- Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
Degeneration of articular cartilage is the hallmark pathologic change in osteoarthritis (OA). Cartilage not only serves as a shock-absorbing structure for movement but is also regulated by organs other than bone, while chondrocytes secrete cytokines that influence these organs. The concept of organ axis refers to the regulatory pathways formed between organs via cytokine signaling. The communication network established between cartilage and other organs constitutes the cartilage-organ axis. Through this axis, these organs regulate articular chondrocyte proliferation and apoptosis. It is evident that chondrocytes play a central role in connecting various organs to the progression of OA, prompting interest in strategies to intervene in cartilage damage by modulating the organ axis. This review presents, for the first time, a system summarizing the effects of the extraosseous system on cartilage through various factors that alter OA progression. The aim is to fully elucidate the effects of different organs on cartilage, thus providing insights into the treatment of OA and systemic diseases.
1 Introduction
1.1 Pathologic changes of OA
OA is one of the fastest-growing health issues globally (1), with the knee being the most commonly affected joint. Knee osteoarthritis (KOA) is a chronic and disabling condition with multiple etiologies that impacts the entire knee joint (2, 3). The burden of OA continues to rise globally, with studies suggesting that the disease will affect approximately 7.6 per cent of the world’s population by 2024 (4), and KOA ranks as the 11th leading cause of disability globally (5), particularly as the population ages and the prevalence of obesity increases. As a bearing or hinge structure, joints not only provide attachment and support for muscles and ligaments but also facilitate various movements. Traditionally, the primary pathological change in OA has been recognized as the degeneration of articular cartilage. However, a more comprehensive perspective is now emerging, viewing OA as a disease that encompasses the entire joint structure, including cartilage, synovium, subchondral bone, ligaments, and menisci (6). Despite this broader understanding, degeneration of articular cartilage remains the earliest and most characteristic pathological change observed in the progression of OA. In this article, we aim to provide an overview of the pathological changes that occur in cartilage as OA advances. The degeneration of cartilage is the most characteristic pathological change observed in OA (7, 8). When articular cartilage is subjected to mechanical stress, inflammation, metabolic disturbances, immune responses, and genetic factors (9), signaling pathways such as Wnt/β-catenin, NF-κB, MAPK, and PI3K/AKT are abnormally activated in chondrocytes. This ultimately results in mitochondrial dysfunction, which leads to chondrocyte destruction (10). Under normal conditions, cartilage functions as a cushion and reduces joint friction. However, when cartilage degenerates, its surface integrity is compromised, resulting in the release of components such as proteoglycans and collagen from the cartilage matrix, which form ‘wear’ particles (11). These particles are phagocytosed by synovial macrophages, activating synovial cells to release various pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1 (IL-1), and interleukin-6 (IL-6), thereby triggering an inflammatory response in the synovium (12). Consequently, cartilage damage is exacerbated, and its protective effect on the subchondral bone is diminished, exposing the subchondral bone to abnormal mechanical stress (13). This altered mechanical stress can incite microfracture and repair responses in the subchondral bone. During the repair process, the activity of cells such as osteoblasts and osteoclasts increases, leading to the remodeling of the subchondral bone and resulting in changes such as osteophyte formation and osteosclerosis (14–16). In the pathogenesis of arthritis, cartilage degeneration, synovial changes, and subchondral bone alterations are interrelated and synergistic, creating a complex pathophysiologic network that drives disease progression. Cartilage is the first part to degenerate, so we focus on cartilage changes. The proliferation, degeneration, and apoptosis of chondrocytes are regulated by inter-organ crosstalk, indicating that the onset and progression of OA are also influenced by other organs (17–20).
1.2 Relationship between organs-crosstalk and OA
Since 2000, with the advancements in systems biology and multi-omics techniques, the term “organ crosstalk” has become increasingly prevalent. Originally, this concept was used to describe the intricate interactions between organs that transmit signaling molecules (e.g., hormones, cytokines, metabolites) via the circulatory system (e.g., blood, lymph) (21–28). Despite the complexity and diversity of inter-organ crosstalk mechanisms, recent years have seen significant progress in elucidating these processes (29, 30). The perspective of inter-organ crosstalk offers novel insights, providing innovative solutions for the treatment of various diseases (31). In recent years, continuous technological innovation has clarified the study of cross-system communication involving cytokines, extracellular vesicles, hormones, and neurotransmitters. This advancement enables scientists to analyze the complex interaction mechanisms between bones, joints, and various organs, including the brain, lungs, liver, heart, and kidneys, in greater depth. Consequently, it promotes the rapid development of research on the bone-organ axis.
Organ crosstalk alters chondrocyte metabolism by delivering cytokines and metabolites or by influencing macrophage polarization (32). Although numerous mechanisms have been identified, there is a notable lack of comprehensive summaries and systematic analyses in this research area. In recent years, significant interactions between organs such as the heart, brain, liver, and lungs with the osteoarticular system have been identified, profoundly affecting bone and cartilage metabolism and related pathologies (Tables 1, 2). Additionally, growing evidence suggests that organs including the intestines, kidneys, gonads, and pancreas also play a regulatory role in metabolic changes within articular cartilage (Figure 1). There is an emerging perspective that fat, muscle, and even bone itself may gradually influence the health of articular cartilage (Table 3). Although these tissues are not classified as organs, their ubiquitous presence in the body underscores their importance in research. Joint tissues, as a crucial component of the skeletal system, are also influenced by other internal organs during homeostasis and disease progression, with alterations in articular cartilage being particularly significant. A deeper understanding of the mechanisms underlying bone-organ-axis interactions could facilitate the development of targeted therapies for bone-related and systemic diseases. For instance, by focusing on key molecules or signaling pathways within a specific bone-organ axis, more effective therapeutic drugs and strategies can be devised to enhance treatment efficacy while minimizing adverse effects. In this review, we summarize the relevant mechanisms identified thus far, particularly those related to cartilage damage in OA. By understanding these mechanisms, we aim to provide insights for the prevention and treatment of OA.
2 Cross-talk between cartilage and other organs
2.1 Brain and peripheral nerve-cartilage axis
The influence of the nervous system on cartilage is paramount among all biological systems. The concept of the brain-bone axis has been proposed (33), highlighting the integral roles of both the central and peripheral nervous systems (34–37). Notably, alterations in circadian rhythms can also impact cartilage health. This section summarizes the effects of central and peripheral nerves on chondrocyte proliferation and apoptosis.
2.1.1 Brain and peripheral nerve to cartilage
The brain plays a vital role in regulating cartilage metabolism. This section describes the effects of cellular communication network factor 3 (CCN3), melatonin (MT), and parathyroid hormone (PTH), all of which are produced by the central nervous system and regulated by circadian rhythms, on cartilage. Study finds correlation between OA degenerative changes and CCN3 expression (11). CCN3, a member of the CCN (Cyr61, Ctgf, NOV) family, has been shown to regulate various inflammatory responses (54). Recent findings indicate that CCN3 ameliorates the apoptotic effects on chondrocytes through the activation of the PI3K/AKT/mTOR pathway and enhances the degradation of cartilage matrices (55), including collagen type II and aggregated proteoglycans (56). Furthermore, CCN3 has been identified as being secreted by KISS1 neurons in the arcuate nucleus (ARC KISS1), which increases the potential for bone and cartilage formation, thereby improving cartilage quality (57). The central nervous system is implicated in the regulation of circadian rhythms and has been shown to influence the hypothalamic-pituitary-adrenal axis and hormones, thereby impacting the progression of OA (58–60). Changes in serum concentrations of PTH, regulated by the central nervous system, exhibit diurnal variations, suggesting that cartilage proliferation is also influenced by daily activity rhythms (96). MT, a hormone secreted by the pineal gland, is regulated by circadian rhythms. It has been demonstrated that MT protects chondrocytes by upregulating hyaluronic acid synthesis, inhibiting the NF-kB signaling pathway, and repairing the redox environment and membrane potential within mitochondria (61).
Peripheral nerves not only mediate the production of nociception but also secrete factors that influence cartilage health, including substance P (SP), calcitonin gene-related peptide (CGRP), and semaphorin 3A (Sema3A), which regulate cartilage metabolism. Nerve Growth Factor (NGF) plays a critical role in the development of OA pain by stimulating sensory neurons to produce nociception, which is the primary source of pain in OA (62). Blocking NGF has been shown to reduce pain sensitivity in OA patients (108). Certain factors, such as SP and CGRP, are involved in both nociception production and cartilage metabolism (109, 110). They serve a dual purpose: transducing nociception and regulating chondrocyte metabolism. SP, a peptide primarily secreted by neurons (63), stimulates the secretion of pro-inflammatory chemokines by macrophages and mast cells (64). Additionally, in the context of tissue injury, SP promotes M2 polarization of macrophages, which mitigates inflammatory responses and inhibits chondrocyte apoptosis in OA (65). CGRP exhibits biphasic regulation of chondrocytes: in normal cartilage, it may protect chondrocytes from apoptosis by inhibiting the release of inflammatory factors (e.g., IL-1β, TNF-α), reducing oxidative stress, and counteracting the apoptotic effects of NGF by preventing the abnormal growth of peripheral sensory nerves into cartilage tissue (66). However, CGRP expression is elevated in OA cartilage, where it promotes inflammatory responses and degradation of the cartilage extracellular matrix, with excessive release in degenerating tissues potentially exacerbating chronic pain (67, 110). Sema3A, another protein secreted by sensory neurons (68), maintains cartilage homeostasis and inhibits nerve growth into cartilage during OA. It exerts a protective effect against cartilage degradation through the activation of the PI3K signaling pathway (69).
Neurodegenerative diseases, particularly Alzheimer’s disease (AD), have a significant interplay with OA, with evidence suggesting that AD may promote the development of OA. The pathogenesis of OA is closely associated with AD (33), as indicated by recent studies revealing that patients with KOA are at an elevated risk of developing AD-related dementia (ADRD) (122, 123). Current understanding of the mechanisms underlying the interaction between AD and OA highlights a notable increase in serum IL-6 levels in osteoarthritis, which subsequently leads to heightened levels of inflammatory cytokines in peripheral blood, contributing to brain inflammation (124).
2.1.2 Cartilage to brain
The mechanisms by which healthy chondrocytes influence the nervous system remain largely unexplored; however, cartilage in the context of OA significantly contributes to various neurological disorders. Degenerated chondrocytes associated with OA are implicated in the emergence of lesions characteristic of AD. During the progression of OA, peripheral inflammation persists, marked by elevated levels of inflammatory mediators such as IL-1β, IL-6, and TNF. Notably, these cytokines, along with IL-18, have the capacity to cross the blood-brain barrier (BBB) (125), potentially inducing brain lesions. Moreover, inflammatory cytokines resulting from chondrocyte damage may expedite the deposition of amyloid beta (Aβ) and the advancement of neuroinflammation (126–128). This process leads to the hyperphosphorylation of tau proteins and subsequent neuronal loss. The intracellular aggregation of abnormally phosphorylated tau proteins results in the formation of neurofibrillary tangles (NFTs), ultimately culminating in neuronal degeneration, which, when sufficiently severe, can lead to the development of Alzheimer’s disease (129).
While the potential treatment avenues mentioned above show promise for continued research, several challenges persist. These include the necessity for a more profound understanding of the intricate interactions between the nervous system and osteoarthritis (OA). Given the dynamic nature of brain and peripheral nerve functions, along with the significant individual differences that must be considered, a personalized medicine approach may be essential to optimize treatment outcomes. In other words, targeting cartilage improvement by modulating nervous system function for OA treatment represents an emerging field that may provide new opportunities for integrated brain-joint therapy. Nevertheless, further research is required to fully elucidate the mechanisms of action and to develop targeted and effective treatments.
2.2 Heart-cartilage axis
The correlation between cardiovascular disease (CVD) and OA has been well established, with numerous studies demonstrating that the comorbidities of either condition are directly and significantly associated with an increased risk of developing the other (130). Both conditions often exhibit common pathological mechanisms in their pathogenesis, such as oxidative stress, local microvascular remodeling, and hyperlipidemia. Additionally, the daily behavioral patterns of OA patients contribute to an increased incidence of CVD (131, 132). Similarly, reduced cardiac function can promote the progression of OA (133). There are also molecular mechanisms that interact between the two conditions (134–136), highlighting the importance of exploring the heart-cartilage axis as a potential avenue for new therapeutic options for OA.
2.2.1 Cardiovascular to cartilage
The cardiovascular system produces specific cytokines and vesicles that are modified and released into the bloodstream to influence chondrocytes. Cardiomyocytes exhibit high expression levels of a protein known as fibronectin type III domain-containing protein 5 (FNDC5) (38). FNDC5 is a transmembrane protein composed of an extracellular irisin structural domain and an intracytoplasmic C-terminal structural domain. Irisin, a soluble peptide cleaved from the extracellular structural domain of FNDC5 (39), has been shown to promote chondrocyte growth and extracellular matrix (ECM) accumulation by enhancing mitochondrial autophagy in chondrocytes (40, 41). Vascular endothelial growth factor (VEGF), primarily produced by vascular endothelial cells and the liver, has been implicated in exacerbating the progression of OA. Inhibition of VEGF expression not only enhances chondrogenic differentiation but also protects chondrocytes from tumor necrosis factor-alpha (TNF-α)-induced hypertrophic changes (49). Furthermore, exosomes derived from vascular endothelial cells (EC-Exos) act as negative regulators of chondrocytes, inhibiting chondrocyte autophagy. The entry of EC-Exos into chondrocytes elevates cellular reactive oxygen species (ROS) levels and induces apoptosis by compromising chondrocyte resistance to oxidative stress (95). Factors produced by cardiomyocytes or vascular endothelial cells are transported to the joints via the bloodstream. Collectively, the effects of FNDC5 and VEGF are well-established (137); however, the impact of exosomes produced by the cardiovascular system on chondrocytes remains unclear. Additionally, the influence of exosomes secreted by the cardiovascular system in various states on the joints has yet to be conclusively determined.
