Abstract
Joint inflammation is present in the majority of OA patients and pro-inflammatory mediators, such as IL-6, are actively involved in disease progression. Increased levels of IL-6 in serum or synovial fluid from OA patients correlate with disease incidence and severity, with IL-6 playing a pivotal role in the development of cartilage pathology, e.g. via induction of matrix-degrading enzymes. However, IL-6 also increases expression of anti-catabolic factors, suggesting a protective role. Until now, this dual role of IL-6 is incompletely understood and may be caused by differential effects of IL-6 classic vs trans-signalling. Here, we review current evidence regarding the role of IL-6 classic- and trans-signalling in local joint pathology of cartilage, synovium and bone. Furthermore, we discuss targeting of IL-6 in experimental OA models and provide future perspective for OA treatment by evaluating currently available IL-6 targeting strategies.
Keywords: interleukin-6, osteoarthritis, IL-6 trans-signalling, therapy, cartilage, synovitis
Rheumatology key messages
IL-6 signalling is actively involved in OA pathology, identifying IL-6 as a promising therapeutic target.
Differences between classic- vs trans-signalling explain the protective and degenerative IL-6 effects in joint tissues.
Specific targeting of IL-6 trans-signalling could be a superior treatment strategy in OA.
Introduction
OA is a degenerative joint disease with increasing incidence due to a rise in life expectancy and average body weight in western society [1, 2]. Currently, therapies are focused on pain management or eventually joint replacement. OA affects all joint tissues, resulting in loss of articular cartilage, ectopic bone formation, subchondral bone sclerosis and synovial inflammation [3]. Inflammation is increasingly accepted as a driver of OA pathology, implying the synovium and inflammatory cytokines in driving cartilage degeneration [4–6]. For this reason, treatment strategies have focused on targeting pro-inflammatory cytokines TNF-α and IL-1β in hand and knee OA [7–10], which did not result in clinical applications thus far. Therapies targeting IL-6 are approved and effective in treating RA, juvenile idiopathic arthritis, Castleman’s disease, and giant cell arteritis [11]. Also in OA, IL-6 plays a significant role in joint pathology, but has not been a primary target of interest as research mostly focused on IL-1β and TNF-α. Here, we review the current state of evidence regarding the role of IL-6 in OA pathophysiology, and discuss potential therapeutic approaches to target the IL-6 signalling pathway in OA.
Understanding the complexity of the IL-6 signalling pathway
Intracellular signalling cascades
IL-6 signalling starts by binding of IL-6 to the IL-6 receptor α subunit (IL-6R), followed by complex formation with a homodimer of glycoprotein 130 (gp130) [12]. The IL-6R has no signal transduction capacity and its expression is limited, e.g. to monocytes, hepatocytes and certain leucocyte subsets [13]. In contrast, the signal-transducing receptor gp130 is ubiquitously expressed. Gp130 also functions as a β subunit for other IL-6 family cytokines, like oncostatin-M, IL-11, IL-27 and leukemia inhibitory factor [14]. After IL-6 receptor complex formation, the Janus kinases/signal transducers and activators of transcription (JAK/STAT) pathway is activated (Fig. 1), leading to recruitment and activation of STAT1, STAT3, and to a lesser extent STAT5 [15]. Besides canonical signalling via JAK/STAT, IL-6 activates non-canonical signalling via mitogen-activated protein kinase (MAPK) cascade (Ras-Raf-MEK-ERK pathway) and PI3K- protein kinase B (PkB)/Akt. IL-6-induced JAK/STAT is tightly controlled by negative feedback regulators, such as suppressor of cytokine signalling (SOCS) protein family and protein inhibitors of activated STATs (PIAS) [16, 17]. SOCS proteins are directly induced by gp130 cytokines, resulting in a negative feedback loop. SOCS3 has been identified as a specific inhibitor of IL-6 signalling and directly inhibits JAK-kinase activity [18, 19]. PIAS negative inhibitors are constitutively expressed and inhibit DNA-binding activity by binding to activated STAT-dimers.
Fig. 1.
Overview of IL-6 signalling pathways
After IL-6 binding to the IL-6R, complex formation with gp130 initiates phosphorylation of JAKs resulting in activation of STAT3-, PI3K- and Ras-Raf-MEK-ERK signaling. Activated transcription factors (e.g. STAT3, NF- κβ and NF-IL-6) translocate to the nucleus to regulate target gene expression. SOCS and PIAS proteins negatively regulate IL-6-induced JAK-STAT signal by blocking JAK-mediated activation of STAT3 (SOCS3), or by blocking DNA-binding activity of STAT3 (PIAS). gp130: glycoprotein 130; IL-6: interleukin-6; JAK: janus kinase; MAPK: mitogen-activated protein kinase; NF- κβ: nuclear factor kappa-light-chain-enhancer of activated B cells; NF-IL6: a nuclear factor for IL-6 expression; PIAS: protein inhibitors of activated STATs; PI3K: phosphoinositide 3-kinase; SOCS3: suppressor of cytokine signaling 3; STAT3: signal transducer and activator of transcription 3.
Cytokine interplay and intracellular cross-talk
Interplay between IL-6 signalling pathways and other cytokines exists on multiple levels [14]. For example, other cytokines from the IL-6 family, like ciliary neurotrophic factor (CNTF) and IL-30, can also bind and activate the IL-6R, although with lower binding affinity compared with the CNTF- and IL-30 receptors [20, 21]. Furthermore, interplay between IL-6 and pro-inflammatory cytokine signalling may restrict uncontrolled pro-inflammatory signalling [22]. For instance, IL-1β strongly inhibits IL-6-mediated acute phase reaction in the liver by directly inhibiting p38 MAPK-dependent STAT3 phosphorylation [22, 23]. More specifically, MAPK p38 and the transcription factor NF-κβ were identified as crucial regulators of the IL-6 signalling pathway [22]. Also, interplay between IL-6 and anti-inflammatory cytokines, such as TGF-β, is present at receptor level and at the level of intracellular mediators [24–26]. Crosstalk between STAT3 and Smad3, the main intracellular mediator of TGF-β signalling, exists in diverse pathophysiological conditions and leads to either synergistic or antagonistic actions depending on cell type and context [26].
Modes of IL-6 signalling
IL-6 has the unique ability to initiate signal transduction via different modes of receptor activation. Signalling via membrane-anchored IL-6R (mIL-6R) is termed classic signalling and is important for the acute-phase response, hematopoiesis and central homeostatic processes [27] (Fig. 2a). Interestingly, a soluble form of IL-6R (sIL-6R) can be produced by shedding of membrane-bound receptor or alternative splicing [28, 29]. sIL-6R can bind secreted IL-6, forming a complex that increases the half-life of IL-6 [30]. Signalling via sIL6R is called trans-signalling and greatly broadens the scope of IL-6 responsiveness, as any gp130-expressing cell can bind and respond to the IL-6/sIL-6R complex (Fig. 2b). IL-6 trans-signalling mainly regulates pro-inflammatory events and is implicated in numerous chronic diseases and cancers [27, 31]. Trans-signalling leads to stronger activation of IL-6 intracellular signalling routes, resulting in enhanced target gene expression, but how this works is still unclear [32, 33]. Possibly, restricted expression of mIL-6R limits activation of STAT3 via classic signalling, but not trans-signalling due to additional presence of sIL-6R [32]. Within our circulation, a soluble form of gp130 (sgp130) acts as a natural inhibitor of IL-6 trans-signalling by binding to the IL-6/sIL-6R with high affinity [31, 34–36]. Sgp130 therefore specifically inhibits IL-6 trans-signalling and does not affect classic signalling or recently discovered IL-6 cluster-signalling. IL-6 cluster-signalling involves membrane IL-6/IL-6R complexes on dendritic cells, which activate gp130 receptors on receiving T cells resulting in the generation of pathogenic Th17 cells (Fig. 2c). [37]. Whether IL-6 cluster-signalling is also relevant in other biological settings remains to be investigated.
Fig. 2.
Modes of IL-6 signalling
(a) Classic IL-6 signalling involves cells expressing both membrane (m)IL-6R and gp130; free IL-6 binds to mIL-6R, forming a complex with gp130. (b) IL-6 trans-signalling is activated by pre-formed complexes of IL-6 and soluble IL-6R (IL-6/sIL-6R) and requires only gp130 expression on target cells. Soluble gp130 (sgp130) acts as a natural inhibitor of trans-signaling by specifically binding to IL-6/sIL-6R complexes. (c) IL-6 cluster signaling utilizes gp130 on receiving cells, activated by IL-6/mIL-6R complexes on presenting cells (e.g. dendritic cells). gp130: glycoprotein 130; IL-6: interleukin-6; IL-6R: IL-6 receptor.
