Skip to main content
Erschienen in: International Orthopaedics 5/2021

Open Access 18.01.2021 | Review Article

Molecular mechanisms of mechanical load-induced osteoarthritis

verfasst von: Tianshun Fang, Xianhao Zhou, Mingchao Jin, Jiangbo Nie, XIongfeng Li

Erschienen in: International Orthopaedics | Ausgabe 5/2021

Abstract

Introduction

Mechanical loading enhances the progression of osteoarthritis. However, its molecular mechanisms have not been established.

Objective

The aim of this review was to summarize the probable mechanisms of mechanical load-induced osteoarthritis.

Methods

A comprehensive search strategy was used to search PubMed and EMBASE databases (from the 15th of January 2015 to the 20th of October 2020). Search terms included “osteoarthritis”, “mechanical load”, and “mechanism”.

Results

Abnormal mechanical loading activates the interleukin-1β, tumour necrosis factor-α, nuclear factor kappa-B, Wnt, transforming growth factor-β, microRNAs pathways, and the oxidative stress pathway. These pathways induce the pathological progression of osteoarthritis. Mechanical stress signal receptors such as integrin, ion channel receptors, hydrogen peroxide-inducible clone-5, Gremlin-1, and transient receptor potential channel 4 are present in the articular cartilages.

Conclusion

This review highlights the molecular mechanisms of mechanical loading in inducing chondrocyte apoptosis and extracellular matrix degradation. These mechanisms provide potential targets for osteoarthritis prevention and treatment.
Hinweise
The original online version of this article was revised: With the author(s)’ decision to step back from Open Choice, the copyright of the article was changed on April 2021 to © SICOT aisbl 2021 and the article is forthwith distributed under the terms of copyright.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s00264-021-05055-9.

Publisher’s note

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

Introduction

Osteoarthritis is the most common chronic degenerative joint disease, which is one of the main causes of limited joint mobility and disability in the elderly. The clinical symptoms include pain, swelling, and even deformity of the joint. The main pathological changes of OA are the destruction of articular cartilage, including chondrocyte apoptosis and extracellular matrix (ECM) degradation, subchondral damage, and bone remodeling. Currently, in addition to surgical treatment for end-stage OA, effective treatments to reverse OA are not available [1].
Articular cartilage covers joint surfaces to minimize friction caused by bone movements. Cartilage contains chondrocytes and extracellular matrix. Chondrocytes are the only cells present in cartilage tissue. They synthesize and secrete the extracellular matrix. However, they have little regenerative ability. The ECM of articular cartilage is composed of type II collagen, polymerized proteoglycan, and non-collagen components that are cross-linked to form a porous and permeable fiber complex network that contains a considerable amount of water (60–85% wet weight). Chondrocytes and synovium secrete lubricin that covers joint surfaces and protects against mechanical stress by reducing friction.
Articular cartilage lacks regenerative ability when subjected to acute or long-term abnormal mechanical loads. Under these loads, they are prone to degenerative lesions that lead to osteoarthritis (OA). The specific mechanism of mechanical load inducing OA has not been fully revealed. It is reported that mechanical load activates a variety of inflammatory pathways and their downstream signaling channels, such as IL-1β, TNF-α, NF-κB, Wnt, microRNA, and oxidative stress pathways. These pathways are involved in regulating joint inflammation, activating key degradation enzymes in articular cartilage such as metalloproteinases (MMPs) and aggrecanases (ADAMTs), inducing chondrocyte apoptosis, ECM degradation, subchondral bone dysfunction, and synovial inflammation, ultimately leading to OA [2]. This review focuses on how mechanical load affects the pathogenesis of OA through molecular signaling pathways and discusses the functions of these molecular pathways in OA pathophysiology and cartilage homeostasis. We also summarize recent evidences of therapeutic approaches to treat OA by targeting pathway.

Method

To review the literature on mechanical mechanisms of osteoarthritis induced by mechanical loading, a comprehensive search of information was performed in PubMed and EMBASE databases. The search was done performed between the 15th of January 2015 and the 20th of October 2020. Three groups of search terms composed by all authors during joint discussions were developed. Group A referred to terms associated with “osteoarthritis”, and Group B referred to terms associated with “mechanical load” while Group C referred to terms associated with “molecular mechanisms” (see appendix Table 2).
In each search, a combination of Group A, B, and C terms was used by the Boolean operator AND. Terms within a group were combined using the Boolean operator OR. Both in vitro and in vivo studies were included in the study. The search field was “title and abstract” when using PubMed and EMBASE. A search was conducted based on the title, abstract, and then full text to screen out references containing osteoarthritis, mechanical load, and molecular mechanisms. Only articles written in the English language were included. Articles that were not associated with molecular mechanisms of mechanical load-induced osteoarthritis were excluded (Fig. 1).

Result

The interleukin-1β pathway

Interleukin-1β (IL-1β) is a pro-inflammatory cytokine that is highly expressed in OA chondrocytes compared to normal chondrocytes [3]. Its expression is directly activated by abnormal mechanical stresses. Some experiments stimulated articular cartilage through mechanical overloading in vitro [4] (cyclic compression stimulation (CTS), 6% compression (36 kPa) at 1HZ for 4 h) and in vivo [5] (rats on treadmill five days a week for 60 minutes daily (at a speed of 20 m/min)) and observed a significant upregulation of IL-1β expression. IL-1β mediates OA progression by activating downstream signaling pathways. In addition, IL-1β inducible nitric oxide synthase (iNOS) expression increases nitric oxide (NO) concentration and causes oxidative stress [6]. This cytokine also induces the expression of cyclooxygenase-2 (cox-2) and upregulates prostaglandin E2 (PGE2) concentrations. This enzyme mediates bone synthesis that may be associated with osteophyte formation during OA progression [7]. Furthermore, IL-1β activates all three MAPK pathways (ERK, p38, and JNK). Activation of these pathways generates several matrix-degrading enzymes such as MMP1, MMP3, MMP13, ADAMT4, ADAMT5, and aggrecanase that activate downstream inflammatory factors such as COX-2, iNOS, PGE-2, and IL-6 [8]. Through the nuclear factor kappa-B (NF-κB) pathway activation, downstream matrix degrading enzymes such as MMP1 and MMP13 and inflammatory factors including tumour necrosis factor-α (TNF-α), NO, COX-2, and IL-6 are also upregulated by this cytokine [9]. In addition, IL-1β triggers the Wnt pathway and induces MMP expression [10]. IL-1β upregulates MMP3 and MMP9 by activating the JNK pathway to accelerate synovial senescence [11]. It also induces iNOS and COX-2 by activating PI3K and Akt pathways to upregulate the concentration of NO, PGE2, MMPs, and ADAMTs [12]. IL-1β enhances nuclear factor erythroid-2 related factor 2 (Nrf2) and Heme Oxygenase-1 (HO-1) to upregulate matrix-degrading enzymes and oxidative stress [13]. IL-1β can upregulate MMP3 and MMP9 and ADAMTs by activating IL-17, thereby promoting joint inflammation and inducing chondrocyte apoptosis [14]. Ohtsuki et al. [15] observed that IL-1β is also involved in upregulating Cell migration-inducing hyaluronidase 1 (CEMIP), which induces the degradation of hyaluronan in ECM. Several miRNAs (miR-34a, miR-30a, miR-145, and miR-27a) are activated by IL-1β and participate in progression of OA by activating matrix-degrading enzymes and downstream inflammatory pathways such as MAPK and NF-κB [16]. Notably, IL-1β directly induces the activation of matrix-degrading enzymes, resulting in cartilage damage. Wang et al. [17] showed a significant decrease in the friction coefficient of cartilage after inhibiting IL-1β in OA model rats (IL-1Ra, 500 μg/ml in a total volume of 40 μl per intra-articular injection) and vitro (20 ng/ml, 36 h). These frictions promoted OA progression and upregulated IL-1β concentration. Clinical applications of IL-1β inhibitors against OA can achieve significant effects. IL-1β also stimulates OA pain. The Pannexin-1 (Panx1) channel is an important target for inducing mechanical pain in OA. Activating the Panx1 channel in mouse microglia releases IL-1β, thereby causing mechanical pain. In addition, upregulated IL-1β participates in inducing OA pain by activating neurotrophin (NGF) (Fig. 2) [18].

The tumour necrosis factor-α pathway

The tumour necrosis factor-α (TNF-α) pathway has been reported to be actively involved in the pathogenesis of osteoarthritis that is induced by mechanical loading. This pathway is activated through excessive mechanical load in vivo (90 g compressive mechanical loading was applied to rats for 96 h) and in vitro (CTS;36 kPa at 1HZ for 4 h) that triggers downstream inflammatory pathways to induce chondrocyte apoptosis and ECM degradation [19]. Studies have shown that TNF-α directly induces apoptosis and/or necrosis of rat chondrocytes in vitro (recombinant human 5 ng/ml) and activates matrix-degrading enzymes such as MMP1, MMP13, and ADAMT4/5 [20]. Li et al. [21] reported that activated TNF-α pathway upregulated IL-1β. TNF-α also activates the NF-κB pathway, MAPKs, and c-jun pathway and indirectly upregulates matrix-degrading enzymes such as collagen type 10 (Col10), MMP13, ADAMT5, and ADAMT9 that caused articular cartilage degradation [22]. Furthermore, NOD (nucleotide binding oligomerization domain)-like receptors (NLRP3) play an important role in the TNF-α pathway activation induced by mechanical load [23]. In their study, Grodzinsky et al. [24] documented that after inhibiting TNF-α pathway in chondrocytes, the level of inflammatory factors in articular cartilage was significantly downregulated, thereby alleviating ECM degradation and chondrocyte apoptosis (Fig. 3).

