Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Immunol., 31 October 2025

Sec. Cytokines and Soluble Mediators in Immunity

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1657285

This article is part of the Research TopicAdvancing Musculoskeletal Health: Bridging Basic and Clinical Research on Biomechanical Properties of Joints, Ligaments, Tendons, and Associated StructuresView all 13 articles

Mechanistic insights into IL-1β-mediated progression of tendinopathy

Updated
Yuchi Zhang&#x;Yuchi Zhang1†Jiayue Wang&#x;Jiayue Wang1†Fanbo TangFanbo Tang1Runzhi XianRunzhi Xian1Huanhuan ZhangHuanhuan Zhang1Yanlin YuanYanlin Yuan1Guoqiang Yang*Guoqiang Yang2*Guiquan Chen*Guiquan Chen2*
  • 1School of Physical Education, Southwest Medical University, Luzhou, China
  • 2Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China

Tendinopathy, a chronic degenerative musculoskeletal disorder characterized by pain, edema, and functional impairment, exhibits increased prevalence among elderly populations and athletes. Despite extensive research efforts targeting the attenuation of this degenerative process, clinical outcomes frequently remain suboptimal. Recent evidence underscores the critical need for more precisely targeted modulation of inflammatory pathways to improve therapeutic efficacy. Notably, the proinflammatory cytokine interleukin-1β (IL-1β) has been implicated as a prominent mediator in the pathogenesis of tendinopathy. This review synthesizes current understanding of IL-1β synthesis and its downstream signaling transduction pathways, with the primary objective of elucidating the mechanisms by which IL-1β contributes to tendinopathy progression. Through this approach, we seek to reveal novel therapeutic targets and inform improved management strategies. Although IL-1β represents a promising therapeutic candidate for tendinopathy, as evidenced by numerous investigations, current understanding of its pathogenic role is limited by several factors, including the heterogeneity of experimental models, a lack of translational studies, and insufficient evidence linking IL-1β signaling to specific clinical manifestations. Consequently, further research is essential to delineate the precise mechanisms of IL-1β involvement in tendinopathy.

1 Introduction

Tendinopathy is a chronic degenerative disorder characterized by debilitating pain, functional impairment, and localized edema, exhibiting heightened prevalence among elderly individuals and athletes (1). Current therapeutic strategies for tendinopathy encompass conservative and surgical interventions (2, 3). Conservative management, primarily involving rest, physical therapy, and extracorporeal shockwave therapy, remains the first-line clinical recommendation owing to its non-invasiveness (4, 5). However, these approaches exhibit notable limitations: they act primarily as symptomatic interventions—alleviating pain temporarily but failing to reverse the underlying pathological changes of tendinopathy (6). Additionally, their long-term efficacy is further compromised in chronic or severe cases, where they cannot prevent disease progression to tendon rupture; in some instances, prolonged rest may even lead to tendon atrophy, exacerbating functional impairment (7). Surgical intervention is typically reserved for cases unresponsive to conservative measures, yet it carries inherent risks (e.g., infection, adhesion formation, and prolonged post-operative recovery) and does not guarantee satisfactory long-term functional outcomes, particularly in elderly or active patients (8). Given these limitations, the development of more effective, mechanism-driven therapeutic agents has long been hampered by an incomplete understanding of tendinopathy pathogenesis. Consequently, elucidating the key molecular mediators and signaling pathways underlying tendinopathy progression is essential to overcome the shortcomings of current treatments and establish evidence-based management strategies.

Advances in tendon biology have now firmly established inflammation as a core pathogenic driver of tendinopathy, rather than a secondary byproduct (9). Specifically, dysregulated activation of inflammatory signaling molecules is implicated at every stage of the disease: from the initial phase of tenocyte dysfunction and extracellular matrix (ECM) disorganization, to progressive ECM degradation, and ultimately to the increased risk of tendon rupture (10). This recognition has spurred the exploration of therapeutic strategies targeting inflammation; however, interventions designed to achieve complete inflammatory blockade have yielded inconsistent and often disappointing clinical results. A key reason for this failure is the growing consensus that controlled, transient inflammation is not merely non-harmful but functionally essential for tendinopathy repair (11). Against this backdrop, recent evidence has shifted the therapeutic paradigm: instead of broad, non-specific suppression of inflammation, precisely targeted modulation of inflammatory responses has emerged as a more scientifically sound and clinically viable approach (12). This strategy aims to selectively abrogate the pathogenic inflammatory cascades that drive tissue destruction, while preserving the reparative inflammatory processes critical for tendon healing—addressing the fundamental limitation of earlier anti-inflammatory interventions and aligning with the need for mechanism-based therapies.

Interleukin-1β (IL-1β), a prototypical proinflammatory cytokine, plays a critical role in the pathogenesis of tendinopathy (13, 14). Accumulating evidence from cellular and animal studies underscore the therapeutic promise of IL-1β inhibition in its treatment (15, 16). However, a comprehensive understanding of the mechanisms through which IL-1β contributes to tendinopathy remains incomplete, hindering the translation of these findings into clinical applications. To address this challenge, rodent models of induced tendinopathy and in vitro tendon culture systems have become indispensable preclinical tools for advancing therapeutic strategies. This review begins by consolidating current knowledge on fundamental characteristics of IL-1β, including its production and activation. It then examines recent advances in elucidating the pathogenetic roles and underlying mechanisms of IL-1β in tendinopathy. By integrating these insights, this review seeks to deepen the mechanistic understanding of tendinopathy and facilitate the development of targeted therapies, thereby reinforcing the rationale for IL-1β as a therapeutic target. Further investigation is nevertheless essential to fully delineate the precise actions of IL-1β in tendinopathy.

1.1 Search strategy

1) Search site: Articles are forming PubMed, a database of papers on biomedical science. 2) Database: MEDLINE. 3) Keywords: IL-1β, tendinopathy, tendon disorders, inflammation, mechanisms. 4) Boolean algorithm: (“IL-1β”) OR (“Tendinopathy” OR “Tendon disorders”). 5) Retrieval timeframe: the articles we mainly searched for were published between 2015 and 2025. When referring to classic literature to describe the basic mechanisms, the publication time of these studies was before 2015, but did not exceed 20% of the publication time of the cited literature.

1.1.1 Inclusion and exclusion criteria

Articles were included if the topic is related to IL-1β or tendinopathy, and the article type was a review or experimental paper. The search process was performed as presented in Figure 1.

Figure 1
Flowchart showing the identification of studies via databases. Initially, 397 records were identified. After removing 143 duplicates, 254 remained. An additional 56 were removed due to unrelated titles and abstracts, leaving 198 full articles. Seventy-five full-text articles were excluded for not being about IL-1β or tendinopathy. Finally, 123 articles were selected for review.

Figure 1. Article retrieval flow chart with inclusion and exclusion process. This flowchart illustrates the systematic procedure for identifying and selecting relevant studies. The initial database search yielded 397 records. After the removal of 143 duplicates, 254 records underwent title and abstract screening, which led to the exclusion of 56 unrelated studies. The remaining 198 full-text articles were assessed for eligibility, of which 90 were excluded for not focusing on IL-1β or tendinopathy. Ultimately, a total of 108 studies were deemed suitable and included in the review.

2 IL-1β processing and production

IL-1β is predominantly synthesized by macrophages and dendritic cells (17). However, it can also be secreted by tenocytes, tendon stem/progenitor cells (TSPCs), and osteoblasts (18). Initially, IL-1β is translated as an inactive precursor protein, pro-IL-1β, which requires proteolytic cleavage to generate the bioactive mature cytokines (19). Consequently, the production of IL-1β in most cell types follows a tightly regulated two-step process: (1) a priming step that induces the expression of IL-1β precursor and inflammasome components, and (2) an activation step that triggers NLRP3 inflammasome assembly, caspase-1 activation, and subsequent IL-1β maturation (Figure 2) (20). The priming cascade is initiated when pattern recognition receptors (PRRs)—most commonly Toll-like receptors (TLRs, e.g., TLR4) on the cell surface or endosomes—bind to damage-associated molecular patterns (DAMPs, such as fragmented extracellular matrix or mitochondrial DNA released by injured tendon cells) (21). Upon DAMP binding, TLRs recruit intracellular adaptor proteins (e.g., MyD88), which activate downstream signaling cascades, predominantly the NF-κB pathway. Specifically, this activation leads to phosphorylation and degradation of the NF-κB inhibitor (IκB), freeing NF-κB to translocate into the nucleus (22). Once in the nucleus, NF-κB drives the transcriptional upregulation of two key targets: (a) the inactive precursor form of IL-1β (pro-IL-1β) and (b) components of the NOD-like receptor protein 3 (NLRP3) inflammasome, including NLRP3 itself and the adaptor protein ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain (23). This priming step is essential, as it ensures the cell accumulates sufficient pro-IL-1β and inflammasome components to respond to subsequent activation signals. Subsequently, a diverse range of stimuli—including reactive oxygen species (ROS) and extracellular adenosine triphosphate (ATP)—act through but complementary mechanisms to activate the NLRP3 inflammasome complex (24). Notably, ROS, generated by mitochondrial dysfunction or NADPH oxidase activation in response to tissue injury, induces oxidative stress that damages mitochondrial membranes. This damage releases mitochondrial DAMPs into the cytoplasm, which then directly interact with NLRP3, further stabilizing its oligomerization and enhancing inflammasome activation (25). On the other hand, extracellular ATP binds to the P2X7 receptor (a cation channel) on the cell membrane. This binding induces rapid opening of the P2X7 receptor, leading to massive efflux of intracellular potassium ions (K+). A decrease in cytosolic K+ concentration is a critical “danger signal” that directly promotes the oligomerization of NLRP3 proteins (a key step in inflammasome assembly) (26). Following NLRP3 oligomerization, the adaptor protein ASC is recruited via homotypic interactions (NLRP3’s pyrin domain binds ASC’s pyrin domain). ASC then acts as a scaffold to recruit pro-caspase-1, forming a large multiprotein complex known as the “NLRP3 inflammasome (27).” The assembly of this complex induces conformational changes in pro-caspase-1, driving its autocatalytic cleavage into the enzymatically active form, caspase-1 (28). Once activated, caspase-1 catalyzes the proteolytic maturation of IL-1β by cleaving its inactive precursor, pro-IL-1β, at specific aspartate residues. This cleavage generates the mature, biologically active form of IL-1β (29). Notably, since mature IL-1β lacks a conventional signal peptide, it cannot be released via the classical endoplasmic reticulum–Golgi secretory pathway. Instead, its export depends on a non-classical secretion mechanism mediated by Gasdermin D (GSDMD), which is itself a substrate of caspase-1 (30). Upon cleavage by caspase-1, the N-terminal fragment of GSDMD is liberated and subsequently oligomerizes to form pores in the plasma membrane (31). These pores serve as conduits for the release of mature IL-1β into the extracellular milieu (32). Once outside the cell, IL-1β can bind to its cognate receptors on neighboring cells, initiating and amplifying pro-inflammatory signaling cascades that contribute to the pathogenesis of tendinopathy and other inflammatory conditions (33).