Local and systemic perspective: levels of IL-6 and its soluble receptors in OA
IL-6 levels relate to OA incidence and pathology
IL-6 was detected in OA synovial fluid (SF) as early as 1988. However, this did not result in follow-up studies, as IL-6 levels were lower compared with RA SF and healthy controls were not included [38, 39]. Later, it became clear that IL-6 levels were significantly increased in OA SF and serum compared with healthy individuals [40–42]. Furthermore, additional studies showed a clear relation between systemic IL-6 levels and OA incidence [43–45]. Increased circulating levels of IL-6 were predictive for knee OA and cartilage loss in 3 and 15 years in two independent follow-up studies [43, 44]. Moreover, a high innate capacity to produce IL-6, in response to lipopolysaccharide stimulation, was associated with hand OA development in 90-year-old individuals [45]. Higher IL-6 levels in OA serum or SF also correlate with disease progression or severity of cartilage pathology [42, 46–48]. This suggests that IL-6 levels may reflect cartilage damage, which is supported by the fact that SF IL-6 levels are strongly increased in individuals with cartilage defects but no macroscopic signs of OA [49, 50]. When local vs systemic levels of IL-6 were compared in the same patients, IL-6 concentrations were higher in OA SF (119.8 ± 193.5 pg/ml) compared with plasma samples (3.1 ± 2.7 pg/ml) [51]. Furthermore, two patient subgroups can be identified based on IL-6 levels in OA SF, high producers (2022 ± 526 pg/ml) vs average producers (132 ± 19 pg/ml), of which high producers may particularly benefit from IL-6 targeted therapy [52].
Local production of IL-6 by joint tissues
It is now recognized that synovial inflammation is important in OA, and synovitis is observed in ∼50% of OA patients [5, 53]. The synovium is an important producer of IL-6 in OA, for instance via (activated) synovial fibroblasts or plasma cells in the synovial lining [52, 54–56]. Besides the synovium, the infrapatellar fat pad (IFP) is an important source of IL-6. The IFP is the main fat tissue within the knee, and actively contributes to OA pathophysiology via production of pro-inflammatory mediators and adipokines [57]. Interestingly, the IFP from knee OA patients secreted significantly higher levels of IL-6, but not TNF-α and IL-1β, compared with abdominal fat tissue from the same patients [58]. Furthermore, IFP-conditioned medium cultured with traumatized cartilage explants caused IL-6-dependent glycosaminoglycan release [59]. Also, synovial fibroblasts from obese OA patients secreted higher levels of IL-6 compared with normal-weight patients [60], indicating that IL-6 may be particularly relevant in obesity-derived OA, especially as IL-6 plays a central role in cell metabolism [61].
Broadening our horizon: levels of soluble IL-6 receptors in OA
The ratio of IL-6 classic- vs trans-signalling is regulated by sIL-6 receptors [62]. Increased levels of sIL-6R in OA patients may direct future treatment towards specific inhibition of IL-6 trans-signalling, while decreased sgp130 levels could indicate reduced negative feedback capacity. Unfortunately, studies investigating soluble IL-6 receptors in OA are scarce. Systemically, no differences were detected in sIL-6R levels in serum of healthy donors and OA patients [63], and as far as we know there is no study investigating systemic changes in sgp130 in OA. In OA SF, both sIL-6R and sgp130 are present, but a comparison to healthy individuals is lacking [64–66]. Despite high levels of IL-6 production, it remains unclear if the synovium is a source of sIL-6 receptors in OA. In RA-derived material, cultured SF mononuclear cells produced sIL-6R, but not cultured chondrocytes or synovial cells [64]. However, this was not confirmed in OA. A potential source of sIL-6R in OA could be the IFP, which was shown to produce both IL-6 and sIL-6R [58], possibly resulting in IL-6 trans-signalling.
Direct effects of IL-6 on local joint tissues
Cartilage
As cartilage is the main OA affected tissue, previous studies mostly focused on identifying IL-6 effects in cartilage. However, IL-6 has both catabolic and protective effects in cartilage, which is still not completely understood. Early studies focused on IL-6 classic signalling and generally found protective effects of IL-6, such as production of tissue inhibitor of metalloproteinases (TIMPs) [67, 68]. Furthermore, IL-6 classic signalling slightly stimulated proteoglycan synthesis in human OA chondrocytes [49], and did not affect proteoglycan synthesis of human or bovine chondrocytes [69, 70]. However, detrimental effects of IL-6 classic signalling in cartilage have also been reported. IL-6 inhibited proteoglycan synthesis in human cartilage explants and rabbit chondrocytes [71–73]. Moreover, IL-6 suppressed collagen type II neo-synthesis and enhanced IL-1β-mediated proteoglycan degeneration in rabbit chondrocytes [73, 74]. Several studies show that IL-6 induces metalloproteinase (MMP)-3, MMP-13 and A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) enzyme expression, which mediate cartilage degeneration [75–77]. Besides regulating matrix synthesis and degeneration, IL-6 induces matrix mineralization via formation of basic calcium phosphate crystals leading to proteoglycan loss [78]. Furthermore, IL-6 disturbs several other chondrocyte functions, resulting in decreased proliferation or increased oxidative stress generation [73, 79]. Of note, IL-6 directly induces SOCS3 expression, which can lead to insulin-like growth factor 1 desensitization in cartilage [80]. However, enhanced SOCS3 may also be protective as it restricts pro-inflammatory signalling in chondrocytes [81]. Altogether, this indicates that the definition of IL-6 classic signalling as only ‘protective’ is probably too simplified.
Generally, chondrocytes are considered to have low levels of mIL-6R, which may limit STAT3 activation, but strong evidence for this is missing [32, 82]. In murine epiphysial chondrocytes, no mIL-6R expression was observed with flow cytometry [82]. In contrast, expression of mIL-6R was observed in four donors of OA human chondrocytes on both mRNA and protein level [25]. Chondrocyte sensitivity for classic IL-6 signalling is determined by mIL-6R levels, which can be altered by hormones, cytokines and epigenetic factors [13]. The cytokine IL-1β increases mIL-6R expression in chondrocytes and hepatocytes [25, 83], which may explain synergistic effects of IL-1β and IL-6 in mediating cartilage degradation or collagen breakdown [71, 73, 84]. Cartilage injury induced by blunt trauma also potentiated IL-6-mediated expression of catabolic markers in bovine cartilage, but the underlying mechanism was not investigated [59, 85]. On the other hand, TGF-β decreases mIL-6R expression in chondrocytes, resulting in inhibition of classic IL-6 signalling [25]. Also, mechanical compression of cartilage, which leads to active TGF-β signalling [86], inhibited catabolic effects of IL-6 and TNF-α combined [87]. Changes in mIL-6R expression in chondrocytes alter their sensitivity towards IL-6 classic signalling and may partly explain previous discrepancies regarding IL-6 effects in cartilage.
With respect to IL-6 trans-signalling in cartilage, functional studies also show contradictive results. Most studies conclude that IL-6 trans-signalling is detrimental for cartilage, as it inhibits proteoglycan synthesis and stimulates proteoglycan loss [72, 88]. Moreover, sIL-6R was required for full activation of JAK1/2 in bovine chondrocytes, resulting in decreased expression of matrix components and increased levels of cartilage degrading enzymes [89, 90]. On the other hand, soluble IL-6R also augmented production of anti-catabolic TIMPs in chondrocytes [67], which suggests that sIL-6R stimulates general IL-6 signalling and not only the catabolic response. In conclusion, there is substantial evidence for a direct role of IL-6 in regulating chondrocyte function and cartilage metabolism. While most studies report catabolic effects of IL-6 on cartilage, protective effects are also found. These discrepancies may be explained by functional differences in IL-6 classic vs trans-signalling, or disturbed expression of mIL-6R levels on chondrocytes.
