Home Life Sciences Effect of cell receptors in the pathogenesis of osteoarthritis: Current insights
Article Open Access

Effect of cell receptors in the pathogenesis of osteoarthritis: Current insights

  • Li Lei , Li Meng , Xu Changqing , Zhu Chen , Yao Gang and Fang Shiyuan EMAIL logo
Published/Copyright: July 7, 2022

Abstract

Osteoarthritis (OA) is a chronic arthritic disease characterized by cartilage degradation, synovial inflammation, and subchondral bone lesions. The studies on the pathogenesis of OA are complex and diverse. The roles of receptors signaling in chondrocyte anabolism, inflammatory factors expression of synovial fibroblast, and angiogenesis in subchondral bone are particularly important for exploring the pathological mechanism of OA and clinical diagnosis and treatment. By reviewing the relevant literature, this article elaborates on the abnormal expression of receptors and the signaling transduction pathways from different pathological changes of OA anatomical components, aiming to provide new research ideas and clinical therapeutic value for OA pathogenesis.

1 Introduction

Osteoarthritis (OA) is one of the most common joint diseases threatening the health of middle-aged and elderly people worldwide, with main clinical manifestations such as joint swelling, pain, deformity, and dysfunction [1,2,3]. According to the statistics, the number of patients with OA in the United States has continued to rise during the past decade, with a forecast of 67 million by 2030. However, the number of patients with OA in China is not statistically comprehensive, but it is also increasing year by year, according to the current reports in various regions [4,5]. Therefore, the study and treatment of the pathogenesis of OA are still facing severe challenges. OA is not a simple process of cartilage degeneration, and its pathological changes almost involve all anatomical structures within the joint. Chondrocyte matrix catabolism disorder, synovial inflammatory response, abnormal subchondral bone remodeling, meniscal degeneration, inflammation, and fibrosis of the infrapatellar fat pad, and bone-chondral crosstalk and the new emerging morpho-functional unit of infrapatellar fat pad-synovial membrane act as crucial parts in OA progression [6,7,8,9]. The pathogenesis of OA has not been totally elucidated, and the current findings provide some new evidence for a comprehensive understanding of OA.

Receptors are macromolecular substances that can bind to ligands, including hormones, neurotransmitters, or intracellular signaling molecules, and induce specific changes in the target cell. Meanwhile, receptors are divided into cell-membrane receptors and intracellular receptors according to the position in cells, which can play important roles in the onset of diseases such as cardiovascular disease, tumorigenesis, and neurodegeneration, together with some mediated signal changes [10,11,12]. Thus, studies targeting exploring evolution are focused. This review aimed to clarify the effect of receptors on OA pathogenesis, and these studies provided evidence that cell receptors may be a potential target for OA treatment.

2 Methods

A PubMed search was performed for articles published between December 5, 1998 and September 02, 2022. Search terms were (OA AND receptor) OR (cartilage AND receptor) OR (synovium AND receptor) OR (subchondral bone AND receptor) OR (meniscus AND receptor) OR (infrapatellar fat pad AND receptor), with results limited to English language studies. Titles were checked and excluded if they paid attention to similar content topics. The impact of the study was highlighted in furthering novel areas of OA research and suggestions solicited from experts in the field.

2.1 Receptors and cartilage degeneration

The cartilage degeneration is the most important pathological feature of OA, which is often characterized by surface cracking, stripping, and thinning [13]. With mechanical pressure, the cartilage wear is serious, and the velocity of chondrocyte metabolism is slower than that of the catabolism. The imbalance of the two processes leads to cartilage degeneration and facilitates inflammatory reaction, cartilage matrix, and collagen degradation, resulting in joint pain and swelling [14]. The biological responses mediated by receptor signaling are particularly crucial in this process.

2.1.1 Inflammatory factor-associated receptors and cartilage degeneration

OA is inextricably linked to inflammation, while interleukin (IL) is an indispensable bioactive molecule in the inflammatory response. IL-1β is considered the most critical cytokine in OA initiation and progression [15]. The overexpression of IL-1β in OA cartilage can induce the cascade reaction of other proinflammatory factors and promote the synthetic release of metalloproteinases (MMPs), which can inhibit the normal anabolic response of chondrocytes and destroy the cartilage homeostasis [16,17]. IL-1 receptors include IL-1RI and IL-1RII, and a study has reported that the number of IL-1R in OA chondrocytes is twice as high as in normal cells. Thus, IL-1R and OA chondrocytes are more easily affinitive to exert biological effects [18]. The application of IL-1R antagonists in the OA rabbit model significantly alleviated the development of cartilage degradation, showing a promising approach for proteoglycan synthesis and cartilage repair [19]. Endogenous IL-1β binds to IL-1RI to form a receptor complex on the surface of chondrocytes membranes, as well as a high-affinity trimer structure combined with IL-1RAcP (IL-1β accessory protein). Meanwhile, IL-1RI and IL-1RAcP form heterodimeric structures and transduce signaling by cAMP, GTP, and other second messengers, activating IL-1-induced cartilage matrix degradation. A large number of transcription factors, chemokines, and protein kinases, like p38MAPK, NF-κB, and ERK1/2 signaling pathways, are also involved in this process to regulate apoptosis, abnormal differentiation, and oxidative stress in OA chondrocyte [20,21]. Not only that, but IL-1RⅡ signaling can also promote the secretion of IL-8, nitric oxide (NO), and Prostaglandin E2 (PGE2) in vitro induced by IL-1β [22]. Therefore, the research and drug development of targeted IL-1R are of great significance for OA treatment.

Tumor necrosis factor (TNF)-α is another kind of important cytokine affecting cartilage matrix metabolism, which can often be used to prepare the OA chondrocyte model in vitro. The upregulated MMP13 has been detected in OA chondrocytes through multiple cellular signaling pathways induced by TNF-α. Additionally, the high expression of TNF-α was observed in synovial fluid and serum, which accelerated the OA progression [23]. In particular, TNF-α and other proinflammatory factors were highly expressed with CD4+ T cell infiltration in the advanced OA [24]. TNF-RⅠ and TNF-RⅡ are different subtypes of TNF-α membrane receptors on the chondrocyte surface, whose expression is upregulated in OA. The signaling of binding TNF-α to TNF-RⅠ mediated the production of COX-2 (cyclooxygenase-2), eventually leading to the cartilage matrix degradation [25]. In fact, soluble receptors isoforms of TNF-R, which result from alternative splicing, include sTNF-RI and sTNF-RII. They were competitively bound to TNF-R on the cell surface with TNF-α, which can inhibit cartilage degeneration and delay the OA initiation. Other proinflammatory factors (i.e., IL-2, IL-5, IL-6, IL-8, IL-17, and IL-18) and anti-inflammatory factors (i.e., IL-4, IL-10, and IL-13) were also involved in the cartilage degeneration of OA. Especially, as a precursor inflammatory cytokine, the combination of IL-17 and IL-17R (IL-17 receptor) played an important role in regulating the balance of proinflammatory and proinflammatory factors in OA while inducing the overexpression of (inducible nitric oxide synthase) iNOS and NO to accelerate cartilage oxidation [26].

Factly, IL-1β and TNF-α are considered vital proinflammatory cytokines in the OA progression. The two mediators, produced partly by OA chondrocytes, induce the synthesis of a mass of inflammatory and catabolic factors [27,28]. Levels of IL-1β and TNF-α are prominently elevated in OA patients compared to those in healthy individuals [29]. Therefore, in most studies, appropriate concentrations of IL-1β or TNF-α are usually induced by chondrocytes to simulate OA in vitro [30,31]. In addition, the OA model could also be prepared by exposing primary chondrocytes to lipopolysaccharide or a conditioned medium of human-activated monocytes [32,33]. Notably, cytokines failed to model the complexity of the OA pathology in vivo, and it is only an acceptable research technique in vitro.

2.1.2 Growth factor-associated receptors and cartilage degeneration

Various cell growth factors are important mediators in regulating the formation of bone and cartilage, which can influence the occurrence of osteochondrogenesis in the embryo. The proliferation and differentiation of chondrocytes and cartilage homeostasis were positively or negatively regulated by these growth factors in OA development [34,35].

The transforming growth factor (TGF)-β signaling pathway superfamily includes many functional cytokines, such as bone morphogenetic proteins, which can stimulate chondrocyte proliferation, terminal differentiation, and extracellular matrix deposition [36]. TGF-β receptors are classified into TβRI, TβRII, and TβRIII types. The classical TβRI receptor activin receptor-like kinase 5 (ALK5) can phosphorylate the Smad 2/3 protein after combing with TGF-β to transmit the TGF-β signal from the cell surface to the nucleus, regulating the transcription of targeted genes and onsetting the initial differentiation of chondrocyte. However, a report has demonstrated that terminal differentiation on chondrocytes was regulated by Smad 1, 5, and 8 signaling pathways by ALK1 receptors [37]. Deleting ALK1, 5 can lead to the aging of chondrocytes and cartilage dysfunction, whose receptors were reduced during OA progression [38]. The decreased ratio of ALK5/ALK1 potentiated the terminal hypertrophy of OA chondrocyte, leading to proteoglycan decomposing with many soluble small molecules secreted to decompose extracellular matrix [39]. TGFβ receptor activation also promoted synovial inflammation and subchondral bone vascular infiltration, thereby promoting OA progression [40,41].

Vascular endothelial growth factor (VEGF) has a fundamental role in the proliferation, maturation, migration, and survival of vascular endothelial cells, stimulating the angiogenesis in tissues. Vascular infiltration is a vital pathological change in OA cartilage degeneration. During OA evolution, the new microvessels break through the articular tide line and invade the cartilage surface from the subchondral bone growth plate. Vascularization at the osteochondral unit is a key positive factor in OA progression [42]. The microangiogenesis allows the transport of MMPs, growth factors, and chemokines, which exposes articular cartilage to an inflammatory environment. Moreover, the finding of Zupan et al. has proved the positive role of VEGF in the OA development [43], and it is associated with the activation of MMP-1, MMP-3, MMP-9, and MMP-13 expressions in chondrocytes. The biological impact of VEGF was reflected through binding to the receptor tyrosine kinases of VEGFR-1 (Flt-1), VEGFR-2 (KDR/Flk-1), and VEGFR-3 (Flt-4) [44]. Both the upregulated expression of VEGFR-1 and VEGFR-2 in OA chondrocytes induce the release of MMPs and inhibit the expression of TIMPs (tissue MMP inhibitors), disrupting the intrinsic balance of these two processes and degrading the cartilage matrix [45]. A recent in vivo study has examined that VEGF induces vascularization mainly by coupling the VEGFR-2 on the surface of vascular endothelial cells, and the recruitment and activation of macrophages mediated by VEGFR-1 signaling also positively increased theVEGF secretion in macrophages and other angiogenesis-related cytokines to further stimulate vascularization [46]. Furthermore, the levels of phosphorylated VEGFR2 in chondrocytes have been contributors to OA development [47]. Therefore, targeted VEGF and its receptor signaling can be a new treatment in the field of OA therapy.

Insulin-like growth factor (IGF)-1 is a chemoattractant activator that regulates the chondroitin-glycan synthesis and the formation of the collagen network. IGF-1R, an IGF-1 receptor on the cartilage cell membrane, which the proteoglycans and type II collagen synthesis in a paracrine and autocrine manner, are mediated by activation, and the biological effect can inhibit cartilage matrix degradation and maintain the homeostasis of articular cartilage [48]. The elevated IGF-1 concentration in serum of OA patients is caused by the stimulation of IL-1 and TNF-α [49]. Additionally, the imbalance of synergistic relationship between IGF-1 and various cytokines such as TGF-β inhibits cartilage matrix synthesis. Meanwhile, IGF-1 binding protein (IGFBP) are also upregulated in OA, which competes with IGF-1 to bind to the IGF-1R on the chondrocyte surface, reducing the sensitivity of degenerative chondrocytes for IGF-1 and interfering with the synthetic process of cartilage matrix [50]. Therefore, IGF-I/IGFBP disorders may initiate and are involved in OA development. Correspondingly, a study that exogenous IGF-1 transfection in an extracorporeal cartilage defect model has indicated the unique biological function for repairing OA cartilage [51].