The nuclear factor kappa-B pathway

In vivo (rats on treadmill, 20 m/min for 45 minutes daily for four weeks) and in vitro (10% CTS, 0.5 Hz, 12 h) experiments have shown that overloading significantly upregulates the NF-κβ pathway and its downstream inflammatory factors [25], such as TNF-α, IL-1, IL-4, IL-6, and NO. It also activates matrix-degrading enzymes such as MMP3, MMP9, and MMP13 and ADAMT4, ADAMT5, and ADAMT9, thereby, enhancing OA progression [26]. Yang et al. [27] reported that physiological mechanical stress downregulated the NF-κβ pathway and reduced the inflammatory responses of chondrocytes. Inhibiting the NF-κβ pathway can effectively reduce overload-induced osteoarthritis incidences [28]. Chang et al. [29] reported that Gremlin-1, an extracellular antagonist of bone morphogenetic proteins (BMPs), is a key regulator of the NF-κβ pathway in overload-induced OA. A study by Suzuki et al. [30] found that overload-induced NF-κβ pathway significantly upregulated cathepsin K in cartilage matrix degradation.
In addition, NF-κβ activates downstream oxidative stress signals that indirectly induce ECM degradation and chondrocyte apoptosis. The NF-κβ pathway activates iNOS to upregulate NO levels in chondrocytes [31]. Excessive reactive oxide species (ROS) causes chondrocyte DNA mismatches, protein oxidative modification, and cell membrane lipid peroxidation that eventually result in chondrocyte apoptosis. The NF-κβ pathway also induces COX-2 and downregulates PGE2 to promote inflammation. NF-κβ and IL-1β pathways have a synergistic effect in the progression of OA. High IL-1β expression activates the NF-κβ pathway to induce chondrocyte apoptosis. Furthermore, NF-κβ upregulates downstream inflammatory cytokines such as hypoxia inducible factor-2α (HIF-2α), E74-like factor 3 (ELF3), SOX9, BMP2, IL-6, and TNF-α that promote chondrocyte apoptosis and ECM degradation [32]. Murahashi et al. [33] observed that overload-induced NF-κβ activates HIF-2α signals and upregulates downstream MMP13 and ADAMT5. This promotes cartilage matrix degradation and HIF-2α pathway. Furthermore, the Fas pathway is activated by NF-κβ to induce chondrocyte apoptosis. The overexpression of SOX9 downregulates OA factors such as MMPs and IL-1, which have been proven to induce OA in vivo and in vitro. The expression of SOX9 is antagonized by NF-κβ pathway in OA cartilage [34]. Besides, NF-κβ activates the ELF3 pathway and downstream MMPs causing cartilage matrix degradation [32]. The ELF3 pathway inhibits the SOX9 activator, thereby inhibiting the synthesis of SOX9-driven COL2A1, and promotes the progression of post-traumatic osteoarthritis (Fig. 4) [35].

The Wnt pathway

Wnt signals include canonical β-catenin-dependent and non-canonical β-catenin-independent signaling pathways. Wnt signals of different types and strengths regulate each other and maintain a delicate balance. Wnt16 signaling inhibits Wnt3a from over-activating the typical Wnt pathway, thereby preventing OA risk factors such as chondrocyte apoptosis and lubricin reduction [36].
It has been shown that under the same mechanical load, Wnt5a knockout rats are more susceptible to OA [37]. A study by Yuan reported that Wnt pathway inhibition by overload is characterized by OA manifestations such as induced ECM degradation, upregulated inflammatory factors, and trabecular bone erosion. The activation of the Wnt pathway can reverse these symptoms [38]. Physiological stress can activate Wnt signaling to inhibit osteoclast production and promote COL2A1 synthesis [39]. Wnt3a-RANKL signaling plays an antagonistic role in ECM degradation while Wnt16 plays a protective role by activating the PCP-mTORC1-PTHrP axis (a signal regulating cell proliferation) that inhibits ECM degradation [40]. The Wnt/β-catenin pathway significantly downregulates downstream coding products (MMP13 and ADAMT5) by regulating specific OA-related microRNAs such as miR-27a/b, miR-140, miR-146a/b, and miR365 [41]. The physiological mechanical load induces the upregulation of the Wnt5b/9a pathway thereby activating proteoglycan 4 (Prg4) to induce lubricin expression in the superficial area of articular cartilage [37], which significantly reduces joint friction.
Moreover, abnormal activation of the Wnt signal plays a role in cartilage destruction. Wnt1/3a signals that are induced by overloading significantly upregulate matrix degradation enzymes such as MMP13 and ADAMT5, ADAMT7, and ADAMT7 [40]. Wnt-5a phosphorylates JNK and PKC by activating atypical Wnt pathways thus inducing chondrocyte apoptosis. The overexpression of Wnt5a significantly upregulates MMPs and ADAMTs, thereby inducing chondrocyte apoptosis [42]. A study by Huang et al. reported that MIR-29a, a Wnt1 pathway transcription factor, upregulated caspase-3 expression causing chondrocyte apoptosis [43]. Moreover, Deshmukh et al. [44] observed that injecting SM04690 (a strong Wnt signal blocker) into a joint with a high expression of the Wnt signal effectively alleviated the overloading-induced damage [45]. Excessive Wnt signal levels upregulates ADAMT4, ADAMT5, ADAMT7, and ADAMT12 and promotes the degradation of ECM. However, IWP-2 (a Wnt signaling inhibitor) suppresses overloading-induced chondrocyte catabolism [46].
Different types and intensities of Wnt signals exert distinct effects on the articular cartilage. This may explain the different effects of excessive load and moderate load on the survival of articular cartilage. Moderate Wnt signal induced by physiological stress maintains the normal survival of articular cartilage. Meanwhile, excessive mechanical loads result in the abnormal expression of Wnt signal and induces changes associated with osteoarthritis such as chondrocyte apoptosis, matrix destruction, and osteophyte formation (Fig. 5).

The transforming growth factor-β pathway

Upregulation of transforming growth factor-β (TGF-β) in chondrocytes antagonizes chondrocyte apoptosis and matrix degradation caused by excessive mechanical load in vitro (36 kPa CTS at 0.5HZ for 2 h) and in vivo (OA model rats) [47]. TGF-β maintains functions of articular cartilage by transducing Smad2/3P signals thereby promote collagen (Col2a1) and fibronectin (Fn1) synthesis and prevent the degradation of cartilage matrix induced by overload [48]. Moreover, TGF-β inhibits MMP13 activation by stimulating IL-2/12 [49]. It has also been reported that TGF-β inhibits cartilage matrix degradation by upregulating SOX9 signals. 2,3′-Phosphoadenosine 5′-phosphosulfate synthase 2 (PAPSS2; one of TGF-β transcripts) regulates the physiological synthesis of proteoglycans in ECM [50]. Boyan et al. [51] showed that activating the TGF-β pathway with 25-hydroxyvitaminD3 [25 (OH) D3] can inhibit the IL-1β inflammatory pathway, antagonize chondrocyte apoptosis, and downregulate matrix-degrading enzymes. Elsewhere, Chavez et al. found that Prg4, one of the transcripts of TGF-β, promoted the secretion of articular cartilage lubricant to reduce the damage of articular cartilage caused by mechanical friction [52]. In addition, TGF-β confers anti-OA effect by activating some MIRNAs. Of note, TGF-β downregulates MIR-92a to inhibit adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK) and p38 pathway whereas it upregulates MIR-135b and MIR-140-5p to prevent chondrocyte apoptosis and cartilage matrix degradation (Fig. 6) [53].

MicroRNA and osteoarthritis

Numerous studies have reported that mechanical loading causes differential expression of microRNA (MIRNA) in articular cartilage and regulates the levels of matrix-degrading enzymes and inflammatory factors in osteoarthritis cartilage [54]. Specifically, it targets the expression of downstream transcripts. A study by Yang et al. [55] found that mechanical loading upregulated MIR-365 expression in chondrocytes. In addition, another study showed that MIR-365 overexpression activated the IL-1β pathway and increased the expression of matrix degrading enzyme (MMP13) and collagen type X (ColX). A study by De Palma et al. [56] reported that mechanical loading downregulated MIR-155 and MIR-181a in OA cartilage and activated the NF-κB pathway. Mechanical loading increased MIR-590-5p expression and activated TGF-β thereby enhancing chondrocyte apoptosis and autophagy [57]. Overloading activates the IL-1 β pathway and ADAMT5 and upregulates MIR-145 to promote ECM degradation [16]. Mechanical load upregulates MIR-365 in chondrocytes and promotes the expression of osteogenic genes and induce osteoarthritis osteophyte formation. Overloading upregulates MIR-204 expression, which then activates NF-κB and blocks the synthesis of proteoglycans, thus affecting ECM anabolism [58]. Overloading also upregulates miR-125a-5P, which in turn activates Smad2 to protect against Osteoarthritis [59]. Cheleschi et al. [41] indicated that mechanical loading upregulated MIR27a/b, MIR-140, and MIR-146a/b and downregulated MIR-365 expression, resulting in high expression of MMP13, ADAMT5, and HDAC-4. Changes in MIRNAs induced by mechanical loading do not always cause damage to chondrocytes. A study by Guan et al. [60] found that physiological mechanical load inhibited the Notch1, IL-6, and IL-1 signaling pathways and upregulated MIR-146a to alleviate osteoarthritis. It also upregulates MIR-204 and MIR211 leading to decreased expression of matrix-degrading enzymes in chondrocytes. It activates downstream Runx2 signals that maintain ECM homeostasis [61].

