Skip to main content

Research progress on the mechanism of interleukin-1β on epiphyseal plate chondrocytes

Abstract

Epiphyseal plate injury, a common problem in pediatric orthopedics, may result in poor bone repair or growth defects. Epiphyseal plate, also known as growth plate is a layer of hyaline cartilage tissue between the epiphysis and metaphyseal and has the ability to grow longitudinally. Under normal physiological conditions, the epiphyseal plate has a certain axial resistance to stress, but it is fragile in growth phase and can be damaged by excessive stress, leading to detachment or avulsion of the epiphysis, resulting in life-long devastating consequences for patients. There is an obvious inflammatory response in the phase of growth plate injury, the limited physiological inflammatory response locally favors tissue repair and the organism, but uncontrolled chronic inflammation always leads to tissue destruction and disease progression. Interleukin-1β (IL-1β), as representative inflammatory factors, not only affect the inflammatory phase response to bone and soft tissue injury, but have a potentially important role in the later repair phase, though the exact mechanism is not fully understood. At present, epiphyseal plate injuries are mainly treated by corrective and reconstructive surgery, which is highly invasive with limited effectiveness, thus new therapeutic approaches are urgently needed, so a deeper understanding and exploration of the pathological mechanisms of epiphyseal plate injuries at the cellular molecular level is an entry point. In this review, we fully introduced the key role of IL-1 in the progression of epiphyseal plate injury and repair, deeply explored the mechanism of IL-1 on the molecular transcript level and endocrine metabolism of chondrocytes from multiple aspects, and summarized other possible mechanisms to provide theoretical basis for the clinical treatment and in-depth study of epiphyseal plate injury in children.

Introduction

The growth plate assumes responsibility for children’s longitudinal growth of long bones. However, as the weakest part of long bone, it is vulnerable to damage and have limited regenerative capacity, which may often fail to be fully repaired after injury, resulting in some patients being severely affected for the later growth failure. Inhibition of the growth function of the epiphyseal plate generally occurs for two reasons: (1) due to the cartilage damage or blood supply disorder, the ability of the epiphyseal plate growth zone is reduced, leading to premature closure; (2) special types of fracture like Salter–Harris type III and IV epiphyseal plate fractures misaligned healing and local formation of bone bridges, resulting in restrained growth. Incomplete closure or even premature arthrogryposis may result from complete growth failure, differences in limb lengths, and angulation deformities, a series of complications that can significantly affect growth and development in children. It is therefore necessary to properly assess and manage growth plate injuries. The procedure of fracture healing involves complex processes such as inflammation inflammatory reaction, endochondral ossification and bone reconstruction [1, 2]. The immune response at the injured site is of critical importance to affect fracture healing, which rapidly forms hematoma, chemotactic and recruit a large number of inflammatory cells [3], releasing the cytokine IL-1β, TNF-α, etc. It can be metabolized physiologically when pro-inflammatory and anti-inflammatory cytokines work together to maintain homeostasis. However, due to the limited regeneration and self-repair ability of this site, without intervention, lots of inflammatory factors infiltrate like IL-1β, causing dynamic imbalance of pro-inflammatory and anti-inflammatory cytokines, triggering chondrocyte apoptosis and metabolic abnormalities resulting in bone growth defects of the involved limb [4, 5]. It is consistent with the research using growth plate injured rats by Fiona H. Zhou et al., which showed that IL-1β may play an important role in early acute inflammatory events and later bone bridge formation and remodeling [6]. However, there are few research about the detailed mechanism of IL-1β on epiphyseal plate chondrocytes at present. With the continuous development of studies on cartilage-derived diseases such as traumatic epiphyseal plate closure and osteoarthritis, increasing attention is being gained to IL-1β action. Therefore, this article focuses on clarifying the mechanism of action of IL-1β on the effect on epiphyseal plate chondrocytes, provides new perspectives and ideas to furnish a theoretical basis for clinical applications.

Biological function of IL-1

The IL-1 family concludes 11 members, and these cytokines have the same c-terminal three-dimensional structure. In 1984, IL-1 cDNA isolated from human body first proved that IL-1 has two different biochemical forms, namely IL-1α and IL-1β [7]. IL-1β is mainly secreted from macrophages, with a molecular weight of about 17 kDa and widely present in various tissues and organs in human body. IL-1β is an inflammatory cytokine with the same receptor complex as IL-1α and acts on the same cytokine receptor for signal regulation, which can trigger inflammation, especially when tissues are damaged. But the effects also differ due to differences in how they are produced, where IL-1β is mainly synthesized by phagocytes [8]. The pathogenic effect of IL-1β has been fully verified in auto-inflammatory diseases, mainly on account of gain-of-function mutations in genes encoding mature inflammasome. Studies have also confirmed that IL-1β is locally induced in the bone marrow niche in response to injury, which contributes to bone marrow emergency generation [9, 10]. Autoimmune diseases, diabetes mellitus, gout, and neurodegenerative diseases, among others, also have its involvement [8, 11, 12]. As the best-characterized member of this family [8], the secretion of IL-1β is tightly regulated, requiring inflammatory activation as a second stimulus. Inflammatory activation makes maturation of inflammatory caspases, followed by cleavage of the pro-IL-1β into active forms, and secret subsequently [13, 14]. In addition, due to the polymorphism of IL-1β, its function is not only limited to the body inflammatory response, but is associated with multiple system pathological functions [15,16,17]. In 1989, Yamashita et al. confirmed that IL-1β can be produced by mature chondrocytes by immunohistochemical localization of IL-1 in human epiphyseal plate and cartilage canal tissue [18]. Normal joint synovial fluid contains trace amount of IL-1β. It can also be observed in cultured chondrocytes and synovial cells [19]. It indicates that IL-1β derived from chondrocytes may have an important impact in hypertrophy, cartilage revascularization and bone formation. Jenei-Lanzl Z et al. described a link between IL-1β and bone disorders in different subpopulations [20], IL-1β has effect on chondrocytes through catabolism, a process that involves upregulation of polymerases and matrix metalloproteinase (MMPs) and is further self-upregulated in chondrocytes through a positive feedback mechanism. In addition, a certain degree of mechanical strain can also induce IL-1β expression in chondrocytes in osteoarthritis. The function of IL-1β is far more than these, and it was the availability of its correlative studies with chondrocytes that gave us a new entry point/direction into the study of poor healing after epiphyseal plate injury.