Other joint tissues
As OA is a whole joint disease, direct effects of IL-6 on other joint tissues such as synovium, subchondral bone, and muscle tissue are also of interest [3]. Comparable to IL-6 effects in cartilage, mainly IL-6 trans-signalling was associated with detrimental effects in synovium. In OA- or RA-digested synovium, both IL-6 classic and trans-signalling increase production of TIMP, but only trans-signalling induces expression of ADAMTS4 [67, 68, 91, 92]. Moreover, IL-6 trans- but not classic signalling caused strong proliferation of RA synovial fibroblasts that could indicate a role in synovial hyperplasia or fibrosis [93]. However, in bone this distinction is less clear. On the one hand, IL-6 trans-signalling promotes osteoclast formation and consequently bone resorption, while classic signalling inhibits osteoclastogenesis [39, 94, 95]. On the other hand, IL-6 trans-signalling has been shown to promote bone formation [96]. The dual effect of IL-6 trans-signalling on bone resorption and formation may be explained by variation in levels of the pro-osteoclastogenic factor receptor activator of nuclear factor kappa-B ligand (RANKL). While high-levels of RANKL promote osteoclastogenesis, lower RANKL levels result in inhibition of osteoclast formation [97]. Finally, IL-6 may be directly involved in the process of OA-related muscle degeneration [98, 99]. Indeed, elevated levels of IL-6 are associated with reduced muscle endurance in elderly women with knee OA [100]. Moreover, increased levels of IL-6, STAT3 and SOCS3 have been detected in muscle tissue of knee OA patients [101]. However, IL-6 also mediates important anabolic processes in muscle tissue, such as muscle growth and myogenic differentiation [102]. This dual role of IL-6 in muscle tissue might be a result of functional differences in IL-6 classic- vs trans-signalling, but this is yet unknown. Thus, IL-6 classic and trans-signalling can affect joint tissues besides articular cartilage, but their respective functional effects and role in OA development remain elusive.
Evidence for a role of IL-6 in OA pathology: lessons learned from animal studies
The role of IL-6 in OA pathophysiology has been studied in several experimental OA models and mostly show a destructive role for IL-6 (Table 1). Induction of the destabilization of the medial meniscus (DMM) model in IL-6-/- mice resulted in marked reduction of cartilage destruction compared to wildtype mice [75]. Moreover, expression levels of MMP-3 and MMP-13 were significantly decreased in IL-6-/- mice compared to wildtype, indicating that IL-6 induces catabolic mediators in OA. This catabolic role of IL-6 was supported by overexpression of HIF-2α in wildtype vs IL-6-/- mice, causing OA-like cartilage destruction in wildtype but not IL-6-/- mice. Local injection of IL-6 into the knee joint caused significantly increased cartilage degeneration and MMP-3 and -13 expression, revealing direct evidence for deleterious effects of IL-6 in OA [75]. Strikingly, despite marked evidence of IL-6 involvement in OA, only one study blocked IL-6 itself. Both systemic administration of a neutralizing antibody as well as anti-IL-6 siRNA resulted in decreased cartilage lesions and subchondral bone sclerosis in the anterior cruciate ligament transaction (ACLT) OA model [103]. Moreover, systemic treatment with an anti-IL-6-receptor neutralizing antibody (MR16) in the DMM model ameliorated the extent of cartilage pathology, synovial inflammation and osteophyte development [105]. This antibody is similar to tocilizumab, which directly targets the human IL-6R and is clinically effective in several inflammatory diseases [11]. Moreover, blocking of the IL-6R using tocilizumab resulted in cartilage preservation in a mouse model of ischemic osteonecrosis and significantly increased bone volume [106].
Table 1.
Effect of targeting IL-6 signalling in experimental OA models
| IL-6 targeting strategy | Animal (sex, strain, age) | OA model | Observations | Effect | References |
|---|---|---|---|---|---|
| General knock-out (IL-6-/-) | Male C57BL/6 mice (10–12 weeks) | Surgically-induced post-traumatic OA (DMM) | ↓ cartilage pathology | + | [75] |
| Systemic treatment with anti-IL-6 antibody | Male C57BL/6 mice (8 weeks) | Surgically-induced post-traumatic OA (ACLT) | ↓ cartilage pathology | + | [98] |
| Systemic treatment with STAT3 inhibitor(AG490) | Male C57BL/6 mice (8 weeks) | Surgically-induced post-traumatic OA (ACLT) |
|
+ | [98] |
| Systemic treatment with anti-IL-6R antibody(MR16-1) | Male C57BL/6 mice (10 weeks) | Surgically-induced post-traumatic OA (DMM) |
|
+ | [99] |
| Systemic treatment with STAT3 inhibitor(Stattic) | Male C57BL/6 mice (10 weeks) | Surgically-induced post-traumatic OA (DMM) |
|
+ | [99] |
| General knock-out (IL-6-/-) | Male C57BL/6 mice (18–23 months) | Age-related OA (Spontaneous OA) |
|
[103] | |
| General knock-out (IL-6-/-) | Female C57BL/6 mice (18–23 months) | Age-related OA (Spontaneous OA) | No difference | No effect | [103] |
| General knock-out (IL-6-/-) | Male and female C57BL/6 mice (3–4 months) | Chemically-induced joint instability OA (CiOA) | No difference | No effect | [103] |
| Intra-articular treatment with gp130 modulator (RCGD 423) | Male Sprague-Dawley rats (3–4 months) | Surgically-induced post-traumatic OA (Partial menisectomy) |
|
+ | [104] |
ACTL: anterior cruciate ligament transection; CiOA: collagenase-induced OA; DMM: destabilization of the medial meniscus.
IL-6 is possibly mainly involved in trauma-related OA, as both the DMM and ACLT models reflect trauma-induced OA development. Also, in humans, local IL-6 levels strongly increase upon cartilage trauma and associate with knee OA progression after previous meniscectomy [49, 50]. Systemic IL-6 levels also increase during natural ageing and are associated with several age-related diseases [107, 108]. However, there was no difference in cartilage degradation in wildtype or IL-6-/- mice after age-related OA development [109]. Male IL-6-/- mice even developed more cartilage damage, ectopic bone formation and subchondral bone sclerosis compared to male wildtype mice, while there was no difference in pathology of females. This suggests that IL-6 has no pathological role in age-related murine OA, and even ameliorates OA pathology in male mice. Multiple studies show interplay between IL-6 and sex hormones such as testosterone and oestrogen [110–112], but it is unclear how sex hormones affect IL-6 function within the joint. Of note, no markable inflammation was detected in these mice including inflammatory infiltrate, exudate or synovitis [109]. This raises the question whether murine ageing fully reflects human age-related OA development, in which synovitis is commonly observed and levels of inflammatory mediators, amongst which IL-6, are systemically increased [5, 53, 107]. Besides age-related OA, there was no difference in OA pathology caused by collagenase-induced OA (CiOA) between wildtype or IL-6-/- mice [109]. Although using conditional IL-6-/- mice, instead of constitutive knockouts, would more closely resemble physiological conditions, this suggests that other mediators may cause OA pathology in this model. However, to really exclude a role for IL-6 in CiOA and age-related OA, lack of IL-6 effects should be confirmed by independent studies. Functional differences in IL-6 classic vs trans-signalling may explain the contradictive results obtained in the different OA models. Unfortunately, all of the employed blocking strategies block both the classic- and trans-signalling pathway and current studies do not report sIL-6R levels. Specific inhibition of IL-6 trans-signalling in OA models might be extremely helpful to dissect detrimental vs protective effects of IL-6 in the future.
Blocking downstream of IL-6: targeting STAT3 in experimental OA
STAT3 is the most specific downstream mediator of the IL-6 signalling pathway, but is not solely activated by IL-6. Therefore, STAT3 activation in OA results from synergistic actions of several gp130 cytokines [14]. Hypothetically, targeting of STAT3 may be more successful compared with blocking IL-6, as other STAT3-activating cytokines also have catabolic and inflammatory effects on cartilage [113]. Indeed, repeated administration of a small molecule inhibitor against STAT3 (Stattic) in the DMM model resulted in stronger protection against cartilage degeneration and osteophyte formation compared with blocking IL-6R [105]. This additional effect may result from blockade of both IL-6 and oncostatin M (OSM) signalling via STAT3, based on the role of OSM in osteophyte proliferation and synovial inflammation [104, 114]. Inhibition of JAK2/STAT3 signalling in the ACLT model using the AG490 inhibitor also led to considerable protection against cartilage degeneration and subchondral bone sclerosis [103]. However, mesoderm-specific deletion of STAT3 caused expansion of growth plate hypertrophic chondrocytes and severe dysregulation of endochondral ossification, caused by STAT3-mediated activation of Sox9 in chondrocytes [115]. This phenotype is not observed in IL-6-/- animals [116], suggesting that other STAT3-activating cytokines may cause dysregulation of cartilage and bone development, such as LIF which is associated with reduced skeletal growth [117]. Recently, a novel gp130-small molecule modulator (RCGD 423) was discovered, which directed gp130 towards proliferative STAT3/c-Myc signalling, while inhibiting ERK/NF-κβ signalling. Therapeutic administration of the RCGD 423 compound, leading to STAT3 activation, resulted in reduced cartilage degeneration in a rat partial meniscectomy model [118]. This contradicts the earlier finding that STAT3 inhibition using Stattic protects against cartilage degeneration [105]. It is possible that the proliferative effect of the RCGD423 inhibitor is caused by the strong activation of c-Myc, as LIF-driven c-Myc activation is critical for chondrocyte survival and proliferation in fetal cartilage [118]. These opposing results indicate that the ultimate result of STAT3 inhibition, beneficial or detrimental, is strongly context-dependent and determined by the integrated signal of multiple gp130 cytokines. While targeting of STAT3 may seem promising in experimental OA, this might prove difficult in OA patients due to large differences in severity and incidence of inflammation, and heterogeneity in STAT3-activating cytokines or growth factors [53]. This makes it difficult to predict the outcome of STAT3 inhibition in OA and argues for the simpler approach of directly targeting gp130 cytokines, such as IL-6, upstream of STAT3.