Oxidative stress pathway

Several recent studies have revealed that oxidative stress plays an important role in mechanical stress-induced OA. The ROS in chondrocytes is mainly produced by the mitochondrial electron transport chain. A study by Coleman et al. [62] reported that overload (1.0 MPa, 0.5 Hz) on bovine articular cartilage decreased proton membrane potential, respiratory activity and ATP production of chondrocytes, and elevated ROS production. It was reported that levels of antioxidant enzymes such as SOD, CAT, GSH, LDHA, and GPX in the cartilage of OA patients were significantly lower than those in normal articular cartilage, and excessive ROS level altered the redox balance in chondrocytes [63]. Excessive ROS generation can damage DNA and protein and cause lipid peroxidation in chondrocytes. ROS induces mitochondrial dysfunction and activates pro-inflammatory mediators and matrix degradation enzymes, resulting in OA. ROS has been found to participate in several inflammatory pathways in chondrocytes. Excessive ROS inhibits the PI3K/Akt pathway and activates MEK/ERK-MAPK/JNK pathway to upregulate caspase-3 and thereby causing chondrocyte apoptosis. Excessive ROS induced by overload activates inflammatory factors such as NF-κβ, protein kinase C, TNF-α, and IL-1β and eventually activates downstream iNOS, IL-8, and COX-2. Besides, excessive ROS activates matrix degradation enzymes such as MMP3, MMP9, MMP13, ADAMT4, and ADAMT5 in chondrocytes and degrade ECM components such as collagen type II, aggrecan, and proteoglycans [64]. Besides, antioxidants reduce overload-induced osteoarthritis which hence can be used for the clinical treatment of OA. Application of N-acetylcysteine (a common antioxidant) to overloaded chondrocytes effectively reversed the expression of MMPs and ADAMT4 and downregulation of type II collagen in chondrocytes of OA rats induced by mechanical load [65].
These mechanical stress-induced signal pathways are not isolated from each other. For example, IL-1β, TNF-α, NF-κB, and ROS activate each other and promote cartilage inflammation. Some experiments confirm that IL-1β, TNF-α, NF-κB, and ROS can downregulate TGF-β in articular cartilage. Different Wnt signal factors play different roles in mediating OA. For example, IL-1β, TNF-α, NF-κB, and ROS all upregulate Wnt5a/9a level, while inhibiting the expression of Wnt3a/16. Overexpression of Wnt3a/16 suppresses the levels of IL-1β, TNF-α, NF-κB, and ROS. Different types of microRNAs have completely different effects on the OA-related inflammatory factors (Table 1 and Fig. 7).
Table 1
Mechanical load regulates OA progress through MIRNAs
MIRs
Activated targets
Suppressed targets
MIR-365
MIR-155
MIR-181a
MIR-590-5p
MIR-146a
MIR-145
MIR-365
MIR-27a/b
MIR-140
MIR-204
MIR-211
IL-1/MMP13/Col X
 
NF-κB
 
NF-κB
 
TGF-β
 
 
Notch1/IL-6/IL-1
IL-1β/ADAMTs
 
Osteogenic gene
 
MMP13
 
ADAMT5
 
Runx2/NF-κB/
Synthetic PG/Runx2
 
miR-125a-5P
Smad2
 

Receptors of mechanical load in cartilage

There are receptors in cartilage that detect mechanical stress and regulate downstream signal pathways. Integrin (a cell adhesion molecule), for example, detects the mechanical signal in ECM and then activates the integrin-FAK-MAPKs axis to the upregulate OA-promoting factors such as MMPs, ADAMTs, and IL-6. Besides, integrin-fibrin-α5β1 axis detects mechanical signals and increases the expression of matrix degrading enzymes, MMP3, and MMP13 [66]. Hirose et al. [67] showed that integrin inhibitor cilengitide significantly downregulated several inflammatory factors such as IL-1β, TNF-α, MMPs, p-FAK, p-ERK, JNK, and p-p38 that were upregulated by overloading.
Moreover, ion channels play important roles in mechanical signal transduction. Mechanical signals are converted into electrical signals which then activate inflammatory signal pathways. A study by Lee et al. [68] observed that mechanical load generated Ca2+ transient using an atomic force microscope. Blocking this calcium transient with GsMTx4 (a piezoresistive peptide) significantly inhibited the apoptosis of chondrocytes after mechanical injury. Elsewhere, Xu et al. [69] reported that the mechanosensitive ion channel transient receptor potential channel 4 (TRPV4) of chondrocytes could detect mechanical stress and induce extracellular Ca2+ influx thereby upregulating levels of fas-related proteins and caspase-3, caspase-6, caspase-7, and caspase-8 triggering chondrocyte apoptosis. Some studies reported that voltage-dependent calcium channels are involved in the perception of persistent OA pain by modulating the CGRP and AC-PKA/PKC/MAPK signaling pathways in the dorsal root ganglion [70]. Besides, TRPA1 and RPV4 are closely associated with modulation of mechanical pain in OA. Mechanical loading activates matrix-degrading enzymes such as ADAMT5, ADAMT9, and MMP13 through TRPV1 and TRPV4 ion channels [71]. It has been reported that P2X7 activates Ca2+ and K + channels in chondrocytes under mechanical loading, causing chondrocyte injury [72].
Primary cilia are a key mechanical signal transduction factor in chondrocytes, which is a cytoskeletal organelle that extends from the cell surface into the external environment of the cell. Primary cilia are sensitive to mechanical stress on chondrocytes; when exposed to cyclic tensile strain, its length significantly shortens to absorb the stimulation of mechanical stress on chondrocytes. The main biological activity of primary cilia is the intraflagellar transport (IFT), a process in which primary cilia form microtubules so that soluble proteins and transmembrane receptors move along the microtubules [73]. In addition, primary cilia are also involved in mechanical load-related growth plate chondrocyte development. The dysfunction of primary cilia significantly impairs mechanical signal transduction. Some experiments have shown that animals with primary cilia damage, such as IFT-80 and IFT-88 knockout rats, have lower mechanical signal transduction efficiency and develop OA earlier [74].
Some other receptors in chondrocytes sense mechanical load. Gremlin-1 (an extracellular antagonist that secretes protein and bone morphogenetic protein (BMPs)) has been proposed to play a key role in perceiving mechanical load. Intra-articular injection of gremlin-1 antibody or chondrocyte-specific deletion gremlin-1 in OA model mice significantly reduces the sensitivity of articular cartilage to mechanical load [29]. Furthermore, hydrogen peroxide-inducible clone-5 (Hic-5) is considered to be a mechanically sensitive adaptor. The catabolic gene expression of chondrocytes induced by overloading significantly decreases after Hic-5 knockout [75].

The molecular mechanism of OA pain induced by mechanical load

Pain is the most prominent clinical symptom of osteoarthritis, which is also the main reason for patients to seek medical treatment. Long-term abnormal mechanical load induces structural changes in weight-bearing joints, which lead to joint pain. MRI studies have been used to confirm correlations between OA pain and these changes, including cartilage loss, osteophyte formation, and synovitis [76]. In addition, OA pain is also closely related to joint inflammation exacerbated by abnormal mechanical load, anti-inflammatory treatments, including glucocorticoids, and IL-1β/TNF-α inhibitors, which can effectively relieve pain symptoms during walking. In addition, there are multiple nociceptors in OA joints, which receive stimulus including mechanical/chemical/inflammatory mediators and convert harmful signals into pain. The transient receptor potential (TRP) family, possessing transmembrane domains, plays an important role in signal reception and conversion, which functions as ion channels when stimulated [77]. Some recent evidence suggests that nerve growth factor (NGF) increases the sensitivity of nociceptors in joints to mediate OA pain. NGF inhibitors can effectively alleviate OA pain [78]. A report by Carcolé et al. [79] indicated that sigma-1 receptors perceived and modulated mechanical pain in osteoarthritis. Furthermore, OA patients usually have peripheral nociceptors, a phenomenon in which the sensitivity of receptors increases and the excitability threshold of pain nerves decreases, which is clinically manifested as hyperalgesia (physiological stimulation can also cause pain).

Targets of molecular pathways for OA therapy

There are already quite a few promising molecular targeted therapies for OA clinically. It is reported that IL-1β inhibitors, such as Diacerein/AMG108/anakinra, effectively protected chondrocytes through reducing MMPs and ROS and preventing ECM degradation both in vitro (Diacerein, 20 μg/ml) and in vivo (rats, Diacerein at 5, 15 or 50 mg/kg) [34]. Adalimumab, a TNF-α inhibitor, has been shown to be efficacious in relieving the pathological symptoms of RA in clinic (DE001/003). However, it was less efficacious in reducing symptoms and inflammation factors of OA in a RCT(NCT00597623) compared with RA [80]. An iNOS inhibitor (SD-6010) had achieved significant effects on relieving OA symptoms of an RCT (NCT00565812). P38-MAPK pathway inhibitors (PH-797804) have also been verified effective anti-inflammatory and symptom relief in vitro (50 ng/ml) and vivo (rats,1 μM/kg) [81]. In addition to these, many other cytokines associated with OA have also become therapeutic targets. Some siRNA targeting pro-inflammatory mediators (including TNFα, IL-1β, IL-6, IL-18, and NF-κβ) effectively reduced the inflammation of OA [82].

Conclusion

It is currently believed that mechanical load affects osteoarthritis progression through a wide range of molecular pathways. Overload activates its downstream molecular pathways including IL-1β pathway, TNF-α pathway, NF-κB pathway, Wnt pathway, microRNA pathway, and oxidative stress pathway which induce apoptosis of chondrocytes and ECM degradation. This eventually causes osteoarthritis. It is important to note, however, that the mechanical load does not always cause harmful effects on articular the cartilage. In fact, physiological mechanical load can activate the TGF-β pathway to protect the cartilage. Many receptors detect mechanical signals in articular cartilage including integrins, ion channel receptors, Hic-5, Gremlin-1, and TRPV-4. These molecular mechanisms provide potential targets for clinical prevention and treatment of osteoarthritis induced by mechanical load.

Compliance with ethical standards

Conflict of interest

The authors declare that there are no conflicts of interest.

Ethical approval

Not applicable. This review does not conduct human or animal experiments.
Not applicable.
Not applicable.