How IL-1β affects the chondrocytes

Degradation of the cartilage extracellular matrix

Downregulation of IL-1β on SOX-9 expression

Earlier studies by M et al. found that IL-1β inhibited mRNA expression of proteoglycan and collagen type II, and significantly prevented synthesis of proteoglycan, resulting in ATDC5 chondrocyte dynamics and metatarsal growth restriction [21]. Collagen type II, one of the principal extracellular matrix components abundant in cartilage, combines with proteoglycan to maintain the cellular structure acting as a skeleton for chondrocyte attachment and migration, and interacts with chondrocytes to effectively maintain the chondrogenic phenotype, While the decrease of their synthesis and expression will lower the potential for recovery results in loss of the chondrocyte phenotype. As a key transcription factor in chondrocytes, sox-9 plays a key role in the process of cartilage development, activating the transcription of many cartilage specific genes, such as collagen-2 (COL2A1) and cartilage oligomeric protein (COMP), which encode the extracellular matrix (ECM) component, and directly regulating Col2a1 transcription process, especially by targeting specific binding sites located in introns [22, 23]. Specific deletion of SOX9 leads to severe cartilage dysplasia in mice before and after mesenchymal condensation [24]. This was further supported by the fact that SOX9 transcription levels were significantly reduced in osteoarthritis with degradation of extracellular matrix, while when transduced to overexpress in human chondrocytes, it significantly stimulated the synthesis of proteoglycan and collagen II to restore extracellular matrix and promote the re-expression of cartilage phenotype [25, 26]. In addition, silencing of SOX9 reversed the protective effect of etomidate on ECM degradation components in an in vitro injury model of chondrocytes stimulated by glycation end-products (AGEs) [27]. Machiyuan et al. observed for the first time that degradation of type II collagen and SOX-9 was only regulated by of nuclear receptor subfamily 4A group a member 3 (NR4A3). Further studies showed that IL-induced changes in NR4A3 in chondrocytes affected cartilage matrix degradation [28, 29]. What’s more, the expression of collagen II and SOX-9 decreased in chondrocytes under IL-1β intervention, while F-box/WD repeat-containing protein 7 (FBW7) promoted the integral role of collagen type II, proteoglycan and Sox-9 in cartilage to correct chondrocytic disorders [30]. Xu Z et al. found that the activation of G protein coupled receptor 120 (GPR120) after exposure to IL-1β in ATDC5 chondrocytes can reverse the expression of collagen type II and proteoglycan via blocking the downregulation of Sox-9, to reduce the inflammation induced by IL-1β [31]. The above studies illustrate that the elevation of IL-1β content can negatively regulate SOX-9 after injury occurs, which in turn leads to inhibition of proteoglycan and collagen type II synthesis, finally causing cartilage extracellular matrix degradation.

Induction of IL-1β on MMP synthesis

Sox-9 expression in chondrocyte in response to IL-1β was reduced, as was the matrix metalloproteinase family (MMPs). MMPs, a super family of protease widely found in connective tissues, whose activity is regulated by zinc ions, are mainly responsible for extracellular matrix degradation and tissue remodeling. Luteolin has been proven to exert anti-inflammatory properties, and Junliang Fei et al., by evaluating the expression of various indicators in chondrocytes that were intervened with IL-1β for 24 h, suggested that luteolin significantly decreased MMP9 and MMP13 synthesis which promoted by IL-1β, effectively reversed collagen type II degradation [32]. Bone morphogenetic protein 2 (BMP2) is a known indicator of osteogenesis. IL-1β stimulates chondrocytes and increases the expression levels of BMP2 and MMP13 by targeting the MEK/ERK/SP1 pathway, enhancing cartilage structural remodeling and cartilage degradation, which leads to chondrocyte degeneration [33]. In the investigation of osteogenic differentiation process luteolin was found that, in periodontal ligament cells, on the one hand, was able to dose-dependently increase BMP2 expression to promote osteogenic differentiation, on the other hand, was to simultaneously antagonize the negative effect of IL-1β-promoted MMP production on cartilage production [34, 35]. In fibrocartilage-derived cells of the temporomandibular joint (TMJ), IL-1β increased the fibro-chondrocyte proteoglycan ADAMTS4 and ADAMTS5 expression, as well as strongly increased MMP-13 expression, then inducing cartilage damage [36]. In addition, disruption of collagen II and proteoglycan is an essential feature of cartilage in patients with intervertebral disc degeneration (IDD). It is found that reduction of cartilage-derived morphogenetic protein-1 (CDMP-1) was dose-dependent, while appropriate supplementation of CDMP-1 contributed to collagen II and proteoglycan synthesis and inhibited MMP-9 and MMP-13 breakdown after using IL-1β to intervene in nucleus pulposus cells [37]. Also, Wei Qi et al. treated human nucleus pulposus cells (HNPC) derived from the notochord under IL-1β intervention with tyrosol, a multi-component compound with anti-inflammatory properties, and found that tyrosol inhibited IL-1 through SIRT1/PI3K/AKT pathway to reverse the upregulation of MMP-3, MMP-9 and MMP-13, which can just reduce the degradation of type II collagen in chondrocytes [38]. In addition, Elsa Mével et al. study showed that hydroxytyrosol (HT), an olive major extract, exerted anti-osteoarthritis effects in a post-traumatic animal model and exhibited anti-inflammatory and chondroprotective effects in IL-1β-stimulated primary cultured rabbit chondrocytes [39]. Exploring the effects of tyrosol and similar compounds in chondrocyte culture in vitro or in an epiphyseal plate injury model in vivo would hopefully represent a breakthrough point for the treatment of this condition in the clinic. On the whole, IL-1β ultimately leads to cellular abnormalities by increasing the synthesis of MMPs, causing decomposition in collagen type II and thereby degrading cartilage extracellular matrix.