Fibroblast growth factor (FGF) is involved in the regulation of cell proliferation and differentiation during the growth and development of organisms, especially in the maintenance of osteochondrogenesis homeostasis [52]. FGF family contains at least 23 members, which transduce FGF signaling. Factly, FGF 2, 8, 9, and 18 have been shown to act relatively with OA progression in chondrocytes, and FGF receptors are divided into four types, including FGFR1, FGFR2, FGFR3, and FGFR4. FGF can bind to the extracellular domain of FGFR and induce phosphorylation of FGFR tyrosine residues. Next, activated FGFR recruits targeted proteins, which trigger downstream signaling pathways through phosphorylation, including RAS/MAPK, PI3K/AKT, phospholipase C, and protein kinase C signaling pathways. All FGFR subtypes are examined in adult articular chondrocytes, while the expressed levels of FGFR1 and FGFR3 are significantly higher than FGFR2 and FGFR4 in OA [53], suggesting that FGFR1 and FGFR3 signaling pathways participate in the process of chondrocyte anabolism. Studies have found that FGFR1 signaling promotes chondrocyte proliferation and ADAMTS5 (platelet-reactive protein integrin metallopeptidase 5) expression by binding FGF2 [54], which is particularly important for abnormal differentiation of chondrocytes. Moreover, the absence of FGFR3 contributes to cartilage degeneration and increased MMP13 expression, targeting the metabolic regulation of chondrocytes after binding to FGF2, 9, and 18 [55]. The effects of local articular injection of recombinant FGF18 on the repair of cartilage injury have also been confirmed, which has completed the phase II clinical trial [56].

The involvement of epidermal growth factor receptor (EGFR) signaling in the OA progression and cartilage homeostasis has been reported in previous studies [57,58]. TGF-α and HB-EGF (Heparin Binding EGF Like Growth Factor) are activators of EGFR signaling, and the mRNA of these two ligands is upregulated and expressed in OA articular cartilage. On the one hand, the high activity of EGFR signaling in OA can sustain cartilage homeostasis and lubricate cartilage surface by promoting the survival of chondrocytes, which can prevent cartilage from being injured in the early stage of OA, but the effect of contributing to chondrocytes proliferation showed little benefit [59]. On the other hand, EGFR signaling can also inhibit the synthesis of cartilage matrix proteins such as type II collagen and proteoglycan, which affect cartilage metabolism by blocking the expression of transcription factor Sox9 and induce the MMP13 upregulation in OA chondrocytes [60]. Therefore, the final role of EGFR signaling transduction in OA development is influenced by age, the OA stage, and so on. Sun et al. [61]. have confirmed that autophagy was also involved in activating EGFR signaling as a role in regulating OA chondrocyte phenotypic changes through the animal model with Egfr gene knockout. Intraarticular injection of gefitinib, an EGFR inhibitor, delays the degradation of cartilage matrix and provides a new clinical strategy for conservative treatment of OA.

2.1.3 Hormone-associated receptors and cartilage degeneration

Investigations have shown that women are at increased risk of OA occurrence after oophorectomy and natural menopause, suggesting that decreased estrogen levels are closely tied to OA onset [62]. The protective effect of estrogen replacement therapy on cartilage has also been highlighted in OA treatment [63]. The estrogen receptor (ER), which act as a major role in chondrocytes, regulates the maturation and development of chondrocytes by the combination of ER-α or ER-β with ER in chondrocytes. At the same time, Yazdi et al. [64] also drew a conclusion that the polymorphism sites Pvu II of ER-α genes were highly related to the susceptibility of female OA. The expression of ER was downregulated in OA chondrocytes. Selective estrogen receptor modulators significantly reduced the expression of MMP-2, MMP-3, MMP-13, and NO, which also inhibited OA chondrocyte apoptosis and the release of IL-1, TNF-α, COX-2, and promoted the synthesis of alkaline phosphatase, proteoglycan, collagen II, collagen X in the later stage of OA. Thus, these terminal outcomes positively regulate the synthesis and metabolism of the chondrocyte matrix to repair the OA cartilage [65,66].

Progesterone is another sex hormone that protects cartilage in OA. Progesterone receptors (PR) contain two nuclear receptors, PRA and PRB, as well as cell membrane receptors. The binding of progesterone to its nuclear receptor in OA chondrocytes inhibits some overexpressed inflammatory factors, including IL-1, TGF-β, and TNF-α induced by MMPs [67], thereby alleviating the cleavage of collagen II. PR signaling often exerts its functions through NF-κB (Nuclear Factor Kappa B), and the NF-κβp65 subunit is inversely associated with the interaction of progesterone receptor or progesterone-induced intranuclear translocation of Iκβ protein. Additionally, progesterone also inhibits iNOS, COX-2 mRNA expression, NO, and PGE2 production [68].

2.1.4 Toll-like receptors (TLRs) and cartilage degeneration

TLRs are a kind of pattern recognition receptors in non-specific immunity, which activate the inflammatory reaction and immune response by identifying the pathogen-related molecular patterns and endogenous damage-associated molecular patterns (DAMPs), and this innate immunity also is involved in OA lesions [69]. A report has assessed that activation of TLRs in human chondrocytes contributed to the OA development via NF-κB or JNK signaling pathway [70]. The study of Kim et al. [71] has consistently confirmed the significant increase of TLR2 and TLR4 expressions in OA cartilage tissue. TLR-dependent signaling transduction can motivate NF-κB, interferon regulatory factors (IRF), or MAPK, thereby leading to the excessive secretion of pro-inflammatory factors, chemokines, interferons, and adhesion molecules. The onset of TLR2 and TLR4 signaling triggers the expression of seven genes of IL-1 and MMPs, which stimulates NO and PGE2 overexpression and prevents cartilage matrix deposition [72]. The current study found that IL receptor 1 (IL-1R) has high homology with TLR4, which provides a stronger basis for exploring the mechanism of TLRs in OA. Both are mainly involved in OA by an inflammatory response and innate immune reaction by TLR/Myd88 (myeloid differentiation factor 88)/NF-κB signaling pathway [73].

On the other hand, the binding of DAMPs with TLRs facilitates sterile inflammation and triggers chondrocyte senescence, characterized by cartilage degradation and aging [74]. Meanwhile, TLR activation via Myd88 (TLR2, TLR7, TLR8, and TLR9), TRIF (TLR3), or both (TLR4) can induce chondrocytes apoptosis. Eventually, the chondrocyte proliferation is retardant, thereby leading to chondrocyte death, hypocellularity, and cartilage cracking [75].

2.1.5 Other types of receptors and cartilage degeneration

The mechanical load plays a critical role in the OA development, which activates a variety of inflammatory signaling pathways mediated by TNF-α, NF-κB, Wnt, TGF-β, and oxidative stress [76]. The stress-sensing receptors in cartilage were involved in regulating the process of the conversion of mechanical signals to electrical signals, which then release inflammatory factors. Transient receptor potential vanilloid 4 (TRPV4), an important ion channel in mechanical signal transduction, has been reported that its activation of chondrocytes mediated OA pain and facilitated chondrocytes apoptosis by inducing extracellular Ca2+ influx [77,78]. Recent studies have shown that TRPV4 is not only a potential therapeutic target for cartilage repair in OA but also a promising tool for the development of regenerative medicine, and the TRPV4 activation in chondrocytes promotes the formation of tissue-engineered constructs with improved mechanical properties without the bioreactors, especially cartilaginous tissue formation and mechanical properties [79,80].

As the major receptors of the extracellular matrix (ECM), the integrins can transduce biological information from the matrix to the cell [81]. Integrin binding of ECM ligands induces the formation of signaling complexes, which regulate the chondrocyte survival, adhesion, proliferation, differentiation and matrix remodeling [82]. The integrins α5β1, αVβ3, αVβ5, α6β1, α1β1, α2β1, and α10β1 were detected in chondrocytes. For example, integrin α10β1 is the main collagen type II binding receptor on chondrocytes, and a recent study from Delco ML et al. demonstrated that intra-articular administration of integrin a10β1-selected MSCs attenuates OA progression in an animal model [83].

The complex and diverse receptor signaling is involved in OA, some of which have not been fully elucidated but also play a vital role in the pathophysiology of OA. The peroxisome proliferator-activated receptor (PPAR), a kind of ligand-dependent transcription factor, belongs to the nuclear hormone receptor superfamily, which includes three subtypes of PPARα, PPARβ/δ, and PPARγ, and PPAR signaling mainly mediated the glucose, lipid metabolism and immune response [84]. The inhibition of PPARα by advanced glycation end products (AGEs) has been demonstrated to reduce the MMP-9 and TGF-β expressions in OA rabbit chondrocytes [85]. The PPARα activation was confirmed to reduce senescent cells and inflammatory response and increase autophagy in aging human OA chondrocytes [86]. Additionally, PPARβ/δ signaling activation promotes chondrogenic differentiation of mesenchymal stem cells [87]. PPAR-γ plays a role in anti-inflammatory and anti-chondral degeneration by inhibiting the NF-κB signaling pathway [88]. Therefore, activators of three subtypes of PPAR have emerged as potential therapeutic modalities for OA.

The renin-angiotensin system (RAS) activation has been gradually clarified in recent years, and this signaling system is composed of angiotensin I (Ang I), angiotensin enzyme (ACE), angiotensin II (Ang II), and angiotensin receptors, respectively. AT1R and AT2R act the opposite role in OA chondrocytes, of which the chondrocyte hypertrophy, cartilage matrix degeneration, and vascular infiltration are mediated by the former, as well as promoting the production of local RAS-related proteins, while the activation of the latter significantly downregulate the expression of IL-1β and IL-6 in OA and reduce the DNA binding activity to inhibit the inflammatory response [89]. OA rats treated with captopril (ACE inhibitor) showed the results of AT2R overproduction and downregulated expression of AT1R [90].

Moreover, vitamin D receptors (VDR) are involved in the metabolic activities of various cells during skeleton development and maturation, which is also closely related to the OA onset. A study has found that VDR BsmI polymorphism leads to changes in VDR structure, which induces vitamin D resistance in bone tissue and the disorder of calcium and phosphorus metabolism [91]. And abnormalities in VDR gene expression in damaged cartilage are associated with inflammatory response, which can also contribute to chondrocyte apoptosis in OA [92,93]. As a nuclear receptor, NR4A1 is a recently identified class of transcription factors highly correlated to the apoptosis in OA chondrocytes, expressing upregulation of translocation from the nucleus to mitochondria and inducing the release of cytochrome c, which participates in the process of TNF-induced chondrocytes apoptosis as a cofactor. Shi et al. also showed that mitochondrial apoptosis involved in TNF/p38MAPK/NR4A1 signaling pathway might be a vital link in the OA chondrocytes degradation [94].

2.2 Receptors and synovitis

Synovium is composed of matrix, cells, and fibers, with the smooth inner surface layer including 1∼3 layers of connective tissue cells. The main cellular components are fibroblasts, macrophages, mast cells, and adipocytes. Synovitis is another prominent pathological feature of OA. Activation and release of inflammatory factors induced by hyperplastic and hypertrophic synovium are the main causes of joint swelling, pain, and dysfunction [95]. Therefore, the mediation of receptor signaling also plays a crucial role in the inflammatory response of OA synovium.

2.2.1 Inflammatory factor-associated receptors and synovitis

Synovial inflammation response is more serious than cartilage. IL-1, IL-6, IL-7, IL-16, IL-17, IL-18, and TNF receptor signaling act similarly in synovioblasts and cartilage, which accelerates the release of inflammatory factors and regulates the synthetic expression of MMPs [96,97], further aggravating the cartilage degeneration. Among these, the interleukin-7 receptor (IL-7R) is a member of the type I cytokine receptor family, suggesting the down-regulated expression in OA synovial cells. The function of the IL-7/IL-7R signaling pathway is not clear in OA, which is mainly referred to as the inflammatory factors synthesis by sensitized T cells. It has also been reported that IL-7R-expressed B cells exert the effect of proinflammatory progression in collagen-induced arthritis [98].