Under construction: IL-6 targeted therapy in OA
Currently, multiple therapeutic strategies exist to effectively target the IL-6 signalling pathway and are safely applied for the treatment of several inflammatory diseases. For example, the IL-6R targeting antibody tocilizumab has been approved for treatment of RA, juvenile idiopathic arthritis, Castleman’s disease and recently also for giant cell arteritis [62, 119]. Currently, no therapies targeting IL-6 signalling are approved for treatment of OA. This may change in the near future, as tocilizumab is being tested in a phase 3 randomized controlled trial in patients with refractory hand OA (ClinicalTrials.gov NCT02477059).
Previous studies showed that levels of IL-6 in SF can vary between different joints, which could direct future IL-6 targeted therapy towards relevant patient subgroups. For example, levels of IL-6 were strikingly higher in knee OA SF compared with carpometacarpal joint fluid [120]. Moreover, more IL-6 was detected in post-traumatic wrist OA compared with knee OA patients [121]. As inflammation is strongly linked to structural damage in hand OA patients [122–124], this patient group may be very suitable to study the consequences of blocking pro-inflammatory cytokines, such as TNF-α [125] and IL-1β [7] and now IL-6. Besides stratification of patients based on joint location, treatment choice could also be based on OA subtypes. Post-traumatic OA is a common form of OA, developing after joint injury (e.g. anterior cruciate ligament or meniscus injury) [126]. In these patients, there may be a therapeutic window after injury, in which targeting of inflammatory mediators may prevent the development of further damage. During joint injury, such as anterior cruciate ligament rupture, levels of IL-6 in SF are highly increased up to 1000-fold [127, 128], and a sudden increase in IL-6 levels has also been found after focal cartilage damage [49, 50]. This suggests that inhibition of IL-6 shortly after joint injury may be a promising treatment strategy to prevent the development of post-traumatic OA; however, the optimal therapeutic window to prevent further damage is still unknown.
Due to the success of tocilizumab, novel IL-6 pathway inhibitors have been developed, such as biologics targeting IL-6R (vobarilizumab, satralizumab, sarilumab), IL-6 (siltuximab, olokizumab, sirukumab, clazakizumab and MEDI 5117), IL-6 trans-signalling (olamkicept), or small molecule inhibitors directed against JAKs or STAT3 [129]. Multiple inhibitors have been developed and clinically tested that target JAK-kinases or STAT3 directly [11]. Although some of these compounds are clinically effective in RA, such as tofacitinib and baricitinib (pan-JAK inhibitors), they have not been tested in OA patients [130]. Yet, there are pre-clinical indications that JAK/STAT inhibition could be effective in OA. For instance, tofacitinib inhibited cytokine-induced proteoglycan loss and restored collagen type II synthesis in bovine cartilage explants [131]. In addition, animal studies indicate protective effects of JAK/STAT inhibition in experimental OA [103, 105]. However, targeting of JAK/STAT signalling also results in inhibition of multiple cytokines including IL-10, IL-4 and IGF-1, which have a beneficial role in joint biology and OA development [14, 132, 133]. As OA is a very heterogeneous disease with large differences in severity of inflammation and cytokine profile [53], it will be difficult to predict outcome of JAK/STAT inhibition in OA patients. Therefore, the simpler approach of targeting one cytokine, like IL-6, might be a safer strategy. As several therapeutics have been developed that target IL-6 signalling via a different mechanism, the comparison of these treatments will greatly enhance knowledge about the role of IL-6 in disease. Tocilizumab, for instance, blocks all IL-6 signalling pathways (classic and trans-signalling, and potentially also cluster-signalling [37]), because it inhibits IL-6 binding to both mIL-6R and sIL-6R (Fig. 3) [134]. In contrast, olamkicept specifically targets the IL-6/sIL-6R complex, thereby only inhibiting IL-6 trans-signalling and not classic signalling. Olamkicept is a fusion protein consisting of two soluble human gp130 proteins fused with the Fc region of human IgG (sgp130Fc) [129]. Accordingly, olamkicept blocks pro-inflammatory events of IL-6 trans-signalling, while simultaneously allowing homeostatic effects of IL-6 classic signalling. Olamkicept was already successfully used in treating experimental Crohn’s disease and is now in clinical trials for inflammatory bowel disease and active ulcerative colitis [11, 135]. Specific inhibition of IL-6 trans-signalling could be a preferred future treatment strategy, as several side effects of tocilizumab have already been reported that may result from inhibition of IL-6 classic signalling, such as impairment of the acute phase response [129]. Olamkicept might also be a promising therapy for OA, as several studies demonstrated catabolic and pro-inflammatory effects of IL-6 trans-signalling in cartilage and synovial tissue [72, 88, 92, 93]. Thus, even if tocilizumab does not prove effective in hand OA, specific inhibition of IL-6 trans-signalling may hold additional promise. Nonetheless, additional pre-clinical research will first be needed as the relevance of IL-6 trans-signalling in experimental OA has not yet been demonstrated.
Fig. 3.
Current IL-6 targeting strategies
Anti-IL-6 monoclonal antibodies (e.g. siltuximab) directly target IL-6, thus blocking both classic and IL-6 trans-signaling. IL-6R targeting antibodies (e.g. tocilizumab) block binding of IL-6 to the IL-6R (both mIL-6R as well as sIL-6R), thereby inhibiting IL-6 classic and trans-signaling pathways. The sgp130Fc fusion protein (e.g. olamkicept) was developed to specifically target IL-6 trans-signaling, and only binds to the IL-6/sIL-6R complex. Sgp130Fc does not bind to membrane IL-6R or free IL-6, therefore allowing classic IL-6 signaling to continue. IL-6: interleukin 6; mIL-6R: membrane IL-6 receptor; sgp130: soluble glycoprotein 130; sIL-6R: soluble IL-6 receptor.
Conclusions and future perspectives
In this review, we focus on the unique ability of IL-6 to signal via the classic- and trans-signalling pathway, and discuss the opposing effects of these signalling routes in OA pathophysiology and treatment. Levels of IL-6 are increased in SF and serum of OA patients, and relate to disease incidence and pathology. In contrast, regulation of sIL-6R in OA, which controls activation of IL-6 trans-signalling, has been overlooked until now and warrants further research. In local joint tissues such as cartilage, synovium and bone, IL-6 classic signalling results mainly in protective effects, while trans-signalling leads to pro-inflammatory and catabolic effects. However, it is highly likely that local regulation of IL-6R levels also determines IL-6 outcome to a great extent. Current evidence of IL-6 blockade in experimental OA shows that therapeutic targeting of the IL-6 pathway could be a promising treatment strategy to reduce cartilage damage, synovial inflammation and subchondral bone pathology in OA patients. Moreover, we propose that specific blockade of IL-6 trans-signalling could be a superior treatment strategy, which may result in inhibition of deleterious IL-6 effects in OA, while maintaining protective IL-6 signalling via the classic pathway.
Search strategy
Articles were selected using the PubMed search engine. To select articles regarding IL-6 in OA, we used following search terms present in title or abstract [tiab]: ‘interleukin-6’ in combination with search terms covering the topics in this review including ‘osteoarthritis’, ‘cartilage’, ‘synovium’, ‘bone’, ‘muscle’, ‘infrapatellar fat pad’ and ‘therapy’. Relevant synonyms were included using MeSH terms. Title and abstract of articles were screened for relevant topics as listed in this review. Non-English articles, and articles in which IL-6 was only used as a marker of inflammation were excluded. In addition, reference lists of cited articles and articles in our personal databases were screened for eligibility. Search includes articles published up to January 2020.
Funding: This study was supported by ReumaNederland (Dutch Arthritis Society, with grant number: 15–2-404 and 19–1-204).
Disclosure statement: The authors have declared no conflicts of interest.