Publisher’s note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Orthopädie & Unfallchirurgie

Kombi-Abonnement

Mit e.Med Orthopädie & Unfallchirurgie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Anhänge

Appendix

Table 2
List of the terms used in the searches
A
Terms related to osteoarthritis
B
Terms related to mechanical load
C
Terms related to molecular mechanism
Osteoarthritis
Mechanical load
Mechanism
Osteoarthritides
Mechanical loading
Mechanisms
Osteoarthrosis
Mechanical stress
Molecular mechanism
Osteoarthroses
Mechanical stresses
Molecular mechanisms
Arthritis, degenerative
Stresses, mechanical
Molecular
Arthritides, degenerative
Workloads
Pathway
Degenerative arthritides
Workload
Pathways
Degenerative arthritis
Work load
Signal
Arthrosis
Work loads
Signaling
Arthroses
Load
Receptor
Osteoarthrosis deformans
Loading
Cytokine
Pressure
Cytokines
Pressures
Interleukin
Mechanical pressures
IL
Mechanical pressure
Transforming growth factor
 
TGF
 
Tumor necrosis factors
 
TNF
 
Nuclear factor
 
NF-κβ
 
Wnt
 
MicroRNA
 
Activating factors
 
Suppressor factors
Literatur
2.
Zurück zum Zitat Zhu J, Zhu Y, Xiao W, Hu Y, Li Y (2020) Instability and excessive mechanical loading mediate subchondral bone changes to induce osteoarthritis. Ann Transl Med 8:350PubMedPubMedCentralCrossRef Zhu J, Zhu Y, Xiao W, Hu Y, Li Y (2020) Instability and excessive mechanical loading mediate subchondral bone changes to induce osteoarthritis. Ann Transl Med 8:350PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Jenei-Lanzl Z, Meurer A, Zaucke F (2019) Interleukin-1β signaling in osteoarthritis - chondrocytes in focus. Cell Signal 53:212–223PubMedCrossRef Jenei-Lanzl Z, Meurer A, Zaucke F (2019) Interleukin-1β signaling in osteoarthritis - chondrocytes in focus. Cell Signal 53:212–223PubMedCrossRef
4.
Zurück zum Zitat Dolzani P, Assirelli E, Pulsatelli L, Meliconi R, Mariani E, Neri S (2019) Ex vivo physiological compression of human osteoarthritis cartilage modulates cellular and matrix components. PLoS One 14:e0222947PubMedPubMedCentralCrossRef Dolzani P, Assirelli E, Pulsatelli L, Meliconi R, Mariani E, Neri S (2019) Ex vivo physiological compression of human osteoarthritis cartilage modulates cellular and matrix components. PLoS One 14:e0222947PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Szychlinska MA, Castrogiovanni P, Trovato FM, Nsir H, Zarrouk M, Lo Furno D, Di Rosa M, Imbesi R, Musumeci G (2019) Physical activity and Mediterranean diet based on olive tree phenolic compounds from two different geographical areas have protective effects on early osteoarthritis, muscle atrophy and hepatic steatosis. Eur J Nutr 58:565–581PubMedCrossRef Szychlinska MA, Castrogiovanni P, Trovato FM, Nsir H, Zarrouk M, Lo Furno D, Di Rosa M, Imbesi R, Musumeci G (2019) Physical activity and Mediterranean diet based on olive tree phenolic compounds from two different geographical areas have protective effects on early osteoarthritis, muscle atrophy and hepatic steatosis. Eur J Nutr 58:565–581PubMedCrossRef
6.
Zurück zum Zitat Wang Y, Chen Y, Chen Y, Zhou B, Shan X, Yang G (2018) Eriodictyol inhibits IL-1β-induced inflammatory response in human osteoarthritis chondrocytes. Biomed Pharmacother 107:1128–1134PubMedCrossRef Wang Y, Chen Y, Chen Y, Zhou B, Shan X, Yang G (2018) Eriodictyol inhibits IL-1β-induced inflammatory response in human osteoarthritis chondrocytes. Biomed Pharmacother 107:1128–1134PubMedCrossRef
7.
Zurück zum Zitat Wu D, Zhu X, Kang X, Huang H, Yu J, Pan J, Zhang X (2019) The protective effect of sophocarpine in osteoarthritis: an in vitro and in vivo study. Int Immunopharmacol 67:145–151PubMedCrossRef Wu D, Zhu X, Kang X, Huang H, Yu J, Pan J, Zhang X (2019) The protective effect of sophocarpine in osteoarthritis: an in vitro and in vivo study. Int Immunopharmacol 67:145–151PubMedCrossRef
8.
Zurück zum Zitat Feng Z, Li X, Lin J, Zheng W, Hu Z, Xuan J, Ni W, Pan X (2017) Oleuropein inhibits the IL-1β-induced expression of inflammatory mediators by suppressing the activation of NF-κB and MAPKs in human osteoarthritis chondrocytes. Food Funct 8:3737–3744PubMedCrossRef Feng Z, Li X, Lin J, Zheng W, Hu Z, Xuan J, Ni W, Pan X (2017) Oleuropein inhibits the IL-1β-induced expression of inflammatory mediators by suppressing the activation of NF-κB and MAPKs in human osteoarthritis chondrocytes. Food Funct 8:3737–3744PubMedCrossRef
9.
Zurück zum Zitat Huang X, Xi Y, Pan Q, Mao Z, Zhang R, Ma X, You H (2018) Caffeic acid protects against IL-1β-induced inflammatory responses and cartilage degradation in articular chondrocytes. Biomed Pharmacother 107:433–439PubMedCrossRef Huang X, Xi Y, Pan Q, Mao Z, Zhang R, Ma X, You H (2018) Caffeic acid protects against IL-1β-induced inflammatory responses and cartilage degradation in articular chondrocytes. Biomed Pharmacother 107:433–439PubMedCrossRef
10.
Zurück zum Zitat Wu J, Huang JF, Qin XX, Hu F, Chen ZF, Zheng Y, Liu YX, Cai XH (2018) Platelet-rich plasma inhibits Wnt/β-catenin signaling in rabbit cartilage cells activated by IL-1β. Int Immunopharmacol 55:282–289PubMedCrossRef Wu J, Huang JF, Qin XX, Hu F, Chen ZF, Zheng Y, Liu YX, Cai XH (2018) Platelet-rich plasma inhibits Wnt/β-catenin signaling in rabbit cartilage cells activated by IL-1β. Int Immunopharmacol 55:282–289PubMedCrossRef
11.
Zurück zum Zitat Zhang Y, Zhou S, Cai W, Han G, Li J, Chen M, Li H (2020) Hypoxia/reoxygenation activates the JNK pathway and accelerates synovial senescence. Mol Med Rep 22:265–276PubMedPubMedCentralCrossRef Zhang Y, Zhou S, Cai W, Han G, Li J, Chen M, Li H (2020) Hypoxia/reoxygenation activates the JNK pathway and accelerates synovial senescence. Mol Med Rep 22:265–276PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Lu C, Li Y, Hu S, Cai Y, Yang Z, Peng K (2018) Scoparone prevents IL-1β-induced inflammatory response in human osteoarthritis chondrocytes through the PI3K/Akt/NF-κB pathway. Biomed Pharmacother 106:1169–1174PubMedCrossRef Lu C, Li Y, Hu S, Cai Y, Yang Z, Peng K (2018) Scoparone prevents IL-1β-induced inflammatory response in human osteoarthritis chondrocytes through the PI3K/Akt/NF-κB pathway. Biomed Pharmacother 106:1169–1174PubMedCrossRef
13.
Zurück zum Zitat Wu D, Jin S, Lin Z, Chen R, Pan T, Kang X, Huang H, Lin C, Pan J (2018) Sauchinone inhibits IL-1β induced catabolism and hypertrophy in mouse chondrocytes to attenuate osteoarthritis via Nrf2/HO-1 and NF-κB pathways. Int Immunopharmacol 62:181–190PubMedCrossRef Wu D, Jin S, Lin Z, Chen R, Pan T, Kang X, Huang H, Lin C, Pan J (2018) Sauchinone inhibits IL-1β induced catabolism and hypertrophy in mouse chondrocytes to attenuate osteoarthritis via Nrf2/HO-1 and NF-κB pathways. Int Immunopharmacol 62:181–190PubMedCrossRef
14.
Zurück zum Zitat Na HS, Park JS, Cho KH, Kwon JY, Choi J, Jhun J, Kim SJ, Park SH, Cho ML (2020) Interleukin-1-interleukin-17 signaling axis induces cartilage destruction and promotes experimental osteoarthritis. Front Immunol 11:730PubMedPubMedCentralCrossRef Na HS, Park JS, Cho KH, Kwon JY, Choi J, Jhun J, Kim SJ, Park SH, Cho ML (2020) Interleukin-1-interleukin-17 signaling axis induces cartilage destruction and promotes experimental osteoarthritis. Front Immunol 11:730PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Ohtsuki T, Hatipoglu OF, Asano K, Inagaki J, Nishida K, Hirohata S (2020) Induction of CEMIP in chondrocytes by inflammatory cytokines: underlying mechanisms and potential involvement in osteoarthritis. Int J Mol Sci 21 Ohtsuki T, Hatipoglu OF, Asano K, Inagaki J, Nishida K, Hirohata S (2020) Induction of CEMIP in chondrocytes by inflammatory cytokines: underlying mechanisms and potential involvement in osteoarthritis. Int J Mol Sci 21
16.