Effect of IL-1β at gene level

Accumulating evidence so far proves that RNA plays important roles in various diseases. Numerous studies have shown that IL-1β autophagy in cartilage was stimulated at the initial stage of inflammation, but was eventually significantly inhibited. It was found that upregulating ciRS-7 abnormally expressed in OA could enhance IL-cartilage degradation induced by IL-1β [40]. Beyond that, circRNA.33186 was frequently upregulated in chondrocytes treated with IL-1β, by knocking down circRNA.33816, MMP-13 was found decreased in chondrocytes by Zhou et al., while collagen type II increased, which accelerated proliferation of chondrocytes but simultaneously inhibited apoptosis [41]. circRNA.33186 is circular and mainly exists in the cytoplasm. Early studies found that circular RNA can competitively bind to miRNA [42]. Some scholars screened that there was an obviously negative interaction between miR-127-5P and circRNA.33186 through luciferase, which targeted MMP13 to regulate the catabolic function of IL-1β-treated chondrocytes, confirming that circRNA.33186 could directly or indirectly affect MMP13 through miR-127-5P, resulting in abnormal chondrocyte function [43]. At present, it has been recognized that miRNA and lncRNA are indispensable in the occurrence and development of diseases. They are of great importance to life activities, such as cell cycle regulation and cellular differentiation. Concretely, LncRNA MALAT1 was shown to be upgraded in IL-1β-treated chondrocytes. Ying Zhang et al. demonstrated that in the same cell model, MALAT1 targets miR-150-5p to regulate Akt3 indirectly, and competitively binds with miR-150-5p to inhibit proteoglycan and collagen II expression, reduce cell proliferation as well [44]. Moreover, MALAT1 directly binds to MiR-145, which is negatively regulated. Overexpression of MALAT1 suppressed chondrocyte viability after IL-1β promotion and degraded extrachondral matrix, which was opposite to the effect results after miR-145 upregulation [45]. Besides, MALAT1 has been shown to regulate chondrocytes through the regulation of miR-515-3P or miR-181a-5p [46, 47]. There is another RNA worth mentioning called LncRNA snhg5, which was downregulated in OA and targeted to regulate H3 histone family 3B (H3F3B) expression through miR-10a-5p to enhance apoptosis caused by IL-1β [48]. It is also interesting to note that both SNHG5 and MALAT1 could protect stimulated chondrocytes with IL-1β by regulating miR-181a-5p, but the two target proteins were different [49]. In addition, biochemical analysis predicted high mobility group box 1 (HMGB1) was a target agent of miR-140-5p, and its overexpression reduced HMGB1, thereby suppressing inflammatory responses and apoptosis in IL-1β-treated chondrocytes [50]. In a study by Jing Wang et al., increased miR-98 expression was found in chondrocytes under IL-1β intervention, while inhibition of miR-98 effectively reduced cell apoptosis, suggesting that IL-1β regulated chondrocyte apoptosis-related proteins through mir-98 [51]. The above studies proved that IL-1β could affect chondrocyte matrix catabolism by affecting different proteins of interest with RNA targeting, and in the future, RNA regulation could be used as an entry point to provide a theoretical reference for clinical prevention and curing abnormal changes in chondrocytes.

Facilitation of adipogenesis

Among patients with orthopedic diseases, obese individuals deserve our attention for numerous studies have found that mediators of joint degeneration are derived from adipose paracrine signaling [52,53,54]. IL-1β plays a vital role on lipid metabolism via regulating lipase activity and negatively adjusts cartilage, there are reports of an association between IL-1β and obesity independent of population [55, 56]. In obese humans, adipocytokines, like leptin and adiponectin secretion by adipocytes is increased [57,58,59,60], which regulates inflammatory response, cartilage catabolic activity and bone remodeling, and is involved in the occurrence and development of obesity-induced osteoarthritis [61,62,63,64]. Leptin, an adipose tissue-derived adipokine with multiple immune and physiological functions [65], suppressing eating and increasing thermogenesis, and participates in multiple immune inflammatory responses [66]. T Simopoulou et al. demonstrated that leptin and its receptors were outstandingly enhanced at progressive stages, and the expression of leptin mRNA is closely related to BMI, moreover, IL-1β, MMP-9 and MMP-13 protein were also increased, with adverse effects on chondrocytes [67]. Adiponectin is another factor secreted by adipose tissue. Early studies established that adiponectin reduces cartilage extracellular matrix degradation and cartilage destruction resulting from increased MMP-13 induced by IL-1β [68]. In 2007, Simons P j et al. also found that adiponectin secretion was significantly downregulated by chronic exposure of adipocytes to IL-1β [69]. Later, T Delessa Challa et al. found that adiponectin (0.5 ug/ml) could promote mouse ATDC5 chondrocyte cell line proliferation and elevated the expression of collagen II, proteoglycan, Runx2, etc., demonstrating that low-level adiponectin effected positively on chondrocyte proliferation and differentiation [70]. It follows that leptin secretion is increased in obesity, bringing about pro-inflammatory effects, whereas adiponectin synthesis, which is anti-inflammatory, is decreased. While an in-depth study of its specific mechanism may be able to provide a new direction for us in the clinic for the treatment of obese patients with epiphyseal plate fractures as well as the prevention of complications.

What’s more, adipose tissue, made up of developing and mature adipocytes as well as a wide range of immune cells [71], though of a low-grade nature, is sufficient to cause negative effects on distant organ function [72]. Initially, obesity-related comorbidities were thought to dominate in diabetes mellitus type 2 (T2DM) due to growth of the global economy [73]. Obesity may lead to disturbed homeostasis between adipocytes and immune cells, and cause M1 macrophages fragmentation and polarization, activating the NLRP3 inflammatory to massively secrete IL-1β to exacerbate pro-inflammatory responses, a process that has been found to be involved in type 1 diabetes mellitus (T1DM) [74,75,76]. High levels of IL-1β, positively correlated with the severity of diabetes, confer insulin resistance in obese individuals [77, 78]. Evidence suggests that prolonged elevation of IL-1β promoted insulin expression, stimulated glucose uptake and aggravated macrophage inflammation, resulting in severe pathological metabolic processes [79]. In a study of the relationship between T2MD, obesity and skeleton, Francesca Vigevano et al. collected and analyzed data from 112 obese men and found that group with T2DM and obesity had more bone disease than those without T2DM [80]. This may indicate that high levels of IL in obesity-induced chronic inflammation leads to adverse skeletal reactions.

Since the twenty-first century, the problem of adolescent obesity has become increasingly severe, as a matter of fact, low-grade chronic inflammation leads to insulin resistance and diabetes, including type I diabetes mellitus(T1DM) [81]. Abnormal fat metabolism in obese children has become an early manifestation of diabetes, T1DM, also called insulin-dependent diabetes. Clinical statistics found that T1DM accounted for 5–10% of all diabetes cases and has a predilection for children or adolescents [82]. In general, T1DM is considered an immune disease [83], and the role of NLRP3 in T1DM remains to be investigated; whereas, recent studies point to a greater incidence of T1DM with obesity [73], possibly for that obesity leads to immunogenicity and glucose dysregulation, thereby contributing to insulin resistance in patients with T1DM. In addition, toll like receptors (TLRs), one pattern recognition molecule, which may be a biomarker in the early stage of T1DM, induce the production of IL-1β [84]. So further understanding of the role of IL-1β in T1DM may improve prognosis of fracture patients with juvenile T1DM. Earlier studies detected significantly higher IL-1β levels and lower insulin levels in patients with T1DM [84, 85] which correlated with IL-1β inducing pro-inflammatory factors migration to pancreatic islets and exerting cytotoxic effects [86], IL-1β reduced insulin-induced glucose transport in adipocytes, leading to lipid accumulation in muscle versus liver and deleterious effects. Studies have shown that skeletal and muscle health was affected by poorly controlled T1DM disease course in children [87].