References
- 1. Arden N, Nevitt MC.. Osteoarthritis: epidemiology. Best Pract Res Clin Rheumatol 2006;20:3–25. [DOI] [PubMed] [Google Scholar]
- 2. Johnson VL, Hunter DJ.. The epidemiology of osteoarthritis. Best Pract Res Clin Rheumatol 2014;28:5–15. [DOI] [PubMed] [Google Scholar]
- 3. Loeser RF, Goldring SR, Scanzello CR, Goldring MB.. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum 2012;64:1697–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. 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. [DOI] [PubMed] [Google Scholar]
- 5. Ayral X, Pickering EH, Woodworth TG, Mackillop N, Dougados M.. Synovitis: a potential predictive factor of structural progression of medial tibiofemoral knee osteoarthritis – results of a 1 year longitudinal arthroscopic study in 422 patients. Osteoarthritis Cartilage 2005;13:361–7. [DOI] [PubMed] [Google Scholar]
- 6. Berenbaum F. Osteoarthritis as an inflammatory disease (osteoarthritis is not osteoarthrosis!). Osteoarthritis Cartilage 2013;21:16–21. [DOI] [PubMed] [Google Scholar]
- 7. Kloppenburg M, Peterfy C, Haugen IK. et al. Phase IIa, placebo-controlled, randomised study of lutikizumab, an anti-interleukin-1alpha and anti-interleukin-1beta dual variable domain immunoglobulin, in patients with erosive hand osteoarthritis. Ann Rheum Dis 2019;78:413–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Fleischmann RM, Bliddal H, Blanco FJ. et al. A phase II trial of lutikizumab, an anti-interleukin-1alpha/beta dual variable domain immunoglobulin, in knee osteoarthritis patients with synovitis. Arthritis Rheumatol 2019;71:1056–69. [DOI] [PubMed] [Google Scholar]
- 9. Magnano MD, Chakravarty EF, Broudy C. et al. A pilot study of tumor necrosis factor inhibition in erosive/inflammatory osteoarthritis of the hands. J Rheumatol 2007;34:1323–7. [PubMed] [Google Scholar]
- 10. Maksymowych WP, Russell AS, Chiu P. et al. Targeting tumour necrosis factor alleviates signs and symptoms of inflammatory osteoarthritis of the knee. Arthritis Res Ther 2012;14:R206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Kang S, Tanaka T, Narazaki M, Kishimoto T.. Targeting interleukin-6 signaling in clinic. Immunity 2019;50:1007–23. [DOI] [PubMed] [Google Scholar]
- 12. Kishimoto T, Akira S, Taga T.. Interleukin-6 and its receptor: a paradigm for cytokines. Science 1992;258:593–7. [DOI] [PubMed] [Google Scholar]
- 13. Wolf J, Rose-John S, Garbers C.. Interleukin-6 and its receptors: a highly regulated and dynamic system. Cytokine 2014;70:11–20. [DOI] [PubMed] [Google Scholar]
- 14. Garbers C, Hermanns HM, Schaper F. et al. Plasticity and cross-talk of interleukin 6-type cytokines. Cytokine Growth Factor Rev 2012;23:85–97. [DOI] [PubMed] [Google Scholar]
- 15. Luo Y, Zheng SG.. Hall of fame among pro-inflammatory cytokines: interleukin-6 gene and its transcriptional regulation mechanisms. Front Immunol 2016;7:604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Naka T, Narazaki M, Hirata M. et al. Structure and function of a new STAT-induced STAT inhibitor. Nature 1997;387:924–9. [DOI] [PubMed] [Google Scholar]
- 17. Liu B, Liao J, Rao X. et al. Inhibition of Stat1-mediated gene activation by PIAS1. Proc Natl Acad Sci USA 1998;95:10626–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Babon JJ, Varghese LN, Nicola NA.. Inhibition of IL-6 family cytokines by SOCS3. Semin Immunol 2014;26:13–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Croker BA, Krebs DL, Zhang JG. et al. SOCS3 negatively regulates IL-6 signaling in vivo. Nat Immunol 2003;4:540–5. [DOI] [PubMed] [Google Scholar]
- 20. Schuster B, Kovaleva M, Sun Y. et al. Signaling of human ciliary neurotrophic factor (CNTF) revisited. The interleukin-6 receptor can serve as an alpha-receptor for CTNF. J Biol Chem 2003;278:9528–35. [DOI] [PubMed] [Google Scholar]
- 21. Crabe S, Guay-Giroux A, Tormo AJ. et al. The IL-27 p28 subunit binds cytokine-like factor 1 to form a cytokine regulating NK and T cell activities requiring IL-6R for signaling. J Immunol 2009;183:7692–702. [DOI] [PubMed] [Google Scholar]
- 22. Radtke S, Wuller S, Yang XP. et al. Cross-regulation of cytokine signalling: pro-inflammatory cytokines restrict IL-6 signalling through receptor internalisation and degradation. J Cell Sci 2010;123:947–59. [DOI] [PubMed] [Google Scholar]
- 23. Ahmed ST, Mayer A, Ji JD, Ivashkiv LB.. Inhibition of IL-6 signaling by a p38-dependent pathway occurs in the absence of new protein synthesis. J Leukoc Biol 2002;72:154–62. [PubMed] [Google Scholar]
- 24. Zhang XL, Topley N, Ito T, Phillips A.. Interleukin-6 regulation of transforming growth factor (TGF)-beta receptor compartmentalization and turnover enhances TGF-beta1 signaling. J Biol Chem 2005;280:12239–45. [DOI] [PubMed] [Google Scholar]
- 25. Wiegertjes R, van Caam A, van Beuningen H. et al. TGF-beta dampens IL-6 signaling in articular chondrocytes by decreasing IL-6 receptor expression. Osteoarthritis Cartilage 2019;27:1197–207. [DOI] [PubMed] [Google Scholar]
- 26. Itoh Y, Saitoh M, Miyazawa K.. Smad3-STAT3 crosstalk in pathophysiological contexts. Acta Biochim Biophys Sin 2018;50:82–90. [DOI] [PubMed] [Google Scholar]
- 27. Rose-John S. The soluble interleukin 6 receptor: advanced therapeutic options in inflammation. Clin Pharmacol Ther 2017;102:591–8. [DOI] [PubMed] [Google Scholar]
- 28. Lust JA, Donovan KA, Kline MP. et al. Isolation of an mRNA encoding a soluble form of the human interleukin-6 receptor. Cytokine 1992;4:96–100. [DOI] [PubMed] [Google Scholar]
- 29. Mülberg J, Schooltink H, Stoyan T. et al. The soluble interleukin-6 receptor is generated by shedding. Eur J Immunol 1993;23:473–80. [DOI] [PubMed] [Google Scholar]
- 30. Peters M, Jacobs S, Ehlers M. et al. The function of the soluble interleukin 6 (IL-6) receptor in vivo: sensitization of human soluble IL-6 receptor transgenic mice towards IL-6 and prolongation of the plasma half-life of IL-6. J Exp Med 1996;183:1399–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Jones SA, Jenkins BJ.. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat Rev Immunol 2018;18:773–89. [DOI] [PubMed] [Google Scholar]
- 32. Reeh H, Rudolph N, Billing U. et al. Response to IL-6 trans- and IL-6 classic signalling is determined by the ratio of the IL-6 receptor alpha to gp130 expression: fusing experimental insights and dynamic modelling. Cell Commun Signal 2019;17:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Zegeye MM, Lindkvist M, Falker K. et al. Activation of the JAK/STAT3 and PI3K/AKT pathways are crucial for IL-6 trans-signaling-mediated pro-inflammatory response in human vascular endothelial cells. Cell Commun Signal 2018;16:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Garbers C, Thaiss W, Jones GW. et al. Inhibition of classic signaling is a novel function of soluble glycoprotein 130 (sgp130), which is controlled by the ratio of interleukin 6 and soluble interleukin 6 receptor. J Biol Chem 2011;286:42959–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Jostock T, Mullberg J, Ozbek S. et al. Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur J Biochem 2001;268:160–7. [DOI] [PubMed] [Google Scholar]
- 36. Narazaki M, Yasukawa K, Saito T. et al. Soluble forms of the interleukin-6 signal-transducing receptor component gp130 in human serum possessing a potential to inhibit signals through membrane-anchored gp130. Blood 1993;82:1120–6. [PubMed] [Google Scholar]
- 37. Heink S, Yogev N, Garbers C. et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat Immunol 2017;18:74–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Houssiau FA, Devogelaer JP, Van Damme J, de Deuxchaisnes CN, Van Snick J.. Interleukin-6 in synovial fluid and serum of patients with rheumatoid arthritis and other inflammatory arthritides. Arthritis Rheum 1988;31:784–8. [DOI] [PubMed] [Google Scholar]