Zurück zum Zitat Liu C, Ren S, Zhao S, Wang Y (2019) LncRNA MALAT1/MiR-145 adjusts IL-1β-induced chondrocytes viability and cartilage matrix degradation by regulating ADAMTS5 in human osteoarthritis. Yonsei Med J 60:1081–1092PubMedPubMedCentralCrossRef Liu C, Ren S, Zhao S, Wang Y (2019) LncRNA MALAT1/MiR-145 adjusts IL-1β-induced chondrocytes viability and cartilage matrix degradation by regulating ADAMTS5 in human osteoarthritis. Yonsei Med J 60:1081–1092PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Wang F, Liu J, Chen X, Zheng X, Qu N, Zhang B, Xia C (2019) IL-1β 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 21:171PubMedPubMedCentralCrossRef Wang F, Liu J, Chen X, Zheng X, Qu N, Zhang B, Xia C (2019) IL-1β 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 21:171PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Pecchi E, Priam S, Gosset M, Pigenet A, Sudre L, Laiguillon MC, Berenbaum F, Houard X (2014) Induction of nerve growth factor expression and release by mechanical and inflammatory stimuli in chondrocytes: possible involvement in osteoarthritis pain. Arthritis Res Ther 16:R16PubMedPubMedCentralCrossRef Pecchi E, Priam S, Gosset M, Pigenet A, Sudre L, Laiguillon MC, Berenbaum F, Houard X (2014) Induction of nerve growth factor expression and release by mechanical and inflammatory stimuli in chondrocytes: possible involvement in osteoarthritis pain. Arthritis Res Ther 16:R16PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Zhang C, Lin S, Li T, Jiang Y, Huang Z, Wen J, Cheng W, Li H (2017) Mechanical force-mediated pathological cartilage thinning is regulated by necroptosis and apoptosis. Osteoarthr Cartil 25:1324–1334CrossRef Zhang C, Lin S, Li T, Jiang Y, Huang Z, Wen J, Cheng W, Li H (2017) Mechanical force-mediated pathological cartilage thinning is regulated by necroptosis and apoptosis. Osteoarthr Cartil 25:1324–1334CrossRef
20.
Zurück zum Zitat Wang C, Al-Ani MK, Sha Y, Chi Q, Dong N, Yang L, Xu K (2019) Psoralen protects chondrocytes, exhibits anti-inflammatory effects on synoviocytes, and attenuates monosodium iodoacetate-induced osteoarthritis. Int J Biol Sci 15:229–238PubMedPubMedCentralCrossRef Wang C, Al-Ani MK, Sha Y, Chi Q, Dong N, Yang L, Xu K (2019) Psoralen protects chondrocytes, exhibits anti-inflammatory effects on synoviocytes, and attenuates monosodium iodoacetate-induced osteoarthritis. Int J Biol Sci 15:229–238PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Li YN, Fan ML, Liu HQ, Ma B, Dai WL, Yu BY, Liu JH (2019) Dihydroartemisinin derivative DC32 inhibits inflammatory response in osteoarthritic synovium through regulating Nrf2/NF-κB pathway. Int Immunopharmacol 74:105701PubMedCrossRef Li YN, Fan ML, Liu HQ, Ma B, Dai WL, Yu BY, Liu JH (2019) Dihydroartemisinin derivative DC32 inhibits inflammatory response in osteoarthritic synovium through regulating Nrf2/NF-κB pathway. Int Immunopharmacol 74:105701PubMedCrossRef
22.
Zurück zum Zitat Yan L, Zhou L, Xie D, Du W, Chen F, Yuan Q, Tong P, Shan L, Efferth T (2019) Chondroprotective effects of platelet lysate towards monoiodoacetate-induced arthritis by suppression of TNF-α-induced activation of NF-ĸB pathway in chondrocytes. Aging 11:2797–2811PubMedPubMedCentralCrossRef Yan L, Zhou L, Xie D, Du W, Chen F, Yuan Q, Tong P, Shan L, Efferth T (2019) Chondroprotective effects of platelet lysate towards monoiodoacetate-induced arthritis by suppression of TNF-α-induced activation of NF-ĸB pathway in chondrocytes. Aging 11:2797–2811PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat McAllister MJ, Chemaly M, Eakin AJ, Gibson DS, McGilligan VE (2018) NLRP3 as a potentially novel biomarker for the management of osteoarthritis. Osteoarthr Cartil 26:612–619CrossRef McAllister MJ, Chemaly M, Eakin AJ, Gibson DS, McGilligan VE (2018) NLRP3 as a potentially novel biomarker for the management of osteoarthritis. Osteoarthr Cartil 26:612–619CrossRef
24.
Zurück zum Zitat Grodzinsky AJ, Wang Y, Kakar S, Vrahas MS, Evans CH (2017) Intra-articular dexamethasone to inhibit the development of post-traumatic osteoarthritis. J Orthop Res: Off Publ Orthop Res Soc 35:406–411CrossRef Grodzinsky AJ, Wang Y, Kakar S, Vrahas MS, Evans CH (2017) Intra-articular dexamethasone to inhibit the development of post-traumatic osteoarthritis. J Orthop Res: Off Publ Orthop Res Soc 35:406–411CrossRef
25.
Zurück zum Zitat Choi MC, Jo J, Park J, Kang HK, Park Y (2019) NF-κB signaling pathways in osteoarthritic cartilage destruction. Cells 8 Choi MC, Jo J, Park J, Kang HK, Park Y (2019) NF-κB signaling pathways in osteoarthritic cartilage destruction. Cells 8
26.
Zurück zum Zitat Liang Y, Chen S, Yang Y, Lan C, Zhang G, Ji Z, Lin H (2018) Vasoactive intestinal peptide alleviates osteoarthritis effectively via inhibiting NF-κB signaling pathway. J Biomed Sci 25:25PubMedPubMedCentralCrossRef Liang Y, Chen S, Yang Y, Lan C, Zhang G, Ji Z, Lin H (2018) Vasoactive intestinal peptide alleviates osteoarthritis effectively via inhibiting NF-κB signaling pathway. J Biomed Sci 25:25PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Yang Y, Wang Y, Kong Y, Zhang X, Zhang H, Gang Y, Bai L (2019) Mechanical stress protects against osteoarthritis via regulation of the AMPK/NF-κB signaling pathway. J Cell Physiol 234:9156–9167PubMedCrossRef Yang Y, Wang Y, Kong Y, Zhang X, Zhang H, Gang Y, Bai L (2019) Mechanical stress protects against osteoarthritis via regulation of the AMPK/NF-κB signaling pathway. J Cell Physiol 234:9156–9167PubMedCrossRef
28.
Zurück zum Zitat Yan H, Duan X, Collins KH, Springer LE, Guilak F, Wickline SA, Rai MF, Pan H, Pham CTN (2019) Nanotherapy targeting NF-κB attenuates acute pain after joint injury. Precis Nanomed 2:245–248PubMedPubMedCentralCrossRef Yan H, Duan X, Collins KH, Springer LE, Guilak F, Wickline SA, Rai MF, Pan H, Pham CTN (2019) Nanotherapy targeting NF-κB attenuates acute pain after joint injury. Precis Nanomed 2:245–248PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Chang SH, Mori D, Kobayashi H, Mori Y, Nakamoto H, Okada K, Taniguchi Y, Sugita S, Yano F, Chung UI et al (2019) Excessive mechanical loading promotes osteoarthritis through the gremlin-1-NF-κB pathway. Nat Commun 10:1442PubMedPubMedCentralCrossRef Chang SH, Mori D, Kobayashi H, Mori Y, Nakamoto H, Okada K, Taniguchi Y, Sugita S, Yano F, Chung UI et al (2019) Excessive mechanical loading promotes osteoarthritis through the gremlin-1-NF-κB pathway. Nat Commun 10:1442PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Suzuki M, Takahashi N, Sobue Y, Ohashi Y, Kishimoto K, Hattori K, Ishiguro N, Kojima T (2020) Hyaluronan suppresses enhanced cathepsin K expression via activation of NF-κB with mechanical stress loading in a human chondrocytic HCS-2/8 cells. Sci Rep 10:216PubMedPubMedCentralCrossRef Suzuki M, Takahashi N, Sobue Y, Ohashi Y, Kishimoto K, Hattori K, Ishiguro N, Kojima T (2020) Hyaluronan suppresses enhanced cathepsin K expression via activation of NF-κB with mechanical stress loading in a human chondrocytic HCS-2/8 cells. Sci Rep 10:216PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Ostojic M, Soljic V, Vukojevic K, Dapic T (2017) Immunohistochemical characterization of early and advanced knee osteoarthritis by NF-κB and iNOS expression. J Orthop Res: Off Publ Orthop Res Soc 35:1990–1997CrossRef Ostojic M, Soljic V, Vukojevic K, Dapic T (2017) Immunohistochemical characterization of early and advanced knee osteoarthritis by NF-κB and iNOS expression. J Orthop Res: Off Publ Orthop Res Soc 35:1990–1997CrossRef
32.
Zurück zum Zitat Wondimu EB, Culley KL, Quinn J, Chang J, Dragomir CL, Plumb DA, Goldring MB, Otero M (2018) Elf3 contributes to cartilage degradation in vivo in a surgical model of post-traumatic osteoarthritis. Sci Rep 8:6438PubMedPubMedCentralCrossRef Wondimu EB, Culley KL, Quinn J, Chang J, Dragomir CL, Plumb DA, Goldring MB, Otero M (2018) Elf3 contributes to cartilage degradation in vivo in a surgical model of post-traumatic osteoarthritis. Sci Rep 8:6438PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Murahashi Y, Yano F, Kobayashi H, Makii Y, Iba K, Yamashita T, Tanaka S, Saito T (2018) Intra-articular administration of IκBα kinase inhibitor suppresses mouse knee osteoarthritis via downregulation of the NF-κB/HIF-2α axis. Sci Rep 8:16475PubMedPubMedCentralCrossRef Murahashi Y, Yano F, Kobayashi H, Makii Y, Iba K, Yamashita T, Tanaka S, Saito T (2018) Intra-articular administration of IκBα kinase inhibitor suppresses mouse knee osteoarthritis via downregulation of the NF-κB/HIF-2α axis. Sci Rep 8:16475PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Nishimura R, Hata K, Takahata Y, Murakami T, Nakamura E, Ohkawa M, Ruengsinpinya L (2020) Role of signal transduction pathways and transcription factors in cartilage and joint diseases. Int J Mol Sci 21 Nishimura R, Hata K, Takahata Y, Murakami T, Nakamura E, Ohkawa M, Ruengsinpinya L (2020) Role of signal transduction pathways and transcription factors in cartilage and joint diseases. Int J Mol Sci 21