Figure 2
Diagram illustrating the NLRP3 inflammasome signaling pathway. It shows two stages: priming and activation. Priming begins with TLR activation, leading to NF-kB-dependent transcription of pro-IL-1β and NLRP3 in the nucleus. During activation, potassium efflux, ATP, calcium, and ROS trigger NLRP3 inflammasome assembly. This activates Caspase-1, converting pro-IL-1β to IL-1β.

Figure 2. This schematic illustrates the canonical two-step mechanism underlying the maturation and release of the pro-inflammatory cytokine IL-1β. Signal 1 (Priming): Pathogen- or damage-associated molecular patterns (PAMPs/DAMPs) engage pattern-recognition receptors (e.g., Toll-like Receptors, TLRs), initiating a signaling cascade that activates the transcription factor NF-κB. This leads to the transcriptional upregulation of both pro-IL-1β and the NLRP3 protein, preparing the necessary components for inflammasome assembly. Signal 2 (Activation): Diverse stimuli, including extracellular ATP (leading to P2X7 receptor activation and K+ efflux) and crystalline structures, trigger the assembly of the NLRP3 inflammasome. This multi-protein complex recruits the adapter protein ASC (Apoptosis-associated speck-like protein containing a CARD), which then recruits and activates pro-caspase-1. Activated caspase-1 cleaves the inactive precursor pro-IL-1β into its biologically active form, IL-1β, which is subsequently released from the cell to drive inflammatory responses.

3 IL-1β signal transduction

Upon release, IL-1β activity is tightly regulated through its receptors. IL-1 receptor type I (IL-1RI), expressed ubiquitously on nucleated cells, is essential for initiating signal transduction (34). IL-1β also binds to a second receptor, IL-1 receptor type II (IL-1RII), which functions as a decoy receptor by competing with IL-1RI for ligand binding, thereby negatively regulating IL-1βsignaling. Furthermore, the endogenous IL-1 receptor antagonist (IL-1Ra) binds to IL-1RI with high affinity, preventing downstream signaling activation (35). Ligand binding to IL-1RI initiates the formation of a binary IL-1β:IL-1RI complex. This complex subsequently recruits the interleukin-1 receptor accessory protein (IL-1RAcP), forming the ternary signaling complex IL-1β:IL-1RI: IL-1RAcP (36). The Toll/interleukin-1 receptor (TIR) domains within this complex then engage the TIR domain of the adaptor protein myeloid differentiation primary response 88 (MyD88). MyD88 recruits interleukin-1 receptor-associated kinases (IRAKs), specifically IRAK4, IRAK1, and IRAK2. IRAK4 autophosphorylates and subsequently phosphorylates IRAK1 and IRAK2, enabling their association with tumor necrosis factor receptor-associated factor 6 (TRAF6) (37). TRAF6 serves as an E3 ubiquitin ligase that recruits and activates TGF-β-activated kinase 1 (TAK1). Activated TAK1 phosphorylates components of the IκB kinase (IKK) complex and mitogen-activated protein kinase (MAPK) cascades (38). This leads to the activation of key transcription factors, including: Nuclear factor kappa B (NF-κB), Activator protein-1 (AP-1), and Members of the MAPK family: p38 MAPK, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) (39). The activation of these transcription factors induces the expression of target genes involved in diverse cellular responses, ultimately contributing to the pathogenesis and progression of various diseases in a cell-type-specific manner (Figure 3).

Figure 3
Diagram of IL-1 signaling pathways. IL-1 binds to IL-1RI, activating MyD88 and IRAK4, leading to phosphorylation of IRAK1 and IRAK2. TRAF6 and TAK-1 are engaged, activating MAPK and NF-kB, impacting the nucleus and gene expression. IL-1Ra and IL-1RII do not signal.

Figure 3. Schematic presentation of IL-1β signal transduction. Left: The IL-1 receptor antagonist (IL-1Ra) binds to the IL-1 type I receptor (IL-1RI), which fails to initiate signal transduction; Middle: IL-1β binding to IL-1RI triggers a functional signaling pathway. This recruits myeloid differentiation primary response protein 88 (MyD88), followed by phosphorylation (“P”) and activation of interleukin-1 receptor-associated kinase 4 (IRAK4). Subsequent phosphorylation of IRAK1, IRAK2, and tumor necrosis factor receptor-associated factor 6 (TRAF6) occurs, with activated TRAF6 inducing the activation of transforming growth factor-β-activated kinase 1 (TAK-1). TAK-1 then activates downstream cascades, including the mitogen-activated protein kinase (MAPK) pathway and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. These pathways translocate to the nucleus to regulate gene transcription, such as the induction of activator protein 1 (AP-1); Right: IL-1β binding to the IL-1 type II receptor (IL-1RII) does not transduce a signal.

4 Main roles of IL-1β in tendinopathy

The pathophysiology of tendinopathy remains incompletely understood. Proposed pathogenic mechanisms include aberrant mechanical loading, sustained inflammatory responses, imbalance between matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs), aberrant differentiation of tendon stem/progenitor cells (TSPCs), dysregulated apoptosis and cellular senescence, as well as disruption of collagen fiber architecture characterized by diminished type I collagen synthesis and disorganized deposition of type III collagen (40, 41). Accumulating evidence implicates IL-1β as a significant mediator in these pathological processes (Figure 4), as elaborated below.

Figure 4
Diagram illustrating the impact of IL-1β on tendon structure and cellular responses. Collagen molecules, fibrils, fascicles, and tendons are labeled. IL-1β influences tenocyte proinflammatory phenotypes, inflammatory factors, M1 macrophages, and T cells, leading to persistent inflammation. It causes ECM degradation by increasing MMPs and reducing TIMPs. It reduces tenogenic and increases osteogenic differentiation, affecting TSPCs. NF-κB and PI3K/Akt pathways are activated; SIRT1 is reduced. Caspase-3 and Bax are elevated, Bcl-2 is decreased, resulting in increased cell apoptosis and senescence.

Figure 4. The roles and mechanisms of IL-1β in the progression of tendinopathy. Exposure to IL-1β disrupts tendon homeostasis by triggering a pro-inflammatory microenvironment and key pathological processes. This leads to an imbalance in extracellular matrix (ECM) remodeling, a shift in tendon stem/progenitor cell (TSPC) differentiation away from tenogenesis, and increased cellular apoptosis and senescence. Collectively, these mechanisms drive the progression of tendinopathy. (↑: increase; ↓: decrease).

4.1 IL-1β amplifies the inflammatory responses

Persistent inflammation is now recognized as a fundamental driver of tendinopathy pathogenesis, orchestrated by dynamic crosstalk between three interconnected cellular compartments: the stromal (tenocytes, tendon stem/progenitor cells), immune-sensing (resident macrophages, mast cells), and infiltrating (recruited immune cells) compartments. Rather than a transient response, inflammation in tendinopathy reflects a dysregulated, self-sustaining network that disrupts tendon homeostasis (42). Within this network, the cytokine IL-1β emerges as a critical signaling node, mediating a feed-forward inflammatory cascade that engages all three compartments and propagates chronicity (43).

The inflammatory cascade in tendinopathy is frequently initiated within the immune-sensing compartment, where resident mast cells and macrophages recognize microdamage or other pathological stimuli via pattern recognition receptors. Upon activation, these cells release IL-1β, which subsequently acts on both infiltrating immune cells and local stromal components. A central pathogenic mechanism is the capacity of IL-1β to polarize macrophages toward a pro-inflammatory M1 phenotype. These M1 macrophages further amplify IL-1β production and recruit additional immune cells, establishing a self-sustaining inflammatory loop that contributes to chronic tendon degeneration (44). However, the influence of IL-1β on M2 macrophages—a phenotype generally associated with anti-inflammatory and reparative functions—is less clearly defined. Whether IL-1β suppresses M2 differentiation, alters the functional properties of existing M2 macrophages, or disrupts the balance between M1 and M2 populations represents a significant knowledge gap. Elucidating these mechanisms will be essential for understanding the dysregulated repair processes in tendinopathy. Future studies should aim to clarify the temporal and contextual effects of IL-1β on macrophage. Most critically, IL-1β directly reprograms the stromal compartment, driving tenocytes into an activated, pro-inflammatory state (4547). This phenotypic shift is not merely a passive response but represents a fundamental change in tenocyte identity and function. Evidence from human studies indicates that tenocytes from tendinopathic tissues exhibit a sustained activated phenotype, characterized by elevated expression of markers like podoplanin (PDPN) (45). Notably, this inflammatory memory can persist long-term, as tenocytes isolated from patients’ years after clinical intervention display heightened sensitivity to IL-1β challenge, secreting elevated levels of IL-6 and IL-8. This suggests that IL-1β exposure can epigenetically imprint a hyper-responsive state on tenocytes, a potential mechanism for disease chronicity and recurrence (45). At the molecular level, the sustained inflammatory phenotype is underpinned by constitutive activation of the NF-κB pathway (48). IL-1β signaling robustly activates NF-κB, which not only drives the immediate expression of classic inflammatory mediators (IL-6, IL-8, COX-2, PGE-2) but also appears to lower the threshold for subsequent activation, thereby sensitizing tenocytes (49). Inhibition of NF-κB potently reduces IL-1β-induced cytokine production, confirming its central role (50). Furthermore, IL-1β orchestrates a multi-faceted attack on tendon homeostasis by upregulating prostaglandin E synthase (mPGES) and specifically enhancing the expression of the EP4 receptor for PGE2, creating an autocrine loop that may amplify inflammatory and catabolic signals (5052). The cytokine also stimulates the release of nociceptive mediators like Substance P, directly linking the inflammatory process to pain (53). The cycle of chronicity is further reinforced by IL-1β’s impact on immune cell recruitment (54). By inducing tenocytes to produce chemokines such as CCL20 and CCL5, IL-1β facilitates the recruitment and activation of T cells. These activated T cells reciprocally produce TNF-α and IL-1β, which further activate tenocytes, establishing a vicious paracrine loop that is difficult to resolve (55). This T cell-tenocyte crosstalk not only perpetuates inflammation but also directly contributes to pathological matrix remodeling, as evidenced by an increased collagen III/I ratio (56).