2.2.2 Fat factor receptor and synovitis

The etiology of OA covers obesity, type Ⅱ diabetes, and lipase metabolism disorder, to name a few. Nowadays, a large number of studies focus on adipokines in OA pathogenesis, among which adiponectin, leptin, and resistin are more important [99]. The adiponectin is also found to be structurally homologous to complement C1q and TNF-α belonging to the C1q-TNF superfamily, and similar biological effects are displayed with TNF in OA [100]. The adiponectin receptors of AdipoR1 and AdipoR2 are found in OA synovial membrane, and the increased expression of adiponectin induces the synthesis of IL-6 and PGE2 and the release of MMP-1 through adipor1-mediated p38MAPK and NF-κB signaling pathways in synovial fibroblasts [101]. Besides, leptin is involved in a variety of inflammatory responses and is detected in OA synovial fluid. The results of reverse transcriptase-quantitative polymerase chain reaction have confirmed that long (OBRI) and short (OBRs) subtype receptors of leptin are overexpressed. The binding of leptin with OBRl can activate JAK2 (Janus kinase 2), which in turn phosphorylates tyrosine residues in the tail of the receptor. Thus, the recruitment of STAT-3 (signal transduction and transcriptional activator protein-3) is induced, which can also regulate tyrosine-phosphorylated IRS-1 (insulin receptor substrate 1) binding to the regulatory subunit p85 of PI3K, participating in the synthesis and release IL-8 in OA synovial fibroblasts [102]. Resistin is also upregulated in OA synovial tissues, which promotes the expression of MCP-1 (monocyte chemoattractant protein-1) and initiates the chemotaxis of monocytes to migrate across the skin to the site of inflammatory lesions. The release of MCP-1 also aggravates the progress of synovitis [103]. However, there are few studies on the receptor of resistin, so it is uncertain to determine the existence of its specific receptor. Nevertheless, resistin can bind to the TLR4 of the human leukocyte (THP-1) membrane surface and CAP1 (adenylate cyclase-associated protein) to activate the receptor signaling pathway and elevate the expressed levels of IL-1, IL-6, and TNF-α [104]. Additionally, it also can stimulate synovial fibroblasts to induce the phosphorylation of IGF-1R and AKT, thereby activating PI3K/AKT signaling pathway [105].

2.2.3 Bradykinin receptor B2 and synovitis

Bradykinin receptor B2 (BDKRB2) signaling has been demonstrated to mediate bradykinin-induced inflammatory responses, whereas bradykinin often exists in synovial fluid. BDKRB2 in OA synovial cells can enhance the production of proinflammatory factors, initiate the inflammatory reaction, stimulate nerve fibers, and improve nerve sensitivity, leading to joint pain, as well as the synthesis and release of NO, and strong vasodilation effects [106]. The study on the relationship between BDKRB2 gene polymorphism and OA severity indicated that BDKRB2 +9/−9 and other genetic polymorphisms promote inflammatory factors by inducing the TLR-2 expression, while BDKRB2 antagonist MEN16132 or TLR-2 knockdown inhibited the secretion of IL-6 and IL-8 in synovial fibroblasts [107].

2.2.4 AXL receptors and synovitis

AXL receptors are widely expressed in various tissues and cells, and GAS6 (growth arrest protein 6) is the only ligand for it. The proliferation and migration of cells are regulated by the binding of AXL receptors and GAS6. A study found that synovial fibroblast proliferation was activated, and the number of macrophages polarized toward M1 was increased. Upon stimulation of the obesity-derived macrophages by an inflammatory response, the AXL receptors were reduced, resulting in more severe inflammation in OA. The GAS6 synthesized by M2 macrophages binding to AXL receptors on M1 macrophages, inhibiting the M1-induced inflammation [108].

2.3 Receptors and subchondral bone lesions

Factly, subchondral bone undergoes abnormal bone resorption and remodeling in response to imbalanced load during the OA, which were mediated by osteoblast and osteoclast, respectively. Moreover, the crosstalk of bone cartilage is attributed to the formation of subchondral fissures, vascular invasion, and neurogenesis, suggesting a close association between aberrant subchondral bone remodeling and OA progression. Moreover, an increasing amount of studies confirmed that the TGF-β, Wnt, MAPK, and other signaling pathways are involved in this complicated process [109, 110]. Since subchondral bone cells are composed of osteoclasts, osteoblasts, bone cells, and bone marrow mesenchymal stem cells, there is little understanding of the underlying mechanism of cell surface receptors and nuclear receptors in OA. Nevertheless, some important receptors signaling transduction are known to provide new ideas for the study of OA pathogenesis.

2.3.1 Parathyroid hormone (PTH) receptor and subchondral bone lesions

PTH mainly regulates calcium and phosphorus metabolism to increase blood calcium and decrease blood phosphorus. The study has shown that PTH can delay the progress of OA, inhibit terminal differentiation of chondrocytes, protect cartilage matrix, and improve the subchondral bone remodeling [111]. PTH receptors are divided into three subtypes of PTH1R, PTH2R, and PTH3R, with the corresponding ligands of PTH, PTHrP (PTH-related peptide), and tuberoinfundibular peptide 39. PTH1R is mainly distributed in osteoblasts and osteocytes, playing the most critically biological role in the binding with PTH. Hilal et al. [112] confirmed that OA osteoblasts were resistant to PTH stimulation and PTHR expression was decreased in osteoblasts, but IGF-1 significantly inhibited the expression level of PTHR mRNA, which may clarify the causes of subchondral bone sclerosis and abnormal bone remodeling. PTH [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34] upregulates the PTH1R expression of chondrocytes and osteocytes, and PTH/PTH1R signaling pathway participates in repairing cartilage defects and increases the thickness of the subchondral bone plate in the treatment of rabbit cartilage defect model [113]. Additionally, PTH [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34] binding to PTH1R can inhibit Dkk-1 expression in osteoblasts [114]. Dkk-1 is an important negative regulator of the Wnt/β-catenin signaling pathway involved in OA cartilage destruction, which further proves the protective effect of the PTH/PTHR signaling pathway on OA.

2.3.2 Thyroid hormone receptor (THR) and subchondral bone lesions

The biological characteristics of thyroid hormones are mainly regulated by THRs encoded by the THRA and THRB genes and mediate many tissues’ growth and maturity, especially the process of skeletal development and remodeling. THRs are divided into two subtypes, THRα and THRβ. The relationship between THR and OA remains unclear. Our findings have revealed that the upregulated expression of THRs in OA osteoblasts due to the abnormal proliferation of osteoblasts and THRα knockdown repressed angiogenesis in subchondral bone, thereby alleviating OA progression [115]. The microvasculature formation of the bone-cartilage unit allows the transport of detrimental small molecules and inflammatory mediators.

2.3.3 ER and subchondral bone lesions

Estrogen replacement therapy is primarily used in postmenopausal-related diseases. The biological effect of ER combined with its receptor (ER) not only represses the degeneration of OA articular cartilage but also reduces the conversion rate of subchondral bone, as well as inhibition of bone resorption mediated by osteoclasts [66]. Sniekers et al. [116] confirmed that ER was examined in osteoblasts and osteoclasts, and the knockout of ER genes expedited osteophyte formation and thinned subchondral bone plates through micro-CT analysis. Therefore, selective ER agonists can contribute to relieving OA symptoms and pathological evolution.

The role of all of the above receptors is not simple in the occurrence and development of OA. For example, adiponectin and leptin receptors are also upregulated expression in OA chondrocytes to mediate cartilage degradation. ER agonists also inhibit the release of OA chondrocyte inflammatory factors. Factly, TGF-β receptor signaling facilitates matrix degradation in OA chondrocytes and represses bone loss and vascularization in the subchondral bone. The inflammatory factor of TNF-α also enables osteoclast activation to promote bone resorption and inhibit collagen I synthesis. Numerous growth factor receptors are also involved in regulating aberrant bone remodeling in OA osteoblasts. In addition, the investigation of subchondral bone marrow mesenchymal stem cells is of great value for elucidating OA pathological changes and cartilage repairment in the future [117,118].

Tables 1 and 2, respectively, summarize the related studies demonstrating the association between different cell receptors and OA progression in human and OA animal models. Figure 1 illustrates the role of receptors in cartilage degeneration and synovium inflammation, as well as the angiogenesis of subchondral bone.

Table 1

Different cell receptors are correlated with pathological changes in human OA tissues

Receptors Changes in OA tissue Ref.
“↓” Inhibition “↑” Promotion
IL-1R ↑ Cartilage degradation [16,17,18,20,21,22]
↑ Synovits [96,97]
TNF-R ↑ Cartilage degradation [23,25]
↑ Synovits [96,97]
TβR ↑ Cartilage degradation [37,38,39]
↑ Synovits [40]
↑ Vascular invasion in subchondral bone [41]
VEGFR ↑ Cartilage degradation [43,45]
↑ Vascular invasion in subchondral bone [43]
IGF-1R ↓Cartilage degradation [48,49,51]
FGFR ↓ Cartilage degradation [53,54,55]
EGFR ↓ Cartilage degradation (Early OA) [59]
↑ Cartilage degradation (Advanced OA) [60]
ER ↓ Cartilage degradation [64,65,66]
↓ Osteophyte formation [66,116]
PR ↓ Cartilage degradation [67,68]
TLR ↑ Cartilage degradation [70,71,72,73,74]
TRPV4 ↓ Cartilage degradation [80]
Integrin ↓ Cartilage degradation [82]
PPARα ↓ Cartilage degradation [86]
VDR ↓ Cartilage degradation [92,93]
NR4A1 ↑ Cartilage degradation [94]
AdipoR ↑ Synovits [101]
OBRI/OBRs ↑ Synovits [102]
BDKRB2 ↑ Synovits [106,107]
AXLR ↑ Synovits [108]
PTHR ↓ Cartilage degradation [111]
↓ Subchondral bone lesion [111,112]
THRα ↑ Vascular invasion in subchondral bone [115]
Table 2

Different cell receptors are correlated with pathological changes in OA animal model

Receptors Changes in OA tissue Ref.
“↓ Inhibition “↑ ” Promotion
IL-1R ↑ Cartilage degradation (rabbit) [19]
IL-17R ↑ Cartilage degradation (mice) [26]
VEGFR ↑ Cartilage degradation (mice) [47]
↑ Synovits (mice) [47]
IGF-1R ↓ Cartilage degradation (mice) [51]
EGFR ↑ Cartilage degradation (mice) [61]
TRPV4 ↑ OA pain (rat) [77]
↑ Cartilage degradation (mice) [78]
Integrin α10β1 ↓ Cartilage degradation (calve) [79]
PPARα ↓ Cartilage degradation (rabbit) [85]
PPARγ ↓ Cartilage degradation (mice) [88]
AT1R ↑ Cartilage degradation (rat) [90]
AT2R ↓ Cartilage degradation (rat) [90]
PTHR ↓ Cartilage degradation (rabbit) [113]
Figure 1 
                     Different roles of receptors in cartilage degeneration and synovium inflammation, as well as the pathological changes of subchondral bone in OA.
Figure 1

Different roles of receptors in cartilage degeneration and synovium inflammation, as well as the pathological changes of subchondral bone in OA.