35.
Zurück zum Zitat Otero M, Peng H, Hachem KE, Culley KL, Wondimu EB, Quinn J, Asahara H, Tsuchimochi K, Hashimoto K, Goldring MB (2017) ELF3 modulates type II collagen gene (COL2A1) transcription in chondrocytes by inhibiting SOX9-CBP/p300-driven histone acetyltransferase activity. Connect Tissue Res 58:15–26PubMedCrossRef Otero M, Peng H, Hachem KE, Culley KL, Wondimu EB, Quinn J, Asahara H, Tsuchimochi K, Hashimoto K, Goldring MB (2017) ELF3 modulates type II collagen gene (COL2A1) transcription in chondrocytes by inhibiting SOX9-CBP/p300-driven histone acetyltransferase activity. Connect Tissue Res 58:15–26PubMedCrossRef
36.
Zurück zum Zitat Nalesso G, Thomas BL, Sherwood JC, Yu J, Addimanda O, Eldridge SE, Thorup AS, Dale L, Schett G, Zwerina J et al (2017) WNT16 antagonises excessive canonical WNT activation and protects cartilage in osteoarthritis. Ann Rheum Dis 76:218–226PubMedCrossRef Nalesso G, Thomas BL, Sherwood JC, Yu J, Addimanda O, Eldridge SE, Thorup AS, Dale L, Schett G, Zwerina J et al (2017) WNT16 antagonises excessive canonical WNT activation and protects cartilage in osteoarthritis. Ann Rheum Dis 76:218–226PubMedCrossRef
37.
Zurück zum Zitat Xuan F, Yano F, Mori D, Chijimatsu R, Maenohara Y, Nakamoto H, Mori Y, Makii Y, Oichi T, Taketo MM et al (2019) Wnt/β-catenin signaling contributes to articular cartilage homeostasis through lubricin induction in the superficial zone. Arthritis Res Ther 21:247PubMedPubMedCentralCrossRef Xuan F, Yano F, Mori D, Chijimatsu R, Maenohara Y, Nakamoto H, Mori Y, Makii Y, Oichi T, Taketo MM et al (2019) Wnt/β-catenin signaling contributes to articular cartilage homeostasis through lubricin induction in the superficial zone. Arthritis Res Ther 21:247PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Jiang YY, Wen J, Gong C, Lin S, Zhang CX, Chen S, Cheng W, Li H (2018) BIO alleviated compressive mechanical force-mediated mandibular cartilage pathological changes through Wnt/β-catenin signaling activation. J Orthop Res: Off Publ Orthop Res Soc 36:1228–1237 Jiang YY, Wen J, Gong C, Lin S, Zhang CX, Chen S, Cheng W, Li H (2018) BIO alleviated compressive mechanical force-mediated mandibular cartilage pathological changes through Wnt/β-catenin signaling activation. J Orthop Res: Off Publ Orthop Res Soc 36:1228–1237
39.
Zurück zum Zitat Lietman C, Wu B, Lechner S, Shinar A, Sehgal M, Rossomacha E, Datta P, Sharma A, Gandhi R, Kapoor M et al (2018) Inhibition of Wnt/β-catenin signaling ameliorates osteoarthritis in a murine model of experimental osteoarthritis. JCI Insight 3 Lietman C, Wu B, Lechner S, Shinar A, Sehgal M, Rossomacha E, Datta P, Sharma A, Gandhi R, Kapoor M et al (2018) Inhibition of Wnt/β-catenin signaling ameliorates osteoarthritis in a murine model of experimental osteoarthritis. JCI Insight 3
40.
Zurück zum Zitat Pérez-García S, Carrión M, Villanueva-Romero R, Hermida-Gómez T, Fernández-Moreno M, Mellado M, Blanco FJ, Juarranz Y, Gomariz RP (2019) Wnt and RUNX2 mediate cartilage breakdown by osteoarthritis synovial fibroblast-derived ADAMTS-7 and -12. J Cell Mol Med 23:3974–3983PubMedPubMedCentralCrossRef Pérez-García S, Carrión M, Villanueva-Romero R, Hermida-Gómez T, Fernández-Moreno M, Mellado M, Blanco FJ, Juarranz Y, Gomariz RP (2019) Wnt and RUNX2 mediate cartilage breakdown by osteoarthritis synovial fibroblast-derived ADAMTS-7 and -12. J Cell Mol Med 23:3974–3983PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Cheleschi S, De Palma A, Pecorelli A, Pascarelli NA, Valacchi G, Belmonte G, Carta S, Galeazzi M, Fioravanti A (2017) Hydrostatic pressure regulates microRNA expression levels in osteoarthritic chondrocyte cultures via the Wnt/β-catenin pathway. Int J Mol Sci 18 Cheleschi S, De Palma A, Pecorelli A, Pascarelli NA, Valacchi G, Belmonte G, Carta S, Galeazzi M, Fioravanti A (2017) Hydrostatic pressure regulates microRNA expression levels in osteoarthritic chondrocyte cultures via the Wnt/β-catenin pathway. Int J Mol Sci 18
42.
Zurück zum Zitat Huang G, Chubinskaya S, Liao W, Loeser RF (2017) Wnt5a induces catabolic signaling and matrix metalloproteinase production in human articular chondrocytes. Osteoarthr Cartil 25:1505–1515CrossRef Huang G, Chubinskaya S, Liao W, Loeser RF (2017) Wnt5a induces catabolic signaling and matrix metalloproteinase production in human articular chondrocytes. Osteoarthr Cartil 25:1505–1515CrossRef
43.
Zurück zum Zitat Hu S, Mao G, Zhang Z, Wu P, Wen X, Liao W, Zhang Z (2019) MicroRNA-320c inhibits development of osteoarthritis through downregulation of canonical Wnt signaling pathway. Life Sci 228:242–250PubMedCrossRef Hu S, Mao G, Zhang Z, Wu P, Wen X, Liao W, Zhang Z (2019) MicroRNA-320c inhibits development of osteoarthritis through downregulation of canonical Wnt signaling pathway. Life Sci 228:242–250PubMedCrossRef
44.
Zurück zum Zitat Deshmukh V, Hu H, Barroga C, Bossard C, Kc S, Dellamary L, Stewart J, Chiu K, Ibanez M, Pedraza M et al (2018) A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee. Osteoarthr Cartil 26:18–27CrossRef Deshmukh V, Hu H, Barroga C, Bossard C, Kc S, Dellamary L, Stewart J, Chiu K, Ibanez M, Pedraza M et al (2018) A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee. Osteoarthr Cartil 26:18–27CrossRef
45.
Zurück zum Zitat Praxenthaler H, Krämer E, Weisser M, Hecht N, Fischer J, Grossner T, Richter W (2018) Extracellular matrix content and WNT/β-catenin levels of cartilage determine the chondrocyte response to compressive load. Biochim Biophys Acta Mol basis Dis 1864:851–859PubMedCrossRef Praxenthaler H, Krämer E, Weisser M, Hecht N, Fischer J, Grossner T, Richter W (2018) Extracellular matrix content and WNT/β-catenin levels of cartilage determine the chondrocyte response to compressive load. Biochim Biophys Acta Mol basis Dis 1864:851–859PubMedCrossRef
46.
Zurück zum Zitat Huang X, Chen Z, Shi W, Zhang R, Li L, Liu H, Wu L (2019) TMF inhibits miR-29a/Wnt/β-catenin signaling through upregulating Foxo3a activity in osteoarthritis chondrocytes. Drug Des Dev Ther 13:2009–2019CrossRef Huang X, Chen Z, Shi W, Zhang R, Li L, Liu H, Wu L (2019) TMF inhibits miR-29a/Wnt/β-catenin signaling through upregulating Foxo3a activity in osteoarthritis chondrocytes. Drug Des Dev Ther 13:2009–2019CrossRef
47.
Zurück zum Zitat Luo Z, Jiang L, Xu Y, Li H, Xu W, Wu S, Wang Y, Tang Z, Lv Y, Yang L (2015) Mechano growth factor (MGF) and transforming growth factor (TGF)-β3 functionalized silk scaffolds enhance articular hyaline cartilage regeneration in rabbit model. Biomaterials 52:463–475PubMedCrossRef Luo Z, Jiang L, Xu Y, Li H, Xu W, Wu S, Wang Y, Tang Z, Lv Y, Yang L (2015) Mechano growth factor (MGF) and transforming growth factor (TGF)-β3 functionalized silk scaffolds enhance articular hyaline cartilage regeneration in rabbit model. Biomaterials 52:463–475PubMedCrossRef
48.
Zurück zum Zitat Madej W, Buma P, van der Kraan P (2016) Inflammatory conditions partly impair the mechanically mediated activation of Smad2/3 signaling in articular cartilage. Arthritis Res Ther 18:146PubMedPubMedCentralCrossRef Madej W, Buma P, van der Kraan P (2016) Inflammatory conditions partly impair the mechanically mediated activation of Smad2/3 signaling in articular cartilage. Arthritis Res Ther 18:146PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Lu W, He Z, Shi J, Wang Z, Wu W, Liu J, Kang H, Li F, Liang S (2019) AMD3100 attenuates post-traumatic osteoarthritis by maintaining transforming growth factor-β1-induced expression of tissue inhibitor of metalloproteinase-3 via the phosphatidylinositol 3-kinase/Akt pathway. Front Pharmacol 10:1554PubMedCrossRef Lu W, He Z, Shi J, Wang Z, Wu W, Liu J, Kang H, Li F, Liang S (2019) AMD3100 attenuates post-traumatic osteoarthritis by maintaining transforming growth factor-β1-induced expression of tissue inhibitor of metalloproteinase-3 via the phosphatidylinositol 3-kinase/Akt pathway. Front Pharmacol 10:1554PubMedCrossRef
50.