Metabolites associated with CVD)are significant factors influencing chondrocyte metabolism, particularly those resulting from abnormal lipid metabolism, which exert the most profound effects. CVD is characterized by elevated blood levels of low-density lipoprotein (LDL) and cholesterol. Excess LDL undergoes oxidation, resulting in oxidized LDL (oxLDL). Both oxLDL and cholesterol contribute to processes such as synovial inflammation, cartilage degeneration, and bone deformation (42–45). Nakagawa et al. demonstrated that oxLDL induces damage to chondrocytes in vitro (138), subsequent studies indicated that oxLDL enhances cartilage degradation by increasing the expression of monocyte chemoattractant protein 1(MCP-1) in chondrocytes (46). Furthermore, oxLDL impairs autophagic flux, leading to autophagic dysfunction and promoting chondrocyte apoptosis by elevating p62/SQSTM1 levels and inhibiting the activity of transcription factor EB (TFEB) (139). Elevated cholesterol levels significantly enhance synovial activation and heterotopic bone formation in early collagenase-induced OA (140, 141). In chondrocytes, cholesterol and its oxidative metabolites directly activate retinoic acid-related orphan receptor α (RORα), contributing to OA by upregulating matrix-degrading enzymes (142). Additionally, increased blood cholesterol levels, and consequently higher cholesterol concentrations in joint fluid, downregulate low-density lipoprotein receptor-related protein 3(LRP3) expression in chondrocytes while upregulating syndecan-4 through the activation of the Ras signaling pathway, thereby inducing degeneration of the cartilaginous ECM (47, 48). Disturbances in lipid metabolism within the cardiovascular system can lead to increased oxidative stress and exacerbated inflammatory responses. These changes may result in damage and degradation of articular cartilage, ultimately inducing OA.
Finally, we found that altered hemodynamics is also a significant indicator of chondrocyte activity. Recent statistical studies have demonstrated that hypertension and osteoarthritis are interrelated conditions (143). Articular cartilage lacks blood vessels and nerve endings; therefore, the nutrition necessary for its growth and maintenance primarily derives from the subchondral bone and synovium (144, 145). The synovium is richly vascularized and exhibits a high density of capillary distribution (146, 147). The metaphyseal trabecular bone within the subchondral region is also highly vascularized, containing a network of capillaries and sinuses (148, 149). Hypertension modifies the state of blood flow, leading to abnormal perfusion of these capillaries and sinuses, which in turn alters the hemodynamics of the subchondral bone and impairs the nutritional supply to the cartilage (150–152).
2.2.2 Cartilage to cardiovascular
Chondrocytes can influence the heart and blood vessels by regulating specific cytokines. In healthy chondrocytes, the proliferation and differentiation of vascular smooth muscle cells are modulated through the Wnt/β-catenin signaling pathway. MiR-214-3p, a microRNA secreted by chondrocytes, plays a significant role in this process. Overexpression of miR-214-3p inhibits endothelial cell migration and angiogenesis, a process mediated through the regulation of the TrkB pathway. Activation of TrkB results in elevated levels of VEGF, which promotes endothelial cell migration and angiogenesis. In contrast, in osteoarthritic chondrocytes, the expression of miR-214-3p is down-regulated while TrkB levels are up-regulated, leading to increased VEGF secretion that facilitates endothelial cell migration and angiogenesis (97). Furthermore, in studies of cardiac fibrosis, miR-214-3p has been shown to prevent cardiac fibrosis by targeting NOD-like receptor family CARD domain containing 5 (NLRC5). It remains to be further investigated whether the risk of myocardial fibrosis is reduced in OA patients (98). Additionally, chondrocytes produce metabolites that influence the heart and blood vessels. The metabolic profile of chondrocytes in OA differs from that of normal chondrocytes, characterized by markedly increased glycolytic activity, which leads to elevated lactate production (99). This excess lactate enters the circulation, promoting angiogenesis by activating the PI3K/Akt pathway and subsequently upregulating VEGF expression (100).
The cardiovascular-cartilage axis represents a complex yet under-explored domain, with potential implications for both cardiovascular and cartilage health. The interactions within this axis are multifaceted, involving various factors such as metabolism, inflammation, and mechanical forces. Notably, shared influences, including angiogenic factors like VEGF, exhibit dual roles in cardiovascular repair and cartilage degradation. Thus, advancing strategies for tissue-specific modulation of angiogenesis, while balancing the requirements for cardiovascular and cartilage repair, is paramount for future research. Overcoming challenges related to target specificity, model limitations, and clinical translation bottlenecks is essential, with interdisciplinary collaboration and precision medicine technologies playing a crucial role.
2.3 Lung− cartilage axis
There exists a significant correlation between osteoarthritis and lung disease, as evidenced by numerous studies that highlight the interaction between lung and bone. Conditions such as lung cancer and pneumonia produce factors that influence cartilage cells (153, 154). Conversely, articular cartilage also generates factors that impact lung diseases. Therefore, the objective of this section is to elucidate the interactions between lung disease and articular cartilage.
2.3.1 Lung to cartilage
Lung tumor cells can express lncRNA-MIR4435-2HG (50), which is associated with lymph node metastasis and enhances the metastatic capabilities of tumor cells (51). Additionally, this lncRNA promotes the proliferation of chondrocytes while inhibiting their apoptosis (50, 52). The COVID-19 pandemic represents the most significant global public health threat in the past five years (155), with infected individuals exhibiting markedly different primary conditions based on individual variability (156). Research indicates that patients with COVID-19 pneumonitis show elevated expression levels of the CXC chemokine receptor 4 (CXCR4) gene (157). CXCR4, a receptor located on the surface of cartilage (53), is activated by the binding of upstream inflammatory factors, which in turn activates the TGF-β/Smad2/3 signaling pathway and subsequently induces the release of matrix metalloproteinases, leading to cartilage degradation in osteoarthritis. Chondroitin sulfate (CS), an essential component of the ECM of cartilage, is closely linked to the metabolism of chondrocytes (158, 159).
2.3.2 Cartilage to lung
Chondrocyte-derived factors and ECM components have been demonstrated to influence the development of lung diseases. The secreted glycoprotein YKL-40, produced by chondrocytes (101), is significantly elevated in the serum of patients with asthma and chronic obstructive pulmonary disease (COPD), where it serves as a crucial serum predictor for COPD (102). Additionally, YKL-40 is secreted by chondrocytes in osteoarthritic cartilage, which correlates with a higher prevalence of asthma and COPD (103). CS, an essential component of the cartilage extracellular matrix, is closely linked to chondrocyte metabolism (104). Evidence suggests that the higher the level of chondroitin sulfate in the lungs, the more severe the airway obstruction (105, 106). CS has been identified as playing a crucial role in certain severe lung diseases. Eleni et al. conducted an immunohistochemical analysis of lung tissues from patients who succumbed to COVID-19, revealing a significant elevation of chondroitin sulfate, particularly in the diffuse alveolar damage (DAD) region. Subsequent studies indicated that during instances of severe lung injury, the activity and expression of arylsulfatase B (ARSB), an enzyme essential for the degradation of chondroitin sulfate, were diminished (105). CS has been implicated in tissue remodeling across various diseases (160). Moreover, most current pharmacological treatments for OA aim to repair cartilage by promoting the regeneration of its ECM (161). However, these treatments may inadvertently lead to the non-specific deposition of CS in the lung interstitium, potentially increasing the risk of pulmonary fibrosis.
The interaction mechanism of the lung-cartilage axis involves inflammation, metabolism, and various other factors. Effective treatment must navigate the complexities of inter-organ regulation, model limitations, and potential drug side effects. The systemic effects of chronic inflammation and oxidative stress in the lungs are challenging to regulate locally; when targeted therapies are applied, they may inadvertently lead to dual lung-cartilage side effects. Lung hypoxia, as seen in conditions like COPD, results in elevated blood lactate and systemic lactate levels (162), which may activate chondrocyte MMP-13 expression by acidifying the joint microenvironment, potentially promoting cartilage degradation (163). Developing novel anti-inflammatory drugs that can simultaneously control lung inflammation and mitigate metabolite accumulation resulting from lung disease, without adversely impacting cartilage repair, presents a significant challenge.
2.4 Liver−cartilage axis
The liver is a crucial digestive organ in the human body, responsible for the production of various digestive enzymes and serving as a key hub for numerous physiological processes (164). However, the interactions between liver and cartilage metabolism remain underexplored and vary significantly under both physiological and pathological conditions. The following sections will summarize this organ axis.
2.4.1 Liver to cartilage
Insulin-like growth factor-1 (IGF-1) is secreted by hepatocytes and plays a critical role in regulating the metabolism, proliferation, and growth of various cells in the body (70, 71). Studies have demonstrated its ability to influence the progression of OA by altering IGF-1 concentrations (72). IGF-1 inhibits NF-κB signaling by regulating the MAPK and PI3K/Akt signaling pathways in chondrocytes, thereby preventing chondrocyte apoptosis through the inhibition of ROS production, which in turn inhibits the progression of OA (73). Furthermore, non-alcoholic liver disease has been positively correlated with OA (165, 166), warranting further exploration of the mechanisms underlying the interaction between liver disease and cartilage. A long non-coding RNA, known as Long non-coding RNA highly up-regulated in liver cancer (lncRNA-HULC), is highly expressed in hepatocellular carcinoma. It sponges miR-101 by inhibiting the activation of the NF-κB and MAPK signaling pathways, ultimately protecting in vitro chondrocytes from TNF-α-induced inflammatory injury.
2.4.2 Cartilage to liver
The mechanisms by which chondrocytes directly secrete cytokines to influence liver function in vivo remain inadequately explored. However, it is now well established that CS, which is produced from the degradation of the cartilage extracellular matrix in patients with OA, as well as certain drugs that promote the synthesis of the cartilage extracellular matrix, can adversely affect liver health. Notably, glucosamine and CS have been shown to exacerbate hepatocellular damage in individuals with chronic liver diseases (167) and may even obscure the risk associated with pre-existing hepatitis (168). This indicates that the use of chondrotherapeutic agents should be approached with caution in patients with cartilage damage and concurrent hepatitis, as it may hinder the efficacy of cartilage repair.
As a crucial metabolic organ, liver dysfunction can directly or indirectly harm cartilage through abnormalities in metabolites. Although several potential therapeutic targets have been identified, including IGF-1 and IL-6, which play significant roles in the liver-bone axis, the specific functions and efficacies of these targets in various liver diseases and conditions associated with the liver-cartilage axis require further investigation. Additionally, multiple compensatory mechanisms may influence the therapeutic outcomes. Therefore, identifying therapeutic targets that exhibit high specificity, effective efficacy, and minimal side effects remains a pressing challenge, necessitating more fundamental research and clinical trials to accurately screen and identify truly effective targets.
The influence of other organs on cartilage is unidirectional, and no evidence has yet been found to suggest that cartilage interacts reciprocally with these organs. Nonetheless, we summarize this information for reference (Figure 2).
2.5 Intestine− cartilage axis
The intestinal tract serves multiple functions, including the digestion and absorption of food, as well as an endocrine role. It is also the body’s largest site of bacterial colonization. Intestinal peptides produced by the intestinal tract and microbial metabolites can significantly affect cartilage health. Maintaining a dynamic balance of intestinal flora is essential for cartilage health; any disruption of this balance can trigger various pathological processes throughout the body (169–173). Recent studies have indicated a significant impact of intestinal flora on the existence and progression of OA (174, 175). The gut microbiota(GM) and its metabolites can initiate systemic inflammation that impacts the joints through various pathways. For instance, bacterial components, such as lipopolysaccharides, can traverse the compromised intestinal barrier and enter the bloodstream, thereby activating the immune system and prompting the release of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6. These cytokines accumulate locally in the joints, resulting in joint inflammation and cartilage degradation (176).
Glucagon-like peptide (GLP) is a hormone primarily produced by intestinal L-cells (74), classified as an enteroglucagon. In vivo, it activates the glucagon-like peptide-1 receptor (GLP-1R), leading to various downstream effects (75). Activation of GLP-1R has been shown to protect chondrocytes from endoplasmic reticulum stress and apoptosis induced by IL-1β or triglycerides (TGs). Furthermore, GLP inhibits apoptosis and endoplasmic reticulum stress via the PI3K/Akt signaling pathway. The activation of GLP-1R also suppresses the nuclear factor κB pathway in TG-treated chondrocytes, thereby reducing the release of inflammatory mediators such as IL-6 and tumor necrosis factor α, and decreasing matrix catabolism (76, 77). Recent studies have elucidated the mechanism by which the activation of GLP-1R alleviates osteoarthritis (OA), providing evidence that farnesoid X receptor (FXR) serves as a viable therapeutic target for this purpose through the enhancement of GLP-1 levels. Further research has identified ursodeoxycholic acid (UDCA) as a drug that targets FXR activation, which subsequently promotes intestinal production of GLP-1 and activates GLP-1R, thereby facilitating the repair of chondrocytes (78).
The impact of the GM on the systems and tissues of the human body is profound. GM influences cartilage health through multifaceted mechanisms, including metabolites, immune regulation, barrier maintenance, and neuroendocrine interactions. We summarize the effects of several factors—namely, short-chain fatty acids(SCFAs), capsiates(CAT), tryptophan, and extracellular vesicles (EVs)—on cartilage production. The gut microbiota produces SCFAs, which have been demonstrated to modulate inflammation and may confer protective effects on chondrocytes. Notably, levels of total fatty acids, saturated fatty acids, and linoleic acid entering the gut have been significantly associated with a reduced risk of osteoarthritis. Following their breakdown and absorption, short-chain fatty acids, among others, have been found to exert significant anti-osteoarthritis effects (177, 178). SCFAs can act directly on chondrocytes after entering the bloodstream or indirectly protect against chondrocyte apoptosis and matrix degradation by inhibiting osteoclast activity and modulating the inflammatory response, thereby enhancing cartilage repair (79, 80). CAT, another metabolite produced by intestinal flora, inhibit HIF-1α expression and reduce iron-death-dependent chondrocyte damage through the activation of Solute Carrier Family 2 Member 1 (SLC2A1) (81). Intestinal tryptophan metabolites, such as indole-3-acetic acid (IAA) and indole-3-pyruvate (IPA), function as aryl hydrocarbon receptor (AhR) ligands, activating the AhR and subsequently initiating the Wnt/β-catenin signaling pathway, which significantly promotes chondrocyte proliferation (82).
GM serves as a double-edged sword; when in a balanced state, it protects joint health. Conversely, when the gut flora becomes imbalanced, various harmful substances or factors can enter the bloodstream, ultimately affecting the joints and promoting the formation of OA. While some studies have indicated that probiotics and prebiotics may confer benefits for arthritis, there remains a lack of uniform standards and consensus regarding the most effective probiotic strains, their combinations, dosages, and duration of use. Fecal transplants represent an emerging strategy for modulating gut microbiota; however, their application in the treatment of arthritis is still in the exploratory phase.