- 39. Kotake S, Sato K, Kim KJ. et al. Interleukin-6 and soluble interleukin-6 receptors in the synovial fluids from rheumatoid arthritis patients are responsible for osteoclast-like cell formation. J Bone Miner Res 2009;11:88–95. [DOI] [PubMed] [Google Scholar]
- 40. Beekhuizen M, Gierman LM, van Spil WE. et al. An explorative study comparing levels of soluble mediators in control and osteoarthritic synovial fluid. Osteoarthritis Cartilage 2013;21:918–22. [DOI] [PubMed] [Google Scholar]
- 41. Sohn DH, Sokolove J, Sharpe O. et al. Plasma proteins present in osteoarthritic synovial fluid can stimulate cytokine production via toll-like receptor 4. Arthritis Res Ther 2012;14:R7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Wang ZW, Chen L, Hao XR. et al. Elevated levels of interleukin-1beta, interleukin-6, tumor necrosis factor-alpha and vascular endothelial growth factor in patients with knee articular cartilage injury. World J Clin Cases 2019;7:1262–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Livshits G, Zhai G, Hart DJ. et al. Interleukin-6 is a significant predictor of radiographic knee osteoarthritis: the Chingford Study. Arthritis Rheum 2009;60:2037–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Stannus O, Jones G, Cicuttini F. et al. Circulating levels of IL-6 and TNF-alpha are associated with knee radiographic osteoarthritis and knee cartilage loss in older adults. Osteoarthritis Cartilage 2010;18:1441–7. [DOI] [PubMed] [Google Scholar]
- 45. Goekoop RJ, Kloppenburg M, Kroon HM. et al. Low innate production of interleukin-1beta and interleukin-6 is associated with the absence of osteoarthritis in old age. Osteoarthritis Cartilage 2010;18:942–7. [DOI] [PubMed] [Google Scholar]
- 46. Siqueira MB, Frangiamore S, Klika AK. et al. Comparison of synovial fluid cytokine levels between traumatic knee injury and end-stage osteoarthritis. J Knee Surg 2016;30:128–33. [DOI] [PubMed] [Google Scholar]
- 47. Cuellar VG, Cuellar JM, Kirsch T, Strauss EJ.. Correlation of synovial fluid biomarkers with cartilage pathology and associated outcomes in knee arthroscopy. Arthroscopy 2016;32:475–85. [DOI] [PubMed] [Google Scholar]
- 48. Larsson S, Englund M, Struglics A, Lohmander LS.. Interleukin-6 and tumor necrosis factor alpha in synovial fluid are associated with progression of radiographic knee osteoarthritis in subjects with previous meniscectomy. Osteoarthritis Cartilage 2015;23:1906–14. [DOI] [PubMed] [Google Scholar]
- 49. Tsuchida AI, Beekhuizen M, Rutgers M. et al. Interleukin-6 is elevated in synovial fluid of patients with focal cartilage defects and stimulates cartilage matrix production in an in vitro regeneration model. Arthritis Res Ther 2012;14:R262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Tsuchida AI, Beekhuizen M, t Hart MC. et al. Cytokine profiles in the joint depend on pathology, but are different between synovial fluid, cartilage tissue and cultured chondrocytes. Arthritis Res Ther 2014;16:441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Laavola M, Leppanen T, Hamalainen M. et al. IL-6 in osteoarthritis: effects of pine stilbenoids. Molecules 2018;24:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Doß F, Menard J, Hauschild M. et al. Elevated IL-6 levels in the synovial fluid of osteoarthritis patients stem from plasma cells. Scand J Rheumatol 2007;36:136–9. [DOI] [PubMed] [Google Scholar]
- 53. D’Agostino MA, Conaghan P, Le Bars M. et al. EULAR report on the use of ultrasonography in painful knee osteoarthritis. Part 1: prevalence of inflammation in osteoarthritis. Ann Rheum Dis 2005;64:1703–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Guerne PA, Zuraw BL, Vaughan JH, Carson DA, Lotz M.. Synovium as a source of interleukin 6 in vitro. Contribution to local and systemic manifestations of arthritis. J Clin Invest 1989;83:585–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Bondeson J, Wainwright SD, Lauder S, Amos N, Hughes CE.. The role of synovial macrophages and macrophage-produced cytokines in driving aggrecanases, matrix metalloproteinases, and other destructive and inflammatory responses in osteoarthritis. Arthritis Res Ther 2006;8:R187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Nguyen HN, Noss EH, Mizoguchi F. et al. Autocrine loop involving IL-6 family member LIF, LIF receptor, and STAT4 drives sustained fibroblast production of inflammatory mediators. Immunity 2017;46:220–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Clockaerts S, Bastiaansen-Jenniskens YM, Runhaar J. et al. The infrapatellar fat pad should be considered as an active osteoarthritic joint tissue: a narrative review. Osteoarthritis Cartilage 2010;18:876–82. [DOI] [PubMed] [Google Scholar]
- 58. Distel E, Cadoudal T, Durant S. et al. The infrapatellar fat pad in knee osteoarthritis: an important source of interleukin-6 and its soluble receptor. Arthritis Rheum 2009;60:3374–7. [DOI] [PubMed] [Google Scholar]
- 59. He J, Jiang Y, Alexander PG. et al. Infrapatellar fat pad aggravates degeneration of acute traumatized cartilage: a possible role for interleukin-6. Osteoarthritis Cartilage 2017;25:138–45. [DOI] [PubMed] [Google Scholar]
- 60. Pearson MJ, Herndler-Brandstetter D, Tariq MA. et al. IL-6 secretion in osteoarthritis patients is mediated by chondrocyte-synovial fibroblast cross-talk and is enhanced by obesity. Sci Rep 2017;7:3451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Lehrskov LL, Christensen RH.. The role of interleukin-6 in glucose homeostasis and lipid metabolism. Semin Immunopathol 2019;41:491–9. [DOI] [PubMed] [Google Scholar]
- 62. Tanaka T, Narazaki M, Kishimoto T.. IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol 2014;6:a016295–a016295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Desgeorges A, Gabay C, Silacci P. et al. Concentrations and origins of soluble interleukin 6 receptor-alpha in serum and synovial fluid. J Rheumatol 1997;24:1510–6. [PubMed] [Google Scholar]
- 64. Fujikawa Y, Shingu M, Torisu T, Itonaga I, Masumi S.. Synovial fluid and serum levels of soluble interleukin-6 receptor in patients with rheumatoid arthritis. Jpn J Rheumatol 1998;8:27–36. [Google Scholar]
- 65. Richards PJ, Nowell MA, Horiuchi S. et al. Functional characterization of a soluble gp130 isoform and its therapeutic capacity in an experimental model of inflammatory arthritis. Arthritis Rheum 2006;54:1662–72. [DOI] [PubMed] [Google Scholar]
- 66. Tanaka M, Kishimura M, Ozaki S. et al. Cloning of novel soluble gp130 and detection of its neutralizing autoantibodies in rheumatoid arthritis. J Clin Invest 2000;106:137–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Silacci P, Dayer JM, Desgeorges A. et al. Interleukin (IL)-6 and its soluble receptor induce TIMP-1 expression in synoviocytes and chondrocytes, and block IL-1-induced collagenolytic activity. J Biol Chem 1998;273:13625–9. [DOI] [PubMed] [Google Scholar]
- 68. Lotz M, Guerne PA.. Interleukin-6 induces the synthesis of tissue inhibitor of metalloproteinases-1/erythroid potentiating activity (TIMP-1/EPA). J Biol Chem 1991;266:2017–20. [PubMed] [Google Scholar]
- 69. Malfait AM, Verbruggen G, Veys EM. et al. Comparative and combined effects of interleukin 6, interleukin 1 beta, and tumor necrosis factor alpha on proteoglycan metabolism of human articular chondrocytes cultured in agarose. J Rheumatol 1994;21:314–20. [PubMed] [Google Scholar]
- 70. Kandel RA, Petelycky M, Dinarello CA. et al. Comparison of the effect of interleukin 6 and interleukin 1 on collagenase and proteoglycan production by chondrocytes. J Rheumatol 1990;17:953–7. [PubMed] [Google Scholar]
- 71. Nietfeld JJ, Wilbrink B, Helle M. et al. Interleukin-1-induced interleukin-6 is required for the inhibition of proteoglycan synthesis by interleukin-1 in human articular cartilage. Arthritis Rheum 1990;33:1695–701. [DOI] [PubMed] [Google Scholar]