It can be seen that obesity may cause an inflammatory response with elevated IL-1β levels, increasing the probability of developing diabetes while improving the risk of fracture and affecting fracture healing. Once the epiphyseal plate is injured, more IL-1β infiltration will increase the possibility of poor prognosis. In general, IL-1β is closely related to lipid and cartilage, especially in the context of the general increase of obese children, thus continued exploration of the relationship between adipocytes and chondrocytes and deeper investigation of obesity and bone diseases may in the future clinically provide a new direction for treating bone fractures in obese people, especially adolescents.

Promotion of oxidative stress

The chief reason for oxidative stress is disequilibrium between the generation of intracellular reactive oxygen species (ROS) and the scavenging effects of antioxidant ability, which is one main cause of chronic inflammation. In OA, oxidative stress has a certain influence on chondrocytes. Cellular ROS originates from mitochondrial respiratory chain [88], mainly produced by reduced NADPH oxidases (NOX) in chondrocytes [89], which usually exists in cells at a low level and is vital to maintain cell function and stability [90]. Previous studies have found that ROS oxidative stress make IL-1β and other inflammatory mediators highly up-regulated [91,92,93], which can induce the production of ROS and MMPs to degrade extracellular matrix [94, 95]. As a key link of classical inflammatory pathway, NF-κB participates in the regulation of a variety of genes, and it is a necessary molecule involved in the pathophysiological changes of cartilage [96].

Chondrocyte with hyperoside (Hyp) preconditioning saved ROS overproduction and chondrocyte apoptosis induced by IL-1β, playing an anti-inflammatory role by partially inhibiting NF-κB signaling pathway [97], so as simvastatin[98] and theobromine [99]. Garlic derived S-Allyl mercaptocysteine (SAMC) also has cartilage protection, but it mainly activates nuclear factor-E2 related factor (Nrf2), accompanied by downregulation of NOX4, resulting in improvement of collagen damage and maintenance of redox homeostasis [100]. At present, it is known that transcription factors that maintain cell redox balance and signal transduction can reduce intracellular oxidative stress damage [101]. Yao x et al. observed that Ferrostatin-1, a specific inhibitor of iron death, can reduce IL-1β-induced ROS accumulation, activate Nrf2 antioxidant system and rescue the expression of type II collagen [102]. Similarly, studies have found that a natural naphthoquinone compound β-hydroxyisovalerylshikonin (β-HIVs) can also inhibit IL-induced ROS production and chondrocyte metabolism through Nrf2, and downregulate the expression of ADAMTS5 and MMP13 [103]. Activation of nuclear receptor subfamily 1 group D member 1 (NR1D1) enhance Nrf2 pathway as well [104]. What’s more, Recent studies have shown that Licochalcone a (Lico a) can reduce the level of IL-1β and NLRP3 in vitro, and inhibit cell death via Nrf2/HO-1/NF-κB signal axis, to improve the degradation of cartilage extracellular matrix [105]. Normoline (NOM)-pretreated chondrocytes were also proved to inhibit NF-κB signal transduction by dissociating kelch like ECH associated protein 1 (Keap1) /Nrf2 path, effectively inhibiting inflammatory factor recruitment and ROS over regulation [106]. In conclusion, the weakening of NF-κB pathway and the activation of Nrf2 pathway can reduce IL-1β and ROS, reducing the degree of oxidative stress and protect chondrocytes.

It is worth mentioning, NOX4 is the only subtype expressed in chondrocytes in the NOX family, mainly cause ROS overproduction in chondrocytes after IL-1β stimulation [107, 108]. Besides, Heme oxygenase-1 (HO-1) is very important for NOX4 activity, which significantly downregulates the expression of MMP-1and NOX4 in IL-1β-treated chondrocytes [109]. Later studies further confirmed that IL-1β mediated NOX4 to stimulate the upregulation of MMP-1 and MMP-13 [110], which is consistent with the research results of Fu D et al. [111]. The regulation of oxidative stress and the relief of neuroinflammation may provide an effective reference for clinical treatment.

Conclusion

The repair of epiphyseal cartilage injury is regulated by many factors, and the inflammatory response is directly related to the growth, development and structural function of the repaired bone. Studies have shown that IL-1β levels are significantly elevated which can affect chondrocytes through a variety of ways, including the reduction of SOX-9 expression, the promotion of MMP synthesis, and further inhibit the interaction between proteoglycan and collagen II to achieve EMC degradation. In addition, IL-1β acts directly or indirectly on chondrocytes through lipids to promote lipolysis, resulting in abnormal levels of adiponectin and leptin, affecting lipid metabolism, and promoting the development of inflammation. In addition, IL-1β also causes irreversible damage to chondrocytes through multiple miRNA and circRNA targeting effects or enhancement of the response to oxidative stress, which has been effectively demonstrated in a variety of cellular or animal experiments and represents some of the advance in the understanding of the mechanisms of osteoarticular chondrocytes and growth plates repair. Further experimental work is also needed to deeply investigate the exact mechanism by which IL-1β affects growth plate chondrocytes to reduce or eliminate the effects of various harmful factors on the epiphyseal plate as early as possible to give new research directions in repair after epiphyseal plate injury.

Availability of data and materials

Not applicable.

References

  1. Marsell R, Einhorn TA. The biology of fracture healing. Injury. 2011;42(6):551–5.

    Article  Google Scholar 

  2. Gorter EA, Reinders CR, Krijnen P, et al. The effect of osteoporosis and its treatment on fracture healing a systematic review of animal and clinical studies. Bone Rep. 2021;15:101117.

    Article  CAS  Google Scholar 

  3. Claes L, Recknagel S, Ignatius A. Fracture healing under healthy and inflammatory conditions. Nat Rev Rheumatol. 2012;8(3):133–43.

    Article  CAS  Google Scholar 

  4. Dinarello CA. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev. 2018;281(1):8–27.

    Article  CAS  Google Scholar 

  5. Liu S, Deng Z, Chen K, et al. Cartilage tissue engineering: from proinflammatory and anti-inflammatory cytokines to osteoarthritis treatments (Review). Mol Med Rep. 2022. https://doi.org/10.3892/mmr.2022.12615.

    Article  Google Scholar 

  6. Zhou FH, Foster BK, Sander G, Xian CJ. Expression of proinflammatory cytokines and growth factors at the injured growth plate cartilage in young rats. Bone. 2004;35(6):1307–15.

    Article  CAS  Google Scholar 

  7. Cameron P, Limjuco G, Rodkey J, et al. Amino acid sequence analysis of human interleukin 1 (IL-1). Evidence for biochemically distinct forms of IL-1. J Exp Med. 1985;162(3):790–801.