Collectively, IL-1β is a critical node connecting the three cellular compartments and driving chronic inflammation in tendinopathy. It sustains M1 macrophage polarization through a positive feedback loop, induces and maintains the activated inflammatory phenotype of tenocytes, and promotes T cell-mediated immune responses. Future therapeutic strategies could focus on targeting this cytokine network, particularly disrupting the chronic inflammatory state maintained by the IL-1β/NF-κB axis, potentially offering new avenues for the fundamental treatment of tendinopathy (Figure 5).

Figure 5
Diagram illustrating the interaction between infiltrating and stromal immune compartments. On the left, infiltrating compartment cells, including pro-resolving and proinflammatory macrophages, mast cells, and T cells. Collagen fibers and tenocytes are centrally depicted. The stromal compartment involves acquiring an inflammatory phenotype and producing mediators. The immune-sensing compartment responds to insults with mediators and chemokines. IL-1β is shown as a mediator between compartments.

Figure 5. The role of IL-1β in chronic inflammation during degenerative tendinopathy. In tendinopathy, IL-1β—whether derived from stromal tenocytes or immune cells of the immune-sensing or infiltrating compartments—sustains chronic inflammation by promoting macrophage polarization toward an M1 phenotype. This process, in turn, drives stromal tenocytes toward a proinflammatory phenotype, enhances their capacity to secrete proinflammatory mediators, and stimulates chemokine production, thereby facilitating T cell recruitment and activation.

4.2 IL-1β induces ECM degradation

The progressive degradation of the extracellular matrix (ECM) is a hallmark of degenerative tendinopathy, with matrix metalloproteinases (MMPs) serving as central executors of ECM catabolism (57). A substantial body of evidence indicates that the pro-inflammatory cytokine IL-1β acts as a primary upstream regulator of MMP expression in tenocytes, thereby disrupting ECM homeostasis. Rather than merely cataloging individual findings, this section synthesizes the mechanistic pathways through which IL-1β drives pathological matrix remodeling and examines its critical interplay with mechanical stimuli—a relationship that underpins a self-perpetuating cycle of tendon degeneration.

IL-1β exerts its catabolic effects by shifting the delicate balance between MMPs and their endogenous inhibitors, the tissue inhibitors of metalloproteinases (TIMPs) (58). In healthy tendons, TIMP levels predominate, ensuring controlled turnover. However, upon IL-1β stimulation, tenocytes from multiple species exhibit a coordinated upregulation of key collagenases and stromelysins—notably MMP-1, MMP-3, and MMP-13—without a commensurate increase in TIMP-1 or TIMP-2 (59, 60). This selective induction creates a proteolytic imbalance favoring net matrix degradation. Mechanistically, IL-1β signaling converges on several key pathways: binding to the IL-1 receptor activates the JNK/SAPK and broader MAPK cascades (ERK1/2, p38, JNK), which are requisite for maximal MMP transcription (61, 62). The pathological relevance of this axis is further underscored by the ability of IL-1Ra to attenuate collagen breakdown, albeit at a higher concentration due to competitive receptor binding (63). A particularly insightful concept emerging from the literature is the positive feedback loop initiated by IL-1β. Beyond directly inducing MMPs, IL-1β can stimulate tenocytes to produce more endogenous IL-1β, thereby amplifying and prolonging the catabolic signal (9). This autocrine/paracrine loop may explain the progressive nature of tendinopathy even after the initial insult has subsided. Furthermore, the relative potency of IL-1β compared to other cytokines like TNF-α suggests a specialized role: while TNF-α may initiate degradation, IL-1β appears more critical for driving its progression, as evidenced by its superior capacity to upregulate MMP-3 and MMP-13 in ex vivo models (64).

Perhaps the most significant advance in understanding IL-1β’s role in tendinopathy lies in its integration with the condition’s biomechanical etiology (65). Mechanical overload, a primary risk factor, is no longer viewed as merely causing structural fatigue, but as a trigger for a complex biologic response (66). In this context, tenocytes function as key mechanosensory units that transduce mechanical stimuli into molecular signals, modulating ECM synthesis and degradation to establish novel homeostatic setpoints adapted to their mechanical environment (67). This mechanoresponsive capacity is governed by the cytoskeletal architecture and its dynamic interactions with the ECM (68), where α-smooth muscle actin (αSMA)-mediated organization generates intrinsic cellular tension (69). This tension propagates to the ECM as contractile traction forces, establishing a biomechanical equilibrium within the tissue microenvironment (70). However, pathological overload disrupts this equilibrium: microdamage from overloading relieves mechanical tension on tenocytes, effectively creating a state of “unloading” at the cellular level (71). This aberrant mechanical environment not only stimulates IL-1β production—potentially via mechanisms such as H2O2-induced NLRP3 inflammasome activation (72, 73)—but also critically determines cellular responsiveness to the cytokine. It has been reported that tenocytes with diminished intrinsic tension exhibit exaggerated catabolic responses to IL-1β, including markedly increased MMP expression and apoptosis, ultimately leading to ECM destruction (74). Conversely, tenocytes maintaining high cellular tension demonstrate markedly attenuated responses to IL-1β-induced catabolic events (75), establishing cellular tension state as a pivotal regulator of IL-1β-driven pathology.

Emerging evidence further suggests that dysregulation of mechanosensitive signaling pathways, such as the Yes-associated protein (YAP) pathway, may link abnormal mechanical loading to inflammatory and catabolic responses in tenocytes. Under physiological loading, YAP shuttles between the cytoplasm and nucleus to regulate cell proliferation and matrix homeostasis; however, excessive mechanical stress can lead to aberrant YAP activation, which has been associated with pro-inflammatory gene expression and matrix remodeling (76). Although direct crosstalk between YAP and IL-1β in tendinopathy remains underexplored, studies in other musculoskeletal tissues suggest that YAP signaling can modulate IL-1β production and subsequent MMP activation (77). The resulting cytokine surge promotes MMP secretion, degrading the pericellular matrix and further compromising tissue integrity. Thereby, a self-perpetuating vicious cycle is established: mechanical damage induces IL-1β expression and reduces cellular tension, which synergistically promotes ECM breakdown; this degradation, in turn, weakens the tendon structure and increases its susceptibility to additional mechanical injury. Notably, both excessive strain and stress-shielding upregulate IL-1β and MMPs, thereby driving degenerative processes. The magnitude of mechanical stretch plays a decisive role in determining cellular responses: while low-magnitude strain may exert protective or anti-inflammatory effects, high-magnitude strain acts synergistically with IL-1β to markedly upregulate MMP-1 and MMP-3 expression (78). This synergy underscores that mechanical stress and inflammatory signaling are integrated at the molecular level, collectively governing the net catabolic outcome in tendon tissue. Importantly, pharmacological restoration of cytoskeletal tension (e.g., via calyculin A administration (79)) has been shown to reverse IL-1β-driven catabolism by reestablishing physiological tension homeostasis, highlighting targeted modulation of tenocyte mechanobiology as a promising therapeutic strategy for counteracting inflammatory degradation in tendinopathy.

Collectively, these studies firmly establish IL-1β as a master regulator of ECM degradation in tendinopathy, primarily by disrupting the critical balance between MMPs and TIMPs. The interplay between IL-1β and mechanical loading is particularly noteworthy, suggesting a vicious cycle where microdamage from overloading induces IL-1β expression, which in turn promotes further ECM cleavage, weakening the tendon and making it more susceptible to additional damage. Critically, the cellular response to IL-1β is governed by the mechanobiological state of tenocytes: loss of intrinsic cytoskeletal tension following microdamage hypersensitizes cells to IL-1β, exacerbating matrix destruction. Future therapeutic strategies should extend beyond anti-inflammation to target this mechanobiological dysregulation. Elucidating the crosstalk between pathways like YAP and IL-1β signaling, and developing interventions to “re-tension” tenocytes, hold promise for breaking the degenerative cycle and promoting tendon regeneration.

4.3 IL-1β inhibits tenogenic differentiation of tendon stem/progenitor cell

Beyond the well-established role of tenocytes in tendon homeostasis, the identification of tendon stem/progenitor cells (TSPCs) has fundamentally expanded our understanding of tendon biology and pathology (80). TSPCs, distinct from mature tenocytes, possess the critical capacities for self-renewal and differentiation, serving as a reservoir for tissue maintenance and repair (81). In healthy tendon healing, TSPCs undergo tenogenic differentiation to regenerate functional tissue (82). However, in the pathological microenvironment of tendinopathy, TSPCs are driven toward aberrant differentiation lineages—namely chondrogenic, osteogenic, and adipogenic—leading to the characteristic degenerative features of lipid deposition, proteoglycan accumulation, and ectopic calcification (83). A key insight emerging from recent research is that the cytokine IL-1β acts as a potent pathological switch that redirects TSPC fate away from regeneration and toward degeneration (84).