2.6 Kidney-cartilage axis
The kidney is the primary organ responsible for regulating calcium and phosphorus metabolism in the body, thereby influencing calcium and phosphorus homeostasis, which in turn affects bone mineral deposition and the quality of bone. Although there are fewer studies examining the effects of the kidney and chronic kidney disease (CKD) on cartilage, fibroblast growth factor 23 (FGF23) has emerged as a significant factor linking bone metabolism with renal calcium and phosphorus metabolism. FGF23, a hormone predominantly secreted by osteoblasts (107), regulates blood phosphate levels by promoting renal phosphate excretion and inhibiting the production of 1,25-dihydroxyvitamin D3 [1,25(OH)2D] (179, 180). It achieves this by suppressing the expression of renal phosphate transporter proteins in the proximal renal tubules, thus maintaining the balance between bone synthesis and catabolism. Recent studies have increasingly illuminated the relationship between FGF23 and arthritis, with evidence suggesting that FGF23 enhances the expression of ColX, MMP13, and MMP9 in chondrocytes through the modulation of the Wnt/β-catenin signaling pathway, ultimately contributing to OA (181, 182). Furthermore, it has been demonstrated that FGF23 plays a role in preserving the chondrocyte phenotype in OA (182). Here, we propose the hypothesis that the kidney influences FGF23 expression by modulating calcium and phosphorus metabolism; however, the validity of this hypothesis and the underlying mechanisms merit further investigation.
2.7 Gonad-cartilage axis
Sex hormones, particularly estrogen, play a crucial role in the development and progression of OA (183, 184). As research on OA continues to advance, the gender differences in prevalence and symptom burden are increasingly concerning, particularly given that postmenopausal women exhibit a higher prevalence of OA. Subsequent studies have demonstrated that estrogen has a protective effect on chondrocytes (185). Animal studies indicate that estrogen supplementation significantly reduces the expression of matrix metalloproteinase 13 (MMP13) and decreases chondrocyte apoptosis in female mice (186). In vitro studies have demonstrated that estrogen activates AMP-activated protein kinase (AMPK) and inhibits the phosphorylation of mammalian target of rapamycin (mTOR) by enhancing the expression of SIRT1 in chondrocytes. This process subsequently activates AMPK and suppresses mTOR phosphorylation, leading to a signaling cascade that promotes mitochondrial autophagy in chondrocytes. Consequently, this cascade enhances chondrocyte proliferation and contributes to the improvement of OA (187). In vivo, Estrogen exerts its effects by binding to estrogen receptors (ER) present in the tissues surrounding the joints, specifically binding to estrogen receptor beta (ERβ), which negatively regulates arthritis progression by inhibiting the activation of the NF-κB pathway in synoviocytes (188). Recent studies have revealed a mutual antagonism between estrogen and IL-1β, with IL-1β down-regulating the quantity and functionality of ERα in chondrocytes. The activation of ERα by estradiol (E2) or selective estrogen receptor modulators (SERMs) induces anti-inflammatory effects in these cells, ultimately inhibiting nitric oxide (NO) production and the expression of inducible nitric oxide synthase (iNOS), thereby mitigating chondrocyte damage (83). Previously, it was believed that estrogen could influence chondrocyte metabolism solely through ER (84, 85). However, recent findings have demonstrated that estrogen can also exert chondroprotective effects by activating the G protein-coupled estrogen receptor (GPER). On one hand, by inhibiting the mechanical stress-mediated activation of Piezo1 channels, estrogen reduces actin polymerization, thus inhibiting chondrocyte apoptosis, which suggests a protective role for estrogen in post-traumatic arthritis (84, 85). On the other hand, the activation of GPER decreases intracellular levels of ROS and lipid peroxides, which in turn alleviates intracellular iron overload in chondrocytes, thereby providing additional protection to these cells (86).
2.8 Pancreas−cartilage axis
The pancreas serves as a crucial digestive organ and a regulator of blood glucose levels in the body. Insulin, one of the most significant hormones for chondrocytes, plays a pivotal role in this context. Previous research has established a link between insulin and OA, highlighting that conditions such as diabetes and metabolic syndrome alter the secretion of insulin and glucagon. These alterations can lead to changes in body weight, where weight gain exacerbates weight-bearing stress on the hip and knee joints, potentially resulting in cartilage damage and accelerating the development of OA (189–193). Recent studies have revealed that insulin influences the metabolism of chondrocytes and synovial cells in the joints through various mechanisms, thereby impacting joint health. In recent years, it has been discovered that insulin influences the metabolism of chondrocytes and synovial cells in the joints in various ways, thereby impacting joint health. Researchers have identified insulin-associated receptors on chondrocytes and synovial cells (87–89). An increasing number of studies have demonstrated that the effects of insulin on chondrocytes are complex and biphasic (90). Some studies indicate that moderate levels of insulin exhibit anti-apoptotic effects on chondrocytes and promote their proliferation in animal models (88, 91). However, high concentrations of insulin may down-regulate cartilage proliferation and repair by inhibiting chondrocyte autophagy (92). Additionally, insulin can exacerbate the progression of OA by influencing fibroblast-like synoviocytes (FLS), leading to increased secretion of pro-inflammatory cytokines (93). Interestingly, insulin appears to selectively protect hand joints by inhibiting the release of catabolic enzymes from FLS into the synovial fluid; however, it remains to be determined whether this effect extends to systemic joints (94) (Figure 3).

Figure 3. Overview of Intestinal, renal, gonadal, pancreatic, adipose, muscular, and skeletal effects on cartilage.
2.9 Adipose tissue-cartilage axis
Adiposity is closely linked to the progression of OA. A high body fat percentage in obese patients constitutes a significant risk factor for the development of OA, as the increased pressure on the knee joint exacerbates the wear and tear on articular cartilage. Furthermore, adipose tissue not only influences chondrocyte metabolism but has also been implicated in the progression of OA through the action of adipokines (194). Adipose tissue regulates chondrocyte metabolism via the secretion of free fatty acids (FFA) (195, 196), lipocalin (197–199), and leptin (200). Notably, lipocalin has been shown to protect chondrocytes from hydrogen peroxide (H2O2)-induced apoptosis by activating autophagy through the AMPK/mTOR signaling pathway (111). Additionally, infrapatellar fat pad (IPFP) mesenchymal stromal cell-derived exosomes (MSCIPFP-Exos) transport miR-100-5p into chondrocytes, thereby inhibiting apoptosis by suppressing the mTOR signaling pathway (117). FFA has been found to cause chondrocyte damage by upregulating the expression of nuclear protein 1 (Nupr1) on the surface of chondrocytes (112). In an in vitro assay, an investigator discovered that leptin promotes chondrocyte proliferation and gene expression in a dose-dependent manner, with maximal effects observed at a leptin concentration of 100 ng/mL. Leptin is detectable in the synovial fluid of patients with OA (113); however, contrary to in vitro findings, its levels in synovial fluid are positively correlated with the severity of OA (114). Additionally, leptin has been shown to promote the catabolism of articular cartilage and functions as a pro-inflammatory factor within this tissue (115). It mediates the production of NLRP3 inflammatory vesicles in chondrocytes through reactive ROS production, ultimately leading to the upregulation of various inflammatory factors (e.g., IL-1β, IL-6, IL-8) and resulting in cartilage degradation and joint inflammation (116).
2.10 Muscle−cartilage axis
The relationship between muscle strength and OA has garnered considerable attention in recent years (201). Exercise therapy has long been recognized as an effective treatment for musculoskeletal disorders. It is suggested that aerobic exercise reduces the accumulation of lipid peroxides in the body (202). Furthermore, numerous studies have demonstrated that stronger lower extremity muscles protect the knee joint, thereby inhibiting the progression of OA (203). However, for patients who have already developed OA, pain often limits their movement and exercise, leading to further muscle weakening and exacerbating the progression of the disease (204–207). An increasing body of research indicates that the functional state of muscle can significantly influence skeletal metabolism (208). Folliculin interacting protein 1 (FNIP1) has been identified as a crucial regulator of skeletal muscle activity and bone metabolism within muscle tissue (118, 119). Additionally, skeletal muscle mesenchymal progenitors (MPs) play a vital role in the regenerative processes of skeletal muscle (209). It has been observed that muscle MP cells can exuberantly produce ECM under specific conditions (210), representing a promising therapeutic avenue for cartilage damage repair.
2.11 Bone−cartilage axis
The structure adjacent to the articular cartilage is the subchondral bone. Healthy cartilage is spatially interconnected with its subchondral bone, and studies have demonstrated a biosignaling relationship between the two (211). Subchondral bone primarily consists of cancellous bone and adjacent bone marrow, where osteoblasts and osteoclasts exist in a dynamic equilibrium. When this equilibrium is disrupted, it can induce apoptosis of chondrocytes and accelerate the progression of OA (212). Osteoblasts in physiological subchondral bone enhance glycolytic metabolism in chondrocytes via the MAPK/HIF-1 pathway (213). However, transforming growth factor β (TGF-β) secreted in subchondral bone serves as a crucial regulator (120); abnormal elevation of TGF-β accelerates cartilage degeneration and subchondral bone resorption (121). Similarly, osteoclasts, under physiological conditions, exert a mutual protective effect with chondrocytes (214). Yet, when osteoclasts interact with abnormal cartilage, they degrade the cartilage matrix by secreting MMP8 and MMP9, further exacerbating cartilage degeneration in osteoarthritis and other diseases (215). It is evident that the health of the subchondral bone directly influences the health of the cartilage. The relationship between the two can be likened to that of soil and crops; the health of the former is conducive to the healthy proliferation of cartilage in terms of both spatial and mechanistic support. Furthermore, the dynamics of osteoblasts and osteoclasts in the bone marrow represent a promising area for future research.
3 Strategies for restoring cartilage by intervening in extraosseous organs
Research on cartilage repair by modulating the functional status of extraosseous organs is gaining traction. An analysis of the aforementioned studies indicates that chondrocytes are significantly involved in crosstalk with the brain, heart, lungs, and other systems. Interventions aimed at promoting chondrocyte growth, inhibiting apoptosis, and enhancing the production of extracellular matrix in cartilage through the modulation of organ-specific or highly expressed factors may be pivotal in preventing OA.
Transorganic cytokines exhibiting chondroprotective effects and the mechanisms by which they facilitate chondrocyte proliferation have been elucidated. Sema3A, secreted by sensory neurons, serves to protect chondrocytes and maintains chondrocyte homeostasis through the activation of the PI3K pathway (69). Nerve growth factor (NGF) is a significant mediator of pain in OA, and its antibodies have been extensively validated as a therapeutic approach for pain relief (216). Recent research has achieved a breakthrough in the regulation of biological circadian rhythms; bone morphogenetic protein 2 (BMP2) and runt-related transcription factor 2 (RUNX2) are key initiators of osteocalcin transcription, and their expression levels fluctuate with photoperiod and melatonin, thereby promoting chondrocyte repair (217). MT protects cells from pathological death by scavenging reactive oxygen species in tissues and promoting the expression of glutathione and antioxidant enzymes (92). Intracellularly, MT inhibits the release of TNFα and interleukin-1 beta (IL-1β) in the joints (218), while simultaneously promoting the production of the ECM by activating the TGF-β signaling pathway (219). The mechanism of action of irisin involves enhancing mitochondrial function in chondrocytes. Research has demonstrated that appropriate exercise stimulates the synthesis and release of irisin in vivo. The specific repair mechanism of irisin on chondrocytes is linked to the reduction of the expression of the chondrocyte senescence marker P21 through the inhibition of the PI3K/Akt signaling pathway, thereby alleviating chondrocyte degeneration (41, 220). lncRNAs and microRNAs, which are typically produced by non-chondrocytes and often associated with tumor cells, have been shown to increase tumor aggressiveness. However, extracting these RNAs and transferring them to chondrocytes promotes chondrocyte proliferation. Collectively, these cytokines exhibit chondrocyte-protective effects. Future research will focus on exploring their modes of delivery, synthesizing stable compounds, and constructing suitable drug delivery systems, which is of significant interest.
Cellular receptors in their non-free state, along with the downstream effects triggered by their activation, can rejuvenate chondrocytes without isolating them from the body. The activation of α-1-adrenergic receptors is known to promote chondrocyte apoptosis, while α-adrenergic receptor (α-AR) antagonists, particularly phentolamine, have been shown to stimulate chondrogenesis and induce hypertrophy (221). Prostaglandin E2 (PGE2), which regulates vasodilation and permeability, and Signal Transducer and Activator of Transcription 3 (STAT3), secreted by vascular endothelial cells, both promote H-type angiogenesis in the subchondral bone, exacerbating OA (222, 223). Recent studies indicate that the mechanosensitive receptor Piezo1 plays a pivotal role in both angiogenesis and cartilage homeostasis (223). Specifically, Piezo1 promotes the redistribution of blood flow and increases microvessel density in muscle, while sensing changes in mechanical stress on the surface of chondrocytes, which in turn activates downstream signaling pathways altered by Ca2+ influx, thereby promoting chondrocyte apoptosis (224, 225). The activation of the GPER has been shown to inhibit the mechanical stress-mediated RhoA/LIMK/cofilin pathway, ultimately reducing the expression of the Piezo1 protein and alleviating apoptosis in chondrocytes (84). β-Hydroxybutyrate (β-HB), a readily available energy carrier produced by adipocytes, enhances chondrocyte autophagy and diminishes chondrocyte apoptosis by activating the Erb-B2 receptor tyrosine kinase 3 (ERBB3) signaling pathway. This action ameliorates OA without leading to hepatic pathology. Furthermore, liraglutide has been demonstrated to activate downstream pathways upon interaction with the GLP-1R, subsequently inducing chondrogenesis and reducing chondrocyte apoptosis. GLP-1 agonists also contribute to the improvement of sarcopenia and the increase of bone density in the elderly, indirectly enhancing and preventing the progression of OA (226). It is important to note that these receptors are not exclusively expressed by chondrocytes; different receptor proteins exhibit distinct activation effects in various cell types. Consequently, the investigation of drugs targeting these chondrocyte receptors remains a significant focus within pharmacological research.
Optimizing the structure of intestinal flora and enhancing the autonomous absorption of nutrients conducive to cartilage repair through improvements in dietary composition is a treatment approach that requires time to manifest its effects. Supplementing the daily diet with vitamin B1, which is absorbed into the bloodstream via the intestinal tract, can significantly alleviate the symptoms of OA by modulating macrophage polarization and thereby inhibiting chondrocyte apoptosis. Furthermore, promoting the production of SCFAs in the intestinal tract is crucial; dietary fiber serves as the primary substrate for the fermentative production of SCFAs by intestinal flora. A high-fiber diet, defined as an intake of more than 30 grams of fiber per day (227), can substantially increase the concentration of SCFAs in feces. Additionally, the intake of probiotics (e.g., Lactobacillus, Bifidobacterium) can synergistically enhance these fermentation effects, resulting in increased SCFA production and inhibiting the proliferation of harmful bacteria (228, 229). This interplay can effectively promote cartilage repair through the action of SCFAs in the bloodstream.