    Article  CAS  Google Scholar 

  8. Migliorini P, Italiani P, Pratesi F, et al. The IL-1 family cytokines and receptors in autoimmune diseases. Autoimmun Rev. 2020;19(9):102617.

    Article  CAS  Google Scholar 

  9. Pietras EM, Mirantes-Barbeito C, Fong S, et al. Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal. Nat Cell Biol. 2016;18(6):607–18.

    Article  CAS  Google Scholar 

  10. Mitroulis I, Ruppova K, Wang B, et al. Modulation of myelopoiesis progenitors is an integral component of trained immunity. Cell. 2018;172(1–2):147-161.e112.

    Article  CAS  Google Scholar 

  11. Lopez-Rodriguez AB, Hennessy E, Murray CL, et al. Acute systemic inflammation exacerbates neuroinflammation in Alzheimer’s disease: IL-1β drives amplified responses in primed astrocytes and neuronal network dysfunction. Alzheimer’s Dementia. 2021;17(10):1735–55.

    Article  CAS  Google Scholar 

  12. Klück V, Liu R, Joosten LAB. The role of interleukin-1 family members in hyperuricemia and gout. Joint Bone Spine. 2021;88(2):105092.

    Article  Google Scholar 

  13. Evavold CL, Ruan J, Tan Y, et al. The pore-forming protein gasdermin D regulates interleukin-1 secretion from living macrophages. Immunity. 2018;48(1):35–44.

    Article  CAS  Google Scholar 

  14. Zhang J, Liu X, Wan C, et al. NLRP3 inflammasome mediates M1 macrophage polarization and IL-1beta production in inflammatory root resorption. J Clin Periodontol. 2020;47(4):451–60.

    Article  CAS  Google Scholar 

  15. Majcher S, Ustianowski P, Tarnowski M, et al. IL-1beta and IL-10 gene polymorphisms in women with gestational diabetes. J Matern Fetal Neonatal Med. 2021;34(19):3169–74.

    Article  CAS  Google Scholar 

  16. Yao M, Cui B, Zhang W, et al. Exosomal miR-21 secreted by IL-1beta-primed-mesenchymal stem cells induces macrophage M2 polarization and ameliorates sepsis. Life Sci. 2021;264:118658.

    Article  CAS  Google Scholar 

  17. Kiss M, Vande WL, Saavedra P, et al. IL1beta promotes immune suppression in the tumor microenvironment independent of the inflammasome and gasdermin D. Cancer Immunol Res. 2021;9(3):309–23.

    Article  CAS  Google Scholar 

  18. Yamashita F, Sakakida K, Kusuzaki K, et al. Immunohistochemical localization of interleukin 1 in human growth cartilage. Nihon Seikeigeka Gakkai Zasshi. 1989;63(5):562–8.

    CAS  Google Scholar 

  19. Marks PH, Donaldson ML. Inflammatory cytokine profiles associated with chondral damage in the anterior cruciate ligament-deficient knee. Arthroscopy. 2005;21(11):1342–7.

    Article  Google Scholar 

  20. Jenei-Lanzl Z, Meurer A, Zaucke F. Interleukin-1beta signaling in osteoarthritis—chondrocytes in focus. Cell Signal. 2019;53:212–23.

    Article  CAS  Google Scholar 

  21. MacRae VE, Farquharson C, Ahmed SF. The restricted potential for recovery of growth plate chondrogenesis and longitudinal bone growth following exposure to pro-inflammatory cytokines. J Endocrinol. 2006;189(2):319–28.

    Article  CAS  Google Scholar 

  22. Im GI, Kim HJ, Lee JH. Chondrogenesis of adipose stem cells in a porous PLGA scaffold impregnated with plasmid DNA containing SOX trio (SOX-5,-6 and -9) genes. Biomaterials. 2011;32(19):4385–92.

    Article  CAS  Google Scholar 

  23. Pritchett J, Athwal V, Roberts N, et al. Understanding the role of SOX9 in acquired diseases: lessons from development. Trends Mol Med. 2011;17(3):166–74.

    Article  CAS  Google Scholar 

  24. Akiyama H, Chaboissier MC, Martin JF, et al. The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev. 2002;16(21):2813–28.

    Article  CAS  Google Scholar 

  25. Cucchiarini M, Thurn T, Weimer A, et al. Restoration of the extracellular matrix in human osteoarthritic articular cartilage by overexpression of the transcription factor SOX9. Arthritis Rheum. 2007;56(1):158–67.

    Article  CAS  Google Scholar 

  26. Tew SR, Li Y, Pothacharoen P, et al. Retroviral transduction with SOX9 enhances re-expression of the chondrocyte phenotype in passaged osteoarthritic human articular chondrocytes. Osteoarthritis Cartilage. 2005;13(1):80–9.

    Article  Google Scholar 

  27. Sun X, Zhang J, Li Y, et al. Etomidate ameliorated advanced glycation end-products (AGEs)-induced reduction of extracellular matrix genes expression in chondrocytes. Bioengineered. 2021;12(1):4191–200.

    Article  CAS  Google Scholar 

  28. Ma C, Wu L, Song L, et al. The pro-inflammatory effect of NR4A3 in osteoarthritis. J Cell Mol Med. 2020;24(1):930–40.

    Article  CAS  Google Scholar 

  29. Xiong Y, Ran J, Xu L, et al. Reactivation of NR4A1 restrains chondrocyte inflammation and ameliorates osteoarthritis in rats. Front Cell Dev Biol. 2020;8:158.

    Article  Google Scholar 

  30. Zhu WJ, Chang BY, Wang XF, et al. FBW7 regulates HIF-1alpha/VEGF pathway in the IL-1beta induced chondrocytes degeneration. Eur Rev Med Pharmacol Sci. 2020;24(11):5914–24.

    Google Scholar 

  31. Xu Z, Ke T, Zhang Y, et al. Agonism of GPR120 prevented IL-1beta-induced reduction of extracellular matrix through SOX-9. Aging (Albany NY). 2020;12(12):12074–85.

    Article  CAS  Google Scholar 

  32. Fei J, Liang B, Jiang C, et al. Luteolin inhibits IL-1beta-induced inflammation in rat chondrocytes and attenuates osteoarthritis progression in a rat model. Biomed Pharmacother. 2019;109:1586–92.

    Article  CAS  Google Scholar 

  33. Chien SY, Tsai CH, Liu SC, et al. Noggin inhibits IL-1beta and BMP-2 expression, and attenuates cartilage degeneration and subchondral bone destruction in experimental osteoarthritis. Cells. 2020;9(4):927.