It has been reported that IL-1β consistently suppresses the expression of key tenogenic markers such as scleraxis (Scx), tenomodulin (Tnmd), and type I collagen, while simultaneously promoting markers of non-tenogenic lineages. What makes this effect particularly consequential for chronic tendinopathy is its potential persistence. Studies indicate that even transient exposure to IL-1β can cause a lasting impairment of TSPCs’ tenogenic potential, suggesting that an early inflammatory insult may irreversibly compromise the tendon’s intrinsic repair capacity long after the initial cytokine signal has subsided (85). This provides a plausible cellular mechanism for the progressive and often irreversible nature of advanced tendinopathy. The mechanistic underpinnings of this fate switch involve multiple signaling pathways. IL-1β has been shown to act through the downregulation of microRNAs such as miR-337-3p, leading to the activation of Nox4-JNK and IRS1-ERK cascades that favor chondro-osteogenic differentiation (86). Other work implicates the Rac1 signaling pathway in IL-1β-mediated induction of osteogenic markers (SOX9, COL2α1) and suppression of tenogenic factors (87). Furthermore, the source of IL-1β is also of pathological relevance. Recent evidence links pyroptotic macrophages—a highly inflammatory form of cell death—to IL-1β release, which in turn drives TSPCs toward osteogenic differentiation, creating a direct bridge between immune cell activity and degenerative tissue remodeling (88).

In summary, the impact of IL-1β on TSPCs represents a critical paradigm shift in tendinopathy pathogenesis. It moves the focus beyond the catabolic degradation of the existing matrix by tenocytes to include the failure of regenerative potential through the maldifferentiation of the progenitor cell pool. This dual attack—simultaneously breaking down mature tissue and corrupting the cells meant to rebuild it—explains the progressive and structurally disastrous nature of the disease. Therefore, therapeutic strategies that not only block IL-1β’s catabolic effects but also safeguard or restore the tenogenic differentiation capacity of TSPCs could be pivotal in achieving true tendon regeneration rather than mere symptom mitigation. Future research should prioritize elucidating the epigenetic mechanisms behind IL-1β’s long-lasting effects on TSPCs and identifying strategies to therapeutically reverse this pathological reprogramming.

4.4 IL-1β accelerates cell apoptosis

Beyond its well-characterized roles in promoting inflammation and matrix degradation, IL-1β contributes to tendinopathy pathogenesis by accelerating tenocyte death through the induction of apoptotic pathways (89). While physiological apoptosis is essential for tissue turnover, its dysregulation represents a critical mechanism of cellular depletion and functional decline in degenerative tendons (90). A growing body of evidence positions IL-1β as a master regulator of this pathological apoptosis, primarily through its orchestration of the NF-κB and PI3K/Akt signaling axes.

The pro-apoptotic effect of IL-1β is executed via the classic mitochondrial pathway, culminating in the activation of caspase-3, the key effector protease of apoptosis. IL-1β signaling shifts the delicate balance between pro-apoptotic and anti-apoptotic proteins, consistently upregulating Bax while suppressing Bcl-2 (91). This imbalance promotes mitochondrial membrane permeabilization, triggering the caspase cascade and committing the cell to death (91). What elevates this finding from a simple observation to a mechanistically insightful one is the identification of the specific signaling pathways involved and their regulatory nodes. Research demonstrates that IL-1β concurrently activates both the NF-κB and PI3K/Akt pathways to drive apoptosis. Notably, the caspase-3-dependent apoptotic process appears specifically tied to NF-κB activation (92). A crucial upstream regulator of this pathway is Sirtuin 1 (SIRT1), a NAD+-dependent deacetylase associated with cellular stress responses (93). IL-1β downregulates SIRT1, which in turn unleashes NF-κB activity, leading to the pro-apoptotic Bax/Bcl-2 dysregulation (93). This mechanistic hierarchy is substantiated by interventional studies: compounds like chitosan and resveratrol can attenuate IL-1β-induced apoptosis by activating SIRT1, thereby suppressing NF-κB signaling and restoring cell survival (92, 94).

In summary, the induction of apoptosis solidifies IL-1β’s role as a central mediator of tendon degeneration. Its ability to activate specific, interconnected pathways like NF-κB and PI3K/Akt, finely tuned by regulators like SIRT1, provides a sophisticated mechanism for excessive cell death. Viewing IL-1β through the lens of apoptosis unveils new therapeutic opportunities aimed at preserving the tenocyte population by targeting these regulatory nodes, potentially slowing disease progression by maintaining the cellular workforce essential for tendon homeostasis.

4.5 IL-1β stimulates cell senescence

Of note, cellular senescence has emerged as a pivotal mechanism in tendinopathy, operating in concert with apoptosis to drive tissue dysfunction (95). Senescent cells, characterized by irreversible growth arrest and a distinct secretory phenotype, accumulate in degenerative tendons and contribute to an impaired regenerative environment (96). Key biomarkers of this state include elevated expression of cyclin-dependent kinase inhibitors p16 and p21, tumor suppressor p53, and increased senescence-associated β-galactosidase (SA-β-gal) activity (97). A compelling body of evidence now identifies IL-1β as a potent inducer of this deleterious state in both tenocytes and tendon stem/progenitor cells (TSPCs).

The mechanistic link between IL-1β and senescence is robustly demonstrated across multiple studies. Stimulation with IL-1β reliably triggers a canonical senescent phenotype in tenocytes, manifesting as altered morphology, growth arrest, and upregulated expression of p16, p21, and p53, alongside enhanced SA-β-gal activity (98). A key insight is the central role of the NF-κB signaling pathway in mediating this response. The pathological relevance of this axis is confirmed by interventional data: inhibition of NF-κB signaling effectively attenuates IL-1β-induced senescence markers in TSPCs. Furthermore, the microRNA miR-146a, a known negative regulator of inflammation, has been shown to confer protection against IL-1β-driven senescence by suppressing the upstream IRAK4/TRAF6/NF-κB cascade (99). This not only reinforces the role of NF-κB but also suggests the existence of endogenous regulatory mechanisms that become overwhelmed in the diseased state.

In summary, IL-1β’s capacity to induce cellular senescence solidifies its position as a master regulator of tendon degeneration, acting through a well-defined NF-κB-dependent pathway. This mechanism contributes to the failure of tissue homeostasis not merely by reducing cell numbers, as in apoptosis, but by creating a population of dysfunctional, senescent cells that actively degrade the tissue milieu. Therapeutic strategies aimed at selectively eliminating senescent cells (senolytics) or modulating their secretory phenotype (senomorphics) may therefore hold promise for disrupting this vicious cycle in IL-1β-driven tendinopathy.

5 Conclusions and future perspective

In summary, the collective evidence establishes IL-1β as a pivotal mediator in tendinopathy pathogenesis. The data synthesized herein demonstrate that IL-1β contributes to disease progression through multiple mechanisms: potentiating inflammatory responses, mediating extracellular matrix (ECM) degradation, suppressing tenogenic differentiation of tendon stem/progenitor cells (TSPCs), accelerating cellular apoptosis, and promoting senescence. While the reviewed studies provide valuable insights into IL-1β’s pathogenic mechanisms and identify promising therapeutic targets, significant knowledge gaps persist.

A key challenge in interpreting current research on IL-1β mechanisms in tendinopathy lies in the widespread use of heterogeneous tenocyte populations lacking subtype-specific markers. Although in vitro models offer valuable molecular insights that may inform clinical prevention strategies, the physiological relevance of findings can be limited when tenocytes are isolated using conventional methods, typically enzymatic digestion of whole tendon tissue, which captures a mixed population of cells with potentially distinct roles (100). Without markers to distinguish tenocyte subtypes (e.g., resident tendon stem/progenitor cells, mature tenocytes, or fibrotic precursors), it remains difficult to attribute IL-1β responses to specific subpopulations (101). This heterogeneity may obscure important functional differences in inflammatory sensitivity, matrix turnover, and mechanoresponsiveness, thereby complicating the extrapolation of in vitro results to in vivo pathology. Furthermore, many studies focus predominantly on transcriptional-level alterations induced by IL-1β, while comprehensive analyses of functional protein dynamics, including post-translational modifications, protein activity, and turnover, are still limited. This is particularly relevant in the context of IL-1β-driven tendinopathy, as IL-1β not only regulates gene expression but also influences the activation and stability of key effector proteins such as matrix MMPs and inflammatory mediators through post-translational mechanisms. For example, MMP activity is often controlled by proteolytic activation and inhibition, processes that are not fully captured by mRNA measurements (102). A greater emphasis on protein-level dynamics will be essential to fully understand the functional impact of IL-1β signaling on tendon degradation and repair. Collectively, future studies should prioritize the identification of tenocyte subtype-specific markers through single-cell transcriptomic or proteomic approaches, enabling more precise cell sorting and functional characterization. In parallel, integrating multi-omics strategies that assess not only transcriptional changes but also protein expression, modification, and metabolic activity will provide a more holistic view of IL-1β’s role in tendinopathy.

Secondly, chronic inflammation in tendinopathy demonstrates intricate interplay with other proinflammatory cytokines, notably TNF-α and IL-6 (103, 104). Consequently, therapeutic strategies targeting a single cytokine may prove inadequate for complete disease resolution, potentially explaining limited clinical efficacy. This premise is substantiated by recent findings wherein concomitant exposure of equine tenocytes to IFN-γ, TNF-α and IL-1β synergistically amplified MMP-1, MMP-3, and MMP-13 expression—an effect unmitigated by IL-1Ra administration (15). This indicates IL-1β inhibition alone is insufficient to counterbalance the catabolic influence of other inflammatory mediators. Furthermore, independent inhibition of TNF-α or IL-6 signaling has demonstrated therapeutic potential in tendinopathy models (105, 106). Given this collective evidence, synergistic inhibition of IL-1β with other pathogenic cytokines represents a promising therapeutic approach warranting systematic investigation.

Finally, while the primary focus of this review has been on the mechanistic role of IL-1β in tendinopathy, the translational prospects of targeting this cytokine warrant discussion. Currently, there are no IL-1β-targeted therapies specifically approved for the treatment of tendinopathy. However, potent biologic agents that neutralize IL-1β, such as the monoclonal antibody Canakinumab, or block its receptor, such as Anakinra (a recombinant IL-1 receptor antagonist), are clinically available and successfully used for other inflammatory conditions, including rheumatoid arthritis and gouty arthritis (107, 108). This established safety profile presents a compelling rationale for investigating their repurposing for tendinopathy. Despite this potential, their application to tendinopathy faces challenges. The translation of IL-1β blockade to tendinopathy is not straightforward, as the pathology often involves a complex interplay of inflammatory, degenerative, and failed healing processes. A key question is identifying the patient subgroup most likely to benefit from anti-IL-1β therapy, presumably those in the earlier, more inflammatory stages of the disease. Furthermore, optimal delivery methods—systemic versus localized, single injection versus sustained-release formulations—need careful evaluation to maximize efficacy and minimize systemic side effects.