2.4 OA treatment targeting receptors

For many years, strong and cost-intensive efforts have been undertaken to develop therapies to improve care, quality of life, and pain relief for OA patients. A variety of clinical studies targeting receptors have also emerged in OA treatment. Due to the vital effect of IL-1 in OA pathogenesis, a clinical trial of intraarticular injections of recombinant human IL-1 receptor antagonist (anakinra) in patients with knee OA was safe without inflammatory reaction [119]. Additionally, the Food and Drug Administration-approved drug gefitinib could efficiently inhibit EGFR activation in OA and restore cartilage structure and matrix synthesis in the mouse model [61]. Hydroxychloroquine (HC), a chloroquine derivative, exerts its effects via TLR7/9, which may interfere with signaling molecules to inhibit inflammation in OA [120]. Fenofibrate, a PPARα agonist, could reduce the number of senescent cells via apoptosis, increase the autophagic flux, and protect against cartilage degradation in clinical OA treatment [86]. Apart from inflammatory inhibition in OA joints, pain management is a dominant treatment strategy for OA. It is well known that opioid receptor antagonists, such as tramadol, are commonly used for OA pain relief. As the selective agonist of Kappa-opioid receptor, CR845, the dose-dependent pain reduction was observed in hip and knee OA patients [121]. New therapy method to inhibit nerve growth factor (NGF)-induced pain concentrate on the antagonization of its receptors tropomyosin-related kinase A (TrkA) and p75NTR, and Pan Trk inhibitor GZ389988 has represented the short-term moderate pain reduction in knee OA [122]. In clinical trials, trans-capsaicin (CNTX-4975) can reduce moderate-to-severe OA by blocking TRPV1 channels expressed in sensory neurons [123]. Moreover, FGFR3 was proved that exerts the protective effects in OA by activating RAS-MAPK and PI3K-AKT signaling pathways [124]. FGF18 is the high-affinity ligand of FGFR3 and the only FGF-based drug currently in clinical trials for OA treatment [125]. The intra-articular application of sprifermin (recombinant human FGF18) can increase cartilage thickness and inhibit ECM degradation without discernible local or systemic reverse reaction [126]. A report has revealed that the use of low-dose teriparatide (PTH 1-34) contributed to cartilage repair and improved subchondral bone microstructure in OA [111]. Table 3 summarizes the related pre-clinical/clinical studies focused on the use of treatments targeting the receptors. Therefore, given the essential but complex roles of receptor signaling in the maintenance of articular cartilage, targeting the receptors system to treat OA is a promising direction in the future.

Table 3

Current OA treatments targeting receptors

Receptors Targeting receptors Biological effect Ref.
“↓” Inhibitor “↑ ” Activator “↓” Inhibition
Anakinra ↓ IL-1R ↓ Cartilage degradation [119]
↓ Synovits
↓ OA pain
Gefitibib ↑ EGFR ↓ Cartilage degradation [61]
HC ↑ TLR7/9 ↓ Cartilage degradation [120]
Tenofibrate ↑ PPRAα ↓ Cartilage degradation [86]
CR845 ↑ Kappa-opioid receptor ↓ OA pain [121]
GZ389988 ↓ TrkA/p75NTR ↓ OA pain [122]
CNTX-4975 ↓ TRPV1 ↓ OA pain [123]
Sprifermin (FGF18) ↑ FGFR3 ligand ↓ Cartilage degradation [125,126]
Teriparatide (PTH [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34]) ↑ PTHR ↓ Cartilage degradation [111]
↓ Subchondral bone lesion

3 Conclusion

The researchers on OA are trying to explore the essential link of its pathogenesis, whether from the molecular level or from the current genomics and proteomics, to formulate targeted and precise prevention and treatment programs. With the transformation of research results, conservative treatment approaches of OA have been gradually applied to clinical practice and achieved certain effects. Notably, promising advances in cartilage repair therapy were focused on the field of regenerative medicine in large animal models [127]. The injection of allogeneic mesenchymal stem cells showed excellent clinical efficacy and pain relief in horse degenerative joint disease [128]. This provides valuable experimental evidence for the clinical treatment of OA in the future. However, the medical treatment and gene therapy for OA based on the significant pathological changes such as cartilage, synovial membrane, subchondral bone, and osteophyte should not be isolated but attenuate the progression of two or more of anatomical components in OA at least so that the drug efficacy can be confirmed. In conclusion, the exploration of OA pathogenesis and the replacement of clinical treatment strategies have always been a great challenge for medical workers, but the individualization and comprehensive treatment for OA will become the main research direction. Of course, there is no doubt that what has to be considered is the role of receptors in OA.


tel: +86-15856957697

Acknowledgements

This study was supported by the Fundamental Research Funds for the Central Universities (No. WK9110000178).

  1. Funding information: Authors state no funding involved.

  2. Conflict of interest: Authors state no conflict of interest.

  3. Data availability statement: Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

[1] Nelson AE, Allen KD, Golightly YM, Goode AP, Jordan JM. A systematic review of recommendations and guidelines for the management of osteoarthritis: the chronic osteoarthritis management initiative of the US bone and joint initiative. Semin Arthritis Rheum. 2014;43:701–12.10.1016/j.semarthrit.2013.11.012Search in Google Scholar

[2] Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380:2163–96.10.1016/S0140-6736(12)61729-2Search in Google Scholar

[3] Bunzli S, O'brien P, Ayton D, Dowsey M, Gunn J, Choong P, et al. Misconceptions and the acceptance of evidence-based nonsurgical interventions for knee osteoarthritis. A qualitative study. Clin Orthop Relat Res. 2019;477:1975–83.10.1097/CORR.0000000000000784Search in Google Scholar

[4] Tang X, Wang S, Zhan S, Niu J, Tao K, Zhang Y, et al. The prevalence of symptomatic knee osteoarthritis in China: Results from the china health and retirement longitudinal study. Arthritis Rheumatol. 2016;68:648–53.10.1002/art.39465Search in Google Scholar

[5] Hunter DJ, March L, Chew M. Osteoarthritis in 2020 and beyond: a lancet commission. Lancet. 2020;396:1711–2.10.1016/S0140-6736(20)32230-3Search in Google Scholar

[6] Onuora S. Osteoarthritis: Cartilage matrix stiffness regulates chondrocyte metabolism and OA pathogenesis. Nat Rev Rheumatol. 2015;9:504.10.1038/nrrheum.2015.107Search in Google Scholar

[7] Scanzello CR, Goldring SR. The role of synovitis in osteoarthritis pathogenesis. Bone. 2012;51:249–57.10.1016/j.bone.2012.02.012Search in Google Scholar PubMed PubMed Central

[8] Aaron RK, Racine J, Dyke JP. Contribution of circulatory disturbances in subchondral bone to the pathophysiology of osteoarthritis. Curr Rheumatol Rep. 2017;19:49.10.1007/s11926-017-0660-xSearch in Google Scholar PubMed

[9] Macchi V, Stocco E, Stecco C, Belluzzi E, Favero M, Porzionato A, et al. The infrapatellar fat pad and the synovial membrane: an anatomo-functional unit. J Anat. 2018;233:146–54.10.1111/joa.12820Search in Google Scholar PubMed PubMed Central

[10] Yao C, Veleva T, Scott L Jr, Cao S, Li L, Chen G, et al. Enhancedcardiomyocyte NLRP3 inflammasome signaling promotes atrial fibrillation. Circulation. 2018;138:2227–42.10.1161/CIRCULATIONAHA.118.035202Search in Google Scholar PubMed PubMed Central

[11] Kuo WT, Lee TC, Yu LC. Eritoran suppresses colon cancer by altering a founctional balance in Toll-like receptors that bind lipopolysaccharide. Cancer Res. 2016;76:4684–95.10.1158/0008-5472.CAN-16-0172Search in Google Scholar PubMed

[12] Ghezzi L, Carandini T, Arighi A, Fenoglio C, Arcaro M, De Riz M, et al. Evidence of CNS β-amyloid deposition in Nasu-Hakola disease due to the TREM2 Q33X mutation. Neurology. 2017;89:2503–5.10.1212/WNL.0000000000004747Search in Google Scholar PubMed

[13] Mobasheri A, Batt M. An update on the pathophysiology of osteoarthritis. Ann Phys Rehabil Med. 2016;59:333–9.10.1016/j.rehab.2016.07.004Search in Google Scholar PubMed

[14] Abramoff B, Caldera FE. Osteoarthritis: Pathology, diagnosis, and treatment options. Med Clin North Am. 2020;104:293–311.10.1016/j.mcna.2019.10.007Search in Google Scholar PubMed

[15] Wojdasiewicz P, Poniatowski ŁA, Szukiewicz D. The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm. 2014;2014:561459.10.1155/2014/561459Search in Google Scholar PubMed PubMed Central

[16] Wang F, Liu J, Chen X, Zheng X, Qu N, Zhang B, et al. IL-1beta receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes. Arthritis Res Ther. 2019;21:171.10.1186/s13075-019-1952-5Search in Google Scholar PubMed PubMed Central

[17] Ding QH, Cheng Y, Chen WP, Zhong HM, Wang XH. Celastrol, an inhibitor of heat shock protein 90β potently suppresses the expression of matrix metalloproteinases, inducible nitric oxide synthase and cyclooxygenase-2 in primary human osteoarthritic chondrocytes. Eur J Pharmacol. 2013;708:1–7.10.1016/j.ejphar.2013.01.057Search in Google Scholar PubMed

[18] Murata M, Trahan C, Hirahashi J, Mankin HJ, Towle CA. Intracellular interleukin-1 receptor antagonist in osteoarthritis chondrocytes. Clin Orthop Relat Res. 2003;409:285–95.10.1097/01.blo.0000059582.08469.acSearch in Google Scholar PubMed

[19] Zhao R, Wang S, Jia L, Li Q, Qiao J, Peng X. Interleukin-1 receptor antagonist protein (IL-1Ra) and miR-140 overexpression via pNNS-conjugated chitosan-mediated gene transfer enhances the repair of full-thickness cartilage defects in a rabbit model. Bone Jt Res. 2019;8:165–78.10.1302/2046-3758.83.BJR-2018-0222.R1Search in Google Scholar PubMed PubMed Central

[20] Sun HY, Hu KZ, Yin ZS. Inhibition of the p38-MAPK signaling pathway suppresses the apoptosis and expression of proinflammatory cytokines in human osteoarthritis chondrocytes. Cytokine. 2017;90:135–43.10.1016/j.cyto.2016.11.002Search in Google Scholar PubMed

[21] Chen X, Wang Y, Qu N, Zhang B, Xia C. PLCgamma1 inhibition-driven autophagy of IL-1beta-treated chondrocyte confers cartilage protection against osteoarthritis, involving AMPK, Erk and Akt. J Cell Mol Med. 2021;25:1531–45.10.1111/jcmm.16245Search in Google Scholar PubMed PubMed Central

[22] Grieshaber-Bouyer R, Kämmerer T, Rosshirt N, Nees TA, Koniezke P, Tripel E, et al. Divergent mononuclear cell participation and cytokine release profiles define hip and knee osteoarthritis. J Clin Med. 2019;8:1631.10.3390/jcm8101631Search in Google Scholar

[23] Ene R, Sinescu RD, Ene P, Cîrstoiu MM, Cîrstoiu FC. Synovial inflammation in patients with different stages of knee osteoarthritis. Rom J Morphol Embryol. 2015;56:169–73.Search in Google Scholar

[24] Hussein MR, Fathi NA, El-Din AME, Hassan HI, Abdullah F, AL-Hakeem E, et al. Alterations of the CD4 +, CD8 + T cell subsets, interleukins-1β, IL-10, IL-17, tumor necrosis factor-α and soluble intercellular adhesion molecule-1 in rheumatoid arthritis and osteoarthritis: preliminary observations. Pathol Oncol Res. 2008;14:321–8.10.1007/s12253-008-9016-1Search in Google Scholar

[25] Webb GR, Westacott CI, Elson CJ. Chondrocyte tumor necrosis factor receptors and focal loss of cartilage in osteoarthritis. Osteoarthr Cartil. 1997;5:427–37.10.1016/S1063-4584(97)80047-7Search in Google Scholar

[26] Na HS, Park JS, Cho KH, Kwon JY, Choi J, Jhun J, et al. Interleukin-1-Interleukin-17 signaling axis induces cartilage destruction and promotes experimental osteoarthritis. Front Immunol. 2020;5(11):730.10.3389/fimmu.2020.00730Search in Google Scholar PubMed PubMed Central

[27] Wojdasiewicz P, Poniatowski ŁA, Szukiewicz D. The role of inflammatory and anti-inflammatory cytokines in the pathogenesis of osteoarthritis. Mediators Inflamm. 2014;2014:561459.10.1155/2014/561459Search in Google Scholar PubMed PubMed Central

[28] Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7:33–42.10.1038/nrrheum.2010.196Search in Google Scholar PubMed

[29] Wang T, He C. Proinflammatory cytokines: The link between obesity and osteoarthritis. Cytokine Growth Factor Rev. 2018;44:38–50.10.1016/j.cytogfr.2018.10.002Search in Google Scholar PubMed

[30] Jenei-Lanzl Z, Meurer A, Zaucke F. Interleukin-1beta signaling in osteoarthritis- chondrocytes in focus. Cell Signal. 2019;53:212–23.10.1016/j.cellsig.2018.10.005Search in Google Scholar PubMed