- 72. Guerne PA, Desgeorges A, Jaspar JM. et al. Effects of IL-6 and its soluble receptor on proteoglycan synthesis and NO release by human articular chondrocytes: comparison with IL-1. Modulation by dexamethasone. Matrix Biol 1999;18:253–60. [DOI] [PubMed] [Google Scholar]
- 73. Jikko A, Wakisaka T, Iwamoto M. et al. Effects of interleukin-6 on proliferation and proteoglycan metabolism in articular chondrocyte cultures. Cell Biol Int 1998;22:615–21. [DOI] [PubMed] [Google Scholar]
- 74. Poree B, Kypriotou M, Chadjichristos C. et al. Interleukin-6 (IL-6) and/or soluble IL-6 receptor down-regulation of human type II collagen gene expression in articular chondrocytes requires a decrease of Sp1.Sp3 ratio and of the binding activity of both factors to the COL2A1 promoter. J Biol Chem 2008;283: 4850–65. [DOI] [PubMed] [Google Scholar]
- 75. Ryu JH, Yang S, Shin Y. et al. Interleukin-6 plays an essential role in hypoxia-inducible factor 2alpha-induced experimental osteoarthritic cartilage destruction in mice. Arthritis Rheum 2011;63:2732–43. [DOI] [PubMed] [Google Scholar]
- 76. Zanotti S, Canalis E.. Interleukin 6 mediates selected effects of Notch in chondrocytes. Osteoarthritis Cartilage 2013;21:1766–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Sahu N, Viljoen HJ, Subramanian A.. Continuous low-intensity ultrasound attenuates IL-6 and TNFalpha-induced catabolic effects and repairs chondral fissures in bovine osteochondral explants. BMC Musculoskelet Disord 2019;20:193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Nasi S, So A, Combes C, Daudon M, Busso N.. Interleukin-6 and chondrocyte mineralisation act in tandem to promote experimental osteoarthritis. Ann Rheum Dis 2016;75:1372–9. [DOI] [PubMed] [Google Scholar]
- 79. Mathy-Hartert M, Hogge L, Sanchez C. et al. Interleukin-1beta and interleukin-6 disturb the antioxidant enzyme system in bovine chondrocytes: a possible explanation for oxidative stress generation. Osteoarthritis Cartilage 2008;16:756–63. [DOI] [PubMed] [Google Scholar]
- 80. Smeets RL, Veenbergen S, Arntz OJ. et al. A novel role for suppressor of cytokine signaling 3 in cartilage destruction via induction of chondrocyte desensitization toward insulin-like growth factor. Arthritis Rheum 2006;54:1518–28. [DOI] [PubMed] [Google Scholar]
- 81. Gui T, He BS, Gan Q, Yang C.. Enhanced SOCS3 in osteoarthiritis may limit both proliferation and inflammation. Biotech Histochem 2017;92:107–14. [DOI] [PubMed] [Google Scholar]
- 82. Liu X, Croker BA, Campbell IK. et al. Key role of suppressor of cytokine signaling 3 in regulating gp130 cytokine-induced signaling and limiting chondrocyte responses during murine inflammatory arthritis. Arthritis Rheumatol 2014;66:2391–402. [DOI] [PubMed] [Google Scholar]
- 83. Bauer J, Lengyel G, Bauer TM, Acs G, Gerok W.. Regulation of interleukin-6 receptor expression in human monocytes and hepatocytes. FEBS Lett 1989;249:27–30. [DOI] [PubMed] [Google Scholar]
- 84. Rowan AD, Koshy PJ, Shingleton WD. et al. Synergistic effects of glycoprotein 130 binding cytokines in combination with interleukin-1 on cartilage collagen breakdown. Arthritis Rheum 2001;44:1620–32. [DOI] [PubMed] [Google Scholar]
- 85. Sui Y, Lee JH, DiMicco MA. et al. Mechanical injury potentiates proteoglycan catabolism induced by interleukin-6 with soluble interleukin-6 receptor and tumor necrosis factor alpha in immature bovine and adult human articular cartilage. Arthritis Rheum 2009;60:2985–96. [DOI] [PubMed] [Google Scholar]
- 86. Madej W, van Caam A, Blaney Davidson EN, van der Kraan PM, Buma P.. Physiological and excessive mechanical compression of articular cartilage activates Smad2/3P signaling. Osteoarthritis Cartilage 2014;22:1018–25. [DOI] [PubMed] [Google Scholar]
- 87. Li Y, Frank EH, Wang Y. et al. Moderate dynamic compression inhibits pro-catabolic response of cartilage to mechanical injury, tumor necrosis factor-alpha and interleukin-6, but accentuates degradation above a strain threshold. Osteoarthritis Cartilage 2013;21:1933–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Flannery CR, Little CB, Hughes CE. et al. IL-6 and its soluble receptor augment aggrecanase-mediated proteoglycan catabolism in articular cartilage. Matrix Biol 2000;19:549–53. [DOI] [PubMed] [Google Scholar]
- 89. Legendre F, Dudhia J, Pujol JP, Bogdanowicz P.. JAK/STAT but not ERK1/ERK2 pathway mediates interleukin (IL)-6/soluble IL-6R down-regulation of Type II collagen, aggrecan core, and link protein transcription in articular chondrocytes. Association with a down-regulation of SOX9 expression. J Biol Chem 2003;278:2903–12. [DOI] [PubMed] [Google Scholar]
- 90. Legendre F, Bogdanowicz P, Boumediene K, Pujol JP.. Role of interleukin 6 (IL-6)/IL-6R-induced signal tranducers and activators of transcription and mitogen-activated protein kinase/extracellular. J Rheumatol 2005;32:1307–16. [PubMed] [Google Scholar]
- 91. Ito A, Itoh Y, Sasaguri Y, Morimatsu M, Mori Y.. Effects of interleukin-6 on the metabolism of connective tissue components in rheumatoid synovial fibroblasts. Arthritis Rheum 1992;35:1197–201. [DOI] [PubMed] [Google Scholar]
- 92. Mimata Y, Kamataki A, Oikawa S. et al. Interleukin-6 upregulates expression of ADAMTS-4 in fibroblast-like synoviocytes from patients with rheumatoid arthritis. Int J Rheum Dis 2012;15:36–44. [DOI] [PubMed] [Google Scholar]
- 93. Mihara M, Moriya Y, Kishimoto T, Ohsugi Y.. Interleukin-6 (IL-6) induces the proliferation of synovial fibroblastic cells in the presence of soluble IL-6 receptor. Br J Rheumatol 1995;34:321–5. [DOI] [PubMed] [Google Scholar]
- 94. Tamura T, Udagawa N, Takahashi N. et al. Soluble interleukin-6 receptor triggers osteoclast formation by interleukin 6. Proc Natl Acad Sci USA 1993;90:11924–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Blanchard F, Duplomb L, Baud’huin M, Brounais B.. The dual role of IL-6-type cytokines on bone remodeling and bone tumors. Cytokine Growth Factor Rev 2009;20:19–28. [DOI] [PubMed] [Google Scholar]
- 96. McGregor NE, Murat M, Elango J. et al. IL-6 exhibits both cis- and trans-signaling in osteocytes and osteoblasts, but only trans-signaling promotes bone formation and osteoclastogenesis. J Biol Chem 2019;294:7850–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Feng W, Liu H, Luo T. et al. Combination of IL-6 and sIL-6R differentially regulate varying levels of RANKL-induced osteoclastogenesis through NF-kappaB, ERK and JNK signaling pathways. Sci Rep 2017;7:41411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Perez-Baos S, Prieto-Potin I, Roman-Blas JA. et al. Mediators and patterns of muscle loss in chronic systemic inflammation. Front Physiol 2018;9:409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. De Ceuninck F, Fradin A, Pastoureau P.. Bearing arms against osteoarthritis and sarcopenia: when cartilage and skeletal muscle find common interest in talking together. Drug Discov Today 2014;19:305–11. [DOI] [PubMed] [Google Scholar]
- 100. Santos ML, Gomes WF, Pereira DS. et al. Muscle strength, muscle balance, physical function and plasma interleukin-6 (IL-6) levels in elderly women with knee osteoarthritis (OA). Arch Gerontol Geriatr 2011;52:322–6. [DOI] [PubMed] [Google Scholar]
- 101. Levinger P, Caldow MK, Feller JA. et al. Association between skeletal muscle inflammatory markers and walking pattern in people with knee osteoarthritis. Arthritis Care Res 2011;63:1715–21. [DOI] [PubMed] [Google Scholar]
- 102. Munoz-Canoves P, Scheele C, Pedersen BK, Serrano AL.. Interleukin-6 myokine signaling in skeletal muscle: a double-edged sword? FEBS J 2013;280:4131–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Wu X, Cao L, Li F. et al. Interleukin-6 from subchondral bone mesenchymal stem cells contributes to the pathological phenotypes of experimental osteoarthritis. Am J Transl Res 2018;10:1143–54. [PMC free article] [PubMed] [Google Scholar]