Author contributions

YZ: Writing – review & editing, Writing – original draft. JW: Writing – review & editing, Methodology. FT: Validation, Writing – review & editing. RX: Conceptualization, Methodology, Writing – review & editing. HZ: Formal analysis, Investigation, Visualization, Writing – review & editing. YY: Resources, Writing – review & editing. GY: Project administration, Writing – review & editing. GC: Supervision, Investigation, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This article is supported by the following funding: Health Commission of Sichuan Province Medical Science and Technology Program (24QNMP012); Southwest Medical University Technology Program (2024ZXYZX14); Southwest Medical University Technology Program (2024ZKZ016).

Acknowledgments

The authors thank all who contributed their time and expertise to this project, particularly the participants. Furthermore, the authors are really grateful to all those who devote much time to reading this thesis and give me much advice, which will benefit me in my later study.

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 no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

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.

References

1. Traweger A, Scott A, Kjaer M, Wezenbeek E, Scattone Silva R, Kennedy JG, et al. Achilles tendinopathy. Nat Rev Dis Primers. (2025) 11:20. doi: 10.1038/s41572-025-00602-9

PubMed Abstract | Crossref Full Text | Google Scholar

2. Saul H, Cassidy S, Swaithes L, and Pavlova A. Weights, resistance bands, and rest days are best for tendinopathy. BMJ. (2024) 384:q513. doi: 10.1136/bmj.q513

PubMed Abstract | Crossref Full Text | Google Scholar

3. Hall S, Kaplan JRM, Schipper ON, Vulcano E, Johnson AH, Jackson JB, et al. Minimally invasive approaches to Haglund’s deformity and insertional achilles tendinopathy: A contemporary review. Foot Ankle Int. (2024) 45:664–75. doi: 10.1177/10711007241237529

PubMed Abstract | Crossref Full Text | Google Scholar

4. Cooper K, Alexander L, Brandie D, Brown VT, Greig L, Harrison I, et al. Exercise therapy for tendinopathy: a mixed-methods evidence synthesis exploring feasibility, acceptability and effectiveness. Health Technol Assess. (2023) 27:1–389. doi: 10.3310/TFWS2748

PubMed Abstract | Crossref Full Text | Google Scholar

5. Perveen W, Anwar S, Hashmi R, Ali MA, Raza A, Ilyas U, et al. Effects of extracorporeal shockwave therapy versus ultrasonic therapy and deep friction massage in the management of lateral epicondylitis: a randomized clinical trial. Sci Rep. (2024) 14:16535. doi: 10.1038/s41598-024-67313-1

PubMed Abstract | Crossref Full Text | Google Scholar

6. Charles R, Fang L, Zhu R, and Wang J. The effectiveness of shockwave therapy on patellar tendinopathy, Achilles tendinopathy, and plantar fasciitis: a systematic review and meta-analysis. Front Immunol. (2023) 14:1193835. doi: 10.3389/fimmu.2023.1193835

PubMed Abstract | Crossref Full Text | Google Scholar

7. Taik FZ, Karkouri S, Tahiri L, Aachari I, Moulay Berkchi J, Hmamouchi I, et al. Effects of kinesiotaping on disability and pain in patients with rotator cuff tendinopathy: double-blind randomized clinical trial. BMC Musculoskelet Disord. (2022) 23:90. doi: 10.1186/s12891-022-05046-w

PubMed Abstract | Crossref Full Text | Google Scholar

8. Angileri HS, Gohal C, Comeau-Gauthier M, Owen MM, Shanmugaraj A, Terry MA, et al. Chronic calcific tendonitis of the rotator cuff: a systematic review and meta-analysis of randomized controlled trials comparing operative and nonoperative interventions. J Shoulder Elbow Surg. (2023) 32:1746–60. doi: 10.1016/j.jse.2023.03.017

PubMed Abstract | Crossref Full Text | Google Scholar

9. Gehwolf R, Tempfer H, Cesur NP, Wagner A, Traweger A, and Lehner C. Tendinopathy: the interplay between mechanical stress, inflammation, and vascularity. Adv Sci (Weinh). (2025) 12:e06440. doi: 10.1002/advs.202506440

PubMed Abstract | Crossref Full Text | Google Scholar

10. Ma Z, Lee AYW, Kot CH, Yung PSH, Chen SC, and Lui PPY. Upregulation of FABP4 induced inflammation in the pathogenesis of chronic tendinopathy. J Orthop Translat. (2024) 47:105–15. doi: 10.1016/j.jot.2024.06.007

PubMed Abstract | Crossref Full Text | Google Scholar

11. Sanchez-Mila Z, Abuin-Porras V, Fidalgo-Gomez H, Minguez-Esteban I, Almazan-Polo J, and Velazquez-Saornil J. Efficacy of dry needling combined with eccentric exercise versus oral and topical NSAID treatment in patients with tennis elbow: A randomized controlled trial. J Evid Based Integr Med. (2025) 30:2515690X251355017. doi: 10.1177/2515690X251355017

PubMed Abstract | Crossref Full Text | Google Scholar

12. Li D, Li S, He S, He H, Yuan G, Ma B, et al. Restoring tendon microenvironment in tendinopathy: Macrophage modulation and tendon regeneration with injectable tendon hydrogel and tendon-derived stem cells exosomes. Bioact Mater. (2025) 47:152–69. doi: 10.1016/j.bioactmat.2025.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

13. Zhang AY, Xie QZ, Guo SZ, Liu X, Yu YH, Tang H, et al. Platelet-rich plasma-derived exosomes have the novel ability to alleviate insertional Achilles tendinopathy by promoting tenogenesis in tendon stem/progenitor cells. Biomater Adv. (2025) 173:214272. doi: 10.1016/j.bioadv.2025.214272

PubMed Abstract | Crossref Full Text | Google Scholar

14. Yuan Z, Yao S, Yao X, Zhou C, Li J, and Fan C. The IL-1beta/NETs/AIM2 axis participates in the formation of trauma-induced heterotopic ossification by orchestrating crosstalk between neutrophils and macrophages. J Adv Res. (2025). doi: 10.1016/j.jare.2025.06.069

PubMed Abstract | Crossref Full Text | Google Scholar

15. Smith EJ, Beaumont RE, McClellan A, Sze C, Palomino Lago E, Hazelgrove L, et al. Tumour necrosis factor alpha, interleukin 1 beta and interferon gamma have detrimental effects on equine tenocytes that cannot be rescued by IL-1RA or mesenchymal stromal cell-derived factors. Cell Tissue Res. (2023) 391:523–44. doi: 10.1007/s00441-022-03726-6

PubMed Abstract | Crossref Full Text | Google Scholar

16. Yang J, Chen J, Liu Y, Zhao X, Chen Z, Zheng H, et al. Multifunctional PRussian lue nanozymes ameliorate tendinopathy via modulating tissue homeostasis. Mater Today Bio. (2025) 34:102187. doi: 10.1016/j.mtbio.2025.102187

PubMed Abstract | Crossref Full Text | Google Scholar

17. Exconde PM, Hernandez-Chavez C, Bourne CM, Richards RM, Bray MB, Lopez JL, et al. The tetrapeptide sequence of IL-18 and IL-1beta regulates their recruitment and activation by inflammatory caspases. Cell Rep. (2023) 42:113581. doi: 10.1016/j.celrep.2023.113581

PubMed Abstract | Crossref Full Text | Google Scholar

18. Jiang L, Liu T, Lyu K, Chen Y, Lu J, Wang X, et al. Inflammation-related signaling pathways in tendinopathy. Open Life Sci. (2023) 18:20220729. doi: 10.1515/biol-2022-0729

PubMed Abstract | Crossref Full Text | Google Scholar

19. Chen H, Zhao S, Liu S, Wei Y, Guo Y, Gao K, et al. FCV activates NLRP3 inflammasome through ion-dependent pathway to promote IL-1beta secretion. Vet Microbiol. (2025) 307:110628. doi: 10.1016/j.vetmic.2025.110628

PubMed Abstract | Crossref Full Text | Google Scholar

20. Ford SG, Caswell P, Brough D, and Seoane PI. The secretion of interleukin-1beta. Cytokine Growth Factor Rev. (2025) 84:101–13. doi: 10.1016/j.cytogfr.2025.05.005

PubMed Abstract | Crossref Full Text | Google Scholar

21. Gupta S, Cassel SL, Sutterwala FS, and Dagvadorj J. Regulation of the NLRP3 inflammasome by autophagy and mitophagy. Immunol Rev. (2025) 329:e13410. doi: 10.1111/imr.13410

PubMed Abstract | Crossref Full Text | Google Scholar

22. Fu J and Wu H. Structural mechanisms of NLRP3 inflammasome assembly and activation. Annu Rev Immunol. (2023) 41:301–16. doi: 10.1146/annurev-immunol-081022-021207

PubMed Abstract | Crossref Full Text | Google Scholar

23. Xu J and Nunez G. The NLRP3 inflammasome: activation and regulation. Trends Biochem Sci. (2023) 48:331–44. doi: 10.1016/j.tibs.2022.10.002

PubMed Abstract | Crossref Full Text | Google Scholar

24. Meier DT, de Paula Souza J, and Donath MY. Targeting the NLRP3 inflammasome-IL-1beta pathway in type 2 diabetes and obesity. Diabetologia. (2025) 68:3–16. doi: 10.1007/s00125-024-06306-1

PubMed Abstract | Crossref Full Text | Google Scholar

25. Chen Y, Ye X, Escames G, Lei W, Zhang X, Li M, et al. The NLRP3 inflammasome: contributions to inflammation-related diseases. Cell Mol Biol Lett. (2023) 28:51. doi: 10.1186/s11658-023-00462-9