Bone-cartilage grafting represents a promising approach for the repair of arthropathic lesions, integrating both mechanical and molecular biological strategies. This technique utilizes healthy cartilage and subchondral bone tissues, wherein healthy cartilage-bone plugs are transplanted into the injured area. These transplanted bone plugs not only provide initial mechanical support and reduce joint stress concentration but also supply active chondrocytes and bone matrix, thereby promoting chondrocyte migration and matrix deposition in the affected region. In OA, the balance between osteoblasts and osteoclasts in cartilage and subchondral bone is disrupted; however, the transplantation of bone-cartilage grafts from healthy areas can enhance the subchondral microenvironment and facilitate the formation of peripheral cartilage. Future research is focused on improving graft survival rates through gene editing techniques, such as CRISPR technology, which can knock down IL-1 receptor expression in chondrocytes to enhance their anti-inflammatory capabilities. Moreover, the potential discovery of new loci associated with pathological chondrocyte death underscores the importance of gene editing in this field.
Exosomes derived from various sources present a universal therapeutic solution. As previously discussed, numerous organs and tissues can ameliorate OA by enhancing chondrocyte viability and promoting their proliferation through characteristic mesenchymal stem cell (MSC)-derived exosomes. Additionally, exosomes exhibit extremely low immunogenicity, which renders allogeneic interspecies therapy feasible and underscores their significant therapeutic potential. Future advancements in the efficient purification of exosomes from diverse sources, as well as in the engineering and targeting of these exosomes, promise to enhance their therapeutic efficacy.
Future studies should investigate the mechanisms underlying the effects of various organs on cartilage. Numerous pathways remain to be explored to enhance these therapeutic approaches and to develop precise therapeutic strategies aimed at optimizing clinical efficacy. Such advancements will make the prevention of OA a more attainable goal in the future.
The inter-crosstalk between multiple organs constitutes a more complex system that is critical to the health impact of the joints. The brain-gut-bone-joint axis has been extensively studied, with numerous studies confirming its close relationship with neurodegenerative diseases. Dysbiosis of the intestinal microbiota can lead to abnormalities in metabolites, such as SCFAs and lipopolysaccharides (LPS), which may affect the synthesis and release of neurotransmitters, resulting in neuroinflammation and neurodegeneration (230). Furthermore, dysbiosis or the accumulation of undesirable metabolites can contribute to the degeneration of articular cartilage. The brain-gut-bone-joint axis encompasses complex interactions among multiple systems, with significant variations in genes, composition of intestinal microbiota, lifestyles, dietary habits, and other factors among individuals. These variations complicate the exploration of this axis, although the potential for future research in this area is promising. Additionally, multifactorial interactions with other organs warrant further investigation.
4 Conclusions and future perspectives
Contrary to traditional perceptions of OA, recent studies characterize OA more as a metabolic syndrome disease (231). In this context, various extraosseous organs throughout the body influence chondrocyte value addition and apoptosis through multiple cytokines, EVs, hormones, and metabolites. Therefore, assessing the functional status of different organ systems is crucial when focusing on OA.
Numerous potential therapeutic approaches and the potential interactions between various systemic diseases have emerged from the perspective of the organ-cartilage axis. The administration of chondroitin sulfate should be individualized for OA patients with concurrent pulmonary fibrotic disease, as sensory neurons not only play a role in the generation of nociceptive sensations but also enhance the viability of chondrocytes.
Exosomes derived from various organ sources represent promising therapeutic tools, yet our understanding of them remains limited. Most exosomes enter chondrocytes by delivering miRNAs and proteins that promote cellular proliferation. In future research, we will selectively utilize exosomes from specific sources based on the distinct damage mechanisms affecting chondrocytes. However, it is important to note that not all exosomes facilitate chondrocyte proliferation or inhibit apoptosis; for instance, EC-Exos can induce chondrocyte apoptosis. Furthermore, exosomes derived from probiotic sources exhibit significant therapeutic potential for human diseases, presenting a promising avenue for investigating the influence of gut flora on human health.
OA involves lesions beyond mere cartilage damage, including synovial inflammation, bony changes in the subchondral bone, ligamentous damage, and degeneration of the surrounding muscles. There remains a significant research gap regarding the connection between pathological changes in these tissues and the organ axis. Comprehensive exploration of the mechanisms underlying these complex interactions is an essential task in biomedical research.
This review proposes that OA should be perceived as a systemic disease, where its progression reflects the functional state of all organs in the body. Future treatment strategies and research on OA may extend beyond bone metabolism, cartilage repair, and synovial regeneration. Instead, multidisciplinary and cross-disciplinary exploration, along with mechanistic research, is likely to become a mainstream approach.
Author contributions
YQ: Writing – original draft. JD: Writing – review & editing. ZG: Conceptualization, Writing – review & editing. SC: Writing – review & editing. CX: Methodology, Writing – review & editing, Supervision.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This research is fully sponsored by the National Natural Science Foundation of China (81972075), Central-led Local Science and Technology Development Funds (YDZJSX20231A062), Shanxi Provincial Scientific and Technological Achievement Transformation Guidance Special Programme (202204021301067).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that Generative AI was used in the creation of this manuscript. During the preparation of this work the used ChatGPT in order to improve authors’ language. After using this tool, the authors reviewed and edited the content as needed and take full responsibility for the content of the publication.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
Glossary
1,25(OH)2D: 1,25-dihydroxyvitamin D3
AD: Alzheimer’s disease
ADRD: AD-related dementia
AhR: Aryl hydrocarbon receptor
AR: Androgen receptor
ARC KISS1: KISS1 neurons in the arcuate nucleus
Aβ: Amyloid beta
BBB: Blood-brain barrier
BMD: Bone mineral density
BMP2: Bone morphogenetic protein 2
CAT: Capsiates
CCN3: Cellular communication network factor 3
CGRP: Calcitonin gene-related peptide
CKD: Chronic kidney disease
COPD: Chronic obstructive pulmonary disease
CS: Chondroitin sulfate
CVD: Cardiovascular disease
CXCR4: CXC chemokine receptor 4
E2: Estradiol
EC-Exos: Exosomes derived from vascular endothelial cells
ECM: Extracellular matrix
ER: Estrogen receptor
ERBB3: Erb-B2 receptor tyrosine kinase 3
ERβ: Estrogen receptor beta
EVs: Extracellular vesicles
FFA: Free fatty acids
FGF23: Fibroblast growth factor 23
FLS: Fibroblast-like synoviocytes
FNDC5: Fibronectin type III domain-containing protein 5
FNIP1: Folliculin interacting protein 1
GLP: Glucagon-like peptide
GLP-1R: Glucagon-like peptide-1 receptor
GM: Gut microbiota
GPER: G protein-coupled estrogen receptor
H2O2: Hydrogen peroxide
IAA: Indole-3-acetic acid
IGF-1: Insulin-like growth factor-1
IL-1β: Interleukin-1 beta
iNOS: Inducible nitric oxide synthase
IPA: Indole-3-pyruvate
IPFP: Infrapatellar fat pad
KOA: Knee osteoarthritis
LDL: Low-density lipoprotein
LPS: Lipopolysaccharides
lncRNA-HULC: Long non-coding RNA highly up-regulated in liver cancer
LRP3: Low-density lipoprotein receptor-related protein 3
MCP-1: Monocyte chemoattractant protein 1
MMP13: Matrix metalloproteinase 13
MPs: Mesenchymal progenitors
MSC: Mesenchymal stem cell
MSCIPFP-Exos: Infrapatellar fat pad mesenchymal stromal cell-derived exosomes
MT: Melatonin
NFTs: Neurofibrillary tangles
NGF: Nerve Growth Factor
NLRC5: NOD-like receptor family CARD domain containing 5
NO: Nitric oxide
Nupr1: Nuclear protein 1
OA: Osteoarthritis
oxLDL: Oxidized LDL
PGE2: Prostaglandin E2
PTH: Parathyroid hormone
RORα: Retinoic acid-related orphan receptor α
ROS: Reactive oxygen species
RUNX2: Runt-related transcription factor 2
SCFAs: Short-chain fatty acids
Sema3A: Semaphorin 3A
SERMs: Selective estrogen receptor modulators
SLC2A1: Solute Carrier Family 2 Member 1
SP: Substance P
STAT3: Signal Transducer and Activator of Transcription 3
TFEB: Transcription factor EB
TGF-β: Transforming growth factor β
TGs: Triglycerides
TNF-α: Tumor necrosis factor-alpha
VEGF: Vascular endothelial growth factor
α-AR: α-Adrenergic receptor
β-HB: β-Hydroxybutyrate.
References
1. Global incidence, prevalence, years lived with disability (YLDs), disability-adjusted life-years (DALYs), and healthy life expectancy (HALE) for 371 diseases and injuries in 204 countries and territories and 811 subnational locations, 1990-2021: a systematic analysis for the Global Burden of Disease Study 2021. Lancet (London England). (2024) 403:2133–61. doi: 10.1016/S0140-6736(24)00757-8
2. Park JH, Hong JY, Han K, Suh SW, Park SY, Yang JH, et al. Prevalence of symptomatic hip, knee, and spine osteoarthritis nationwide health survey analysis of an elderly Korean population. Med (Baltimore). (2017) 96:e6372. doi: 10.1097/MD.0000000000006372
3. Dasa V, Mihalko W, Rivadeneyra A, Urban J, Wickline A, Rogenmoser D, et al. Innovations in Genicular Outcomes Registry (IGOR): protocol for a real-world registry study of treatments for knee osteoarthritis. Ther Adv musculoskel Dis. (2024) 16:1759720x241304193. doi: 10.1177/1759720X241304193
4. Courties A, Kouki I, Soliman N, Mathieu S, and Sellam J. Osteoarthritis year in review 2024: Epidemiology and therapy. Osteoarthr cartil. (2024) 32:1397–404. doi: 10.1016/j.joca.2024.07.014
5. Ren J-L, Yang J, and Hu W. The global burden of osteoarthritis knee: a secondary data analysis of a population-based study. Clin Rheumatol. (2025) 44(4):1769–810. doi: 10.1007/s10067-025-07347-6
6. Chen D, Shen J, Zhao W, Wang T, Han L, Hamilton JL, et al. Osteoarthritis: toward a comprehensive understanding of pathological mechanism. Bone Res. (2017) 5:16044. doi: 10.1038/boneres.2016.44
7. Oo WM. Prospects of disease-modifying osteoarthritis drugs. Clinics geriatric Med. (2022) 38:397–432. doi: 10.1016/j.cger.2021.11.010
8. Fujii Y, Liu L, Yagasaki L, Inotsume M, Chiba T, and Asahara H. Cartilage homeostasis and osteoarthritis. Int J Mol Sci. (2022) 23:6316. doi: 10.3390/ijms23116316
9. Busija L, Bridgett L, Williams SR, Osborne RH, Buchbinder R, March L, et al. Osteoarthritis. Best Pract Res Clin Rheumatol. (2010) 24:757–68. doi: 10.1016/j.berh.2010.11.001
10. Deng Y, Hou M, Wu Y, Liu Y, Xia X, Yu C, et al. SIRT3-PINK1-PKM2 axis prevents osteoarthritis via mitochondrial renewal and metabolic switch. Bone Res. (2025) 13:36. doi: 10.1038/s41413-025-00413-4
11. Lavalle S, Scapaticci R, Masiello E, Salerno VM, Cuocolo R, Cannella R, et al. Beyond the surface: nutritional interventions integrated with diagnostic imaging tools to target and preserve cartilage integrity: A narrative review. Biomedicines. (2025) 13:570. doi: 10.3390/biomedicines13030570
12. Wang M, Tan G, Jiang H, Liu A, Wu R, Li J, et al. Molecular crosstalk between articular cartilage, meniscus, synovium, and subchondral bone in osteoarthritis. Bone Joint Res. (2022) 11:862–72. doi: 10.1302/2046-3758.1112.BJR-2022-0215.R1
13. Largo R, Mediero A, Villa-Gomez C, Bermejo-Alvarez I, and Herrero-Beaumont G. Aberrant anabolism hinders constructive metabolism of chondrocytes by pharmacotherapy in osteoarthritis. Bone Joint Res. (2025) 14:199–207. doi: 10.1302/2046-3758.143.BJR-2024-0241.R1
14. Ren P, Niu H, Cen H, Jia S, Gong H, and Fan Y. Biochemical and morphological abnormalities of subchondral bone and their association with cartilage degeneration in spontaneous osteoarthritis. Calcif Tissue Int. (2021) 109:179–89. doi: 10.1007/s00223-021-00834-3
15. Donell S. Subchondral bone remodelling in osteoarthritis. EFORT Open Rev. (2019) 4:221–9. doi: 10.1302/2058-5241.4.180102
16. Boyde A. The bone cartilage interface and osteoarthritis. Calcif Tissue Int. (2021) 109:303–28. doi: 10.1007/s00223-021-00866-9