- 104. Langdon C, Kerr C, Hassen M. et al. Murine oncostatin M stimulates mouse synovial fibroblasts in vitro and induces inflammation and destruction in mouse joints in vivo. Am J Pathol 2000;157:1187–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Latourte A, Cherifi C, Maillet J. et al. Systemic inhibition of IL-6/Stat3 signalling protects against experimental osteoarthritis. Ann Rheum Dis 2017;76:748–55. [DOI] [PubMed] [Google Scholar]
- 106. Kamiya N, Kuroyanagi G, Aruwajoye O, Kim H.. IL6 receptor blockade preserves articular cartilage and increases bone volume following ischemic osteonecrosis in immature mice. Osteoarthritis Cartilage 2019;27:326–35. [DOI] [PubMed] [Google Scholar]
- 107. Greene MA, Loeser RF.. Aging-related inflammation in osteoarthritis. Osteoarthritis Cartilage 2015;23:1966–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Maggio M, Guralnik JM, Longo DL, Ferrucci L.. Interleukin-6 in aging and chronic disease: a magnificent pathway. J Gerontol A Biol Sci Med Sci 2006;61:575–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. de Hooge AS, van de Loo FA, Bennink MB. et al. Male IL-6 gene knock out mice developed more advanced osteoarthritis upon aging. Osteoarthritis Cartilage 2005;13:66–73. [DOI] [PubMed] [Google Scholar]
- 110. Bellido T, Jilka RL, Boyce BF. et al. Regulation of interleukin-6, osteoclastogenesis, and bone mass by androgens. The role of the androgen receptor. J Clin Invest 1995;95:2886–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Wang LH, Yang XY, Mihalic K. et al. Activation of estrogen receptor blocks interleukin-6-inducible cell growth of human multiple myeloma involving molecular cross-talk between estrogen receptor and STAT3 mediated by co-regulator PIAS3. J Biol Chem 2001;276:31839–44. [DOI] [PubMed] [Google Scholar]
- 112. Fontanini G, Campani D, Roncella M. et al. Expression of interleukin 6 (IL-6) correlates with oestrogen receptor in human breast carcinoma. Br J Cancer 1999;80:579–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Liu X, Liu R, Croker BA. et al. Distinctive pro-inflammatory gene signatures induced in articular chondrocytes by oncostatin M and IL-6 are regulated by suppressor of cytokine signaling-3. Osteoarthritis Cartilage 2015;23:1743–54. [DOI] [PubMed] [Google Scholar]
- 114. de Hooge AS, van de Loo FA, Bennink MB. et al. Adenoviral transfer of murine oncostatin M elicits periosteal bone apposition in knee joints of mice, despite synovial inflammation and up-regulated expression of interleukin-6 and receptor activator of nuclear factor-kappa B ligand. Am J Pathol 2002;160:1733–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Hall MD, Murray CA, Valdez MJ, Perantoni AO.. Mesoderm-specific Stat3 deletion affects expression of Sox9 yielding Sox9-dependent phenotypes. PLoS Genet 2017;13:e1006610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Sims NA, Jenkins BJ, Nakamura A. et al. Interleukin-11 receptor signaling is required for normal bone remodeling. J Bone Miner Res 2005;20:1093–102. [DOI] [PubMed] [Google Scholar]
- 117. Ware CB, Horowitz MC, Renshaw BR. et al. Targeted disruption of the low-affinity leukemia inhibitory factor receptor gene causes placental, skeletal, neural and metabolic defects and results in perinatal death. Development 1995;121:1283–99. [DOI] [PubMed] [Google Scholar]
- 118. Shkhyan R, Van Handel B, Bogdanov J. et al. Drug-induced modulation of gp130 signalling prevents articular cartilage degeneration and promotes repair. Ann Rheum Dis 2018;77:760–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Stone JH, Tuckwell K, Dimonaco S. et al. Trial of tocilizumab in giant-cell arteritis. N Engl J Med 2017;377:317–28. [DOI] [PubMed] [Google Scholar]
- 120. Barreto G, Soliymani R, Baumann M. et al. Functional analysis of synovial fluid from osteoarthritic knee and carpometacarpal joints unravels different molecular profiles. Rheumatology 2019;58:897–907. [DOI] [PubMed] [Google Scholar]
- 121. Teunis T, Beekhuizen M, Van Osch GV. et al. Soluble mediators in posttraumatic wrist and primary knee osteoarthritis. Arch Bone Jt Surg 2014;2:146–50. [PMC free article] [PubMed] [Google Scholar]
- 122. Scanzello CR. Role of low-grade inflammation in osteoarthritis. Curr Opin Rheumatol 2017;29:79–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Damman W, Liu R, Bloem JL. et al. Bone marrow lesions and synovitis on MRI associate with radiographic progression after 2 years in hand osteoarthritis. Ann Rheum Dis 2017;76:214–7. [DOI] [PubMed] [Google Scholar]
- 124. Haugen IK, Mathiessen A, Slatkowsky-Christensen B. et al. Synovitis and radiographic progression in non-erosive and erosive hand osteoarthritis: is erosive hand osteoarthritis a separate inflammatory phenotype? Osteoarthritis Cartilage 2016;24:647–54. [DOI] [PubMed] [Google Scholar]
- 125. Chevalier X, Ravaud P, Maheu E. et al. Adalimumab in patients with hand osteoarthritis refractory to analgesics and NSAIDs: a randomised, multicentre, double-blind, placebo-controlled trial. Ann Rheum Dis 2015;74:1697–705. [DOI] [PubMed] [Google Scholar]
- 126. Thomas AC, Hubbard-Turner T, Wikstrom EA, Palmieri-Smith RM.. Epidemiology of posttraumatic osteoarthritis. J Athl Train 2017;52:491–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Struglics A, Larsson S, Kumahashi N, Frobell R, Lohmander LS.. Changes in cytokines and aggrecan ARGS neoepitope in synovial fluid and serum and in C-terminal crosslinking telopeptide of type II collagen and n-terminal crosslinking telopeptide of type I collagen in urine over five years after anterior cruciate ligament rupture: an exploratory analysis in the knee anterior cruciate ligament, nonsurgical versus surgical treatment trial. Arthritis Rheumatol 2015;67:1816–25. [DOI] [PubMed] [Google Scholar]
- 128. Watt FE, Paterson E, Freidin A. et al. Acute molecular changes in synovial fluid following human knee injury: association with early clinical outcomes. Arthritis Rheumatol 2016;68:2129–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Garbers C, Heink S, Korn T, Rose-John S.. Interleukin-6: designing specific therapeutics for a complex cytokine. Nat Rev Drug Discov 2018;17:395–412. [DOI] [PubMed] [Google Scholar]
- 130. O'Shea JJ, Kontzias A, Yamaoka K, Tanaka Y, Laurence A.. Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis 2013;72:ii111–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Kjelgaard-Petersen CF, Sharma N, Kayed A. et al. Tofacitinib and TPCA-1 exert chondroprotective effects on extracellular matrix turnover in bovine articular cartilage ex vivo. Biochem Pharmacol 2019;165:91–8. [DOI] [PubMed] [Google Scholar]
- 132. Steen-Louws C, Popov-Celeketic J, Mastbergen SC. et al. IL4-10 fusion protein has chondroprotective, anti-inflammatory and potentially analgesic effects in the treatment of osteoarthritis. Osteoarthritis Cartilage 2018;26:1127–35. [DOI] [PubMed] [Google Scholar]
- 133. Zhang Z, Li L, Yang W. et al. The effects of different doses of IGF-1 on cartilage and subchondral bone during the repair of full-thickness articular cartilage defects in rabbits. Osteoarthritis Cartilage 2017;25:309–20. [DOI] [PubMed] [Google Scholar]
- 134. Mihara M, Kasutani K, Okazaki M. et al. Tocilizumab inhibits signal transduction mediated by both mIL-6R and sIL-6R, but not by the receptors of other members of IL-6 cytokine family. Int Immunopharmacol 2005;5:1731–40. [DOI] [PubMed] [Google Scholar]
- 135. Atreya R, Mudter J, Finotto S. et al. Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo. Nat Med 2000;6:583–8. [DOI] [PubMed] [Google Scholar]