[31] Chen-An P, Andreassen KV, Henriksen K, Karsdal MA, Bay-Jensen AC. Investigation of chondrocyte hypertrophy and cartilage calcification in a full-depth articular cartilage explants model. Rheumatol Int. 2013;33:401–11.10.1007/s00296-012-2368-6Search in Google Scholar PubMed

[32] Yan Z, Qi W, Zhan J, Lin Z, Lin J, Xue X, et al. Activating Nrf2 signalling alleviates osteoarthritis development by inhibiting inflammasome activation. J Cell Mol Med. 2020;24:13046–57.10.1111/jcmm.15905Search in Google Scholar PubMed PubMed Central

[33] Tarricone E, Mattiuzzo E, Belluzzi E, Elia R, Benetti A, Venerando R, et al. Anti-inflammatory performance of lactose-modified chitosan and hyaluronic acid mixtures in an in vitro macrophage- mediated inflammation osteoarthritis model. Cells. 2020;9:1328.10.3390/cells9061328Search in Google Scholar PubMed PubMed Central

[34] Huang J, Zhao L, Chen D. Growth factor signalling in osteoarthritis. Growth Factors. 2018;36:187–95.10.1080/08977194.2018.1548444Search in Google Scholar PubMed PubMed Central

[35] Hayes AJ, Whitelock J, Melrose J. Regulation of FGF-2, FGF-18 and transcription factor activity by perlecan in the maturational development of transitional rudiment and growth plate cartilages and in the maintenance of permanent cartilage homeostasis. Int J Mol Sci. 2022;23:1934.10.3390/ijms23041934Search in Google Scholar PubMed PubMed Central

[36] Thielen NGM, van der Kraan PM, van Caam APM. TGFβ/BMP signaling pathway in cartilage homeostasis. Cells. 2019;8:969–98.10.3390/cells8090969Search in Google Scholar PubMed PubMed Central

[37] van der Kraan PM. Age-related alterations in TGF beta signaling as a causal factor of cartilage degeneration in osteoarthritis. Biomed Mater Eng. 2014;24:75–80.10.3233/BME-140976Search in Google Scholar PubMed

[38] van der Kraan PM, Blaney Davidson EN, van den Berg WB. A role for age-related changes in TGFbeta signaling in aberrant chondrocyte differentiation and osteoarthritis. Arthritis Res Ther. 2010;12:201.10.1186/ar2896Search in Google Scholar PubMed PubMed Central

[39] Blaney Davidson EN, Remst DF, Vitters EL, van Beuningen HM, Blom AB, Goumans MJ, et al. Increase in ALK1/ALK5 ratio as a cause for elevated MMP-13 expression in osteoarthritis in humans and mice. J Immunol. 2009;182:7937–45.10.4049/jimmunol.0803991Search in Google Scholar PubMed

[40] Baugé C, Girard N, Lhuissier E, Bazille C, Boumediene K. Regulation and role of TGFbeta signaling pathway in aging and osteoarthritis joints. Aging Dis. 2013;5:394–405.Search in Google Scholar

[41] Zhen G, Wen C, Jia X, Li Y, Crane JL, Mears SC, et al. Inhibition of TGF-beta signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nat Med. 2013;19:704–12.10.1038/nm.3143Search in Google Scholar PubMed PubMed Central

[42] Yuan XL, Meng HY, Wang YC, Peng J, Guo QY, Wang AY, et al. Bone-cartilage interface crosstalk in osteoarthritis: potential pathways and future therapeutic strategies. Osteoarthr Cartil. 2014;22:1077–89.10.1016/j.joca.2014.05.023Search in Google Scholar PubMed

[43] Zupan J, Vrtačnik P, Cör A, Haring G, Weryha G, Visvikis-Siest S, et al. VEGF-A is associated with early degenerative changes in cartilage and subchondral bone. Growth Factors. 2018;36:263–73.10.1080/08977194.2019.1570926Search in Google Scholar PubMed

[44] Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling-in control of vascular function. Nat Rev Mol Cell Biol. 2006;7:359–71.10.1038/nrm1911Search in Google Scholar

[45] Enomoto H, Inoki I, Komiya K, Shiomi T, Ikeda E, Obata K, et al. Vascular endothelial growth factor isoforms and their receptors are expressed in human osteoarthritic cartilage. Am J Pathol. 2003;162:171–81.10.1016/S0002-9440(10)63808-4Search in Google Scholar

[46] Hamilton JL, Nagao M, Levine BR, Chen D, Olsen BR, Im HJ. Targeting VEGF and its receptors for the treatment of osteoarthritis and associated pain. J Bone Min Res. 2016;31:911–24.10.1002/jbmr.2828Search in Google Scholar PubMed PubMed Central

[47] Nagao M, Hamilton JL, Kc R, Berendsen AD, Duan X, Cheong CW, et al. Vascular endothelial growth factor in cartilage development and osteoarthritis. Sci Rep. 2017;7:13027.10.1038/s41598-017-13417-wSearch in Google Scholar PubMed PubMed Central

[48] Middleton J, Manthey A, Tyler J. Insulin-like growth factor (IGF) receptor, IGF-I, interleukin-1 beta (IL-1 beta), and IL-6 mRNA expression in osteoarthritic and normal human cartilage. J Histochem Cytochem. 1996;44:133–41.10.1177/44.2.8609369Search in Google Scholar PubMed

[49] Yi C, Ma C, Xie Z, Zhang G, Song W, Zhou X, et al. Down-regulation of programmed cell death 5by insulin-like growth factor 1in osteoarthritis chondrocytes. Int Orthop. 2013;37:937–43.10.1007/s00264-012-1744-xSearch in Google Scholar PubMed PubMed Central

[50] Galasso O, De Gori M, Nocera A, Brunetti A, Gasparini G. Regulatory functions of insulin-like growth factor binding proteins in osteoarthritis. Int J Immunopathol Pharmacol. 2011;24:55–9.10.1177/03946320110241S211Search in Google Scholar PubMed

[51] Boushell MK, Mosher CZ, Suri GK, Doty SB, Strauss EJ, Hunziker EB, et al. Polymeric mesh and insulin-like growth factor 1 delivery enhance cell homing and graft-cartilage integration. Ann N Y Acad Sci. 2019;1442(1):138–52.10.1111/nyas.14054Search in Google Scholar PubMed PubMed Central

[52] Wang J, Liu S, Li J, Yi Z. The role of the fibroblast growth factor family in bone-related diseases. Chem Biol Drug Des. 2019;94:1740–9.10.1111/cbdd.13588Search in Google Scholar PubMed

[53] Yan D, Chen D, Cool SM, van Wijnen AJ, Mikecz K, Murphy G, et al. Fibroblast growth factor receptor 1 is principally responsible for fibroblast growth factor 2-induced catabolic activities in human articular chondrocytes. Arthritis Res Ther. 2011;13:R130.10.1186/ar3441Search in Google Scholar PubMed PubMed Central

[54] Liu Z, Xu J, Colvin JS, Ornitz DM. Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18. Genes Dev. 2015;16:859–69.10.1101/gad.965602Search in Google Scholar PubMed PubMed Central

[55] Correa D, Somoza RA, Lin P, Greenberg S, Rom E, Duesler L, et al. Sequential exposure to fibroblast growth factors (FGF) 2, 9 and 18 enhances hMSC chondrogenic differentiation. Osteoarthr Cartil. 2015;23:443–53.10.1016/j.joca.2014.11.013Search in Google Scholar PubMed PubMed Central

[56] Lohmander LS, Hellot S, Dreher D, Krantz EF, Kruger DS, Guermazi A, et al. Intraarticular sprifermin (recombinant human fibroblast growth factor 18) in knee osteoarthritis: a randomized, double-blind, placebocontrolled trial. Arthritis Rheumatol. 2014;66:1820–31.10.1002/art.38614Search in Google Scholar PubMed

[57] Jia H, Ma X, Tong W, Doyran B, Sun Z, Wang L, et al. EGFR signaling is critical for maintaining the superficial layer of articular cartilage and preventing osteoarthritis initiation. Proc Natl Acad Sci U SA. 2016;113:14360–5.10.1073/pnas.1608938113Search in Google Scholar PubMed PubMed Central

[58] Zhang X, Zhu J, Li Y, Lin T, Siclari VA, Chandra A, et al. Epidermal growth factor receptor (EGFR) signaling regulates epiphyseal cartilage development through beta-catenin-dependent and -independent pathways. J Biol Chem. 2013;288:32229–40.10.1074/jbc.M113.463554Search in Google Scholar PubMed PubMed Central

[59] Qin L, Beier F. EGFR signaling: Friend or foe for cartilage? JBMR Plus. 2019;3:e10177.10.1002/jbm4.10177Search in Google Scholar PubMed PubMed Central

[60] Long DL, Ulici V, Chubinskaya S, Loeser RF. Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is increased in osteoarthritis and regulates chondrocyte catabolic and anabolic activities. Osteoarthr Cartil. 2015;23:1523–31.10.1016/j.joca.2015.04.019Search in Google Scholar PubMed PubMed Central

[61] Sun H, Wu Y, Pan Z, Yu D, Chen P, Zhang X, et al. Gefitinib for epidermal growth factor receptor activated osteoarthritis subpopulation treatment. EBioMedicine. 2018;32:223–33.10.1016/j.ebiom.2018.06.002Search in Google Scholar PubMed PubMed Central

[62] Roos EM, Arden NK. Strategies for the prevention of knee osteoarthritis. Nat Rev Rheumatol. 2016;12:92–101.10.1038/nrrheum.2015.135Search in Google Scholar PubMed

[63] Liang Y, Duan L, Xiong J, Zhu W, Liu Q, Wang D, et al. E2 regulates MMP-13 via targeting miR-140 in IL-1β-induced extracellular matrix degradation in human chondrocytes. Arthritis Res Ther. 2016;18:105.10.1186/s13075-016-0997-ySearch in Google Scholar PubMed PubMed Central

[64] Yazdi MM, Jamalaldini MH, Sobhan MR, Jafari M, Mazaheri M, Zare-Shehneh M, et al. Association of ESRalpha gene Pvu II T > C, XbaI A > G and BtgI G > A polymorphisms with knee osteoarthritis susceptibility: a systematic review and meta-analysis based on 22 case-control studies. Arch Bone JtSurg. 2017;5:351–62.Search in Google Scholar

[65] Xiao YP, Tian FM, Dai MW, Wang WY, Shao LT, Zhang L. Are estrogen-related drugs new alternatives for the management of osteoarthritis? Arthritis Res & Ther. 2016;18:151.10.1186/s13075-016-1045-7Search in Google Scholar PubMed PubMed Central

[66] Xu X, Li X, Liang Y, Ou Y, Huang J, Xiong J, et al. Estrogen modulates cartilage and subchondral bone remodeling in an ovariectomized rat model of postmenopausal osteoarthritis. Med Sci Monit. 2019;25:3146–53.10.12659/MSM.916254Search in Google Scholar PubMed PubMed Central

[67] Chen Q, Jin M, Yang F, Zhu J, Xiao Q, Zhang L. Matrix metalloproteinases: inflammatory regulators of cell behaviors in vascular formation and remodeling. Med Inflamm. 2013;2013:928315.10.1155/2013/928315Search in Google Scholar PubMed PubMed Central

[68] Powell BS, Dhaher YY, Szleifer IG. Review of the multiscale effects of female sex hormones on matrix metalloproteinase-mediated collagen degradation. Crit Rev Biomed Eng. 2015;43:401–28.10.1615/CritRevBiomedEng.2016016590Search in Google Scholar PubMed

[69] Woodell-May JE, Sommerfeld SD. Role of inflammation and the immune system in the progression of osteoarthritis. J Orthop Res. 2020;38:253–7.10.1002/jor.24457Search in Google Scholar PubMed

[70] Millerand M, Berenbaum F, Jacques C. Danger signals and inflammaging in osteoarthritis. Clin Exp Rheumatol. 2019;37(Suppl 120):48–56.Search in Google Scholar

[71] Kim HA, Cho ML, Choi HY, Yoon CS, Jhun JY, Oh HJ, et al. The catabolic pathway mediated by Toll-like receptors in human osteoarthritic chondrocytes. Arthritis Rheum. 2006;54:2152–63.10.1002/art.21951Search in Google Scholar PubMed