17. Wan QQ, Qin WP, Ma YX, Shen MJ, Li J, Zhang ZB, et al. Crosstalk between bone and nerves within bone. Adv Sci (Weinh). (2021) 8:2003390. doi: 10.1002/advs.202003390
18. Deng A-F, Wang F-X, Wang S-C, Zhang Y-Z, Bai L, and Su J-C. Bone-organ axes: bidirectional crosstalk. Military Med Res. (2024) 11:37. doi: 10.1186/s40779-024-00540-9
19. Zhao J, Lei H, Wang T, and Xiong X. Liver-bone crosstalk in non-alcoholic fatty liver disease: Clinical implications and underlying pathophysiology. Front Endocrinol (Lausanne). (2023) 14:1161402. doi: 10.3389/fendo.2023.1161402
20. Li Z, Wen X, Li N, Zhong C, Chen L, Zhang F, et al. The roles of hepatokine and osteokine in liver-bone crosstalk: Advance in basic and clinical aspects. Front Endocrinol (Lausanne). (2023) 14:1149233. doi: 10.3389/fendo.2023.1149233
21. Collino M, Rudich A, and Konrad D. Mediators of interorgan crosstalk in metabolic inflammation. Mediators Inflammation. (2013) 2013:609096. doi: 10.1155/2013/609096
22. Piletic K, Alsaleh G, and Simon AK. Autophagy orchestrates the crosstalk between cells and organs. EMBO Rep. (2023) 24:e57289. doi: 10.15252/embr.202357289
23. Rissel R, Schaefer M, Kamuf J, Ruemmler R, Riedel J, Mohnke K, et al. Lung-brain ‘cross-talk’: systemic propagation of cytokines in the ARDS via the bloodstream using a blood transfusion model does not influence cerebral inflammatory response in pigs. PeerJ. (2022) 10:e13024. doi: 10.7717/peerj.13024
24. Armutcu F. Organ crosstalk: the potent roles of inflammation and fibrotic changes in the course of organ interactions. Inflammation Res. (2019) 68:825–39. doi: 10.1007/s00011-019-01271-7
25. Besedovsky H, del Rey A, Sorkin E, Da Prada M, Burri R, and Honegger C. The immune response evokes changes in brain noradrenergic neurons. Science. (1983) 221:564–6. doi: 10.1126/science.6867729
26. Stead RH, Tomioka M, Quinonez G, Simon GT, Felten SY, and Bienenstock J. Intestinal mucosal mast cells in normal and nematode-infected rat intestines are in intimate contact with peptidergic nerves. Proc Natl Acad Sci U S A. (1987) 84:2975–9. doi: 10.1073/pnas.84.9.2975
27. Veiga-Fernandes H and Pachnis V. Neuroimmune regulation during intestinal development and homeostasis. Nat Immunol. (2017) 18:116–22. doi: 10.1038/ni.3634
28. Audrit KJ, Delventhal L, Aydin Ö, and Nassenstein C. The nervous system of airways and its remodeling in inflammatory lung diseases. Cell Tissue Res. (2017) 367:571–90. doi: 10.1007/s00441-016-2559-7
29. Li X, Hassoun HT, Santora R, and Rabb H. Organ crosstalk: the role of the kidney. Curr Opin Crit Care. (2009) 15:481–7. doi: 10.1097/MCC.0b013e328332f69e
30. Quílez ME, López-Aguilar J, and Blanch L. Organ crosstalk during acute lung injury, acute respiratory distress syndrome, and mechanical ventilation. Curr Opin Crit Care. (2012) 18:23–8. doi: 10.1097/MCC.0b013e32834ef3ea
31. Yang Y, Pei T, Liu C, Cao M, Hu X, Yuan J, et al. Glutamine metabolic competition drives immunosuppressive reprogramming of intratumour GPR109A+ myeloid cells to promote liver cancer progression. Gut. (2025) 74:255. doi: 10.1136/gutjnl-2024-332429
32. van den Bosch MHJ, Blom AB, and van der Kraan PM. Inflammation in osteoarthritis: Our view on its presence and involvement in disease development over the years. Osteoarthr cartil. (2024) 32:355–64. doi: 10.1016/j.joca.2023.12.005
33. Zhang F and Zhang W. Research progress in Alzheimer’s disease and bone-brain axis. Ageing Res Rev. (2024) 98:102341. doi: 10.1016/j.arr.2024.102341
34. Kelly RR, Sidles SJ, and LaRue AC. Effects of neurological disorders on bone health. Front Psychol. (2020) 11:612366. doi: 10.3389/fpsyg.2020.612366
35. Xiao Y, Han C, Wang Y, Zhang X, Bao R, Li Y, et al. Interoceptive regulation of skeletal tissue homeostasis and repair. Bone Res. (2023) 11:48. doi: 10.1038/s41413-023-00285-6
36. Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, et al. Leptin inhibits bone formation through a hypothalamic relay a central control of bone mass. Cell. (2000) 100(2):197–207. doi: 10.1016/s0092-8674(00)81558-5
37. Xia W, Xie J, Cai Z, Liu X, Wen J, Cui Z-K, et al. Damaged brain accelerates bone healing by releasing small extracellular vesicles that target osteoprogenitors. Nat Commun. (2021) 12:6043. doi: 10.1038/s41467-021-26302-y
38. Maak S, Norheim F, Drevon CA, and Erickson HP. Progress and challenges in the biology of FNDC5 and irisin. Endocr Rev. (2021) 42:436–56. doi: 10.1210/endrev/bnab003
39. Bi JA-O, Yang L, Wang T, Zhang JA-O, Li T, Ren YA-O, et al. Irisin improves autophagy of aged hepatocytes via increasing telomerase activity in liver injury. Oxid Med Cell Longev. (2020) 2020:6946037. doi: 10.1155/2020/6946037
40. Vadalà G, Di Giacomo G, Ambrosio L, Cannata F, Cicione C, Papalia R, et al. Irisin recovers osteoarthritic chondrocytes in vitro. Cells. (2020) 9:1478. doi: 10.3390/cells9061478
41. Wang FS, Kuo CW, Ko JY, Chen YS, Wang SY, Ke HJ, et al. Irisin mitigates oxidative stress, chondrocyte dysfunction and osteoarthritis development through regulating mitochondrial integrity and autophagy. Antioxid (Basel Switzerland). (2020) 9:810. doi: 10.3390/antiox9090810
42. de Munter W, van der Kraan PM, van den Berg WB, and van Lent PL. High systemic levels of low-density lipoprotein cholesterol: fuel to the flames in inflammatory osteoarthritis? Rheumatol (Oxford England). (2016) 55:16–24. doi: 10.1093/rheumatology/kev270
43. Kishimoto H, Akagi M, Zushi S, Teramura T, Onodera Y, Sawamura T, et al. Induction of hypertrophic chondrocyte-like phenotypes by oxidized LDL in cultured bovine articular chondrocytes through increase in oxidative stress. Osteoarthr cartil. (2010) 18:1284–90. doi: 10.1016/j.joca.2010.05.021
44. Villalvilla A, Gómez R, Largo R, and Herrero-Beaumont G. Lipid transport and metabolism in healthy and osteoarthritic cartilage. Int J Mol Sci. (2013) 14:20793–808. doi: 10.3390/ijms141020793
45. Gkretsi V, Simopoulou T, and Tsezou A. Lipid metabolism and osteoarthritis: lessons from atherosclerosis. Prog Lipid Res. (2011) 50:133–40. doi: 10.1016/j.plipres.2010.11.001
46. Akagi M, Ueda A, Teramura S, Kanata T, Sawamura C, and Hamanishi C. Oxidized LDL binding to LOX-1 enhances MCP-1 expression in cultured human articular chondrocytes. Osteoarthritis Cartilage. (2009) 17(2):271–5. doi: 10.1016/j.joca.2008.06.019
47. Cao C, Shi Y, Zhang X, Li Q, Zhang J, Zhao F, et al. Cholesterol-induced LRP3 downregulation promotes cartilage degeneration in osteoarthritis by targeting Syndecan-4. Nat Commun. (2022) 13:7139. doi: 10.1038/s41467-022-34830-4
48. Sanchez C, Lambert C, Dubuc JE, Bertrand J, Pap T, and Henrotin Y. Syndecan-4 is increased in osteoarthritic knee, but not hip or shoulder, articular hypertrophic chondrocytes. Cartilage. (2021) 13:862s–71s. doi: 10.1177/1947603519870855
49. Zhang X, Crawford R, and Xiao Y. Inhibition of vascular endothelial growth factor with shRNA in chondrocytes ameliorates osteoarthritis. J Mol Med. (2016) 94:787–98. doi: 10.1007/s00109-016-1425-0
50. Zhang M, Yu X, Zhang Q, Sun Z, He Y, and Guo W. MIR4435-2HG: A newly proposed lncRNA in human cancer. Biomed Pharmacother. (2022) 150:112971. doi: 10.1016/j.biopha.2022.112971
51. Qian H, Chen L, Huang J, Wang X, Ma S, Cui F, et al. The lncRNA MIR4435-2HG promotes lung cancer progression by activating β-catenin signalling. J Mol Med (Berlin Germany). (2018) 96:753–64. doi: 10.1007/s00109-018-1654-5
52. Xiao Y, Bao Y, Tang L, and Wang L. LncRNA MIR4435-2HG is downregulated in osteoarthritis and regulates chondrocyte cell proliferation and apoptosis. J Orthop Surg Res. (2019) 14:247. doi: 10.1186/s13018-019-1278-7
53. Thomas NP, Li P, Fleming BC, Chen Q, Wei X, Xiao-Hua P, et al. Attenuation of cartilage pathogenesis in post-traumatic osteoarthritis (PTOA) in mice by blocking the stromal derived factor 1 receptor (CXCR4) with the specific inhibitor, AMD3100. J Orthop Res. (2015) 33:1071–8. doi: 10.1002/jor.22862
54. Jun JI and Lau LF. Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nature reviews. Drug Discov. (2011) 10:945–63. doi: 10.1038/nrd3599
55. Huang X, Ni B, Mao Z, Xi Y, Chu X, Zhang R, et al. NOV/CCN3 induces cartilage protection by inhibiting PI3K/AKT/mTOR pathway. J Cell Mol Med. (2019) 23:7525–34. doi: 10.1111/jcmm.v23.11
56. Janune D, Abd El Kader T, Aoyama E, Nishida T, Tabata Y, Kubota S, et al. Novel role of CCN3 that maintains the differentiated phenotype of articular cartilage. J Bone Mineral Metab. (2017) 35:582–97. doi: 10.1007/s00774-016-0793-4
57. Babey ME, Krause WC, Chen K, Herber CB, Torok Z, Nikkanen J, et al. A maternal brain hormone that builds bone. Nature. (2024) 632:357–65. doi: 10.1038/s41586-024-07634-3
58. Kalsbeek A, van der Spek R, Lei J, Endert E, Buijs RM, and Fliers E. Circadian rhythms in the hypothalamo–pituitary–adrenal (HPA) axis. Mol Cell Endocrinol. (2012) 349:20–9. doi: 10.1016/j.mce.2011.06.042
59. Joseph F, Ahmad AM, Ul-Haq M, Durham BH, Whittingham P, Fraser WD, et al. Effects of growth hormone administration on bone mineral metabolism, PTH sensitivity and PTH secretory rhythm in postmenopausal women with established osteoporosis*. J Bone Mineral Res. (2009) 23:721–9. doi: 10.1359/jbmr.071117
60. Berenbaum F and Meng QJ. The brain-joint axis in osteoarthritis: nerves, circadian clocks and beyond. Nat Rev Rheum. (2016) 12:508–16. doi: 10.1038/nrrheum.2016.93
61. Liu Z-Q, Wang J, Thapa A, Xie Y, Guo J, Li X, et al. Melatonin enhances chondrocytes survival through hyaluronic acid production and mitochondria protection. Arch Med Sci. (2021). doi: 10.5114/aoms/136343
62. Shang X, Wang Z, and Tao H. Mechanism and therapeutic effectiveness of nerve growth factor in osteoarthritis pain. Ther Clin Risk Manage. (2017) 13:951–6. doi: 10.2147/TCRM.S139814
63. Mashaghi A, Marmalidou A, Tehrani M, Grace PM, Pothoulakis C, and Dana R. Neuropeptide substance P and the immune response. Cell Mol Life Sci. (2016) 73:4249–64. doi: 10.1007/s00018-016-2293-z
64. Lauritano D, Mastrangelo F, D’Ovidio C, Ronconi G, Caraffa A, Gallenga CE, et al. Activation of mast cells by neuropeptides: the role of pro-inflammatory and anti-inflammatory cytokines. Int J Mol Sci. (2023) 24:4811. doi: 10.3390/ijms24054811
65. Zou Y, Liu C, Wang Z, Li G, and Xiao J. Neural and immune roles in osteoarthritis pain: Mechanisms and intervention strategies. J Orthop Translat. (2024) 48:123–32. doi: 10.1016/j.jot.2024.07.010
66. Wei X, Sun C, Zhou R-P, Ma G-G, Yang Y, Lu C, et al. Nerve growth factor promotes ASIC1a expression via the NF-κB pathway and enhances acid-induced chondrocyte apoptosis. Int Immunopharmacol. (2020) 82:106340. doi: 10.1016/j.intimp.2020.106340
67. Muschter D, Fleischhauer L, Taheri S, Schilling AF, Clausen-Schaumann H, and Grässel S. Sensory neuropeptides are required for bone and cartilage homeostasis in a murine destabilization-induced osteoarthritis model. Bone. (2020) 133:115181. doi: 10.1016/j.bone.2019.115181
68. Dontchev VD and Letourneau PC. Nerve growth factor and semaphorin 3A signaling pathways interact in regulating sensory neuronal growth cone motility. J Neurosci. (2002) 22:6659–69. doi: 10.1523/JNEUROSCI.22-15-06659.2002
69. Huang S, Gao D, Li Z, He H, Yu X, You X, et al. Neuronal guidance factor Sema3A inhibits neurite ingrowth and prevents chondrocyte hypertrophy in the degeneration of knee cartilage in mice, monkeys and humans. Bone Res. (2025) 13:4. doi: 10.1038/s41413-024-00382-0
70. Tsugawa Y, Handa H, and Imai T. Arginine induces IGF-1 secretion from the endoplasmic reticulum. Biochem Biophys Res Commun. (2019) 514:1128–32. doi: 10.1016/j.bbrc.2019.05.044
71. Lara-Diaz VJ, Castilla-Cortazar I, Martín-Estal I, García-Magariño M, Aguirre GA, Puche JE, et al. IGF-1 modulates gene expression of proteins involved in inflammation, cytoskeleton, and liver architecture. J Physiol Biochem. (2017) 73:245–58. doi: 10.1007/s13105-016-0545-x
72. Hartley A, Sanderson E, Paternoster L, Teumer A, Kaplan RC, Tobias JH, et al. Mendelian randomization provides evidence for a causal effect of higher serum IGF-1 concentration on risk of hip and knee osteoarthritis. Rheumatology. (2020) 60:1676–86. doi: 10.1093/rheumatology/keaa597
73. Hossain MA, Adithan A, Alam MJ, Kopalli SR, Kim B, Kang CW, et al. IGF-1 facilitates cartilage reconstruction by regulating PI3K/AKT, MAPK, and NF-kB signaling in rabbit osteoarthritis. J Inflammation Res. (2021) 14:3555–68. doi: 10.2147/JIR.S316756
74. Brubaker PL. The molecular determinants of glucagon-like peptide secretion by the intestinal L cell. Endocrinology. (2022) 163:bqac159. doi: 10.1210/endocr/bqac159
75. Holst JJ, Andersen DB, and Grunddal KV. Actions of glucagon-like peptide-1 receptor ligands in the gut. Br J Pharmacol. (2022) 179:727–42. doi: 10.1111/bph.v179.4
76. Chen J, Xie J-J, Shi K-S, Gu Y-T, Wu C-C, Xuan J, et al. Glucagon-like peptide-1 receptor regulates endoplasmic reticulum stress-induced apoptosis and the associated inflammatory response in chondrocytes and the progression of osteoarthritis in rat. Cell Death Dis. (2018) 9:212. doi: 10.1038/s41419-017-0217-y