PubMed Abstract | Crossref Full Text | Google Scholar

26. Yuan XY, Zhang Y, Zhao X, Chen A, and Liu P. IL-1beta, an important cytokine affecting Helicobacter pylori-mediated gastric carcinogenesis. Microb Pathog. (2023) 174:105933. doi: 10.1016/j.micpath.2022.105933

PubMed Abstract | Crossref Full Text | Google Scholar

27. Ma Q. Pharmacological inhibition of the NLRP3 inflammasome: structure, molecular activation, and inhibitor-NLRP3 interaction. Pharmacol Rev. (2023) 75:487–520. doi: 10.1124/pharmrev.122.000629

PubMed Abstract | Crossref Full Text | Google Scholar

28. Li Y and Jiang Q. Uncoupled pyroptosis and IL-1beta secretion downstream of inflammasome signaling. Front Immunol. (2023) 14:1128358. doi: 10.3389/fimmu.2023.1128358

PubMed Abstract | Crossref Full Text | Google Scholar

29. Kelly SB, Green E, Hunt RW, Nold-Petry CA, Gunn AJ, Nold MF, et al. Interleukin-1: an important target for perinatal neuroprotection? Neural Regener Res. (2023) 18:47–50. doi: 10.4103/1673-5374.341044

PubMed Abstract | Crossref Full Text | Google Scholar

30. Jacobs N and Griffin GK. Throwing IL-1beta for a loop. Sci Immunol. (2024) 9:eado2158. doi: 10.1126/sciimmunol.ado2158

PubMed Abstract | Crossref Full Text | Google Scholar

31. Ren W, Sun Y, Zhao L, and Shi X. NLRP3 inflammasome and its role in autoimmune diseases: A promising therapeutic target. BioMed Pharmacother. (2024) 175:116679. doi: 10.1016/j.biopha.2024.116679

PubMed Abstract | Crossref Full Text | Google Scholar

32. Vande Walle L and Lamkanfi M. Drugging the NLRP3 inflammasome: from signalling mechanisms to therapeutic targets. Nat Rev Drug Discov. (2024) 23:43–66. doi: 10.1038/s41573-023-00822-2

PubMed Abstract | Crossref Full Text | Google Scholar

33. Penin-Franch A, Hurtado-Navarro L, Garcia-Vidal JA, Escolar-Reina P, Medina-Mirapeix F, and Pelegrin P. Multiple ASC-dependent inflammasomes drive differential pro-inflammatory cytokine production in a mouse model of tendinopathy. Biosci Rep. (2024) 44. doi: 10.1042/BSR20241282

PubMed Abstract | Crossref Full Text | Google Scholar

34. Ortega MA, De Leon-Oliva D, Garcia-Montero C, Fraile-Martinez O, Boaru DL, de Castro AV, et al. Reframing the link between metabolism and NLRP3 inflammasome: therapeutic opportunities. Front Immunol. (2023) 14:1232629. doi: 10.3389/fimmu.2023.1232629

PubMed Abstract | Crossref Full Text | Google Scholar

35. Bogdanova D, Samsonov MY, Lebedeva S, Bukhanova D, Materenchuk M, and Mutig K. Targeting interleukin-1 signaling for renoprotection. Front Immunol. (2025) 16:1591197. doi: 10.3389/fimmu.2025.1591197

PubMed Abstract | Crossref Full Text | Google Scholar

36. Fok ET, Moorlag S, Negishi Y, Groh LA, Dos Santos JC, Grawe C, et al. A chromatin-regulated biphasic circuit coordinates IL-1beta-mediated inflammation. Nat Genet. (2024) 56:85–99. doi: 10.1038/s41588-023-01598-2

PubMed Abstract | Crossref Full Text | Google Scholar

37. Malkova AM, Gubal AR, Petrova AL, Voronov E, Apte RN, Semenov KN, et al. Pathogenetic role and clinical significance of interleukin-1beta in cancer. Immunology. (2023) 168:203–16. doi: 10.1111/imm.13486

PubMed Abstract | Crossref Full Text | Google Scholar

38. Manoj H, Gomes SM, Thimmappa PY, Nagareddy PR, Jamora C, and Joshi MB. Cytokine signalling in formation of neutrophil extracellular traps: Implications for health and diseases. Cytokine Growth Factor Rev. (2025) 81:27–39. doi: 10.1016/j.cytogfr.2024.12.001

PubMed Abstract | Crossref Full Text | Google Scholar

39. Morita W, Snelling SJB, Wheway K, Watkins B, Appleton L, Carr AJ, et al. ERK1/2 drives IL-1beta-induced expression of TGF-beta1 and BMP-2 in torn tendons. Sci Rep. (2019) 9:19005. doi: 10.1038/s41598-019-55387-1

PubMed Abstract | Crossref Full Text | Google Scholar

40. Oliva F, Marsilio E, Asparago G, Frizziero A, Berardi AC, and Maffulli N. The impact of hyaluronic acid on tendon physiology and its clinical application in tendinopathies. Cells. (2021) 10. doi: 10.3390/cells10113081

PubMed Abstract | Crossref Full Text | Google Scholar

41. Liang W, Zhou C, Deng Y, Fu L, Zhao J, Long H, et al. The current status of various preclinical therapeutic approaches for tendon repair. Ann Med. (2024) 56:2337871. doi: 10.1080/07853890.2024.2337871

PubMed Abstract | Crossref Full Text | Google Scholar

42. Zhang X, Li M, Mao X, Yao Z, Zhu W, Yuan Z, et al. Small intestinal submucosa hydrogel loaded with gastrodin for the repair of achilles tendinopathy. Small. (2024) 20:e2401886. doi: 10.1002/smll.202401886

PubMed Abstract | Crossref Full Text | Google Scholar

43. Wang Z, Dong Z, Li Y, Jiao X, Liu Y, Chang H, et al. Verapamil attenuates the severity of tendinopathy by mitigating mitochondrial dysfunction through the activation of the Nrf2/HO-1 pathway. Biomedicines. (2024) 12. doi: 10.3390/biomedicines12040904

PubMed Abstract | Crossref Full Text | Google Scholar

44. Chen R, Ai L, Zhang J, and Jiang D. Dendritic cell-derived exosomes promote tendon healing and regulate macrophage polarization in preventing tendinopathy. Int J Nanomedicine. (2024) 19:11701–18. doi: 10.2147/IJN.S466363

PubMed Abstract | Crossref Full Text | Google Scholar

45. Dakin SG, Buckley CD, Al-Mossawi MH, Hedley R, Martinez FO, Wheway K, et al. Persistent stromal fibroblast activation is present in chronic tendinopathy. Arthritis Res Ther. (2017) 19:16. doi: 10.1186/s13075-016-1218-4

PubMed Abstract | Crossref Full Text | Google Scholar

46. Dakin SG, Martinez FO, Yapp C, Wells G, Oppermann U, Dean BJ, et al. Inflammation activation and resolution in human tendon disease. Sci Transl Med. (2015) 7:311ra173. doi: 10.1126/scitranslmed.aac4269

PubMed Abstract | Crossref Full Text | Google Scholar

47. Fu W, Yang R, and Li J. Single-cell and spatial transcriptomics reveal changes in cell heterogeneity during progression of human tendinopathy. BMC Biol. (2023) 21:132. doi: 10.1186/s12915-023-01613-2

PubMed Abstract | Crossref Full Text | Google Scholar

48. Xu B, Wang Y, He G, Tao X, Gao S, Zhou M, et al. An aligned-to-random PLGA/Col1-PLGA/nHA bilayer electrospun nanofiber membrane enhances tendon-to-bone healing in a murine model. Am J Sports Med. (2025) 53:885–99. doi: 10.1177/03635465241310530

PubMed Abstract | Crossref Full Text | Google Scholar

49. Abraham AC, Shah SA, Golman M, Song L, Li X, Kurtaliaj I, et al. Targeting the NF-kappaB signaling pathway in chronic tendon disease. Sci Transl Med. (2019) 11. doi: 10.1126/scitranslmed.aav4319

PubMed Abstract | Crossref Full Text | Google Scholar

50. Lin X, Tian X, Jiang H, Li W, Wang C, Wu J, et al. Carpaine alleviates tendinopathy in mice by promoting the ubiquitin-proteasomal degradation of p65 via targeting the E3 ubiquitin ligase LRSAM1. Phytomedicine. (2024) 124:155323. doi: 10.1016/j.phymed.2023.155323

PubMed Abstract | Crossref Full Text | Google Scholar

51. Zhang J, Middleton KK, Fu FH, Im HJ, and Wang JH. HGF mediates the anti-inflammatory effects of PRP on injured tendons. PLoS One. (2013) 8:e67303. doi: 10.1371/journal.pone.0067303

PubMed Abstract | Crossref Full Text | Google Scholar

52. Zhang J, Maloney D, Pastukh V, Hattori S, Hogan MV, and Wang JH. Metformin lotion as a novel approach to prevent tendinopathy induced by mechanical overuse. Sci Rep. (2025) 15:31474. doi: 10.1038/s41598-025-16279-9

PubMed Abstract | Crossref Full Text | Google Scholar

53. Mousavizadeh R, Backman L, McCormack RG, and Scott A. Dexamethasone decreases substance P expression in human tendon cells: an in vitro study. Rheumatol (Oxford). (2015) 54:318–23. doi: 10.1093/rheumatology/keu315

PubMed Abstract | Crossref Full Text | Google Scholar

54. Garcia-Melchor E, Cafaro G, MacDonald L, Crowe LAN, Sood S, McLean M, et al. Novel self-amplificatory loop between T cells and tenocytes as a driver of chronicity in tendon disease. Ann Rheum Dis. (2021) 80:1075–85. doi: 10.1136/annrheumdis-2020-219335

PubMed Abstract | Crossref Full Text | Google Scholar

55. Bakht SM, Pardo A, Gomez-Florit M, Caballero D, Kundu SC, Reis RL, et al. Human tendon-on-chip: unveiling the effect of core compartment-T cell spatiotemporal crosstalk at the onset of tendon inflammation. Adv Sci (Weinh). (2024) 11:e2401170. doi: 10.1002/advs.202401170