Zurück zum Zitat Chavez RD, Coricor G, Perez J, Seo HS, Serra R (2017) SOX9 protein is stabilized by TGF-β and regulates PAPSS2 mRNA expression in chondrocytes. Osteoarthr Cartil 25:332–340CrossRef Chavez RD, Coricor G, Perez J, Seo HS, Serra R (2017) SOX9 protein is stabilized by TGF-β and regulates PAPSS2 mRNA expression in chondrocytes. Osteoarthr Cartil 25:332–340CrossRef
51.
Zurück zum Zitat Boyan BD, Hyzy SL, Pan Q, Scott KM, Coutts RD, Healey R, Schwartz Z (2016) 24R,25-Dihydroxyvitamin D3 protects against articular cartilage damage following anterior cruciate ligament transection in male rats. PLoS One 11:e0161782PubMedPubMedCentralCrossRef Boyan BD, Hyzy SL, Pan Q, Scott KM, Coutts RD, Healey R, Schwartz Z (2016) 24R,25-Dihydroxyvitamin D3 protects against articular cartilage damage following anterior cruciate ligament transection in male rats. PLoS One 11:e0161782PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Chavez RD, Sohn P, Serra R (2019) Prg4 prevents osteoarthritis induced by dominant-negative interference of TGF-ß signaling in mice. PLoS One 14:e0210601PubMedPubMedCentralCrossRef Chavez RD, Sohn P, Serra R (2019) Prg4 prevents osteoarthritis induced by dominant-negative interference of TGF-ß signaling in mice. PLoS One 14:e0210601PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Kuo SJ, Liu SC, Huang YL, Tsai CH, Fong YC, Hsu HC, Tang CH (2019) TGF-β1 enhances FOXO3 expression in human synovial fibroblasts by inhibiting miR-92a through AMPK and p38 pathways. Aging 11:4075–4089PubMedPubMedCentralCrossRef Kuo SJ, Liu SC, Huang YL, Tsai CH, Fong YC, Hsu HC, Tang CH (2019) TGF-β1 enhances FOXO3 expression in human synovial fibroblasts by inhibiting miR-92a through AMPK and p38 pathways. Aging 11:4075–4089PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Kwak YH, Kwak DK, Kim NY, Kim YJ, Lim JS, Yoo JH (2020) Significant changes in synovial fluid microRNAs after high tibial osteotomy in medial compartmental knee osteoarthritis: identification of potential prognostic biomarkers. PLoS One 15:e0227596PubMedPubMedCentralCrossRef Kwak YH, Kwak DK, Kim NY, Kim YJ, Lim JS, Yoo JH (2020) Significant changes in synovial fluid microRNAs after high tibial osteotomy in medial compartmental knee osteoarthritis: identification of potential prognostic biomarkers. PLoS One 15:e0227596PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Yang X, Guan Y, Tian S, Wang Y, Sun K, Chen Q (2016) Mechanical and IL-1β responsive miR-365 contributes to osteoarthritis development by targeting histone deacetylase 4. Int J Mol Sci 17:436PubMedPubMedCentralCrossRef Yang X, Guan Y, Tian S, Wang Y, Sun K, Chen Q (2016) Mechanical and IL-1β responsive miR-365 contributes to osteoarthritis development by targeting histone deacetylase 4. Int J Mol Sci 17:436PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat De Palma A, Cheleschi S, Pascarelli NA, Giannotti S, Galeazzi M, Fioravanti A (2018) Hydrostatic pressure as epigenetic modulator in chondrocyte cultures: a study on miRNA-155, miRNA-181a and miRNA-223 expression levels. J Biomech 66:165–169PubMedCrossRef De Palma A, Cheleschi S, Pascarelli NA, Giannotti S, Galeazzi M, Fioravanti A (2018) Hydrostatic pressure as epigenetic modulator in chondrocyte cultures: a study on miRNA-155, miRNA-181a and miRNA-223 expression levels. J Biomech 66:165–169PubMedCrossRef
57.
Zurück zum Zitat Wang J, Zhang Y, Song W, Ma T, Wang K (2018) microRNA-590-5p targets transforming growth factor β1 to promote chondrocyte apoptosis and autophagy in response to mechanical pressure injury. J Cell Biochem 119:9931–9940PubMedCrossRef Wang J, Zhang Y, Song W, Ma T, Wang K (2018) microRNA-590-5p targets transforming growth factor β1 to promote chondrocyte apoptosis and autophagy in response to mechanical pressure injury. J Cell Biochem 119:9931–9940PubMedCrossRef
58.
Zurück zum Zitat Kang D, Shin J, Cho Y, Kim HS, Gu YR, Kim H, You KT, Chang MJ, Chang CB, Kang SB et al (2019) Stress-activated miR-204 governs senescent phenotypes of chondrocytes to promote osteoarthritis development. Sci Transl Med 11 Kang D, Shin J, Cho Y, Kim HS, Gu YR, Kim H, You KT, Chang MJ, Chang CB, Kang SB et al (2019) Stress-activated miR-204 governs senescent phenotypes of chondrocytes to promote osteoarthritis development. Sci Transl Med 11
59.
Zurück zum Zitat Shen P, Yang Y, Liu G, Chen W, Chen J, Wang Q, Gao H, Fan S, Shen S, Zhao X (2020) CircCDK14 protects against osteoarthritis by sponging miR-125a-5p and promoting the expression of Smad2. Theranostics 10:9113–9131PubMedPubMedCentralCrossRef Shen P, Yang Y, Liu G, Chen W, Chen J, Wang Q, Gao H, Fan S, Shen S, Zhao X (2020) CircCDK14 protects against osteoarthritis by sponging miR-125a-5p and promoting the expression of Smad2. Theranostics 10:9113–9131PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Guan YJ, Li J, Yang X, Du S, Ding J, Gao Y, Zhang Y, Yang K, Chen Q (2018) Evidence that miR-146a attenuates aging- and trauma-induced osteoarthritis by inhibiting Notch1, IL-6, and IL-1 mediated catabolism. Aging Cell 17:e12752PubMedPubMedCentralCrossRef Guan YJ, Li J, Yang X, Du S, Ding J, Gao Y, Zhang Y, Yang K, Chen Q (2018) Evidence that miR-146a attenuates aging- and trauma-induced osteoarthritis by inhibiting Notch1, IL-6, and IL-1 mediated catabolism. Aging Cell 17:e12752PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Huang J, Zhao L, Fan Y, Liao L, Ma PX, Xiao G, Chen D (2019) The microRNAs miR-204 and miR-211 maintain joint homeostasis and protect against osteoarthritis progression. Nat Commun 10:2876PubMedPubMedCentralCrossRef Huang J, Zhao L, Fan Y, Liao L, Ma PX, Xiao G, Chen D (2019) The microRNAs miR-204 and miR-211 maintain joint homeostasis and protect against osteoarthritis progression. Nat Commun 10:2876PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Coleman MC, Ramakrishnan PS, Brouillette MJ, Martin JA (2016) Injurious loading of articular cartilage compromises chondrocyte respiratory function. Arthritis Rheumatol 68:662–671PubMedPubMedCentralCrossRef Coleman MC, Ramakrishnan PS, Brouillette MJ, Martin JA (2016) Injurious loading of articular cartilage compromises chondrocyte respiratory function. Arthritis Rheumatol 68:662–671PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Arra M, Swarnkar G, Ke K, Otero JE, Ying J, Duan X, Maruyama T, Rai MF, O'Keefe RJ, Mbalaviele G et al (2020) LDHA-mediated ROS generation in chondrocytes is a potential therapeutic target for osteoarthritis. Nat Commun 11:3427PubMedPubMedCentralCrossRef Arra M, Swarnkar G, Ke K, Otero JE, Ying J, Duan X, Maruyama T, Rai MF, O'Keefe RJ, Mbalaviele G et al (2020) LDHA-mediated ROS generation in chondrocytes is a potential therapeutic target for osteoarthritis. Nat Commun 11:3427PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Cao B, Li Z, Peng R, Ding J (2015) Effects of cell-cell contact and oxygen tension on chondrogenic differentiation of stem cells. Biomaterials 64:21–32PubMedCrossRef Cao B, Li Z, Peng R, Ding J (2015) Effects of cell-cell contact and oxygen tension on chondrogenic differentiation of stem cells. Biomaterials 64:21–32PubMedCrossRef
65.
Zurück zum Zitat Kaneko Y, Tanigawa N, Sato Y, Kobayashi T, Nakamura S, Ito E, Soma T, Miyamoto K, Kobayashi S, Harato K et al (2019) Oral administration of N-acetyl cysteine prevents osteoarthritis development and progression in a rat model. Sci Rep 9:18741PubMedPubMedCentralCrossRef Kaneko Y, Tanigawa N, Sato Y, Kobayashi T, Nakamura S, Ito E, Soma T, Miyamoto K, Kobayashi S, Harato K et al (2019) Oral administration of N-acetyl cysteine prevents osteoarthritis development and progression in a rat model. Sci Rep 9:18741PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Almonte-Becerril M, Gimeno LI, Villarroya O, Benito-Jardón M, Kouri JB, Costell M (2018) Genetic abrogation of the fibronectin-α5β1 integrin interaction in articular cartilage aggravates osteoarthritis in mice. PLoS One 13:e0198559PubMedPubMedCentralCrossRef Almonte-Becerril M, Gimeno LI, Villarroya O, Benito-Jardón M, Kouri JB, Costell M (2018) Genetic abrogation of the fibronectin-α5β1 integrin interaction in articular cartilage aggravates osteoarthritis in mice. PLoS One 13:e0198559PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Hirose N, Okamoto Y, Yanoshita M, Asakawa Y, Sumi C, Takano M, Nishiyama S, Su SC, Mitsuyoshi T, Kunimatsu R et al (2020) Protective effects of cilengitide on inflammation in chondrocytes under excessive mechanical stress. Cell Biol Int 44:966–974PubMedCrossRef Hirose N, Okamoto Y, Yanoshita M, Asakawa Y, Sumi C, Takano M, Nishiyama S, Su SC, Mitsuyoshi T, Kunimatsu R et al (2020) Protective effects of cilengitide on inflammation in chondrocytes under excessive mechanical stress. Cell Biol Int 44:966–974PubMedCrossRef