77. Zhang X, Jiang J, Xu J, Chen J, Gu Y, and Wu G. Liraglutide, a glucagon-like peptide-1 receptor agonist, ameliorates inflammation and apoptosis via inhibition of receptor for advanced glycation end products signaling in AGEs induced chondrocytes. BMC Musculoskel Disord. (2024) 25:601. doi: 10.1186/s12891-024-07640-6
78. Yang Y, Hao C, Jiao T, Yang Z, Li H, Zhang Y, et al. Osteoarthritis treatment via the GLP-1-mediated gut-joint axis targets intestinal FXR signaling. Science. (2025) 388:eadt0548. doi: 10.1126/science.adt0548
79. Wallimann A, Magrath W, Thompson K, Moriarty T, Richards RG, Akdis CA, et al. Gut microbial-derived short-chain fatty acids and bone: a potential role in fracture healing. Eur Cells mater. (2021) 41:454–70. doi: 10.22203/eCM.v041a29
80. Yang KL, Mullins BJ, Lejeune A, Ivanova E, Shin J, Bajwa S, et al. Mitigation of osteoclast-mediated arthritic bone remodeling by short chain fatty acids. Arthritis Rheum. (2024) 76:647–59. doi: 10.1002/art.42765
81. Guan Z, Jin X, Guan Z, Liu S, Tao K, and Luo L. The gut microbiota metabolite capsiate regulate SLC2A1 expression by targeting HIF-1α to inhibit knee osteoarthritis-induced ferroptosis. Aging Cell. (2023) 22:e13807. doi: 10.1111/acel.13807
82. Chen C, Cao Z, Lei H, Zhang C, Wu M, Huang S, et al. Microbial tryptophan metabolites ameliorate ovariectomy-induced bone loss by repairing intestinal ahR-mediated gut-bone signaling pathway. Adv Sci. (2024) 11(36):e2404545. doi: 10.1002/advs.202404545
83. Richette P, Dumontier MF, Tahiri K, Widerak M, Torre A, Benallaoua M, et al. Oestrogens inhibit interleukin 1beta-mediated nitric oxide synthase expression in articular chondrocytes through nuclear factor-kappa B impairment. Ann rheum Dis. (2007) 66:345–50. doi: 10.1136/ard.2006.059550
84. Sun Y, Leng P, Guo P, Gao H, Liu Y, Li C, et al. G protein coupled estrogen receptor attenuates mechanical stress-mediated apoptosis of chondrocyte in osteoarthritis via suppression of Piezo1. Mol Med (Cambridge Mass.). (2021) 27:96. doi: 10.1186/s10020-021-00360-w
85. Wang N, Lu Y, Rothrauff BB, Zheng A, Lamb A, Yan Y, et al. Mechanotransduction pathways in articular chondrocytes and the emerging role of estrogen receptor-α. Bone Res. (2023) 11:13. doi: 10.1038/s41413-023-00248-x
86. Chuang SC, Chen CH, Chou YS, Ho ML, and Chang JK. G Protein-Coupled Estrogen Receptor Mediates Cell Proliferation through the cAMP/PKA/CREB Pathway in Murine Bone Marrow Mesenchymal Stem Cells. Int J Mol Sci. (2020) 21:6490. doi: 10.3390/ijms21186490
87. Rosa SC, Rufino AT, Judas F, Tenreiro C, Lopes MC, and Mendes AF. Expression and function of the insulin receptor in normal and osteoarthritic human chondrocytes: modulation of anabolic gene expression, glucose transport and GLUT-1 content by insulin. Osteoarthr cartil. (2011) 19:719–27. doi: 10.1016/j.joca.2011.02.004
88. Phornphutkul C, Wu K-Y, and Gruppuso PA. The role of insulin in chondrogenesis. Mol Cell Endocrinol. (2006) 249:107–15. doi: 10.1016/j.mce.2006.02.002
89. Hamada D, Maynard R, Schott E, Drinkwater CJ, Ketz JP, Kates SL, et al. Suppressive effects of insulin on tumor necrosis factor-dependent early osteoarthritic changes associated with obesity and type 2 diabetes mellitus. Arthritis Rheum. (2016) 68:1392–402. doi: 10.1002/art.39561
90. Torres ES, Andrade CV, Fonseca EC, Mello MA, and Duarte ME. Insulin impairs the maturation of chondrocytes in vitro. Braz J Med Biol Res = Rev Bras pesquisas medicas e biol. (2003) 36:1185–92. doi: 10.1590/S0100-879X2003000900007
91. Alarid ET, Schlechter NL, Russell SM, and Nicoll CS. Evidence suggesting that insulin-like growth factor-I is necessary for the trophic effects of insulin on cartilage growth in vivo. Endocrinology. (1992) 130:2305–9. doi: 10.1210/endo.130.4.1547741
92. Ribeiro M, López de Figueroa P, Blanco FJ, Mendes AF, and Caramés B. Insulin decreases autophagy and leads to cartilage degradation. Osteoarthr cartil. (2016) 24:731–9. doi: 10.1016/j.joca.2015.10.017
93. Qiao L, Li Y, and Sun S. Insulin exacerbates inflammation in fibroblast-like synoviocytes. Inflammation. (2020) 43:916–36. doi: 10.1007/s10753-020-01178-0
94. Hamada D, Maynard R, Fransworth C, Kates S, Hilton M, Zuscik M, et al. Insulin selectively suppresses TNF&x3b1;/IL-1&x3b2;-induced catabolic enzymes in Ostezoarthritic fibroblast-like synovziocytes. Osteoarthr cartil. (2014) 22:S451–2. doi: 10.1016/j.joca.2014.02.856
95. Yang RZ, Zheng HL, Xu WN, Zheng XF, Li B, Jiang LS, et al. Vascular endothelial cell-secreted exosomes facilitate osteoarthritis pathogenesis by promoting chondrocyte apoptosis. Aging. (2021) 13:4647–62. doi: 10.18632/aging.202506
96. Adams SL, Cohen Aj Fau - Lassová L, and Lassová L. Integration of signaling pathways regulating chondrocyte differentiation during endochondral bone formation. J Cell Physiol. (2007) 213(3):635–41. doi: 10.1002/jcp.21262
97. Xiao P, Zhu X, Sun J, Zhang Y, Qiu W, Li J, et al. Cartilage tissue miR-214-3p regulates the TrkB/ShcB pathway paracrine VEGF to promote endothelial cell migration and angiogenesis. Bone. (2021) 151:116034. doi: 10.1016/j.bone.2021.116034
98. Yang K, Shi J, Hu Z, and Hu X. The deficiency of miR-214-3p exacerbates cardiac fibrosis via miR-214-3p/NLRC5 axis. Clin Sci. (2019) 133:1845–56. doi: 10.1042/CS20190203
99. Adam MS, Zhuang H, Ren X, Zhang Y, and Zhou P. The metabolic characteristics and changes of chondrocytes in vivo and in vitro in osteoarthritis. Front Endocrinol (Lausanne). (2024) 15:1393550. doi: 10.3389/fendo.2024.1393550
100. Zhang S, Wu F, Zhan L, Lin W, Liang C, Pang Y, et al. Exercise regulates the lactate receptor HCAR1 and ERK1/2-PI3K/akt pathways to promote cerebral angiogenesis. Iran J Public Health. (2022) 51:2298–307. doi: 10.18502/ijph.v51i10.10988
101. Salomon J, Matusiak Ł, Nowicka-Suszko D, and Szepietowski JC. Chitinase-3-like protein 1 (YKL-40) reflects the severity of symptoms in atopic dermatitis. J Immunol Res. (2017) 2017:5746031. doi: 10.1155/2017/5746031
102. Gao J, Iwamoto H, Koskela J, Alenius H, Hattori N, Kohno N, et al. Characterization of sputum biomarkers for asthma-COPD overlap syndrome. Int J chronic obstruct pulmon Dis. (2016) 11:2457–65. doi: 10.2147/COPD.S113484
103. Einarsson JM, Bahrke S, Sigurdsson BT, Ng C-H, Petersen PH, Sigurjonsson OE, et al. Partially acetylated chitooligosaccharides bind to YKL-40 and stimulate growth of human osteoarthritic chondrocytes. Biochem Biophys Res Commun. (2013) 434:298–304. doi: 10.1016/j.bbrc.2013.02.122
104. Bishnoi M, Jain A, Hurkat P, and Jain SK. Chondroitin sulphate: a focus on osteoarthritis. Glycoconjugate J. (2016) 33:693–705. doi: 10.1007/s10719-016-9665-3
105. Tzankov A, Bhattacharyya S, Kotlo K, and Tobacman JK. Increase in chondroitin sulfate and decline in arylsulfatase B may contribute to pathophysiology of COVID-19 respiratory failure. Pathobiol: J immunopathol Mol Cell Biol. (2022) 89:81–91. doi: 10.1159/000519542
106. Papakonstantinou E, Klagas I, Roth M, Tamm M, and Stolz D. Acute exacerbations of COPD are associated with increased expression of heparan sulfate and chondroitin sulfate in BAL. Chest. (2016) 149:685–95. doi: 10.1378/chest.14-2868
107. Kovesdy CP and Quarles LD. FGF23 from bench to bedside. American journal of physiology. Renal Physiol. (2016) 310:F1168–74. doi: 10.1152/ajprenal.00606.2015
108. Gowler PRW, Li L, Woodhams SG, Bennett AJ, Suzuki R, Walsh DA, et al. Peripheral brain-derived neurotrophic factor contributes to chronic osteoarthritis joint pain. Pain. (2020) 161:61–73. doi: 10.1097/j.pain.0000000000001694
109. Miller RE, Tran PB, Das R, Ghoreishi-Haack N, Ren D, Miller RJ, et al. CCR2 chemokine receptor signaling mediates pain in experimental osteoarthritis. Proc Natl Acad Sci U S A. (2012) 109:20602–7. doi: 10.1073/pnas.1209294110
110. Stöckl S, Eitner A, Bauer RJ, König M, Johnstone B, Grässel S, et al. and alpha-calcitonin gene-related peptide differentially affect human osteoarthritic and healthy chondrocytes. Front Immunol. (2021) 12:722884. doi: 10.3389/fimmu.2021.722884
111. Hu J, Cui W, Ding W, Gu Y, Wang Z, and Fan W. Globular adiponectin attenuated H2O2-induced apoptosis in rat chondrocytes by inducing autophagy through the AMPK/mTOR pathway. Cell Physiol Biochem. (2017) 43:367–82. doi: 10.1159/000480416
112. Tan L, Armstrong AR, Rosas S, Patel CM, Vander Wiele SS, Willey JS, et al. Nuclear protein-1 is the common link for pathways activated by aging and obesity in chondrocytes: A potential therapeutic target for osteoarthritis. FASEB J. (2023) 37:e23133. doi: 10.1096/fj.202201700RR
113. Fu R, Han F, Liu L, Yu F, Gui Z, Wang X, et al. The effects of leptin on the proliferation and differentiation of primary chondrocytes in vitro and cartilage regeneration in vivo. ACS biomater Sci Eng. (2019) 5:1907–19. doi: 10.1021/acsbiomaterials.8b01168
114. Izquierdo AG, Crujeiras AB, Casanueva FF, and Carreira MC. Leptin, obesity, and leptin resistance: where are we 25 years later? Nutrients. (2019) 11:2704. doi: 10.3390/nu11112704
115. Lambova SN, Batsalova T, Moten D, Stoyanova S, Georgieva E, Belenska-Todorova L, et al. Serum leptin and resistin levels in knee osteoarthritis-clinical and radiologic links: towards precise definition of metabolic type knee osteoarthritis. Biomedicines. (2021) 9:1019. doi: 10.3390/biomedicines9081019
116. Mourmoura E, Papathanasiou I, Trachana V, Konteles V, Tsoumpou A, Goutas A, et al. Leptin-depended NLRP3 inflammasome activation in osteoarthritic chondrocytes is mediated by ROS. Mech Ageing Dev. (2022) 208:111730. doi: 10.1016/j.mad.2022.111730
117. Wu J, Kuang L, Chen C, Yang J, Zeng W-N, Li T, et al. miR-100-5p-abundant exosomes derived from infrapatellar fat pad MSCs protect articular cartilage and ameliorate gait abnormalities via inhibition of mTOR in osteoarthritis. Biomaterials. (2019) 206:87–100. doi: 10.1016/j.biomaterials.2019.03.022
118. Yan M, Xu Z, Yin Y, Fu T, Zhou Z, Yang L, et al. 1625-P: osteoprotection by FNIP1 signaling in skeletal muscle. Diabetes. (2023) 72(Supplement_1):1625–P. doi: 10.2337/db23-1625-P
119. Xiao L, Liu J, Sun Z, Yin Y, Mao Y, Xu D, et al. AMPK-dependent and -independent coordination of mitochondrial function and muscle fiber type by FNIP1. PloS Genet. (2021) 17:e1009488. doi: 10.1371/journal.pgen.1009488
120. Ping SH, Tian FM, Liu H, Sun Q, Shao LT, Lian QQ, et al. Raloxifene inhibits the overexpression of TGF-β1 in cartilage and regulates the metabolism of subchondral bone in rats with osteoporotic osteoarthritis. Bosnian J basic Med Sci. (2021) 21:284–93. doi: 10.17305/bjbms.2020.5142
121. Zheng L, Pi C, Zhang J, Fan Y, Cui C, Zhou Y, et al. Aberrant activation of latent transforming growth factor-β initiates the onset of temporomandibular joint osteoarthritis. Bone Res. (2018) 6:26. doi: 10.1038/s41413-018-0027-6
122. Liu J, Tao J, Cai G, Chen J, Zhao L, Wang Y, et al. The altered hippocampal functional connectivity and serum brain-derived neurotrophic factor level predict cognitive decline in patients with knee osteoarthritis. Cereb Cortex. (2023) 33:10584–94. doi: 10.1093/cercor/bhad305
123. Innes KE and Sambamoorthi U. The Association of Osteoarthritis and Related Pain Burden to Incident Alzheimer’s Disease and Related Dementias: A Retrospective Cohort Study of U.S. Medicare Beneficiaries. J Alzheimers Dis. (2020) 75(3):789–805. doi: 10.3233/JAD-191311
124. Kyrkanides S, Tallents RH, Miller J-NH, Olschowka ME, Johnson R, Yang M, et al. Osteoarthritis accelerates and exacerbates Alzheimer’s disease pathology in mice. J Neuroinflamm. (2011) 8:112. doi: 10.1186/1742-2094-8-112
125. Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, and Ramos-Escobar N. Neuroinflammation as a common feature of neurodegenerative disorders. Front Pharmacol. (2019) 10:1008. doi: 10.3389/fphar.2019.01008
126. Cao W and Zheng H. Correction to: Peripheral immune system in aging and Alzheimer’s disease. Mol Neurodegener. (2018) 13(1):58. doi: 10.1186/s13024-018-0290-4. Erratum for: Mol Neurodegener. 2018 Oct 3;13(1):51. doi: 10.1186/s13024-018-0284-2
127. Banks WA. The blood-brain barrier in neuroimmunology: Tales of separation and assimilation. Brain Behav Immun. (2015) 44:1–8. doi: 10.1016/j.bbi.2014.08.007
128. Gupta DP, Lee Y-S, Choe Y, Kim K-T, Song GJ, and Hwang S-C. Knee osteoarthritis accelerates amyloid beta deposition and neurodegeneration in a mouse model of Alzheimer’s disease. Mol Brain. (2023) 16:1. doi: 10.1186/s13041-022-00986-9
129. Ittner LM and Götz J. Amyloid-β and tau–a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci. (2011) 12(2):65–72. doi: 10.1038/nrn2967
130. Perruccio AV, Kral C, Wilfong JM, Eder L, Power JD, Canizares M, et al. Cardiovascular disease onset in osteoarthritis and the role of sex and systemic inflammation. Osteoarthr Cartil. (2024) 32:S326–7. doi: 10.1016/j.joca.2024.02.481