[72] Herrero-Beaumont G, Pérez-Baos S, Sánchez-Pernaute O, Roman-Blas JA, Lamuedra A, Largo R. Targeting chronic innate inflammatory pathways, the main road to prevention of osteoarthritis progression. Biochem Pharmacol. 2019;165:24–32.10.1016/j.bcp.2019.02.030Search in Google Scholar PubMed

[73] Su SL, Tsai CD, Lee CH, Salter DM, Lee HS. Expression and regulation of Toll-like receptor 2 by IL-1beta and fibronectin fragments in human articular chondrocytes. Osteoarthr Cartil. 2005;13:879–86.10.1016/j.joca.2005.04.017Search in Google Scholar PubMed

[74] Barreto G, Sandelin J, Salem A, Nordström DC, Waris E. Toll-like receptors and their soluble forms differ in the knee and thumb basal osteoarthritic joints. Acta Orthop. 2017;88:326–33.10.1080/17453674.2017.1281058Search in Google Scholar PubMed PubMed Central

[75] Barreto G, Manninen M, Eklund K, K. Osteoarthritis and Toll-like receptors: When innate immunity meets chondrocyte apoptosis. Biology (Basel). 2020;9:6510.3390/biology9040065Search in Google Scholar PubMed PubMed Central

[76] Fang T, Zhou X, Jin M, Nie J, Li X. Molecular mechanisms of mechanical load-induced osteoarthritis. Int Orthop. 2021;45:1125–36.10.1007/s00264-021-04938-1Search in Google Scholar PubMed

[77] Soga M, Izumi T, Nanchi I, Horita N, Yamamoto M, Kawasaki S, et al. Suppression of joint pain in transient receptor potential vanilloid 4 knockout rats with monoiodoacetate-induced osteoarthritis. Pain Rep. 2021;6:e951.10.1097/PR9.0000000000000951Search in Google Scholar PubMed PubMed Central

[78] Xu B, Xing R, Huang Z, Yin S, Li X, Zhang L, et al. Excessive mechanical stress induces chondrocyte apoptosis through TRPV4 in an anterior cruciate ligament-transected rat osteoarthritis model. Life Sci. 2019;228:158–66.10.1016/j.lfs.2019.05.003Search in Google Scholar PubMed

[79] Eleswarapu SV, Athanasiou KA. TRPV4 channel activation improves the tensile properties of self-assembled articular cartilage constructs. Acta Biomater. 2013;9:5554–61.10.1016/j.actbio.2012.10.031Search in Google Scholar PubMed PubMed Central

[80] McNulty AL, Leddy HA, Liedtke W, Guilak F. TRPV4 as a therapeutic target for joint diseases. Naunyn-Schmiedeberg’s Arch Pharmacol. 2015;388:437–50.10.1007/s00210-014-1078-xSearch in Google Scholar PubMed PubMed Central

[81] Loeser RF. Integrins and chondrocyte-matrix interactions in articular cartilage. Matrix Biol. 2014;39:11–6.10.1016/j.matbio.2014.08.007Search in Google Scholar PubMed PubMed Central

[82] Tian J, Zhang FJ, Lei GH. Role of integrins and their ligands in osteoarthritic cartilage. Rheumatol Int. 2015;35:787–98.10.1007/s00296-014-3137-5Search in Google Scholar PubMed

[83] Delco ML, Goodale M, Talts JF, Pownder SL, Koff MF, Miller AD, et al. Integrin alpha10beta1-Selected mesenchymal stem cells mitigate the progression of osteoarthritis in an equine talar impact model. Am J Sports Med. 2020;48:612–23.10.1177/0363546519899087Search in Google Scholar PubMed

[84] Christofides A, Konstantinidou E, Jani C, Boussiotis VA. The role of peroxisome proliferator- activated receptors (PPAR) in immune responses. Metabolism. 2021;114:154338.10.1016/j.metabol.2020.154338Search in Google Scholar PubMed PubMed Central

[85] Wang J, Wang G, Sun GW. Role of PPARα in down-regulating AGE-induced TGF-β and MMP-9 expressions in chondrocytes. Genet Mol Res. 2016;15:15.10.4238/gmr.15027963Search in Google Scholar PubMed

[86] Nogueira-Recalde U, Lorenzo-Gómez I, Blanco FJ, Loza MI, Grassi D, Shirinsky V, et al. Fibrates as drugs with senolytic and autophagic activity for osteoarthritis therapy. EBioMedicine. 2019;45:588–605.10.1016/j.ebiom.2019.06.049Search in Google Scholar PubMed PubMed Central

[87] Heck BE, Park JJ, Makani V, Kim EC, Kim DH. PPAR-δ agonist with mesenchymal stem cells induces typeⅡcollagen-producing chondrocytes in human arthritic synovial fluid. Cell Transpl. 2017;26:1405–17.10.1177/0963689717720278Search in Google Scholar PubMed PubMed Central

[88] Wang JS, Xiao WW, Zhong YS, Li XD, Du SX, Xie P, et al. Galectin-3 deficiency protects lipopolysaccharide-induced chondrocytes injury via regulation of TLR4 and PPAR-γ-mediated NF-κB signaling pathway. J Cell Biochem. 2019;120:10195–204.10.1002/jcb.28304Search in Google Scholar PubMed

[89] Wu Y, Lu X, Li M, Zeng J, Zeng J, Shen B, et al. Renin-angiotensin system in osteoarthritis: A new potential therapy. Int Immunopharmacol. 2019;75:105796.10.1016/j.intimp.2019.105796Search in Google Scholar PubMed

[90] Tang Y, Hu XP, Lu XW. Captopril, an angiotensin-converting enzyme inhibitor, possesses chondroprotective efficacy in a rat model of osteoarthritis through suppression local renin-angiotensin system. Int J Clin Exp Med. 2015;8:12584–92.Search in Google Scholar

[91] Mukhtar M, Sheikh N, Suqaina SK, Batool A, Fatima N, Mehmood R, et al. Vitamin D receptor gene polymorphism: An important predictor of arthritis development. Biomed Res Int. 2019;2019:8326246.10.1155/2019/8326246Search in Google Scholar PubMed PubMed Central

[92] Zhang L, Yin X, Wang J, Xu D, Wang Y, Yang J, et al. Associations between VDR gene polymorphisms and osteoporosis risk and bone mineral density in postmenopausal women: A systematic review and meta-analysis. Sci Rep. 2018;8:981.10.1038/s41598-017-18670-7Search in Google Scholar PubMed PubMed Central

[93] Garfinkel RJ, Dilisio MF, Agrawal DK. Vitamin D and its effects on articular cartilage and osteoarthritis. Orthop J Sports Med. 2017;5:2325967117711376.10.1177/2325967117711376Search in Google Scholar PubMed PubMed Central

[94] Shi X, Ye H, Yao X, Gao Y. The involvement and possible mechanism of NR4A1 in chondrocyte apoptosis during osteoarthritis. Am J Transl Res. 2017;9:746–54.Search in Google Scholar

[95] Mathiessen A, Conaghan PG. Synovitis in osteoarthritis: current understanding with therapeutic implications. Arthritis Res Ther. 2017;19:18.10.1186/s13075-017-1229-9Search in Google Scholar PubMed PubMed Central

[96] Martel-Pelletier J, Mineau F, Jolicoeur, Cloutier JM, Pelletier JPFC. In vitro effects of diacerhein and rhein on interleukin 1 and tumor necrosis factor-alpha systems in human osteoarthritic synovium and chondrocytes. J Rheumatol. 1998;25:753–62.Search in Google Scholar

[97] Appleton CT. Osteoarthritis year in review 2017: biology. Osteoarthr Cartil. 2018;26:296–303.10.1016/j.joca.2017.10.008Search in Google Scholar PubMed

[98] Pongratz G, Anthofer JM, Melzer M, Anders S, Grässel S, Straub RH. IL-7 receptor α expressing B cells act proinflammatory in collagen-induced arthritis and are inhibited by sympathetic neurotransmitters. Ann Rheum Dis. 2014;73:306–12.10.1136/annrheumdis-2012-202944Search in Google Scholar PubMed

[99] Han W, Aitken D, Zheng S, Wang B, Wluka AE, Zhu Z, et al. Higher serum levels of resistin are associated with knee synovitis and structural abnormalities in patients with symptomatic knee osteoarthritis. J Am Med Dir Assoc. 2019;20:1242–6.10.1016/j.jamda.2019.07.001Search in Google Scholar

[100] Shapiro L, Scherer PE. The crystal structure of a complement-1q family protein suggests an evolutionary link to tumor necrosis factor. Curr Biol. 1998;8:335–8.10.1016/S0960-9822(98)70133-2Search in Google Scholar

[101] Tang CH, Chiu YC, Tan TW, Yang RS, Fu WM. Adiponectin enhances IL-6 production in human synovial fibroblast via an Adipo R1 receptor, AMPK, p38, and NF-kappa B pathway. J Immunol. 2007;179:5483–92.10.4049/jimmunol.179.8.5483Search in Google Scholar

[102] Tong KM, Shieh DC, Chen CP, Tzeng CY, Wang SP, Huang KC, et al. Leptin induces IL-8 expression via leptin receptor, IRS-1, PI3K, Akt cascade and promotion of NF-kappaB/p300 binding in human synovial fibroblasts. Cell Signal. 2008;20:1478–88.10.1016/j.cellsig.2008.04.003Search in Google Scholar

[103] Conti P, Reale M, Barbacane RC, Castellani ML, Orso C. Differential production of RANTES and MCP-1 in synovial fluid from the inflamed human knee. Immunol Lett. 2002;80:105–11.10.1016/S0165-2478(01)00303-0Search in Google Scholar

[104] Lee S, Lee HC, Kwon YW, Lee SE, Cho Y, Kim J, et al. Adenylyl cyclase-associated protein 1 is a receptor for human resistin and mediates inflammatory actions of human monocytes. Cell Metab. 2014;19:484–97.10.1016/j.cmet.2014.01.013Search in Google Scholar PubMed PubMed Central

[105] Boström EA, Svensson M, Andersson S, Jonsson IM, Ekwall AK, Eisler T, et al. Resistin and insulin/insulin-like growth factor signaling in rheumatoid arthritis. Arthritis Rheum. 2011;63:2894–904.10.1002/art.30527Search in Google Scholar PubMed

[106] Chen S, Zhang L, Xu R, Ti Y, Zhao Y, Zhou L, et al. BDKRB2 + 9/-9 bp polymorphisms influence BDKRB2 expression levels and NO production in knee osteoarthritis. Exp Biol Med (Maywood). 2017;242:422–8.10.1177/1535370215625471Search in Google Scholar PubMed PubMed Central

[107] Chen S, Zhang L, Xu R, Ti Y, Zhao Y, Zhou L, et al. The BDKRB2 + 9/-9 polymorphisms influence pro-inflammatory cytokine levels in knee osteoarthritis by altering TLR-2 expression: clinical and in vitro studies. Cell Physiol Biochem. 2016;38:1245–56.10.1159/000443072Search in Google Scholar PubMed

[108] Waterborg CEJ, Broeren MGA, Blaney Davidson EN, Koenders MI, van Lent P, van den Berg WB, et al. The level of synovial AXL expression determines the outcome of inflammatory arthritis, possibly depending on the upstream role of TGF-beta1. Rheumatol (Oxf). 2019;58:536–46.10.1093/rheumatology/key337Search in Google Scholar PubMed

[109] Goldring SR, Goldring MB. Changes in the osteochondral unit during osteoarthritis: structure, function and cartilage-bone crosstalk. Nat Rev Rheumatol. 2016;12:632–44.10.1038/nrrheum.2016.148Search in Google Scholar PubMed

[110] van der Kraan PM. Differential role of transforming growth factor-beta in an osteoarthritic or a healthy joint. J Bone Metab. 2018;25:65–72.10.11005/jbm.2018.25.2.65Search in Google Scholar PubMed PubMed Central

[111] Ng PY, Ong AJ, Gale LS, Dass CR. Treatment of bone disorders with parathyroid hormone: success and pitfalls. Pharmazie. 2016;71:427–33.Search in Google Scholar