    Article  CAS  Google Scholar 

  34. Quan H, Dai X, Liu M, et al. Luteolin supports osteogenic differentiation of human periodontal ligament cells. BMC Oral Health. 2019;19(1):229.

    Article  Google Scholar 

  35. Balci YH, Toker H, Yildirim A, et al. The effect of luteolin in prevention of periodontal disease in Wistar rats. J Periodontol. 2019;90(12):1481–9.

    Article  Google Scholar 

  36. Tabeian H, Betti BF, Dos SCC, et al. IL-1beta damages fibrocartilage and upregulates MMP-13 expression in fibrochondrocytes in the condyle of the temporomandibular joint. Int J Mol Sci. 2019;20(9):2260.

    Article  CAS  Google Scholar 

  37. Yang Z, Gao XJ, Zhao X. CDMP1 promotes type II collagen and aggrecan synthesis of nucleus pulposus cell via the mediation of ALK6. Eur Rev Med Pharmacol Sci. 2020;24(21):10975–83.

    CAS  Google Scholar 

  38. Qi W, Ren D, Wang P, et al. Upregulation of Sirt1 by tyrosol suppresses apoptosis and inflammation and modulates extracellular matrix remodeling in interleukin-1beta-stimulated human nucleus pulposus cells through activation of PI3K/Akt pathway. Int Immunopharmacol. 2020;88:106904.

    Article  CAS  Google Scholar 

  39. Mével E, Merceron C, Vinatier C, et al. Olive and grape seed extract prevents post-traumatic osteoarthritis damages and exhibits in vitro anti IL-1β activities before and after oral consumption. Sci Rep. 2016;6:33527.

    Article  Google Scholar 

  40. Zhou X, Li J, Zhou Y, et al. Down-regulated ciRS-7/up-regulated miR-7 axis aggravated cartilage degradation and autophagy defection by PI3K/AKT/mTOR activation mediated by IL-17A in osteoarthritis[J]. Aging (Albany NY). 2020;12(20):20163–83.

    Article  CAS  Google Scholar 

  41. Zhou ZB, Huang GX, Fu Q, et al. circRNA.33186 contributes to the pathogenesis of osteoarthritis by sponging miR-127-5p. Mol Ther. 2019;27(3):531–41.

    Article  CAS  Google Scholar 

  42. Zheng Q, Bao C, Guo W, et al. Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging multiple miRNAs. Nat Commun. 2016;7:11215.

    Article  CAS  Google Scholar 

  43. Park SJ, Cheon EJ, Lee MH, et al. MicroRNA-127-5p regulates matrix metalloproteinase 13 expression and interleukin-1beta-induced catabolic effects in human chondrocytes. Arthritis Rheum. 2013;65(12):3141–52.

    Article  CAS  Google Scholar 

  44. Zhang Y, Wang F, Chen G, et al. LncRNA MALAT1 promotes osteoarthritis by modulating miR-150-5p/AKT3 axis. Cell Biosci. 2019;9:54.

    Article  Google Scholar 

  45. Liu C, Ren S, Zhao S, et al. LncRNA MALAT1/MiR-145 adjusts IL-1beta-induced chondrocytes viability and cartilage matrix degradation by regulating adamts5 in human osteoarthritis. Yonsei Med J. 2019;60(11):1081–92.

    Article  CAS  Google Scholar 

  46. Wang Y, Zhang Q. Long noncoding RNA MALAT1 knockdown inhibits proliferation, migration, and invasion and promotes apoptosis in non-small-cell lung cancer cells through regulating miR-515-3p/TRIM65 axis. Cancer Biother Radiopharm. 2020. https://doi.org/10.1089/cbr.2020.3730.

    Article  Google Scholar 

  47. Lu Z, Luo T, Pang T, et al. MALAT1 promotes gastric adenocarcinoma through the MALAT1/miR-181a-5p/AKT3 axis. Open Biol. 2019;9(9):190095.

    Article  CAS  Google Scholar 

  48. Jiang H, Pang H, Wu P, et al. LncRNA SNHG5 promotes chondrocyte proliferation and inhibits apoptosis in osteoarthritis by regulating miR-10a-5p/H3F3B axis. Connect Tissue Res. 2020;62:1–10.

    Google Scholar 

  49. Yue Y, Zhibo S, Feng L, et al. SNHG5 protects chondrocytes in interleukin-1beta-stimulated osteoarthritis via regulating miR-181a-5p/TGFBR3 axis. J Biochem Mol Toxicol. 2021;35:e22866.

    Article  CAS  Google Scholar 

  50. Wang Y, Shen S, Li Z, et al. MIR-140-5p affects chondrocyte proliferation, apoptosis, and inflammation by targeting HMGB1 in osteoarthritis. Inflamm Res. 2020;69(1):63–73.

    Article  CAS  Google Scholar 

  51. Wang J, Chen L, Jin S, et al. Altered expression of microRNA-98 in IL-1beta-induced cartilage degradation and its role in chondrocyte apoptosis. Mol Med Rep. 2017;16(3):3208–16.

    Article  CAS  Google Scholar 

  52. Collins KH, Lenz KL, Pollitt EN, et al. Adipose tissue is a critical regulator of osteoarthritis. Proc Natl Acad Sci. 2020;118(1):e2021096118.

    Article  Google Scholar 

  53. Kuroda K, Kabata T, Hayashi K, et al. The paracrine effect of adipose-derived stem cells inhibits osteoarthritis progression. BMC Musculoskelet Disord. 2015;16:236.

    Article  Google Scholar 

  54. Platas J, Guillén MI, Pérez Del Caz MD, et al. Paracrine effects of human adipose-derived mesenchymal stem cells in inflammatory stress-induced senescence features of osteoarthritic chondrocytes. Aging. 2016;8(8):1703–17.

    Article  CAS  Google Scholar 

  55. Matsuki T, Horai R, Sudo K, et al. IL-1 plays an important role in lipid metabolism by regulating insulin levels under physiological conditions. J Exp Med. 2003;198(6):877–88.

    Article  CAS  Google Scholar 

  56. Blaue D, Schedlbauer C, Starzonek J, et al. The influence of equine body weight gain on inflammatory cytokine expressions of adipose tissue in response to endotoxin challenge. Acta Vet Scand. 2020;62(1):17.

    Article  CAS  Google Scholar 

  57. Seth M, Biswas R, Ganguly S, et al. Leptin and obesity. Physiol Int. 2020;107(4):455–68.

    Article  CAS  Google Scholar 

  58. Mathias LS, Rodrigues BM, Goncalves BM, et al. Triiodothyronine activated extranuclear pathways upregulate adiponectin and leptin in murine adipocytes. Mol Cell Endocrinol. 2020;503:110690.