131. Uematsu T, Nojiri S, Ishijima M, and Nishizaki Y. Association between osteoarthritis and cardiovascular disease in elderly in Japan: an administrative claims database analysis. BMJ Open. (2024) 14:e080387. doi: 10.1136/bmjopen-2023-080387
132. Athow NF, Morgan PM, and Brown GA. Hip and knee osteoarthritis, not nonsteroidal anti-inflammatory drugs, are linked to cardiac disease. J Arthrop. (2023) 38:2455–63. doi: 10.1016/j.arth.2023.05.026
133. Yin M, Xu W, Pang J, Xie S, Xiang M, Shi B, et al. Causal relationship between osteoarthritis with atrial fibrillation and coronary atherosclerosis: a bidirectional Mendelian randomization study of European ancestry. Front Cardiovasc Med. (2023) 10:1213672. doi: 10.3389/fcvm.2023.1213672
134. Fernandes GS and Valdes AM. Cardiovascular disease and osteoarthritis: common pathways and patient outcomes. Eur J Clin Invest. (2015) 45:405–14. doi: 10.1111/eci.2015.45.issue-4
135. Atiquzzaman M, Karim ME, Kopec J, Wong H, and Anis AH. Role of nonsteroidal antiinflammatory drugs in the association between osteoarthritis and cardiovascular diseases: A longitudinal study. Arthritis Rheum. (2019) 71:1835–43. doi: 10.1002/art.v71.11
136. Wang Y, Pontoh EW, Hussain SM, Lim YZ, Jones G, Hill CL, et al. Association between popliteal artery wall thickness and structural progression in patients with symptomatic knee osteoarthritis. Rheumatol (Oxford England). (2023) 62:1645–51. doi: 10.1093/rheumatology/keac469
137. Zhang M, Xiong W, Qiao R, Li M, Zhang C, Yang C, et al. Irisin in the modulation of bone and cartilage homeostasis: a review on osteoarthritis relief potential. Front Physiol. (2025) 16:1570157. doi: 10.3389/fphys.2025.1570157
138. Nakagawa T, Yasuda T, Hoshikawa H, Shimizu M, Kakinuma T, Chen M, et al. LOX-1 expressed in cultured rat chondrocytes mediates oxidized LDL-induced cell death-possible role of dephosphorylation of Akt. Biochem Biophys Res Commun. (2002) 299:91–7. doi: 10.1016/S0006-291X(02)02597-4
139. Lee JS, Kim YH, Jhun J, Na HS, Um IG, Choi JW, et al. Oxidized LDL accelerates cartilage destruction and inflammatory chondrocyte death in osteoarthritis by disrupting the TFEB-regulated autophagy-lysosome pathway. Immune netw. (2024) 24:e15. doi: 10.4110/in.2024.24.e15
140. de Munter W, van den Bosch MH, Slöetjes AW, Croce KJ, Vogl T, Roth J, et al. High LDL levels lead to increased synovial inflammation and accelerated ectopic bone formation during experimental osteoarthritis. Osteoarthr cartil. (2016) 24:844–55. doi: 10.1016/j.joca.2015.11.016
141. de Munter W, Blom AB, Helsen MM, Walgreen B, van der Kraan PM, Joosten LA, et al. Cholesterol accumulation caused by low density lipoprotein receptor deficiency or a cholesterol-rich diet results in ectopic bone formation during experimental osteoarthritis. Arthritis Res Ther. (2013) 15:R178. doi: 10.1186/ar4367
142. Choi W-S, Lee G, Song W-H, Koh J-T, Yang J, Kwak J-S, et al. The CH25H–CYP7B1–RORα axis of cholesterol metabolism regulates osteoarthritis. Nature. (2019) 566:254–8. doi: 10.1038/s41586-019-0920-1
143. Haugen IK, Ramachandran VS, Misra D, Neogi T, Niu J, Yang T, et al. Hand osteoarthritis in relation to mortality and incidence of cardiovascular disease: data from the Framingham heart study. Ann rheum Dis. (2015) 74:74–81. doi: 10.1136/annrheumdis-2013-203789
144. Havelka S, Horn V, Spohrová D, and Valouch P. The calcified-noncalcified cartilage interface: the tidemark. Acta biol Hungarica. (1984) 35:271–9.
145. Oegema TR Jr, Carpenter RJ, Hofmeister F, and Thompson RC Jr. The interaction of the zone of calcified cartilage and subchondral bone in osteoarthritis. Microsc Res techn. (1997) 37:324–32. doi: 10.1002/(SICI)1097-0029(19970515)37:4<324::AID-JEMT7>3.0.CO;2-K
146. Knight AD and Levick JR. Morphometry of the ultrastructure of the blood-joint barrier in the rabbit knee. Q J Exp Physiol (Cambridge England). (1984) 69:271–88. doi: 10.1113/expphysiol.1984.sp002805
147. Dryll A, Lansaman J, Cazalis P, Peltier AP, and De Seze S. Light and electron microscopy study of capillaries in normal and inflammatory human synovial membrane. J Clin Pathol. (1977) 30:556–62. doi: 10.1136/jcp.30.6.556
148. Chappard C, Peyrin F, Bonnassie A, Lemineur G, Brunet-Imbault B, Lespessailles E, et al. Subchondral bone micro-architectural alterations in osteoarthritis: a synchrotron micro-computed tomography study. Osteoarthr cartil. (2006) 14:215–23. doi: 10.1016/j.joca.2005.09.008
149. Chiba K, Nango N, Kubota S, Okazaki N, Taguchi K, Osaki M, et al. Relationship between microstructure and degree of mineralization in subchondral bone of osteoarthritis: a synchrotron radiation µCT study. J Bone mineral Res. (2012) 27:1511–7. doi: 10.1002/jbmr.1609
150. Findlay DM. Vascular pathology and osteoarthritis. Rheumatol (Oxford England). (2007) 46:1763–8. doi: 10.1093/rheumatology/kem191
151. James J and Steijn-Myagkaya GL. Death of osteocytes. Electron microscopy after in vitro ischaemia. J Bone Joint Surg. (1986) 68:620–4. doi: 10.1302/0301-620X.68B4.3733842
152. Hussain SM, Dawson C, Wang Y, Tonkin AM, Chou L, Wluka AE, et al. Vascular pathology and osteoarthritis: A systematic review. J Rheumatol. (2020) 47:748–60. doi: 10.3899/jrheum.181236
153. Wshah A, Guilcher SJ, Goldstein R, and Brooks D. Prevalence of osteoarthritis in individuals with COPD: a systematic review. Int J Chron Obstruct Pulmon Dis. (2018) 13:1207–16. doi: 10.2147/COPD.S158614
154. Koo H-K, Song P, and Lee J-H. Novel association between asthma and osteoarthritis: a nationwide health and nutrition examination survey. BMC Pulmon Med. (2021) 21:59. doi: 10.1186/s12890-021-01425-6
155. Yuan Y, Jiao B, Qu L, Yang D, and Liu R. The development of COVID-19 treatment. Front Immunol. (2023) 14:1125246. doi: 10.3389/fimmu.2023.1125246
156. To KK, Sridhar S, Chiu KH, Hung DL, Li X, Hung IF, et al. Lessons learned 1 year after SARS-CoV-2 emergence leading to COVID-19 pandemic. Emerg microbes infect. (2021) 10:507–35. doi: 10.1080/22221751.2021.1898291
157. Daoud S and Taha M. Ligand-based modeling of CXC chemokine receptor 4 and identification of inhibitors of novel chemotypes as potential leads towards new anti- COVID-19 treatments. Med Chem (Shariqah (United Arab Emirates)). (2022) 18:871–83. doi: 10.2174/1573406418666220118153541
158. Li P, Wei L, and Wei X. Blocking hypoxia-induced CXCR4 by AMD3100 inhibits production of OA associated catabolic mediators il-1β and MMP-13. J Orthop Translation. (2016) 7:137. doi: 10.1016/j.jot.2016.06.212
159. Zheng H, Fang J, Lu W, Liu Y, Chen S, Huang G, et al. TCF12 regulates the TGF-β/Smad2/3 signaling pathway to accelerate the progression of osteoarthritis by targeting CXCR4. J Orthop Translation. (2024) 44:35–46. doi: 10.1016/j.jot.2023.11.006
160. Kai Y, Yoneyama H, Yoshikawa M, Kimura H, and Muro S. Chondroitin sulfate in tissue remodeling: Therapeutic implications for pulmonary fibrosis. Respir Invest. (2021) 59:576–88. doi: 10.1016/j.resinv.2021.05.012
161. Bruyère O. Pharmaceutical-grade chondroitin sulfate in the management of knee osteoarthritis. Expert Opin pharmacother. (2018) 19:409–12. doi: 10.1080/14656566.2018.1442438
162. Durmuş U, Doğan N, Pekdemir M, Yılmaz S, Yaka E, Karadaş A, et al. The value of lactate clearance in admission decisions of patients with acute exacerbation of COPD. Am J Emergency Med. (2018) 36:972–6. doi: 10.1016/j.ajem.2017.11.002
163. Huang Y-F, Wang G, Ding L, Bai Z-R, Leng Y, Tian J-W, et al. Lactate-upregulated NADPH-dependent NOX4 expression via HCAR1/PI3K pathway contributes to ROS-induced osteoarthritis chondrocyte damage. Redox Biol. (2023) 67:102867. doi: 10.1016/j.redox.2023.102867
164. Trefts E, Gannon M, and Wasserman DH. The liver. Curr Biol. (2017) 27:R1147–51. doi: 10.1016/j.cub.2017.09.019
165. Lu Y, Zhang J, Li H, and Li T. Association of non-alcoholic fatty liver disease with self-reported osteoarthritis among the US adults. Arthritis Res Ther. (2024) 26:40. doi: 10.1186/s13075-024-03272-2
166. Habib GS and Saliba WR. Arthritis associated with non-alcoholic steatohepatitis. Ann rheum Dis. (2001) 60:815–6. doi: 10.1136/ard.60.8.815a
167. Cerda C, Bruguera M, and Parés A. Hepatotoxicity associated with glucosamine and chondroitin sulfate in patients with chronic liver disease. World J gastroenterol. (2013) 19:5381–4. doi: 10.3748/wjg.v19.i32.5381
168. Barakat M, Niknam N, Valceanu A, and Kasturi L. Chondroitin sulphate: unmasking autoimmune hepatitis. Prz Gastroenterol. (2019) 14:302–3. doi: 10.5114/pg.2019.88173
169. Clemente JC, Ursell LK, Parfrey LW, and Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. (2012) 148:1258–70. doi: 10.1016/j.cell.2012.01.035
170. Reigstad CS and Kashyap PC. Beyond phylotyping: understanding the impact of gut microbiota on host biology. Neurogastroenterol Motil. (2013) 25:358–72. doi: 10.1111/nmo.2013.25.issue-5
171. Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G, et al. The gut microbiota and host health: a new clinical frontier. Gut. (2016) 65:330–9. doi: 10.1136/gutjnl-2015-309990
172. Zhang Y, Wang H, Sang Y, Liu M, Wang Q, Yang H, et al. Gut microbiota in health and disease: advances and future prospects. MedComm. (2024) 5:e70012. doi: 10.1002/mco2.70012
173. de Sire A, de Sire R, Petito V, Masi L, Cisari C, Gasbarrini A, et al. Gut-joint axis: the role of physical exercise on gut microbiota modulation in older people with osteoarthritis. Nutrients. (2020) 12:574. doi: 10.3390/nu12020574
174. Maeda Y, Kurakawa T, Umemoto E, Motooka D, Ito Y, Gotoh K, et al. Dysbiosis contributes to arthritis development via activation of autoreactive T cells in the intestine. Arthritis Rheum. (2016) 68:2646–61. doi: 10.1002/art.v68.11
175. 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. (2021) 23:42. doi: 10.1186/s13075-021-02427-9
176. Pan B, Guo Q, Cai J, Chen L, Zhao Z, Shen P, et al. Investigating the causal impact of gut microbiota on arthritis via inflammatory proteins using mendelian randomization. Sci Rep. (2024) 14:27433. doi: 10.1038/s41598-024-79336-9
177. Lu Y, Zhou Z, and Pan D. Genetic insights into the roles of fatty acids and gut microbiota in osteoarthritis: A Mendelian randomization study. Medicine. (2024) 103:e40674. doi: 10.1097/MD.0000000000040674
178. Yao Y, Cai X, Zheng Y, Zhang M, Fei W, Sun D, et al. Short-chain fatty acids regulate B cells differentiation via the FFA2 receptor to alleviate rheumatoid arthritis. Br J Pharmacol. (2022) 179:4315–29. doi: 10.1111/bph.v179.17
179. Bacchetta J, Sea JL, Chun RF, Lisse TS, Wesseling-Perry K, Gales B, et al. Fibroblast growth factor 23 inhibits extrarenal synthesis of 1,25-dihydroxyvitamin D in human monocytes. J Bone mineral Res. (2013) 28:46–55. doi: 10.1002/jbmr.1740
180. Martin A, David V, and Quarles LD. Regulation and function of the FGF23/klotho endocrine pathways. Physiol Rev. (2012) 92:131–55. doi: 10.1152/physrev.00002.2011
181. Meo Burt P, Xiao L, and Hurley MM. FGF23 regulates wnt/β-catenin signaling-mediated osteoarthritis in mice overexpressing high-molecular-weight FGF2. Endocrinology. (2018) 159:2386–96. doi: 10.1210/en.2018-00184
182. Bianchi A, Guibert M, Cailotto F, Gasser A, Presle N, Mainard D, et al. Fibroblast Growth Factor 23 drives MMP13 expression in human osteoarthritic chondrocytes in a Klotho-independent manner. Osteoarthr cartil. (2016) 24:1961–9. doi: 10.1016/j.joca.2016.06.003
183. Engdahl C, Börjesson AE, Forsman HF, Andersson A, Stubelius A, Krust A, et al. The role of total and cartilage-specific estrogen receptor alpha expression for the ameliorating effect of estrogen treatment on arthritis. Arthritis Res Ther. (2014) 16:R150. doi: 10.1186/ar4612
184. Ziemian SN, Ayobami OO, Rooney AM, Kelly NH, Holyoak DT, Ross FP, et al. Low bone mass resulting from impaired estrogen signaling in bone increases severity of load-induced osteoarthritis in female mice. Bone. (2021) 152:116071. doi: 10.1016/j.bone.2021.116071
185. Pan Q, O’Connor MI, Coutts RD, Hyzy SL, Olivares-Navarrete R, Schwartz Z, et al. Characterization of osteoarthritic human knees indicates potential sex differences. Biol Sex Dif. (2016) 7:27. doi: 10.1186/s13293-016-0080-z
186. Lu Z, Zhang A, Wang J, Han K, and Gao H. Estrogen alleviates post-traumatic osteoarthritis progression and decreases p-EGFR levels in female mouse cartilage. BMC Musculoskelet Disord. (2022) 23:685. doi: 10.1186/s12891-022-05608-y
187. Mei R, Lou P, You G, Jiang T, Yu X, and Guo L. 17β-estradiol induces mitophagy upregulation to protect chondrocytes via the SIRT1-mediated AMPK/mTOR signaling pathway. Front Endocrinol (Lausanne). (2020) 11:615250. doi: 10.3389/fendo.2020.615250
188. Zhang E, Zhang H, Liu F, Dong C, Yao Y, Yun Z, et al. Estrogen exerts anti-inflammatory effects by inhibiting NF-κB pathway through binding with estrogen receptor β on synovicytes of osteoarthritis. Xi bao yu fen zi mian yi xue za zhi = Chin J Cell Mol Immunol. (2016) 32:1605–9.