68.
Zurück zum Zitat Lee W, Leddy HA, Chen Y, Lee SH, Zelenski NA, McNulty AL, Wu J, Beicker KN, Coles J, Zauscher S et al (2014) Synergy between Piezo1 and Piezo2 channels confers high-strain mechanosensitivity to articular cartilage. Proc Natl Acad Sci U S A 111:E5114–E5122PubMedPubMedCentralCrossRef Lee W, Leddy HA, Chen Y, Lee SH, Zelenski NA, McNulty AL, Wu J, Beicker KN, Coles J, Zauscher S et al (2014) Synergy between Piezo1 and Piezo2 channels confers high-strain mechanosensitivity to articular cartilage. Proc Natl Acad Sci U S A 111:E5114–E5122PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Xu B, Xing R, Huang Z, Yin S, Li X, Zhang L, Ding L, Wang P (2019) Excessive mechanical stress induces chondrocyte apoptosis through TRPV4 in an anterior cruciate ligament-transected rat osteoarthritis model. Life Sci 228:158–166PubMedCrossRef Xu B, Xing R, Huang Z, Yin S, Li X, Zhang L, Ding L, Wang P (2019) Excessive mechanical stress induces chondrocyte apoptosis through TRPV4 in an anterior cruciate ligament-transected rat osteoarthritis model. Life Sci 228:158–166PubMedCrossRef
70.
Zurück zum Zitat Sun L, Wang G, He M, Mei Z, Zhang F, Liu P (2020) Effect and mechanism of the CACNA2D1-CGRP pathway in osteoarthritis-induced ongoing pain. Biomed Pharmacother 129:110374PubMedCrossRef Sun L, Wang G, He M, Mei Z, Zhang F, Liu P (2020) Effect and mechanism of the CACNA2D1-CGRP pathway in osteoarthritis-induced ongoing pain. Biomed Pharmacother 129:110374PubMedCrossRef
71.
Zurück zum Zitat Zelenski NA, Leddy HA, Sanchez-Adams J, Zhang J, Bonaldo P, Liedtke W, Guilak F (2015) Type VI collagen regulates pericellular matrix properties, chondrocyte swelling, and mechanotransduction in mouse articular cartilage. Arthritis Rheumatol 67:1286–1294PubMedPubMedCentralCrossRef Zelenski NA, Leddy HA, Sanchez-Adams J, Zhang J, Bonaldo P, Liedtke W, Guilak F (2015) Type VI collagen regulates pericellular matrix properties, chondrocyte swelling, and mechanotransduction in mouse articular cartilage. Arthritis Rheumatol 67:1286–1294PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Zeng D, Yao P, Zhao H (2019) P2X7, a critical regulator and potential target for bone and joint diseases. J Cell Physiol 234:2095–2103PubMedCrossRef Zeng D, Yao P, Zhao H (2019) P2X7, a critical regulator and potential target for bone and joint diseases. J Cell Physiol 234:2095–2103PubMedCrossRef
73.
Zurück zum Zitat Thompson CL, Plant JC, Wann AK, Bishop CL, Novak P, Mitchison HM, Beales PL, Chapple JP, Knight MM (2017) Chondrocyte expansion is associated with loss of primary cilia and disrupted hedgehog signalling. Eur Cells Mater 34:128–141CrossRef Thompson CL, Plant JC, Wann AK, Bishop CL, Novak P, Mitchison HM, Beales PL, Chapple JP, Knight MM (2017) Chondrocyte expansion is associated with loss of primary cilia and disrupted hedgehog signalling. Eur Cells Mater 34:128–141CrossRef
75.
Zurück zum Zitat Miyauchi A, Kim-Kaneyama JR, Lei XF, Chang SH, Saito T, Haraguchi S, Miyazaki T, Miyazaki A (2019) Alleviation of murine osteoarthritis by deletion of the focal adhesion mechanosensitive adapter, Hic-5. Sci Rep 9:15770PubMedPubMedCentralCrossRef Miyauchi A, Kim-Kaneyama JR, Lei XF, Chang SH, Saito T, Haraguchi S, Miyazaki T, Miyazaki A (2019) Alleviation of murine osteoarthritis by deletion of the focal adhesion mechanosensitive adapter, Hic-5. Sci Rep 9:15770PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Hayashi D, Roemer FW, Guermazi A (2016) Imaging for osteoarthritis. Ann Phys Rehab Med 59:161–169CrossRef Hayashi D, Roemer FW, Guermazi A (2016) Imaging for osteoarthritis. Ann Phys Rehab Med 59:161–169CrossRef
77.
Zurück zum Zitat Krupkova O, Zvick J, Wuertz-Kozak K (2017) The role of transient receptor potential channels in joint diseases. Eur Cells Mater 34:180–201CrossRef Krupkova O, Zvick J, Wuertz-Kozak K (2017) The role of transient receptor potential channels in joint diseases. Eur Cells Mater 34:180–201CrossRef
78.
Zurück zum Zitat Xu L, Nwosu LN, Burston JJ, Millns PJ, Sagar DR, Mapp PI, Meesawatsom P, Li L, Bennett AJ, Walsh DA et al (2016) The anti-NGF antibody muMab 911 both prevents and reverses pain behaviour and subchondral osteoclast numbers in a rat model of osteoarthritis pain. Osteoarthr Cartil 24:1587–1595CrossRef Xu L, Nwosu LN, Burston JJ, Millns PJ, Sagar DR, Mapp PI, Meesawatsom P, Li L, Bennett AJ, Walsh DA et al (2016) The anti-NGF antibody muMab 911 both prevents and reverses pain behaviour and subchondral osteoclast numbers in a rat model of osteoarthritis pain. Osteoarthr Cartil 24:1587–1595CrossRef
79.
Zurück zum Zitat Carcolé M, Kummer S, Gonçalves L, Zamanillo D, Merlos M, Dickenson AH, Fernández-Pastor B, Cabañero D, Maldonado R (2019) Sigma-1 receptor modulates neuroinflammation associated with mechanical hypersensitivity and opioid tolerance in a mouse model of osteoarthritis pain. Br J Pharmacol 176:3939–3955PubMedPubMedCentralCrossRef Carcolé M, Kummer S, Gonçalves L, Zamanillo D, Merlos M, Dickenson AH, Fernández-Pastor B, Cabañero D, Maldonado R (2019) Sigma-1 receptor modulates neuroinflammation associated with mechanical hypersensitivity and opioid tolerance in a mouse model of osteoarthritis pain. Br J Pharmacol 176:3939–3955PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Chevalier X, Ravaud P, Maheu E, Baron G, Rialland A, Vergnaud P, Roux C, Maugars Y, Mulleman D, Lukas C et al (2015) Adalimumab in patients with hand osteoarthritis refractory to analgesics and NSAIDs: a randomised, multicentre, double-blind, placebo-controlled trial. Ann Rheum Dis 74:1697–1705PubMedCrossRef Chevalier X, Ravaud P, Maheu E, Baron G, Rialland A, Vergnaud P, Roux C, Maugars Y, Mulleman D, Lukas C et al (2015) Adalimumab in patients with hand osteoarthritis refractory to analgesics and NSAIDs: a randomised, multicentre, double-blind, placebo-controlled trial. Ann Rheum Dis 74:1697–1705PubMedCrossRef
81.
Zurück zum Zitat Maudens P, Seemayer CA, Pfefferlé F, Jordan O, Allémann E (2018) Nanocrystals of a potent p38 MAPK inhibitor embedded in microparticles: therapeutic effects in inflammatory and mechanistic murine models of osteoarthritis. J Control Release 276:102–112PubMedCrossRef Maudens P, Seemayer CA, Pfefferlé F, Jordan O, Allémann E (2018) Nanocrystals of a potent p38 MAPK inhibitor embedded in microparticles: therapeutic effects in inflammatory and mechanistic murine models of osteoarthritis. J Control Release 276:102–112PubMedCrossRef
82.
Zurück zum Zitat Rai MF, Pan H, Yan H, Sandell LJ, Pham CTN, Wickline SA (2019) Applications of RNA interference in the treatment of arthritis. Transl Res 214:1–16PubMedPubMedCentralCrossRef Rai MF, Pan H, Yan H, Sandell LJ, Pham CTN, Wickline SA (2019) Applications of RNA interference in the treatment of arthritis. Transl Res 214:1–16PubMedPubMedCentralCrossRef
Metadaten
Titel
Molecular mechanisms of mechanical load-induced osteoarthritis
verfasst von
Tianshun Fang
Xianhao Zhou
Mingchao Jin
Jiangbo Nie
XIongfeng Li
Publikationsdatum
18.01.2021
Verlag
Springer Berlin Heidelberg
Erschienen in
International Orthopaedics / Ausgabe 5/2021
Print ISSN: 0341-2695
Elektronische ISSN: 1432-5195
DOI
https://doi.org/10.1007/s00264-021-04938-1

Weitere Artikel der Ausgabe 5/2021

International Orthopaedics 5/2021 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Arthroskopie kann Knieprothese nicht hinauszögern

25.04.2024 Gonarthrose Nachrichten

Ein arthroskopischer Eingriff bei Kniearthrose macht im Hinblick darauf, ob und wann ein Gelenkersatz fällig wird, offenbar keinen Unterschied.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Ärztliche Empathie hilft gegen Rückenschmerzen

23.04.2024 Leitsymptom Rückenschmerzen Nachrichten

Personen mit chronischen Rückenschmerzen, die von einfühlsamen Ärzten und Ärztinnen betreut werden, berichten über weniger Beschwerden und eine bessere Lebensqualität.

Update Orthopädie und Unfallchirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.