PubMed Abstract | Crossref Full Text | Google Scholar

56. Dai W, Xu Q, Li Q, Wang X, Zhang W, Zhou G, et al. Piezoelectric nanofilms fabricated by coaxial electrospun polycaprolactone/Barium titanate promote Achilles tendon regeneration by reducing IL-17A/NF-kappaB-mediated inflammation. Bioact Mater. (2025) 49:1–22. doi: 10.1016/j.bioactmat.2025.02.008

PubMed Abstract | Crossref Full Text | Google Scholar

57. Stowe EJ, Keller MR, and Connizzo BK. Cellular senescence impairs tendon extracellular matrix remodeling in response to mechanical unloading. Aging Cell. (2024) 23:e14278. doi: 10.1111/acel.14278

PubMed Abstract | Crossref Full Text | Google Scholar

58. Lee MJ, Park K, Yeon Lee S, Jang KH, Won S, and Hyunchul Jo C. Effects of conditioned media from human umbilical cord-derived mesenchymal stem cells on tenocytes from degenerative rotator cuff tears in an interleukin 1beta-induced tendinopathic condition. Orthop J Sports Med. (2024) 12:23259671241286412. doi: 10.1177/23259671241286412

PubMed Abstract | Crossref Full Text | Google Scholar

59. Lee AY, Park JY, Hwang SJ, Jang KH, and Jo CH. Effects of late-passage small umbilical cord-derived fast proliferating cells on tenocytes from degenerative rotator cuff tears under an interleukin 1beta-induced tendinopathic environment. Tissue Eng Regener Med. (2024) 21:1217–31. doi: 10.1007/s13770-024-00673-x

PubMed Abstract | Crossref Full Text | Google Scholar

60. Belacic ZA, Sullivan SN, Rice HC, and Durgam SS. Interleukin-1beta and methylprednisolone acetate demonstrate differential effects on equine deep digital flexor tendon and navicular bone fibrocartilage cells in vitro. Am J Vet Res. (2023) 84. doi: 10.2460/ajvr.22.08.0128

PubMed Abstract | Crossref Full Text | Google Scholar

61. Corps AN, Curry VA, Buttle DJ, Hazleman BL, and Riley GP. Inhibition of interleukin-1beta-stimulated collagenase and stromelysin expression in human tendon fibroblasts by epigallocatechin gallate ester. Matrix Biol. (2004) 23:163–9. doi: 10.1016/j.matbio.2004.05.001

PubMed Abstract | Crossref Full Text | Google Scholar

62. Jiao X, Wang Z, Li Y, Wang T, Xu C, Zhou X, et al. Fullerenol inhibits tendinopathy by alleviating inflammation. Front Bioeng Biotechnol. (2023) 11:1171360. doi: 10.3389/fbioe.2023.1171360

PubMed Abstract | Crossref Full Text | Google Scholar

63. McClellan A, Evans R, Sze C, Kan S, Paterson Y, and Guest D. A novel mechanism for the protection of embryonic stem cell derived tenocytes from inflammatory cytokine interleukin 1 beta. Sci Rep. (2019) 9:2755. doi: 10.1038/s41598-019-39370-4

PubMed Abstract | Crossref Full Text | Google Scholar

64. de Mos M, Joosten LA, Oppers-Walgreen B, van Schie JT, Jahr H, van Osch GJ, et al. Tendon degeneration is not mediated by regulation of Toll-like receptors 2 and 4 in human tenocytes. J Orthop Res. (2009) 27:1043–7. doi: 10.1002/jor.20834

PubMed Abstract | Crossref Full Text | Google Scholar

65. Gogele C, Pattappa G, Tempfer H, Docheva D, and Schulze-Tanzil G. Tendon mechanobiology in the context of tendon biofabrication. Front Bioeng Biotechnol. (2025) 13:1560025. doi: 10.3389/fbioe.2025.1560025

PubMed Abstract | Crossref Full Text | Google Scholar

66. Pentzold S and Wildemann B. Mechanical overload decreases tenogenic differentiation compared to physiological load in bioartificial tendons. J Biol Eng. (2022) 16:5. doi: 10.1186/s13036-022-00283-y

PubMed Abstract | Crossref Full Text | Google Scholar

67. Gracey E, Burssens A, Cambre I, Schett G, Lories R, McInnes IB, et al. Tendon and ligament mechanical loading in the pathogenesis of inflammatory arthritis. Nat Rev Rheumatol. (2020) 16:193–207. doi: 10.1038/s41584-019-0364-x

PubMed Abstract | Crossref Full Text | Google Scholar

68. Leahy TP, Chenna SS, Soslowsky LJ, and Dyment NA. Focal adhesion kinase regulates tendon cell mechanoresponse and physiological tendon development. FASEB J. (2024) 38:e70050. doi: 10.1096/fj.202400151R

PubMed Abstract | Crossref Full Text | Google Scholar

69. Gardner K, Lavagnino M, Egerbacher M, and Arnoczky SP. Re-establishment of cytoskeletal tensional homeostasis in lax tendons occurs through an actin-mediated cellular contraction of the extracellular matrix. J Orthop Res. (2012) 30:1695–701. doi: 10.1002/jor.22131

PubMed Abstract | Crossref Full Text | Google Scholar

70. Holmes DF, Yeung CC, Garva R, Zindy E, Taylor SH, Lu Y, et al. Synchronized mechanical oscillations at the cell-matrix interface in the formation of tensile tissue. Proc Natl Acad Sci U S A. (2018) 115:E9288–97. doi: 10.1073/pnas.1801759115

PubMed Abstract | Crossref Full Text | Google Scholar

71. Stanczak M, Kacprzak B, and Gawda P. Tendon cell biology: effect of mechanical loading. Cell Physiol Biochem. (2024) 58:677–701. doi: 10.33594/000000743

PubMed Abstract | Crossref Full Text | Google Scholar

72. Chen Q, Zhou J, Zhang B, Chen Z, Luo Q, and Song G. Cyclic stretching exacerbates tendinitis by enhancing NLRP3 inflammasome activity via F-actin depolymerization. Inflammation. (2018) 41:1731–43. doi: 10.1007/s10753-018-0816-5

PubMed Abstract | Crossref Full Text | Google Scholar

73. Chen Z, Li M, Chen P, Tai A, Li J, Bassonga EL, et al. Mechanical overload-induced release of extracellular mitochondrial particles from tendon cells leads to inflammation in tendinopathy. Exp Mol Med. (2024) 56:583–99. doi: 10.1038/s12276-024-01183-5

PubMed Abstract | Crossref Full Text | Google Scholar

74. Lavagnino M, Brooks AE, Oslapas AN, Gardner KL, and Arnoczky SP. Crimp length decreases in lax tendons due to cytoskeletal tension, but is restored with tensional homeostasis. J Orthop Res. (2017) 35:573–9. doi: 10.1002/jor.23489

PubMed Abstract | Crossref Full Text | Google Scholar

75. Maeda E, Kuroyanagi K, Ando Y, and Matsumoto T. Effects of substrate stiffness on morphology and MMP-1 gene expression in tenocytes stimulated with interleukin-1beta. J Orthop Res. (2020) 38:150–9. doi: 10.1002/jor.24403

PubMed Abstract | Crossref Full Text | Google Scholar

76. Di X, Gao X, Peng L, Ai J, Jin X, Qi S, et al. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther. (2023) 8:282. doi: 10.1038/s41392-023-01501-9

PubMed Abstract | Crossref Full Text | Google Scholar

77. Pan C, Lu F, Hao X, Deng X, Liu J, Sun K, et al. Low-intensity pulsed ultrasound delays the progression of osteoarthritis by regulating the YAP-RIPK1-NF-kappaB axis and influencing autophagy. J Transl Med. (2024) 22:286. doi: 10.1186/s12967-024-05086-x

PubMed Abstract | Crossref Full Text | Google Scholar

78. Lazarczuk SL, Maniar N, Opar DA, Duhig SJ, Shield A, Barrett RS, et al. Mechanical, material and morphological adaptations of healthy lower limb tendons to mechanical loading: A systematic review and meta-analysis. Sports Med. (2022) 52:2405–29. doi: 10.1007/s40279-022-01695-y

PubMed Abstract | Crossref Full Text | Google Scholar

79. Liu H, Li Y, An Y, He P, Wu L, Gao Y, et al. Endothelium-independent hypoxic contraction is prevented specifically by nitroglycerin via inhibition of Akt kinase in porcine coronary artery. Stem Cells Int. (2016) 2016:2916017. doi: 10.1155/2016/2916017

PubMed Abstract | Crossref Full Text | Google Scholar

80. Lu J, Yang X, He C, Chen Y, Li C, Li S, et al. Rejuvenation of tendon stem/progenitor cells for functional tendon regeneration through platelet-derived exosomes loaded with recombinant Yap1. Acta Biomater. (2023) 161:80–99. doi: 10.1016/j.actbio.2023.02.018

PubMed Abstract | Crossref Full Text | Google Scholar

81. Lu K, Zhou M, Wang L, Wang Y, Tang H, He G, et al. N-Acetyl-L-cysteine facilitates tendon repair and promotes the tenogenic differentiation of tendon stem/progenitor cells by enhancing the integrin alpha5/beta1/PI3K/AKT signaling. BMC Mol Cell Biol. (2023) 24:1. doi: 10.1186/s12860-022-00463-0

PubMed Abstract | Crossref Full Text | Google Scholar

82. Sakai T and Kumagai K. Molecular dissection of tendon development and healing: Insights into tenogenic phenotypes and functions. J Biol Chem. (2025) 301:108353. doi: 10.1016/j.jbc.2025.108353

PubMed Abstract | Crossref Full Text | Google Scholar

83. Gao Y, Wang H, Shi L, Lu P, Dai G, Zhang M, et al. Erroneous differentiation of tendon stem/progenitor cells in the pathogenesis of tendinopathy: current evidence and future perspectives. Stem Cell Rev Rep. (2025) 21:423–53. doi: 10.1007/s12015-024-10826-z