189. Li Z, Yin S, Zhao G, and Cao X. Association between sarcopenic obesity and osteoarthritis: The potential mediating role of insulin resistance. Exp Gerontol. (2024) 197:112611. doi: 10.1016/j.exger.2024.112611
190. Yu H-J, Ho M, Liu X, Yang J, Chau PH, and Fong YT. Association of weight status and the risks of diabetes in adults: a systematic review and meta-analysis of prospective cohort studies. Int J Obes. (2022) 46:1101–13. doi: 10.1038/s41366-022-01096-1
191. Visser AW, de Mutsert R, le Cessie S, den Heijer M, Rosendaal FR, and Kloppenburg M. The relative contribution of mechanical stress and systemic processes in different types of osteoarthritis: the NEO study. Ann rheum Dis. (2015) 74:1842–7. doi: 10.1136/annrheumdis-2013-205012
192. Yan W and Li X. Impact of diabetes and its treatments on skeletal diseases. Front Med. (2013) 7:81–90. doi: 10.1007/s11684-013-0243-9
193. Courties A and Sellam J. Osteoarthritis and type 2 diabetes mellitus: What are the links? Diabetes Res Clin Pract. (2016) 122:198–206. doi: 10.1016/j.diabres.2016.10.021
194. King LK, Henneicke H, Seibel MJ, March L, and Anandacoomarasmy A. Association of adipokines and joint biomarkers with cartilage-modifying effects of weight loss in obese subjects. Osteoarthr cartil. (2015) 23:397–404. doi: 10.1016/j.joca.2014.11.020
195. Frommer KW, Hasseli R, Schäffler A, Lange U, Rehart S, Steinmeyer J, et al. Free fatty acids in bone pathophysiology of rheumatic diseases. Front Immunol. (2019) 10:2757. doi: 10.3389/fimmu.2019.02757
196. Loef M, van de Stadt L, Böhringer S, Bay-Jensen AC, Mobasheri A, Larkin J, et al. The association of the lipid profile with knee and hand osteoarthritis severity: the IMI-APPROACH cohort. Osteoarthritis Cartilage. (2022) 30(8):1062–9. doi: 10.1016/j.joca.2022.05.008
197. Krumbholz G, Junker S, Meier FMP, Rickert M, Steinmeyer J, Rehart S, et al. Response of human rheumatoid arthritis osteoblasts and osteoclasts to adiponectin. Clin Exp Rheumatol. (2017) 35(3):406–14.
198. Chen TH, Chen L, Hsieh MS, Chang CP, Chou DT, and Tsai SH. Evidence for a protective role for adiponectin in osteoarthritis. Biochim Biophys Acta. (2006) 1762:711–8. doi: 10.1016/j.bbadis.2006.06.008
199. Khoramipour K, Chamari K, Hekmatikar AA, Ziyaiyan A, Taherkhani S, Elguindy NM, et al. Adiponectin: structure, physiological functions, role in diseases, and effects of nutrition. Nutrients. (2021) 13:1180. doi: 10.3390/nu13041180
200. Wada N, Hirako S, Takenoya F, Kageyama H, Okabe M, and Shioda S. Leptin and its receptors. J Chem Neuroanat. (2014) 61-62:191–9. doi: 10.1016/j.jchemneu.2014.09.002
201. Lee JY, Han K, McAlindon TE, Park YG, and Park SH. Lower leg muscle mass relates to knee pain in patients with knee osteoarthritis. Int J rheum Dis. (2018) 21:126–33. doi: 10.1111/apl.2018.21.issue-1
202. Wu Y, Hou M, Deng Y, Xia X, Liu Y, Yu J, et al. Swimming exercise induces redox-lipid crosstalk to ameliorate osteoarthritis progression. Redox Biol. (2025) 81:103535. doi: 10.1016/j.redox.2025.103535
203. Tuna S, Balcı N, and Özçakar L. The relationship between femoral cartilage thickness and muscle strength in knee osteoarthritis. Clin Rheumatol. (2016) 35:2073–7. doi: 10.1007/s10067-016-3271-4
204. Xu C, Nie Y, Tan C, Zeng Y, Si H, Zhou Z, et al. Role of lower limb muscle strength in knee osteoarthritis progression for patients with mild and moderate knee osteoarthritis. Am J Phys Med Rehabil. (2022) 101:433–8. doi: 10.1097/PHM.0000000000001843
205. Mohajer B, Dolatshahi M, Moradi K, Najafzadeh N, Eng J, Zikria B, et al. Role of thigh muscle changes in knee osteoarthritis outcomes: osteoarthritis initiative data. Radiology. (2022) 305:169–78. doi: 10.1148/radiol.212771
206. Awad K, Moore L, Huang J, Brotto L, and Brotto M. Can acute muscle damage lead to osteoarthritis? Innovation Aging. (2023) 7:768–8. doi: 10.1093/geroni/igad104.2481
207. Tung EV, Marriott KA, Laing AC, Mourtzakis M, and Maly MR. The relationship between muscle capacity utilization during gait and pain in people with symptomatic knee osteoarthritis. Gait Post. (2022) 94:58–66. doi: 10.1016/j.gaitpost.2022.02.021
208. Sheng R, Cao M, Song M, Wang M, Zhang Y, Shi L, et al. Muscle-bone crosstalk via endocrine signals and potential targets for osteosarcopenia-related fracture. J Orthop Translat. (2023) 43:36–46. doi: 10.1016/j.jot.2023.09.007
209. Kamizaki K, Katsukawa M, Yamamoto A, Fukada SI, Uezumi A, Endo M, et al. Ror2 signaling regulated by differential Wnt proteins determines pathological fate of muscle mesenchymal progenitors. Cell Death Dis. (2024) 15:784. doi: 10.1038/s41419-024-07173-9
210. Julien A, Kanagalingam A, Martínez-Sarrà E, Megret J, Luka M, Ménager M, et al. Direct contribution of skeletal muscle mesenchymal progenitors to bone repair. Nat Commun. (2021) 12:2860. doi: 10.1038/s41467-021-22842-5
211. Pan J, Zhou X, Li W, Novotny JE, Doty SB, and Wang L. In situ measurement of transport between subchondral bone and articular cartilage. J orthop Res. (2009) 27:1347–52. doi: 10.1002/jor.v27:10
212. Zhang S, Li T, Feng Y, Zhang K, Zou J, Weng X, et al. Exercise improves subchondral bone microenvironment through regulating bone-cartilage crosstalk. Front Endocrinol (Lausanne). (2023) 14:1159393. doi: 10.3389/fendo.2023.1159393
213. Li J, Fu X, Zhang D, Guo D, Xu S, Wei J, et al. Co-culture with osteoblasts up-regulates glycolysis of chondrocytes through MAPK/HIF-1 pathway. Tissue Cell. (2022) 78:101892. doi: 10.1016/j.tice.2022.101892
214. Odgren PR, Witwicka H, and Reyes-Gutierrez P. The cast of clasts: catabolism and vascular invasion during bone growth, repair, and disease by osteoclasts, chondroclasts, and septoclasts. Connect Tissue Res. (2016) 57:161–74. doi: 10.3109/03008207.2016.1140752
215. Larrouture QC, Cribbs AP, Rao SR, Philpott M, Snelling SJ, and Knowles HJ. Loss of mutual protection between human osteoclasts and chondrocytes in damaged joints initiates osteoclast-mediated cartilage degradation by MMPs. Sci Rep. (2021) 11:22708. doi: 10.1038/s41598-021-02246-7
216. Schmelz M, Mantyh P, Malfait AM, Farrar J, Yaksh T, Tive L, et al. Nerve growth factor antibody for the treatment of osteoarthritis pain and chronic low-back pain: mechanism of action in the context of efficacy and safety. Pain. (2019) 160:2210–20. doi: 10.1097/j.pain.0000000000001625
217. Ze Y, Wu Y, Tan Z, Li R, Li R, Gao W, et al. Signaling pathway mechanisms of circadian clock gene Bmal1 regulating bone and cartilage metabolism: a review. Bone Res. (2025) 13:19. doi: 10.1038/s41413-025-00403-6
218. Lim HD, Kim YS, Ko SH, Yoon IJ, Cho SG, Chun YH, et al. Cytoprotective and anti-inflammatory effects of melatonin in hydrogen peroxide-stimulated CHON-001 human chondrocyte cell line and rabbit model of osteoarthritis via the SIRT1 pathway. J pineal Res. (2012) 53:225–37. doi: 10.1111/j.1600-079X.2012.00991.x
219. Pei M, He F, Wei L, and Rawson A. Melatonin enhances cartilage matrix synthesis by porcine articular chondrocytes. J pineal Res. (2009) 46:181–7. doi: 10.1111/j.1600-079X.2008.00646.x
220. Jia S, Yang Y, Bai Y, Wei Y, Zhang H, Tian Y, et al. Mechanical stimulation protects against chondrocyte pyroptosis through irisin-induced suppression of PI3K/akt/NF-κB signal pathway in osteoarthritis. Front Cell Dev Biol. (2022) 10:797855. doi: 10.3389/fcell.2022.797855
221. Wei X, Qiu J, Lai R, Wei T, Lin Z, Huang S, et al. A human organoid drug screen identifies &x3b1;2-adrenergic receptor signaling as a therapeutic target for cartilage regeneration. Cell Stem Cell. (2024) 31:1813–1830.e8. doi: 10.1016/j.stem.2024.09.001
222. Li J, Zhang W, Liu X, Li G, Gu Y, Zhang K, et al. Endothelial Stat3 activation promotes osteoarthritis development. Cell prolifer. (2023) 56:e13518. doi: 10.1111/cpr.v56.12
223. Liang W, Chen Q, Cheng S, Wei R, Li Y, Yao C, et al. Skin chronological aging drives age-related bone loss via secretion of cystatin-A. Nat Aging. (2022) 2:906–22. doi: 10.1038/s43587-022-00285-x
224. Li J, Wang X, Li X, Liu D, Zhai L, Wang X, et al. Mechanical loading promotes the migration of endogenous stem cells and chondrogenic differentiation in a mouse model of osteoarthritis. Calcif Tissue Int. (2023) 112:363–76. doi: 10.1007/s00223-022-01052-1
225. Ren X, Zhuang H, Li B, Jiang F, Zhang Y, and Zhou P. Gsmtx4 Alleviated Osteoarthritis through Piezo1/Calcineurin/NFAT1 Signaling Axis under Excessive Mechanical Strain. Int J Mol Sci. (2023) 24. doi: 10.3390/ijms24044022
226. Tufail E, Sanyal S, Mithal A, Sanyal S, and Chattopadhyay N. The role of glucagon-like peptides in osteosarcopenia. J Endocrinol. (2025) 264. doi: 10.1530/JOE-24-0210
227. Singh RK, Chang HW, Yan D, Lee KM, Ucmak D, Wong K, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. (2017) 15:73. doi: 10.1186/s12967-017-1175-y
228. Chen C, Zhang Y, Yao X, Li S, Wang G, Huang Y, et al. Characterizations of the gut bacteriome, mycobiome, and virome in patients with osteoarthritis. Microbiol Spectr. (2023) 11:e0171122. doi: 10.1128/spectrum.01711-22
229. Kim DS, Song L, Wang J, Wu H, Gu G, Sugi Y, et al. GRP94 is an essential regulator of pancreatic β-cell development, mass, and function in male mice. Endocrinology. (2018) 159:1062–73. doi: 10.1210/en.2017-00685
230. Doenyas C, Clarke G, and Cserjési R. Gut-brain axis and neuropsychiatric health: recent advances. Sci Rep. (2025) 15:3415. doi: 10.1038/s41598-025-86858-3
Keywords: osteoarthritis, organs-crosstalk, cartilage, chondrocyte, therapeutic strategy
Citation: Qin Y, Di J, Guo Z, Chen S and Xiang C (2025) Association of organs-crosstalk with the pathogenesis of osteoarthritis: cartilage as a key player. Front. Endocrinol. 16:1593658. doi: 10.3389/fendo.2025.1593658
Received: 14 March 2025; Accepted: 13 May 2025;
Published: 05 June 2025.
Edited by:
Xuesong Zhu, The First Affiliated Hospital of Soochow University, ChinaCopyright © 2025 Qin, Di, Guo, Chen and Xiang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Chuan Xiang, Y2h1YW54aWFuZ0BzeG11LmVkdS5jbg==
†These authors have contributed equally to this work and share first authorship