[112] Hilal G, Massicotte F, Martel-Pelletier J, Fernandes JC, Pelletier JP, Lajeunesse D. Endogenous prostaglandin E2 and insulin-like growth factor 1 can modulate the levels of parathyroid hormone receptor in human osteoarthritic osteoblasts. J Bone Min Res. 2001;16:713–21.10.1359/jbmr.2001.16.4.713Search in Google Scholar PubMed

[113] Orth P, Cucchiarini M, Zurakowski D, Menger MD, Kohn DM, Madry H. Parathyroid hormone [1-34] improves articular cartilage surface architecture and integration and subchondral bone reconstitution in osteochondral defects in vivo. Osteoarthr Cartil. 2013;21:614–24.10.1016/j.joca.2013.01.008Search in Google Scholar PubMed

[114] Guo J, Liu M, Yang D, Bouxsein ML, Saito H, Galvin RJ, et al. Suppression of Wnt signaling by Dkk1 attenuates PTH-mediated stromal cell response and new bone formation. Cell Metab. 2010;11:161–71.10.1016/j.cmet.2009.12.007Search in Google Scholar PubMed PubMed Central

[115] Li L, Li M, Pang Y, Wang J, Wan Y, Zhu C, et al. Abnormal thyroid hormone receptor signaling in osteoarthritic osteoblasts regulates microangiogenesis in subchondral bone. Life Sci. 2019;239C:116975.10.1016/j.lfs.2019.116975Search in Google Scholar PubMed

[116] Sniekers YH, van Osch GJ, Ederveen AG, Inzunza J, Gustafsson JA, van Leeuwen JP, et al. Development of osteoarthritic features in estrogen receptor knockout mice. Osteoarthr Cartil. 2009;17:1356–61.10.1016/j.joca.2009.04.008Search in Google Scholar PubMed

[117] Ruiz M, Toupet K, Maumus M, Rozier P, Jorgensen C, Noël D. TGFBI secreted by mesenchymal stromal cells ameliorates osteoarthritis and is detected in extracellular vesicles. Biomaterials. 2019;226:119544.10.1016/j.biomaterials.2019.119544Search in Google Scholar PubMed

[118] Zhao X, Ruan J, Tang H, Li J, Shi Y, Li M, et al. Multi-compositional MRI evaluation of repair cartilage in knee osteoarthritis with treatment of allogeneic human adipose-derived mesenchymal progenitor cells. Stem Cell Res Ther. 2019;10:308.10.1186/s13287-019-1406-7Search in Google Scholar PubMed PubMed Central

[119] Chevalier X, Giraudeau B, Conrozier T, Marliere J, Kiefer P, Goupille P. Safety study of intraarticular injection of interleukin 1 receptor antagonist in patients with painful knee osteoarthritis: a multicenter study. J Rheumatol. 2005;32:1317–23.Search in Google Scholar

[120] Sun S, Rao NL, Venable J, Thurmond R, Karlsson L. TLR7/9 antagonists as therapeutics for immune-mediated inflammatory disorders. Inflamm Allergy Drug Targets. 2007;6:223–35.10.2174/187152807783334300Search in Google Scholar PubMed

[121] Bagal MCS, Brady P, Stauffer J. A Phase 2, Randomized, Double-Blind, PlaceboControlled, Titration-to-Effect Study of Orally Administered CR845 in Patients with Osteoarthritis of the Hip or Knee. [abstract], ACR/ARHP Annual Meeting. Arthritis Rheumatol; 2017.Search in Google Scholar

[122] Krupka E, Jiang GL, Jan C. Efficacy and safety of intra-articular injection of tropomyosin receptor kinase A inhibitor in painful knee osteoarthritis: a randomized, double-blind and placebo-controlled study. Osteoarthr Cartil. 2019;27:1599–607.10.1016/j.joca.2019.05.028Search in Google Scholar PubMed

[123] Stevens RM, Ervin J, Nezzer J, Nieves Y, Guedes K, Burges R, et al. Randomized, double-blind, placebocontrolled trial of intraarticular trans-capsaicin for pain associated with osteoarthritis of the knee. Arthritis Rheumatol. 2019;71:1524–33.10.1002/art.40894Search in Google Scholar PubMed PubMed Central

[124] Ellman MB, Yan D, Ahmadinia K, Chen D, An HS, Im HJ. Fibroblast growth factor control of cartilage homeostasis. J Cell Biochem. 2013;114:735–42.10.1002/jcb.24418Search in Google Scholar PubMed PubMed Central

[125] Xie Y, Zinkle A, Chen L, Mohammadi M. Fibroblast growth factor signalling in osteoarthritis and cartilage repair. Nat Rev Rheumatol. 2020;16:547–64.10.1038/s41584-020-0469-2Search in Google Scholar PubMed

[126] Lohmander LS, Hellot S, Dreher D, Krantz EF, Kruger DS, Guermazi A, et al. Intraarticular sprifermin (recombinant human fibroblast growth factor 18) in knee osteoarthritis: a randomized, double-blind, placebo-controlled trial. Arthritis Rheumatol. 2014;66:1820–31.10.1002/art.38614Search in Google Scholar PubMed

[127] Ribitsch I, Baptista PM, Lange-Consiglio A, Melotti L, Patruno M, Jenner F, et al. Large animal models in regenerative medicine and tissue engineering: To do or not to do. Front Bioeng Biotechnol. 2020;8:972.10.3389/fbioe.2020.00972Search in Google Scholar PubMed PubMed Central

[128] Van Hecke L, Magri C, Duchateau L, Beerts C, Geburek F, Suls M, et al. Repeated intra-articular administration of equine allogeneic peripheral blood-derived mesenchymal stem cells does not induce a cellular and humoral immune response in horses. Vet Immunol Immunopathol. 2021;239:110306.10.1016/j.vetimm.2021.110306Search in Google Scholar PubMed

Received: 2022-01-11
Revised: 2022-03-16
Accepted: 2022-03-21
Published Online: 2022-07-07