    Article  CAS  Google Scholar 

  59. Cartier A, Cote M, Lemieux I, et al. Age-related differences in inflammatory markers in men: contribution of visceral adiposity. Metabolism. 2009;58(10):1452–8.

    Article  CAS  Google Scholar 

  60. Gao YH, Zhao CW, Liu B, et al. An update on the association between metabolic syndrome and osteoarthritis and on the potential role of leptin in osteoarthritis. Cytokine. 2020;129:155043.

    Article  CAS  Google Scholar 

  61. Kroon F, Veenbrink AI, de Mutsert R, et al. The role of leptin and adiponectin as mediators in the relationship between adiposity and hand and knee osteoarthritis. Osteoarthritis Cartilage. 2019;27(12):1761–7.

    Article  CAS  Google Scholar 

  62. Xie C, Chen Q. Adipokines: new therapeutic target for osteoarthritis? Curr Rheumatol Rep. 2019;21(12):71.

    Article  CAS  Google Scholar 

  63. Min S, Shi T, Han X, et al. Serum levels of leptin, osteopontin, and sclerostin in patients with and without knee osteoarthritis. Clin Rheumatol. 2021;40(1):287–94.

    Article  Google Scholar 

  64. Cordero-Barreal A, Gonzalez-Rodriguez M, Ruiz-Fernandez C, et al. An update on the role of leptin in the immuno-metabolism of cartilage. Int J Mol Sci. 2021;22(5):2411.

    Article  CAS  Google Scholar 

  65. Moraes-Vieira PM, Bassi EJ, Araujo RC, et al. Leptin as a link between the immune system and kidney-related diseases: leading actor or just a coadjuvant? Obes Rev. 2012;13(8):733–43.

    Article  CAS  Google Scholar 

  66. Jequier E. Leptin signaling, adiposity, and energy balance. Ann N Y Acad Sci. 2002;967:379–88.

    Article  CAS  Google Scholar 

  67. Simopoulou T, Malizos KN, Iliopoulos D, et al. Differential expression of leptin and leptin’s receptor isoform (Ob-Rb) mRNA between advanced and minimally affected osteoarthritic cartilage; effect on cartilage metabolism. Osteoarthritis Cartilage. 2007;15(8):872–83.

    Article  CAS  Google Scholar 

  68. Chen TH, Chen L, Hsieh MS, et al. Evidence for a protective role for adiponectin in osteoarthritis. Biochim Biophys Acta. 2006;1762(8):711–8.

    Article  CAS  Google Scholar 

  69. Simons PJ, van den Pangaart PS, Aerts JM, et al. Pro-inflammatory delipidizing cytokines reduce adiponectin secretion from human adipocytes without affecting adiponectin oligomerization. J Endocrinol. 2007;192(2):289–99.

    Article  CAS  Google Scholar 

  70. Challa TD, Rais Y, Ornan EM. Effect of adiponectin on ATDC5 proliferation, differentiation and signaling pathways. Mol Cell Endocrinol. 2010;323(2):282–91.

    Article  CAS  Google Scholar 

  71. Dam V, Sikder T, Santosa S. From neutrophils to macrophages: differences in regional adipose tissue depots. Obes Rev. 2016;17(1):1–17.

    Article  CAS  Google Scholar 

  72. Kawai T, Autieri MV, Scalia R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am J Physiol Cell Physiol. 2021;320(3):C375–91.

    Article  CAS  Google Scholar 

  73. Polsky S, Ellis SL. Obesity, insulin resistance, and type 1 diabetes mellitus. Curr Opin Endocrinol Diabetes Obes. 2015;22(4):277–82.

    Article  CAS  Google Scholar 

  74. Soares J, Fernandes FP, Patente TA, et al. Gain-of-function variants in NLRP1 protect against the development of diabetic kidney disease: NLRP1 inflammasome role in metabolic stress sensing? Clin Immunol. 2018;187:46–9.

    Article  CAS  Google Scholar 

  75. Sun X, Xia Y, Liu Y, et al. Polymorphisms in NLRP1 gene are associated with type 1 diabetes. J Diabetes Res. 2019;2019:7405120.

    Article  Google Scholar 

  76. Orliaguet L, Ejlalmanesh T, Alzaid F. Metabolic and molecular mechanisms of macrophage polarisation and adipose tissue insulin resistance. Int J Mol Sci. 2020;21(16):5731.

    Article  CAS  Google Scholar 

  77. Vandanmagsar B, Youm YH, Ravussin A, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17(2):179–88.

    Article  CAS  Google Scholar 

  78. Esser N, L’Homme L, De Roover A, et al. Obesity phenotype is related to NLRP3 inflammasome activity and immunological profile of visceral adipose tissue. Diabetologia. 2013;56(11):2487–97.

    Article  CAS  Google Scholar 

  79. Dror E, Dalmas E, Meier DT, et al. Postprandial macrophage-derived IL-1β stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat Immunol. 2017;18(3):283–92.

    Article  CAS  Google Scholar 

  80. Vigevano F, Gregori G, Colleluori G, et al. In men with obesity, T2DM is associated with poor trabecular microarchitecture and bone strength and low bone turnover. J Clin Endocrinol Metab. 2021;106(5):1362–76.

    Article  Google Scholar 

  81. Kumar V, Kiran S, Kumar S, Singh UP. Extracellular vesicles in obesity and its associated inflammation. Int Rev Immunol. 2022;41(1):30–44.

    Article  CAS  Google Scholar 

  82. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2011;34:S62-69.

    Article  Google Scholar 

  83. Atkinson MA. The pathogenesis and natural history of type 1 diabetes. Cold Spring Harb Perspect Med. 2012;2(11):a007641.

    Article  Google Scholar 

  84. Dogan Y, Akarsu S, Ustundag B, et al. Serum IL-1beta, IL-2, and IL-6 in insulin-dependent diabetic children. Mediators Inflamm. 2006;2006(1):59206.

    Google Scholar 

  85. Hermann C, Krikovszky D, Fust G, et al. Association between interleukin-6 polymorphism and age-at-onset of type 1 diabetes. Epistatic influences of the tumor necrosis factor-alpha and interleukin-1beta polymorphisms. Eur Cytokine Netw. 2005;16(4):277–81.

    CAS  Google Scholar 

  86. Pang H, Luo S, Huang G, et al. Advances in knowledge of candidate genes acting at the beta-cell level in the pathogenesis of T1DM. Front Endocrinol (Lausanne). 2020;11:119.

    Article  Google Scholar 

  87. Dongare-Bhor S, Lohiya N, Maheshwari A, et al. Muscle and bone parameters in underprivileged Indian children and adolescents with T1DM. Bone. 2020;130:115074.

    Article  CAS  Google Scholar 

  88. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87(1):245–313.

