcari S, Fusco W, Spivak I, Fiorani M, Gasbarrini A, Elinav E, Cammarota G and Ianiro G: Fine-tuning the gut ecosystem: The current landscape and outlook of artificial microbiome therapeutics. Lancet Gastroenterol Hepatol. 9:460–475. 2024. View Article : Google Scholar : PubMed/NCBI

70 

Yu M, Yang Y, Sykes M and Wang S: Small-molecule inhibitors of tankyrases as prospective therapeutics for cancer. J Med Chem. 65:5244–5273. 2022. View Article : Google Scholar : PubMed/NCBI

71 

Zhong X, Zhang F, Yin X, Cao H, Wang X, Liu D, Chen J and Chen X: Bone homeostasis and gut microbial-dependent signaling pathways. J Microbiol Biotechnol. 31:765–774. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Ticinesi A, Siniscalchi C, Meschi T and Nouvenne A: Gut microbiome and bone health: Update on mechanisms, clinical correlations, and possible treatment strategies. Osteoporos Int. 36:167–191. 2025. View Article : Google Scholar : PubMed/NCBI

73 

Liu Y, Da W, Xu MJ, Xiao CX, Deng T, Zhou SL, Chen XT, Zhou YJ, Tang L, Nie Y, et al: Single-cell transcriptomics reveals novel chondrocyte and osteoblast subtypes and their role in knee osteoarthritis pathogenesis. Signal Transduct Target Ther. 10:402025. View Article : Google Scholar : PubMed/NCBI

74 

Li J, Yang X, Chu Q, Xie L, Ding Y, Xu X, Timko MP and Fan L: Multi-omics molecular biomarkers and database of osteoarthritis. Database (Oxford). 2022:baac0522022. View Article : Google Scholar : PubMed/NCBI

75 

Wang Y, Zeng T, Tang D, Cui H, Wan Y and Tang H: Integrated multi-omics analyses reveal lipid metabolic signature in osteoarthritis. J Mol Biol. 437:1688882025. View Article : Google Scholar : PubMed/NCBI

76 

Ji Q, Zheng Y, Zhang G, Hu Y, Fan X, Hou Y, Wen L, Li L, Xu Y, Wang Y and Tang F: Single-cell RNA-seq analysis reveals the progression of human osteoarthritis. Ann Rheum Dis. 78:100–110. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Feng B, Lu J, Han Y, Han Y, Qiu X and Zeng Z: The role of short-chain fatty acids in the regulation of osteoporosis: new perspectives from gut microbiota to bone health: A review. Medicine (Baltimore). 103:e394712024. View Article : Google Scholar : PubMed/NCBI

78 

Kumar SS, Fathima A, Srihari P and Jamma T: Host-gut microbiota derived secondary metabolite mediated regulation of Wnt/β-catenin pathway: A potential therapeutic axis in IBD and CRC. Front Oncol. 14:13925652024. View Article : Google Scholar : PubMed/NCBI

79 

Choi YR, Collins KH, Lee JW, Kang HJ and Guilak F: Genome engineering for osteoarthritis: From designer cells to disease-modifying drugs. Tissue Eng Regen Med. 16:335–343. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Tyagi AM, Yu M, Darby TM, Vaccaro C, Li JY, Owens JA, Hsu E, Adams J, Weitzmann MN, Jones RM and Pacifici R: The microbial metabolite butyrate stimulates bone formation via T regulatory cell-mediated regulation of WNT10B expression. Immunity. 49:1116–1131.e7. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Chaudhry N, Muhammad H, Seidl C, Downes D, Young DA, Hao Y, Zhu L and Vincent TL: Highly efficient CRISPR-Cas9-mediated editing identifies novel mechanosensitive microRNA-140 targets in primary human articular chondrocytes. Osteoarthritis Cartilage. 30:596–604. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Seidl CI, Fulga TA and Murphy CL: CRISPR-Cas9 targeting of MMP13 in human chondrocytes leads to significantly reduced levels of the metalloproteinase and enhanced type II collagen accumulation. Osteoarthritis Cartilage. 27:140–147. 2019. View Article : Google Scholar : PubMed/NCBI

83 

Ortinski PI, O'Donovan B, Dong X and Kantor B: Integrase-deficient lentiviral vector as an all-in-one platform for highly efficient CRISPR/Cas9-mediated gene editing. Mol Ther Methods Clin Dev. 5:153–164. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Zhang R, Zhang L, Tian B, Wang Y, Kang X and Zheng J: The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review). Mol Med Rep 33: 23, 2026.
APA
Zhang, R., Zhang, L., Tian, B., Wang, Y., Kang, X., & Zheng, J. (2026). The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review). Molecular Medicine Reports, 33, 23. https://doi.org/10.3892/mmr.2025.13733
MLA
Zhang, R., Zhang, L., Tian, B., Wang, Y., Kang, X., Zheng, J."The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review)". Molecular Medicine Reports 33.1 (2026): 23.
Chicago
Zhang, R., Zhang, L., Tian, B., Wang, Y., Kang, X., Zheng, J."The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review)". Molecular Medicine Reports 33, no. 1 (2026): 23. https://doi.org/10.3892/mmr.2025.13733
Copy and paste a formatted citation
x
Spandidos Publications style
Zhang R, Zhang L, Tian B, Wang Y, Kang X and Zheng J: The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review). Mol Med Rep 33: 23, 2026.
APA
Zhang, R., Zhang, L., Tian, B., Wang, Y., Kang, X., & Zheng, J. (2026). The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review). Molecular Medicine Reports, 33, 23. https://doi.org/10.3892/mmr.2025.13733
MLA
Zhang, R., Zhang, L., Tian, B., Wang, Y., Kang, X., Zheng, J."The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review)". Molecular Medicine Reports 33.1 (2026): 23.
Chicago
Zhang, R., Zhang, L., Tian, B., Wang, Y., Kang, X., Zheng, J."The gut‑bone‑cartilage triad: Microbial regulation of the Wnt/β‑catenin signaling pathway in osteoarthritis joint remodeling (Review)". Molecular Medicine Reports 33, no. 1 (2026): 23. https://doi.org/10.3892/mmr.2025.13733
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team