© 2022 Li Lei et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Biomedical Sciences
  2. Effects of direct oral anticoagulants dabigatran and rivaroxaban on the blood coagulation function in rabbits
  3. The mother of all battles: Viruses vs humans. Can humans avoid extinction in 50–100 years?
  4. Knockdown of G1P3 inhibits cell proliferation and enhances the cytotoxicity of dexamethasone in acute lymphoblastic leukemia
  5. LINC00665 regulates hepatocellular carcinoma by modulating mRNA via the m6A enzyme
  6. Association study of CLDN14 variations in patients with kidney stones
  7. Concanavalin A-induced autoimmune hepatitis model in mice: Mechanisms and future outlook
  8. Regulation of miR-30b in cancer development, apoptosis, and drug resistance
  9. Informatic analysis of the pulmonary microecology in non-cystic fibrosis bronchiectasis at three different stages
  10. Swimming attenuates tumor growth in CT-26 tumor-bearing mice and suppresses angiogenesis by mediating the HIF-1α/VEGFA pathway
  11. Characterization of intestinal microbiota and serum metabolites in patients with mild hepatic encephalopathy
  12. Functional conservation and divergence in plant-specific GRF gene family revealed by sequences and expression analysis
  13. Application of the FLP/LoxP-FRT recombination system to switch the eGFP expression in a model prokaryote
  14. Biomedical evaluation of antioxidant properties of lamb meat enriched with iodine and selenium
  15. Intravenous infusion of the exosomes derived from human umbilical cord mesenchymal stem cells enhance neurological recovery after traumatic brain injury via suppressing the NF-κB pathway
  16. Effect of dietary pattern on pregnant women with gestational diabetes mellitus and its clinical significance
  17. Potential regulatory mechanism of TNF-α/TNFR1/ANXA1 in glioma cells and its role in glioma cell proliferation
  18. Effect of the genetic mutant G71R in uridine diphosphate-glucuronosyltransferase 1A1 on the conjugation of bilirubin
  19. Quercetin inhibits cytotoxicity of PC12 cells induced by amyloid-beta 25–35 via stimulating estrogen receptor α, activating ERK1/2, and inhibiting apoptosis
  20. Nutrition intervention in the management of novel coronavirus pneumonia patients
  21. circ-CFH promotes the development of HCC by regulating cell proliferation, apoptosis, migration, invasion, and glycolysis through the miR-377-3p/RNF38 axis
  22. Bmi-1 directly upregulates glucose transporter 1 in human gastric adenocarcinoma
  23. Lacunar infarction aggravates the cognitive deficit in the elderly with white matter lesion
  24. Hydroxysafflor yellow A improved retinopathy via Nrf2/HO-1 pathway in rats
  25. Comparison of axon extension: PTFE versus PLA formed by a 3D printer
  26. Elevated IL-35 level and iTr35 subset increase the bacterial burden and lung lesions in Mycobacterium tuberculosis-infected mice
  27. A case report of CAT gene and HNF1β gene variations in a patient with early-onset diabetes
  28. Study on the mechanism of inhibiting patulin production by fengycin
  29. SOX4 promotes high-glucose-induced inflammation and angiogenesis of retinal endothelial cells by activating NF-κB signaling pathway
  30. Relationship between blood clots and COVID-19 vaccines: A literature review
  31. Analysis of genetic characteristics of 436 children with dysplasia and detailed analysis of rare karyotype
  32. Bioinformatics network analyses of growth differentiation factor 11
  33. NR4A1 inhibits the epithelial–mesenchymal transition of hepatic stellate cells: Involvement of TGF-β–Smad2/3/4–ZEB signaling
  34. Expression of Zeb1 in the differentiation of mouse embryonic stem cell
  35. Study on the genetic damage caused by cadmium sulfide quantum dots in human lymphocytes
  36. Association between single-nucleotide polymorphisms of NKX2.5 and congenital heart disease in Chinese population: A meta-analysis
  37. Assessment of the anesthetic effect of modified pentothal sodium solution on Sprague-Dawley rats
  38. Genetic susceptibility to high myopia in Han Chinese population
  39. Potential biomarkers and molecular mechanisms in preeclampsia progression
  40. Silencing circular RNA-friend leukemia virus integration 1 restrained malignancy of CC cells and oxaliplatin resistance by disturbing dyskeratosis congenita 1
  41. Endostar plus pembrolizumab combined with a platinum-based dual chemotherapy regime for advanced pulmonary large-cell neuroendocrine carcinoma as a first-line treatment: A case report
  42. The significance of PAK4 in signaling and clinicopathology: A review
  43. Sorafenib inhibits ovarian cancer cell proliferation and mobility and induces radiosensitivity by targeting the tumor cell epithelial–mesenchymal transition
  44. Characterization of rabbit polyclonal antibody against camel recombinant nanobodies
  45. Active legumain promotes invasion and migration of neuroblastoma by regulating epithelial-mesenchymal transition
  46. Effect of cell receptors in the pathogenesis of osteoarthritis: Current insights
  47. MT-12 inhibits the proliferation of bladder cells in vitro and in vivo by enhancing autophagy through mitochondrial dysfunction
  48. Study of hsa_circRNA_000121 and hsa_circRNA_004183 in papillary thyroid microcarcinoma
  49. BuyangHuanwu Decoction attenuates cerebral vasospasm caused by subarachnoid hemorrhage in rats via PI3K/AKT/eNOS axis
  50. Effects of the interaction of Notch and TLR4 pathways on inflammation and heart function in septic heart
  51. Monosodium iodoacetate-induced subchondral bone microstructure and inflammatory changes in an animal model of osteoarthritis
  52. A rare presentation of type II Abernethy malformation and nephrotic syndrome: Case report and review
  53. Rapid death due to pulmonary epithelioid haemangioendothelioma in several weeks: A case report
  54. Hepatoprotective role of peroxisome proliferator-activated receptor-α in non-cancerous hepatic tissues following transcatheter arterial embolization
  55. Correlation between peripheral blood lymphocyte subpopulations and primary systemic lupus erythematosus
  56. A novel SLC8A1-ALK fusion in lung adenocarcinoma confers sensitivity to alectinib: A case report
  57. β-Hydroxybutyrate upregulates FGF21 expression through inhibition of histone deacetylases in hepatocytes
  58. Identification of metabolic genes for the prediction of prognosis and tumor microenvironment infiltration in early-stage non-small cell lung cancer
  59. BTBD10 inhibits glioma tumorigenesis by downregulating cyclin D1 and p-Akt
  60. Mucormycosis co-infection in COVID-19 patients: An update
  61. Metagenomic next-generation sequencing in diagnosing Pneumocystis jirovecii pneumonia: A case report
  62. Long non-coding RNA HOXB-AS1 is a prognostic marker and promotes hepatocellular carcinoma cells’ proliferation and invasion
  63. Preparation and evaluation of LA-PEG-SPION, a targeted MRI contrast agent for liver cancer
  64. Proteomic analysis of the liver regulating lipid metabolism in Chaohu ducks using two-dimensional electrophoresis
  65. Nasopharyngeal tuberculosis: A case report
  66. Characterization and evaluation of anti-Salmonella enteritidis activity of indigenous probiotic lactobacilli in mice
  67. Aberrant pulmonary immune response of obese mice to periodontal infection
  68. Bacteriospermia – A formidable player in male subfertility
  69. In silico and in vivo analysis of TIPE1 expression in diffuse large B cell lymphoma
  70. Effects of KCa channels on biological behavior of trophoblasts
  71. Interleukin-17A influences the vulnerability rather than the size of established atherosclerotic plaques in apolipoprotein E-deficient mice
  72. Multiple organ failure and death caused by Staphylococcus aureus hip infection: A case report
  73. Prognostic signature related to the immune environment of oral squamous cell carcinoma
  74. Primary and metastatic squamous cell carcinoma of the thyroid gland: Two case reports
  75. Neuroprotective effects of crocin and crocin-loaded niosomes against the paraquat-induced oxidative brain damage in rats
  76. Role of MMP-2 and CD147 in kidney fibrosis
  77. Geometric basis of action potential of skeletal muscle cells and neurons
  78. Babesia microti-induced fulminant sepsis in an immunocompromised host: A case report and the case-specific literature review
  79. Role of cerebellar cortex in associative learning and memory in guinea pigs
  80. Application of metagenomic next-generation sequencing technique for diagnosing a specific case of necrotizing meningoencephalitis caused by human herpesvirus 2
  81. Case report: Quadruple primary malignant neoplasms including esophageal, ureteral, and lung in an elderly male
  82. Long non-coding RNA NEAT1 promotes angiogenesis in hepatoma carcinoma via the miR-125a-5p/VEGF pathway
  83. Osteogenic differentiation of periodontal membrane stem cells in inflammatory environments
  84. Knockdown of SHMT2 enhances the sensitivity of gastric cancer cells to radiotherapy through the Wnt/β-catenin pathway
  85. Continuous renal replacement therapy combined with double filtration plasmapheresis in the treatment of severe lupus complicated by serious bacterial infections in children: A case report
  86. Simultaneous triple primary malignancies, including bladder cancer, lymphoma, and lung cancer, in an elderly male: A case report
  87. Preclinical immunogenicity assessment of a cell-based inactivated whole-virion H5N1 influenza vaccine
  88. One case of iodine-125 therapy – A new minimally invasive treatment of intrahepatic cholangiocarcinoma
  89. S1P promotes corneal trigeminal neuron differentiation and corneal nerve repair via upregulating nerve growth factor expression in a mouse model
  90. Early cancer detection by a targeted methylation assay of circulating tumor DNA in plasma
  91. Calcifying nanoparticles initiate the calcification process of mesenchymal stem cells in vitro through the activation of the TGF-β1/Smad signaling pathway and promote the decay of echinococcosis
  92. Evaluation of prognostic markers in patients infected with SARS-CoV-2
  93. N6-Methyladenosine-related alternative splicing events play a role in bladder cancer
  94. Characterization of the structural, oxidative, and immunological features of testis tissue from Zucker diabetic fatty rats
  95. Effects of glucose and osmotic pressure on the proliferation and cell cycle of human chorionic trophoblast cells
  96. Investigation of genotype diversity of 7,804 norovirus sequences in humans and animals of China
  97. Characteristics and karyotype analysis of a patient with turner syndrome complicated with multiple-site tumors: A case report
  98. Aggravated renal fibrosis is positively associated with the activation of HMGB1-TLR2/4 signaling in STZ-induced diabetic mice
  99. Distribution characteristics of SARS-CoV-2 IgM/IgG in false-positive results detected by chemiluminescent immunoassay
  100. SRPX2 attenuated oxygen–glucose deprivation and reperfusion-induced injury in cardiomyocytes via alleviating endoplasmic reticulum stress-induced apoptosis through targeting PI3K/Akt/mTOR axis
  101. Aquaporin-8 overexpression is involved in vascular structure and function changes in placentas of gestational diabetes mellitus patients
  102. Relationship between CRP gene polymorphisms and ischemic stroke risk: A systematic review and meta-analysis
  103. Effects of growth hormone on lipid metabolism and sexual development in pubertal obese male rats
  104. Cloning and identification of the CTLA-4IgV gene and functional application of vaccine in Xinjiang sheep
  105. Antitumor activity of RUNX3: Upregulation of E-cadherin and downregulation of the epithelial–mesenchymal transition in clear-cell renal cell carcinoma
  106. PHF8 promotes osteogenic differentiation of BMSCs in old rat with osteoporosis by regulating Wnt/β-catenin pathway
  107. A review of the current state of the computer-aided diagnosis (CAD) systems for breast cancer diagnosis
  108. Bilateral dacryoadenitis in adult-onset Still’s disease: A case report
  109. A novel association between Bmi-1 protein expression and the SUVmax obtained by 18F-FDG PET/CT in patients with gastric adenocarcinoma
  110. The role of erythrocytes and erythroid progenitor cells in tumors
  111. Relationship between platelet activation markers and spontaneous abortion: A meta-analysis
  112. Abnormal methylation caused by folic acid deficiency in neural tube defects
  113. Silencing TLR4 using an ultrasound-targeted microbubble destruction-based shRNA system reduces ischemia-induced seizures in hyperglycemic rats
  114. Plant Sciences
  115. Seasonal succession of bacterial communities in cultured Caulerpa lentillifera detected by high-throughput sequencing
  116. Cloning and prokaryotic expression of WRKY48 from Caragana intermedia
  117. Novel Brassica hybrids with different resistance to Leptosphaeria maculans reveal unbalanced rDNA signal patterns
  118. Application of exogenous auxin and gibberellin regulates the bolting of lettuce (Lactuca sativa L.)
  119. Phytoremediation of pollutants from wastewater: A concise review
  120. Genome-wide identification and characterization of NBS-encoding genes in the sweet potato wild ancestor Ipomoea trifida (H.B.K.)
  121. Alleviative effects of magnetic Fe3O4 nanoparticles on the physiological toxicity of 3-nitrophenol to rice (Oryza sativa L.) seedlings
  122. Selection and functional identification of Dof genes expressed in response to nitrogen in Populus simonii × Populus nigra
  123. Study on pecan seed germination influenced by seed endocarp
  124. Identification of active compounds in Ophiopogonis Radix from different geographical origins by UPLC-Q/TOF-MS combined with GC-MS approaches
  125. The entire chloroplast genome sequence of Asparagus cochinchinensis and genetic comparison to Asparagus species
  126. Genome-wide identification of MAPK family genes and their response to abiotic stresses in tea plant (Camellia sinensis)
  127. Selection and validation of reference genes for RT-qPCR analysis of different organs at various development stages in Caragana intermedia
  128. Cloning and expression analysis of SERK1 gene in Diospyros lotus
  129. Integrated metabolomic and transcriptomic profiling revealed coping mechanisms of the edible and medicinal homologous plant Plantago asiatica L. cadmium resistance
  130. A missense variant in NCF1 is associated with susceptibility to unexplained recurrent spontaneous abortion
  131. Assessment of drought tolerance indices in faba bean genotypes under different irrigation regimes
  132. The entire chloroplast genome sequence of Asparagus setaceus (Kunth) Jessop: Genome structure, gene composition, and phylogenetic analysis in Asparagaceae
  133. Food Science
  134. Dietary food additive monosodium glutamate with or without high-lipid diet induces spleen anomaly: A mechanistic approach on rat model
  135. Binge eating disorder during COVID-19
  136. Potential of honey against the onset of autoimmune diabetes and its associated nephropathy, pancreatitis, and retinopathy in type 1 diabetic animal model
  137. FTO gene expression in diet-induced obesity is downregulated by Solanum fruit supplementation
  138. Physical activity enhances fecal lactobacilli in rats chronically drinking sweetened cola beverage
  139. Supercritical CO2 extraction, chemical composition, and antioxidant effects of Coreopsis tinctoria Nutt. oleoresin
  140. Functional constituents of plant-based foods boost immunity against acute and chronic disorders
  141. Effect of selenium and methods of protein extraction on the proteomic profile of Saccharomyces yeast
  142. Microbial diversity of milk ghee in southern Gansu and its effect on the formation of ghee flavor compounds
  143. Ecology and Environmental Sciences
  144. Effects of heavy metals on bacterial community surrounding Bijiashan mining area located in northwest China
  145. Microorganism community composition analysis coupling with 15N tracer experiments reveals the nitrification rate and N2O emissions in low pH soils in Southern China
  146. Genetic diversity and population structure of Cinnamomum balansae Lecomte inferred by microsatellites
  147. Preliminary screening of microplastic contamination in different marine fish species of Taif market, Saudi Arabia
  148. Plant volatile organic compounds attractive to Lygus pratensis
  149. Effects of organic materials on soil bacterial community structure in long-term continuous cropping of tomato in greenhouse
  150. Effects of soil treated fungicide fluopimomide on tomato (Solanum lycopersicum L.) disease control and plant growth
  151. Prevalence of Yersinia pestis among rodents captured in a semi-arid tropical ecosystem of south-western Zimbabwe
  152. Effects of irrigation and nitrogen fertilization on mitigating salt-induced Na+ toxicity and sustaining sea rice growth
  153. Bioengineering and Biotechnology
  154. Poly-l-lysine-caused cell adhesion induces pyroptosis in THP-1 monocytes
  155. Development of alkaline phosphatase-scFv and its use for one-step enzyme-linked immunosorbent assay for His-tagged protein detection
  156. Development and validation of a predictive model for immune-related genes in patients with tongue squamous cell carcinoma
  157. Agriculture
  158. Effects of chemical-based fertilizer replacement with biochar-based fertilizer on albic soil nutrient content and maize yield
  159. Genome-wide identification and expression analysis of CPP-like gene family in Triticum aestivum L. under different hormone and stress conditions
  160. Agronomic and economic performance of mung bean (Vigna radiata L.) varieties in response to rates of blended NPS fertilizer in Kindo Koysha district, Southern Ethiopia
  161. Influence of furrow irrigation regime on the yield and water consumption indicators of winter wheat based on a multi-level fuzzy comprehensive evaluation
  162. Discovery of exercise-related genes and pathway analysis based on comparative genomes of Mongolian originated Abaga and Wushen horse
  163. Lessons from integrated seasonal forecast-crop modelling in Africa: A systematic review
  164. Evolution trend of soil fertility in tobacco-planting area of Chenzhou, Hunan Province, China
  165. Animal Sciences
  166. Morphological and molecular characterization of Tatera indica Hardwicke 1807 (Rodentia: Muridae) from Pothwar, Pakistan
  167. Research on meat quality of Qianhua Mutton Merino sheep and Small-tail Han sheep
  168. SI: A Scientific Memoir
  169. Suggestions on leading an academic research laboratory group
  170. My scientific genealogy and the Toronto ACDC Laboratory, 1988–2022
  171. Erratum
  172. Erratum to “Changes of immune cells in patients with hepatocellular carcinoma treated by radiofrequency ablation and hepatectomy, a pilot study”
  173. Erratum to “A two-microRNA signature predicts the progression of male thyroid cancer”
  174. Retraction
  175. Retraction of “Lidocaine has antitumor effect on hepatocellular carcinoma via the circ_DYNC1H1/miR-520a-3p/USP14 axis”
Downloaded on 5.12.2025 from https://www.degruyterbrill.com/document/doi/10.1515/biol-2022-0075/html
Scroll to top button