PubMed Abstract | Crossref Full Text | Google Scholar

84. Liu H, Zhu M, Yang H, Chai L, Han J, Ning L, et al. Monitoring endoplasmic reticulum peroxynitrite fluctuations in primary tendon-derived stem cells and insights into tendinopathy. ACS Sens. (2024) 9:6750–8. doi: 10.1021/acssensors.4c02452

PubMed Abstract | Crossref Full Text | Google Scholar

85. Zhang K, Asai S, Yu B, and Enomoto-Iwamoto M. IL-1beta irreversibly inhibits tenogenic differentiation and alters metabolism in injured tendon-derived progenitor cells in vitro. Biochem Biophys Res Commun. (2015) 463:667–72. doi: 10.1016/j.bbrc.2015.05.122

PubMed Abstract | Crossref Full Text | Google Scholar

86. Geng Y, Zhao X, Xu J, Zhang X, Hu G, Fu SC, et al. Overexpression of mechanical sensitive miR-337-3p alleviates ectopic ossification in rat tendinopathy model via targeting IRS1 and Nox4 of tendon-derived stem cells. J Mol Cell Biol. (2020) 12:305–17. doi: 10.1093/jmcb/mjz030

PubMed Abstract | Crossref Full Text | Google Scholar

87. Yang L, Tang C, Chen Y, Ruan D, Zhang E, Yin Z, et al. Pharmacological inhibition of rac1 activity prevents pathological calcification and enhances tendon regeneration. ACS Biomater Sci Eng. (2019) 5:3511–22. doi: 10.1021/acsbiomaterials.9b00335

PubMed Abstract | Crossref Full Text | Google Scholar

88. Li J, Wang X, Yao Z, Yuan F, Liu H, Sun Z, et al. NLRP3-dependent crosstalk between pyroptotic macrophage and senescent cell orchestrates trauma-induced heterotopic ossification during aberrant wound healing. Adv Sci (Weinh). (2023) 10:e2207383. doi: 10.1002/advs.202207383

PubMed Abstract | Crossref Full Text | Google Scholar

89. Ko PY, Hsu CC, Chen SY, Kuo LC, Su WR, Jou IM, et al. Cross-linked hyaluronate and corticosteroid combination ameliorate the rat experimental tendinopathy through anti-senescent and -apoptotic effects. Int J Mol Sci. (2022) 23. doi: 10.3390/ijms23179760

PubMed Abstract | Crossref Full Text | Google Scholar

90. Chen H, Chen X, Yang L, Sheng S, Yang J, Lu Y, et al. TRIM54 alleviates inflammation and apoptosis by stabilizing YOD1 in rat tendon-derived stem cells. J Biol Chem. (2024) 300:105510. doi: 10.1016/j.jbc.2023.105510

PubMed Abstract | Crossref Full Text | Google Scholar

91. Vinhas A, Rodrigues MT, Goncalves AI, and Gomes ME. Immunomodulatory Behavior of Tendon Magnetic Cell Sheets can be Modulated in Hypoxic Environments under Magnetic Stimulus. ACS Appl Mater Interfaces. (2024) 16:44440–50. doi: 10.1021/acsami.4c08154

PubMed Abstract | Crossref Full Text | Google Scholar

92. Busch F, Mobasheri A, Shayan P, Lueders C, Stahlmann R, and Shakibaei M. Resveratrol modulates interleukin-1beta-induced phosphatidylinositol 3-kinase and nuclear factor kappaB signaling pathways in human tenocytes. J Biol Chem. (2012) 287:38050–63. doi: 10.1074/jbc.M112.377028

PubMed Abstract | Crossref Full Text | Google Scholar

93. Chen Q, Lu H, and Yang H. Chitosan prevents adhesion during rabbit flexor tendon repair via the sirtuin 1 signaling pathway. Mol Med Rep. (2015) 12:4598–603. doi: 10.3892/mmr.2015.4007

PubMed Abstract | Crossref Full Text | Google Scholar

94. Buhrmann C, Mobasheri A, Busch F, Aldinger C, Stahlmann R, Montaseri A, et al. Curcumin modulates nuclear factor kappaB (NF-kappaB)-mediated inflammation in human tenocytes in vitro: role of the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem. (2011) 286:28556–66. doi: 10.1074/jbc.M111.256180

PubMed Abstract | Crossref Full Text | Google Scholar

95. Dai G, Li Y, Zhang M, Lu P, Zhang Y, Wang H, et al. The regulation of the AMPK/mTOR axis mitigates tendon stem/progenitor cell senescence and delays tendon aging. Stem Cell Rev Rep. (2023) 19:1492–506. doi: 10.1007/s12015-023-10526-0

PubMed Abstract | Crossref Full Text | Google Scholar

96. Wang H, Dai GC, Li YJ, Chen MH, Lu PP, Zhang YW, et al. Targeting senescent tendon stem/progenitor cells to prevent or treat age-related tendon disorders. Stem Cell Rev Rep. (2023) 19:680–93. doi: 10.1007/s12015-022-10488-9

PubMed Abstract | Crossref Full Text | Google Scholar

97. Korcari A, Przybelski SJ, Gingery A, and Loiselle AE. Impact of aging on tendon homeostasis, tendinopathy development, and impaired healing. Connect Tissue Res. (2023) 64:1–13. doi: 10.1080/03008207.2022.2102004

PubMed Abstract | Crossref Full Text | Google Scholar

98. Chen SY, Jou IM, Ko PY, Hsu KL, Su WR, Kuo LC, et al. Amelioration of experimental tendinopathy by lentiviral CD44 gene therapy targeting senescence-associated secretory phenotypes. Mol Ther Methods Clin Dev. (2022) 26:157–68. doi: 10.1016/j.omtm.2022.06.006

PubMed Abstract | Crossref Full Text | Google Scholar

99. Hsu CC, Chen SY, Ko PY, Kwan FC, Su WR, Jou IM, et al. MicroRNA-146a gene transfer ameliorates senescence and senescence-associated secretory phenotypes in tendinopathic tenocytes. Aging (Albany NY). (2024) 16:2702–14. doi: 10.18632/aging.205505

PubMed Abstract | Crossref Full Text | Google Scholar

100. Dede Eren A, Vermeulen S, Schmitz TC, Foolen J, and de Boer J. The loop of phenotype: Dynamic reciprocity links tenocyte morphology to tendon tissue homeostasis. Acta Biomater. (2023) 163:275–86. doi: 10.1016/j.actbio.2022.05.019

PubMed Abstract | Crossref Full Text | Google Scholar

101. DiIorio SE, Young B, Parker JB, Griffin MF, and Longaker MT. Understanding tendon fibroblast biology and heterogeneity. Biomedicines. (2024) 12. doi: 10.3390/biomedicines12040859

PubMed Abstract | Crossref Full Text | Google Scholar

102. de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, et al. Matrix metalloproteinases: from molecular mechanisms to physiology, pathophysiology, and pharmacology. Pharmacol Rev. (2022) 74:712–68. doi: 10.1124/pharmrev.121.000349

PubMed Abstract | Crossref Full Text | Google Scholar

103. Dang J, Zhang Z, Fu J, Sun L, Shi Y, Wang L, et al. Regulating inflammation microenvironment and tenogenic differentiation as sequential therapy promotes tendon healing in diabetic rats. J Orthop Translat. (2025) 53:63–81. doi: 10.1016/j.jot.2025.04.015

PubMed Abstract | Crossref Full Text | Google Scholar

104. Najafi Z, Moosavi Z, Baradaran Rahimi V, Hashemitabar G, and Askari VR. Evaluation of Anti-Nociceptive, Anti-Inflammatory, and Anti-Fibrotic effects of noscapine against a rat model of Achilles tendinopathy. Int Immunopharmacol. (2024) 130:111704. doi: 10.1016/j.intimp.2024.111704

PubMed Abstract | Crossref Full Text | Google Scholar

105. Kim DS, Kim JH, Baek SW, Lee JK, Park SY, Choi B, et al. Controlled vitamin D delivery with injectable hyaluronic acid-based hydrogel for restoration of tendinopathy. J Tissue Eng. (2022) 13:20417314221122089. doi: 10.1177/20417314221122089

PubMed Abstract | Crossref Full Text | Google Scholar

106. Wei B, Ji M, Lin Y, Wang S, Liu Y, Geng R, et al. Mitochondrial transfer from bone mesenchymal stem cells protects against tendinopathy both in vitro and in vivo. Stem Cell Res Ther. (2023) 14:104. doi: 10.1186/s13287-023-03329-0

PubMed Abstract | Crossref Full Text | Google Scholar

107. Wang K, Luo H, Liu L, Gao H, Song Y, and Li D. Blockade of IL-1 family cytokines in the treatment of rheumatoid arthritis. Front Pharmacol. (2025) 16:1577628. doi: 10.3389/fphar.2025.1577628

PubMed Abstract | Crossref Full Text | Google Scholar

108. Yao TK, Lee RP, Wu WT, Chen IH, Yu TC, and Yeh KT. Advances in gouty arthritis management: integration of established therapies, emerging treatments, and lifestyle interventions. Int J Mol Sci. (2024) 25. doi: 10.3390/ijms251910853

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: IL-1β, tendinopathy, pathogenesis, therapeutic targets, proinflammatory cytokines

Citation: Zhang Y, Wang J, Tang F, Xian R, Zhang H, Yuan Y, Yang G and Chen G (2025) Mechanistic insights into IL-1β-mediated progression of tendinopathy. Front. Immunol. 16:1657285. doi: 10.3389/fimmu.2025.1657285

Received: 01 July 2025; Accepted: 15 October 2025;
Published: 31 October 2025.

Edited by:

Mengcun Chen, University of Pennsylvania, United States

Reviewed by:

Lin Zhiqi, Shanghai Jiao Tong University, China
Yuanyin Teng, Zhejiang University, China
Kai Yang, Washington University in St. Louis, United States

Copyright © 2025 Zhang, Wang, Tang, Xian, Zhang, Yuan, Yang and Chen. 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: Guiquan Chen, Y2hlbmd1aXF1YW4xOTczQDEyNi5jb20=; Guoqiang Yang, eWd1b3FpYW5nMTk4NEBzd211LmVkdS5jbg==

These authors have contributed equally to this work and share first authorship

Disclaimer: 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.