    Article  CAS  Google Scholar 

  89. Schroder K. NADPH oxidase-derived reactive oxygen species: dosis facit venenum. Exp Physiol. 2019;104(4):447–52.

    Article  Google Scholar 

  90. Lismont C, Nordgren M, Van Veldhoven PP, et al. Redox interplay between mitochondria and peroxisomes. Front Cell Dev Biol. 2015;3:35.

    Article  Google Scholar 

  91. Shin HJ, Park H, Shin N, et al. p47phox siRNA-loaded PLGA nanoparticles suppress ROS/oxidative stress-induced chondrocyte damage in osteoarthritis. Polymers (Basel). 2020;12(2):443.

    Article  CAS  Google Scholar 

  92. Lou C, Deng A, Zheng H, et al. Pinitol suppresses TNF-alpha-induced chondrocyte senescence. Cytokine. 2020;130:155047.

    Article  CAS  Google Scholar 

  93. Zahan OM, Serban O, Gherman C, et al. The evaluation of oxidative stress in osteoarthritis. Med Pharm Rep. 2020;93(1):12–22.

    Google Scholar 

  94. Ansari MY, Ahmad N, Haqqi TM. Oxidative stress and inflammation in osteoarthritis pathogenesis: Role of polyphenols. Biomed Pharmacother. 2020;129:110452.

    Article  CAS  Google Scholar 

  95. Arra M, Swarnkar G, Ke K, et al. LDHA-mediated ROS generation in chondrocytes is a potential therapeutic target for osteoarthritis. Nat Commun. 2020;11(1):3427.

    Article  Google Scholar 

  96. Jimi E, Fei H, Nakatomi C. NF-kappaB signaling regulates physiological and pathological chondrogenesis. Int J Mol Sci. 2019;20(24):6275.

    Article  CAS  Google Scholar 

  97. Sun K, Luo J, Jing X, et al. Hyperoside ameliorates the progression of osteoarthritis: an in vitro and in vivo study. Phytomedicine. 2021;80:153387.

    Article  CAS  Google Scholar 

  98. Yu SM, Han Y, Kim SJ. Simvastatin abolishes nitric oxide- and reactive oxygen species-induced cyclooxygenase-2 expression by blocking the nuclear factor kappaB pathway in rabbit articular chondrocytes. Cell Biol Int. 2020;44(10):2153–62.

    Article  CAS  Google Scholar 

  99. Gu R, Shi Y, Huang W, et al. Theobromine mitigates IL-1beta-induced oxidative stress, inflammatory response, and degradation of type II collagen in human chondrocytes. Int Immunopharmacol. 2020;82:106226.

    Article  CAS  Google Scholar 

  100. Yang G, Sun S, Wang J, et al. S-allylmercaptocysteine targets Nrf2 in osteoarthritis treatment through NOX4/NF-kappaB pathway. Drug Des Devel Ther. 2020;14:4533–46.

    Article  CAS  Google Scholar 

  101. Yuan H, Xu Y, Luo Y, et al. Role of Nrf2 in cell senescence regulation. Mol Cell Biochem. 2021;476(1):247–59.

    Article  CAS  Google Scholar 

  102. Yao X, Sun K, Yu S, et al. Chondrocyte ferroptosis contribute to the progression of osteoarthritis. J Orthop Translat. 2021;27:33–43.

    Article  Google Scholar 

  103. Chen X, Gu M, Jin J, et al. beta-Hydroxyisovalerylshikonin inhibits IL-1beta-induced chondrocyte inflammation via Nrf2 and retards osteoarthritis in mice. Food Funct. 2020;11(11):10219–30.

    Article  CAS  Google Scholar 

  104. Liu H, Zhu Y, Gao Y, et al. NR1D1 modulates synovial inflammation and bone destruction in rheumatoid arthritis. Cell Death Dis. 2020;11(2):129.

    Article  CAS  Google Scholar 

  105. Yan Z, Qi W, Zhan J, et al. Activating Nrf2 signalling alleviates osteoarthritis development by inhibiting inflammasome activation. J Cell Mol Med. 2020;24(22):13046–57.

    Article  CAS  Google Scholar 

  106. Xue XH, Xue JX, Hu W, et al. Nomilin targets the Keap1-Nrf2 signalling and ameliorates the development of osteoarthritis. J Cell Mol Med. 2020;24(15):8579–88.

    Article  CAS  Google Scholar 

  107. Tiftik RN, Temiz-Resitoglu M, Guden DS, et al. Involvement of Rho-kinase/IkappaB-alpha/NF-kappaB activation in IL-1beta-induced inflammatory response and oxidative stress in human chondrocytes. Can J Physiol Pharmacol. 2021;99(4):418–26.

    Article  CAS  Google Scholar 

  108. Liu G, Liu Q, Yan B, et al. USP7 inhibition alleviates H2O2-induced injury in chondrocytes via inhibiting NOX4/NLRP3 pathway. Front Pharmacol. 2020;11:617270.

    Article  CAS  Google Scholar 

  109. Rousset F, Nguyen MV, Grange L, et al. Heme oxygenase-1 regulates matrix metalloproteinase MMP-1 secretion and chondrocyte cell death via Nox4 NADPH oxidase activity in chondrocytes. PLoS ONE. 2013;8(6):e66478.

    Article  CAS  Google Scholar 

  110. Rousset F, Hazane-Puch F, Pinosa C, et al. IL-1beta mediates MMP secretion and IL-1beta neosynthesis via upregulation of p22(phox) and NOX4 activity in human articular chondrocytes. Osteoarthritis Cartilage. 2015;23(11):1972–80.

    Article  CAS  Google Scholar 

  111. Fu D, Lu J, Yang S. Oleic/palmitate induces apoptosis in human articular chondrocytes via upregulation of NOX4 expression and ROS production. Ann Clin Lab Sci. 2016;46(4):353–9.

    CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to acknowledge Shengjing Hospital of China Medical University.

Funding

This work was supported by the National Natural Science Foundation of China (Grant Numbers: 8197081040).

Author information

Authors and Affiliations

Authors

Contributions

ZT contributed to relevant literature analysis, manuscript preparation, XY helped to write and revise the manuscript, JL provided research directions and writing ideas. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Jianjun Li.

Ethics declarations

Ethics approval and consent to participate

No ethics approval was required for this review that did not involve patients or patient data.

Consent for publication

We have obtained consents to publish this paper from all the participants of this study.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tong, Z., Yang, X. & Li, J. Research progress on the mechanism of interleukin-1β on epiphyseal plate chondrocytes. Eur J Med Res 27, 313 (2022). https://doi.org/10.1186/s40001-022-00893-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-022-00893-8

Keywords